Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Front Cardiovasc Med ; 10: 1150011, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37469478

RESUMEN

Malignant vasovagal reflex syndrome can be induced by pulling of cardiac tissue during percutaneous transcatheter closure of patent foramen ovale. In this case, a patient presented with a malignant vasovagal reflex syndrome characterized by decreased heart rate, cardiac arrest, and ventricular tachycardia. Therefore, it's particularly important to observe patients' heart rate and timely deal with vasovagal reflex syndrome during the operation.

2.
Front Immunol ; 13: 894002, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35634320

RESUMEN

Macrophages play an important role in clearing necrotic myocardial tissues, myocardial ischemia-reperfusion injury, and ventricular remodeling after myocardial infarction. M1 macrophages not only participate in the inflammatory response in myocardial tissues after infarction, which causes heart damage, but also exert a protective effect on the heart during ischemia. In contrast, M2 macrophages exhibit anti-inflammatory and tissue repair properties by inducing the production of high levels of anti-inflammatory cytokines and fibro-progenitor cells. Interleukin (IL)-38, a new member of the IL-1 family, has been reported to modulate the IL-36 signaling pathway by playing a role similar to that of the IL-36 receptor antagonist, which also affects the production and secretion of macrophage-related inflammatory factors that play an anti-inflammatory role. IL-38 can relieve myocardial ischemia-reperfusion injury by promoting the differentiation of M1 macrophages into M2 macrophages, inhibit the activation of NOD-like receptor thermal protein domain-associated protein 3 (NLRP3) inflammasome, and increase the secretion of anti-inflammatory cytokines, such as IL-10 and transforming growth factor-ß. The intact recombinant IL-38 can also bind to interleukin 1 receptor accessory protein-like 1 (IL-1RAPL1) to activate the c-jun N-terminal kinase/activator protein 1 (JNK/AP1) pathway and increase the production of IL-6. In addition, IL-38 regulates dendritic cell-induced cardiac regulatory T cells, thereby regulating macrophage polarization and improving ventricular remodeling after myocardial infarction. Accordingly, we speculated that IL-38 and macrophage regulation may be therapeutic targets for ameliorating myocardial ischemic injury and ventricular remodeling after myocardial infarction. However, the specific mechanism of the IL-38 action warrants further investigation.


Asunto(s)
Lesiones Cardíacas , Infarto del Miocardio , Daño por Reperfusión Miocárdica , Antiinflamatorios/farmacología , Citocinas/metabolismo , Lesiones Cardíacas/metabolismo , Humanos , Interleucinas/metabolismo , Macrófagos/metabolismo , Infarto del Miocardio/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Remodelación Ventricular
3.
Oxid Med Cell Longev ; 2022: 1806513, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35602104

RESUMEN

Background: Our previous studies have shown that interleukin- (IL-) 37 plays a protective role in patients and animal models with coronary artery disease. However, the role of IL-37 in patients with abdominal aortic aneurysm (AAA), another artery disease, is yet to be elucidated. Methods and Results: AAA tissues and plasma samples were obtained from patients with or without surgical intervention. Normal renal aortic tissues were collected from kidney transplant donors. Our findings established that in AAA, IL-37 was distributed in endothelial cells, macrophages, and vascular smooth muscle cells (VSMCs) and that it was chiefly concentrated in VSMCs. Furthermore, the expression was found to be downregulated compared with that in normal artery tissues. Immunofluorescence showed that, unlike normal arteries, IL-37 was translocated to the nucleus of VSMCs in AAA. Moreover, in patients with AAA, the expressions of IL-37, IL-6, and tumor necrosis factor- (TNF-) α were increased in the plasma in comparison with the healthy controls. Correlation analysis revealed that IL-37 was positively correlated with IL-6, TNF-α, age, aneurysm diameter, and blood pressure. Furthermore, human aortic vascular smooth muscle cells (HASMCs) were stimulated with angiotensin II (AngII) in vitro to simulate smooth muscle cell (SMC) damage in AAA. A decrease in IL-37 expression and an increase in receptor-interacting serine/threonine-protein kinase 3 (RIPK3) expression were observed in HASMCs stimulated with AngII. On this basis, inhibition of RIPK3 with GSK'872 significantly attenuated necroptosis. Moreover, the necroptosis rates were significantly lowered in HASMCs treated with recombinant IL-37, whereas the rates were enhanced when the cells were depleted of the interleukin. Immunoblotting results showed that both exogenous and endogenous IL-37 could affect the expressions of RIPK3, NLRP3, and IL-1ß. Also, the phosphorylation of RIPK3 and p65 was affected. Meanwhile, IL-37 promoted the transition of SMC from proliferative type to contractile type. Conclusions: The expression of IL-37 in VSMCs decreases in patients with AAA, whereas IL-37 supplementation suppresses RIPK3-mediated necroptosis and promotes the transition of VSMCs from proliferative to contractile type.


Asunto(s)
Aneurisma de la Aorta Abdominal , Interleucina-1 , Miocitos del Músculo Liso , Necroptosis , Angiotensina II/metabolismo , Angiotensina II/farmacología , Aneurisma de la Aorta Abdominal/patología , Células Endoteliales/metabolismo , Humanos , Interleucina-1/metabolismo , Interleucina-6/metabolismo , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Factor de Necrosis Tumoral alfa/metabolismo
4.
J Am Heart Assoc ; 9(19): e016796, 2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-32794415

RESUMEN

Background The coronavirus disease 2019 (COVID-19) has developed into a global outbreak. Patients with cardiovascular disease (CVD) with COVID-19 have different clinical characteristics and prognostic outcomes. This study aimed to summarize the clinical characteristics and laboratory indicators of patients with COVID-19 with CVD, especially the critically ill patients. Methods and Results This study included 244 patients diagnosed with COVID-19 and CVD (hypertension, coronary heart disease, or heart failure). The patients were categorized into critical (n=36) and noncritical (n=208) groups according to the interim guidance of China's National Health Commission. Clinical, laboratory, and outcome data were collected from the patients' medical records and compared between the 2 groups. The average body mass index of patients was significantly higher in the critical group than in the noncritical group. Neutrophil/lymphocyte ratio, and C-reactive protein, procalcitonin, and fibrinogen, and d-dimer levels at admission were significantly increased in the critical group. The all-cause mortality rate among cases of COVID-19 combined with CVD was 19.26%; the proportion of coronary heart disease and heart failure was significantly higher in deceased patients than in recovered patients. High body mass index, previous history of coronary heart disease, lactic acid accumulation, and a decrease in the partial pressure of oxygen were associated with death. Conclusions All-cause mortality in patients with COVID-19 with CVD in hospitals is high. The high neutrophil/lymphocyte ratio may be a predictor of critical patients. Overweight/obesity combined with coronary heart disease, severe hypoxia, and lactic acid accumulation resulting from respiratory failure are related to poor outcomes. Registration URL: https://www.chictr.org.cn; Unique identifier: ChiCTR2000029865.


Asunto(s)
Betacoronavirus , Enfermedades Cardiovasculares/epidemiología , Infecciones por Coronavirus/epidemiología , Neumonía Viral/epidemiología , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Proteína C-Reactiva/metabolismo , COVID-19 , Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/diagnóstico , China/epidemiología , Comorbilidad , Infecciones por Coronavirus/diagnóstico , Femenino , Fibrinógeno/metabolismo , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Pandemias , Neumonía Viral/diagnóstico , Polipéptido alfa Relacionado con Calcitonina/sangre , Pronóstico , Estudios Retrospectivos , SARS-CoV-2 , Tasa de Supervivencia/tendencias , Tomografía Computarizada por Rayos X
5.
Front Pharmacol ; 11: 257, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32269523

RESUMEN

BACKGROUND: The antitumor effect of doxorubicin (DOX) is limited by its acute and chronic toxicity to the heart, which causes heart injury. Heat shock protein 22 (Hsp22) is a protein proved to exert anti-apoptosis and anti-inflammatory effects in other diseases and physical conditions. In this study, we aim to explore whether Hsp22 could exert a protective role during cardiac injury in response to DOX. METHODS: The overexpression of Hsp22 was mediated via adenovirus vector to clarify the role of Hsp22 in the cardiac injury caused by DOX. DOX-induced acute heart injury mouse model was established by single intraperitoneal injection of DOX (15 mg/kg). Subsequently, cardiac staining and molecular biological analysis were performed to analyze the morphological and biochemical effects of Hsp22 on cardiac injury. H9c2 cells were used for validation in vitro. RESULTS: An increase in the expression level of Hsp22 was observed in DOX-treated heart tissue. Furthermore, cardiac-specific overexpression of Hsp22 showed reduced cardiac dysfunction, decrease in inflammatory response, and reduction in cell apoptosis in injury heart and cardiomyocytes induced by DOX in vivo and in vitro. Moreover, the suppression of Toll-like receptor (TLR)4/NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) was associated with the protective effect of Hsp22. Finally, the protective effect of Hsp22 cardiac function was almost abolished by overexpression of NLRP3 in DOX-treated mice. CONCLUSION: In summary, Hsp22 overexpression in the heart could suppress cardiac injury in response to DOX treatment through blocking TLR4/NLRP3 activation. Hsp22 may become a new therapeutic method for treating cardiac injury induced by DOX in cancer patients.

6.
Oxid Med Cell Longev ; 2020: 1645249, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32256945

RESUMEN

Oxidative stress and subsequent cardiac myocyte apoptosis play central roles in the initiation and progression of myocardial ischemia-reperfusion (I/R) injury. Homeobox transcript antisense intergenic RNA (Hotair) was previously implicated in various heart diseases, yet its role in myocardial I/R injury has not been clearly demonstrated. Mice with cardiac-restricted knockdown or overexpression of Hotair were exposed to I/R surgery. H9c2 cells were cultured and subjected to hypoxia/reoxygenation (H/R) stimulation to further verify the role and underlying mechanisms of Hotair in vitro. Histological examination, molecular detection, and functional parameters were determined in vivo and in vitro. In response to I/R or H/R treatment, Hotair expression was increased in a bromodomain-containing protein 4-dependent manner. Cardiac-restricted knockdown of Hotair exacerbated, whereas Hotair overexpression prevented I/R-induced oxidative stress, cardiac myocyte apoptosis, and cardiac dysfunction. Mechanistically, we observed that Hotair exerted its beneficial effects via activating AMP-activated protein kinase alpha (AMPKα). Further detection revealed that Hotair activated AMPKα through regulating the enhancer of zeste homolog 2/microRNA-451/calcium-binding protein 39 (EZH2/miR-451/Cab39) axis. We provide the evidence that endogenous lncRNA Hotair is an essential negative regulator for oxidative stress and cardiac myocyte apoptosis in myocardial I/R injury, which is dependent on AMPKα activation via the EZH2/miR-451/Cab39 axis.


Asunto(s)
Daño por Reperfusión Miocárdica/genética , Miocitos Cardíacos/metabolismo , Estrés Oxidativo/genética , ARN Largo no Codificante/metabolismo , Animales , Apoptosis , Masculino , Ratones
7.
Chem Biol Interact ; 311: 108772, 2019 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-31351049

RESUMEN

Atherosclerosis is a common type of cardiovascular disease (CVD), remaining one of the leading causes of global death. Tripartite motif-containing 28 (TRIM28) is a member of TRIM family that has been found to be involved in atherosclerosis. However, the role of TRIM28 in atherosclerosis remains unknown. This study aimed to investigate the effects of TRIM28 on the phenotypic switching of human aortic smooth muscle cells (HASMCs), which is considered as a fundamental event during the development of atherosclerosis. The results showed that TRIM28 was highly expressed in human atherosclerotic tissues, as well in cultured HASMCs stimulated by platelet-derived growth factor subunit B homodimer (PDGF-BB). Knockdown of TRIM28 by transfection with siRNA targeting TRIM28 (si-TRIM28) significantly suppressed the PDGF-BB-induced cell proliferation and migration of HASMCs. Besides, knockdown of TRIM28 inhibited the expressions of matrix metalloproteinase (MMP)-2 and MMP-9. The VSMC markers including α-smooth muscle actin (α-SMA), calponin and SM22α were upregulated in TRIM28 knocked down HASMCs. Furthermore, knockdown of TRIM28 blocked PDGF-BB-induced NF-κB activation in HASMCs. Collectively, knockdown of TRIM28 prevented PDGF-BB-induced phenotypic switching of HASMCs, which might be mediated by the regulation of NF-κB signaling pathway.


Asunto(s)
Becaplermina/farmacología , Proliferación Celular/efectos de los fármacos , Proteína 28 que Contiene Motivos Tripartito/metabolismo , Aterosclerosis/metabolismo , Aterosclerosis/patología , Proteínas de Unión al Calcio/metabolismo , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Humanos , Proteínas de Microfilamentos/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Factor de Transcripción ReIA/metabolismo , Proteína 28 que Contiene Motivos Tripartito/antagonistas & inhibidores , Proteína 28 que Contiene Motivos Tripartito/genética , Regulación hacia Arriba/efectos de los fármacos , Calponinas
8.
Nat Commun ; 10(1): 959, 2019 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-30814518

RESUMEN

Myocardial microRNAs (myo-miRs) are released into the circulation after acute myocardial infarction (AMI). How they impact remote organs is however largely unknown. Here we show that circulating myo-miRs are carried in exosomes and mediate functional crosstalk between the ischemic heart and the bone marrow (BM). In mice, we find that AMI is accompanied by an increase in circulating levels of myo-miRs, with miR-1, 208, and 499 predominantly in circulating exosomes and miR-133 in the non-exosomal component. Myo-miRs are imported selectively to peripheral organs and preferentially to the BM. Exosomes mediate the transfer of myo-miRs to BM mononuclear cells (MNCs), where myo-miRs downregulate CXCR4 expression. Injection of exosomes isolated from AMI mice into wild-type mice downregulates CXCR4 expression in BM-MNCs and increases the number of circulating progenitor cells. Thus, we propose that myo-miRs carried in circulating exosomes allow a systemic response to cardiac injury that may be leveraged for cardiac repair.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , MicroARNs/sangre , Infarto del Miocardio/sangre , Miocardio/metabolismo , Anciano , Animales , Regulación hacia Abajo , Exosomas/metabolismo , Femenino , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Persona de Mediana Edad , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Receptores CXCR4/metabolismo
9.
Biosci Rep ; 37(4)2017 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-28615347

RESUMEN

Endothelial cells' (EC) injury is a major step for the pathological progression of atherosclerosis. Recent study demonstrated that thymic stromal lymphopoietin (TSLP) exerts a protective role in atherosclerosis. However, the effect of TSLP and the exact molecular mechanism involved in EC remains unknown. In the present study, we found that long noncoding RNA (lncRNA) HOTAIR was much lower in EC from atherosclerotic plaque. Functional assays showed that HOTAIR facilitated cell proliferation and migration, and suppressed apoptosis in EC. Moreover, we demonstrated that TSLP functions upstream of HOTAIR. We found that serum level of TSLP was decreased in atherosclerosis patients and serum TSLP level positively correlated with HOTAIR expression in EC. Further investigation demonstrated that TSLP activated HOTAIR transcription through PI3K/AKT-IRF1 pathway and then regulates the EC proliferation and migration. TSLP-HOTAIR axis also plays a protective role in low-density lipoprotein (ox-LDL)-induced EC injury. Taken together, TSLP-HOTAIR may be a potential therapy for EC dysfunction in atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , Movimiento Celular , Proliferación Celular , Citocinas/metabolismo , ARN Largo no Codificante/biosíntesis , Aterosclerosis/patología , Línea Celular , Células Endoteliales de la Vena Umbilical Humana , Humanos , Factor 1 Regulador del Interferón/metabolismo , Lipoproteínas LDL/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Linfopoyetina del Estroma Tímico
10.
Med Sci Monit ; 23: 2007-2016, 2017 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-28445445

RESUMEN

BACKGROUND Malvidin (alvidin-3-glucoside) is a polyphenol that belongs to the class of natural anthocyanin, which is abundantly found in red wines, colored fruits, and the skin of red grapes. Therefore, the current investigation was intended to evaluate the effect of malvidin against myocardial infarction induced by isoproterenol in the rats. MATERIAL AND METHODS The cardioprotective effects was assessed by determining the effect of malvidin on the activities of endogenous antioxidants - catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GSH) - and on the levels of lipid peroxidation and serum marker enzymes. The serum levels of IL-6 and TNF-α were also determined using an enzyme-linked immunosorbent assay (ELISA) kit. RESULTS The present study demonstrated a significant cardioprotective effect of malvidin by restoring the defensive activities of endogenous antioxidants - catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GSH) - and by reducing the levels of lipid peroxidation and serum marker enzymes lactate dehydrogenase (LD) and creatine kinase (CK). Malvidin significantly ameliorated the histopathological changes and impaired mitochondria in the cardiac necrosis stimulated with isoproterenol. Additionally, the results also demonstrated that nuclear translocation of Nrf-2 and subsequent HO-1 expression might be associated with nuclear factor kappa B (NF-κB) pathway activation. CONCLUSIONS Our findings suggest that malvidin exerts cardioprotective effects that might be due to possible strong antioxidant and anti-inflammatory activities. Therefore, this study provides the basis for the development of malvidin as a safe and effective treatment of myocardial infarction.


Asunto(s)
Antocianinas/uso terapéutico , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/prevención & control , Animales , Antocianinas/farmacología , Antioxidantes/farmacología , Catalasa/efectos de los fármacos , Catalasa/metabolismo , Ensayo de Inmunoadsorción Enzimática , Glutatión Peroxidasa/efectos de los fármacos , Glutatión Peroxidasa/metabolismo , Corazón/efectos de los fármacos , Isoproterenol/farmacología , Peroxidación de Lípido/efectos de los fármacos , Masculino , Infarto del Miocardio/metabolismo , Miocardio/patología , Fitoterapia , Extractos Vegetales/farmacología , Ratas , Ratas Wistar , Superóxido Dismutasa/efectos de los fármacos , Superóxido Dismutasa/metabolismo
11.
Circ Res ; 120(11): 1754-1767, 2017 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-28348007

RESUMEN

RATIONALE: Acute coronary syndrome (ACS) is a leading cause of death worldwide. Immune functions play a vital role in ACS development; however, whether epigenetic modulation contributes to the regulation of blood immune cells in this disease has not been investigated. OBJECTIVE: We conducted an epigenome-wide analysis with circulating immune cells to identify differentially methylated genes in ACS. METHODS AND RESULTS: We examined genome-wide methylation of whole blood in 102 ACS patients and 101 controls using HumanMethylation450 array, and externally replicated significant discoveries in 100 patients and 102 controls. For the replicated loci, we further analyzed their association with ACS in 6 purified leukocyte subsets, their correlation with the expressions of annotated genes, and their association with cardiovascular traits/risk factors. We found novel and reproducible association of ACS with blood methylation at 47 cytosine-phosphoguanine sites (discovery: false discovery rate <0.005; replication: Bonferroni corrected P<0.05). The association of methylation levels at these cytosine-phosphoguanine sites with ACS was further validated in at least 1 of the 6 leukocyte subsets, with predominant contributions from CD8+ T cells, CD4+ T cells, and B cells. Blood methylation of 26 replicated cytosine-phosphoguanine sites showed significant correlation with expressions of annotated genes (including IL6R, FASLG, and CCL18; P<5.9×10-4), and differential gene expression in case versus controls corroborated the observed differential methylation. The replicated loci suggested a role in ACS-relevant functions including chemotaxis, coronary thrombosis, and T-cell-mediated cytotoxicity. Functional analysis using the top ACS-associated methylation loci in purified T and B cells revealed vital pathways related to atherogenic signaling and adaptive immune response. Furthermore, we observed a significant enrichment of the replicated cytosine-phosphoguanine sites associated with smoking and low-density lipoprotein cholesterol (Penrichment≤1×10-5). CONCLUSIONS: Our study identified novel blood methylation alterations associated with ACS and provided potential clinical biomarkers and therapeutic targets. Our results may suggest that immune signaling and cellular functions might be regulated at an epigenetic level in ACS.


Asunto(s)
Síndrome Coronario Agudo/sangre , Síndrome Coronario Agudo/genética , Metilación de ADN/fisiología , Epigénesis Genética/fisiología , Estudio de Asociación del Genoma Completo/métodos , Síndrome Coronario Agudo/epidemiología , Anciano , Estudios de Casos y Controles , China/epidemiología , Femenino , Humanos , Masculino , Persona de Mediana Edad
12.
PLoS One ; 10(10): e0141464, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26517374

RESUMEN

Abnormal expression of thymic stromal lymphopoietin (TSLP) and its receptor (TSLPR) was found in patients with acute coronary syndrome. Ticagrelor, an oral platelet ADP P2Y12 receptor antagonist, is widely used in these patients. The aim of this study was to verify whether different doses of ticagrelor regulated plaque progression and platelet activity by modulating TSLP/TSLPR. Seventy-five ApoE-/- mice were randomly divided into five groups: (1) high-cholesterol diet (HCD, n = 15); (2) HCD plus ticagrelor 25 mg/kg/d (T1, n = 15); (3) HCD plus ticagrelor 50 mg/kg/d (T2, n = 15); (4) HCD plus ticagrelor 100 mg/kg/d (T3, n = 15); and (5) a normal diet group (ND, n = 15). At day 0 and at week 16, blood lipids and serum TSLP levels, expression of TSLPR, CD62, and CD63, platelet aggregation, platelet ATP release, PI3K/Akt signaling pathway, and plaque morphology were assessed. HCD increased TSLPR expression and atherosclerosis progression but high-dose ticagrelor (100 mg/kg) moderated this trend. TSLPR was positively correlated with Akt1, platelet aggregation, corrected plaque area, and vulnerability index in the T3 group (P<0.01). In conclusion, low-dose ticagrelor only inhibited platelet activity. Besides this inhibition, high-dose ticagrelor modulated platelet activity and atherosclerosis mediated by TSLPR, potentially through the PI3K/Akt signal pathway.


Asunto(s)
Adenosina/análogos & derivados , Aterosclerosis/tratamiento farmacológico , Citocinas/fisiología , Inmunoglobulinas/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Agregación Plaquetaria/efectos de los fármacos , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores de Citocinas/efectos de los fármacos , Adenosina/administración & dosificación , Adenosina/farmacología , Animales , Aorta/metabolismo , Aorta/patología , Apolipoproteínas E/deficiencia , Aterosclerosis/sangre , Aterosclerosis/prevención & control , Colesterol en la Dieta/toxicidad , Citocinas/sangre , Relación Dosis-Respuesta a Droga , Hiperlipoproteinemia Tipo II/sangre , Hiperlipoproteinemia Tipo II/complicaciones , Hiperlipoproteinemia Tipo II/tratamiento farmacológico , Inmunoglobulinas/biosíntesis , Inmunoglobulinas/fisiología , Lípidos/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Selectina-P/análisis , Fosfatidilinositol 3-Quinasas/fisiología , Inhibidores de Agregación Plaquetaria/administración & dosificación , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/fisiología , Antagonistas del Receptor Purinérgico P2Y/administración & dosificación , Distribución Aleatoria , Receptores de Citocinas/biosíntesis , Receptores de Citocinas/fisiología , Transducción de Señal/efectos de los fármacos , Tetraspanina 30/sangre , Ticagrelor , Linfopoyetina del Estroma Tímico
13.
Mediators Inflamm ; 2015: 563951, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26161004

RESUMEN

Chemerin15 (C15), an endogenous anti-inflammatory component, inhibits the activity of neutrophils and macrophages through G protein-coupled receptor ChemR23; however, its role as well as functional mechanism in mouse myocardial ischemia/reperfusion (I/R) injury remains unknown. Methods. Sham or I/R operations were performed on C57BL/6J mice. The I/R mice received an injection of C15 immediately before reperfusion. Serum troponin T levels, infarct size, cardiomyocyte apoptosis, reactive oxygen species (ROS) production, and infiltration of neutrophils were assessed 24 h after reperfusion, while the macrophage phenotypes, macrophage infiltration, and inflammatory cytokine levels were assessed 48 h after reperfusion. Results. Compared with the control group, the C15-treated mice showed an obvious amelioration of I/R injury and displayed less ROS, accompanied by reduced neutrophil recruitment. C15 decreased the tumor necrosis factor- (TNF-) α and interleukin- (IL-) 6 levels and increased the IL-10 levels in the serum of the I/R mice, which suggested a suppressed inflammatory response that could be related to elevated alternatively activated M2 macrophages with characteristic skewed expression of M2 markers and inhibition of classically activated M1 marker expression. Conclusion. C15 may induce alternatively activated M2 macrophage polarization and suppress the inflammatory response to protect against myocardial I/R injury in mice.


Asunto(s)
Factores Quimiotácticos/uso terapéutico , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , Activación de Macrófagos/efectos de los fármacos , Daño por Reperfusión Miocárdica/prevención & control , Fragmentos de Péptidos/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/efectos de los fármacos , Infiltración Neutrófila/efectos de los fármacos
14.
Atherosclerosis ; 238(2): 278-88, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25544178

RESUMEN

Regulatory T-Cells (Tregs) play a protective role against the development of atherosclerosis. Moreover, thymic stromal lymphopoietin (TSLP)/thymic stromal lymphopoietin receptor (TSLPR) signaling in myeloid dendritic cells (DCs) promote Treg differentiation. Here, we examined the potential role of TSLP/TSLPR on Treg homeostasis in atherosclerosis. The frequencies of both latency-associated peptide (LAP)(+) and Foxp3(+) Tregs were reduced in the thymus and spleen of ApoE(-/-) mice compared with C57BL/6 mice, and this effect was associated with decreased thymic output. The tolerogenic function of DCs obtained from ApoE(-/-) mice was compromised compared with those from C57BL/6 mice. The expression of TSLP and TSLPR was also inhibited in ApoE(-/-) mice. In addition, we found that ox-LDL attenuated TSLP expression in cultured thymic epithelial cells (TECs) through the activation of retinoid X receptor alpha (RXRA) and IL-1ß and decreased LAP and PD-L1 expression in oxLDL-activated DCs while both were up-regulated in TSLP-activated DCs. We also observed that the TSLP-DCs mediated differentiation of Tregs was abrogated through LAP neutralization. Furthermore, TSLP injection rescued Treg defects in ApoE(-/-) mice. These findings suggest that Treg defects in ApoE(-/-) mice might partially be attributed to the disruption of TSLP-TSLPR-LAP signaling in epithelial cells (ECs) and DCs.


Asunto(s)
Aterosclerosis/metabolismo , Comunicación Celular , Citocinas/metabolismo , Células Dendríticas/inmunología , Células Epiteliales/metabolismo , Hiperlipidemias/metabolismo , Inmunoglobulinas/metabolismo , Péptidos/metabolismo , Receptores de Citocinas/metabolismo , Transducción de Señal , Linfocitos T Reguladores/metabolismo , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/patología , Comunicación Celular/efectos de los fármacos , Diferenciación Celular , Células Cultivadas , Técnicas de Cocultivo , Citocinas/administración & dosificación , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/inmunología , Factores de Transcripción Forkhead/metabolismo , Hiperlipidemias/genética , Hiperlipidemias/inmunología , Hiperlipidemias/patología , Tolerancia Inmunológica , Lipoproteínas LDL/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Péptidos/aislamiento & purificación , Receptores de Citocinas/agonistas , Transducción de Señal/efectos de los fármacos , Bazo/inmunología , Bazo/metabolismo , Linfocitos T Reguladores/inmunología , Timo/inmunología , Timo/metabolismo , Linfopoyetina del Estroma Tímico
15.
J Immunol Res ; 2014: 342693, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25762441

RESUMEN

Our study intended to prove whether agonistic autoantibodies to angiotensin II type 1 receptor (AT1-AAs) exist in patients with coronary heart disease (CHD) and affect the human endothelial cell (HEC) by upregulating proinflammatory cytokines expression involved in NF-κB pathway. Antibodies were determined by chronotropic responses of cultured neonatal rat cardiomyocytes coupled with receptor-specific antagonists (valsartan and AT1-EC2) as described previously. Interleukin-6 (IL-6), vascular cell adhesion molecule-1 (VCAM-1), and monocyte chemotactic protein-1 (MCP-1) expression were improved at both mRNA and protein levels in HEC, while NF-κB in the DNA level was improved detected by electrophoretic mobility shift assays (EMSA). These improvements could be inhibited by specific AT1 receptor blocker valsartan, NF-κB blocker pyrrolidine dithiocarbamate (PDTC), and specific short peptides from the second extracellular loop of AT1 receptor. These results suggested that AT1-AAs, via the AT1 receptor, induce expression of proinflammatory cytokines involved in the activation of NF-κB. AT1-AAs may play a great role in the pathogenesis of the acute coronary syndrome by mediating vascular inflammatory effects involved in the NF-κB pathway.


Asunto(s)
Síndrome Coronario Agudo/inmunología , Autoanticuerpos/metabolismo , Células Endoteliales/inmunología , Miocitos Cardíacos/metabolismo , FN-kappa B/metabolismo , Antagonistas de Receptores de Angiotensina/farmacología , Animales , Autoantígenos/inmunología , Células Cultivadas , Quimiocina CCL2/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-6/metabolismo , FN-kappa B/genética , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/inmunología , Prolina/análogos & derivados , Prolina/farmacología , Ratas , Receptor de Angiotensina Tipo 1/agonistas , Receptor de Angiotensina Tipo 1/química , Receptor de Angiotensina Tipo 1/inmunología , Tiocarbamatos/farmacología , Valsartán/farmacología , Molécula 1 de Adhesión Celular Vascular/metabolismo
16.
Mediators Inflamm ; 2013: 635672, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24453425

RESUMEN

BACKGROUND: CD4+ T helper (Th) cells play critical roles in the development and progression of atherosclerosis and the onset of acute coronary syndromes (ACS, including acute myocardial infarction (AMI) and unstable angina pectoris (UAP)). In addition to Th1, Th2, and Th17 cells, Th22 and Th9 subsets have been identified in humans. In the present study, we investigated whether Th22 cells and Th9 cells are involved in the onset of ACS. METHODS: The frequencies of Th22 and Th9 cells were detected using a flow cytometric analysis and their related cytokine and transcription factor were measured in the AMI, UAP, stable angina pectoris (SAP), and control groups. RESULTS: The results revealed a significant increase in the peripheral Th22 number, AHR expression, and IL-22 levels in patients with ACS compared with those in the SAP and control groups. Although there was no difference in the peripheral Th9 number among the four groups, the PU.1 expression and IL-9 levels were significantly increased in patients with ACS compared with the SAP and control groups. CONCLUSIONS: Circulating Th22 and Th9 type responses may play a potential role in the onset of ACS symptom.


Asunto(s)
Síndrome Coronario Agudo/inmunología , Linfocitos T Colaboradores-Inductores/fisiología , Anciano , Angina Estable/inmunología , Femenino , Citometría de Flujo , Humanos , Interleucina-9/sangre , Interleucinas/sangre , Sistema de Señalización de MAP Quinasas , Masculino , Persona de Mediana Edad , Proteínas Proto-Oncogénicas/análisis , Receptores de Hidrocarburo de Aril/análisis , Transactivadores/análisis , Función Ventricular Izquierda , Interleucina-22
17.
Zhonghua Yi Xue Za Zhi ; 91(28): 1982-6, 2011 Jul 26.
Artículo en Chino | MEDLINE | ID: mdl-22093895

RESUMEN

OBJECTIVE: To explore the therapeutic effect of transplantation of bone marrow mesenchymal stem cells (BMSCs) over-expressing Cx43 on heart failure in post-infarction rats. METHODS: Sixty SD rats were randomly divided equally into 4 groups: sham group, DMEM/F12 group injected with DMEM/F12, EGFP group with transplanted EGFP transfected BMSCs and Cx43 group with transplanted Cx43 transfected BMSCs. Myocardial infarction model was established by ligating anterior descending branch and the cells were transplanted after 30 minutes. At Week 4 post-infarction, the heart functions of rats were evaluated by echocardiography. After the rats were sacrificed, their tissue samples were collected. The areas of myocardial infarction and the levels of collagen fiber content were measured. And the expressions of EGFR and Cx43 were observed under laser confocal microscopy. The level of Cx43 mRNA was measured by reverse transcription-polymerase chain reaction (RT-PCR). RESULTS: As compared with the DMEM/F12 group, cardiac function was improved significantly, myocardial infarct area shrunk and collagen fiber content decreased significantly in the EGFP and group in Cx43 groups the. Survival of BMSCs and the formation of gap junction between BMSCs and the host myocardium could be observed under laser confocal microscopy both in EGFP group and Cx43 groups. And the post-infarction, expression of Cx43 mRNA in myocardial tissue decreased significantly in the group DMEM/F12, when compared with sham group (0.18 ± 0.05 vs 0.78 ± 0.14, P < 0.01). There was no significant difference on expression of Cx43 mRNA between DMEM/F12 group and EGFP group (0.18 ± 0.05 vs 0.20 ± 0.09, P > 0.05). The lever of Cx43 mRNA was higher in group Cx43 than in group DMEM/F12 and group EGFP(0.39 ± 0.14 vs 0.18 ± 0.05, P < 0.01; 0.39 ± 0.14 vs 0.20 ± 0.09, P < 0.05). CONCLUSION: Transplantation of BMSCs attenuates ventricular remodeling and improves cardiac functions. It may result from the over-expression of Cx43 gene through its effects of improving gap junction remodeling and increasing electro-mechanical coupling between myocardial cells in peri-infarct area.


Asunto(s)
Conexina 43/metabolismo , Insuficiencia Cardíaca/cirugía , Trasplante de Células Madre Mesenquimatosas , Animales , Células de la Médula Ósea/metabolismo , Uniones Comunicantes/metabolismo , Masculino , Células Madre Mesenquimatosas/metabolismo , Infarto del Miocardio/complicaciones , Infarto del Miocardio/metabolismo , Infarto del Miocardio/cirugía , Ratas , Ratas Sprague-Dawley , Transfección
18.
Cytokine ; 53(3): 320-6, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21177121

RESUMEN

BACKGROUND: CXCL16 has been shown to be involved in atherosclerotic lesion development, but its role in preexisting lesions is still unclear. This study aims to assess the effect of CXCL16 on the stability of preexisting lesions. METHODS: We firstly measured plasma CXCL16 level in Apolipoprotein E-Knockout (ApoE KO) mice with either high-cholesterol diet (HCD) or normal diet (ND) by enzyme-linked immunosorbent assay (ELISA). Then, silastic collars were placed around the carotid arteries in HCD-ApoE KO mice to accelerate atherosclerotic lesions. Five weeks later, CXCL16 was overexpressed by intravenous injection of lentivirus carrying CXCL16 transgene. Two weeks after infection, lesions were stained with hematoxylin and eosin (HE) and with oil red O. Biomarkers in the lesions, such as MMPs, CCL2, VCAM-1 and TNF-α were measured by real-time polymerase chain reaction (RT-PCR), which indicate the instability of plaques. RESULTS: The level of CXCL16 in plasma was higher in HCD-ApoE KO mice as compared to ND-ApoE KO mice. Circulating CXCL16 overexpression does not affect the size of preexisting plaques, but it leads to vulnerable plaque morphology and increases the expression of markers of plaque destabilization. CONCLUSION: Systemic CXCL16 becomes much higher in atherosclerosis, and it could be a potential atherogenic biomarker. Overexpression of CXCL16 promotes the evolution of preexisting lesions to vulnerable plaques in ApoE KO mice.


Asunto(s)
Apolipoproteínas E/deficiencia , Arterias Carótidas/metabolismo , Quimiocina CXCL6/sangre , Placa Aterosclerótica/sangre , Animales , Apolipoproteínas E/genética , Tirantes/efectos adversos , Arterias Carótidas/patología , Quimiocina CCL2/genética , Quimiocina CXCL16 , Quimiocina CXCL6/biosíntesis , Quimiocina CXCL6/genética , Colesterol en la Dieta/administración & dosificación , Colesterol en la Dieta/efectos adversos , Ensayo de Inmunoadsorción Enzimática , Expresión Génica , Masculino , Metaloproteinasas de la Matriz/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Placa Aterosclerótica/etiología , Placa Aterosclerótica/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Factor de Necrosis Tumoral alfa/genética , Molécula 1 de Adhesión Celular Vascular/genética
19.
J Huazhong Univ Sci Technolog Med Sci ; 30(6): 704-8, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21181358

RESUMEN

This study examined the effect of ischemia-reperfusion injury on the expression of Pim-3 gene in myocardial tissues and their underlying mechanism. Rat models of myocardial ischemia-reperfusion injury were established by ligating the left anterior descending coronary artery of the rats. A total of 30 SD male adult rats were randomly divided into 5 groups: group A (sham operation, n=6); group B (in which the rats were subjected to 15 min of ischemia by ligation of the left anterior descending coronary artery, n=6); group C (in which the rats received 30 min of ischemia, n=6), group D and group E (in which the left anterior descending coronary artery of the rats were ligated for 30 min and then reperfused for 30 min or 120 min, n=6 in each). The left ventricular tissues were removed immediately after the ischemia-reperfusion injury. Neonatal cardiomyocytes were cultured and treated with different concentrations of H(2)O(2) (0, 5, 10, 20 µmol/L) or tumor necrosis factor-α (TNF-α, 0, 1, 5, 10 ng/mL). The mRNA and protein expression of Pim-3 gene was determined by using RT-PCR, western blotting and immunohistochemistry. Additionally, neonatal cardiomyocytes were transfected with Pim-3 siRNA, and induced to develop apoptosis by using H(2)O(2). The results showed that normal myocardial tissues expressed a quantity of Pim-3 gene mRNA and protein. Ischemia-reperfusion injury could up-regulate the mRNA and protein expression of Pim-3 gene in myocardial tissues. Furthermore, H(2)O(2) but not TNF-α up-regulated the Pim-3 gene expression in cultured cardiomyocytes. And Pim-3 silencing failed to strengthen the H(2)O(2)-inducing apoptosis in cardiomyocytes. It was concluded that ischemia-reperfusion injury up-regulated the Pim-3 gene expression through oxidative stress signaling pathway in myocardial tissues.


Asunto(s)
Isquemia Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Animales Recién Nacidos , Masculino , Isquemia Miocárdica/fisiopatología , Estrés Oxidativo/fisiología , Proteínas Serina-Treonina Quinasas/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Regulación hacia Arriba
20.
J Zhejiang Univ Sci B ; 11(12): 895-904, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21121066

RESUMEN

OBJECTIVE: Angiogenic therapy is emerging as a potential strategy for the treatment of ischemic heart disease but is limited by a relatively short half-life of growth factors. Fibrin glue (FG) provides a reservoir for controlled-release of growth factors. The aim of this study was to evaluate the effects of basic fibroblast growth factor (bFGF) incorporating FG on angiogenesis and cardiac performance in a canine infarct model. METHODS: Acute myocardial infarction was induced by ligation of the left anterior descending coronary artery (LAD). Group I (n=6) underwent ligation of LAD alone. In Group II, transmural channels were created in the infarct area (n=6). In Group III, non-transmural channels were created to locate FG cylinders containing bFGF (n=6). Eight weeks after operation, myocardial perfusion was assessed by single photon emission computed tomography, cardiac function by echocardiography, and vascular development by immunohistochemical staining. RESULTS: Total vascular density and the number of large vessels (internal diameter ≥50 µm) were dramatically higher in Group III than in Groups I and II at eight weeks. Only the controlled-release group exhibited an improvement in regional myocardial perfusion associated with lower defect score. Animals in Group III presented improved cardiac regional systolic and diastolic functions as well as global systolic function in comparison with the other two groups. CONCLUSIONS: Enhanced and sustained angiogenic response can be achieved by controlled-release bFGF incorporating FG within transmyocardial laser channels, thus enabling improvement in myocardial perfusion and cardiac function.


Asunto(s)
Adhesivo de Tejido de Fibrina/administración & dosificación , Factor 2 de Crecimiento de Fibroblastos/administración & dosificación , Corazón/fisiopatología , Infarto del Miocardio/tratamiento farmacológico , Imagen de Perfusión Miocárdica , Animales , Preparaciones de Acción Retardada , Modelos Animales de Enfermedad , Perros , Factor VIII/análisis , Masculino , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/fisiopatología , Tomografía Computarizada de Emisión de Fotón Único
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA