Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(4): 167113, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38460862

RESUMEN

Hypertension, a prevalent cardiovascular ailment globally, can precipitate numerous complications, notably hypertensive cardiomyopathy. Meteorin-like (METRNL) is demonstrated to possess potential protective properties on cardiovascular diseases. However, its specific role and underlying mechanism in hypertensive myocardial hypertrophy remain elusive. Spontaneously hypertensive rats (SHRs) served as hypertensive models to explore the effects of METRNL on hypertension and its induced myocardial hypertrophy. The research results indicate that, in contrast to Wistar-Kyoto (WKY) rats, SHRs exhibit significant symptoms of hypertension and myocardial hypertrophy, but cardiac-specific overexpression (OE) of METRNL can partially ameliorate these symptoms. In H9c2 cardiomyocytes, METRNL suppresses Ang II-induced autophagy by controlling the BRCA2/Akt/mTOR signaling pathway. But when BRCA2 expression is knocked down, this effect will be suppressed. Collectively, METRNL emerges as a potential therapeutic target for hypertensive cardiomyopathy.


Asunto(s)
Cardiomiopatías , Hipertensión , Ratas , Animales , Ratas Endogámicas WKY , Cardiomegalia/genética , Cardiomegalia/tratamiento farmacológico , Hipertensión/complicaciones , Hipertensión/genética , Hipertensión/tratamiento farmacológico , Ratas Endogámicas SHR , Miocitos Cardíacos/metabolismo , Cardiomiopatías/metabolismo , Autofagia/genética
2.
Stem Cell Res Ther ; 11(1): 541, 2020 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-33317626

RESUMEN

BACKGROUND: Mesenchymal stem cells (MSCs) have exerted their brilliant potential to promote heart repair following myocardial infarction. However, low survival rate of MSCs after transplantation due to harsh conditions with hypoxic and ischemic stress limits their therapeutic efficiency in treating cardiac dysfunction. ELABELA (ELA) serves as a peptide hormone which has been proved to facilitate cell growth, survival, and pluripotency in human embryonic stem cells. Although ELA works as an endogenous ligand of a G protein-coupled receptor APJ (Apelin receptor, APLNR), whether APJ is an essential signal for the function of ELA remains elusive. The effect of ELA on apoptosis of MSCs is still vague. OBJECTIVE: We studied the role of ELABELA (ELA) treatment on the anti-apoptosis of MSCs in hypoxic/ischemic (H/I) conditions which mimic the impaired myocardial microenvironment and explored the possible mechanisms in vitro. METHODS: MSCs were obtained from donated rats weighing between 80~120 g. MSCs were exposed to serum-free and hypoxic (1% O2) environments for 24 h, which mimics hypoxic/ischemic damage in vivo, using serum-containing normoxic conditions (20% O2) as a negative control. MSCs that were exposed to H/I injury with ELA processing were treated by 5 µM of ELA. Cell viability and apoptosis of MSCs were evaluated by CCK8 and flow cytometry, respectively. Mitochondrial function of MSCs was also assessed according to mitochondrial membrane potential (MMP) and ATP content. The protein expression of key kinases of the PI3K/AKT and ERK1/2 signaling pathways involving t-AKT, p-AKT, t-ERK1/2, and p-ERK1/2, as well as apoptosis-related protein expression of Bcl-2, Bax, and cleaved Caspase 3, were monitored by Western blot. RESULTS: We found that ELA treatment of H/I-induced MSCs improved overall cell viability, enhanced Bcl/Bax expression, and decreased Caspase 3 activity. ELA inhibited H/I-induced mitochondrial dysfunction by increasing ATP concentration and suppressing the loss of mitochondrial transmembrane potential. However, this anti-apoptotic property of ELA was restrained in APJ-silenced MSCs. Additionally, ELA treatment induced the phosphorylation of AKT and ERK, while the blockade of PI3K/AKT and ERK1/2 pathways with respective inhibitors, LY294002 and U0126, suppressed the action of ELA. CONCLUSION: ELA positively affected on the survival of MSCs and exhibited anti-apoptotic characteristics when exposed to hypoxic/ischemic condition in vitro. Also, the function of ELA was correlated with the APJ receptor, reduced mitochondrial damage, and activation of the PI3K/AKT and ERK1/2 signal axes.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Células Madre Mesenquimatosas , Animales , Apoptosis , Células Madre Mesenquimatosas/metabolismo , Mitocondrias/metabolismo , Hormonas Peptídicas , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas
3.
Sci Rep ; 9(1): 17122, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31745106

RESUMEN

This article has been retracted.

4.
Stem Cell Res Ther ; 9(1): 109, 2018 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-29673400

RESUMEN

BACKGROUND: Currently, the overall therapeutic efficiency of mesenchymal stem cells (MSCs) transplantation for the treatment of cardiovascular disease is not satisfactory. The low viability and angiogenic capacity of the implanted cells in the local infarct tissues restrict their further application. Evidence shows that long noncoding RNA H19 (lncRNA-H19) mediates cell survival and angiogenesis. Additionally, it is also involved in MSCs biological activities. This study aimed to explore the functional role of lncRNA-H19 in MSCs survival and angiogenic capacity as well as the underlying mechanism. METHODS: MSCs were obtained from C57BL/6 mice and cultured in vitro. Cells at the third passage were divided into the following groups: MSCs+H19, MSCs+H19 NC, MSCs+si-H19, MSCs+si-H19 NC and MSCs. The MSCs+H19 and MSCs+H19 NC groups were transfected with lncRNA-H19 and lncRNA-H19 scramble RNA respectively. The MSCs+si-H19 and MSCs+si-H19 NC groups were transfected with lncRNA-H19 siRNA and lncRNA-H19 siRNA scramble respectively. MSCs were used as the blank control. All groups were exposed to normoxia (20% O2) and hypoxia (1% O2)/serum deprivation (H/SD) conditions for 24 h. Cell proliferation, apoptosis and vascular densities were assessed. Bioinformatics and dual luciferase reporter assay were performed. Relevant biomarkers were detected in different experimental groups. RESULTS: Overexpression of lncRNA-H19 improved survival and angiogenic capacity of MSCs under both normoxia and H/SD conditions, whereas its knockdown impaired cell viability and their angiogenic potential. MicroRNA-199a-5p (miR-199a-5p) targeted and downregulated vascular endothelial growth factor A (VEGFA). MiR-199a-5p was a target of lncRNA-H19. LncRNA-H19 transfection led to a decreased level of miR-199a-5p, accompanied with an elevated expression of VEGFA. However, both miR-199a-5p and VEGFA presented inverse alterations in the condition of lncRNA-H19 knockdown. CONCLUSIONS: LncRNA-H19 enhanced MSCs survival and their angiogenic potential in vitro. It could directly upregulate VEGFA expression by inhibiting miR-199a-5p as a competing endogenous RNA. This mechanism contributes to a better understanding of MSCs biological activities and provides new insights for cell therapy based on MSCs transplantation.


Asunto(s)
MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Humanos , Células Madre Mesenquimatosas , Ratones
5.
Int J Mol Med ; 41(3): 1643-1650, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29286092

RESUMEN

Sepsis is characterized by injury to the microvasculature and the microvascular endothelial cells, leading to barrier dysfunction. However, the specific role of injury in septic endothelial barrier dysfunction remains to be elucidated. In the present study, it was hypothesized that endothelial cell inflammatory injury is likely required for barrier dysfunction under septic conditions in vitro. 2,3,5,4'­Tetrahydroxystilbene­2­O­ß­D­glucoside (TSG), a compound extracted from Chinese herbs, is able to inhibit the inflammatory injury of septic­serum in endothelial cells. In the present study, cell viability was assayed by CCK­8 method; mRNA and protein expression was identified by RT­qPCR, western blot or Elisa, respectively and the production of reactive oxygen species was observed by a fluorescence microscope. The present study indicated that septic serum significantly decreased the cell viability of pulmonary aortic endothelial cells (PAECs) following co­cultivation for 6 h, which occurred in a time­dependent manner. TSG notably increased the viability of PAECs in a time­ and concentration­dependent manner. Further investigations revealed that septic serum increased the secretion of interleukin (IL)­1ß, IL­6 and C­reactive protein in PAECs, whereas pretreatment with TSG significantly decreased the secretion of these inflammatory factors. These data indicated that septic serum increased inflammatory injury to the PAECs, and TSG decreased this injury via the reactive oxygen species­mitogen­activated protein kinase­nuclear factor­κB signaling pathway.


Asunto(s)
Aorta/patología , Células Endoteliales/metabolismo , Glucósidos/farmacología , Inflamación/patología , Pulmón/patología , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sepsis/sangre , Estilbenos/farmacología , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citocinas/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Células Endoteliales/patología , Glucósidos/química , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Transducción de Señal , Estilbenos/química , Superóxidos/metabolismo
6.
Stem Cell Res Ther ; 8(1): 215, 2017 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-28962638

RESUMEN

BACKGROUND: Cardiac stem cells (CSCs) transplantation has been regarded as an optimal therapeutic approach for cardiovascular disease. However, inferior survival and low differentiation efficiency of these cells in the local infarct site reduce their therapeutic efficacy. In this study, we investigated the influence of hypoxia preconditioning (HP) on CSCs survival and cardiogenic differentiation in vitro and explored the relevant mechanism. METHODS: CSCs were obtained from Sprague-Dawley rats and cells of the third passage were cultured in vitro and exposed to hypoxia (1% O2). Cells survival and apoptosis were evaluated by MTS assay and flow cytometry respectively. Cardiogenic differentiation was induced by using 5-azacytidine for another 24 h after the cells experienced HP. Normoxia (20% O2) was used as a negative control during the whole process. Cardiogenic differentiation was assessed 2 weeks after the induction. Relevant molecules were examined after HP and during the differentiation process. Anti-hypoxia-inducible factor-1α (HIF-1α) small interfering RNA (siRNA), anti-apelin siRNA, and anti-putative receptor protein related to the angiotensin receptor AT1 (APJ) siRNA were transfected in order to block their expression, and relevant downstream molecules were detected. RESULTS: Compared with the normoxia group, the hypoxia group presented more rapid growth at time points of 12 and 24 h (p < 0.01). Cells exhibited the highest proliferation rate at the time point of 24 h (p < 0.01). The cell apoptosis rate significantly declined after 24 h of hypoxia exposure (p < 0.01). Expression levels of HIF-1α, apelin, and APJ were all enhanced after HP. The percentage of apelin, α-SA, and cTnT positive cells was greatly increased in the HP group after 2 weeks of induction. The protein level of α-SA and cTnT was also significantly elevated at 7 and 14 days (p < 0.01). HIF-1α, apelin, and APJ were all increased at different time points during the cardiogenic differentiation process (p < 0.01). Knockdown of HIF-1α, apelin or APJ by siRNAs resulted in a significant reduction of α-SA and cTnT. HIF-1α blockage caused a remarkable decrease of apelin and APJ (p < 0.01). Expression levels of apelin and APJ were depressed after the inhibition of apelin (p < 0.01). CONCLUSION: HP could effectively promote CSCs survival and cardiogenic differentiation in vitro, and this procedure involved activation of the HIF-1α/apelin/APJ axis. This study provided a new perspective for exploring novel strategies to enhance CSCs transplantation efficiency.


Asunto(s)
Diferenciación Celular , Miocitos Cardíacos/citología , Oxígeno/metabolismo , Células Madre/citología , Animales , Apelina/genética , Apelina/metabolismo , Apoptosis , Hipoxia de la Célula , Células Cultivadas , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Miocitos Cardíacos/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Células Madre/metabolismo
7.
Exp Mol Pathol ; 102(2): 203-209, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28161441

RESUMEN

BACKGROUND: Mesenchymal stem cells (MSCs) transplantation has been regarded as an optimal therapeutic approach for cardiovascular disease. However, the inferior survival and low vascularization potential of these cells in the local infarct site reduce the therapeutic efficacy. In this study, we investigated the influence of apelin on MSCs survival and vascularization under hypoxic-ischemic condition in vitro and explored the relevant mechanism. METHODS: MSCs were obtained from C57BL/6 mice and cultured in vitro. Cells of the third passage were divided into MSCs and MSCs+apelin groups. In the MSCs+apelin group, MSCs were stimulated with apelin-13 (5µM). The two groups experienced exposure to hypoxia (1% O2) and serum deprivation for 24h, using normoxia (20% O2) as a negative control during the process. Human umbilical vein endothelial cells (HUVECs) were used and incubated with conditioned media from both groups to promote vascularization for another 6h. Vascular densities were assessed and relevant biomarkers were detected thereafter. RESULTS: Compared with MSCs group, MSCs+apelin group presented more rapid growth. The proliferation rate was much higher. Cells apoptosis percentage was significantly declined both under normoxic and hypoxic conditions. Media produced from MSCs+apelin group triggered HUVECs to form a larger number of vascular branches on matrigel. The expression and secretion of vascular endothelial growth factor (VEGF) were significantly increased. CONCLUSION: Apelin could effectively promote MSCs survival and vascularization under hypoxic-ischemic condition in vitro, and this procedure was associated with the upregulation of VEGF. This study provides a new perspective for exploring novel approaches to enhance MSCs survival and vascularization potential.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Neovascularización Patológica/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Apoptosis , Hipoxia de la Célula/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Medios de Cultivo Condicionados/química , Células Endoteliales de la Vena Umbilical Humana , Humanos , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos C57BL , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/genética
8.
Stem Cell Res Ther ; 8(1): 4, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-28095922

RESUMEN

BACKGROUND: Mesenchymal stem cells (MSCs) have limited potential of cardiogenic differentiation. In this study, we investigated the influence of long noncoding RNA Braveheart (lncRNA-Bvht) on cardiogenic differentiation of MSCs in vitro. METHODS: MSCs were obtained from C57BL/6 mice and cultured in vitro. Cells were divided into three groups: blank control, null vector control, and lncRNA-Bvht. All three groups experienced exposure to hypoxia (1% O2) and serum deprivation for 24 h, and 24 h of reoxygenation (20% O2). Cardiogenic differentiation was induced using 5-AZA for another 24 h. Normoxia (20% O2) was applied as a negative control during the whole process. Cardiogenic differentiation was assessed, and expressions of cardiac-specific transcription factors and epithelial-mesenchymal transition (EMT)-associated biomarkers were detected. Anti-mesoderm posterior1 (Mesp1) siRNA was transfected in order to block its expression, and relevant downstream molecules were examined. RESULTS: Compared with the blank control and null vector control groups, the lncRNA-Bvht group presented a higher percentage of differentiated cells of the cardiogenic phenotype in vitro both under the normal condition and after hypoxia/re-oxygenation. There was an increased level of cTnT and α-SA, and cardiac-specific transcription factors including Nkx2.5, Gata4, Gata6, and Isl-1 were significantly upregulated (P < 0.01). Expressions of EMT-associated genes including Snail, Twist and N-cadherin were much higher (P < 0.01). Mesp1 exhibited a distinct augmentation following lncRNA-Bvht transfection. Expressions of relevant cardiac-specific transcription factors and EMT-associated genes all presented a converse alteration in the condition of Mesp1 inhibition prior to lncRNA-Bvht transfection. CONCLUSION: lncRNA-Bvht could efficiently promote MSCs transdifferentation into cells with the cardiogenic phenotype in vitro. It might function via enhancing the expressions of cardiac-specific transcription factors and EMT-associated genes. Mesp1 could be a pivotal intermediary in the procedure.


Asunto(s)
Sistema Cardiovascular/metabolismo , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Células Madre Embrionarias de Ratones/metabolismo , Miocitos Cardíacos/metabolismo , ARN Largo no Codificante/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Sistema Cardiovascular/citología , Sistema Cardiovascular/crecimiento & desarrollo , Diferenciación Celular , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Mesodermo/citología , Mesodermo/crecimiento & desarrollo , Mesodermo/metabolismo , Ratones , Ratones Transgénicos , Células Madre Embrionarias de Ratones/citología , Miocitos Cardíacos/citología , Organogénesis/genética , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , ARN Largo no Codificante/antagonistas & inhibidores , ARN Largo no Codificante/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal
9.
Sci Rep ; 6: 29589, 2016 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-27435826

RESUMEN

Tert-butylhydroquinone (tBHQ), as an antioxidant, has been widely used for many years to prevent oxidization of food products. The aim of this study was to investigate whether tBHQ activates endothelial nitric oxide synthase (eNOS) to prevent endothelial dysfunction and lower blood pressure. The role of Akt in tBHQ-induced eNOS phosphorylation was examined in human umbilical vein endothelial cells (HUVEC) or in mice. tBHQ treatment of HUVEC increased both Akt-Ser473 phosphorylation, accompanied with increased eNOS-Ser1177 phosphorylation and NO release. Mechanically, pharmacologic or genetic inhibition of Akt abolished tBHQ-enhanced NO release and eNOS phosphorylation in HUVEC. Gain-function of PTEN or inhibition of 26S proteasome abolished tBHQ-enhanced Akt phosphorylation in HUVEC. Ex vivo analysis indicated that tBHQ improved Ach-induced endothelium-dependent relaxation in LPC-treated mice aortic arteries, which were abolished by inhibition of Akt or eNOS. In animal study, administration of tBHQ significantly increased eNOS-Ser1177 phosphorylation and acetylcholine-induced vasorelaxation, and lowered AngII-induced hypertension in wildtype mice, but not in mice deficient of Akt or eNOS. In conclusion, tBHQ via proteasome-dependent degradation of PTEN increases Akt phosphorylation, resulting in upregulation of eNOS-derived NO production and consequent improvement of endothelial function in vivo. In this way, tBHQ lowers blood pressure in hypertensive mice.


Asunto(s)
Angiotensina II/efectos adversos , Hidroquinonas/administración & dosificación , Hipertensión/tratamiento farmacológico , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosfohidrolasa PTEN/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hidroquinonas/farmacología , Hipertensión/inducido químicamente , Hipertensión/metabolismo , Masculino , Ratones , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Fosfohidrolasa PTEN/genética , Fosforilación/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/genética
10.
Exp Mol Pathol ; 100(3): 493-501, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27180105

RESUMEN

Remarkable breakthroughs made in genomic technologies have facilitated the discovery of thousands of novel transcripts that do not template protein synthesis. Numerous RNAs termed as long noncoding RNAs (lncRNAs) generated from this pervasive transcription function vividly in gene regulatory networks and a variety of biological and cellular processes. Here, we make a brief description of the known and putative functions of lncRNAs in cardiovascular biology and disease. The association between lncRNAs and stem cells mediated cardiomyocytes differentiation and neovascularization is discussed then. It will provide a new clue for further studies on these novel molecules in cardiovascular disease and bring bright prospects for their future applications in cardiac regenerative medicine.


Asunto(s)
Enfermedades Cardiovasculares/genética , Sistema Cardiovascular/metabolismo , ARN Largo no Codificante/genética , Transducción de Señal/genética , Enfermedades Cardiovasculares/terapia , Diferenciación Celular/genética , Regulación de la Expresión Génica , Humanos , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Medicina Regenerativa/métodos , Medicina Regenerativa/tendencias , Células Madre/citología , Células Madre/metabolismo
11.
Am J Emerg Med ; 34(5): 772-7, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26830216

RESUMEN

BACKGROUND: NLRP3 inflammasome activation is recently reported to be linked to the pathogenesis of sepsis. Artemisinin is shown to play beneficial effects in sepsis. However, the impacts of artemisinin on burn sepsis have not been investigated. This study is designed to investigate the role of artemisinin in burn sepsis and the involvement of NLRP3 inflammasome activation. METHODS: Male BALB/c mice were randomly divided into sham burn, burn, burn sepsis, and artemisinin treated groups. Inflammatory cytokines were measured by enzyme-linked immunosorbent assay. Adhesion molecules and neutrophil infiltration in lung and heart were detected by real-time polymerase chain reaction. Mortality rates were monitored. Artemisinin was added to Raw 264.7 cells that were stimulated with burn sepsis serum in the presence/absence of an inhibitor of NLRP3 inflammasome, 3, 4-methylenedioxy-ß-nitrostyrene. Interleukin (IL) 1ß and IL-18 messenger RNA expression as well as NLRP3 and caspase 1 protein were measured. RESULTS: Production of inflammatory cytokines in serum, levels of adhesion molecules and neutrophil infiltration in lung and heart, and mortality rate of burn septic mice were significantly higher than those of control. These effects were attenuated by artemisinin. Artemisinin down-regulated protein levels of NLRP3 and caspase 1 and inhibited the increases of IL-1ß and IL-18 messenger RNA expression from Raw 264.7 cells that were stimulated with burn sepsis serum. These effects of artemisinin were not further strengthened in the presence of 4-methylenedioxy-ß-nitrostyrene. CONCLUSION: Artemisinin protects mice from burn sepsis by attenuating the inflammatory response and alleviating inflammatory infiltration in vital organs, likely through inhibiting the activation of NLRP3 inflammasome.


Asunto(s)
Antiinfecciosos/uso terapéutico , Artemisininas/uso terapéutico , Quemaduras/complicaciones , Proteínas Portadoras/antagonistas & inhibidores , Inflamasomas/efectos de los fármacos , Sepsis/prevención & control , Animales , Antiinfecciosos/farmacología , Artemisininas/farmacología , Biomarcadores/metabolismo , Proteínas Portadoras/metabolismo , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Inflamasomas/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Proteína con Dominio Pirina 3 de la Familia NLR , Células RAW 264.7 , Distribución Aleatoria , Reacción en Cadena en Tiempo Real de la Polimerasa , Sepsis/etiología , Sepsis/metabolismo
12.
World J Emerg Surg ; 10: 53, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26550025

RESUMEN

INTRODUCTION: Metallothioneins (MTs) are a family of cysteine-rich and low molecular-weight proteins that can regulate metal metabolism and act as antioxidants. Recent studies showed that MTs played a protective role in excessive inflammation and sepsis. However, the role of MTs in burn sepsis remains unclear. This study is designed to investigate the role of MTs in burn sepsis in an experimental mouse model. METHODS: MT-I/II knockout (-/-) mice on a C57BL/6 background and their wild-type (WT) littermates were randomly divided into sham burn, burn, burn sepsis, Zn treated and Zn-MT-2 treated groups. Levels of inflammatory cytokines were measured by enzyme-linked immunosorbent assay (ELISA). Myeloperoxidase (MPO) activity was detected by spectrophotometry. In in vitro study, exogenous MT was added to macrophages that stimulated with serum from burn sepsis mice with or without Akt inhibitor LY294002. The IL-1 ß and IL-6 mRNA expression were detected by quantitative real-time polymerase chain reaction. The levels of Akt expression were determined by western blot. RESULTS: Burn sepsis induced significantly elevated levels of inflammatory cytokines in serum and increased inflammatory infiltration in the liver and lung. These effects were more prominent in MT (-/-) mice than in WT mice. Furthermore, exogenous MT-2 inhibited these elevated inflammatory response in both WT and MT (-/-) mice. MT-2 up-regulated Akt phosphorylation and abrogated the increase of IL-1ß and IL-6 mRNA expression from macrophages that stimulated with burn sepsis serum. These effects of MT-2 were abolished in the presence of LY294002. CONCLUSION: MT-2 ameliorates burn sepsis by attenuating inflammatory response and diminishing inflammatory organ damage, which is at least partly mediated by activation of Akt signaling pathway.

13.
Exp Mol Pathol ; 99(3): 693-701, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26554848

RESUMEN

BACKGROUND: In this study, we hypothesized that CSCs mediated the expression of Cx43 after transplantation post MI via the ANG II/AT1R/TGF-beta1 signaling pathway. METHODS: Myocardial infarction (MI) was induced in twenty male Sprague-Dawley rats. The rats were randomized into two groups and were then received the injection of 5 × 10(6) CSCs labeled with PKH26 in phosphate buffer solution (PBS) or equal PBS alone into the infarct anterior ventricular free wall two weeks after MI. Six weeks later, relevant signaling molecules involved were all examined. RESULTS: In the CSCs group, an increased expression of Cx43 could be observed in different zones of the left ventricle (P<0.01). There was a significant reduction of the angiotensin II (ANG II) level in plasma and different regions of the left ventricular cardiac tissues (P<0.05; P<0.01). The angiotensin II type I receptor (AT1R) was decreased accompanied with an enhanced expression of angiotensin II type II receptor (AT2R) (P<0.01). Transforming growth factor beta-1(TGF-beta1) was downregulated (P<0.01). The expression of mothers against decapentaplegic homolog (SMAD) proteins including SMAD2 and SMAD3 was attenuated whereas SMAD7 was elevated (P<0.01, P<0.01, P<0.05). In addition, the expression of mitogen-activated protein kinases (MAPKs) including extracellular kinases 1/2 (ERK1/2) and p38 was also found to be reduced (P<0.01). CONCLUSION: CSCs transplantation could enhance the level of Cx43 after MI. They might function through intervening the ANGII/AT1R/TGF-beta1 signaling pathway to regulate the expression of Cx43.


Asunto(s)
Conexina 43/biosíntesis , Infarto del Miocardio/terapia , Miocitos Cardíacos/trasplante , Transducción de Señal/fisiología , Trasplante de Células Madre/métodos , Angiotensina II/metabolismo , Animales , Modelos Animales de Enfermedad , Masculino , Infarto del Miocardio/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
14.
Stem Cell Rev Rep ; 11(6): 885-99, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26275398

RESUMEN

BACKGROUND: In this study, we hypothesized that activation of PPAR-γ enhanced MSCs survival and their therapeutic efficacy via upregulating the expression of Cx43. METHODS: MI was induced in 50 male Sprague-Dawley rats. The rats were randomized into five groups: MI group and four intervention groups, including the MSCs group, combined therapy group (MSCs+ pioglitazone), pioglitazone group and PBS group. Two weeks later, 5 × 10(6) MSCs labeled with PKH26 in PBS were injected into the infarct anterior ventricular free wall in the MSCs and combined therapy groups, and PBS alone was injected into the infarct anterior ventricular free wall in the PBS group. Pioglitazone (3 mg/kg/day) was given to the combined therapy and pioglitazone groups by oral gavage at the same time for another 2 weeks. Myocardial function and relevant signaling molecules involved were all examined thereafter. RESULTS: Heart function was enhanced after MSCs treatment for 2 weeks post MI. A significant improvement of heart function was observed in the combined therapy group in contrast to the other three intervention groups. Compared with the MSCs group, there was a higher level of PPAR-γ in the combined therapy group; Cx43 was remarkably increased in different regions of the left ventricle; TGF-ß1 was decreased in the infarct zone and border zone. To the downstream signaling molecules, mothers against Smad proteins including Smad2 and Smad3 presented a synchronized alteration with TGF-ß1; no differences of the expressions of ERK1/2 and p38 could be discovered in the left ventricular cardiac tissue. CONCLUSIONS: MSCs transplantation combined with pioglitazone administration improved cardiac function more effectively after MI. Activation of PPAR-γ could promote MSCs to express Cx43. Inhibition of TGF-ß1/Smads signaling pathway might be involved in the process.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Conexina 43/biosíntesis , Trasplante de Células Madre Mesenquimatosas , Infarto del Miocardio/terapia , PPAR gamma/metabolismo , Tiazolidinedionas/uso terapéutico , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Modelos Animales de Enfermedad , Activación Enzimática , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , PPAR gamma/agonistas , Pioglitazona , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Proteína Smad2/metabolismo , Proteína smad3/metabolismo
15.
Int J Surg ; 21: 22-7, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26163884

RESUMEN

INTRODUCTION: This study aims to investigate whether apelin would regulate inflammatory response and promote survival in an experimental burn sepsis model through a phosphatidylinositol 3-kinase/protein kinase B dependent pathway. METHODS: Male BALB/c mice were divided into the following groups: sham, burn, burn sepsis, burn sepsis treated with apelin, burn sepsis treated with apelin plus LY294002, and burn sepsis treated with LY294002 alone. Apelin level and inflammatory cytokines in serum were detected by enzyme-linked immuno sorbent assay. Apelin/APJ (apelin receptor, gene symbol APLNR) mRNA expression in spleen and adhesion molecules levels in lung was detected by real-time polymerase chain reaction. Neutrophil infiltration in lung was determined by myeloperoxidase assay. Phosphorylation of protein kinase B in lung was determined by western blot. Mortality rate was monitored. RESULTS: Burn sepsis induced decreased apelin/APJ mRNA expression in spleen and reduced apelin level in plasma, which were both restored by exogenous apelin treatment. Burn sepsis treated with apelin resulted in decreased interleukin-6, tumor-necrosis factor-alpha, interleukin -1ß and monocyte chemotactic protein-1 levels in plasma. Mice with apelin treatment also showed decreased neutrophil infiltration and adhesion molecules expression, accompanied by a remarkable increased protein kinase B phosphorylation in lung tissue. The mortality rate in apelin treated animals was also significantly reduced. Importantly, the above effects of apelin were abolished by LY294002 treatment. CONCLUSION: Apelin regulates inflammatory response, diminishes inflammatory remote organ damage and improves survival in an experimental model of burn sepsis, which is at least partly mediated by a phosphatidylinositol 3-kinase/protein kinase B dependent pathway.


Asunto(s)
Adipoquinas/genética , Quemaduras/complicaciones , Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/genética , Fosfatidilinositol 3-Quinasa/genética , Proteínas Proto-Oncogénicas c-akt/genética , ARN Mensajero/genética , Sepsis/genética , Adipoquinas/biosíntesis , Animales , Apelina , Western Blotting , Quemaduras/genética , Quemaduras/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Masculino , Ratones , Ratones Endogámicos BALB C , Fosfatidilinositol 3-Quinasa/biosíntesis , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Reacción en Cadena en Tiempo Real de la Polimerasa , Sepsis/etiología , Sepsis/metabolismo
16.
Cell Biochem Biophys ; 71(2): 735-40, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25304741

RESUMEN

To observe the effect of simvastatin in patients with acute myocardial infarction in rabbits against myocardial apoptosis, and to explore its possible mechanism. Male New Zealand white rabbits were randomized into three groups, including the myocardial infarction group (12 rabbits), the simvastatin treatment group (15 rabbits), and the sham group (12 rabbits). In the simvastatin treatment and myocardial infarction groups, the rabbits received myocardial infarction surgeries. While in the sham group, loose knots were tied in the left anterior descending coronary artery branches. The simvastatin treatment group was given simvastatin by oral gavage 24 h after surgery. Parameters, which included left ventricular end-diastolic diameter, left ventricular end-systolic diameter, left ventricular ejection fraction, and left ventricular mass index, were recorded in these three groups. Edge myocardial infarction and myocardial cell apoptosis were analyzed using TUNEL assay, and Bcl-2, Bax, and Caspase-3 protein levels were detected by Western blot. Acute myocardial infarction model was successfully established in rabbits by ligation of the left anterior descending coronary artery. Compared with the myocardial infarction group, left ventricular end-diastolic diameter (LVEDD) and left ventricular end systolic diameter (LVESD) were significantly reduced and left ventricular ejection fraction (LVEF) increased in the simvastatin treatment group. Compared with the sham group, LVEDD and LVESD were significantly increased and LVEF decreased in the simvastatin treatment group. All the differences were statistically significant (P < 0.05). Left ventricular mass index in the simvastatin treatment group was statistically lower than the myocardial infarction group. Compared with the sham group, left ventricular mass index in both the simvastatin treatment and myocardial infarction groups was significantly increased. The differences of the above comparisons were statistically significant (P < 0.05). Compared with the sham group, the apoptosis rate of the myocardial infarction group and the simvastatin treatment groups was significantly increased as shown by TUNEL assay, however, the apoptosis rate of the simvastatin treatment group was significantly lower than that of the myocardial infarction group. All the differences among above comparisons were statistically significant (P < 0.05). Bcl-2 levels significantly increased in the simvastatin treatment group compared with the myocardial infarction group, but Bcl-2 levels in both groups were significantly lower than the sham group. However, Bax protein levels showed inverse expression with Bcl-2. Meanwhile, Caspase-3 protein expression showed similar trend with Bcl-2. Simvastatin can improve cardiac function after myocardial infarction and reduce apoptosis of myocardial cells, possibly by decreasing Bax and Caspase-3 expression and increasing the expression level of Bcl-2.


Asunto(s)
Apoptosis/efectos de los fármacos , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/patología , Simvastatina/farmacología , Enfermedad Aguda , Animales , Caspasa 3/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Masculino , Infarto del Miocardio/metabolismo , Tamaño de los Órganos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Conejos , Simvastatina/uso terapéutico , Proteína X Asociada a bcl-2/metabolismo
17.
Pak J Med Sci ; 30(6): 1314-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25674130

RESUMEN

OBJECTIVE: To investigate the role of Brachial ankle Pulse Wave Relocity (baPWV) and cfPWV on the risk of Coronary artery disease and the interaction between baPWV and risk factors of Coronary artery disease (CAD). METHODS: A case-control study was conducted at Department of Emergency, SunYat-Sen memorial Hospital, China. We collected 332 cases with coronary artery disease and 328 subjects without CAD between February 2012 and October 2013. A multivariate logistic regression analysis was performed to analyze the risk factors of CAD. RESULTS: CAD subjects were more likely to be old age, and have higher BMI, waist-hip ratio, hypertension, fasting glucose, TG, carotid-femoral PWV (cfPWV) and baPWV, and CAD subjects had a lower TC, HDL-C and LDL-C. We found that older age, smoking, higher hypertension, TC, TG, HDL-C, LDL-C, carotid-femoral PWV (CfPWV) and baPWV were associated with risk of CAD. baPWV had significant interaction with age, TC, TG, HDL-C and LDL-C, carotid-femoral PWV (cfWV) was correlated with age, HDL-C and LDL-C. CONCLUSION: This study showed that baPWV and cfPWV are two independent factors for the risk of Coronary artery disease, and baPWV and cfPWV have interaction with age, TC, TG, HDL-C and LDL-C.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA