Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Mol Reprod Dev ; 85(1): 46-61, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29219221

RESUMEN

In vitro embryo development remains suboptimal compared to in vivo development due to the challenge from various stressors associated with in vitro culturing of oocytes. When 0.2 µM lycopene was added to oocyte in vitro maturation and embryo culture media, to assess its antioxidant effects on embryo development, we observed a significant (p < 0.05) increase in cleavage and blastocyst development rates compared to the corresponding controls (84.3 ± 0.6% vs. 73.1 ± 1.9% and 41.0 ± 1.4% vs. 33.4 ± 0.7%, respectively). Lycopene also significantly reduced (p < 0.05) intracellular reactive oxygen species concentrations in oocytes and blastocysts, whereas lipid peroxidation and mitochondrial activity increased compared to control conditions. The number of apoptotic nuclei was significantly reduced in the lycopene-treated compared to the control group (1.7 ± 0.1 vs. 4.7 ± 0.3), and the quantity of cells in the trophectoderm (207.1 ± 1.6 vs. 171.3 ± 1.0, respectively) and inner cell mass (41.9 ± 0.4 vs. 36.7 ± 0.4, respectively) was higher following treatment-although the inner cell mass-to-trophectoderm ratio was unchanged (1:3.3 vs. 1:3.4 for lycopene vs. control, respectively). Lycopene supplementation also significantly (p < 0.05) attenuated expression of IKBKB (Inhibitor of nuclear factor kappa B kinase, subunit beta) and reduced Caspase 9 and Caspase 3 protein abundance, while up-regulating GDF9 (Growth and differentiation factor 9), BMP15 (Bone morphogenetic protein 15), SOD2 (Superoxide dismutase 2), NDUFA2 (NADH dehydrogenase), ACADL (Acyl-CoA dehydrogenase, long chain), and ACSL3 (Acyl-CoA synthetase 3, long-chain membrane 3) transcription compared to control. Therefore, co-culturing with lycopene during oocyte maturation improved bovine embryo developmental potential during in vitro culture by improving embryonic resilience to stress.


Asunto(s)
Antioxidantes/farmacología , Técnicas de Cultivo de Embriones , Desarrollo Embrionario/efectos de los fármacos , Licopeno/farmacología , Oocitos/crecimiento & desarrollo , Acil-CoA Deshidrogenasa de Cadena Larga/biosíntesis , Animales , Blastocisto/citología , Proteína Morfogenética Ósea 15/biosíntesis , Caspasa 3/análisis , Caspasa 9/análisis , Bovinos , Coenzima A Ligasas/biosíntesis , Factor 9 de Diferenciación de Crecimiento/biosíntesis , Quinasa I-kappa B/biosíntesis , NADH Deshidrogenasa/biosíntesis , Superóxido Dismutasa/biosíntesis
2.
Oncol Rep ; 36(5): 2771-2776, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27667548

RESUMEN

The nuclear factor-κB (NF-κB) transcription factors control many physiological processes including inflammation, immunity, apoptosis and angiogenesis. In our search for NF-κB inhibitors from natural resources, we identified baicalein from Scutellaria baicalensis as an inhibitor of NF-κB activation. As examined by the NF-κB luciferase reporter assay, we found that baicalein suppressed TNF-α-induced NF-κB activation in a dose-dependent manner. It also inhibited TNF-α-induced nuclear translocation of p65 through inhibition of phosphorylation and degradation of IκBα. Furthermore, baicalein blocked the TNF-α-induced expression of NF-κB target genes involved in anti-apoptosis (cIAP-1, cIAP-2, FLIP and BCL-2), proliferation (COX-2, cyclin D1 and c-Myc), invasion (MMP­9), angiogenesis (VEGF) and major inflammatory cytokines (IL-8 and MCP1). The flow cytometric analysis indicated that baicalein potentiated TNF-α-induced apoptosis and induced G1 phase arrest in HeLa cells. Moreover, baicalein significantly blocked activation of p38, extracellular signal-regulated kinase 1/2 (ERK1/2). Our results imply that baicalein could be a lead compound for the modulation of inflammatory diseases as well as certain cancers in which inhibition of NF-κB activity may be desirable.


Asunto(s)
Flavanonas/administración & dosificación , Extractos Vegetales/administración & dosificación , Factor de Transcripción ReIA/biosíntesis , Factor de Necrosis Tumoral alfa/genética , Apoptosis/genética , Proliferación Celular/efectos de los fármacos , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Quinasa I-kappa B/biosíntesis , Quinasa I-kappa B/genética , FN-kappa B/biosíntesis , FN-kappa B/genética , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Fosforilación , Scutellaria baicalensis , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/genética , Proteínas Quinasas p38 Activadas por Mitógenos/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/genética
3.
Toxicol Appl Pharmacol ; 287(2): 119-127, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25981167

RESUMEN

As the importance of allergic disorders such as atopic dermatitis and allergic asthma, research on potential drug candidates becomes more necessary. Mast cells play an important role as initiators of allergic responses through the release of histamine; therefore, they should be the target of pharmaceutical development for the management of allergic inflammation. In our previous study, anti-allergic effect of extracts of Amomum xanthioides was demonstrated. To further investigate improved candidates, 1,2,4,5-tetramethoxybenzene (TMB) was isolated from methanol extracts of A. xanthioides. TMB dose-dependently attenuated the degranulation of mast cells without cytotoxicity by inhibiting calcium influx. TMB decreased the expression of pro-inflammatory cytokines such as tumor necrosis factor-α and interleukin (IL)-4 at both the transcriptional and translational levels. Increased expression of these cytokines was caused by translocation of nuclear factor-κB into the nucleus, and it was hindered by suppressing activation of IκB kinase complex. To confirm the effect of TMB in vivo, the ovalbumin (OVA)-induced active systemic anaphylaxis (ASA) and IgE-mediated passive cutaneous anaphylaxis (PCA) models were used. In the ASA model, hypothermia was decreased by oral administration of TMB, which attenuated serum histamine, OVA-specific IgE, and IL-4 levels. Increased pigmentation of Evans blue was reduced by TMB in a dose-dependent manner in the PCA model. Our results suggest that TMB is a possible therapeutic candidate for allergic inflammatory diseases that acts through the inhibition of mast cell degranulation and expression of pro-inflammatory cytokines.


Asunto(s)
Anisoles/farmacología , Inflamación/tratamiento farmacológico , Inflamación/fisiopatología , Amomum , Animales , Degranulación de la Célula/efectos de los fármacos , Citocinas/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Hipersensibilidad , Quinasa I-kappa B/biosíntesis , Mediadores de Inflamación/antagonistas & inhibidores , Masculino , Mastocitos/efectos de los fármacos , Ratones , Extractos Vegetales/farmacología , Ratas , Ratas Sprague-Dawley
4.
Oncol Rep ; 33(6): 2940-6, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25845556

RESUMEN

Tetraarsenic hexoxide (As4O6) has been used in Korean traditional medicine for the treatment of cancer since the late 1980's, and arsenic trioxide (As2O3) is currently used as a chemotherapeutic agent. Previous studies suggest that the As4O6-induced cell death pathway is different from that of As2O3 and its mechanism of anticancer activity remains unclear. Nuclear factor (NF)-κB is a well-known transcription factor involved in cell proliferation, invasion and metastasis. Hence, in the present study, we investigated the effects of As4O6 on NF-κB activity and NF-κB-regulated gene expression in vitro and in vivo. The cytotoxicity assay revealed that As4O6 inhibited the growth of SW620 cells in a dose-dependent manner, and the half maximal inhibitory concentration (IC50) was ~1 µM after a 48 h treatment. As4O6 suppressed NF-κB activation and suppressed inhibitory κBα (IκBα) phosphorylation stimulated by tumor necrosis factor (TNF). As4O6 also suppressed downstream NF-κB-regulated proteins involved in cancer anti-apoptosis, proliferation, invasion and metastasis. In addition, As4O6 marginally suppressed tumor growth and the anti-NF-κB activity was confirmed using an in vivo xenograft mouse model in which animals were injected with SW620 cells. The present study provides evidence that As4O6 has anticancer properties through suppression of NF-κB activity and NF-κB-mediated cellular responses.


Asunto(s)
Antineoplásicos/administración & dosificación , Arsenicales/administración & dosificación , Neoplasias del Colon/tratamiento farmacológico , FN-kappa B/biosíntesis , Óxidos/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Trióxido de Arsénico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Quinasa I-kappa B/biosíntesis , Ratones , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , Fosforilación , Factor de Necrosis Tumoral alfa/biosíntesis
5.
Pancreas ; 43(8): 1291-8, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25036911

RESUMEN

OBJECTIVES: This study aimed to investigate the influence of rosiglitazone on hepatic insulin resistance and the expressions of IκB kinase-ß (IKK-ß)/nuclear factor-κB (NF-κB) in chronic pancreatitis (CP). METHODS: After CP was induced in rats, rosiglitazone and GW9662 were administered at the doses of 4 and 2 mg/kg per day for 4 weeks, respectively. Then, glucose and insulin tolerance tests were performed. Hepatocytes were isolated for the glucose release experiments. Determination of the IKK-ß, NF-κB, and Ser307p-insulin receptor substrates-1 (Ser307p-IRS-1) expression in the liver was performed. RESULTS: The increased plasma glucose, reduced insulin sensitivity, and the capacity of insulin to suppress glucose release in hepatocytes were observed in CP rats. The IKK-ß, NF-κB, and Ser307p-IRS-1 expressions were significantly higher in the liver of CP rats than in sham-operated rats (P < 0.05). Rosiglitazone treatment significantly improved hepatic insulin sensitivity and inhibited the IKK-ß, NF-κB, and Ser307p-IRS-1 expressions in the liver (P < 0.05). Counteraction with peroxisome proliferator-activated receptor-γ by GW9662 attenuated the aforementioned effects of rosiglitazone. CONCLUSIONS: Rosiglitazone attenuates hepatic insulin resistance induced by CP. The inhibition of hepatic IKK-ß and NF-κB expressions via peroxisome proliferator-activated receptor-γ may be involved in the therapeutic effect of rosiglitazone.


Asunto(s)
Resistencia a la Insulina , Hígado/efectos de los fármacos , Pancreatitis Crónica/tratamiento farmacológico , Tiazolidinedionas/uso terapéutico , Anilidas/farmacología , Anilidas/uso terapéutico , Animales , Evaluación Preclínica de Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Quinasa I-kappa B/biosíntesis , Quinasa I-kappa B/genética , Insulina/sangre , Proteínas Sustrato del Receptor de Insulina/biosíntesis , Proteínas Sustrato del Receptor de Insulina/genética , Hígado/metabolismo , Masculino , PPAR gamma/agonistas , Pancreatitis Crónica/metabolismo , Ratas , Ratas Wistar , Rosiglitazona , Tiazolidinedionas/farmacología , Factor de Transcripción ReIA/biosíntesis , Factor de Transcripción ReIA/genética
6.
Cancer Sci ; 105(4): 473-80, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24450414

RESUMEN

Constitutive activation of the signal transducer and activator of transcription 3 (STAT3) or the nuclear factor-κB (NF-κB) pathway occurs frequently in cancer cells and contributes to oncogenesis. The activation of Janus kinase 2 (JAK2) and IκB kinase (IKK) are key events in STAT3 and NF-κB signaling, respectively. We have identified 2-methoxystypandrone (2-MS) from a traditional Chinese medicinal herb Polygonum cuspidatum as a novel dual inhibitor of JAK2 and IKK. 2-MS inhibits both interleukin-6-induced and constitutively-activated STAT3, as well as tumor necrosis factor-α-induced NF-κB activation. 2-MS specifically inhibits JAK and IKKß kinase activities but has little effect on activities of other kinases tested. The inhibitory effects of 2-MS on STAT3 and NF-κB signaling can be eliminated by DTT or glutathione and can last for 4 h after a pulse treatment. Furthermore, 2-MS inhibits growth and induces death of tumor cells, particularly those with constitutively-activated STAT3 or NF-κB signaling. We propose that the natural compound 2-MS, as a potent dual inhibitor of STAT3 and NF-κB pathways, is a promising anticancer drug candidate.


Asunto(s)
Quinasa I-kappa B/biosíntesis , Janus Quinasa 2/biosíntesis , FN-kappa B/genética , Naftoquinonas/administración & dosificación , Factor de Transcripción STAT3/biosíntesis , Apoptosis/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Quinasa I-kappa B/genética , Interleucina-6/biosíntesis , Janus Quinasa 2/genética , Medicina Tradicional China , Fosforilación/efectos de los fármacos , Factor de Transcripción STAT3/genética , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/biosíntesis
7.
Acta Haematol ; 131(2): 102-11, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24157922

RESUMEN

Celastrol is an active compound extracted from the root bark of the traditional Chinese medicine Tripterygium wilfordii Hook F. In this study, we investigated the effect of celastrol on lipopolysaccharide (LPS)-activated LP-1 human multiple myeloma cell-induced angiogenesis, and identified its molecular mechanism of action. Migration of human umbilical vein endothelial cells (HUVECs) was tested using a wound-healing assay. HUVEC invasion was assayed using a Transwell chamber. Cell surface expression of Toll-like receptor 4 (TLR4) was analyzed by flow cytometry. Angiogenic factor vascular endothelial growth factor (VEGF) level was quantified by LUMINEX and protein expression was analyzed by Western blot. Translocation of nuclear factor-kappa B (NF-κB) was observed by fluorescence microscopy. Celastrol inhibited LPS-stimulated LP-1 human multiple myeloma-induced HUVEC migration and invasion in a concentration-dependent manner. Wound diameters increased by 72.9, 165.4 and 246.2% at 0.025, 0.05 and 0.1 µM, respectively, compared to LPS alone. A 45-74% inhibition of LPS-dependent cell invasion was achieved in the presence of 0.025-0.1 µM celastrol. Celastrol significantly downregulated LPS-induced TLR4 expression and inhibited LPS-induced VEGF secretion in LP-1 cells. VEGF levels decreased by 64.8, 84.4 and 92.9% after coexposure to celastrol at 0.025, 0.05 and 0.1 µM, respectively, compared to LPS alone. Celastrol also inhibited the IκB kinase (IKK)/NF-κB pathway induced by LPS. Protein levels of NF-κB p65, IKKα and IκB-α decreased in a dose-dependent manner after coexposure to celastrol. Celastrol also blocked nuclear translocation of the p65 subunit. These results suggest that celastrol inhibits LPS-induced angiogenesis by suppressing TLR4-triggered NF-κB activation.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , FN-kappa B/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Receptor Toll-Like 4/antagonistas & inhibidores , Triterpenos/farmacología , Transporte Activo de Núcleo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Regulación hacia Abajo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Quinasa I-kappa B/antagonistas & inhibidores , Quinasa I-kappa B/biosíntesis , Quinasa I-kappa B/genética , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Lipopolisacáridos/toxicidad , Estructura Molecular , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Inhibidor NF-kappaB alfa , Triterpenos Pentacíclicos , Receptor Toll-Like 4/biosíntesis , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/fisiología , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Transcripción Genética/efectos de los fármacos , Triterpenos/química , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas
8.
Inflammation ; 35(2): 560-5, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21603970

RESUMEN

Parthenolide, a naturally occurring sesquiterpene lactone derived from feverfew (Tanacetum parthenium), exhibits exceptional anti-cancer and anti-inflammatory properties, making it a prominent candidate for further studies and drug development. In this review, we briefly investigate molecular events and cell-specific activities of this chemical in relation to cytochrome c, nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB), signal transduction and activation of transcription (STAT), reactive oxygen species (ROS), TCP, HDACs, microtubules, and inflammasomes. This paper reports that parthenolide shows strong NF-κB- and STAT-inhibition-mediated transcriptional suppression of pro-apoptotic genes. This compound acts both at the transcriptional level and by direct inhibition of associated kinases (IKK-ß). Similarly, this review discusses parthenolide-induced ROS-mediated apoptosis of tumor cells via the intrinsic apoptotic signaling pathway. The unique ability of this compound to not harm normal cells but at the same time induce sensitization to extrinsic as well as intrinsic apoptosis signaling in cancer cells provides an important, novel therapeutic strategy for treatment of cancer and inflammation-related disorders.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Antineoplásicos/farmacología , Inflamación/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Sesquiterpenos/farmacología , Apoptosis/efectos de los fármacos , Citocromos c/metabolismo , Humanos , Quinasa I-kappa B/antagonistas & inhibidores , Quinasa I-kappa B/biosíntesis , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Extractos Vegetales/farmacología , Especies Reactivas de Oxígeno , Factores de Transcripción STAT/antagonistas & inhibidores , Factores de Transcripción STAT/metabolismo , Sesquiterpenos/uso terapéutico , Transducción de Señal/efectos de los fármacos , Tanacetum parthenium/química , Tanacetum parthenium/metabolismo , Transcripción Genética/efectos de los fármacos
9.
J Gerontol A Biol Sci Med Sci ; 67(1): 3-10, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22016358

RESUMEN

We have investigated whether the quality of dietary fat and supplementation with coenzyme Q(10) (CoQ) modifies expression of genes related with inflammatory response and endoplasmic reticulum stress in elderly persons. Twenty participants received three diets for 4 weeks each: Mediterranean diet + CoQ (Med + CoQ), Mediterranean diet (Med), and saturated fatty acid-rich diet (SFA). After 12-hour fast, volunteers consumed a breakfast with a fat composition similar to that consumed in each of the diets. Med and Med + CoQ diets produced a lower fasting calreticulin, IL-1b, and JNK-1 gene expression; a lower postprandial p65, IKK-b, MMP-9, IL-1b, JNK-1, sXBP-1, and BiP/Grp78 gene expression; and a higher postprandial IkB-a gene expression compared with the SFA diet. Med + CoQ diet produced a lower postprandial decrease p65 and IKK-b gene expression compared with the other diets. Our results support the anti-inflammatory effect of Med diet and that exogenous CoQ supplementation in synergy with a Med diet modulates the inflammatory response and endoplasmic reticulum stress.


Asunto(s)
Dieta Mediterránea , Suplementos Dietéticos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Inflamación/metabolismo , Ubiquinona/análogos & derivados , Anciano , Índice de Masa Corporal , Calreticulina/biosíntesis , Calreticulina/genética , Estudios Cruzados , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Grasas de la Dieta/metabolismo , Chaperón BiP del Retículo Endoplásmico , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de Choque Térmico/biosíntesis , Proteínas de Choque Térmico/genética , Humanos , Quinasa I-kappa B/biosíntesis , Quinasa I-kappa B/genética , Inflamación/tratamiento farmacológico , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/genética , Proteína Quinasa 8 Activada por Mitógenos/biosíntesis , Proteína Quinasa 8 Activada por Mitógenos/genética , Factores de Transcripción del Factor Regulador X , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Ubiquinona/administración & dosificación , Ubiquinona/sangre , Ubiquinona/metabolismo
10.
Am J Physiol Endocrinol Metab ; 298(6): E1122-30, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20354158

RESUMEN

Hypothalamic inflammation induced by high-fat feeding causes insulin and leptin resistance and contributes to the pathogenesis of obesity. Since in vitro exposure to saturated fatty acids causes inflammation and insulin resistance in many cultured cell types, we determined how cultured hypothalamic neurons respond to this stimulus. Two murine hypothalamic neuronal cell cultures, N43/5 and GT1-7, were exposed to escalating concentrations of saturated fatty acids for up to 24 h. Harvested cells were evaluated for activation of inflammation by gene expression and protein content. Insulin-treated cells were evaluated for induction of markers of insulin receptor signaling (p-IRS, p-Akt). In both hypothalamic cell lines, inflammation was induced by prototypical inflammatory mediators LPS and TNFalpha, as judged by induction of IkappaBalpha (3- to 5-fold) and IL-6 (3- to 7-fold) mRNA and p-IkappaBalpha protein, and TNFalpha pretreatment reduced insulin-mediated p-Akt activation by 30% (P < 0.05). By comparison, neither mixed saturated fatty acid (100, 250, or 500 microM for

Asunto(s)
Ácidos Grasos/administración & dosificación , Hipotálamo/metabolismo , Resistencia a la Insulina/inmunología , Obesidad/metabolismo , Animales , Western Blotting , Línea Celular , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Perfilación de la Expresión Génica/métodos , Hipotálamo/patología , Quinasa I-kappa B/biosíntesis , Quinasa I-kappa B/genética , Inflamación/metabolismo , Inflamación/patología , Insulina/metabolismo , Interleucina-6/biosíntesis , Interleucina-6/genética , Ratones , Obesidad/patología , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Proteínas Proto-Oncogénicas c-akt/genética , ARN Mensajero/química , ARN Mensajero/genética , Receptor de Insulina/metabolismo
11.
Int J Hyperthermia ; 21(7): 603-14, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16304713

RESUMEN

AIM: The aim of the present study was to explore whether heme oxygenase-1 (HO-1) is involved in the hyperthermia-provided protection of the small intestine from ischemia/reperfusion injury in rats. METHODS: Intestinal damage was induced in male Sprague-Dawley rats by clamping both the superior mesenteric artery and the celiac trunk for 30 min, followed by reperfusion. Whole-body hyperthermia was induced in anesthetized rats by placement in a temperature-controlled water bath. Whole-body hyperthermia to a core temperature of 42-43 degrees C for 15 min was followed by passive cooling. We started the hyperthermic treatment 6 h before the vascular clamping. The severity of the mucosal injury was evaluated by several biochemical markers and histological findings. Hyperthermia-induced heat-shock proteins were detected by Western blotting. We also investigated the effect of zinc protoporphyrin IX (an HO-1 inhibitor) on the protective effect of hyperthermia. RESULTS: The rats, which were killed after ischemia/reperfusion, had severe intestinal inflammation. Hyperthermia significantly induced the production of Hsp70 and HO-1 in intestinal mucosa and significantly reduced ischemia/reperfusion-induced mucosal injury. The combination of zinc protoporphyrin IX with hyperthermia extinguished the protective effects of hyperthermia on ischemia/reperfusion injury. CONCLUSION: Hyperthermia protects against ischemia/reperfusion injury in rat small intestine through the expression of heat-shock proteins, especially HO-1.


Asunto(s)
Hemo Oxigenasa (Desciclizante)/fisiología , Hipertermia Inducida , Daño por Reperfusión/prevención & control , Animales , Quimiocinas CXC/análisis , Inducción Enzimática , Proteínas HSP70 de Choque Térmico/biosíntesis , Hemo Oxigenasa (Desciclizante)/biosíntesis , Quinasa I-kappa B/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/análisis , Mucosa Intestinal/química , Mucosa Intestinal/efectos de los fármacos , Intestino Delgado/citología , Intestino Delgado/efectos de los fármacos , Masculino , Peroxidasa/metabolismo , Protoporfirinas/farmacología , Ratas , Ratas Sprague-Dawley , Sustancias Reactivas al Ácido Tiobarbitúrico/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA