Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Cardiovasc Ther ; 2021: 9956814, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34354763

RESUMEN

OBJECTIVE: Necrostatin-1 (Nec-1), an inhibitor of necroptosis, has been reported to protect against myocardial ischemia-reperfusion (MI/R) injury. However, the contribution of the potential antinecroptotic effect of Nec-1 on its infarct limitation and cardiac function improvement effects after MI/R has not been investigated. METHODS: The present study investigated the effect of Nec-1 on myocardial infarct size, necroptosis, and cardiac functional recovery in rats subjected to myocardial ischemia-reperfusion (MI/R 30 min/12, 24, 48, and 72 h). RESULTS: The study showed that Nec-1 might reduce myocardial cell death and maintain myoarchitectonic integrity, consequently inhibiting the reactive fibrosis process in rats in myocardial ischemia/late reperfusion. Moreover, the administration of Nec-1 (0.6 mg/kg) at the onset of reperfusion significantly reduced the release of creatine kinase and downregulation of autophagy within 24 h after reperfusion, and there was a significantly positive correlation between them. CONCLUSION: These results suggest that antinecroptosis treatment may improve the clinical outcomes of patients with ischemic heart disease.


Asunto(s)
Apoptosis , Autofagia , Regulación hacia Abajo , Daño por Reperfusión Miocárdica , Animales , Muerte Celular , Humanos , Imidazoles , Indoles , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos , Ratas , Ratas Sprague-Dawley , Reperfusión
2.
Biochem Biophys Res Commun ; 535: 60-65, 2021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33341674

RESUMEN

Nonalcoholic steatohepatitis (NASH) is characterized by inflammation, hepatocellular injury, and different degrees of fibrosis. Previous studies have indicated that the transcriptional coactivator with PDZ-binding motif TAZ (WWTR1) is correlated with the increased level of liver cholesterol which suppresses TAZ proteasomal degradation and promotes fibrotic NASH by activating soluble adenylyl cyclase -calcium-RhoA pathway. However, the exact mechanism by which TAZ promotes inflammatory and hepatocyte injury has not yet been fully addressed. Reportedly, p62/Sqstm1plays a pivotal role in inflammatory and hepatocyte injury during NASH development. Here, we demonstrated that p62/Sqstm1 was overexpressed in the livers of mouse NASH models in a TAZ-dependent manner. In addition, hepatocyte-specific TAZ deletion reduced p62/Sqstm1 both in vitro and in vivo. Strikingly, luciferase reporter data demonstrated that p62/Sqstm1 is a TAZ/TEAD target gene and can be transcriptionally regulated by TAZ, indicating that hepatocyte-specific TAZ deletion downregulates p62/Sqstm1 expression in NASH.


Asunto(s)
Regulación hacia Abajo , Eliminación de Gen , Hepatocitos/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Especificidad de Órganos , Proteína Sequestosoma-1/metabolismo , Transactivadores/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Línea Celular , Dieta , Silenciador del Gen , Ratones Endogámicos C57BL , Proteína Sequestosoma-1/genética , Transcripción Genética
3.
Biochem Biophys Res Commun ; 510(4): 495-500, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30658852

RESUMEN

Epithelial-mesenchymal transition (EMT) occurs in the progression of liver fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). The hydroxysteroid sulfotransferase 2B1b (SULT2B1b) promotes the proliferation of hepatocarcinoma cells both in vitro and in vivo. However, the correlation between SULT2B1b and the EMT in hepatocytes has not yet been addressed. The present study demonstrated that the SULT2B1b overexpression promoted the EMT process in mouse primary hepatocytes in the absence or presence of TGF-ß1 treatment. Moreover, SULT2B1b interference suppressed the EMT and attenuated the migration and invasion abilities of human hepatocarcinoma BEL-7402 cells by inhibiting the activation of the ß-catenin/MMP-7 pathway. In summary, SULT2B1b enhanced the EMT of hepatocytes and promoted the migration and invasion abilities of BEL-7402 cells by activing the ß-catenin/MMP-7 pathway. Therefore, inhibition of SULT2B1b has therapeutic potential for the treatment of HCC.


Asunto(s)
Transición Epitelial-Mesenquimal , Metaloproteinasa 7 de la Matriz/metabolismo , Sulfotransferasas/metabolismo , beta Catenina/metabolismo , Animales , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Células Cultivadas , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Vía de Señalización Wnt
4.
Biomed Pharmacother ; 101: 787-791, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29525674

RESUMEN

Cirsium japonicum, a constituent of traditional Chinese medicine, has been shown to exert inflammatory effects as well as to improve the circulation and thus to counteract hematologic stasis. Studies have demonstrated that intermedin (IMD) has protective effects on hypertension in rats by regulating the Ang/NO metabolic pathway. In this study, we investigated whether by regulating the expression of IMD, Cirsium japonicum could improve cardiac function in rats with 2k1c-induced renal hypertension. Renal hypertension was induced in Sprague-Dawley rats by occluding the renal artery. The rats were maintained on a normal diet and randomly divided into four groups: sham, 2k1c, 2k1c with Cirsium japonicum (1.8 g/kg per day) and 2k1c with IMD (n = 10 in each group). Cardiac function, plasma angiotensin II (Ang II), IMD, serum nitric oxide (NO) and nitric oxide synthase (NOS), as well as the expression of IMD and adrenomedullin (ADM) in the aorta and left ventricle were analyzed. Administration of Cirsium japonicum or IMD significantly strengthened cardiac function in 2k1c-induced rats, increased serum NO and NOS levels, reduced plasma Ang II, and upregulated IMD expression in the aorta and left ventricle. These results demonstrate that Cirsium japonicum has cardioprotective effects on 2k1c-induced renal hypertension in rats via the IMD/NO pathway.


Asunto(s)
Adrenomedulina/sangre , Cirsium , Hipertensión Renal/sangre , Neuropéptidos/sangre , Óxido Nítrico/sangre , Extractos Vegetales/uso terapéutico , Función Ventricular Izquierda/fisiología , Animales , Aorta/efectos de los fármacos , Aorta/fisiología , Hipertensión Renal/tratamiento farmacológico , Hipertensión Renal/fisiopatología , Masculino , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/farmacología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Función Ventricular Izquierda/efectos de los fármacos
5.
Mol Med Rep ; 16(6): 9181-9188, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29039510

RESUMEN

Atherosclerosis (AS) is a progressive disease of multifactorial origin, which occurs in response to endothelial injury. Increased homocysteine (Hcy) is considered a major cause of endothelial dysfunction, oxidative stress and DNA methylation; however, the mechanisms remain to be fully elucidated. The aim of the present study was to investigate whether Hcy causes injury to endothelial cells (ECs) by the effect of lectin­like oxidized­low density lipoprotein receptor­1 (LOX­1) DNA methylation through toll­like receptor 4(TLR4)/nuclear factor (NF)­κB/DNA methyltransferase (DNMT)1. The ECs were treated with different concentrations of Hcy, and it was found that Hcy promoted the expression of TLR4, leading to EC injury. The effect of oxidative stress was analyzed by measuring superoxide dismutase, malondialdehyde and hydrogen peroxide in the ECs. In addition, the association between NF­κB and DNMT1 was examined by treatment of the ECs with pyrrolidine dithiocarbamate (PDTC). The results suggested that Hcy induced LOX­1 DNA hypomethyaltion to promote the expression levels of LOX­1. Taken together, Hcy injured the ECs through the effect of methylation and trans­sulfuration metabolism of LOX­1 through TLR4/NF­κB/DNMT1. Following injury to the ECs, lipids, particularly ox­LDL, accumulated in the sub­endothelial layer to promote the formation of AS.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Metilación de ADN , Células Endoteliales/metabolismo , Homocisteína/metabolismo , FN-kappa B/metabolismo , Estrés Oxidativo , Receptores Depuradores de Clase E/genética , Receptor Toll-Like 4/metabolismo , Biomarcadores , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Regulación de la Expresión Génica , Homocisteína/farmacología , Humanos , Peróxido de Hidrógeno , Lipoproteínas LDL , Superóxido Dismutasa/metabolismo , Receptor Toll-Like 4/genética
6.
Mol Med Rep ; 16(5): 7775-7783, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28944836

RESUMEN

Vascular smooth muscle cell (VSMC) proliferation is a primary pathological event in atherosclerosis (AS), and homocysteine (Hcy) is an independent risk factor for AS. However, the underlying mechanisms are still lagging. Studies have used the combination of methylation of promoters of multiple genes to diagnose tumors, thus the aim of the current study was to investigate the role of methylation status of several genes in VSMCs treated with Hcy. CpG islands were identified in the promoters of platelet­derived growth factor (PDGF), p53, phosphatase and tensin homologue on chromosome 10 (PTEN) and mitofusin 2 (MFN2). Hypomethylation was observed to occur in the promoter region of PDGF, hypermethylation in p53, PTEN and MFN2, and hypomethylation in two global methylation indicators, aluminium (Alu) and long interspersed nucleotide element­1 (Line­1). This was accompanied by an increase in the expression of PDGF, and reductions of p53, PTEN and MFN2, both in mRNA and protein levels. An elevation of S­adenosylmethionine (SAM) and a reduction of S­adenosylhomocysteine (SAH) and the SAM/SAH ratio were also identified. In conclusion, Hcy impacted methylation the of AS­associated genes and global methylation status that mediate the cell proliferation, which may be a character of VSMCs treated with Hcy. The data provided evidence for mechanisms of VSMCs proliferation in AS induced by Hcy and may provide a new perspective for AS induced by Hcy.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Epigénesis Genética , Homocisteína/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Regiones Promotoras Genéticas , Elementos Alu , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Proliferación Celular/efectos de los fármacos , Islas de CpG , Femenino , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Humanos , Elementos de Nucleótido Esparcido Largo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Modelos Biológicos , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Cultivo Primario de Células , S-Adenosilhomocisteína/metabolismo , S-Adenosilmetionina/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Venas Umbilicales/citología , Venas Umbilicales/efectos de los fármacos , Venas Umbilicales/metabolismo
7.
Cardiovasc Toxicol ; 17(1): 25-34, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-26679939

RESUMEN

Hypercholesterolemia can increase the risk of cardiac injury, but the underlying mechanisms are not fully understood. The present study aimed to determine whether changes in the fluidity of the cardiomyocyte membrane may contribute to the increased susceptibility to myocardial ischemia/reperfusion (MI/R) injury observed in hypercholesterolemic rats. Male Wistar rats were fed a normal (n = 24) or high-cholesterol diet (n = 32) for 10 weeks. At the 6th week, the rats in the high-cholesterol diet group were treated with vehicle (n = 16, HC + V) or pioglitazone (n = 16, HC + PIO), a peroxisome proliferator-activated receptor-γ (PPARγ) agonist, and treatment lasted for the next 4 weeks. Rats in HC + V group displayed less membrane fluidity, a greater membrane cholesterol-to-phospholipid ratio (C/P), less Na+-K+-ATPase activity, and less cAMP content in their myocardial cells than rats fed a normal diet. A strong positive correlation was observed between membrane fluidity and cardiac injury, i.e., the myocardial infarct size when subjected to MI/R (30 min/24 h). Treatment with PIO restored much of the lost hypercholesterolemia-induced myocardial cell membrane fluidity, decreased membrane C/P ratio, increased Na+-K+-ATPase activity and cardiac cell cAMP content, improved cardiac function, and reduced the sizes of myocardial infarcts. Results demonstrated that hypercholesterolemia-induced decreased myocardial cell membrane fluidity may contribute to the increased susceptibility to cardiac injury, and PPARγ agonists may have therapeutic value in patients with hypercholesterolemia.


Asunto(s)
Membrana Celular/efectos de los fármacos , Hipercolesterolemia/tratamiento farmacológico , Fluidez de la Membrana/efectos de los fármacos , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , PPAR gamma/agonistas , Tiazolidinedionas/farmacología , Animales , Membrana Celular/metabolismo , Membrana Celular/patología , Colesterol/metabolismo , AMP Cíclico/metabolismo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Hipercolesterolemia/complicaciones , Hipercolesterolemia/metabolismo , Hipercolesterolemia/patología , Masculino , Infarto del Miocardio/etiología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/etiología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , PPAR gamma/metabolismo , Fosfolípidos/metabolismo , Pioglitazona , Ratas Wistar , Transducción de Señal/efectos de los fármacos , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Factores de Tiempo
8.
Placenta ; 46: 49-62, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27697222

RESUMEN

Inflammation and dysregulated lipid metabolism are involved in the pathogenesis of preeclampsia, and fatty acid binding protein 4 (FABP4) is known to regulate both inflammation and lipid metabolism. In the present study, we elucidated the role of FABP4 using in vitro and in vivo models of preclampsia. We found increased expression of FABP4 in the placenta of preeclamptic rats, which was further confirmed in HTR-8 cells, an extravillous trophoblast cell line, treated with L-NAME. Overexpression of FABP4 in HTR-8 cells resulted in upregulated expression of pro-inflammatory cytokines IL-6 and TNF-α, and increased lipid accumulation, suggesting that FABP4 plays a role in preeclampsia. Furthermore, downregulation of methylation in the promotor resulted in increased FABP4 expression, which was mediated by downregulated DNA methyltransferase 1 (DNMT1). Bioinformatics analysis showed that miR-148a/152 regulated the expression of DNMT1, and additional in vitro studies revealed that miR-148a/152 inhibited DNMT1 expression by directly binding to its 3'-UTR. Interestingly, DNMT1 enhanced the expression of miR-148a/152 by downregulation of methylation in its promotor. Taken together, our results showed that FABP4 may be involved in the pathogenesis of preeclampsia, and the expression of FABP4 is enhanced by miR-148a/152 mediated inhibition of DNMT1 expression.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Proteínas de Unión a Ácidos Grasos/metabolismo , MicroARNs/metabolismo , Preeclampsia/etiología , Animales , Citocinas/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1 , Modelos Animales de Enfermedad , Femenino , Metabolismo de los Lípidos , Metilación , NG-Nitroarginina Metil Éster , Placenta/metabolismo , Preeclampsia/metabolismo , Embarazo , Regiones Promotoras Genéticas , Ratas Sprague-Dawley , Regulación hacia Arriba
9.
Mol Med Rep ; 14(1): 289-300, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27175774

RESUMEN

The present study aimed to confirm whether the ratio of S-adenosylmethionine (SAM) to S-adenosylhomocysteine (SAH) is a sensitive indicator, and whether it can be used as a biomarker for the clinical diagnosis of atherosclerosis. Apolipoprotein E (ApoE)-/- mice were randomly divided into four groups and fed with a high methionine diet for 15 weeks. Serum levels of homocysteine (Hcy) were measured using an automatic biochemistry analyzer. The concentrations of SAM and SAH were determined using high­performance liquid chromatography. The methylation levels of B1 repetitive elements, adipocyte fatty acid binding protein (FABP4), monocyte chemoattractant protein-1 (MCP-1) and extracellular superoxide dismutase (EC­SOD) were analyzed using nested touchdown-methylation-specific-polymerase chain reaction analysis. After 15 weeks, compared with the normal control group, serum concentrations of Hcy were significantly increased by 1.15­, 2.54­ and 1.17­fold (P<0.05) in the ApoE­/­ control group, Meth group and Meth­F group, respectively. The sizes of the atherosclerotic lesions were increased in the ApoE­/­ control group, Meth group and Meth­F group, by up to 1.44­, 2.40­ and 1.45­fold, respectively, compared with the normal control group (P<0.05). The concentrations of SAM were significantly increased by 3.02­, 3.42­ and 2.46­fold in the ApoE­/­ control group, Meth group and Meth­F group, respectively (P<0.05). The ratios of SAM/SAH were increased by 1.67­ and 2.75­fold in the in ApoE­/­ control group and Meth group, respectively, compared with the normal control group. The methylation levels of B1 repetitive elements, FABP4, MCP­1 and EC­SOD were decreased and exhibited hypomethylation. The methylation statuses of these genes were correlated with the ratio of the serum levels of SAM and SAH. These findings suggested that the SAM/SAH ratio is a biomarker and may provide a sensitive indicator for the clinical diagnosis of atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , S-Adenosilhomocisteína/metabolismo , S-Adenosilmetionina/metabolismo , Animales , Apolipoproteínas E/deficiencia , Aterosclerosis/sangre , Aterosclerosis/genética , Biomarcadores , Quimiocina CCL2/genética , Metilación de ADN , Modelos Animales de Enfermedad , Proteínas de Unión a Ácidos Grasos/genética , Homocisteína/sangre , Homocisteína/metabolismo , Secuencias Repetitivas Esparcidas , Masculino , Ratones , Ratones Noqueados , Placa Aterosclerótica/metabolismo , S-Adenosilhomocisteína/sangre , S-Adenosilmetionina/sangre , Superóxido Dismutasa/genética
10.
Pharmacol Rep ; 68(2): 423-8, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26922548

RESUMEN

OBJECTIVES: To investigate the cytotoxicity of FM-Nov17 against chronic myeloid leukemia (CML) cells, we explored its underlying mechanisms mediating the induction of DNA damage and apoptotic cell death by reactive oxygen species (ROS). METHODS: MTT assays were used to measure the proliferation-inhibition ratio of K562 and K562/G01 cells. Flow cytometry (FCM) was used to test the level of extracellular ROS, DNA damage, cell cycle progression and apoptosis. Western blotting was used to verify the amount of protein. RESULTS: FM-Nov17 significantly inhibited the proliferation of K562 cells, with an IC50 of 58.28±0.304µM, and K562/G01 cells, with an IC50 of 62.36±0.136µM. FM-Nov17 significantly stimulated the generation of intracellular ROS, followed by the induction of DNA damage and the activation of the ATM-p53-r-H2AX pathway and checkpoint-related signals Chk1/Chk2, which led to increased numbers of cells in the S and G2/M phases of the cell cycle. Furthermore, FM-Nov17 induced apoptotic cell death by decreasing mitochondrial membrane potential and activating caspase-3 and PARP. The above effects were all prevented by the ROS scavenger N-acetylcysteine. CONCLUSIONS: FM-Nov17-induces DNA damage and mitochondria-dependent cellular apoptosis in CML cells. The process is mediated by the generation of ROS.


Asunto(s)
Daño del ADN/efectos de los fármacos , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Novobiocina/farmacología , Especies Reactivas de Oxígeno/metabolismo , Acetilcisteína/metabolismo , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo
11.
Mol Med Rep ; 13(1): 483-90, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26573388

RESUMEN

Accumulating evidence has suggested that homocysteine (Hcy) is an independent risk factor for atherosclerosis (AS). Hcy can promote vascular smooth muscle cell (VSMC) proliferation, which is pivotal in the pathogenesis and progression of AS. The aim of the present study was to investigate the epigenetic regulatory mechanism of microRNA (miR)­143­mediated VSMCs proliferation induced by Hcy. The results of a 3­(4,5­dimethylthiazol­2­yl)­2,5­diphe­nyltetrazolium bromide assay revealed that VSMC proliferation was increased by 1.39­fold following treatment with 100 mM Hcy, compared with the control group. The levels of miR­143 were markedly downregulated in the Hcy group, compared with the control group, as determined using reverse transcription­quantitative polymerase chain reaction analysis. In addition, the level of miR­143 methylation was increased markedly in the VSMCs treated with Hcy, compared with the control, and was reduced following transfection with DNA methyltransferase (DNMT)3a small interfering RNA, determined using methylation­specific­PCR. The activities of DNMT3a luciferase were also altered accordingly in VSMCs transfected with pre­miR­143 and miR­143 inhibitor, respectively. In addition, the expression of miR­143 was observed to be inversely correlated with the mRNA and protein expression of DNMT3 in the VSMCs. Taken together, these findings suggest that DNMT3a is a direct target of miR­143, and that the upregulation of DNMT3 is responsible for the hypermethylation of miR­143 in Hcy-induced VSMC proliferation.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Redes Reguladoras de Genes , Homocisteína/farmacología , MicroARNs/metabolismo , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/enzimología , Proliferación Celular/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , ADN Metiltransferasa 3A , Regulación hacia Abajo/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Humanos , Miocitos del Músculo Liso/efectos de los fármacos , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas/genética , Regulación hacia Arriba/efectos de los fármacos
12.
PLoS One ; 10(4): e0123314, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25928540

RESUMEN

XN4 might induce DNA damage and apoptotic cell death through reactive oxygen species (ROS). The inhibition of proliferation of K562 and K562/G01 cells was measured by MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide). The mRNA levels of NADPH oxidase 1-5 (Nox1-5) genes were evaluated by qRT-PCR. The levels of extracellular reactive oxygen species (ROS), DNA damage, apoptosis, and cell cycle progression were examined by flow cytometry (FCM). Protein levels were analyzed by immunoblotting. XN4 significantly inhibited the proliferation of K562 and K562/G01 cells, with IC50 values of 3.75±0.07 µM and 2.63±0.43 µM, respectively. XN4 significantly increased the levels of Nox4 and Nox5 mRNA, stimulating the generation of intracellular ROS, inducing DNA damage and activating ATM-γ-H2AX signaling, which increased the number of cells in the S and G2/M phase of the cell cycle. Subsequently, XN4 induced apoptotic cell death by activating caspase-3 and PARP. Moreover, the above effects were all reversed by the ROS scavenger N-acetylcysteine (NAC). Additionally, XN4 can induce apoptosis in progenitor/stem cells isolated from CML patients' bone marrow. In conclusion, XN4-induced DNA damage and cell apoptosis in CML cells is mediated by the generation of ROS.


Asunto(s)
Daño del ADN/efectos de los fármacos , Novobiocina/farmacología , Antígenos CD34/genética , Antígenos CD34/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Daño del ADN/genética , Citometría de Flujo , Humanos , Células K562 , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , NADPH Oxidasa 4 , NADPH Oxidasa 5 , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Novobiocina/análogos & derivados , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
13.
Acta Biochim Biophys Sin (Shanghai) ; 46(10): 902-10, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25187414

RESUMEN

Endoplasmic reticulum (ER) stress is emerging as an important modulator of different pathological process and as a mechanism contributing to homocysteine (Hcy)-induced hepar injury. However, the molecular event that Hcy-induced ER stress in the hepar under the atherosclerosis background is currently unknown. Endoplasmic reticulum oxidoreductin 1α (ERO1α) plays a crucial role in maintaining ER stress function. In this study, we determined the expression of ERO1α in the hepar in hyperhomocysteinemia and the effect of ERO1α in hepacytes ER stress in the presence of Hcy. HHcy model was established by feeding the methionine diet in apolipoprotein-E-deficient (ApoE-/-) mice, and the hepatocytes were incubated with folate and different concentrations of Hcy. Our results showed that Hcy triggered ER stress characterized by an increased contents of glucose-regulated protein 78 (GRP78), protein kinase RNA-like ER kinase (PERK), activating transcription factor (ATF) 6 and X-box binding protein-1 (XBP-1). The ERO1α expressions in HHcy mice and Hcy-treated hepatocytes were decreased compared with those in ApoE-/- group and control hepacytes (P < 0.05), respectively. Knocking-down the expression of ERO1α with small-interfering RNA significantly augmented Hcy-induced ER stress. Meanwhile, the expressions of ER stress-related factor including GRP78, PERK, ATF6 and XBP-1, were significantly decreased when the ERO1α gene was over-expressed in hepacytes. Our results suggested that ERO1α may be involved in Hcy-induced hepar ER stress, and the inhibition of ERO1α expression can accelerate this process.


Asunto(s)
Aterosclerosis/inducido químicamente , Retículo Endoplásmico/metabolismo , Homocisteína/efectos adversos , Hígado/metabolismo , Glicoproteínas de Membrana/farmacología , Oxidorreductasas/farmacología , Estrés Fisiológico , Animales , Apolipoproteínas E/genética , Aterosclerosis/metabolismo , Chaperón BiP del Retículo Endoplásmico , Masculino , Ratones , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Mol Med Rep ; 10(2): 947-54, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24841643

RESUMEN

It is well established that homocysteine (Hcy) is an independent risk factor for atherosclerosis (AS), which is characterized by vascular smooth muscle cell (VSMC) proliferation. However, the molecular mechanism underlying AS in VSMCs is yet to be elucidated. The aim of this study was to investigate the potential involvement of aberrant DNA methylation of the platelet­derived growth factor (PDGF) gene in Hcy­mediated VSMC proliferation and its underlying mechanism. Cultured human VSMCs were treated with varying concentrations of Hcy. VSMC proliferation, PDGF mRNA and protein expression and PDGF promoter demethylation showed a dose­dependent increase with Hcy concentration, suggesting an association among them. Cell cycle analysis revealed a decreased proportion of VSMCs in G0/G1 and an increased proportion in S phase, indicating that VSMC proliferation was increased under Hcy treatment. Furthermore, S­adenosylhomocysteine (SAH) levels were observed to increase and those of S­adenosylmethionine (SAM) were observed to decrease. The consequent decrease in the ratio of SAM/SAH may partially explain the hypomethylation of PDGF with Hcy treatment. Folate treatment exhibited an antagonistic effect against Hcy­induced VSMC proliferation, aberrant PDGF methylation and PDGF expression. These data suggest that Hcy may stimulate VSMC proliferation through the PDGF signaling pathway by affecting the epigenetic regulation of PDGF through the demethylation of its promoter region. These findings may provide novel insight into the molecular association between aberrant PDGF gene demethylation and the proliferation of VSMCs in Hcy­associated AS.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Homocisteína/farmacología , Músculo Liso Vascular/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/genética , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Ácido Fólico/farmacología , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Humanos , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , S-Adenosilhomocisteína/metabolismo , S-Adenosilmetionina/metabolismo , Cordón Umbilical/citología
15.
Huan Jing Ke Xue ; 34(2): 781-8, 2013 Feb.
Artículo en Chino | MEDLINE | ID: mdl-23668155

RESUMEN

Penicillium simplicissimum(Oudem.) Thomrn BGA can secrete lignocellulolytic enzymes, among these enzymes the highest activities of hemicellulase, cellulase, lignin peroxidase (Lip), manganese peroxidase (Mnp) and laccase are 146.82 Iu.g-1, 2.78 U.g-1, 47.97 U.g-1, 34.56 U.g-1 and 17.94 U.g-1 respectively. According to the results and the statistical analysis of SPSS, the ability of secreting lignocellulolytic enzymes by Penicillium simplicissimum significantly correlated with the structure of lignocellulose, and the biodegradation of lignocellulose was probably a kind of synergistic effect of several lignocellulolytic enzymes. In the solid-state fermentation of 30 days, the hemicellulose content has a significantly negative correlation with the fermentation days(r = -0.946, P < 0.01), there was also a significantly negative correlation between the cellulose-biodegradation and the lignin-biodegradation (r = -0.818, P<0.05). As unselected enzymes, Lip and Mnp can degrade hemicellulose and cellulose corporately when biodegrades lignin. The significant correlation is showed between Lip, Mnp and cellulose (correlation parameters are r = 0. 922, P <0.01; r = 0.807, P<0.05 respectively). In addition, the biosorption is found to have a very important effect in the removal of liquid alkali lignin by Penicillium simplicissimum. Key words:Penicillium simplicissimum; lignocellulolytic enzymes; biodegradation; biosorption; synergistic effect


Asunto(s)
Lignina/metabolismo , Penicillium/enzimología , Penicillium/metabolismo , Eliminación de Residuos/métodos , Biodegradación Ambiental , Celulasa/metabolismo , Peroxidasas/metabolismo , Polisacáridos/metabolismo
16.
Acta Biochim Biophys Sin (Shanghai) ; 45(5): 391-400, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23619570

RESUMEN

Hyperhomocysteinemia (HHcy) is a risk factor for cardiovascular disease and has a strong correlation with heart failure. However, the effects of HHcy on cardiac tissue remain less well understood. To elucidate the role of p53-dependent apoptosis in HHcy-induced cardiac injury, we fed ApoE(-/-) mice with high methionine diet to establish HHcy model. Serum Hcy, cardiac enzymes, and lipids were measured. The protein levels of Noxa, DNMT1, caspases-3/9, and p53 were determined by enzyme-linked immunosorbent assay. Bcl-2 and Bax proteins were detected by immunohistochemistry staining. S-adenosyl methionine and S-adenosyl homocysteine concentrations were determined by high-performance liquid chromatography. The mRNA levels of p53 and DNMT1 were analyzed by real-time polymerase chain reaction (PCR) and the methylation levels of p53 were analyzed by nested methylation-specific-PCR. Our data showed that the concentrations of serum Hcy and lipids were increased in Meth group compared with the N-control group, which indicated that the model was established successfully. The expression levels of p53 and Noxa were increased in Meth group, while the methylation status of p53 was hypomethylation. The activities of caspase-3/9 were increased in Meth group compared with the N-control group. In addition, immunohistochemistry staining showed that the expression of Bax was significantly increased in Meth and Meth-F group compared with the N-control group. In summary, HHcy induces cardiac injury by up-regulation of p53-dependent pro-apoptotic related genes Noxa and Bax, while p53 DNA hypomethylation is a key molecular mechanism in pathological process induced by HHcy.


Asunto(s)
Apolipoproteínas E/deficiencia , Cardiomiopatías/etiología , Hiperhomocisteinemia/fisiopatología , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteína p53 Supresora de Tumor/genética , Proteína X Asociada a bcl-2/biosíntesis , Animales , Cardiomiopatías/fisiopatología , Caspasa 3/metabolismo , Caspasa 9/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/biosíntesis , Metilación de ADN , Masculino , Ratones , Regulación hacia Arriba
17.
Mol Med Rep ; 7(4): 1293-9, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23404057

RESUMEN

Oxymatrine (OMT), an alkaloid extracted from Sophora japonica (kushen), is used to treat inflammatory diseases and various types of cancer in traditional Chinese medicine. However, the cellular and molecular mechanisms underlying the anti­inflammatory activity of OMT remain poorly understood. The present study explored the protective effect of OMT on myocardial injury in rats with septic shock by inhibiting the activation of the janus kinase­signal transducer and activator of transcription (JAK/STAT) signaling pathway. OMT treatment was found to significantly inhibit the activation of JAK2 and STAT3 in myocardial tissue. It also attenuated the expression of pro­inflammatory cytokines, including interleukin­1ß and tumor necrosis factor­α. In addition, OMT exhibited anti­inflammatory properties as heart function and myocardial contractility was improved and pathological and ultrastructural injury was prevented in myocardial tissue induced by septic shock. The results indicate that OMT exhibits substantial therapeutic potential for the treatment of septic shock­induced myocardial injury through inhibition of the JAK2/STAT3 signaling pathway.


Asunto(s)
Alcaloides/administración & dosificación , Lesiones Cardíacas/tratamiento farmacológico , Janus Quinasa 2/metabolismo , Quinolizinas/administración & dosificación , Factor de Transcripción STAT3/metabolismo , Choque Séptico/tratamiento farmacológico , Alcaloides/química , Animales , Lesiones Cardíacas/etiología , Lesiones Cardíacas/metabolismo , Lesiones Cardíacas/patología , Humanos , Interleucina-1beta/metabolismo , Fosforilación/efectos de los fármacos , Extractos Vegetales/administración & dosificación , Extractos Vegetales/química , Quinolizinas/química , Ratas , Choque Séptico/complicaciones , Choque Séptico/metabolismo , Choque Séptico/patología , Transducción de Señal/efectos de los fármacos , Sophora/química
18.
J Cell Physiol ; 228(1): 142-8, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22566240

RESUMEN

Increased vascular resistance in the fetoplacental circulation is a characteristic of preeclampsia. However, the potential molecular mechanisms of this condition remain obscure. The current study aimed to determine the direct effect of the peptide antigen corresponding to the second extracellular loop of the angiotensin II type 1 receptor (AT1R-EC(II) ) activating autoantibody (AT1-AA), a novel risk factor in preeclamptic patients, on fetoplacental villus stem blood vessels. Immunohistochemistry revealed that AT1 receptors were localized in the veins and arteries of human placental villi. Among 58 serum samples from preeclamptic patients, 28 (48.28%) were proved AT1-AA-positive by enzyme-linked immunosorbent assay [P<0.01 vs. 2/51 (3.92%) in the normal pregnancy group]. Total IgGs purified from AT1-AA-positive patients' sera (AT1-AA-IgGs) were added to isolated normal human placental blood vessels. The IgG significantly constricted both the villus veins and arteries in a dose-dependent manner in vitro, which could be blocked by the peptide corresponding to the human AT1R-EC(II) , anti-human IgG or the AT1 receptor antagonist losartan. Additionally, the venous constriction induced by AT1-AA-IgGs remained unchanged even at the end of the experiment (about half an hour), but the vasoconstriction caused by the AT1 receptor agonist angiotensin II underwent desensitization within three minutes. Collectively, our results demonstrated that AT1-AA in preeclamptic sera can directly constrict fetoplacental villus blood vessels without desensitization via the AT1 receptor in vitro, which might contribute to poor fetoplacental perfusion in preeclampsia.


Asunto(s)
Autoanticuerpos/efectos adversos , Circulación Placentaria/inmunología , Preeclampsia/metabolismo , Receptor de Angiotensina Tipo 1/inmunología , Vasoconstricción/inmunología , Adulto , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Femenino , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/aislamiento & purificación , Placenta/irrigación sanguínea , Preeclampsia/inmunología , Embarazo , Receptor de Angiotensina Tipo 2/inmunología , Técnicas de Cultivo de Tejidos , Adulto Joven
19.
Age (Dordr) ; 35(3): 733-46, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22535253

RESUMEN

Survival after acute myocardial infarction is decreased in elderly patients. The enhanced rates of apoptosis in the aging heart exacerbate myocardial ischemia/reperfusion (MI/R) injury. We have recently demonstrated that the X-linked inhibitor of apoptosis protein (XIAP), the most potent endogenous inhibitor of apoptosis, was decreased in aging rats' hearts. XIAP was balanced by two mitochondria proteins, Omi/HtrA2 and Smac/DIABLO. However, the implicative role of XIAP, Omi/HtrA2, and Smac/DIABLO to aging-related MI/R injury has not been previously investigated. In our study, male aging rats (20-24 months) or young adult rats (4-6 months) were subjected to 30 min of myocardial ischemia followed by reperfusion. MI/R-induced cardiac injury was enhanced in aging rats, as evidenced by aggravated cardiac dysfunction, enlarged infarct size, and increased myocardial apoptosis (TUNEL and caspase-3 activity). Then, the XIAP, Omi/HtrA2, and Smac/DIABLO protein and mRNA expression was detected. XIAP protein and mRNA expression was decreased in both aging hearts and aging hearts subjected to MI/R. Meanwhile, myocardial XIAP protein expression was correlated to cardiac function after MI/R. However, Omi/HtrA2, but not Smac/DIABLO, expression was increased in aging hearts. Moreover, the translocation of Omi/HtrA2 from mitochondria to cytosol was increased in both aging hearts and aging hearts subjected to MI/R. Treatment with ucf-101 (a novel and specific Omi/HtrA2 inhibitor) attenuated XIAP degradation and caspase-3 activity and exerted cardioprotective effects. Taken together, these results demonstrated that increased expression and leakage of Omi/HtrA2 enhanced MI/R injury in aging hearts via degrading XIAP and promoting myocardial apoptosis.


Asunto(s)
Envejecimiento/genética , Regulación de la Expresión Génica , Proteínas Mitocondriales/genética , Daño por Reperfusión Miocárdica/genética , Miocitos Cardíacos/patología , Proteínas del Tejido Nervioso/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Serina Endopeptidasas/genética , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Proteínas Reguladoras de la Apoptosis , Western Blotting , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Muerte Celular/genética , Modelos Animales de Enfermedad , Serina Peptidasa A2 que Requiere Temperaturas Altas , Etiquetado Corte-Fin in Situ , Masculino , Mitocondrias Cardíacas/genética , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Proteínas Mitocondriales/biosíntesis , Proteínas Mitocondriales/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , ARN Mensajero/biosíntesis , Proteínas de Unión al ARN/biosíntesis , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Serina Endopeptidasas/biosíntesis , Factores de Empalme Serina-Arginina , Proteína Inhibidora de la Apoptosis Ligada a X/genética , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo
20.
Antioxid Redox Signal ; 17(5): 733-43, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22304458

RESUMEN

AIMS: Abnormal fetal and early postnatal growth is closely associated with adult-onset metabolic syndrome (MetS). However, the underlying etiological factors remain complex. The presence of the autoantibody against the angiotensin II type 1 receptor (AT1-Ab), a known risk factor for pre-eclampsia, may create a suboptimal intrauterine fetal environment. The current study investigated whether middle-aged offspring of AT1-Ab-positive mothers were prone to metabolic disorder development. RESULTS: The AT1-Abs was detected in placental trophoblastic cells, capillary endothelium, and milk of pregnant rats actively immunized with the second extracellular loop of the AT1 receptor. AT1-Abs in newborn rats induced vasoconstriction, increased intracellular-free Ca(2+) in vitro, and was undetectable 7 weeks later. Immunized group offspring exhibited increased weight variability and insulin resistance at 40 weeks of age under a normal diet, evidenced by elevated fasting serum insulin and homeostasis model assessment score compared with the vehicle control. To further observe metabolic alterations, the offspring were given a high-sugar diet (containing 20% sucrose) 40-48 weeks postnatally. The fasting plasma glucose in immunized group offspring was markedly increased. Concomitantly, these offspring manifested increased visceral adipose tissue, increased fatty liver, increased triglycerides, decreased high-density lipoprotein cholesterol, and decreased adiponectin levels, indicative of MetS. INNOVATION: AT1-Abs could be transferred from mother to offspring via the placenta and milk. Moreover, offspring of an AT1-Ab-positive mother were more vulnerable to MetS development in middle age. CONCLUSION: AT1-Ab-positivity of mothers during pregnancy is a previously unrecognized "silent" risk factor for MetS development in their offspring.


Asunto(s)
Autoanticuerpos/inmunología , Síndrome Metabólico/patología , Receptor de Angiotensina Tipo 1/inmunología , Animales , Animales Recién Nacidos , Femenino , Resistencia a la Insulina , Embarazo , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...