Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 188
Filtrar
1.
J Cell Biol ; 223(3)2024 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-38329462

RESUMEN

TNF signaling does not result in cell death unless multiple inhibitory signals are overcome, which can be accomplished by simultaneous signaling through IFNγ. In this issue, Deng and colleagues (http://doi.org/10.1083/jcb.202305026) dissect the mechanisms by which IFNγ signaling combines with TNF to mediate cell death through caspase-8, discussed by James E. Vince.


Asunto(s)
Muerte Celular , Interferón gamma , Transducción de Señal , Interferón gamma/fisiología , Caspasa 8/fisiología , Factores de Necrosis Tumoral/fisiología
2.
Sheng Li Xue Bao ; 69(5): 666-676, 2017 Oct 25.
Artículo en Chino | MEDLINE | ID: mdl-29063114

RESUMEN

Complement-C1q/tumor necrosis factor-related protein 3 (CTRP3) is an adipokine that primarily identified in 2003 and is an important member of CTRP family. CTRP3 is expressed in various tissues and cell types, and is highly conserved among different species. Multiple novel functions of CTRP3 have been reported recently as the further research on this protein develops. CTRP3 not only affects the proliferation of chondrocytes and the pathogenesis of osteoarthritis, but also regulates multiple physiological and pathological processes including the secretion of testosterone and adipokines, glucose and lipid metabolism, mitochondrial biogenesis, inflammatory response, cell apoptosis, angiogenesis, vascular calcification and ventricular remodeling. The present review mainly focuses on the research progresses on CTRP3, including its discovery, gene and protein structure, expression regulation, and biological functions. The progresses summarized may provide new clues for the further investigation of CTRP3.


Asunto(s)
Factores de Necrosis Tumoral/fisiología , Animales , Enfermedades Autoinmunes/fisiopatología , Desarrollo Óseo , Metabolismo de los Hidratos de Carbono , Enfermedades Cardiovasculares/fisiopatología , Humanos , Metabolismo de los Lípidos , Factores de Necrosis Tumoral/química , Factores de Necrosis Tumoral/genética
3.
Medicine (Baltimore) ; 95(26): e3937, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27367991

RESUMEN

Our aim in this study was to determine soluble tumor necrosis factor (TNF)-like weak inducer of apoptosis (sTWEAK) and interleukin-17A (IL-17A) levels in celiac disease, and their association with the gluten diet and autoantibodies. Eighty patients with celiac diagnosis and 80 healthy control individuals with similar age, gender and body mass index to the patient group were included in the study. Serum sTWEAK and IL-17A levels were measured by the serum enzyme-linked immunosorbent assay kit. The median IL-17A (117.5 pg/mL vs. 56.7 pg/mL; P = 0.001) level in celiac patients was higher than in the control group, while the median sTWEAK (543 pg/mL vs. 643 pg/mL; P = 0.016) level in patients was determined to be lower. In the patient group, patients who complied with the gluten diet had a lower level of median IL-17A (98.1 pg/mL vs. 197.5 pg/mL; P = 0.034) and a higher level of sTWEAK (606 pg/mL vs. 522.8 pg/mL; P = 0.031) than those who did not adhere. Furthermore, the IL-17A level was higher and the sTWEAK level was lower in celiac patients with positive antibody than those with negative antibody. A positive correlation was determined among anti-gliadin antibody IgA, anti-gliadin antibody IgG, anti-tissue transglutaminase IgG levels and the IL-17A level, and a negative correlation was determined with the sTWEAK level. In celiac disease, the sTWEAK and IL-17A levels differ between patients who cannot adapt to the gluten diet and who are autoantibody positive, and patients who adapt to the diet and are autoantibody negative. We believe that sTWEAK and IL-17A are associated with the inflammation in celiac pathogenesis.


Asunto(s)
Enfermedad Celíaca/etiología , Interleucina-17/fisiología , Factores de Necrosis Tumoral/fisiología , Adulto , Autoanticuerpos/sangre , Enfermedad Celíaca/sangre , Estudios Transversales , Citocina TWEAK , Dieta , Femenino , Glútenes , Humanos , Interleucina-17/sangre , Masculino , Factores de Necrosis Tumoral/sangre
4.
Rheumatology (Oxford) ; 55(10): 1880-8, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27354685

RESUMEN

OBJECTIVES: Oestrogens have been shown to play key roles in the pathogenesis of SLE. The aim of this study was to investigate the roles and mechanisms of 17ß-estradiol (E2) in TNF-like weak inducer of apoptosis (TWEAK) expression in LN. METHODS: Peripheral blood mononuclear cells (PBMCs) obtained from LN patients were used for in vitro experiments, while female MRL/lpr and MRL/MpJ mice were used for in vivo studies. E2, ICI 182 780 [estrogen receptor (ER)-selective antagonist], methyl-piperidino-pyrazole (MPP, ERα-selective modulator), lentivirus (LV)-TWEAK-short hairpin RNA (shRNA) and LV-control-shRNA treatments were used in this study. RESULTS: TWEAK mRNA expression in PBMCs was significantly increased following E2 treatment and downregulated after incubation with ICI 182 780 or MPP. Compared with sham-operated MRL/lpr mice, ovariectomized mice, treated with dimethyl sulphoxide vehicle alone, showed lower expression levels of renal TWEAK mRNA and protein. The expression of both mRNA and protein in ovariectomized mice was upregulated after E2 treatment and downregulated after ICI 182 780 or MPP co-treatment. Severe renal damage was observed in E2-treated ovariectomized mice, as were higher serum levels of IL-6, compared with dimethyl sulphoxide vehicle-treated ovariectomized mice. Co-treatment with LV-TWEAK-shRNA reversed these changes, and LV-control-shRNA treatment had no effect on them. CONCLUSION: Our results demonstrated that E2 plays an important role in the upregulation of TWEAK expression in LN, most likely through an ERα-dependent pathway, causing kidney damage. This provides a novel insight into the mechanisms of the E2-TWEAK signalling pathway in LN.


Asunto(s)
Estradiol/farmacología , Estrógenos/farmacología , Nefritis Lúpica/etiología , Factores de Necrosis Tumoral/fisiología , Lesión Renal Aguda/etiología , Adolescente , Adulto , Animales , Células Cultivadas , Citocina TWEAK , Regulación hacia Abajo , Receptor alfa de Estrógeno/fisiología , Femenino , Humanos , Leucocitos Mononucleares/metabolismo , Ratones , Persona de Mediana Edad , Ovario/cirugía , ARN Mensajero/metabolismo , Transducción de Señal , Factores de Necrosis Tumoral/farmacología , Regulación hacia Arriba , Adulto Joven
5.
Immunology ; 148(1): 70-82, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27040357

RESUMEN

Uterine natural killer (uNK) cells are the most abundant lymphocyte population in the feto-maternal interface during early gestation, and uNK cells play a significant role in the establishment and maintenance of pregnancy-related vascularization, as well as in tolerance to the fetus. Tumour necrosis factor-like weak inducer of apoptosis (TWEAK) and its receptor, fibroblast growth factor-inducible molecule (Fn14), are involved in preventing local cytotoxicity and counterbalancing the cytotoxic function of uNK cells. Here, we studied the regulation of TWEAK/Fn14-mediated innate immunity in the uterus using a lipopolysaccharide (LPS)-induced model of abortion in pregnant mice. Specifically, we detected the expression of TWEAK and Fn14 in the uterus and in uNK cells following LPS treatment. Our results revealed that TWEAK and Fn14 are expressed by uNK cells in pregnant mice; in particular, it appears that the cytokine TWEAK is primarily derived from uNK cells. Interestingly, the down-regulation of TWEAK in uNK cells and the up-regulation of the Fn14 receptor in the uterus in LPS-treated mice may contribute to the disruption of decidual homeostasis by altering uNK cell cytotoxicity - ultimately leading to fetal rejection. In conclusion, the present study strongly suggests that the TWEAK-Fn14 axis in uNK cells is involved in maintaining the tolerance necessary for successful pregnancy.


Asunto(s)
Aborto Espontáneo/etiología , Células Asesinas Naturales/inmunología , Factores de Necrosis Tumoral/fisiología , Útero/inmunología , Aborto Espontáneo/inmunología , Animales , Citocina TWEAK , Modelos Animales de Enfermedad , Femenino , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Subfamilia K de Receptores Similares a Lectina de Células NK/análisis , Embarazo , Receptores del Factor de Necrosis Tumoral/fisiología , Receptor de TWEAK , Factor de Necrosis Tumoral alfa/análisis
6.
J Am Soc Nephrol ; 27(12): 3639-3652, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27026366

RESUMEN

The identification of the cellular origins of myofibroblasts has led to the discovery of novel pathways that potentially drive myofibroblast perpetuation in disease. Here, we further investigated the role of innate immune signaling pathways in this process. In mice, renal injury-induced activation of pericytes, which are myofibroblast precursors attached to endothelial cells, led to upregulated expression of TNF receptor superfamily member 12a, also known as fibroblast growth factor-inducible 14 (Fn14), by these cells. In live rat kidney slices, administration of the Fn14 ligand, TNF-related weak inducer of apoptosis (TWEAK), promoted pericyte-dependent vasoconstriction followed by pericyte detachment from capillaries. In vitro, administration of TWEAK activated and differentiated pericytes into cytokine-producing myofibroblasts, and further activated established myofibroblasts in a manner requiring canonical and noncanonical NF-κB signaling pathways. Deficiency of Fn14 protected mouse kidneys from fibrogenesis, inflammation, and associated vascular instability after in vivo injury, and was associated with loss of NF-κB signaling. In a genetic model of spontaneous CKD, therapeutic delivery of anti-TWEAK blocking antibodies attenuated disease progression, preserved organ function, and increased survival. These results identify the TWEAK-Fn14 signaling pathway as an important factor in myofibroblast perpetuation, fibrogenesis, and chronic disease progression.


Asunto(s)
Enfermedades Renales/etiología , Riñón/patología , Miofibroblastos/fisiología , Receptores del Factor de Necrosis Tumoral/fisiología , Transducción de Señal , Factores de Necrosis Tumoral/fisiología , Animales , Citocina TWEAK , Progresión de la Enfermedad , Fibrosis/etiología , Ratones , Receptor de TWEAK
7.
Mol Immunol ; 72: 92-102, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27011037

RESUMEN

The differentiation and proper function of thymic epithelial cells (TECs) depend on various tumor necrosis factor superfamily (TNFSF) signals that are needed to maintain the thymic stromal microenvironment. Nevertheless, the direct transcriptional effects of these signals on TECs remain unclear. To address this issue, we stimulated murine embryonic thymus tissue with selected TNFSF ligands and performed a gene expression profiling study. We show that Aire expression is a direct and specific effect of RANKL stimulation, whereas LTß and TNFα are major inducers of chemokines in the thymic stroma and we propose differential NF-κB binding as one possible cause of these gene expression patterns. Our work provides further insight into the complex molecular pathways that shape the thymic microenvironment and maintain central tolerance.


Asunto(s)
Microambiente Celular , Células del Estroma/citología , Timo/citología , Factores de Necrosis Tumoral/fisiología , Animales , Perfilación de la Expresión Génica , Receptor beta de Linfotoxina/biosíntesis , Receptor beta de Linfotoxina/genética , Ratones , Ratones Endogámicos C57BL , Subunidad p50 de NF-kappa B/biosíntesis , Subunidad p50 de NF-kappa B/genética , Técnicas de Cultivo de Órganos , Ligando RANK/genética , Transducción de Señal , Timo/embriología , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética , Factores de Necrosis Tumoral/genética
8.
Cardiovasc Res ; 108(1): 139-47, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26224570

RESUMEN

AIM: The interaction between TNF-like weak inducer of apoptosis (TWEAK, Tnfsf12) and the receptor, fibroblast growth factor-inducible 14 (Fn14), regulates vascular damage through different mechanisms, including inflammation. Oxidative stress plays a major role in inflammation and the development of atherosclerosis, but the relationship between TWEAK and oxidative stress is, however, poorly understood. METHODS AND RESULTS: In this study, we found that TWEAK and Fn14 are co-localized with the NADPH subunits, p22phox and Nox2, in human advanced atherosclerotic plaques. Using primary human macrophages and a murine macrophage cell line, we demonstrate that TWEAK promotes ROS production and enhances NADPH oxidase activity. Hence, we show a direct involvement of the TWEAK-Fn14 axis in oxidative stress, as genetic silencing of Fn14 or Nox2 abrogates the TWEAK-induced ROS production. Furthermore, our results point at Rac1 as an upstream mediator of TWEAK during oxidative stress. Finally, using an in vivo murine model we confirmed the major role of TWEAK in oxidative stress, as genetic silencing of Tnfsf12 in an ApoE(-/-) background reduces the number of DHE and 8-hydroxydeoxyguanosine-positive macrophages by 50%. CONCLUSIONS: Our results suggest that TWEAK regulates vascular damage by stimulating ROS production in an Nox2-dependent manner. These new insights into the TWEAK/Fn14 axis underline their potential use as therapeutic targets in atherosclerosis.


Asunto(s)
Macrófagos/metabolismo , NADPH Oxidasas/fisiología , Estrés Oxidativo , Receptores del Factor de Necrosis Tumoral/fisiología , Factores de Necrosis Tumoral/fisiología , Animales , Enfermedades de las Arterias Carótidas/metabolismo , Células Cultivadas , Citocina TWEAK , Activación Enzimática , Glutatión/análisis , Humanos , Ratones , Receptor de TWEAK , Proteína de Unión al GTP rac1/fisiología
9.
J Invest Dermatol ; 135(8): 1986-1995, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25826425

RESUMEN

Tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK, TNFSF12) and its sole receptor Fn14, belonging to the TNF ligand and receptor superfamilies respectively, are involved in cell survival and cytokine production. The role of TWEAK/Fn14 interactions in the pathogenesis of cutaneous lupus has not been explored. TWEAK treatment of murine PAM212 keratinocytes stimulated the secretion of RANTES via Fn14 and promoted apoptosis. Parthenolide, but not wortmanin or the MAPK inhibitor PD98059, significantly decreased production of RANTES, indicating that this effect of TWEAK is mediated via NF-κB signaling. UVB irradiation significantly upregulated the expression of Fn14 on keratinocytes in vitro and in vivo and increased RANTES production. MRL/lpr Fn14 knockout (KO) lupus mice were compared with MRL/lpr Fn14 wild-type (WT) mice to evaluate for any possible differences in the severity of cutaneous lesions and the presence of infiltrating immune cells. MRL/lpr Fn14 KO mice had markedly attenuated cutaneous disease as compared with their Fn14 WT littermates, as evidenced by the well-maintained architecture of the skin and significantly decreased skin infiltration of T cells and macrophages. Our data strongly implicate TWEAK/Fn14 signaling in the pathogenesis of the cutaneous manifestations in the MRL/lpr model of spontaneous lupus and suggest a possible target for therapeutic intervention.


Asunto(s)
Lupus Eritematoso Cutáneo/etiología , Lupus Eritematoso Cutáneo/fisiopatología , Receptores del Factor de Necrosis Tumoral/fisiología , Transducción de Señal/fisiología , Factores de Necrosis Tumoral/fisiología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Línea Celular , Quimiocina CCL5/metabolismo , Citocina TWEAK , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Técnicas In Vitro , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Queratinocitos/efectos de la radiación , Lupus Eritematoso Cutáneo/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos MRL lpr , Ratones Noqueados , Receptores del Factor de Necrosis Tumoral/deficiencia , Receptores del Factor de Necrosis Tumoral/efectos de la radiación , Receptor de TWEAK , Factores de Necrosis Tumoral/farmacología , Rayos Ultravioleta , Regulación hacia Arriba/efectos de la radiación
10.
FASEB J ; 29(3): 988-1002, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25466899

RESUMEN

Skeletal muscle is responsible for the majority of glucose disposal in body. Impairment in skeletal muscle glucose handling capacity leads to the state of insulin resistance. The TNF-like weak inducer of apoptosis (TWEAK) cytokine has now emerged as a major regulator of skeletal muscle mass and function. However, the role of TWEAK in skeletal muscle metabolic function remains less understood. Here, we demonstrate that with progressive age, skeletal muscle-specific TWEAK-transgenic (TWEAK-Tg) mice gain increased body weight (∼16%) and fat mass (∼64%) and show glucose intolerance and insulin insensitivity. TWEAK-Tg mice also exhibit adipocyte hypertrophy in the epididymal fat. Oxygen uptake, voluntary physical activity, and exercise capacity were significantly reduced in TWEAK-Tg mice compared with controls. Overexpression of TWEAK inhibited (∼31%) 5' AMP-activated protein kinase (AMPK) and reduced (∼31%) the levels of glucose transporter type 4 (GLUT4) without affecting the Akt pathway. TWEAK also inhibited insulin-stimulated glucose uptake (∼32%) and repressed the levels of GLUT4 (∼50%) in cultured myotubes from C57BL6 mice. TWEAK represses the levels of Krüppel-like factor 15; myocyte enhancer factor 2, and peroxisome proliferator-activated receptor-γ coactivator-1α, which are required for the activation of the GLUT4 locus. Collectively our study demonstrates that elevated levels of TWEAK in skeletal muscle cause metabolic abnormalities. Inhibition of TWEAK could be a potential approach to prevent weight gain and type 2 diabetes.


Asunto(s)
Intolerancia a la Glucosa/etiología , Resistencia a la Insulina , Músculo Esquelético/metabolismo , Obesidad Abdominal/etiología , Factores de Necrosis Tumoral/fisiología , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Western Blotting , Células Cultivadas , Citocina TWEAK , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Glucosa/metabolismo , Intolerancia a la Glucosa/metabolismo , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Ratones Transgénicos , Músculo Esquelético/patología , Obesidad Abdominal/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
11.
Nephron Clin Pract ; 127(1-4): 65-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25343824

RESUMEN

Fibrosis is a major hallmark of progressive kidney disease. The cellular mechanisms that lead to kidney tissue fibrosis are complex and include, for example, increased inflammation, increased oxidative stress, and proximal tubule cell death in the form of apoptosis or senescence. Recent studies have identified TWEAK, a tumor necrosis factor-like weak inducer of apoptosis, as a novel cytokine that mediates kidney inflammation in models of renal fibrosis. Inhibition of apoptosis via TWEAK inhibition has been shown to reduce kidney fibrosis. Recent studies using lineage tracing suggest that interstitial pericytes/perivascular fibroblasts differentiate into myofibroblasts and undergo proliferative expansion during fibrosis. Furthermore, increased expression of nuclear peroxisome proliferator-activated receptor-α in proximal tubules can directly reduce increased expression of transforming growth factor-ß1 and interstitial inflammation in models of renal fibrosis, which suggests preservation of proximal tubule cell metabolism and integrity represents an important new therapeutic target. In this review, the current evidence and potential molecular mechanisms involved in the development of kidney fibrosis are discussed.


Asunto(s)
Lesión Renal Aguda/patología , Riñón/patología , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/terapia , Adenosina Trifosfato/metabolismo , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Proteínas Reguladoras de la Apoptosis/fisiología , Senescencia Celular , Cisplatino/toxicidad , Citocina TWEAK , Progresión de la Enfermedad , Células Epiteliales/patología , Ácidos Grasos/metabolismo , Fibroblastos/patología , Fibrosis , Perfilación de la Expresión Génica , Humanos , Inflamación , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Ratones , Ratones Transgénicos , Terapia Molecular Dirigida , Miofibroblastos/patología , PPAR alfa/genética , PPAR alfa/metabolismo , PPAR alfa/fisiología , Pericitos/patología , Receptores del Factor de Necrosis Tumoral/fisiología , Terapia de Reemplazo Renal , Receptor de TWEAK , Factores de Necrosis Tumoral/fisiología
12.
Annu Rev Cell Dev Biol ; 30: 337-56, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25150011

RESUMEN

Cell turnover is a fundamental feature in metazoans. Cells can die passively, as a consequence of severe damage to their structural integrity, or actively, owing to a more confined biological disruption such as DNA damage. Passive cell death is uncontrolled and often harmful to the organism. In contrast, active cell death is tightly regulated and serves to support the organism's life. Apoptosis-the primary form of regulated cell death-is relatively well defined. Necroptosis-an alternative, distinct kind of regulated cell death discovered more recently-is less well understood. Apoptosis and necroptosis can be triggered either from within the cell or by extracellular stimuli. Certain signaling components, including several death ligands and receptors, can regulate both processes. Whereas apoptosis is triggered and executed via intracellular proteases called caspases, necroptosis is suppressed by caspase activity. Here we highlight current understanding of the key signaling mechanisms that control regulated cell death.


Asunto(s)
Muerte Celular/fisiología , Animales , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis/fisiología , Caspasas/fisiología , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/fisiología , Activación Enzimática , Humanos , Modelos Biológicos , Proteína Serina-Treonina Quinasas de Interacción con Receptores/fisiología , Receptores de Muerte Celular/fisiología , Receptores del Factor de Necrosis Tumoral/fisiología , Transducción de Señal/fisiología , Factores de Necrosis Tumoral/fisiología
13.
Ann Neurol ; 76(1): 43-53, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24798682

RESUMEN

OBJECTIVE: Brain damage and ischemia often trigger cortical spreading depression (CSD), which aggravates brain damage. The proinflammatory cytokine tumor necrosis factor (TNF) is significantly upregulated during brain damage, but it is unknown whether TNF influences spreading depression in cerebral cortex in vivo. This question is important because TNF not only furthers inflammatory reactions but might also be neuroprotective. Here we tested the hypothesis that TNF affects CSD, and we explored the direction in which CSD is modified by TNF. METHODS: CSD, elicited by pressure microinjection of KCl, was recorded in anesthetized rats and mice. TNF was administered locally into a trough, providing local TNF treatment of a cortical area. For further analysis, antibodies to TNF receptor (TNFR) 1 or 2 were applied, or CSD was monitored in TNFR1 and TNFR2 knockout mice. γ-Aminobutyric acid (GABA)A receptors were blocked by bicuculline. Immunohistochemistry localized the cortical expression of TNFR1 and TNFR2. RESULTS: Local application of TNF to the cortex reduced dose-dependently the amplitude of CSD. This effect was prevented by blockade or knockout of TNFR2 but not by blockade or knockout of TNFR1. TNFR2 was localized at cortical neurons including parvalbumin-positive inhibitory interneurons, and blockade of GABAA receptors by bicuculline prevented the reduction of CSD amplitudes by TNF. INTERPRETATION: We identified a functional link between TNF and CSD. TNF activates TNFR2 in cortical inhibitory interneurons. The resulting release of GABA reduces CSD amplitudes. In this manner, TNF might be neuroprotective in pathological conditions.


Asunto(s)
Depresión de Propagación Cortical/fisiología , Inhibición Neural/fisiología , Factores de Necrosis Tumoral/fisiología , Animales , Masculino , Ratones , Ratones Noqueados , Ratas , Ratas Wistar , Receptores Tipo I de Factores de Necrosis Tumoral/fisiología , Receptores Tipo II del Factor de Necrosis Tumoral/fisiología , Factores de Necrosis Tumoral/administración & dosificación , Ácido gamma-Aminobutírico/metabolismo
14.
PLoS One ; 9(3): e90399, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24599047

RESUMEN

Peritoneal dialysis (PD) is complicated by peritonitis episodes that cause loss of mesothelium and eventually sclerosing peritonitis. An improved understanding of the molecular contributors to peritoneal injury and defense may increase the therapeutic armamentarium to optimize peritoneal defenses while minimizing peritoneal injury. There is no information on the expression and function of the cytokine TWEAK and its receptor Fn14 during peritoneal injury. Fn14 expression and soluble TWEAK levels were measured in human PD peritoneal effluent cells or fluids with or without peritonitis. Fn14 expression was also analyzed in peritoneal biopsies from PD patients. Actions of intraperitoneal TWEAK were studied in mice in vivo. sTWEAK levels were increased in peritoneal effluent in PD peritonitis. Effluent sTWEAK levels correlated with the number of peritoneal macrophages (r=0.491, p=0.002). Potential TWEAK targets that express the receptor Fn14 include mesothelial cells and macrophages, as demonstrated by flow cytometry of peritoneal effluents and by analysis of peritoneal biopsies. Peritoneal biopsy Fn14 correlated with mesothelial injury, fibrosis and inflammation, suggesting a potential deleterious effect of TWEAK/Fn14. In this regard, intraperitoneal TWEAK administration to mice promoted peritoneal inflammation characterized by increased peritoneal effluent MCP-1, Fn14 and Gr1+ macrophages, increased mesothelial Fn14, MCP-1 and CCL21 expression and submesothelial tissue macrophage recruitment. Taken together these data suggest that the TWEAK/Fn14 system may promote inflammation and tissue injury during peritonitis and PD.


Asunto(s)
Peritonitis/metabolismo , Factores de Necrosis Tumoral/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Estudios de Casos y Controles , Células Cultivadas , Quimiocinas/genética , Quimiocinas/metabolismo , Citocina TWEAK , Femenino , Expresión Génica , Infecciones por Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/metabolismo , Infecciones por Bacterias Grampositivas/inmunología , Infecciones por Bacterias Grampositivas/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Fallo Renal Crónico/terapia , Macrófagos Peritoneales/metabolismo , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Diálisis Peritoneal , Peritonitis/inmunología , Peritonitis/microbiología , Receptores del Factor de Necrosis Tumoral/metabolismo , Receptor de TWEAK
15.
Methods Mol Biol ; 1135: 35-52, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24510853

RESUMEN

The TNF superfamily member TWEAK has emerged as a pleiotropic cytokine that regulates many cellular functions that include immune/inflammatory activity, angiogenesis, cell proliferation, and fate. TWEAK through its inducible receptor, FGF-inducible molecule 14 (Fn14), can induce both beneficial and deleterious activity that has a profound effect on cell survival. Thus it is highly likely that TWEAK and Fn14 expressed by cells of the neurovascular unit help regulate and maintain vascular and tissue homeostasis. In this chapter we discuss the expression of TWEAK and Fn14 signaling in the cerebral microvascular pericyte. Pericytes are a highly enigmatic population of microvascular cells that are important in regulatory pathways that modulate physiological angiogenesis in response to chronic mild hypoxic stress. A brief introduction will identify the microvascular pericyte. A more detailed discussion of pericyte TWEAK signaling during adaptive angioplasticity will follow.


Asunto(s)
Neovascularización Fisiológica , Pericitos/fisiología , Factores de Necrosis Tumoral/fisiología , Animales , Arterias Cerebrales/citología , Arterias Cerebrales/fisiología , Cerebro/irrigación sanguínea , Citocina TWEAK , Humanos , Transducción de Señal
16.
Sci Rep ; 4: 3625, 2014 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-24406502

RESUMEN

Skeletal muscle (SM) regeneration after injury is impaired by excessive inflammation. Particularly, the inflammatory cytokine tumour necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) is a potent inducer of skeletal muscle wasting and fibrosis. In this study we investigated the role of Nrf2, a major regulator of oxidative stress defence, in SM ischemia/reperfusion (I/R) injury and TWEAK induced atrophy. We explored the time-dependent expression of TWEAK after I/R in SM of Nrf2-wildtype (WT) and knockout (KO) mice. Nrf2-KO mice expressed significant higher levels of TWEAK as compared to WT mice. Consequently, Nrf2-KO mice present an insufficient regeneration as compared to Nrf2-WT mice. Moreover, TWEAK stimulation activates Nrf2 in the mouse myoblast cell line C2C12. This Nrf2 activation inhibits TWEAK induced atrophy in C2C12 differentiated myotubes. In summary, we show that Nrf2 protects SM from TWEAK-induced cell death in vitro and that Nrf2-deficient mice therefore have poorer muscle regeneration.


Asunto(s)
Músculo Esquelético/patología , Atrofia Muscular/prevención & control , Factor 2 Relacionado con NF-E2/fisiología , Factores de Necrosis Tumoral/fisiología , Animales , Apoptosis , Secuencia de Bases , Línea Celular , Citocina TWEAK , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa
17.
FASEB J ; 28(3): 1398-411, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24327607

RESUMEN

Skeletal muscle wasting attributed to inactivity has significant adverse functional consequences. Accumulating evidence suggests that peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) and TNF-like weak inducer of apoptosis (TWEAK)-Fn14 system are key regulators of skeletal muscle mass in various catabolic states. While the activation of TWEAK-Fn14 signaling causes muscle wasting, PGC-1α preserves muscle mass in several conditions, including functional denervation and aging. However, it remains unknown whether there is any regulatory interaction between PGC-1α and TWEAK-Fn14 system during muscle atrophy. Here we demonstrate that TWEAK significantly reduces the levels of PGC-1α and mitochondrial content (∼50%) in skeletal muscle. Levels of PGC-1α are significantly increased in skeletal muscle of TWEAK-knockout (KO) and Fn14-KO mice compared to wild-type mice on denervation. Transgenic (Tg) overexpression of PGC-1α inhibited progressive muscle wasting in TWEAK-Tg mice. PGC-1α inhibited the TWEAK-induced activation of NF-κB (∼50%) and dramatically reduced (∼90%) the expression of atrogenes such as MAFbx and MuRF1. Intriguingly, muscle-specific overexpression of PGC-1α also prevented the inducible expression of Fn14 in denervated skeletal muscle. Collectively, our study demonstrates that TWEAK induces muscle atrophy through repressing the levels of PGC-1α. Overexpression of PGC-1α not only blocks the TWEAK-induced atrophy program but also diminishes the expression of Fn14 in denervated skeletal muscle.


Asunto(s)
Músculo Esquelético/patología , Atrofia Muscular/patología , Receptores del Factor de Necrosis Tumoral/fisiología , Factores de Transcripción/fisiología , Factores de Necrosis Tumoral/fisiología , Animales , Citocina TWEAK , Ensayo de Cambio de Movilidad Electroforética , Ratones , Ratones Transgénicos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Receptor de TWEAK
18.
Mol Neurobiol ; 49(1): 78-87, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23873135

RESUMEN

Tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) is a member of the TNF super family that controls many cellular activities including proliferation, migration, differentiation, apoptosis, and inflammation by binding to fibroblast growth factor-inducible 14 (Fn14), a highly inducible cell surface receptor. Recent studies have indicated that TWEAK-Fn14 axis signaling may contribute to chronic autoimmune diseases. TWEAK expression via microglia in cortical lesions, presence of TWEAK(+) macrophages in inflamed leptomeninges, and absence of TWEAK/Fn14 expression in healthy brain implicates importance of this pathway in pathogenesis of multiple sclerosis lesions. TWEAK-Fn14 axis blockade has also shown promise in various multiple sclerosis animal models. Stimulation of the TWEAK/Fn14 pathway can result in activation of both canonical and noncanonical NF-κB signaling and could also stimulate mitogen-activated protein kinase (MAPK) signaling pathways. Here, we have reviewed evidence of the possible role of TWEAK-Fn14 axis in pathophysiology of multiple sclerosis and experimental autoimmune encephalomyelitis (EAE) via neuroinflammation, tissue remodeling, blood-brain barrier (BBB) disruption, neurodegeneration, and astrogliosis.


Asunto(s)
Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/fisiopatología , Transducción de Señal/fisiología , Factores de Necrosis Tumoral/fisiología , Animales , Movimiento Celular , Citocina TWEAK , Modelos Animales de Enfermedad , Humanos , Inflamación/metabolismo , Inflamación/patología , Inflamación/fisiopatología , Esclerosis Múltiple/patología , Ratas , Receptores del Factor de Necrosis Tumoral/fisiología , Receptor de TWEAK , Factores de Necrosis Tumoral/metabolismo
19.
Biochim Biophys Acta ; 1832(10): 1744-55, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23748045

RESUMEN

Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) regulates apoptosis, proliferation and inflammation in renal epithelial cells and plays a role in acute kidney injury. However, there is little information on the chronic effects of TWEAK. We hypothesized that TWEAK may influence renal fibrosis and regulate kidney fibroblast biology, in part, through Ras pathway. We studied a chronic model of experimental unilateral ureteral obstruction in wild type and TWEAK deficient mice, and a murine model of systemic TWEAK overexpression. TWEAK actions were also explored in cultured renal and embryonic fibroblasts. TWEAK and TWEAK receptor expression was increased in the obstructed kidneys. The absence of TWEAK decreased early kidney tubular damage, inflammatory infiltrates and myofibroblast number. TWEAK deficient mice had decreased renal fibrosis 21days after obstruction, as assessed by extracellular matrix staining. In mice without prior underlying kidney disease, systemic overexpression of TWEAK induced kidney inflammation and fibrosis. In cultured fibroblasts, TWEAK induced proliferation through activation of the Ras/ERK pathway. TWEAK also activated nuclear factor κB (NFκB)-dependent inflammatory chemokine production in murine renal fibroblasts. In conclusion, lack of TWEAK reduces renal fibrosis in a model of persistent kidney insult and overexpression of TWEAK led to renal fibrosis. TWEAK actions on renal fibroblasts may contribute to the in vivo observations, as TWEAK promotes inflammatory activity and proliferation in fibroblast cultures.


Asunto(s)
Proliferación Celular , Fibrosis/fisiopatología , Enfermedades Renales/fisiopatología , Riñón/patología , Factores de Necrosis Tumoral/fisiología , Proteínas ras/fisiología , Animales , Línea Celular , Citocina TWEAK , Fibroblastos/patología , Ratones
20.
Br Dent J ; 214(9): 439-51, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23660900

RESUMEN

This paper will explore the pathological process involved in dental resorption as well as its classifications and aetiology. The second subsequent paper will look at its diagnosis and management.


Asunto(s)
Resorción Radicular/patología , Cemento Dental/patología , Enfermedades de la Pulpa Dental/complicaciones , Humanos , Osteoclastos/fisiología , Enfermedades Periapicales/complicaciones , Resorción Radicular/clasificación , Resorción Radicular/etiología , Traumatismos de los Dientes/complicaciones , Factores de Necrosis Tumoral/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...