Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Cells ; 10(9)2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34572081

RESUMEN

Tumor necrosis factor-alpha (TNF-α) signaling regulates phosphorylation of L-plastin, which is involved in forming the nascent sealing zone, a precursor zone for the matured sealing ring. This study aimed to illustrate the molecular mechanisms of L-plastin phosphorylation and the subsequent formation of the nascent sealing zone in osteoclasts treated with TNF-α. Here, we report that anti-TNF-receptor 1, inhibitors of signaling proteins (Src, PI3-K, Rho, and Rho-kinase), and siRNA of TRAF-6 attenuated the phosphorylation of LPL and filamentous actin content significantly in the presence of TNF-α. An inhibitor of integrin αvß3, PKC, or PKA did not inhibit TNF-α-induced L-plastin phosphorylation. Inhibitors of Src and PI3-K and not Rho or Rho-kinase reduced tyrosine phosphorylation of TRAF-6, suggesting that Src and PI3-K regulate TRAF-6 phosphorylation, and Rho and Rho-kinase are downstream of TRAF-6 regulation. Osteoclasts expressing constitutively active or kinase-defective Src proteins were used to determine the role of Src on L-plastin phosphorylation; similarly, the effect of Rho was confirmed by transducing TAT-fused constitutively active (V14) or dominant-negative (N19) Rho proteins into osteoclasts. Pull-down analysis with glutathione S-transferase-fused SH2 and SH3 domains of Src and PI3-K demonstrated coprecipitation of L-plastin and TRAF-6 with the SH3 and SH2 domains of the PI3-K and Src proteins. However, the actual order of the interaction of proteins requires further elucidation; a comprehensive screening should corroborate the initial findings of protein interactions via the SH2/SH3 domains. Ultimately, inhibition of the interaction of proteins with SH2/SH3 could reduce L-plastin phosphorylation and affect NSZ formation and bone resorption in conditions that display osteoclast activation and bone loss.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Osteoclastos/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Actinas/metabolismo , Animales , Resorción Ósea/metabolismo , Ratones , Ratones Endogámicos C57BL , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Dominios y Motivos de Interacción de Proteínas/efectos de los fármacos , Receptores Tipo I de Factores de Necrosis Tumoral/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor 6 Asociado a Receptor de TNF/metabolismo , Inhibidores del Factor de Necrosis Tumoral/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Dominios Homologos src/efectos de los fármacos , Familia-src Quinasas/metabolismo
2.
Cell Death Dis ; 12(4): 360, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33824270

RESUMEN

In the early 1990s, it has been described that LTα and LTß form LTα2ß and LTαß2 heterotrimers, which bind to TNFR1 and LTßR, respectively. Afterwards, the LTαß2-LTßR system has been intensively studied while the LTα2ß-TNFR1 interaction has been ignored to date, presumably due to the fact that at the time of identification of the LTα2ß-TNFR1 interaction one knew already two ligands for TNFR1, namely TNF and LTα. Here, we show that LTα2ß interacts not only with TNFR1 but also with TNFR2. We furthermore demonstrate that membrane-bound LTα2ß (memLTα2ß), despite its asymmetric structure, stimulates TNFR1 and TNFR2 signaling. Not surprising in view of its ability to interact with TNFR2, LTα2ß is inhibited by Etanercept, which is approved for the treatment of rheumatoid arthritis and also inhibits TNF and LTα.


Asunto(s)
Linfotoxina-alfa/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/agonistas , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Humanos , Membranas/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/agonistas , Receptores Tipo I de Factores de Necrosis Tumoral/efectos de los fármacos , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal/inmunología
3.
Inflammation ; 41(6): 2246-2264, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30097812

RESUMEN

Upregulation of chemokine CX3CL1 and its receptor CX3CR1 occurs in the diabetic human placenta. Metformin, an insulin-sensitizing biguanide, is used in the therapy of diabetic pregnancy. By preventing the activation of NF-κB, metformin exhibits anti-inflammatory properties. We examined the influence of hyperglycemia (25 mmol/L glucose; HG group; N = 36) on metformin-mediated effects on CX3CL1 and TNF-α production by placental lobules perfused extracorporeally. Additionally, CX3CR1 expression and contents of CX3CR1, TNF-α receptor 1 (TNFR1), and NF-κB proteins in the placental tissue were evaluated. Placentae perfused under normoglycemia (5 mmol/L glucose; NG group; N = 36) served as the control. Metformin (2.5 and 5.0 mg/L; subgroups B and C) lowered the production of CX3CL1 and TNF-α in a dose-dependent and time-dependent manner. Hyperglycemia did not weaken the strength of these metformin effects. Moreover, CX3CL1 levels after perfusion with 5.0 mg/L metformin were reduced by 33.28 and 33.83% (at 120 and 150 min, respectively) in the HG-C subgroup versus 24.98 and 23.66% in the NG-C subgroup, which indicated an augmentation of the metformin action over time in hyperglycemia. CX3CR1 expression was significantly higher in the HG-B and HG-C subgroups compared to that in the NG-B and NG-C subgroups. Increased CX3CR1 protein content in the placental lysates was observed in subgroups B and C. The two higher metformin concentrations significantly decreased the levels of NF-κBp65 protein content in both groups. However, the decrease was significantly stronger in hyperglycemia. TNFR1 upregulation in the HG group was not affected by metformin. Further studies on metformin therapy during pregnancy are needed, including safety issues.


Asunto(s)
Glucemia , Receptor 1 de Quimiocinas CX3C/metabolismo , Quimiocina CX3CL1/metabolismo , Metformina/farmacocinética , Circulación Placentaria/efectos de los fármacos , Adulto , Animales , Antiinflamatorios/farmacología , Femenino , Humanos , Hiperglucemia , Hipoglucemiantes/farmacología , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Embarazo , Receptores Tipo I de Factores de Necrosis Tumoral/efectos de los fármacos , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Adulto Joven
4.
Artículo en Chino | MEDLINE | ID: mdl-29996218

RESUMEN

Objective: To analyze the relationship between cognitive function and tumor necrosis factor receptor 1 (TNFR1) expression of occupational exposed workers to aluminum and provide the basis for the diagnosis of cognitive impairment. Methods: 140 cases Shanxi aluminum plant workers were collected in 2016 as the research object, including 70 potroom workers for exposure group, 70 non-electrolytic aluminum plant workers in the control group, respectively. Using mini mental status examination (MMSE), digit span test (DST), fuld object memory examination(FOME) and simple reaction time test(SRTT) evaluate the cognitive function of objects. Using graphite furnace atomic absorption method for the determination of plasma aluminum levels as an indicator of aluminum contact exposure of workers. Using RT-PCR method for detection of tumor necrosis factor receptor 1 (TNFR1) mRNA expression levels. And comparison group differences in cognitive and TNFR1 mRNA expression levels. Results: The plasma aluminum content of exposed group (77.12±27.18) µg/L higher than the control group (55.6±28.69)µg/L (P=0.000); Compared to control group, FOME and MMSE score was significantly increased in the exposed group (P=0.000, P=0.000), SRTT scores significantly higher in the exposed group (P=0.001), DST no significant difference in the control group (P=0.893). Compared to control group, The mRNA expression of TNFR1 was significantly higher in the exposed group(P=0.002); Compared to control group, The protein expression of TNFR1 was significantly higher in the exposed group (P=0.002). By correlation analysis in exposure group, plasma aluminum content was negatively correlated with MMSE and the DST (r=-0.284, r=-0.331, P<0.05) and positively correlated with the SRTT, TNFR1 (mRNA) and TNFR1(protein)(r=0.255, r=0.333, r=0.987, P<0.01), MMSE was negatively related to TNFR1 (mRNA) and TNFR1 (protein) (r=-0.268, r=-0.255, P<0.05); DST was negatively correlated with the SRTT and TNFR1 (protein)(r=-0.267, r=-0.330, P<0.05); SRTT was positively correlated with TNFR1 (protein)(r=0.243, P<0.05); TNFR1 (mRNA) was positively correlated with TNFR1 (protein)(r= 0.340, P<0.01). Conclusion: Cognitive function change of occupational exposed workers to aluminum was related to the increase of TNFR1 expression.


Asunto(s)
Aluminio/toxicidad , Cognición/efectos de los fármacos , Exposición Profesional/efectos adversos , Receptores Tipo I de Factores de Necrosis Tumoral/efectos de los fármacos , Trastornos del Conocimiento/inducido químicamente , Humanos
5.
Lung ; 196(3): 321-327, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29516177

RESUMEN

INTRODUCTION: A number of studies indicate that endothelin-1 (ET-1) may act as an inflammatory cell "gatekeeper," by regulating the influx of neutrophils following pulmonary injury. To further examine the role of ET-1 in modulating lung inflammation, hamsters were treated with an endothelin receptor antagonist (ERA), HJP272, either 1 h prior to intratracheal instillation of amiodarone (AM) or 24 h afterwards. METHODS: In both cases, the extent of lung injury and repair was determined by (1) histopathological changes; (2) neutrophil content in bronchoalveolar lavage fluid (BALF); (3) lung collagen content; (4) tumor necrosis factor receptor 1 expression by BALF macrophages; (5) BALF levels of (a) transforming growth factor beta-1, (b) stromal cell-derived factor 1 (commonly referred to as CXCL12), and (c) platelet-derived growth factor BB; (6) alveolar septal cell apoptosis. RESULTS: For each parameter, pretreatment with HJP272 resulted in a significant reduction compared to AM alone, whereas post-treatment was either ineffective or produced only a marginally significant change, suggesting that the course of lung inflammation and repair is programmed at a very early stage. CONCLUSIONS: This finding may explain why ERAs are not an effective treatment for human pulmonary fibrosis. Nevertheless, they may be useful as an adjunct to therapeutic regimens involving drugs that have fibrogenic potential.


Asunto(s)
Amiodarona/toxicidad , Antagonistas de los Receptores de Endotelina/farmacología , Endotelina-1/antagonistas & inhibidores , Hidroxiquinolinas/farmacología , Pulmón/efectos de los fármacos , Fibrosis Pulmonar/inducido químicamente , Vasodilatadores/toxicidad , Animales , Apoptosis/efectos de los fármacos , Becaplermina/efectos de los fármacos , Becaplermina/metabolismo , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Quimiocina CXCL12/efectos de los fármacos , Quimiocina CXCL12/metabolismo , Colágeno/efectos de los fármacos , Colágeno/metabolismo , Femenino , Pulmón/metabolismo , Pulmón/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Mesocricetus , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Alveolos Pulmonares/citología , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/patología , Receptores Tipo I de Factores de Necrosis Tumoral/efectos de los fármacos , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Factor de Crecimiento Transformador beta1/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo
6.
Ann Anat ; 216: 125-134, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29289711

RESUMEN

It is well known that bacterial lipopolysaccharide (LPS) induces migration of several cellular populations within the spleen. However, there are no data about the impact of LPS on B and T lymphocytes present in the red pulp. Therefore, we used an experimental model in which we tested the effects of intravenously injected LPS on the molecular, cellular and structural changes of the spleen, with special reference to the red pulp lymphocytes. We discovered that LPS induced a massive relocation of B and T lymphocytes from the splenic red pulp, which was independent of the tumor necrosis factor receptor-1 signaling axis. Early after LPS treatment, quantitative real-time PCR analysis revealed the elevated levels of mRNA encoding numerous chemokines and proinflammatory cytokines (XCL1, CXCL9, CXCL10, CCL3, CCL4, CCL5, CCL17, CCL20, CCL22, TNFα and LTα) which affect the navigation and activities of B and T lymphocytes in the lymphoid tissues. An extreme increase in mRNA levels for CCL20 was detected in the white pulp of the LPS-treated mice. The CCL20-expressing cells were localized in the PALS. Some smaller CCL20-expressing cells were evenly dispersed in the B cell zone. Thus, our study provides new knowledge of how microbial products could be involved in shaping the structure of lymphatic organs.


Asunto(s)
Lipopolisacáridos/farmacología , Linfocitos/efectos de los fármacos , Receptores Tipo I de Factores de Necrosis Tumoral/efectos de los fármacos , Bazo/citología , Animales , Linfocitos B/efectos de los fármacos , Quimiocina CCL20/genética , Quimiocina CCL20/metabolismo , Quimiocinas/biosíntesis , Citocinas/biosíntesis , Inmunohistoquímica , Recuento de Linfocitos , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal/efectos de los fármacos , Bazo/efectos de los fármacos , Linfocitos T/efectos de los fármacos
7.
Pharmazie ; 71(5): 235-7, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27348964

RESUMEN

Tumor necrosis factor (TNF)/TNF receptors (TNFR1/TNFR2) are considered to be potential drug targets to treat refractory diseases, including autoimmune diseases and malignant tumors. However, their specific functions, especially in the case of TNFR2, are poorly understood. In this study, we constructed a mouse TNFR2 (mTNFR2)-mediated biological assay system that shows no effects of mouse TNFR1 (mTNFR1) in order to screen mTNFR2-selective stimulating agents. Mouse TNFR1(-/-)R2(-/-) preadipocytes were transfected with the gene encoding the mTNFR2/mouse Fas (mFas) chimeric receptor in which the extracellular and transmembrane domains of mTNFR2 were fused to the intracellular domain of mFas. Our results demonstrated that this cell line exhibits highly sensitive mTNFR2-mediated cytotoxic effects. We propose that this mTNFR2-mediated biological assay system would be a useful tool to screen for mTNFR2-selective stimulating agents.


Asunto(s)
Adipocitos/citología , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptor fas/genética , Animales , Bioensayo/métodos , Línea Celular , Ratones , Ratones Noqueados , Receptores Tipo I de Factores de Necrosis Tumoral/efectos de los fármacos , Receptores Tipo II del Factor de Necrosis Tumoral/efectos de los fármacos , Transfección
8.
Medicine (Baltimore) ; 95(19): e3370, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27175635

RESUMEN

We have studied the response to intravenous immunoglobulins (IVIg) by a transcriptomic approach in 11 chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) patients (CIDP duration = 6 [0.83-6.5] years). RNA was extracted from cells in whole blood collected before and 3 weeks after IVIg treatment, and hybridized on Illumina chips. After RNA quality controls, gene expression was analyzed using statistical tests fitted for microarrays (R software, limma package), and a pathway analysis was performed using DAVID software. We identified 52 genes with expression that varied significantly after IVIg (fold change [FC] > 1.2, P < 0.001, false discovery rate [FDR] <0.05). Among these 52 genes, 7 were related to immunity, 3 were related to the tumor necrosis factor (TNF)-α receptor 1 (TNFR1) pathway (inhibitor of caspase-activated DNase (ICAD): FC = 1.8, P = 1.7E-7, FDR = 0.004; p21 protein-activated kinase 2 [PAK2]: FC = 1.66, P = 2.6E-5, FDR = 0.03; TNF-α-induced protein 8-like protein 1 [TNFAIP8L1]: P = 1.00E-05, FDR = 0.026), and 2 were related to Toll-like receptors (TLRs), especially TLRs 7 and 9, and were implicated in autoimmunity. These genes were UNC93B1 (FC = 1.6, P = 2E-5, FDR = 0.03), which transports TLRs 7 and 9 to the endolysosomes, and RNF216 (FC = 1.5, P = 1E-05, FDR = 0.03), which promotes TLR 9 degradation. Pathway analysis showed that the TNFR1 pathway was significantly lessened by IVIg (enrichment score = 24, Fischer exact test = 0.003). TNF-α gene expression was higher in responder patients than in nonresponders; however, it decreased after IVIg in responders (P = 0.04), but remained stable in nonresponders. Our data suggest the actions of IVIg on the TNFR1 pathway and an original mechanism involving innate immunity through TLRs in CIDP pathophysiology and the response to IVIg. We conclude that responder patients have stronger inflammatory activity that is lessened by IVIg.


Asunto(s)
Perfilación de la Expresión Génica , Inmunoglobulinas Intravenosas/farmacología , Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante/genética , Receptores Tipo I de Factores de Necrosis Tumoral/efectos de los fármacos , Transducción de Señal/genética , Receptores Toll-Like/efectos de los fármacos , Anciano , Formación de Anticuerpos/efectos de los fármacos , Formación de Anticuerpos/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Farmacogenética , Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante/sangre , Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante/tratamiento farmacológico , Receptores Tipo I de Factores de Necrosis Tumoral/sangre , Transducción de Señal/efectos de los fármacos , Receptores Toll-Like/sangre , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/efectos de los fármacos
9.
PLoS One ; 10(12): e0143686, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26625143

RESUMEN

Simvastatin (SIM) is anti-inflammatory. We used low density lipoprotein receptor knockout (LDLR-/-) mice and human aortic smooth muscle cells (HASMCs) as model systems to study the effect of SIM on arterial calcification and to explore the potential mechanisms contributing to this protective effect. High-fat diet (HFD) caused the LRLR -/- to develop dyslipidemia, diabetics, atherosclerosis and aortic smooth muscle calcification. SIM, N-acetyl cysteine (NAC, a ROS scavenger) and apocynin (APO, a NADPH oxidase inhibitor) did not significantly retard the development of dyslipidemia or diabetic. However, those treatments were still effective in attenuating the HFD-induced atherosclerosis and aortic smooth muscle calcification. These findings suggest that the protective effect of SIM against aortic calcification is not contributed by the cholesterol lowering effect. SIM, NAC and APO were found to attenuate the HFD induced elevation of serum TNF-α, soluble TNFR1 (sTNFR1), 3-nitro-tyrosine. We hypothesized that the pro-inflammatory cytokine, oxidative stress and TNFR1 played a role in inducing aortic calcification. We used HASMC to investigate the role of TNF-α, oxidative stress and TNFR1 in inducing aortic calcification and to elucidate the mechanism contributes the protective effect of SIM against aortic calcification. We demonstrated that treating HASMC with TNF-α induced cell Ca deposit and result in an increase in ALP, NADPH oxidase activity, NF-kB subunit p65, BMP2, MSX2, and RUNX2 expression. SIM suppressed the TNF-α induced activation of NADPH oxidase subunit p47, the above-mentioned bone markers and TNFR1 expression. Furthermore, p65, p47 and TNFR1 siRNAs inhibited the TNF-α-mediated stimulation of BMP-2, MSX2, RUNX2 expression. SIM, APO, and NAC either partially inhibit or completely block the TNF-α induced H2O2 or superoxide production. These results suggest that SIM may, independent of its cholesterol-lowering effect, suppresses the progression of vascular diseases through the inhibition of the inflammation mediators TNF-α and TNFR1.


Asunto(s)
Arteriosclerosis/metabolismo , Calcinosis/metabolismo , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Simvastatina/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Arteriosclerosis/patología , Dieta Alta en Grasa , Regulación hacia Abajo , Técnicas de Inactivación de Genes , Humanos , Masculino , Ratones , Ratones Noqueados , Receptores de LDL/genética , Receptores Tipo I de Factores de Necrosis Tumoral/efectos de los fármacos , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/genética
10.
Pediatr Res ; 78(6): 650-6, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26331770

RESUMEN

BACKGROUND: Adult human airway smooth muscle (ASM) produce cytokines involved in recruitment and survival of leukocytes within airway walls. Cytokine generation by adult ASM is glucocorticoid-sensitive. Whether developing lung ASM produces cytokines in a glucocorticoid-sensitive fashion is unknown. METHODS: Cultured fetal human ASM cells stimulated with TNF-α (0-20 ng/ml) were incubated with TNF-α receptor-blocking antibodies, fluticasone (1 and 100 nm), or vehicle. Supernatants and cells were assayed for the production of CCL5, CXCL10, and CXCL8 mRNA and protein and glucocorticoid receptor phosphorylation. RESULTS: CCL5, CXCL10, and CXCL8 mRNA and protein production by fetal ASM cell was significantly and dose-dependently following TNF-α treatment. Cytokine mRNA and protein production were effectively blocked by TNF-α R1 and R2 receptor neutralizing antibodies but variably inhibited by fluticasone. TNF-α-induced TNF-R1 and R2 receptor mRNA expression was only partially attenuated by fluticasone. Glucocorticoid receptor phosphorylation at serine (Ser) 211 but not at Ser 226 was enhanced by fluticasone. CONCLUSION: Production of CCL5, CXCL10, and CXCL8 by fetal ASM appears to involve pathways that are both qualitatively and mechanistically distinct to those described for adult ASM. The findings imply developing ASM has potential to recruit leukocyte into airways and, therefore, of relevance to childhood airway diseases.


Asunto(s)
Quimiotaxis de Leucocito/efectos de los fármacos , Citocinas/metabolismo , Fluticasona/farmacología , Glucocorticoides/farmacología , Pulmón/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Anticuerpos/farmacología , Células Cultivadas , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Citocinas/inmunología , Relación Dosis-Respuesta a Droga , Edad Gestacional , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Pulmón/embriología , Pulmón/inmunología , Pulmón/metabolismo , Miocitos del Músculo Liso/inmunología , Miocitos del Músculo Liso/metabolismo , Fosforilación , Receptores de Glucocorticoides/agonistas , Receptores de Glucocorticoides/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/efectos de los fármacos , Receptores Tipo I de Factores de Necrosis Tumoral/inmunología , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/efectos de los fármacos , Receptores Tipo II del Factor de Necrosis Tumoral/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Serina , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
11.
Crit Care Med ; 43(9): 1859-69, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26102252

RESUMEN

OBJECTIVES: There are no current pharmacological therapies for the prevention or treatment of acute respiratory distress syndrome. Early dysregulated inflammation likely plays a role in acute respiratory distress syndrome development and possibly acute respiratory distress syndrome outcomes. p38 mitogen-activated protein kinase is central to the regulation of multiple inflammatory mediators implicated in acute organ dysfunction and is the target for a novel class of cytokine-suppressive anti-inflammatory drugs. In preclinical models, p38 inhibitors reduce lung injury following pancreatitis and burn injury. DESIGN: We conducted a phase IIa, randomized, double-blind, placebo-controlled, parallel-group study to evaluate the safety and tolerability of dilmapimod, a novel p38 mitogen-activated protein kinase inhibitor, in patients at risk for developing acute respiratory distress syndrome admitted with an Injury Severity Score more than 16, excluding head trauma. Enrolled patients received 4- or 24-hour IV dilmapimod infusions at different doses or placebo, daily for 3 days, in four separate cohorts. SETTING: Multicenter randomized clinical trial of large, academic trauma centers. MEASUREMENTS AND MAIN RESULTS: Seventy-seven patients were enrolled. Although adverse events were common in this critically ill population, dilmapimod was well tolerated, with no clinically relevant safety findings. Pharmacokinetic models indicated that the higher dose of 10 mg given as continuous infusion over 24 hours had the most favorable plasma concentration profile. Likewise, measures of soluble inflammatory markers including interleukin-6, C-reactive peptide, interleukin-8, and soluble tumor necrosis factor receptor 1 were most different between this dosing arm and placebo. Although the study was not specifically designed with acute respiratory distress syndrome as an outcome, the number of patients who developed acute respiratory distress syndrome was small (2/77). CONCLUSIONS: The novel p38 mitogen-activated protein kinase inhibitor dilmapimod appears well tolerated and may merit further evaluation for prevention of acute respiratory distress syndrome and other organ injury in larger clinical trials. Furthermore, results of this early-phase trial may aid in design of future studies aimed at prevention of acute respiratory distress syndrome and other organ injury.


Asunto(s)
Mediadores de Inflamación/metabolismo , Piridonas/administración & dosificación , Piridonas/farmacología , Pirimidinas/administración & dosificación , Pirimidinas/farmacología , Síndrome de Dificultad Respiratoria/prevención & control , Heridas y Lesiones/tratamiento farmacológico , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Adulto , Proteína C-Reactiva/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Mortalidad Hospitalaria , Humanos , Interleucina-6/biosíntesis , Interleucina-8/efectos de los fármacos , Masculino , Persona de Mediana Edad , Piridonas/efectos adversos , Pirimidinas/efectos adversos , Receptores Tipo I de Factores de Necrosis Tumoral/efectos de los fármacos , Índices de Gravedad del Trauma
13.
Arterioscler Thromb Vasc Biol ; 34(3): 635-43, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24458710

RESUMEN

OBJECTIVE: Vascular lineage differentiation of stem/progenitor cells can contribute to both tissue repair and exacerbation of vascular diseases such as in vein grafts. The role of macrophages in controlling vascular progenitor differentiation is largely unknown and may play an important role in graft development. This study aims to identify the role of macrophages in vascular stem/progenitor cell differentiation and thereafter elucidate the mechanisms that are involved in the macrophage- mediated process. APPROACH AND RESULTS: We provide in vitro evidence that macrophages can induce endothelial cell (EC) differentiation of the stem/progenitor cells while simultaneously inhibiting their smooth muscle cell differentiation. Mechanistically, both effects were mediated by macrophage-derived tumor necrosis factor-α (TNF-α) via TNF-α receptor 1 and canonical nuclear factor-κB activation. Although the overexpression of p65 enhanced EC (or attenuated smooth muscle cell) differentiation, p65 or TNF-α receptor 1 knockdown using lentiviral short hairpin RNA inhibited EC (or rescued smooth muscle cell) differentiation in response to TNF-α. Furthermore, TNF-α-mediated EC differentiation was driven by direct binding of nuclear factor-κB (p65) to specific VE-cadherin promoter sequences. Subsequent experiments using an ex vivo decellularized vessel scaffold confirmed an increase in the number of ECs and reduction in smooth muscle cell marker expression in the presence of TNF-α. The lack of TNF-α in a knockout mouse model of vein graft decreased endothelialization and significantly increased thrombosis formation. CONCLUSIONS: Our study highlights the role of macrophages in directing vascular stem/progenitor cell lineage commitment through TNF-α-mediated TNF-α receptor 1 and nuclear factor-κB activation that is likely required for endothelial repair in vascular diseases such as vein graft.


Asunto(s)
Células Madre Adultas/citología , Células Endoteliales/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/fisiología , Macrófagos Peritoneales/fisiología , Miocitos del Músculo Liso/efectos de los fármacos , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Células Madre Adultas/efectos de los fármacos , Proteínas Angiogénicas/farmacología , Animales , Antígenos CD/biosíntesis , Antígenos CD/genética , Apoptosis , Cadherinas/biosíntesis , Cadherinas/genética , Línea Celular , Linaje de la Célula , Medios de Cultivo Condicionados/farmacología , Células Endoteliales/citología , Endotelio Vascular/citología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Interleucina-6/farmacología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Neointima/patología , Neovascularización Fisiológica/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/fisiología , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Quimera por Radiación , Receptores Tipo I de Factores de Necrosis Tumoral/efectos de los fármacos , Receptores Tipo I de Factores de Necrosis Tumoral/fisiología , Proteínas Recombinantes/farmacología , Transducción de Señal , Trombofilia/etiología , Trombofilia/fisiopatología , Andamios del Tejido , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/deficiencia , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/farmacología , Venas/trasplante
14.
J Periodontal Res ; 49(4): 508-17, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24102429

RESUMEN

OBJECTIVE AND BACKGROUND: Periodontitis is an inflammatory disorder of the supporting tissue of teeth, which is composed of gingival soft tissue, cementum covering the tooth root, alveolar bone and periodontal ligament. The receptor activator of nuclear factor kappa B ligand (RANKL) is known to be an essential factor for osteoclastogenesis. Recent clinical studies indicate that levels of RANKL in the gingival crevicular fluid are increased while levels of its decoy receptor, osteoprotegerin (OPG), are decreased in patients with periodontitis. Although the gingival sulcus is composed of gingival tissue, RANKL and OPG expression in gingival epithelial cells is not fully understood. The aim of this study is to investigate the expression of RANKL and OPG in gingival tissue and which factors regulate RANKL expression in gingival epithelial cells. MATERIAL AND METHODS: Reverse transcriptase polymerase chain reaction analysis, western blotting and immunohistochemistry were performed to confirm RANKL and OPG expression in gingival epithelial cells (GECs) and in gingival tissue. Immunostaining was also examined to confirm tumor necrosis factor (TNF)-α and TNF receptor type 1 (TNFR1) expression in gingival tissue. Ca9-22 cells, a human gingival epithelial cell line and human primary GECs were treated with TNF-α. Ca9-22 cells were treated by antibodies against TNF receptors, an inhibitor and an activator of protein kinase A (PKA) signaling and inhibitors of p38, Erk and NF-κB signaling to examine TNF-α-RANKL signaling pathways. RESULTS: RANKL mRNA and protein were expressed in GECs. Immunohistochemistry also showed RANKL expression in gingival tissue. On the other hand, the reverse transcriptase polymerase chain reaction and immunohistochemistry assay showed that GECs did not express OPG. In addition, TNF-α and TNFR1 proteins were expressed in junctional epithelium. TNF-α increased RANKL expression in GECs. TNF-α-induced RANKL expression was inhibited by an antibody against TNFR1 and an inhibitor of PKA signaling. Surprisingly, forskolin, a PKA activator, increased TNF-α-induced RANKL expression. CONCLUSION: RANKL, TNF and TNFR1 were coexpressed in junctional epithelium of gingival tissue. TNF-α induced RANKL expression via TNFR1 and PKA signaling in GECs of junctional epithelium.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/efectos de los fármacos , Encía/efectos de los fármacos , Ligando RANK/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Animales , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Colforsina/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Inserción Epitelial/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Femenino , Encía/citología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , FN-kappa B/antagonistas & inhibidores , Osteoprotegerina/efectos de los fármacos , Receptores Tipo I de Factores de Necrosis Tumoral/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
15.
Anesth Analg ; 117(6): 1493-502, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24257399

RESUMEN

BACKGROUND: Glutamate homeostasis and microglia activation play an important role in the development and maintenance of neuropathic pain. We designed this investigation to examine whether ultra-low dose naloxone administered alone or in combination with morphine could alter the concentration of the excitatory amino acids (EAAs) glutamate and aspartate, as well as the expression of tumor necrosis factor-α (TNF-α) and its receptors (TNFR1 and TNFR2) in the spinal cord dorsal horn of rats with partial sciatic nerve transection (PST). METHODS: Male Wistar rats underwent intrathecal catheter implantation for drug delivery and were divided in 7 groups: sham-operated + saline (sham), PST + saline (S), PST + 15 ng naloxone (n), PST + 15 µg naloxone (N), PST + 10 µg morphine (M), PST + 15 ng naloxone + 10 µg morphine (Mn), PST + 15 µg naloxone + 10 µg morphine (MN). Thermal withdrawal latency and mechanical withdrawal threshold, TNF-α and TNFR expression in the spinal cord and dorsal root ganglia, and EAAs glutamate and aspartate concentration in cerebrospinal fluid dialysates were measured. RESULTS: Ten days after PST, rats developed hyperalgesia (P < 0.0001) and allodynia (P < 0.0001), and increased TNF-α (P < 0.0001) and TNFR1 expression (P = 0.0009) were measured in the ipsilateral spinal cord dorsal horn. The antihyperalgesic and antiallodynic effects of morphine (10 µg) were abolished by high-dose naloxone (15 µg; P = 0.0031) but enhanced by ultra-low dose naloxone (15 ng; P = 0.0015), and this was associated with a reduction of TNF-α (P < 0.0001) and TNFR1 (P = 0.0009) expression in the spinal cord dorsal horn and EAAs concentration (glutamate: P = 0.0001; aspartate: P = 0.004) in cerebrospinal fluid dialysate. Analysis of variance (ANOVA) or Student t test with Bonferroni correction were used for statistical analysis. CONCLUSIONS: Ultra-low dose naloxone enhances the antihyperalgesia and antiallodynia effects of morphine in PST rats, possibly by reducing TNF-α and TNFR1 expression, and EAAs concentrations in the spinal dorsal horn. Ultra-low dose naloxone may be a useful adjuvant for increasing the analgesic effect of morphine in neuropathic pain conditions.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Hiperalgesia/tratamiento farmacológico , Morfina/administración & dosificación , Naloxona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Umbral del Dolor/efectos de los fármacos , Células del Asta Posterior/efectos de los fármacos , Receptores Tipo I de Factores de Necrosis Tumoral/efectos de los fármacos , Ciática/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Ácido Aspártico/metabolismo , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación hacia Abajo , Sinergismo Farmacológico , Ácido Glutámico/metabolismo , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Hiperalgesia/psicología , Inyecciones Espinales , Masculino , Células del Asta Posterior/metabolismo , Ratas , Ratas Wistar , Tiempo de Reacción/efectos de los fármacos , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/efectos de los fármacos , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Nervio Ciático/cirugía , Ciática/metabolismo , Ciática/fisiopatología , Ciática/psicología , Factores de Tiempo
16.
J Vasc Res ; 50(1): 21-34, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23095497

RESUMEN

The sex hormone estradiol (E(2)) appears to mediate both anti-atherogenic and pro-inflammatory effects in premenopausal women, suggesting a complex immunomodulatory role. Tumor necrosis factor (TNF) is a key pro-inflammatory cytokine involved in the pathogenesis of atherosclerosis and other inflammatory diseases. Alterations at the TNF receptors (TNFRs) and their downstream signaling/transcriptional pathways can affect inflammatory responses. Given this background, we hypothesized that chronic E(2) exposure would alter endothelial inflammatory response involving modulation at the levels of TNFRs and signaling pathways. HUVECs were used as the model system. Pre-treatment with E(2) did not significantly alter TNF-induced upregulation of pro-inflammatory molecules ICAM-1 (3-6 times) and VCAM-1 (5-7 times). However, pharmacological inhibition of transcriptional pathways suggested a partial shift from NF-ĸB (from 97 to 64%) towards the JNK/AP-1 pathway in ICAM-1 upregulation on E(2) treatment. In contrast, VCAM-1 expression remained NF-ĸB dependent in both control (∼96%) and E(2) treated (∼85%) cells. The pro-inflammatory TNF effects were mediated by TNFR1. Interestingly, E(2) pre-treatment increased TNFR2 levels in these cells. Concomitant TNFR2 activation (but not TNFR1 activation alone) led to the shift towards JNK/AP-1-mediated ICAM-1 upregulation in E(2)-treated cells, suggesting the effects of chronic E(2) to be dependent on TNFR2 signaling.


Asunto(s)
Estradiol/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Inflamación/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Proteínas I-kappa B/metabolismo , Inflamación/genética , Inflamación/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/efectos de los fármacos , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Factor de Transcripción AP-1/metabolismo , Transcripción Genética/efectos de los fármacos , Molécula 1 de Adhesión Celular Vascular/metabolismo
17.
Brain Behav Immun ; 27(1): 133-44, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23072727

RESUMEN

The T-cell derived cytokine CD40 ligand is overexpressed in patients with autoimmune diseases. Through activation of its receptor, CD40 ligand leads to a tumor necrosis factor (TNF) receptor 1 (TNFR1) dependent impairment of locomotor activity in mice. Here we report that this effect is explained through a promotion of sleep, which was specific to non-rapid eye movement (NREM) sleep while REM sleep was suppressed. The increase in NREM sleep was accompanied by a decrease in EEG delta power during NREM sleep and by a decrease in the expression of transcripts in the cerebral cortex known to be associated with homeostatic sleep drive, such as Homer1a, Early growth response 2, Neuronal pentraxin 2, and Fos-like antigen 2. The effect of CD40 activation was mimicked by peripheral TNF injection and prevented by the TNF blocker etanercept. Our study indicates that sleep-wake dysregulation in autoimmune diseases may result from CD40 induced TNF:TNFR1 mediated alterations of molecular pathways, which regulate sleep-wake behavior.


Asunto(s)
Encéfalo , Antígenos CD40/metabolismo , Sueño/fisiología , Factor de Necrosis Tumoral alfa , Animales , Anticuerpos Monoclonales/farmacología , Enfermedades Autoinmunes/complicaciones , Enfermedades Autoinmunes/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Proteína C-Reactiva/efectos de los fármacos , Proteína C-Reactiva/genética , Proteína C-Reactiva/metabolismo , Antígenos CD40/agonistas , Proteínas Portadoras/efectos de los fármacos , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteína 2 de la Respuesta de Crecimiento Precoz/efectos de los fármacos , Proteína 2 de la Respuesta de Crecimiento Precoz/genética , Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , Electroencefalografía/efectos de los fármacos , Electromiografía , Etanercept , Antígeno 2 Relacionado con Fos/efectos de los fármacos , Antígeno 2 Relacionado con Fos/genética , Antígeno 2 Relacionado con Fos/metabolismo , Proteínas de Andamiaje Homer , Inmunoglobulina G/farmacología , Factores Inmunológicos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Proteínas del Tejido Nervioso/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Receptores del Factor de Necrosis Tumoral , Receptores Tipo I de Factores de Necrosis Tumoral/efectos de los fármacos , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Sueño/efectos de los fármacos , Trastornos del Sueño del Ritmo Circadiano/complicaciones , Trastornos del Sueño del Ritmo Circadiano/metabolismo , Sueño REM/efectos de los fármacos , Sueño REM/fisiología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
18.
Neuropsychobiology ; 63(3): 169-76, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21228609

RESUMEN

BACKGROUND: Depressive disorder after myocardial infarction (MI) is associated with increased cardiac morbidity and mortality. Immune activity such as inflammation might be implicated as an underlying mechanism. The purpose of this study is to investigate whether the response to an antidepressant in post-MI depression is associated with changes of inflammatory markers in serum. METHODS: In a double-blind placebo-controlled study with mirtazapine 30 mg/day (50 patients), the antidepressive effect was related to immune activation parameters. The cytokines interleukin 6 (IL-6) and tumor necrosis factor α (TNF-α), the soluble cytokine receptors sIL-6R, sTNF-R1 and sTNF-R2, and the inflammation-sensitive plasma proteins C-reactive protein and neopterin were assessed. RESULTS: Subgroup analyses revealed a highly significant correlation of pronounced sTNF-R1 increase with a decrease in depressive symptoms in antidepressant responders. CONCLUSION: Significant effects on inflammation accompany the therapeutic efficacy of mirtazapine in contrast to the therapeutic efficacy of placebo and the nontherapeutic efficacy of mirtazapine.


Asunto(s)
Antidepresivos Tricíclicos/uso terapéutico , Depresión/tratamiento farmacológico , Depresión/inmunología , Mianserina/análogos & derivados , Infarto del Miocardio/complicaciones , Infarto del Miocardio/inmunología , Receptores Tipo I de Factores de Necrosis Tumoral/efectos de los fármacos , Adulto , Anciano , Antidepresivos Tricíclicos/farmacología , Depresión/sangre , Depresión/complicaciones , Femenino , Humanos , Mediadores de Inflamación/sangre , Masculino , Mianserina/farmacología , Mianserina/uso terapéutico , Persona de Mediana Edad , Mirtazapina , Infarto del Miocardio/sangre , Infarto del Miocardio/tratamiento farmacológico , Receptores Tipo I de Factores de Necrosis Tumoral/sangre
19.
Planta Med ; 77(2): 117-21, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20717874

RESUMEN

Dendrobium alkaloids (DNLA), extracted from Dendrobium nobile Lindl. whose botanical name is Dendrobium moniliforme, Orchidaceae family, were studied for their effect on lipopolysaccharide (LPS)-induced memory impairment in rats. SD rats were pretreated with DNLA (40, 80, 160 mg/kg/d for 7 d), followed by LPS (50 µg) injection into the right lateral ventricle to produce memory impairment. DNLA treatment continued for another 13 days. The spatial behavior was tested by the Morris water maze; the level of tumor necrosis factor receptor 1 (TNFR1) mRNA was detected by real time RT-PCR, and the protein level of TNFR1, nuclear factor kappa-B (NF- κB) and phosphorylated p38 mitogen-activated protein kinases (p-p38 MAPK) by Western blotting. The results showed that DNLA significantly improved the neurobehavioral performance and prevented LPS-induced elevation in TNFR1 mRNA and protein levels. LPS-induced activation of p38 MAPK and NF- κB pathway was also suppressed. In conclusion, DNLA is effective in protecting against LPS-induced brain impairment, and this effect is due, at least in part, to prevent overexpression of TNFR1 via inhibition of p-p38 MAPK and the downstream NF- κB signal pathway.


Asunto(s)
Alcaloides/uso terapéutico , Dendrobium/química , Lipopolisacáridos/toxicidad , Trastornos de la Memoria/prevención & control , Alcaloides/farmacología , Animales , Regulación de la Expresión Génica , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/tratamiento farmacológico , FN-kappa B/biosíntesis , FN-kappa B/efectos de los fármacos , Plantas Medicinales/química , Ratas , Ratas Sprague-Dawley , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Receptores Tipo I de Factores de Necrosis Tumoral/efectos de los fármacos , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos
20.
J Neurosci ; 30(36): 12210-8, 2010 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-20826683

RESUMEN

Tumor necrosis factor alpha (TNF-alpha) is a potent cytokine in neurodegenerative disorders, but its precise role in particular brain disorders is ambiguous. In motor neuron (MN) disease of the mouse, exemplified by the model wobbler (WR), TNF-alpha causes upregulation of the metalloprotease-disintegrin ADAM8 (A8) in affected brain regions, spinal cord, and brainstem. The functional role of A8 during MN degeneration in the wobbler CNS was investigated by crossing WR with A8-deficient mice: a severely aggravated neuropathology was observed for A8-deficient WR compared with WR A8(+/-) mice, judged by drastically reduced survival [7 vs 81% survival at postnatal day 50 (P50)], accelerated force loss in the forelimbs, and terminal akinesis. In vitro protease assays using soluble A8 indicated specific cleavage of a TNF-alpha receptor 1 (p55 TNF-R1) but not a TNF-R2 peptide. Cleavage of TNF-R1 was confirmed in situ, because levels of soluble TNF-R1 were increased in spinal cords of standard WR compared with wild-type mice but not in A8-deficient WR mice. In isolated primary neurons and microglia, TNF-alpha-induced TNF-R1 shedding was dependent on the A8 gene dosage. Furthermore, exogenous TNF-alpha showed higher toxicity for cultured neurons from A8-deficient than for those from wild-type mice, demonstrating that TNF-R1 shedding by A8 is neuroprotective. Our results indicate an essential role for ADAM8 in modulating TNF-alpha signaling in CNS diseases: a feedback loop integrating TNF-alpha, ADAM8, and TNF-R1 shedding as a plausible mechanism for TNF-alpha mediated neuroprotection in situ and a rationale for therapeutic intervention.


Asunto(s)
Proteínas ADAM/metabolismo , Antígenos CD/metabolismo , Regulación de la Expresión Génica/genética , Proteínas de la Membrana/metabolismo , Enfermedades Neurodegenerativas/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas ADAM/deficiencia , Proteínas ADAM/genética , Proteínas ADAM/farmacología , Animales , Animales Recién Nacidos , Antígenos CD/genética , Antígenos CD/farmacología , Recuento de Células/métodos , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática/métodos , Humanos , Estimación de Kaplan-Meier , Antígenos Comunes de Leucocito/metabolismo , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Atrofia Muscular/genética , Proteínas del Tejido Nervioso/metabolismo , Enfermedades Neurodegenerativas/mortalidad , Enfermedades Neurodegenerativas/patología , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , ARN Mensajero/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/efectos de los fármacos , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...