Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
BMJ Open ; 14(9): e088744, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39260836

RESUMEN

INTRODUCTION: Heart failure (HF) is a complex syndrome that affects millions of people worldwide and leads to significant morbidity and mortality. Sacubitril/valsartan, a combination drug consisting of a neprilysin inhibitor and an angiotensin receptor blocker (ARB), has shown a greater improvement in the prognosis of HF than ACE inhibitors (ACEI) or ARB. Recent studies have found that ACEI/ARB or sacubitril/valsartan can increase flow-mediated dilation (FMD) and reduce pulse wave velocity (PWV), which are independent predictors of cardiovascular events and HF prognosis. The purpose of this study is to assess and compare the effect of sacubitril/valsartan and ACEI/ARB on FMD and PWV using meta-analysis and further provide a reference for the role of sacubitril/valsartan in the treatment of HF. METHODS AND ANALYSIS: Clinical randomised controlled trials investigating the effect of sacubitril/valsartan and/or ACEI/ARB on FMD and PWV in patients with HF will be searched in the relevant database, including PubMed, Web of Science, Embase, Cochrane Library and China's National Knowledge Infrastructure up to January 2024. The outcomes of interest are changes in endothelial function assessed by FMD and changes in arterial stiffness assessed by PWV. The risk of bias was evaluated using the revised Cochrane risk of bias tool for randomised trials (RoB2.0). Review Manager V.5.3 software is used for meta-analysis data synthesis, sensitivity analysis, meta-regression analysis, subgroup analysis and risk of bias assessment. The reporting bias of studies will be evaluated using the funnel plot, in which symmetry will be assessed by Begg's and Egger's tests. The evidence quality of the included studies will be evaluated by the Grading of Recommendations Assessment, Development, and Evaluation. ETHICS AND DISSEMINATION: This study only analyses research data from the published literature and therefore does not require ethical approval. We will submit the systematic review to a peer-reviewed journal. PROSPERO REGISTRATION NUMBER: CRD42024538148.


Asunto(s)
Aminobutiratos , Antagonistas de Receptores de Angiotensina , Inhibidores de la Enzima Convertidora de Angiotensina , Compuestos de Bifenilo , Combinación de Medicamentos , Insuficiencia Cardíaca , Metaanálisis como Asunto , Revisiones Sistemáticas como Asunto , Valsartán , Rigidez Vascular , Humanos , Valsartán/uso terapéutico , Aminobutiratos/uso terapéutico , Aminobutiratos/farmacología , Rigidez Vascular/efectos de los fármacos , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/fisiopatología , Antagonistas de Receptores de Angiotensina/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiopatología , Ensayos Clínicos Controlados Aleatorios como Asunto , Tetrazoles/uso terapéutico , Proyectos de Investigación , Análisis de la Onda del Pulso
2.
Arch Virol ; 168(4): 128, 2023 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-37002434

RESUMEN

Due to recurrence and resistance to chemotherapy, the current standard therapeutics are not fully effective against ovarian cancer. Therefore, we aimed to find an effective approach to improve the prognosis and therapy of ovarian cancer. NG34ScFvPD-1 is a modified oncolytic herpes simplex virus NG34 strain that expresses a single-chain antibody against programmed cell death protein 1 (PD-1) (ScFvPD-1). We assessed its efficacy and its regulatory mechanism in a mouse model of ovarian cancer. Enzyme-linked immunosorbent assay and western blot techniques were used to measure protein expression. Oncolysis caused by NG34ScFvPD-1 was examined using cytotoxicity and replication assays. The mechanism by which NG34ScFvPD-1 regulates apoptosis of ovarian cancer cells in vitro was also evaluated. We assessed the antitumor immunity and therapeutic potency of NG34ScFvPD-1 in combination with a phosphoinositide 3-kinase (PI3K) inhibitor. We found that NG34ScFvPD-1-infected ovarian cancer cells expressed and secreted ScFvPD-1, which bound mouse PD-1. The insertion of the ScFvPD-1 sequence did not inhibit the oncolytic activity of NG34ScFvPD-1, which induced apoptosis of ovarian cancer cells via the caspase-dependent pathway in vitro and activated the PI3K/AKT signaling pathway. Synergy was observed between NG34ScFvPD-1 and a PI3K inhibitor, and the combination was able to suppress tumor development, to prolong survival, and to elicit potent antitumor immunity. Thus, inhibition of PI3K enhanced the potent antitumor immunity induced by NG34ScFvPD-1 against ovarian cancer.


Asunto(s)
Herpesvirus Humano 1 , Viroterapia Oncolítica , Virus Oncolíticos , Neoplasias Ováricas , Anticuerpos de Cadena Única , Humanos , Femenino , Ratones , Animales , Fosfatidilinositol 3-Quinasas , Inhibidores de las Quinasa Fosfoinosítidos-3 , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Anticuerpos de Cadena Única/genética , Receptor de Muerte Celular Programada 1 , Neoplasias Ováricas/terapia , Neoplasias Ováricas/patología , Línea Celular Tumoral
3.
Cells ; 11(17)2022 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-36078037

RESUMEN

Atrial fibrillation (AF) is a form of sustained cardiac arrhythmia and microRNAs (miRs) play crucial roles in the pathophysiology of AF. To identify novel miR-mRNA pairs, we performed RNA-seq from atrial biopsies of persistent AF patients and non-AF patients with normal sinus rhythm (SR). Differentially expressed miRs (11 down and 9 up) and mRNAs (95 up and 82 down) were identified and hierarchically clustered in a heat map. Subsequently, GO, KEGG, and GSEA analyses were run to identify deregulated pathways. Then, miR targets were predicted in the miRDB database, and a regulatory network of negatively correlated miR-mRNA pairs was constructed using Cytoscape. To select potential candidate genes from GSEA analysis, the top-50 enriched genes in GSEA were overlaid with predicted targets of differentially deregulated miRs. Further, the protein-protein interaction (PPI) network of enriched genes in GSEA was constructed, and subsequently, GO and canonical pathway analyses were run for genes in the PPI network. Our analyses showed that TNF-α, p53, EMT, and SYDECAN1 signaling were among the highly affected pathways in AF samples. SDC-1 (SYNDECAN-1) was the top-enriched gene in p53, EMT, and SYDECAN1 signaling. Consistently, SDC-1 mRNA and protein levels were significantly higher in atrial samples of AF patients. Among negatively correlated miRs, miR-302b-3p was experimentally validated to suppress SDC-1 transcript levels. Overall, our results suggested that the miR-302b-3p/SDC-1 axis may be involved in the pathogenesis of AF.


Asunto(s)
Fibrilación Atrial , MicroARNs , Fibrilación Atrial/patología , Atrios Cardíacos/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo , ARN Mensajero/genética , Proteína p53 Supresora de Tumor
4.
Am J Respir Cell Mol Biol ; 64(6): 747-759, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33705682

RESUMEN

Smoking-mediated reprogramming of the phenotype and function of airway basal cells (BCs) disrupts airway homeostasis and is an early event in chronic obstructive pulmonary disease (COPD)-associated airway remodeling. Here, we examined the expression and regulation of the transmembrane glycoprotein TROP2 (trophoblast antigen 2), a putative stem cell marker in airway BCs, in lung tissue samples from healthy smokers and healthy nonsmokers and in models in culture to identify therapeutic targets. TROP2 expression was upregulated in the airway epithelia of smokers and positively correlated with the smoking index. In vitro, cigarette smoke extract (CSE) induced TROP2 expression in airway BCs in a time- and dose-dependent manner. The p38 MAPK and NF-κB pathways were also activated by CSE, and their specific antagonists inhibited CSE-induced TROP2 expression. A therapeutic component derived from traditional Chinese medicine, ginsenoside Rb3, inhibited CSE-induced TROP2 expression as well as activation of the p38 MAPK and NF-κB pathways in BCs in monolayer culture. Furthermore, ginsenoside Rb3 prevented the increase in TROP2 expression and antagonized CSE-induced BC hyperplasia and expression of inflammatory factors and epithelial-mesenchymal transition changes in an air-liquid culture model. Thus, CSE-induced TROP2 is a possible biomarker for early changes in the epithelium of smokers, and ginsenoside Rb3 may serve as a therapeutic molecule, preventing the disruption of epithelial homeostasis in COPD.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Moléculas de Adhesión Celular/metabolismo , Ginsenósidos/farmacología , Pulmón/patología , FN-kappa B/metabolismo , Transducción de Señal , Fumar/efectos adversos , Regulación hacia Arriba/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Epitelio/metabolismo , Femenino , Humanos , Hiperplasia , Mediadores de Inflamación/metabolismo , Masculino , Persona de Mediana Edad , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
5.
Artículo en Inglés | MEDLINE | ID: mdl-32371529

RESUMEN

OBJECTIVE: Diabetic cardiomyopathy (DCM) is characterized by cardiac remodeling, dysfunction, and insulin resistance; however, the underlying mechanism has not been fully elucidated. Programmed cell death 4 (PDCD4) is a novel inflammation and apoptosis gene, but its role in type 2 DCM remains elusive. We aimed to determine if PDCD4 intervention improves DCM by affecting left ventricular remodeling, function, and insulin resistance. RESEARCH DESIGN AND METHODS: We designed a PDCD4-/- rat, established a type 2 diabetes animal model, and constructed a PDCD4 overexpressed adenovirus and PDCD4 small interfer RNA (siRNA) vectors to alter PDCD4 expression in H9c2 cardiomyocytes. Thereafter, glucose levels, lipid metabolism, echocardiography, and extent of myocardial fibrosis, inflammation, and apoptosis were compared in vivo and in vitro. RESULTS: PDCD4 deficiency improved insulin resistance, cardiac remodeling, and dysfunction in type 2 DCM rats and improved myocardial hypertrophy, fibrosis, inflammation, and apoptosis. Proliferation and transformation of cardiac fibroblasts was reduced via PDCD4 downregulation in vitro under high-glucose stimulation. Furthermore, PDCD4 regulated the myocardial phosphatidylinositol 3-kinase-protein kinase B (PI3K-AKT) pathway in vivo and in vitro. PDCD4 intervention affected cardiac remodeling, dysfunction, and insulin resistance by influencing fibrosis, inflammation, and apoptosis via the PI3K-AKT pathway in vivo. CONCLUSIONS: PDCD4 knockdown protected against left ventricular remodeling, dysfunction, and insulin resistance in type 2 DCM rats. The underlying mechanisms may involve reducing cardiomyocyte apoptosis, inflammation, fibrosis, and normalized PI3K-AKT phosphorylation. To the best of our knowledge, our study is the first to report the effects and underlying mechanisms of PDCD4 in type 2 DCM. These results provide a potential new treatment avenue for improving the prognosis of patients with type 2 DCM.


Asunto(s)
Diabetes Mellitus Tipo 2 , Cardiomiopatías Diabéticas , Resistencia a la Insulina , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/genética , Cardiomiopatías Diabéticas/genética , Humanos , Resistencia a la Insulina/genética , Fosfatidilinositol 3-Quinasas , Proteínas de Unión al ARN/genética , Ratas , Ratas Sprague-Dawley , Remodelación Ventricular
6.
Genet Test Mol Biomarkers ; 23(11): 807-814, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31638427

RESUMEN

Background: The gene programmed cell death 5 (PDCD5) has recently been characterized as a tumor suppressor gene and is believed to be an important prognostic cancer marker; it is frequently involved in neoplastic transformation and apoptosis of tumor cells. Several studies have demonstrated a decrease or loss of expression of PDCD5 in certain tumors. However, the relevance of PDCD5 expression in human osteoclastoma and its clinicopathological significance have not been extensively studied. Methods: The aim of this study was to explore the relative transcriptional and translational expression levels of PDCD5 in 79 osteoclastoma samples using multi-modal methods of analysis. Results: Our findings showed that 52% (15/29) of osteoclastoma cases exhibited reduced PDCD5 expression at the transcriptional level, and 56% (44/79) exhibited lower PDCD5 expression at the protein level, when compared with nontumor tissue. In addition, the statistical significance of the altered PDCD5 protein expression was examined using the Campanacci grading system for osteoclastoma. More importantly, the decreased expression at the translational level was observed to have a negative association with the Ki-67 staining index. Conclusion: Based on these findings, abnormal PDCD5 expression might be an important biomarker in human osteoclastoma and may contribute to tumor progression and malignant cell proliferation.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Tumor Óseo de Células Gigantes/genética , Proteínas de Neoplasias/genética , Adulto , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Neoplasias Óseas/genética , Línea Celular Tumoral , Proliferación Celular/genética , China , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Tumor Óseo de Células Gigantes/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/metabolismo , Osteoblastoma/genética , Transcriptoma/genética
7.
BMC Neurosci ; 20(1): 41, 2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31387531

RESUMEN

BACKGROUND: Oxidative stress and neuroinflammation are central pathogenic mechanisms common to many neurological diseases. Isoliquiritigenin (ISL) is a flavonoid in licorice with multiple pharmacological properties, including anti-inflammatory activity, and has demonstrated protective efficacy against acute neural injury. However, potential actions against cognitive impairments have not been examined extensively. We established a rat model of cognitive impairment by intracerebroventricular injection of lipopolysaccharide (LPS), and examined the effects of ISL pretreatment on cognitive function, hippocampal injury, and hippocampal expression of various synaptic proteins, antioxidant enzymes, pro-inflammatory cytokines, and signaling factors controlling anti-oxidant and pro-inflammatory responses. RESULTS: Rats receiving LPS alone demonstrated spatial learning deficits in the Morris water maze test as evidenced by longer average escape latency, fewer platform crossings, and shorter average time in the target quadrant than untreated controls. ISL pretreatment reversed these deficits as well as LPS-induced decreases in the hippocampal expression levels of synaptophysin, postsynaptic density-95, brain-derived neurotrophic factor, superoxide dismutase, glutathione peroxidase, and BCL-2. ISL pretreatment also reversed LPS-induced increases in TUNEL-positive (apoptotic) cells, BAX/BCL-2 ratio, and expression levels of tumor necrosis factor-α, interleukin (IL)-1ß, IL-6, and C-C motif chemokine ligand 3. Pretreatment with ISL increased the expression levels of phosphorylated (p)-GSK-3ß, nuclear NRF2, HO-1 mRNA, and NQO1 mRNA, and reversed LPS-induced nuclear translocation of nuclear factor (NF)-κB. CONCLUSIONS: ISL protects against LPS-induced cognitive impairment and neuronal injury by promoting or maintaining antioxidant capacity and suppressing neuroinflammation, likely through phosphorylation-dependent inactivation of GSK-3ß, enhanced expression of NRF2-responsive antioxidant genes, and suppression of NF-κB-responsive pro-inflammatory genes.


Asunto(s)
Antiinflamatorios/farmacología , Antioxidantes/farmacología , Chalconas/farmacología , Disfunción Cognitiva/prevención & control , Animales , Antioxidantes/metabolismo , Apoptosis/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo , Disfunción Cognitiva/inducido químicamente , Citocinas/biosíntesis , Homólogo 4 de la Proteína Discs Large/biosíntesis , Glutatión Peroxidasa/biosíntesis , Hipocampo/metabolismo , Lipopolisacáridos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Ratas , Superóxido Dismutasa/biosíntesis , Sinaptofisina/biosíntesis , Proteína X Asociada a bcl-2/biosíntesis
8.
Inflammopharmacology ; 27(6): 1143-1153, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31037573

RESUMEN

Epileptogenesis is a complex pathological process that occurs after an initial brain injury and involves a series of molecular events. Isoliquiritigenin (ISL), a flavonoid in licorice, is reported to have anti-inflammatory and antioxidant effects in various experimental models, but its specific roles and molecular mechanisms in the epileptogenic process following kainic acid (KA) treatment remain unclear. The purpose of this study was to explore the effects of ISL pretreatment in KA-induced epileptic rats and the underlying mechanisms. Our findings show that ISL pretreatment significantly attenuated the KA-induced expression of ionized calcium-binding adapter molecule 1 (IBα1)-labeled microglia (F(3, 20) = 97.29, p < 0.01, ηp2 = 0.94) and glial fibrillary acidic protein (GFAP)-positive astrocytes (F(3, 20) = 72.48, p < 0.01, ηp2 = 0.92), and the release of inflammatory mediators, such as TNF-α (F(3, 20) = 133.14, p < 0.01, ηp2 = 0.95), IL-1ß, and C-C motif chemokine ligand 3 (CCL3). ISL pretreatment given before KA also significantly prevented apoptotic neuronal injury by upregulating the activities of superoxide dismutase and glutathione peroxidase. It also significantly suppressed the protein levels of Toll-like receptor 4 (TLR4) (F(3, 20) = 63.23, p < 0.01, ηp2 = 0.91) and its downstream molecules, myeloid differentiation primary response 88 (MYD88), phosphorylated (p-)IκBα, and p-NF-κB. Blocking TLR4/MYD88 signaling also attenuated KA-induced neuroinflammation and neuronal damage in the hippocampus. Overall, our study demonstrates that ISL pretreatment plays neuroprotective and anti-inflammatory roles in KA-induced epileptogenesis, which may be mediated by the TLR4/MYD88 signaling pathway.


Asunto(s)
Antiinflamatorios/farmacología , Chalconas/farmacología , Epilepsia/tratamiento farmacológico , Ácido Kaínico/farmacología , Factor 88 de Diferenciación Mieloide/fisiología , Fármacos Neuroprotectores/farmacología , Receptor Toll-Like 4/fisiología , Animales , Citocinas/análisis , Epilepsia/fisiopatología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Microglía/efectos de los fármacos , Microglía/fisiología , Factor 88 de Diferenciación Mieloide/antagonistas & inhibidores , Ratas , Ratas Wistar , Transducción de Señal/fisiología , Receptor Toll-Like 4/antagonistas & inhibidores , Proteína X Asociada a bcl-2/análisis
9.
Int Immunopharmacol ; 72: 358-366, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31030091

RESUMEN

Epileptogenesis is a dynamic process initiated by insults to brain and commonly accompanied by cognitive impairment. Isoliquiritigenin (ISL), a flavonoid in licorice, has a broad spectrum of biological effects including anti-inflammatory and antioxidant activities. However, the protective effects of ISL against cognitive impairment in epileptic processes and the underlying molecular mechanism are not well understood. To address these questions, we established an reproducible seizure model by intracerebroventricular injection of kainic acid (KA) in 21-day-old rats; ISL was intraperitoneally administered three times prior to KA injection, and changes in cognitive function; synaptic plasticity; neuronal injury; number of glial cells; and expression of pro-inflammatory cytokines and nuclear factor-like (NRF)2 signaling and NACHT, LRR, and PYD domains-containing protein (NLRP)3 inflammasome components in the hippocampus were examined. Rats with KA-induced seizures showed longer average escape latency and decreases in the number of platform crossings and average time spent in the target quadrant in the Morris water maze; ISL pretreatment reversed this decline in cognitive impairment and increased the protein levels of synaptophysin, postsynaptic density-95 and brain-derived neurotrophic factor while reducing the number of Fluoro Jade B-positive cells, microglia, and astrocytes; cleaved-Caspase-3 and -9 protein levels; and tumor necrosis factor-α, interleukin (IL)-1ß, and IL-18 production. It also enhanced the nuclear localization of NRF2, hemeoxygenase-1, and NAD(P)H:quinone oxidoreductase (NQO) 1, and reversed the upregulation of NLRP3 inflammasome components NLRP3 and Caspase-1 induced by KA injection. Thus, ISL protects against cognitive impairment in KA-induced epileptic processes possibly through regulation of NRF2 signaling and the NLRP3 inflammasome pathway.


Asunto(s)
Antiinflamatorios/uso terapéutico , Chalconas/uso terapéutico , Disfunción Cognitiva/tratamiento farmacológico , Encefalitis/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Convulsiones/tratamiento farmacológico , Animales , Antiinflamatorios/farmacología , Chalconas/farmacología , Disfunción Cognitiva/metabolismo , Encefalitis/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Inflamasomas/metabolismo , Ácido Kaínico , Masculino , Factor 2 Relacionado con NF-E2/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Ratas Wistar , Convulsiones/inducido químicamente , Convulsiones/metabolismo , Sinapsis/efectos de los fármacos
10.
Oncol Lett ; 16(3): 3805-3811, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30127992

RESUMEN

Programmed cell death 4 (PDCD4) has been recognized as a novel tumor suppressor gene, which inhibits the activation and translation of activator protein (AP)-1. Dysregulated expression of PDCD4 is also involved in various human tumors and is linked to tumor progression and development. However, the function and clinical implication of PDCD4 in giant cell tumors of the bone (GCTBs) has not been previously investigated. In the present study, PDCD4 expression was determined in 83 samples of GCTBs at mRNA and protein levels by quantitative reverse transcription-polymerase chain reaction, western blotting and immunohistochemistry. The results demonstrated that PDCD4 mRNA expression was reduced in 63% of GCTB samples (17/27) and protein expression was decreased in 65% of samples (54/83), compared with adjacent non-tumor tissues. Furthermore, decreased expression of PDCD4 was significantly associated with certain clinicopathological characteristics, including the Campanacci grade and recurrence. A strong negative correlation was determined between PDCD4 expression and the Ki-67 positive rate in GCTBs (r=-0.6392; P<0.001). The results of the present study suggest that PDCD4 may serve a role in the malignant progression of human GCTBs and may be an important prediction factor for prognosis.

11.
J Nutr Biochem ; 51: 91-98, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29107826

RESUMEN

Atherosclerosis is an age-associated disease; however, diabetic atherosclerosis has higher severity beyond age range for accumulative premature senescent cells in diabetes. Recent findings suggest that rutin, a flavonoid, has potential benefits for diabetic individuals. This study was designed to evaluate the effects of rutin on premature senescence and atherosclerosis. Apolipoprotein E knockout mice exhibiting insulin resistance after 6 weeks of high-fat diet were administered with a low dose of streptozotocin (STZ) to induce diabetes. After 8 weeks of STZ administration, rutin (40 mg/kg/d) was supplemented by gavage for the last 6 weeks. We evaluated the prosperity of the plaque and diabetes using serial echocardiography, histopathologic and metabolite analysis. Premature senescence induced by hydrogen peroxide in primary vascular smooth muscle cells (VSMCs) was used to analyze the underlying mechanism. Mice with diabetes showed more severe plaque burden on aortic arteries and less smooth muscle cells but larger senescent cell ratio in plaque compared with mice with control diets. Rutin significantly improves glucose and lipid metabolic disturbance in diabetes. Moreover, rutin decreased the atherosclerotic burden and senescent cell number and increased the VSMC ratio in aortic root plaque. In vitro, we demonstrated that rutin ameliorated premature senescence induced by oxidative stress, and the protective function may be mediated by inhibiting oxidative stress and protecting telomere. Rutin administration attenuates atherosclerosis burden and stabilizes plaque by improving metabolic disturbance and alleviating premature senescence of VSMCs. Inhibition of VSMCs premature senescence with rutin may be an effective therapy for diabetic atherosclerosis.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Antioxidantes/uso terapéutico , Aterosclerosis/dietoterapia , Angiopatías Diabéticas/dietoterapia , Suplementos Dietéticos , Músculo Liso Vascular/metabolismo , Rutina/uso terapéutico , Animales , Aorta , Aterosclerosis/complicaciones , Aterosclerosis/metabolismo , Aterosclerosis/patología , Biomarcadores/sangre , Biomarcadores/metabolismo , Células Cultivadas , Senescencia Celular , Diabetes Mellitus Experimental/complicaciones , Angiopatías Diabéticas/etiología , Angiopatías Diabéticas/metabolismo , Angiopatías Diabéticas/patología , Dieta Alta en Grasa/efectos adversos , Resistencia a la Insulina , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/inmunología , Músculo Liso Vascular/patología , Estrés Oxidativo , Homeostasis del Telómero
12.
Biochem Biophys Res Commun ; 486(3): 693-699, 2017 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-28341121

RESUMEN

The Eph family of receptor tyrosine kinases serves as key modulators of various cellular functions, including inflammation, hypertrophy and fibrosis. Recent analyses have revealed that a member of the Eph family, EphA1, plays a pivotal role in regulating insulin metabolism and kidney injury. However, the importance of EphA1 in diabetic nephropathy has not been recognized. We established a diabetic nephropathy mouse model using a high-fat diet and streptozotocin (STZ) injection. Then, the recombinant adeno-associated virus type 9 (AAV9) overexpressing EphA1 or a negative control was injected locally into the kidney. Metabolite testing and histopathological analyses of kidney fibrosis, pancreatic islet function and signaling pathways were evaluated. Our study showed that hyperglycemia, insulin resistance, and renal fibrosis accompanied the deterioration of kidney function in diabetic mice. The overexpression of EphA1 in the kidney attenuated renal fibrosis and improved kidney function but did not affect systemic glucose metabolism and pancreatic islet function. Furthermore, the overexpression of EphA1 decreased the phosphorylation of ERK1/2, JNK and MYPT1 (a substrate of Rho kinase). The overexpression of EphA1 can be therapeutically targeted to inhibit diabetic renal fibrosis, which suggests that the EphA1-Epha receptor axis may be a novel therapy target for diabetic nephropathy. Mechanistically, the overexpression of EphA1 could inhibit MAPK and the Rho pathway in diabetic kidneys.


Asunto(s)
Diabetes Mellitus Experimental/terapia , Nefropatías Diabéticas/terapia , Efrina-A1/genética , Vectores Genéticos/uso terapéutico , Hiperglucemia/terapia , Receptor EphA1/genética , Animales , Dependovirus/genética , Dependovirus/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/inducido químicamente , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Dieta Alta en Grasa , Efrina-A1/agonistas , Efrina-A1/metabolismo , Fibrosis , Regulación de la Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Hiperglucemia/inducido químicamente , Hiperglucemia/genética , Hiperglucemia/patología , Resistencia a la Insulina , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosfatasa de Miosina de Cadena Ligera/genética , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Receptor EphA1/agonistas , Receptor EphA1/metabolismo , Transducción de Señal , Estreptozocina
13.
Oncotarget ; 7(13): 16194-204, 2016 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-26908443

RESUMEN

Vasa vasorum (VV) neovascularization contributes to atherogenesis and its expansion and distribution is correlated with intraplaque expression of angiogenic factors. The present study investigated the roles of Tongxinluo (TXL), a traditional Chinese medication, on VV proliferation and atherogenesis. In vitro, TXL pre-treatment reversed the tumor necrosis factor-a (TNF-a) induced expression of vascular endothelial growth factor A (VEGF-A) and angiopoietin-1 (ANGPT-1) but not ANGPT-2, leading to increased ratio of ANGPT-1 to ANGPT-2. Consistently, TXL treatment (at a dosage of 0.38, 0.75, 1.5 g/kg/d, respectively) decreased the expression of VEGF-A while increased that of ANGPT-1 in early atherosclerotic lesions of apolipoprotein E deficient (apoE-/-) mice. On aortic ring assay, microvessels sprouting from aortas were significantly inhibited in TXL-treated mice. Moreover, VV neovascularization in plaques was markedly reduced with TXL treatment. Histological and morphological analysis demonstrated that TXL treatment reduced plaque burden, plaque size and changed the plaque composition. These data suggest that TXL inhibits early atherogenesis through regulating angiogenic factor expression and inhibiting VV proliferation in atherosclerotic plaque. Our study shed new light on the anti-atherosclerotic effect of TXL.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Aterosclerosis/patología , Medicamentos Herbarios Chinos/farmacología , Neovascularización Patológica/patología , Angiopoyetina 1/biosíntesis , Animales , Aterosclerosis/metabolismo , Masculino , Ratones , Ratones Noqueados para ApoE , Neovascularización Patológica/metabolismo , Células RAW 264.7 , Vasa Vasorum/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos
14.
Acta Neurobiol Exp (Wars) ; 76(4): 304-317, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28094821

RESUMEN

Growing evidence suggests that macrophage inflammatory protein (MIP)-1alpha (synonym CCL3) is upregulated in the neuroinflammatory processes initiated by some brain disorders, but its precise role and regulatory mechanism remain unclear. The present work aims to evaluate the role of CCL3/MIP-1alpha in lipopolysaccharide (LPS)-induced brain injury, and investigate whether the MAPKs and NF-kappaB regulate CCL3/MIP-1alpha expression. We firstly examined the patterns of CCL3/MIP-1alpha expression and phosphorylation of MAPKs in the brains of rats 6, 24, and 72 h after LPS administration. Additionally, LPS-treated rats were administered an anti-MIP-1alpha neutralizing antibody, and the microglial reaction and the expression of both cyclooxygenase-2 and inducible nitric oxide synthase (iNOS) were analyzed. We finally evaluated the effect of an inhibitor of P38 MAPK, an inhibitor of ERK1/2, or an inhibitor of NF-kappaB, on the levels of CCL3/MIP-1alpha protein and numbers of microglia in the brain. In the observation period, LPS induced CCL3/MIP-1alpha expression, which localized to OX-42-labeled microglia, leading to time-dependent increases in the phosphorylation of P38 MAPK and ERK1/2. The expression pattern of induced CCL3/MIP-1alpha was partly consistent with the phosphorylation of MAPKs (P38 MAPK, ERK1/2). Anti-MIP-1alpha attenuated microglial accumulation and the upregulation of cyclooxygenase-2 and iNOS. The inhibition of P38 MAPK, ERK1/2, or NF-kappaB signaling reduced the induced upregulation of CCL3/MIP-1alpha and the microglial accumulation. Our data suggest that upregulated CCL3/MIP-1alpha mediates the accumulation of microglia and the neuroinflammatory reaction, and its expression may be regulated by MAPKs and NF-kappaB in LPS-induced brain injury.


Asunto(s)
Lesiones Encefálicas/complicaciones , Quimiocina CCL3/metabolismo , Encefalitis/patología , Microglía/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Regulación hacia Arriba/fisiología , Animales , Anticuerpos/farmacología , Antioxidantes/farmacología , Lesiones Encefálicas/inducido químicamente , Lesiones Encefálicas/tratamiento farmacológico , Antígeno CD11b/metabolismo , Quimiocina CCL3/genética , Quimiocina CCL3/inmunología , Modelos Animales de Enfermedad , Encefalitis/etiología , Activadores de Enzimas/farmacología , Inhibidores Enzimáticos/farmacología , Proteína Ácida Fibrilar de la Glía/metabolismo , Lipopolisacáridos/toxicidad , Microglía/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Prolina/análogos & derivados , Prolina/farmacología , Ratas , Ratas Wistar , Tiocarbamatos/farmacología , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos
15.
Oncotarget ; 6(31): 30640-9, 2015 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-26431274

RESUMEN

Tyrosine kinase receptor B (TrkB) is a high-affinity receptor for brain-derived neurotrophic factor (BDNF). In addition to its nervous system functions, TrkB is also expressed in the aortic endothelium. However, the effects of endothelial TrkB signaling on atherosclerosis remained unknown. Immunofluorescence analysis revealed that TrkB expression is downregulated in the endothelium of atherosclerotic lesions from ApoE-/- mice compared with the atheroma-free aorta of WT mice. Endothelial TrkB knockdown led to increased lesion size, lipid deposition and inflammatory responses in the atherosclerotic lesions of the ApoE-/- mice compared with the control mice. Mechanistic studies showed that TrkB activation prevented VE-cadherin shedding by enhancing the interaction between vascular endothelial protein tyrosine phosphatase and VE-cadherin, maintaining VE-cadherin in a dephosphorylated state. Our data demonstrate that TrkB is an endothelial injury-response molecule in atherogenesis. Endothelial BDNF/TrkB signaling reduces VE-cadherin shedding and protects against atherosclerotic lesion development in ApoE-/- mice.


Asunto(s)
Antígenos CD/metabolismo , Apolipoproteínas E/genética , Aterosclerosis/prevención & control , Cadherinas/metabolismo , Endotelio Vascular/patología , Glicoproteínas de Membrana/metabolismo , Placa Aterosclerótica/prevención & control , Proteínas Tirosina Quinasas/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Células Cultivadas , Células Endoteliales/metabolismo , Inflamación/inmunología , Lípidos , Macrófagos/inmunología , Masculino , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Tirosina Quinasas/biosíntesis , Proteínas Tirosina Quinasas/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptor trkB , Transducción de Señal , Uniones Estrechas/fisiología
16.
Brain Res ; 1624: 469-478, 2015 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-26315376

RESUMEN

We describe the sustained delivery of chondroitinase ABC (ChABC) in the hemisected spinal cord using polypropylene carbonate (PPC) electrospun fibers with chitosan (CS) microspheres as a vehicle. PPC and ChABC-loaded CS microspheres were mixed with acetonitrile, and micron fibers were generated by electrospinning. ChABC release was assessed in vitro with high-performance liquid chromatography (HPLC) and revealed stabilized and prolonged release. Moreover, the released ChABC showed sustained activity. PPC-CS micron fibers with or without ChABC were then implanted into a hemisected thoracic spinal cord. In the following 4 weeks, we examined functional recovery and performed immunohistochemical analyses. We found that sustained delivery of ChABC promoted axon sprouting and functional recovery and reduced glial scarring; PPC-CS micron fibers without ChABC did not show these effects. The present findings suggest that PPC-CS micron fibers containing ChABC are a feasible option for spinal cord injury treatment. Furthermore, the system described here may be useful for local delivery of other therapeutic agents.


Asunto(s)
Axones/efectos de los fármacos , Condroitina ABC Liasa/uso terapéutico , Recuperación de la Función/efectos de los fármacos , Regeneración/efectos de los fármacos , Traumatismos de la Médula Espinal/tratamiento farmacológico , Animales , Quitosano/uso terapéutico , Modelos Animales de Enfermedad , Conducta Exploratoria/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Propano/análogos & derivados , Propano/uso terapéutico , Ratas , Ratas Wistar , Estadísticas no Paramétricas , Factores de Tiempo
17.
J Cell Mol Med ; 19(4): 836-49, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25661015

RESUMEN

Macrophage migration inhibitory factor (MIF) involves the pathogenesis of atherosclerosis (AS) and increased plasma MIF levels in diabetes mellitus (DM) patients are associated with AS. Here, we have been suggested that MIF could be a critical contributor for the pathological process of diabetes-associated AS by using adenovirus-mediated RNA interference. First, streptozotocin (STZ)-induced diabetic animal model was constructed in 114 apolipoprotein E-deficient mice (apoE-/- mice) fed on a regular chow diet. Then, the animals were randomly divided into three groups: Adenovirus-mediated MIF interference (Ad-MIFi), Ad-enhanced green fluorescent protein (EGFP) and normal saline (NS) group (n ≈ 33/group). Non-diabetic apoE-/- mice (n = 35) were served as controls. Ad-MIFi, Ad-EGFP and NS were, respectively, injected into the tail vein of mice from Ad-MIFi, Ad-EGFP and NS group, which were injected repeatedly 4 weeks later. Physical, biochemical, morphological and molecular parameters were measured. The results showed that diabetic apoE-/- mice had significantly aggravated atherosclerotic lesions. MIF gene interference attenuated atherosclerotic lesions and stabilized atheromatous plaque, accompanied by the decreased macrophages and lipids deposition and inflammatory cytokines production, improved glucose intolerance and plasma cholesterol level, the decreased ratio of matrix matalloproteinase-2/tissue inhibitor of metalloproteinase-1 and plaque instability index. An increased expression of MIF and its ligand CD74 was also detected in the diabetic patients with coronary artery disease. The results suggest that MIF gene interference is able to inhibit atherosclerotic lesions and increase plaque stability in diabetic apoE-/-mice. MIF inhibition could be a novel and promising approach to the treatment of DM-associated AS.


Asunto(s)
Apolipoproteínas E/metabolismo , Aterosclerosis/metabolismo , Diabetes Mellitus Experimental/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Animales , Antígenos de Diferenciación de Linfocitos B/genética , Antígenos de Diferenciación de Linfocitos B/metabolismo , Apolipoproteínas E/genética , Apoptosis/genética , Aterosclerosis/complicaciones , Aterosclerosis/genética , Western Blotting , Colesterol/sangre , Citocinas/genética , Citocinas/metabolismo , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/genética , Expresión Génica , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/metabolismo , Inmunohistoquímica , Mediadores de Inflamación/metabolismo , Oxidorreductasas Intramoleculares/genética , Receptores X del Hígado , Factores Inhibidores de la Migración de Macrófagos/genética , Ratones Noqueados , Receptores Nucleares Huérfanos/genética , Receptores Nucleares Huérfanos/metabolismo , PPAR alfa/genética , PPAR alfa/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Cardiovasc Pathol ; 23(3): 169-74, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24561077

RESUMEN

BACKGROUND: The inflammatory response of vascular endothelial cells plays important roles in the initiation and progression of atherosclerotic lesions. EphA2 receptor activation promotes the endothelial cell inflammatory response, and its expression is increased in the endothelial cell layer of atherosclerotic plaques. However, the association between EphA2 and atherosclerosis has not been determined. METHODS: Eight-week-old male ApoE(-/-) mice were systemically infected with adenoassociated virus serotype 9 carrying a small hairpin RNA specifically targeting the EphA2 gene to knock down EphA2 expression in aortic endothelial cells. These mice were then fed a high-cholesterol diet for 12 weeks. Blood was collected for the measurement of plasma lipids. The aortas were harvested to evaluate the atherosclerotic lesion size, macrophage components, and expression of proinflammatory genes using Oil Red O staining, immunofluorescence staining, and molecular biology analysis. RESULTS: The lesions formed in the entire aorta and aortic sinus of the ApoE(-/-) mice with EphA2 knockdown were significantly smaller than those in the control mice (10.7%±3.1% versus 25.1%±4.2%; 0.51±0.02mm(2) versus 0.85±0.03mm(2); n=10; P<.05). Furthermore, the lesions in the ApoE(-/-) mice with EphA2 knockdown displayed reduced inflammation compared with the control mice, as reflected by the decreased macrophage infiltration (8.2%±2.9% versus 22.7%±4%; n=10; P<.05); decreased nuclear factor-κß activation; and diminished expression of vascular cell adhesion molecule-1, E-selectin, and monocyte chemotactic protein-1 (all P<.05). CONCLUSIONS: Our data demonstrate that the EphA2 receptor silencing attenuates the extent and inflammation of atherosclerotic lesions in ApoE(-/-) mice. Thus, EphA2 knockdown in endothelial cells represents a novel therapeutic strategy for patients with atherosclerosis.


Asunto(s)
Aorta/metabolismo , Enfermedades de la Aorta/prevención & control , Apolipoproteínas E/deficiencia , Aterosclerosis/prevención & control , Placa Aterosclerótica , Receptor EphA2/deficiencia , Animales , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Colesterol en la Dieta , Modelos Animales de Enfermedad , Mediadores de Inflamación/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones Noqueados , Interferencia de ARN , Receptor EphA2/genética
19.
Mol Cancer Res ; 11(12): 1497-507, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24107295

RESUMEN

UNLABELLED: Sirtuin 1 (SIRT1) is a class III histone/protein deacetylase, and its activation status has been well documented to have physiologic benefits in human health. However, the function of SIRT1 in cancer remains controversial. Here, the expression and role of SIRT1 in gastric cancer is delineated. SIRT1 was present in all normal gastric mucosa specimens; however, it was only present in a portion of the matched gastric cancer tumor specimens. In SIRT1-positive tumors, both mRNA and protein levels were downregulated as compared with the corresponding nonneoplastic tissue. Ectopic expression of SIRT1 inhibited cell proliferation, diminished clonogenic potential, and induced a G1-phase cell-cycle arrest, the effects of which were not apparent when a catalytic-domain mutant form of SIRT1 was introduced, suggesting that SIRT1 functions in gastric cancer are dependent on its deacetylase activity. Further evidence was obtained from depletion of SIRT1. At the molecular level, SIRT1 inhibited the transcription of Cyclin D1 (CCND1), and inhibition of NF-κB in SIRT1-depleted cells rescued Cyclin D1 expression. Furthermore, inhibition of either NF-κB or Cyclin D1 in SIRT1-depleted cells reversed the inhibitory effects of SIRT1. The inhibitory role of SIRT1 was also verified in vivo using xenografts. This work characterizes SIRT1 status and demonstrates its inhibitory function in gastric cancer development, which involves NF-κB/Cyclin D1 signaling, offering a therapeutic role for SIRT1 activators. IMPLICATIONS: The inhibitory functions of SIRT1, which involve NF-κB/Cyclin D1 signaling, suggest the utility of SIRT1 activators in the prevention and therapy of gastric cancer.


Asunto(s)
Ciclina D1/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular , FN-kappa B/metabolismo , Transducción de Señal , Sirtuina 1/genética , Sirtuina 1/metabolismo , Neoplasias Gástricas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Dominio Catalítico , Línea Celular Tumoral , Proliferación Celular , Ciclina D1/genética , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , FN-kappa B/genética , Transducción de Señal/efectos de los fármacos , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 25(7): 424-8, 2013 Jul.
Artículo en Chino | MEDLINE | ID: mdl-23834942

RESUMEN

OBJECTIVE: To investigate the effect of different doses of dexamethasone (DEX) on sepsis induced acute kidney injury (AKI). METHODS: One hundred and thirty healthy male Kunming mice were randomly divided into sham group, sepsis group, physiological-dose DEX group (0.12 mg/kg), stress-dose DEX group (1.2 mg/kg), and high-dose DEX group (12 mg/kg). The sepsis model was reproduced by cecal ligation and puncture (CLP) method. Histopathological changes in the kidney were observed at 24 hours and 48 hours after CLP. The expressions of glucocorticoid receptor-α (GR-α) in the kidney were detected by immunohistochemistry. The levels of GR-α mRNA and nuclear factor-ΚB (NF-ΚB) mRNA were determined by real-time polymerase chain reaction (PCR). The levels of tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) in the plasma were measured by enzyme linked immunosorbent assay (ELISA). RESULTS: Compared with sham group mice, sepsis mice showed serious impairment in renal tubules. The mRNA and protein expression of GR-α were decreased, and NF-ΚB mRNA, plasma TNF-α and IL-1ß were elevated. Compared with sepsis group, the histopathological changes in the kidney were mitigated with different levels in groups treated with different doses of DEX. GR-α protein and mRNA were up-regulated, NF-ΚB mRNA and plasma TNF-α, IL-1ß were down-regulated obviously. The best effect could be seen in physiological DEX group [AKI score: 24 hours 1.480±0.334 vs. 3.040±0.517, 48 hours 1.840±0.167 vs. 3.400±0.400; GR-α protein (A value): 24 hours 0.102±0.009 vs. 0.088±0.005, 48 hours 0.103±0.008 vs. 0.085±0.006; GR-α mRNA: 24 hours 0.0400(0.0300, 0.0400) vs. 0.0100(0.0093, 0.0100), 48 hours 0.0350(0.0300, 0.0475) vs. 0.0100(0.0010, 0.0138); NF-ΚB mRNA: 24 hours 0.009±0.001 vs. 0.012±0.000,48 hours 0.011±0.000 vs. 0.013±0.001; TNF-α: 24 hours 105.84±3.84 ng/L vs. 135.52±4.49 ng/L, 48 hours 111.35±3.67 ng/L vs. 141.22±4.46 ng/L; IL-1ß: 24 hours 45.71±2.93 ng/L vs. 64. 12±3.62 ng/L, 48 hours 57.04±3.04 ng/L vs. 74.87±3.67 ng/L; P<0.05 or P<0.01]. CONCLUSIONS: DEX given in physiological-dose could increase renal GR-α level and alleviate the sepsis induced kidney injury. The protective effect was much better than that of high dose DEX.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Dexametasona/farmacología , Riñón/efectos de los fármacos , Sepsis/metabolismo , Lesión Renal Aguda/etiología , Lesión Renal Aguda/patología , Animales , Dexametasona/administración & dosificación , Interleucina-1beta/metabolismo , Riñón/metabolismo , Masculino , Ratones , Ratones Endogámicos , FN-kappa B/metabolismo , Receptores de Glucocorticoides/metabolismo , Sepsis/complicaciones , Sepsis/patología , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA