Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Anal Cell Pathol (Amst) ; 2019: 7405602, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31428552

RESUMEN

AIMS: Inflammation was closely associated with diabetes-related endothelial dysfunction. C1q/tumor necrosis factor-related protein 3 (CTRP3) is a member of the CTRP family and can provide cardioprotection in many cardiovascular diseases via suppressing the production of inflammatory factors. However, the role of CTRP3 in high glucose- (HG-) related endothelial dysfunction remains unclear. This study evaluates the effects of CTRP3 on HG-induced cell inflammation and apoptosis. MATERIALS AND METHODS: To prevent high glucose-induced cell injury, human umbilical vein endothelial cells (HUVECs) were pretreated with recombinant CTRP3 for 1 hour followed by normal glucose (5.5 mmol/l) or high glucose (33 mmol/l) treatment. After that, cell apoptosis and inflammatory factors were determined. RESULTS: Our results demonstrated that CTRP3 mRNA and protein expression were significantly decreased after HG exposure in HUVECs. Recombinant human CTRP3 inhibited HG-induced accumulation of inflammatory factors and cell loss in HUVECs. CTRP3 treatment also increased the phosphorylation levels of protein kinase B (AKT/PKB) and the mammalian target of rapamycin (mTOR) in HUVECs. CTRP3 lost its inhibitory effects on HG-induced cell inflammation and apoptosis after AKT inhibition. Knockdown of endogenous CTRP3 in HUVECs resulted in increased inflammation and decreased cell viability in vitro. CONCLUSIONS: Taken together, these findings indicated that CTRP3 treatment blocked the accumulation of inflammatory factors and cell loss in HUVECs after HG exposure through the activation of AKT-mTOR signaling pathway. Thus, CTRP3 may be a potential therapeutic drug for the prevention of diabetes-related endothelial dysfunction.


Asunto(s)
Citoprotección , Glucosa/toxicidad , Células Endoteliales de la Vena Umbilical Humana/patología , Sustancias Protectoras/metabolismo , Factores de Necrosis Tumoral/metabolismo , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citoprotección/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Eliminación de Gen , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factores de Necrosis Tumoral/deficiencia
2.
Sci Immunol ; 3(30)2018 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-30578350

RESUMEN

Regulatory T cells (Tregs) are closely related to TH17 cells and use aspects of the TH17-differentiation program for optimal immune regulation. In several chronic inflammatory human diseases, Tregs express IL-17A, suggesting that dysregulation of TH17-associated pathways in Tregs may result in either loss of suppressive function and/or conversion into pathogenic cells. The pathways that regulate the TH17 program in Tregs are poorly understood. We have identified two TNF receptor superfamily (TNFRSF) members, CD27 and OX40, that are preferentially expressed by skin-resident Tregs Both CD27 and OX40 signaling suppressed the expression of TH17-associated genes from Tregs in a cell-intrinsic manner in vitro and in vivo. However, only OX40 played a nonredundant role in promoting Treg accumulation. Tregs that lacked both CD27 and OX40 were defective in controlling skin inflammation and expressed high levels of IL-17A, as well as the master TH17 transcription factor, RORγt. Last, we found that CD27 expression was inversely correlated with Treg IL-17 production in skin of patients with psoriasis and hidradenitis suppurativa. Together, our results suggest that TNFRSF members play both redundant and distinct roles in regulating Treg plasticity in tissues.


Asunto(s)
Glicoproteínas de Membrana/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Factores de Necrosis Tumoral/inmunología , Animales , Diferenciación Celular/inmunología , Glicoproteínas de Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ligando OX40 , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/deficiencia , Factores de Necrosis Tumoral/deficiencia
3.
J Immunol ; 197(10): 4110-4117, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27733552

RESUMEN

Innate immune responses that are crucial for control of infection are often targeted by microbial pathogens. Blockade of NF-κB and MAPK signaling by the Yersinia virulence factor YopJ inhibits cytokine production by innate immune cells but also triggers cell death. This cell death requires RIPK1 kinase activity and caspase-8, which are engaged by TLR4 and the adaptor protein TRIF. Nevertheless, TLR4- and TRIF-deficient cells undergo significant apoptosis, implicating TLR4/TRIF-independent pathways in the death of Yersinia-infected cells. In this article, we report a key role for TNF/TNFR1 in Yersinia-induced cell death of murine macrophages, which occurs despite the blockade of NF-κB and MAPK signaling imposed by Yersinia on infected cells. Intriguingly, direct analysis of YopJ injection revealed a heterogeneous population of injection-high and injection-low cells, and demonstrated that TNF expression came from the injection-low population. Moreover, TNF production by this subpopulation was necessary for maximal apoptosis in the population of highly injected cells, and TNFR-deficient mice displayed enhanced susceptibility to Yersinia infection. These data demonstrate an important role for collaboration between TNF and pattern recognition receptor signals in promoting maximal apoptosis during bacterial infection, and demonstrate that heterogeneity in virulence factor injection and cellular responses play an important role in promoting anti-Yersinia immune defense.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Apoptosis , Macrófagos/microbiología , Macrófagos/fisiología , Factores de Necrosis Tumoral/metabolismo , Yersiniosis/inmunología , Yersinia pseudotuberculosis/patogenicidad , Animales , Proteínas Bacterianas/genética , Caspasa 1/metabolismo , Muerte Celular , Inmunidad Innata , L-Lactato Deshidrogenasa/metabolismo , Ratones , Plásmidos/genética , Transducción de Señal , Receptor Toll-Like 4/inmunología , Factores de Necrosis Tumoral/deficiencia , Factores de Necrosis Tumoral/inmunología , Yersinia pseudotuberculosis/inmunología
4.
J Pathol ; 239(1): 109-21, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26924336

RESUMEN

Tumour necrosis factor-like weak inducer of apoptosis (TWEAK) and its receptor fibroblast growth factor-inducible 14 (Fn14) have been associated with liver regeneration in vivo. To further investigate the role of this pathway we examined their expression in human fibrotic liver disease and the effect of pathway deficiency in a murine model of liver fibrosis. The expression of Fn14 and TWEAK in normal and diseased human and mouse liver tissue and primary human hepatic stellate cells (HSCs) were investigated by qPCR, western blotting and immunohistochemistry. In addition, the levels of Fn14 in HSCs following pro-fibrogenic and pro-inflammatory stimuli were assessed and the effects of exogenous TWEAK on HSCs proliferation and activation were studied in vitro. Carbon tetrachloride (CCl4 ) was used to induce acute and chronic liver injury in TWEAK KO mice. Elevated expression of both Fn14 and TWEAK were detected in acute and chronic human liver injury, and co-localized with markers of activated HSCs. Fn14 levels were low in quiescent HSCs but were significantly induced in activated HSCs, which could be further enhanced with the profibrogenic cytokine TGFß in vitro. Stimulation with recombinant TWEAK induced proliferation but not further HSCs activation. Fn14 gene expression was also significantly up-regulated in CCl4 models of hepatic injury whereas TWEAK KO mice showed reduced levels of liver fibrosis following chronic CCl4 injury. In conclusion, TWEAK/Fn14 interaction leads to the progression of fibrotic liver disease via direct modulation of HSCs proliferation, making it a potential therapeutic target for liver fibrosis.


Asunto(s)
Células Estrelladas Hepáticas/patología , Cirrosis Hepática/etiología , Receptores del Factor de Necrosis Tumoral/metabolismo , Factores de Necrosis Tumoral/deficiencia , Actinas/metabolismo , Animales , Tetracloruro de Carbono/toxicidad , Proliferación Celular , Enfermedad Hepática Inducida por Sustancias y Drogas , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas , Citocina TWEAK , Progresión de la Enfermedad , Fibroblastos/metabolismo , Humanos , Cirrosis Hepática/patología , Masculino , Ratones Noqueados , ARN Mensajero/metabolismo , Factor de Transcripción SOX9/metabolismo , Receptor de TWEAK , Factores de Necrosis Tumoral/metabolismo , Factores de Necrosis Tumoral/farmacología , Regulación hacia Arriba/fisiología
5.
Eur J Clin Invest ; 45(1): 27-35, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25443800

RESUMEN

BACKGROUND: To evaluate the inflammatory axis mediated by tumour necrosis factor-like weak inducer of apoptosis (TWEAK) and its scavenger receptor CD163 during pregnancy and their influence on insulin sensitivity in normal pregnancy and in gestational diabetes mellitus (GDM). MATERIALS AND METHODS: One hundred and thirty seven women with one singleton pregnancy, 71 with normal glucose tolerance (NGT) and 66 with GDM were studied. Glucose metabolism was assessed by oral glucose tolerance test. Serum concentrations of soluble TWEAK (sTWEAK) and CD163 (sCD163) and insulin resistance (HOMA-IR index) were determined in maternal blood drawn at recruitment, in the early third trimester. Offspring weight and height were assessed at birth. RESULTS: Women with GDM had lower circulating sTWEAK concentrations than control NGT group (237·8 (192·1-301·0) pg/mL vs. 277·2 (206·4-355·7) pg/mL; P = 0·013). sTWEAK was negatively associated with the presence of GDM (r = -0·212; P = 0·013), HOMA-IR index (r = -0·197; P = 0·021) and ponderal index of the newborn (r = -0·196; P = 0·025), but positively with HDL cholesterol (r = 0·283; P = 0·001). In multiple regression analysis, sTWEAK concentration emerged as one of the main predictors of insulin resistance, along with BMI, triglycerides and low concentrations of HDL cholesterol (R(2)  = 0·486; P < 0·001). No relationship was found between HOMA-IR index and sCD163 or sCD163/sTWEAK ratio. CONCLUSIONS: sTWEAK concentrations are lower in patients with GDM compared with healthy pregnant women, and low concentrations of sTWEAK are associated with insulin resistance. These findings suggest that insulin resistance during pregnancy is closely linked to inflammatory imbalance and sTWEAK may represent a new candidate associated with GDM.


Asunto(s)
Diabetes Gestacional/etiología , Factores de Necrosis Tumoral/deficiencia , Adulto , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Estudios de Casos y Controles , Citocina TWEAK , Femenino , Humanos , Resistencia a la Insulina/fisiología , Embarazo , Estudios Prospectivos , Receptores de Superficie Celular/metabolismo , Análisis de Regresión
6.
Actas dermo-sifiliogr. (Ed. impr.) ; 105(8): 752-761, oct. 2014. tab, ilus
Artículo en Español | IBECS | ID: ibc-128811

RESUMEN

Paradójicamente se han descrito casos de inducción o empeoramiento de una psoriasis durante el tratamiento con todos los agentes anti-factor de necrosis tumoral α (anti-TNFα) (infliximab, etanercept, adalimumab y certolizumab). Se ha postulado que la alteración del equilibrio entre el TNFα y el interferón α estaría implicada en su etiopatogenia. Clínicamente se distinguen varios patrones de reacciones psoriasiformes paradójicas: la psoriasis de novo en pacientes que no han presentado anteriormente esta enfermedad y que reciben este tratamiento por otra enfermedad inflamatoria, que es la más frecuente y la mejor descrita, y la exacerbación de una psoriasis preexistente durante la terapia anti-TNFα, que puede presentarse con o sin un cambio de morfología. En este trabajo realizamos una revisión de la literatura en relación con las características clínicas e histológicas de este tipo de reacciones, así como de su evolución y tratamiento, y planteamos un esquema de manejo en la práctica clínica


There have been reports of paradoxical induction or worsening of psoriasis during treatment with tumor necrosis factor (TNF) α agents (infliximab, etanercept, adalimumab, and certolizumab). It has been hypothesized that an imbalance between TNF-α and interferon α might have a role in the etiology and pathogenesis of these reactions. Paradoxical psoriasiform reactions can be divided clinically into de novo psoriasis and exacerbation of preexisting psoriasis. The first, which is more common and more extensively described in the literature, occurs in patients without a history of psoriasis who are receiving TNF-α therapy for another inflammatory disorder. The second can occur with or without changes in the morphology of the lesions. In this article, we review the literature on the clinical and histologic features of paradoxical psoriasiform reactions, analyze their clinical course and treatment, and propose a clinical management model for use in routine practice


Asunto(s)
Humanos , Masculino , Femenino , Psoriasis/inducido químicamente , Psoriasis/clasificación , Psoriasis/metabolismo , Psoriasis/diagnóstico , Factores de Necrosis Tumoral/efectos adversos , Factores de Necrosis Tumoral/análisis , Factores de Necrosis Tumoral/deficiencia
7.
J Biol Chem ; 288(49): 35159-69, 2013 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-24151074

RESUMEN

Satellite cell self-renewal is an essential process to maintaining the robustness of skeletal muscle regenerative capacity. However, extrinsic factors that regulate self-renewal of satellite cells are not well understood. Here, we demonstrate that TWEAK cytokine reduces the proportion of Pax7(+)/MyoD(-) cells (an index of self-renewal) on myofiber explants and represses multiple components of Notch signaling in satellite cell cultures. The number of Pax7(+) cells is significantly increased in skeletal muscle of TWEAK knock-out (KO) mice compared with wild-type in response to injury. Furthermore, Notch signaling is significantly elevated in cultured satellite cells and in regenerating myofibers of TWEAK-KO mice. Forced activation of Notch signaling through overexpression of the Notch1 intracellular domain (N1ICD) rescued the TWEAK-mediated inhibition of satellite cell self-renewal. TWEAK also activates the NF-κB transcription factor in satellite cells and inhibition of NF-κB significantly improved the number of Pax7(+) cells in TWEAK-treated cultures. Furthermore, our results demonstrate that a reciprocal interaction between NF-κB and Notch signaling governs the inhibitory effect of TWEAK on satellite cell self-renewal. Collectively, our study demonstrates that TWEAK suppresses satellite cell self-renewal through activating NF-κB and repressing Notch signaling.


Asunto(s)
FN-kappa B/metabolismo , Receptor Notch1/metabolismo , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo , Factores de Necrosis Tumoral/metabolismo , Animales , Apoptosis/fisiología , Proliferación Celular , Células Cultivadas , Citocina TWEAK , Mediadores de Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína MioD/genética , Proteína MioD/metabolismo , FN-kappa B/genética , Factor de Transcripción PAX7/genética , Factor de Transcripción PAX7/metabolismo , Estructura Terciaria de Proteína , Receptor Notch1/química , Receptor Notch1/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Regeneración/genética , Regeneración/fisiología , Transducción de Señal , Factores de Necrosis Tumoral/deficiencia , Factores de Necrosis Tumoral/genética
8.
J Neuroinflammation ; 9: 45, 2012 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-22394384

RESUMEN

BACKGROUND: Cerebral cortical neurons have a high vulnerability to the harmful effects of hypoxia. However, the brain has the ability to detect and accommodate to hypoxic conditions. This phenomenon, known as preconditioning, is a natural adaptive process highly preserved among species whereby exposure to sub-lethal hypoxia promotes the acquisition of tolerance to a subsequent lethal hypoxic injury. The cytokine tumor necrosis factor-like weak inducer of apoptosis (TWEAK) and its receptor fibroblast growth factor-inducible 14 (Fn14) are found in neurons and their expression is induced by exposure to sub-lethal hypoxia. Accordingly, in this work we tested the hypothesis that the interaction between TWEAK and Fn14 induces tolerance to lethal hypoxic and ischemic conditions. METHODS: Here we used in vitro and in vivo models of hypoxic and ischemic preconditioning, an animal model of transient middle cerebral artery occlusion and mice and neurons genetically deficient in TWEAK, Fn14, or tumor necrosis factor alpha (TNF-α) to investigate whether treatment with recombinant TWEAK or an increase in the expression of endogenous TWEAK renders neurons tolerant to lethal hypoxia. We used enzyme-linked immunosorbent assay to study the effect of TWEAK on the expression of neuronal TNF-α, Western blot analysis to investigate whether the effect of TWEAK was mediated by activation of mitogen-activated protein kinases and immunohistochemical techniques and quantitative real-time polymerase chain reaction analysis to study the effect of TWEAK on apoptotic cell death. RESULTS: We found that either treatment with recombinant TWEAK or an increase in the expression of TWEAK and Fn14 induce hypoxic and ischemic tolerance in vivo and in vitro. This protective effect is mediated by neuronal TNF-α and activation of the extracellular signal-regulated kinases 1 and 2 pathway via phosphorylation and inactivation of the B-cell lymphoma 2-associated death promoter protein. CONCLUSIONS: Our work indicate that the interaction between TWEAK and Fn14 triggers the activation of a cell signaling pathway that results in the induction of tolerance to lethal hypoxia and ischemia. These data indicate that TWEAK may be a potential therapeutic strategy to protect the brain from the devastating effects of an ischemic injury.


Asunto(s)
Apoptosis/efectos de los fármacos , Infarto de la Arteria Cerebral Media/patología , Precondicionamiento Isquémico/métodos , Fármacos Neuroprotectores/farmacología , Receptores del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factores de Necrosis Tumoral/farmacología , Animales , Apoptosis/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Corteza Cerebral/citología , Citocina TWEAK , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Glucosa/deficiencia , Hipoxia/tratamiento farmacológico , Etiquetado Corte-Fin in Situ , Técnicas In Vitro , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Receptores del Factor de Necrosis Tumoral/deficiencia , Receptor de TWEAK , Factores de Tiempo , Factor de Necrosis Tumoral alfa/deficiencia , Factores de Necrosis Tumoral/deficiencia , Factores de Necrosis Tumoral/metabolismo
9.
Nature ; 477(7364): 330-4, 2011 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-21804564

RESUMEN

Intestinal immune homeostasis depends on a tightly regulated cross talk between commensal bacteria, mucosal immune cells and intestinal epithelial cells (IECs). Epithelial barrier disruption is considered to be a potential cause of inflammatory bowel disease; however, the mechanisms regulating intestinal epithelial integrity are poorly understood. Here we show that mice with IEC-specific knockout of FADD (FADD(IEC-KO)), an adaptor protein required for death-receptor-induced apoptosis, spontaneously developed epithelial cell necrosis, loss of Paneth cells, enteritis and severe erosive colitis. Genetic deficiency in RIP3, a critical regulator of programmed necrosis, prevented the development of spontaneous pathology in both the small intestine and colon of FADD(IEC-KO) mice, demonstrating that intestinal inflammation is triggered by RIP3-dependent death of FADD-deficient IECs. Epithelial-specific inhibition of CYLD, a deubiquitinase that regulates cellular necrosis, prevented colitis development in FADD(IEC-KO) but not in NEMO(IEC-KO) mice, showing that different mechanisms mediated death of colonic epithelial cells in these two models. In FADD(IEC-KO) mice, TNF deficiency ameliorated colon inflammation, whereas MYD88 deficiency and also elimination of the microbiota prevented colon inflammation, indicating that bacteria-mediated Toll-like-receptor signalling drives colitis by inducing the expression of TNF and other cytokines. However, neither CYLD, TNF or MYD88 deficiency nor elimination of the microbiota could prevent Paneth cell loss and enteritis in FADD(IEC-KO) mice, showing that different mechanisms drive RIP3-dependent necrosis of FADD-deficient IECs in the small and large bowel. Therefore, by inhibiting RIP3-mediated IEC necrosis, FADD preserves epithelial barrier integrity and antibacterial defence, maintains homeostasis and prevents chronic intestinal inflammation. Collectively, these results show that mechanisms preventing RIP3-mediated epithelial cell death are critical for the maintenance of intestinal homeostasis and indicate that programmed necrosis of IECs might be implicated in the pathogenesis of inflammatory bowel disease, in which Paneth cell and barrier defects are thought to contribute to intestinal inflammation.


Asunto(s)
Colitis/patología , Colon/patología , Enteritis/patología , Células Epiteliales/patología , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Apoptosis , Enfermedad Crónica , Colitis/enzimología , Colitis/metabolismo , Colon/enzimología , Colon/metabolismo , Cisteína Endopeptidasas/metabolismo , Enzima Desubiquitinante CYLD , Enteritis/enzimología , Enteritis/metabolismo , Células Epiteliales/enzimología , Células Epiteliales/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/deficiencia , Enfermedades Inflamatorias del Intestino/enzimología , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Metagenoma/fisiología , Ratones , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/metabolismo , Necrosis , Células de Paneth/patología , Transducción de Señal , Factores de Necrosis Tumoral/deficiencia
10.
Neuroscience ; 171(4): 1256-64, 2010 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-20955770

RESUMEN

Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) and its receptor Fibroblast growth factor-inducible 14 (Fn14) are expressed in neurons. Here we demonstrate that TWEAK induces a dose-dependent increase in neuronal death and that this effect is independent of tumor necrosis factor alpha (TNF-α) and mediated by nuclear factor-kappa B (NF-κB) pathway activation. Incubation with TWEAK induces apoptotic cell death in wild-type (Wt) but not in Fn14 deficient (Fn14(-/-)) neurons. Intracerebral injection of TWEAK induces accumulation of poly(ADP-ribose) polymers (PAR) in Wt but not in Fn14(-/-) mice. Exposure to oxygen-glucose deprivation (OGD) conditions increases TWEAK and Fn14 mRNA expression in Wt neurons, and decreases cell survival in Wt but not in Fn14(-/-) or TWEAK deficient (TWEAK(-/-)) neurons. Experimental middle cerebral artery occlusion (MCAO) increases the expression of TWEAK and Fn14 mRNA and active caspase-3, and the cleavage of poly(ADP-ribose) polymerase-1 (PARP-1) with accumulation of PAR in the ischemic area in Wt but not Fn14(-/-) mice. Together, these results suggest a model where in response to hypoxia/ischemia the interaction between TWEAK and Fn14 in neurons induces PARP-1 activation with accumulation of PAR polymers and cell death via NF-κB pathway activation. This is a novel pathway for hypoxia/ischemia-induced TWEAK-mediated cell death and a potential therapeutic target for ischemic stroke.


Asunto(s)
Infarto de la Arteria Cerebral Media/enzimología , Infarto de la Arteria Cerebral Media/patología , Neuronas/fisiología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Células Cultivadas , Corteza Cerebral/citología , Citocina TWEAK , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Glucosa/deficiencia , Peróxido de Hidrógeno/farmacología , Hipoxia/patología , Infarto de la Arteria Cerebral Media/fisiopatología , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , Oxidantes/farmacología , Poli(ADP-Ribosa) Polimerasa-1 , Receptores del Factor de Necrosis Tumoral/deficiencia , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Receptor de TWEAK , Factor de Necrosis Tumoral alfa/farmacología , Factores de Necrosis Tumoral/deficiencia , Factores de Necrosis Tumoral/farmacología
11.
J Cell Biol ; 188(6): 833-49, 2010 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-20308426

RESUMEN

Skeletal muscle atrophy occurs in a variety of clinical settings, including cachexia, disuse, and denervation. Inflammatory cytokines have been shown to be mediators of cancer cachexia; however, the role of cytokines in denervation- and immobilization-induced skeletal muscle loss remains unknown. In this study, we demonstrate that a single cytokine, TNF-like weak inducer of apoptosis (TWEAK), mediates skeletal muscle atrophy that occurs under denervation conditions. Transgenic expression of TWEAK induces atrophy, fibrosis, fiber-type switching, and the degradation of muscle proteins. Importantly, genetic ablation of TWEAK decreases the loss of muscle proteins and spared fiber cross-sectional area, muscle mass, and strength after denervation. Expression of the TWEAK receptor Fn14 (fibroblast growth factor-inducible receptor 14) and not the cytokine is significantly increased in muscle upon denervation, demonstrating an unexpected inside-out signaling pathway; the receptor up-regulation allows for TWEAK activation of nuclear factor kappaB, causing an increase in the expression of the E3 ubiquitin ligase MuRF1. This study reveals a novel mediator of skeletal muscle atrophy and indicates that the TWEAK-Fn14 system is an important target for preventing skeletal muscle wasting.


Asunto(s)
Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Factores de Necrosis Tumoral/metabolismo , Animales , Citocina TWEAK , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Desnervación Muscular , Músculo Esquelético/inervación , Atrofia Muscular/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptor de TWEAK , Factores de Necrosis Tumoral/deficiencia , Factores de Necrosis Tumoral/genética
12.
J Immunol ; 181(9): 5990-6001, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18941188

RESUMEN

A T cell costimulatory molecule, OX40, contributes to T cell expansion, survival, and cytokine production. Although several roles for OX40 in CD8(+) T cell responses to tumors and viral infection have been shown, the precise function of these signals in the generation of memory CD8(+) T cells remains to be elucidated. To address this, we examined the generation and maintenance of memory CD8(+) T cells during infection with Listeria monocytogenes in the presence and absence of OX40 signaling. We used the expression of killer cell lectin-like receptor G1 (KLRG1), a recently reported marker, to distinguish between short-lived effector and memory precursor effector T cells (MPECs). Although OX40 was dispensable for the generation of effector T cells in general, the lack of OX40 signals significantly reduced the number and proportion of KLRG1(low) MPECs, and, subsequently, markedly impaired the generation of memory CD8(+) T cells. Moreover, memory T cells that were generated in the absence of OX40 signals in a host animal did not show self-renewal in a second host, suggesting that OX40 is important for the maintenance of memory T cells. Additional experiments making use of an inhibitory mAb against the OX40 ligand demonstrated that OX40 signals are essential during priming, not only for the survival of KLRG1(low) MPECs, but also for their self-renewing ability, both of which contribute to the homeostasis of memory CD8(+) T cells.


Asunto(s)
Adyuvantes Inmunológicos/fisiología , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Activación de Linfocitos/inmunología , Receptores OX40/fisiología , Transducción de Señal/inmunología , Enfermedad Aguda , Animales , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/microbiología , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Femenino , Memoria Inmunológica/genética , Lectinas Tipo C/antagonistas & inhibidores , Lectinas Tipo C/fisiología , Listeria monocytogenes/genética , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Listeriosis/metabolismo , Activación de Linfocitos/genética , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ligando OX40 , Receptores Inmunológicos , Receptores OX40/deficiencia , Receptores OX40/genética , Transducción de Señal/genética , Células Madre/inmunología , Células Madre/metabolismo , Células Madre/microbiología , Transactivadores/antagonistas & inhibidores , Transactivadores/fisiología , Factores de Necrosis Tumoral/deficiencia , Factores de Necrosis Tumoral/genética
13.
Am J Physiol Cell Physiol ; 294(6): C1436-42, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18385284

RESUMEN

Leukemia inhibitory factor (LIF) is a proinflammatory cytokine mediating a variety of central nervous system (CNS) responses to inflammatory stimuli. During lipopolysaccharide (LPS)-induced inflammation, blood concentrations of LIF increase, correlating with lethality of sepsis. Circulating LIF crosses the blood-brain barrier (BBB) by a saturable transport system. Here we determine how this transport system is regulated in neuroinflammation. Using transport assays that quantify the influx rate and volume of distribution of LIF in mice, we show that LPS facilitated the permeation of LIF from the blood to the brain without compromising the paracellular permeability of the BBB as determined by coadministration of fluorescein. Concurrently, gp130 (shared by the interleukin-6 family of cytokines), but not gp190 (the specific receptor for LIF) or cilliary neutrophic factor (CNTF-Ralpha, a unique receptor for cilliary neurotrophic factor that also uses gp130 and gp190), showed increased levels of mRNA and protein expression in cerebral microvessels from the LPS-treated mice. The upregulation of gp130 by LPS was at least partially mediated by vascular tumor necrosis factor receptor (TNFR)1 and TNFR2. This was shown by elevated TNFR1 and TNFR2 mRNA and protein in cerebral microvessels after LPS and by the absence of the LPS effect on gp130 in knockout mice lacking these receptors. The results show that neuroinflammation by LPS induces endothelial signaling and enhances cytokine transport across the BBB.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Permeabilidad Capilar , Factor Inhibidor de Leucemia/metabolismo , Sepsis/metabolismo , Factores de Necrosis Tumoral/metabolismo , Animales , Subunidad alfa del Receptor del Factor Neurotrófico Ciliar/genética , Subunidad alfa del Receptor del Factor Neurotrófico Ciliar/metabolismo , Receptor gp130 de Citocinas/genética , Receptor gp130 de Citocinas/metabolismo , Modelos Animales de Enfermedad , Fluoresceína/metabolismo , Colorantes Fluorescentes/metabolismo , Factor Inhibidor de Leucemia/sangre , Lipopolisacáridos , Masculino , Ratones , Ratones Noqueados , Microcirculación/metabolismo , ARN Mensajero/metabolismo , Receptores OSM-LIF/genética , Receptores OSM-LIF/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Sepsis/inducido químicamente , Factores de Tiempo , Factores de Necrosis Tumoral/deficiencia , Factores de Necrosis Tumoral/genética
14.
J Clin Invest ; 118(4): 1398-404, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18317595

RESUMEN

Tumors elicit antitumor immune responses, but over time they evolve and can escape immune control through various mechanisms, including the loss of the antigen to which the response is directed. The escape of antigen-loss variants (ALVs) is a major obstacle to T cell-based immunotherapy for cancer. However, cancers can be cured if both the number of CTLs and the expression of antigen are high enough to allow targeting of not only tumor cells, but also the tumor stroma. Here, we showed that IFN-gamma and TNF produced by CTLs were crucial for the elimination of established mouse tumors, including ALVs. In addition, both BM- and non-BM-derived stromal cells were required to express TNF receptors and IFN-gamma receptors for the elimination of ALVs. Although IFN-gamma and TNF were not required by CTLs for perforin-mediated killing of antigen-expressing tumor cells, the strong inference is that tumor antigen-specific CTLs must secrete IFN-gamma and TNF for destruction of tumor stroma. Therefore, bystander killing of ALVs may result from IFN-gamma and TNF acting on tumor stroma.


Asunto(s)
Antígenos/inmunología , Efecto Espectador/inmunología , Interferón gamma/biosíntesis , Neoplasias/inmunología , Neoplasias/metabolismo , Factores de Necrosis Tumoral/biosíntesis , Traslado Adoptivo , Animales , Línea Celular Tumoral , Interferón gamma/deficiencia , Interferón gamma/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trasplante de Neoplasias , Neoplasias/patología , Receptores de Interferón/inmunología , Receptores del Factor de Necrosis Tumoral/inmunología , Células del Estroma/inmunología , Células del Estroma/metabolismo , Linfocitos T/inmunología , Factores de Necrosis Tumoral/deficiencia , Factores de Necrosis Tumoral/genética
15.
Nat Neurosci ; 11(2): 135-42, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18176559

RESUMEN

Nerve growth factor (NGF) has an important role in regulating sympathetic neuron survival and target field innervation during development. Here we show that glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR), a member of the TNF superfamily, and its ligand (GITRL) are co-expressed in mouse sympathetic neurons when their axons are innervating their targets under the influence of target-derived NGF. In culture, GITRL enhanced NGF-promoted neurite growth from neonatal sympathetic neurons, and preventing GITR-GITRL interaction in these neurons or knocking down GITR inhibited NGF-promoted neurite growth without affecting neuronal survival. Tnfrsf18(-/-) (Gitr) neonates have reduced sympathetic innervation density in vivo compared with Gitr(+/+) littermates. GITR activation is required for the phosphorylation of extracellular signal-regulated kinases 1 and 2 by NGF that is necessary for neurite growth. Our results reveal a previously unknown signaling loop in developing sympathetic neurons that is crucial for NGF-dependent axon growth and target innervation.


Asunto(s)
Axones/efectos de los fármacos , Proteína Relacionada con TNFR Inducida por Glucocorticoide/fisiología , Factor de Crecimiento Nervioso/farmacología , Neuronas/citología , Transducción de Señal/fisiología , Factores de Necrosis Tumoral/metabolismo , Factores de Edad , Análisis de Varianza , Animales , Animales Recién Nacidos , Células Cultivadas , Embrión de Mamíferos , Fluoresceínas/metabolismo , Proteína Relacionada con TNFR Inducida por Glucocorticoide/deficiencia , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neuronas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Ganglio Cervical Superior/citología , Factores de Necrosis Tumoral/deficiencia
16.
J Neuroimmunol ; 194(1-2): 97-106, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18207576

RESUMEN

Inflammatory cytokines have been implicated in the pathology of multiple neurologic diseases, including multiple sclerosis. We examined the role of the TNF family member TWEAK in neuroinflammation. Cuprizone-fed mice undergo neuroinflammation and demyelination in the brain, but upon removal of cuprizone from the diet, inflammation is resolved and remyelination occurs. Using this model, we demonstrate that mice lacking TWEAK exhibit a significant delay in demyelination and microglial infiltration. During remyelination, mice lacking the TWEAK gene demonstrate only a marginal delay in remyelination. Thus, this study identifies a primary role of TWEAK in promoting neuroinflammation and exacerbating demyelination during cuprizone-induced damage.


Asunto(s)
Enfermedades Desmielinizantes/fisiopatología , Encefalitis/fisiopatología , Factores de Necrosis Tumoral/fisiología , Animales , Astrocitos/metabolismo , Astrocitos/patología , Quelantes/toxicidad , Cobre , Cuprizona/toxicidad , Citocina TWEAK , Enfermedades Desmielinizantes/inducido químicamente , Método Doble Ciego , Encefalitis/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Microglía/patología , Vaina de Mielina/fisiología , Oligodendroglía/metabolismo , Oligodendroglía/patología , ARN Mensajero/biosíntesis , Receptores del Factor de Necrosis Tumoral/fisiología , Receptor de TWEAK , Factores de Necrosis Tumoral/biosíntesis , Factores de Necrosis Tumoral/deficiencia , Factores de Necrosis Tumoral/genética
17.
J Immunol ; 179(11): 7702-8, 2007 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18025216

RESUMEN

In the absence of TNF, mice infected with Mycobacterium avium suffer a peculiar disintegration of the granulomas, with extensive apoptosis and necrosis of their cells, occurring during the course of the infection and leading to the death of the animals within a few days of its onset. The survival time depends on the virulence of the infecting strain as well as on the dose and route of infection. Intravenously infected mice developed the typical lesions in hepatic granulomas whereas aerosol-infected animals developed them in the lung granulomas. At the onset of the development of pulmonary granuloma disintegration, extensive expansion of T cells, with intense up-regulation of activation markers, massive exacerbation of their ability to secrete IFN-gamma, and increased cytotoxic activity of both CD4(+) and CD8(+) T cells were observed. Forced expression of Bcl2 did not prevent the early death of infected TNF-deficient mice leading merely to a modest increase in survival times. The expression of the FasL on T cells was not affected but there was an intense up-regulation of the TRAIL in T cells and, in particular, myeloid cells. We thus show that an exacerbated immune response occurs in TNF-deficient hosts during M. avium infections that leads to enhanced IFN-gamma production and late up-regulation of TRAIL which may contribute to granuloma disintegration.


Asunto(s)
Infecciones por Mycobacterium/inmunología , Factores de Necrosis Tumoral/deficiencia , Factores de Necrosis Tumoral/inmunología , Animales , Apoptosis/inmunología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Regulación hacia Abajo/inmunología , Granuloma/inmunología , Granuloma/microbiología , Granuloma/patología , Inyecciones Intravenosas , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Mycobacterium/microbiología , Infecciones por Mycobacterium/mortalidad , Mycobacterium avium/inmunología , Mycobacterium avium/aislamiento & purificación , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2 , Tasa de Supervivencia , Linfocitos T/inmunología , Ligando Inductor de Apoptosis Relacionado con TNF/inmunología
18.
J Immunol ; 179(11): 7949-58, 2007 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18025243

RESUMEN

TNF-like weak inducer of apoptosis (TWEAK), a member of the TNF superfamily, is a prominent inducer of proinflammatory cytokines in vitro and in vivo. We previously found that kidney cells display the TWEAK receptor Fn14, and that TWEAK stimulation of mesangial cells and podocytes induces a potent proinflammatory response. Several of the cytokines up-regulated in the kidney in response to TWEAK are instrumental in Lupus nephritis; we therefore hypothesized that TWEAK/Fn14 interactions may be important in the cascade(s) leading to renal damage in systemic Lupus erythematosus. In this study, we analyzed the effects of Fn14 deficiency in the chronic graft-vs-host model of SLE, and the benefits of treatment with an anti-TWEAK mAb in this mouse model. We found that anti-nuclear Ab titers were no different between C57BL/6 Fn14 wild-type and deficient mice injected with alloreactive bm12 splenocytes. However, kidney disease was significantly less severe in Fn14 knockout mice. Furthermore, kidney IgG deposition, IL-6, MCP-1, RANTES, and IP-10, as well as macrophage infiltration, were significantly decreased in Fn14-deficient mice with induced lupus. Similarly, mice with induced Lupus treated with an anti-TWEAK neutralizing mAb had significantly diminished kidney expression of IL-6, MCP-1, IL-10, as well as proteinuria, but similar autoantibody titers, as compared with control-treated mice. We conclude that TWEAK is an important mediator of kidney damage that acts by promoting local inflammatory events, but without impacting adaptive immunity in this experimental LN model. Thus, TWEAK blockade may be a novel therapeutic approach to reduce renal damage in SLE.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Lupus Eritematoso Sistémico/inmunología , Nefritis Lúpica/inmunología , Receptores del Factor de Necrosis Tumoral/metabolismo , Factores de Necrosis Tumoral/metabolismo , Animales , Anticuerpos/administración & dosificación , Anticuerpos/farmacología , Anticuerpos/uso terapéutico , Autoanticuerpos/inmunología , Quimiocinas/efectos de los fármacos , Enfermedad Crónica , Citocina TWEAK , Citocinas/efectos de los fármacos , Citocinas/genética , Modelos Animales de Enfermedad , Femenino , Enfermedad Injerto contra Huésped/patología , Inmunoglobulina G/inmunología , Lupus Eritematoso Sistémico/complicaciones , Lupus Eritematoso Sistémico/patología , Nefritis Lúpica/tratamiento farmacológico , Nefritis Lúpica/patología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteinuria/tratamiento farmacológico , Receptores del Factor de Necrosis Tumoral/deficiencia , Sensibilidad y Especificidad , Receptor de TWEAK , Factores de Necrosis Tumoral/deficiencia
19.
Eur J Immunol ; 37(12): 3597-604, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18022861

RESUMEN

Although corneal transplantation is one of the most common tissue transplantations and is known to have a high graft acceptance rate, occasional corneal graft rejection remains a cause of blindness. OX40, a member of the TNF receptor superfamily, is expressed on activated T cells, and transmits a costimulatory signal by binding to OX40 ligand (OX40L) expressed on several cells with antigen-presenting functions. Using a blocking monoclonal antibody (mAb) against murine OX40L, we investigated the role of OX40 in a murine model of corneal transplantation. C3H/He mouse corneas were transplanted to BALB/c mice orthotopically. Administration of anti-OX40L mAb significantly reduced allograft rejection, and increased graft survival rate to 40% at 8 weeks after transplantation, while all corneas were rejected within 5 weeks in control IgG-treated mice. Similar reduced rejection was observed when wild-type donor corneas were transplanted to OX40L-deficient recipients. In vitro study revealed that the anti-OX40L mAb treatment reduced proliferative response and IFN-gamma production of draining lymph node cells in response to stimulation with donor alloantigen. These results demonstrate that OX40L blockade is effective for prolongation of corneal allograft survival by inhibiting recipient T cell activation.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Trasplante de Córnea , Rechazo de Injerto/prevención & control , Supervivencia de Injerto/efectos de los fármacos , Glicoproteínas de Membrana/antagonistas & inhibidores , Inhibidores del Factor de Necrosis Tumoral , Animales , Anticuerpos Monoclonales/inmunología , Evaluación Preclínica de Medicamentos , Proteínas del Ojo/inmunología , Interferón gamma/biosíntesis , Interleucina-10/biosíntesis , Isoantígenos/inmunología , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Noqueados , Ligando OX40 , Especificidad del Receptor de Antígeno de Linfocitos T , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Donantes de Tejidos , Trasplante Homólogo , Factores de Necrosis Tumoral/deficiencia , Factores de Necrosis Tumoral/genética , Factores de Necrosis Tumoral/fisiología
20.
J Immunol ; 179(6): 3515-23, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17785785

RESUMEN

Costimulatory cross-talk can occur at multiple cellular levels to potentiate expansion and polarization of Th responses. Although OX40L ligand (OX40L) is thought to play a key role in Th2 development, the critical cellular source of this molecule has yet to be identified. In this study, we demonstrate that OX40L expression by the initiating dendritic cell (DC) is a fundamental requirement for optimal induction of primary and memory Th2 responses in vivo. Analysis of the kinetics of the residual Th2 response primed by OX40L-deficient DC suggested a failure to stimulate appropriate expansion and/or survival of T cells, rather than an inability to polarize per se. The dependence upon OX40L was predominantly due to the provision of signaling through OX40 rather than retrograde signaling to the DC. Mechanistically, impaired Th2 priming in the absence of OX40L was not due to exaggerated regulation because there was no evidence of increased expansion or function of regulatory cell populations, suppression through IL-10 production, or hyporesponsiveness to secondary challenge. These data define a critical role for DC-derived OX40L in the induction and development of Th2 responses in vivo.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Memoria Inmunológica , Activación de Linfocitos/inmunología , Glicoproteínas de Membrana/biosíntesis , Transducción de Señal/inmunología , Células Th2/inmunología , Células Th2/metabolismo , Factores de Necrosis Tumoral/biosíntesis , Traslado Adoptivo , Animales , Polaridad Celular/inmunología , Células Cultivadas , Células Dendríticas/trasplante , Regulación de la Expresión Génica/inmunología , Inyecciones Intraperitoneales , Ligandos , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Ratones , Ligando OX40 , Receptores OX40/fisiología , Factores de Necrosis Tumoral/deficiencia , Factores de Necrosis Tumoral/genética , Factores de Necrosis Tumoral/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...