Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 262
Filtrar
1.
Zhongguo Gu Shang ; 37(1): 61-8, 2024 Jan 25.
Artículo en Chino | MEDLINE | ID: mdl-38286453

RESUMEN

OBJECTIVE: To explore the effect of shikonin on the recovery of nerve function after acute spinal cord injury(SCI) in rats. METHODS: 96 male Sprague-Dawley(SD)rats were divided into 4 groups randomly:sham operation group (Group A), sham operation+shikonin group (Group B), SCI+ DMSO(Group C), SCI+shikonin group (Group D).The acute SCI model of rats was made by clamp method in groups C and D . After subdural catheterization, no drug was given in group A. rats in groups B and D were injected with 100 mg·kg-1 of shikonin through catheter 30 min after modeling, and rats in group C were given with the same amount of DMSO, once a day until the time point of collection tissue. Basso-Beattie-Bresnahan(BBB) scores were performed on 8 rats in each group at 6, 12, and 3 d after moneling, and oblique plate tests were performed on 1, 3, 7 and 14 d after modeling, and then spinal cord tissues were collected. Eight rats were intraperitoneally injected with propidine iodide(PI) 1 h before sacrificed to detection PI positive cells at 24 h in each group. Eight rats were sacrificed in each group at 24 h after modeling, the spinal cord injury was observed by HE staining.The Nissl staining was used to observe survivor number of nerve cells. Western-blot technique was used to detect the expression levels of Bcl-2 protein and apoptosis related protein RIPK1. RESULTS: After modeling, BBB scores were normal in group A and B, but in group C and D were significantly higher than those in group A and B. And the scores in group D were higher than those in group C in each time point (P<0.05). At 12 h after modeling, the PI red stained cells in group D were significantly reduced compared with that in group C, and the disintegration of neurons was alleviated(P<0.05). HE and Nissl staining showed nerve cells with normal morphology in group A and B at 24h after operation. The degree of SCI and the number of neuronal survival in group D were better than those in group C, the difference was statistically significant at 24h (P<0.05). The expression of Bcl-2 and RIPK1 proteins was very low in group A and B;The expression of RIPK1 was significantly increased in Group C and decreased in Group D, with a statistically significant difference (P<0.05);The expression of Bcl-2 protein in group D was significantly higher than that in group C (P<0.05). CONCLUSION: Shikonin can alleviate the pathological changes after acute SCI in rats, improve the behavioral score, and promote the recovery of spinal nerve function. The specific mechanism may be related to the inhibition of TNFR/RIPK1 signaling pathway mediated necrotic apoptosis.


Asunto(s)
Dimetilsulfóxido , Naftoquinonas , Traumatismos de la Médula Espinal , Animales , Masculino , Ratas , Dimetilsulfóxido/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas Sprague-Dawley , Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/metabolismo , Receptores del Factor de Necrosis Tumoral/efectos de los fármacos , Receptores del Factor de Necrosis Tumoral/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/efectos de los fármacos , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
2.
Life Sci ; 206: 77-83, 2018 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-29792879

RESUMEN

AIMS: Apoptosis of podocytes plays a crucial role in diabetic nephropathy (DN) development, and astragaloside (AS-IV) has a significant impact on podocyte apoptosis. This study aims to explore the effect of AS-IV on diabetic nephropathy progression. MATERIALS AND METHODS: The diabetic nephropathy model was established in rats with streptozotocin (STZ) injection. The albuminuria was examined by using the enzyme linked immunosorbent assay (ELISA). The expression of miR-378, tumor-necrosis factor (TNF) receptor (TNFR)-associated factor 5 (TRAF5) mRNA and protein was analyzed by qRT-PCR and western blot, respectively. Cell transfection was conducted for modulating endogenous expression of miR-378. Dual luciferase reporter assay was used to evaluate the interaction between miR-378 and TRAF5. The terminal deoxynucleotidy transferase dUTP nick end labeling (TUNEL) staining assay was performed for apoptosis detection. KEY FINDINGS: AS-IV protected diabetic rats from developing into diabetic nephropathy. The expression of miR-378 was down-regulated in diabetic nephropathy rats, which was reversed by AS-IV. AS-IV enhanced the expression of miR-378 in podocytes treated with high glucose. MiR-378 negatively regulated TRAF5. AS-IV inhibited the expression of TRAF5 through miR-378. AS-IV suppressed apoptosis of podocytes via targeting miR-378. SIGNIFICANCE: AS-IV suppresses apoptosis of the podocytes through the miR-378/TRAF5 signaling pathway, and thereby repressing diabetic nephropathy development.


Asunto(s)
Apoptosis/efectos de los fármacos , Nefropatías Diabéticas/patología , MicroARNs/efectos de los fármacos , Podocitos/efectos de los fármacos , Saponinas/farmacología , Factor 5 Asociado a Receptor de TNF/efectos de los fármacos , Triterpenos/farmacología , Albuminuria/inducido químicamente , Animales , Células Cultivadas , Diabetes Mellitus Experimental/patología , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratas , Ratas Sprague-Dawley , Receptores del Factor de Necrosis Tumoral/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transfección
3.
Bratisl Lek Listy ; 118(8): 443-448, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29050480

RESUMEN

OBJECTIVE: Our study aimed to investigate the possible modifying effects of leptin and combined use of resveratrol on rat renal I/R injury and their relationship on signal pathways and apoptosis-related mechanisms. BACKGROUND: Renal ischemia-reperfusion (I/R) injury is an important cause of acute renal failure. METHODS: Male Sprague Dawley rats were divided into 5 groups: Control, I/R, I/R+leptin, I/R+resveratrol and I/R+leptin+resveratrol. Leptin (10 µg/kg BW) was administered (i.p.) 30 min prior to I/R. Resveratrol was administered by gavage at 20 mg/kg BW per d for 12 d prior to I/R. The left renal artery was exposed to 1 h of ischemia and 1 h of reperfusion. RESULTS: Resveratrol treatment alone increased TNF-α, TNF-α R1, NF-κB, SIRT-1, STAT1 and STAT3 mRNA levels and decreased caspase 3 protein levels. Leptin treatment alone significantly decreased the caspase 3 protein levels. The combined use of resveratrol and leptin significantly increased STAT3, and caspase 3 mRNA levels, and decreased the caspase 3 protein levels. Apoptosis was significantly decreased especially in the leptin and leptin+resveratrol groups. CONCLUSION: The present study suggest that a combined use of resveratrol and leptin has preventive and regulatory effects on renal I/R injury; the mechanism involves decreasing apoptosis, likely by altering the JAK/STAT pathway and SIRT1 expression (Fig. 8, Ref. 24).


Asunto(s)
Antioxidantes/farmacología , Riñón/efectos de los fármacos , Leptina/farmacología , Daño por Reperfusión/genética , Sirtuina 1/efectos de los fármacos , Estilbenos/farmacología , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/efectos de los fármacos , Caspasa 3/genética , Caspasa 3/metabolismo , Expresión Génica , Riñón/metabolismo , Masculino , FN-kappa B/efectos de los fármacos , FN-kappa B/genética , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores del Factor de Necrosis Tumoral/efectos de los fármacos , Receptores del Factor de Necrosis Tumoral/genética , Daño por Reperfusión/metabolismo , Resveratrol , Factor de Transcripción STAT1/efectos de los fármacos , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT3/efectos de los fármacos , Factor de Transcripción STAT3/genética , Transducción de Señal , Sirtuina 1/genética , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/genética
4.
Curr Drug Targets ; 16(4): 393-408, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25706111

RESUMEN

The tumor necrosis factor (TNF) superfamily (TNFSF) contains about thirty structurally related receptors (TNFSFRs) and about twenty protein ligands that bind to one or more of these receptors. Almost all of these cell surface protein-protein interactions (PPIs) represent high-value therapeutic targets for inflammatory or immune modulation in autoimmune diseases, transplant recipients, or cancers, and there are several biologics including antibodies and fusion proteins targeting them that are in various phases of clinical development. Small-molecule inhibitors or activators could represent possible alternatives if the difficulties related to the targeting of protein-protein interactions by small molecules can be addressed. Compounds proving the feasibility of such approaches have been identified through different drug discovery approaches for a number of these TNFSFR-TNFSF type PPIs including CD40-CD40L, BAFFR-BAFF, TRAIL-DR5, and OX40-OX40L. Corresponding structural, signaling, and medicinal chemistry aspects are briefly reviewed here. While none of these small-molecule modulators identified so far seems promising enough to be pursued for clinical development, they provide proof-of-principle evidence that these interactions are susceptible to small-molecule modulation and can serve as starting points toward the identification of more potent and selective candidates.


Asunto(s)
Descubrimiento de Drogas , Receptores del Factor de Necrosis Tumoral/metabolismo , Factores de Necrosis Tumoral/metabolismo , Animales , Sitios de Unión , Humanos , Ligandos , Modelos Moleculares , Terapia Molecular Dirigida , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Receptores del Factor de Necrosis Tumoral/química , Receptores del Factor de Necrosis Tumoral/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad , Factores de Necrosis Tumoral/química
5.
J Periodontal Res ; 50(4): 423-33, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25139560

RESUMEN

BACKGROUND AND OBJECTIVE: Smoking cigarettes increases the risk of oral tissue damage leading to periodontal disease. Gingival fibroblasts, the predominant cell type inhabiting gingival connective tissue, play a critical role in remodeling and maintaining gingival structure. The objective of this study was to investigate the effect of long-term exposure to cigarette smoke on human gingival fibroblast survival/apoptosis and the molecular pathways involved in these cell responses. MATERIAL AND METHODS: Human gingival fibroblasts were extracted from healthy non-smokers and cultured in the presence of cigarette smoke condensate (CSC). At the end of each time point, cell growth was evaluated by means of MTT assay. Apoptotic and necrotic gene's expression was investigated by polymerase chain reaction array and by annexin V/propidium iodide staining and cell cycle assays. Western blot was used to investigate Bax and p53 proteins. These tests were supported by caspase 3 activity analyses. RESULTS: High levels of CSC decreased cell growth and deregulated cell cycle progression by increasing the G(0)/G(1) and reducing the S and G(2)/M phases of the gingival fibroblasts. Polymerase chain reaction arrays revealed the activation of several apoptotic genes by CSC, including TNF receptors, caspases, Bax and p53. This was supported by increases in the Bax and p53 protein levels as well as by an elevated activity of caspase-3 in the CSC-exposed cells. Immunofluorescence staining demonstrated that both Bax and caspase-3 displayed a cytosolic and mitochondrial distribution in the CSC-exposed gingival fibroblasts, compared to controls. The damaging effect of CSC on gingival fibroblast growth was also supported by the decrease in interleukin 6 and 8 secretion by the gingival fibroblasts. CONCLUSION: These results suggest that CSC may contribute to deregulating fibroblast functions. This can compromise fibroblast-epithelial cell interactions, which ultimately increases the risk of gingival tissue damage and the onset of periodontitis.


Asunto(s)
Caspasa 3/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Encía/efectos de los fármacos , Humo/efectos adversos , Productos de Tabaco/efectos adversos , Proteína p53 Supresora de Tumor/efectos de los fármacos , Proteína X Asociada a bcl-2/efectos de los fármacos , Apoptosis/efectos de los fármacos , Recuento de Células , Técnicas de Cultivo de Célula , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citosol/efectos de los fármacos , Encía/citología , Humanos , Interleucina-6/análisis , Interleucina-8/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Receptores del Factor de Necrosis Tumoral/efectos de los fármacos , Factores de Tiempo
6.
Toxicol In Vitro ; 29(1): 242-50, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25448810

RESUMEN

Cadmium (Cd) is a human carcinogen that likely acts via epigenetic mechanisms. However, the precise role of Cd in melanoma remains to be defined. The goals of this study are to: (i) examine the effect of Cd on the proliferation rate of cutaneous and uveal melanoma cells; (ii) identify the genes affected by Cd exposure; (iii) understand whether epigenetic changes are involved in the response to Cd. The cell growth capacity increased at 48 h after Cd treatment at doses ranging from 0.5 to 10 µM. The research on the key genes regulating proliferation has shown that aberrant methylation is responsible for silencing of p16(INK4A) and caspase 8 in uveal and cutaneous melanoma cells, respectively. The methylation and expression patterns of p14(ARF), death receptors 4/5, and E-cadherin remained unmodified after Cd treatment in all the cell lines analyzed. Ectopic expression of p16(INK4A) abolished the overgrowth of uveal melanoma cells in response to Cd and the overexpression of caspase 8 drastically increased the apoptotic rate of Cd-treated cutaneous melanoma cells. In conclusion, our data suggest that hypermethylation of p16(INK4A) and caspase 8 represents the most common event linked to Cd-induced stimulation of cell growth and inhibition of cell death pathway in melanoma.


Asunto(s)
Cloruro de Cadmio/toxicidad , Epigénesis Genética/efectos de los fármacos , Melanoma/inducido químicamente , Caspasa 8/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Humanos , Metilación/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/efectos de los fármacos , Receptores del Factor de Necrosis Tumoral/efectos de los fármacos , Neoplasias Cutáneas/inducido químicamente , Proteína p14ARF Supresora de Tumor/efectos de los fármacos , Neoplasias de la Úvea/inducido químicamente
8.
Br J Pharmacol ; 170(4): 748-64, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23957828

RESUMEN

Fibroblast growth factor-inducible 14 (Fn14) is a member of the tumour necrosis factor (TNF) receptor family that is induced in a variety of cell types in situations of tissue injury. Fn14 becomes activated by TNF-like weak inducer of apoptosis (TWEAK), a typical member of the TNF ligand family. TWEAK is constitutively expressed by monocytes and some tumour cell lines and also shows cytokine inducible expression in various other cell types. Fn14 activation results in stimulation of signalling pathways culminating in the activation of NFκB transcription factors and various MAPKs but might also trigger the PI3K/Akt pathway and GTPases of the Rho family. In accordance with its tissue damage-associated expression pattern and its pleiotropic proinflammatory signalling capabilities, the TWEAK-Fn14 system has been implicated in a huge number of pathologies. The use of TWEAK- and Fn14-knockout mice identified the TWEAK-Fn14 system as a crucial player in muscle atrophy, cerebral ischaemia, kidney injury, atherosclerosis and infarction as well as in various autoimmune scenarios including experimental autoimmune encephalitis, rheumatoid arthritis and inflammatory bowel disease. Moreover, there is increasing preclinical evidence that Fn14 targeting is a useful option in tumour therapy. Based on a discussion of the signalling capabilities of TWEAK and Fn14, this review is focused on two major issues. On the one hand, on the molecular and cellular basis of the TWEAK/Fn14-related pathological outcomes in the aforementioned diseases and on the other hand, on the preclinical experience that have been made so far with TWEAK and Fn14 targeting drugs.


Asunto(s)
Diseño de Fármacos , Receptores del Factor de Necrosis Tumoral/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factores de Necrosis Tumoral/metabolismo , Animales , Anticuerpos/farmacología , Citocina TWEAK , Humanos , Ligandos , Terapia Molecular Dirigida , Receptores del Factor de Necrosis Tumoral/metabolismo , Proteínas Recombinantes/farmacología , Receptor de TWEAK , Factores de Necrosis Tumoral/farmacología
9.
Ther Deliv ; 4(3): 369-94, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23442082

RESUMEN

Receptor-based targeting of therapeutics may be a fascinating proposition to improve the therapeutic efficacy of encapsulated drugs. The development of safe and effective nanomedicines is a prerequisite in the current nanotechnological scenario. Currently, the surface engineering of nanocarriers has attracted great attention for targeted therapeutic delivery by selective binding of targeting ligand to the specific receptors present on the surface of cells. In this review, we have discussed the current status of various receptors such as transferrin, lectoferrin, lectin, folate, human EGF receptor, scavenger, nuclear and integrin, which are over-expressed on the surface of cancer cells; along with the relevance of targeted delivery systems such as nanoparticles, polymersomes, dendrimers, liposomes and carbon nanotubes. The review also focuses on the effective utilization of receptor-based targeted delivery systems for the management of cancer in effective ways by minimizing the drug-associated side effects and improving the therapeutic efficacy of developed nano-architectures.


Asunto(s)
Sistemas de Liberación de Medicamentos , Neoplasias/tratamiento farmacológico , Receptores de Superficie Celular/efectos de los fármacos , Animales , Receptores ErbB/efectos de los fármacos , Humanos , Receptores de Hialuranos/efectos de los fármacos , Nanopartículas , Receptores de Leptina/efectos de los fármacos , Receptores de Transferrina/efectos de los fármacos , Receptores del Factor de Necrosis Tumoral/efectos de los fármacos
10.
Curr Opin Support Palliat Care ; 7(1): 54-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23314015

RESUMEN

PURPOSE OF REVIEW: Cancer patients undergoing treatment with cytotoxic chemotherapeutic agents (CCAs) often experience a cluster of treatment-related symptoms, which include fatigue, loss of appetite, disturbed sleep, depressed mood, cognitive difficulties, and changes in body composition. This symptom cluster collectively referred to herein as cancer treatment-related symptoms (CTRSs) decrease quality of life, and physical and social functioning. The preclinical and clinical studies described in this review represent important progress in understanding potential underlying mechanisms of CTRS. RECENT FINDINGS: Recent studies support a role for CCA-induced interleukin-1ß (IL-1ß) signaling in the cause of CTRS. CCAs may share a common ability to activate intracellular stress response pathways to trigger the synthesis, processing, and release of IL-1ß from immune cells. Fatigue, sleep disturbance, and cognitive difficulties in cancer patients exposed to CCAs correlate with plasma levels of IL-6, IL-1 receptor antagonist, and soluble tumor necrosis factor receptor-I/II, surrogate markers of IL-1ß-mediated central nervous system (CNS) inflammation. Additional preclinical work suggests IL-1ß-mediated CNS inflammation may cause CTRS by altering hypothalamic and hippocampal functioning. SUMMARY: Although additional research is necessary to further establish the link between CCA exposure, IL-1ß-mediated inflammatory processes and CTRS, these data provide hints for future studies and therapeutic approaches in ameliorating these symptoms in cancer patients.


Asunto(s)
Antineoplásicos/efectos adversos , Conducta de Enfermedad/efectos de los fármacos , Inflamación/inducido químicamente , Interleucina-1beta/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Calidad de Vida , Transducción de Señal/efectos de los fármacos , Antineoplásicos/uso terapéutico , Trastornos del Conocimiento/inducido químicamente , Trastornos del Conocimiento/complicaciones , Fatiga/inducido químicamente , Fatiga/complicaciones , Humanos , Hipotálamo/efectos de los fármacos , Hipotálamo/fisiología , Inflamación/sangre , Inflamación/complicaciones , Interleucina-1beta/sangre , Interleucina-1beta/fisiología , Interleucina-6/sangre , Interleucina-6/fisiología , Trastornos del Humor/inducido químicamente , Neoplasias/complicaciones , Receptores de Interleucina/sangre , Receptores de Interleucina/efectos de los fármacos , Receptores de Interleucina/fisiología , Receptores del Factor de Necrosis Tumoral/sangre , Receptores del Factor de Necrosis Tumoral/efectos de los fármacos , Receptores del Factor de Necrosis Tumoral/fisiología , Perfil de Impacto de Enfermedad , Síndrome
11.
Future Med Chem ; 5(1): 69-79, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23256814

RESUMEN

The overexpression of TNF has been implicated in a variety of disease conditions including rheumatoid arthritis, Crohn's disease, HIV and cancer. It is presently a therapeutic target for inflammatory diseases. Many of the therapeutics currently used are biologics designed to sequester TNF, preventing it from binding with its receptors. Recent research has been focused on finding small molecules that alter the production of TNF, modulate its signal transduction pathways, or directly inhibit the binding to its receptors. Modulation of a protein-protein interaction with small molecules is an interesting and nontrivial approach. The various strategies used in obtaining small-molecule, nonpeptide, inhibitors of the TNF-TNF receptor interaction through disruption of the TNF trimer or direct inhibition of the TNF-TNF receptor interaction are presented here.


Asunto(s)
Receptores del Factor de Necrosis Tumoral/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Humanos , Modelos Moleculares , Unión Proteica , Receptores del Factor de Necrosis Tumoral/química , Receptores del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/química , Factor de Necrosis Tumoral alfa/metabolismo
12.
J Immunol ; 188(7): 3169-78, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22379032

RESUMEN

Increased rates of tuberculosis (TB) reactivation have been reported in humans treated with TNF-α (TNF)-neutralizing drugs, and higher rates are observed with anti-TNF Abs (e.g., infliximab) as compared with TNF receptor fusion protein (etanercept). Mechanisms driving differential reactivation rates and differences in drug action are not known. We use a computational model of a TB granuloma formation that includes TNF/TNF receptor dynamics to elucidate these mechanisms. Our analyses yield three important insights. First, drug binding to membrane-bound TNF critically impairs granuloma function. Second, a higher risk of reactivation induced from Ab-type treatments is primarily due to differences in TNF/drug binding kinetics and permeability. Apoptotic and cytolytic activities of Abs and pharmacokinetic fluctuations in blood concentration of drug are not essential to inducing TB reactivation. Third, we predict specific host factors that, if augmented, would improve granuloma function during anti-TNF therapy. Our findings have implications for the development of safer anti-TNF drugs to treat inflammatory diseases.


Asunto(s)
Anticuerpos Monoclonales/efectos adversos , Antirreumáticos/efectos adversos , Simulación por Computador , Tuberculosis Latente/fisiopatología , Modelos Biológicos , Mycobacterium tuberculosis/crecimiento & desarrollo , Receptores del Factor de Necrosis Tumoral/efectos de los fármacos , Tuberculoma/fisiopatología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Adalimumab , Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/sangre , Anticuerpos Monoclonales Humanizados/farmacocinética , Antirreumáticos/sangre , Antirreumáticos/clasificación , Antirreumáticos/farmacocinética , Apoptosis/efectos de los fármacos , Certolizumab Pegol , Citotoxicidad Inmunológica , Etanercept , Humanos , Fragmentos Fab de Inmunoglobulinas/efectos adversos , Fragmentos Fab de Inmunoglobulinas/sangre , Inmunoglobulina G/efectos adversos , Inmunoglobulina G/sangre , Infliximab , Tuberculosis Latente/inmunología , Mycobacterium tuberculosis/inmunología , Permeabilidad , Polietilenglicoles/efectos adversos , Polietilenglicoles/farmacocinética , Unión Proteica , Receptores del Factor de Necrosis Tumoral/sangre , Receptores del Factor de Necrosis Tumoral/fisiología , Riesgo , Tuberculoma/inmunología , Tuberculoma/microbiología , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/fisiopatología , Factor de Necrosis Tumoral alfa/fisiología
13.
J Nat Prod ; 74(9): 1916-21, 2011 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-21899269

RESUMEN

In this study, the protective effects of luteolin (1, a major component of Cirsium japonicum) were examined against d-galactosamine (GalN)/lipopolysaccharide (LPS)-induced fulminant hepatic failure. Mice received an intraperitoneal injection of 1 (25, 50, 100, and 200 mg·kg(-1)) 1 h before treatment with GalN (700 mg·kg(-1))/LPS (10 µg·kg(-1)). Treatment with GalN/LPS resulted in increased mortality and serum aminotransferase activity. These increases were attenuated by pretreatment with 1. Treatment with GalN/LPS induced an increase in the serum level of tumor necrosis factor-α (TNF-α) and protein expression of TNF-α receptor-associated death domain, and these increases were prevented by 1. In addition, 1 attenuated apoptosis induced by GalN/LPS treatment, which was analyzed using a caspase-3 and -8 activity assay, as well as by proapoptotic BH3-only protein and cytochrome c protein expression, and by a terminal deoxynuleotidyl transferase-mediated dUTP nick end-labeling method. After GalN/LPS injection, nuclear phosphorylated c-Jun levels showed a significant increase, which were attenuated by 1. The present findings suggest that luteolin ameliorates D-GalN/LPS-induced liver injury and that this protection is likely due to inhibition of the extrinsic and intrinsic apoptotic pathways.


Asunto(s)
Cirsium/química , Galactosamina/farmacología , Luteolina/farmacología , Animales , Apoptosis/efectos de los fármacos , Inyecciones Intraperitoneales , Hígado/efectos de los fármacos , Fallo Hepático Agudo/inducido químicamente , Fallo Hepático Agudo/tratamiento farmacológico , Luteolina/química , Masculino , Ratones , Ratones Endogámicos ICR , Estructura Molecular , Receptores del Factor de Necrosis Tumoral/efectos de los fármacos , Transaminasas/análisis , Transaminasas/sangre , Transaminasas/metabolismo , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/sangre
14.
Nat Rev Gastroenterol Hepatol ; 8(9): 491-501, 2011 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-21826088

RESUMEN

Alcoholic liver disease (ALD) is a major cause of morbidity and mortality worldwide. The spectrum of disease ranges from fatty liver to hepatic inflammation, necrosis, progressive fibrosis and hepatocellular carcinoma. In developed countries, ALD is a major cause of end-stage liver disease that requires transplantation. The most effective therapy for ALD is alcohol abstinence. However, for patients with severe forms of ALD (that is, alcoholic hepatitis) and for those who do not achieve abstinence from alcohol, targeted therapies are urgently needed. The development of new drugs for ALD is hampered by the scarcity of studies and the drawbacks of existing animal models, which do not reflect all the features of the human disease. However, translational research using liver samples from patients with ALD has identified new potential therapeutic targets, such as CXC chemokines, osteopontin and tumor necrosis factor receptor superfamily member 12A. The pathogenetic roles of these targets, however, remain to be confirmed in animal models. This Review summarizes the epidemiology, natural history, risk factors and current knowledge of the pathogenetic mechanisms of ALD. In addition, this article provides a detailed description of the findings of these translational studies and of the animal models used to study ALD.


Asunto(s)
Hepatopatías Alcohólicas/tratamiento farmacológico , Hepatopatías Alcohólicas/etiología , Animales , Quimiocinas CXC/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Hepatopatías Alcohólicas/epidemiología , Osteopontina/efectos de los fármacos , Prevalencia , Receptores del Factor de Necrosis Tumoral/efectos de los fármacos
16.
Brain Behav Immun ; 25(6): 1063-77, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21435392

RESUMEN

In the periphery, exercise induces interleukin (IL)-6 to downregulate tumor necrosis factor (TNF), elevate interleukin-1 receptor antagonist (IL-1RA), decreasing inflammation. Exercise also offers neuroprotection and facilitates brain repair. IL-6 production in the hippocampus following exercise suggests the potential of a similar protective role as in the periphery to down-regulate TNFα and inflammation. Using a chemical-induced model of hippocampal dentate granule cell death (trimethyltin, TMT 2.4 mg/kg, ip) dependent upon TNF receptor signaling, we demonstrate neuroprotection in mice with 2 weeks access to running wheel. Exercise attenuated neuronal death and diminished elevations in TNFα, TNF receptor 1, myeloid differentiation primary response gene (MyD) 88, transforming growth factor ß, chemokine (C-C motif) ligand 2 (CCL2), and CCL3. Elevated mRNA levels for IL-1α, IL-1RA, occurred with injury and protection. mRNA and protein levels of IL-6 and neuronal expression of IL-6 receptor α, were elevated with injury and protection. Microarray pathway analysis supported an up-regulation of TNFα cell death signaling pathways with TMT and inhibition by exercise. IL-6 pathway recruitment occurred in both conditions. IL-6 downstream signal events differed in the level of STAT3 activation. Exercise did not increase mRNA levels of brain derived neurotrophic factor, nerve growth factor, or glial derived neurotrophic factor. In IL-6 deficient mice, exercise did not attenuate TMT-induced tremor and a diminished level of neuroprotection was observed. These data suggest a contributory role for IL-6 induced by exercise for neuroprotection in the CNS similar to that seen in the periphery.


Asunto(s)
Hipocampo/efectos de los fármacos , Interleucina-6/fisiología , Neuronas/efectos de los fármacos , Neurotoxinas/toxicidad , Condicionamiento Físico Animal/fisiología , Receptores del Factor de Necrosis Tumoral/efectos de los fármacos , Compuestos de Trimetilestaño/toxicidad , Animales , Quimiocinas/biosíntesis , Quimiocinas/genética , Epilepsia Tónico-Clónica/inducido químicamente , Hipocampo/metabolismo , Hipocampo/patología , Interleucina-6/biosíntesis , Interleucina-6/deficiencia , Interleucina-6/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Neurológicos , Factor 88 de Diferenciación Mieloide/biosíntesis , Factor 88 de Diferenciación Mieloide/genética , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Neuronas/patología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Quimera por Radiación , Distribución Aleatoria , Receptores del Factor de Necrosis Tumoral/biosíntesis , Receptores del Factor de Necrosis Tumoral/fisiología , Organismos Libres de Patógenos Específicos , Temblor/inducido químicamente , Factor de Necrosis Tumoral alfa/fisiología , Regulación hacia Arriba
17.
Expert Opin Biol Ther ; 10(11): 1555-61, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20919800

RESUMEN

IMPORTANCE OF THE FIELD: The use of biologics as immune modulators in several autoimmune diseases has provided new tools to the physician's therapeutic armamentiarium and has led to improved patients' outcomes and quality of life. By producing autoantibodies, B cells in systemic lupus erythematosus (SLE) are key players in the pathogenesis of the disease and in its clinical manifestations. Therefore, biologics that target B cells in SLE aim at reducing the activity of these cells for the induction of remissions and/or amelioration of disease activity, reduction of organ involvement, and limitation of the complications and side effects caused by immunosuppressive therapies. AREAS COVERED IN THIS REVIEW: This review describes the past and current clinical trials with B-cell-targeted biologics in SLE, to provide a historical perspective and the state-of-the-art on the topic. WHAT THE READER WILL GAIN: We review how the disappointment in the field from promising agents has been instrumental in providing valuable lessons leading to an improved design of new trials that are now giving encouraging results. TAKE HOME MESSAGE: In systemic lupus erythematosus (SLE), the use of B-cell-based biologics in clinical trials has shown both disappointment and promise.


Asunto(s)
Linfocitos B/inmunología , Productos Biológicos/uso terapéutico , Factores Inmunológicos/uso terapéutico , Lupus Eritematoso Sistémico/terapia , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antígenos CD20/inmunología , Factor Activador de Células B/inmunología , Factor Activador de Células B/fisiología , Linfocitos B/efectos de los fármacos , Productos Biológicos/farmacología , Ensayos Clínicos como Asunto/estadística & datos numéricos , Humanos , Factores Inmunológicos/farmacología , Terapia de Inmunosupresión , Lupus Eritematoso Sistémico/inmunología , Estudios Multicéntricos como Asunto/estadística & datos numéricos , Receptores del Factor de Necrosis Tumoral/efectos de los fármacos , Receptores del Factor de Necrosis Tumoral/inmunología , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/uso terapéutico , Lectina 2 Similar a Ig de Unión al Ácido Siálico/inmunología
18.
J Neurochem ; 113(3): 692-703, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20132465

RESUMEN

The alpha-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate receptor (AMPAR) subunit GluR2, which regulates excitotoxicity and the inflammatory cytokine tumour necrosis factor alpha (TNFalpha) have both been implicated in motor neurone vulnerability in amyotrophic lateral sclerosis/motor neurone disease. TNFalpha has been reported to increase cell surface expression of AMPAR subunits to increase synaptic strength and enhance excitotoxicity, but whether this mechanism occurs in motor neurones is unknown. We used primary cultures of mouse motor neurones and cortical neurones to examine the interaction between TNFalpha receptor activation, GluR2 availability, AMPAR-mediated calcium entry and susceptibility to excitotoxicity. Short exposure to a physiologically relevant concentration of TNFalpha (10 ng/mL, 15 min) caused a marked redistribution of both GluR1 and GluR2 to the cell surface as determined by cell surface biotinylation and immunofluorescence. Using fura-2-acetoxymethyl ester microfluorimetry, we showed that exposure to TNFalpha caused a rapid reduction in the peak amplitude of AMPA-mediated calcium entry in a PI3-kinase and p38 kinase-dependent manner, consistent with increased insertion of GluR2-containing AMPAR into the plasma membrane. This resulted in a protection of motor neurones against kainate-induced cell death. Our data therefore, suggest that TNFalpha acts primarily as a physiological regulator of synaptic activity in motor neurones rather than a pathological drive in amyotrophic lateral sclerosis.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Calcio/metabolismo , Neuronas Motoras/efectos de los fármacos , Degeneración Nerviosa/patología , Fármacos Neuroprotectores , Receptores AMPA/antagonistas & inhibidores , Receptores de Superficie Celular/biosíntesis , Factor de Necrosis Tumoral alfa/farmacología , Animales , Biotinilación , Western Blotting , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Agonistas de Aminoácidos Excitadores/toxicidad , Femenino , Técnica del Anticuerpo Fluorescente , Ácido Kaínico/antagonistas & inhibidores , Ácido Kaínico/toxicidad , Ratones , Degeneración Nerviosa/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Embarazo , Receptores AMPA/biosíntesis , Receptores AMPA/genética , Receptores del Factor de Necrosis Tumoral/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrometría de Fluorescencia , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
Cancer Lett ; 291(2): 225-9, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-19900759

RESUMEN

Extracellular adenosine-induced apoptosis of HepG2 cells, a human hepatoma cell line, in a concentration (0.1-20mM)- and treatment (24-72h)-dependent manner by activating caspase-3, -8, and -9. In the gene expression assay using a DNA microalley, adenosine upregulated mRNAs for tumor necrosis factor (TNF), TNF receptor 1-associated death domain protein (TRADD), TNF related apoptosis-inducing ligand receptor 2 (TRAIL-R2), TRADD/receptor-interacting protein kinase 1 (RIPK1), Fas-associated death domain protein (FADD), and caspase-9, involving activation of caspase-8 and -9 followed by the effector caspase-3. The results of the present study suggest that adenosine induces HepG2 cell apoptosis by activating those caspases as a result from tuning apoptosis-mediator gene transcription.


Asunto(s)
Adenosina/farmacología , Apoptosis/fisiología , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Transcripción Genética , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/patología , Caspasa 9/efectos de los fármacos , Caspasa 9/metabolismo , Caspasas/efectos de los fármacos , Caspasas/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Activación Enzimática , Regulación Neoplásica de la Expresión Génica , Humanos , Cinética , Neoplasias Hepáticas/patología , ARN Mensajero/genética , ARN Neoplásico/genética , Receptores del Factor de Necrosis Tumoral/efectos de los fármacos , Receptores del Factor de Necrosis Tumoral/fisiología , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba
20.
J Neuroimmunol ; 218(1-2): 48-56, 2010 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-19913923

RESUMEN

Opioid receptor like-1 receptor (ORL(1)) is selective for orphaninFQ/nociceptin (OFQ/N), a peptide linked to stress. Since immunologic stimuli exert stressor-like effects, the neuroendocrine and behavioral effects of the T-cell superantigen staphylococcal enterotoxin A (SEA) were tested in ORL(1)(-/-) and ORL(1)(+/+) wildtype 129S6 mice. Within 2h of SEA challenge both genotypes showed elevated corticosterone, but only wildtypes were elevated after 4h, and had altered hypothalamic CRH mRNA. Although amygdaloid CRH and TNFalpha mRNA was increased by SEA, this did not vary with genotype. Interestingly, gustatory neophobia due to SEA challenge was augmented in ORL(1)(-/-) mice, although object neophobia tested 4days later was abrogated. These results suggest differential requirements for ORL(1) in the mediation of neuroimmune effects exerted at different times after an immune challenge.


Asunto(s)
Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Enterotoxinas/farmacología , Neuroinmunomodulación/efectos de los fármacos , Receptores Opioides/fisiología , Animales , Encéfalo/inmunología , Encéfalo/metabolismo , Corticosterona/sangre , Hormona Liberadora de Corticotropina/biosíntesis , Hormona Liberadora de Corticotropina/efectos de los fármacos , Citocinas/biosíntesis , Citocinas/efectos de los fármacos , Citocinas/inmunología , Ingestión de Alimentos/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/inmunología , Sistema Hipotálamo-Hipofisario/metabolismo , Masculino , Ratones , Ratones Noqueados , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/inmunología , Sistema Hipófiso-Suprarrenal/metabolismo , ARN Mensajero/análisis , Receptores del Factor de Necrosis Tumoral/biosíntesis , Receptores del Factor de Necrosis Tumoral/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/efectos de los fármacos , Bazo/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Receptor de Nociceptina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...