Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Acta Pharmacol Sin ; 45(4): 728-737, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38086898

RESUMEN

Stimulation of adult cardiomyocyte proliferation is a promising strategy for treating myocardial infarction (MI). Earlier studies have shown increased CCL2 levels in plasma and cardiac tissue both in MI patients and mouse models. In present study we investigated the role of CCL2 in cardiac regeneration and the underlying mechanisms. MI was induced in adult mice by permanent ligation of the left anterior descending artery, we showed that the serum and cardiac CCL2 levels were significantly increased in MI mice. Intramyocardial injection of recombinant CCL2 (rCCL2, 1 µg) immediately after the surgery significantly promoted cardiomyocyte proliferation, improved survival rate and cardiac function, and diminished scar sizes in post-MI mice. Alongside these beneficial effects, we observed an increased angiogenesis and decreased cardiomyocyte apoptosis in post-MI mice. Conversely, treatment with a selective CCL2 synthesis inhibitor Bindarit (30 µM) suppressed both CCL2 expression and cardiomyocyte proliferation in P1 neonatal rat ventricle myocytes (NRVMs). We demonstrated in NRVMs that the CCL2 stimulated cardiomyocyte proliferation through STAT3 signaling: treatment with rCCL2 (100 ng/mL) significantly increased the phosphorylation levels of STAT3, whereas a STAT3 phosphorylation inhibitor Stattic (30 µM) suppressed rCCL2-induced cardiomyocyte proliferation. In conclusion, this study suggests that CCL2 promotes cardiac regeneration via activation of STAT3 signaling, underscoring its potential as a therapeutic agent for managing MI and associated heart failure.


Asunto(s)
Insuficiencia Cardíaca , Infarto del Miocardio , Humanos , Ratones , Animales , Ratas , Quimiocina CCL2/metabolismo , Infarto del Miocardio/metabolismo , Miocitos Cardíacos , Insuficiencia Cardíaca/metabolismo , Regeneración , Ratones Endogámicos C57BL , Apoptosis , Factor de Transcripción STAT3/metabolismo
2.
J Mol Cell Cardiol ; 185: 50-64, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37918322

RESUMEN

Better understanding of the mechanisms regulating the proliferation of pre-existing cardiomyocyte (CM) should lead to better options for regenerating injured myocardium. The absence of a perfect research model to definitively identify newly formed mammalian CMs is lacking. However, methodologies are being developed to identify and enrich proliferative CMs. These methods take advantages of the different proliferative states of CMs during postnatal development, before and after injury in the neonatal heart. New approaches use CMs labeled in lineage tracing animals or single cell technique-based CM clusters. This review aims to provide a timely update on the characteristics of the proliferative CMs, including their structural, functional, genetic, epigenetic and metabolic characteristics versus non-proliferative CMs. A better understanding of the characteristics of proliferative CMs should lead to the mechanisms for inducing endogenous CMs to self-renew, which is a promising therapeutic strategy to treat cardiac diseases that cause CM death in humans.


Asunto(s)
Cardiopatías , Miocitos Cardíacos , Animales , Recién Nacido , Humanos , Miocitos Cardíacos/metabolismo , Proliferación Celular , Corazón/fisiología , Miocardio , Mamíferos , Cardiopatías/metabolismo
3.
Acta Pharmacol Sin ; 44(6): 1149-1160, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36473990

RESUMEN

Heart failure with preserved ejection fraction (HFpEF) is highly prevalent, and lacks effective treatment. The aberration of WNT pathway underlies many pathological processes including cardiac fibrosis and hypertrophy, while porcupine is an acyltransferase essential for the secretion of WNT ligands. In this study we investigated the role of WNT signaling pathway in HFpEF as well as whether blocking WNT signaling by a novel porcupine inhibitor CGX1321 alleviated HFpEF. We established two experimental HFpEF mouse models, namely the UNX/DOCA model and high fat diet/L-NAME ("two-hit") model. The UNX/DOCA and "two-hit" mice were treated with CGX1321 (3 mg·kg-1·d-1) for 4 and 10 weeks, respectively. We showed that CGX1321 treatment significantly alleviated cardiac hypertrophy and fibrosis, thereby improving cardiac diastolic function and exercise performance in both models. Furthermore, both canonical and non-canonical WNT signaling pathways were activated, and most WNT proteins, especially WNT3a and WNT5a, were upregulated during the development of HEpEF in mice. CGX1321 treatment inhibited the secretion of WNT ligands and repressed both canonical and non-canonical WNT pathways, evidenced by the reduced phosphorylation of c-Jun and the nuclear translocation of ß-catenin and NFATc3. In an in vitro HFpEF model, MCM and ISO-treated cardiomyocytes, knockdown of porcupine by siRNA leads to a similar inhibitory effect on WNT pathways, cardiomyocyte hypertrophy and cardiac fibroblast activation as CGX1321 did, whereas supplementation of WNT3a and WNT5a reversed the anti-hypertrophy and anti-fibrosis effect of CGX1321. We conclude that WNT signaling activation plays an essential role in the pathogenesis of HFpEF, and porcupine inhibitor CGX1321 exerts a therapeutic effect on HFpEF in mice by attenuating cardiac hypertrophy, alleviating cardiac fibrosis and improving cardiac diastolic function.


Asunto(s)
Cardiomiopatías , Acetato de Desoxicorticosterona , Insuficiencia Cardíaca , Animales , Ratones , Cardiomegalia/patología , Cardiomiopatías/patología , Acetato de Desoxicorticosterona/farmacología , Acetato de Desoxicorticosterona/uso terapéutico , Fibrosis , Insuficiencia Cardíaca/metabolismo , Miocitos Cardíacos , Volumen Sistólico/fisiología , Vía de Señalización Wnt
4.
Biochem Biophys Res Commun ; 589: 267-274, 2022 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-34933200

RESUMEN

The deprivation of myocardial nutrition causes cardiomyocyte death and disturbance of energy metabolism. IKKε plays an important regulatory role in many biological events such as inflammation, redox reaction, cell death, etc. However, the more in-depth mechanism by which IKKε contributes to cardiomyocytes death in nutrition deprivation remains poorly understood. IKKε expression was knocked down by siRNA in H9c2 cells, and cells were cultured under starvation conditions to simulate ischemic conditions. Starvation triggered greater NLRP3 activation, accompanied by more IL-1ß, IL-18 and caspase-1 release in the siIKKε H9c2 cells compared with the control H9c2 cells. Western blot and immunofluorescence showed that the IKKε konckdown promoted NLRP3 expressions and ROS release under starvation conditions. Furthermore, electron micrography and JC-1 analysis revealed that IKKε konckdown resulted in aggravated mitochondrial damage and more mitochondrial ROS (mtROS) released in vitro. Notably, Western blot analysis showed that IKKε deficiency activated the TBK1 and IRF3 signaling pathways to promote pyroptosis in vitro. Collectively, our results indicate that IKKε protects against cardiomyocyte injury by reducing mitochondrial damage and NLRP3 expression following nutrition deprivation via regulation of the TBK1/IRF3 signaling pathway. This study further revealed the mechanism of IKKε in inflammation and myocardial nutrition deprivation.


Asunto(s)
Citoprotección , Quinasa I-kappa B/metabolismo , Inflamasomas/metabolismo , Mitocondrias/patología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piroptosis , Animales , Línea Celular , Técnicas de Silenciamiento del Gen , Quinasa I-kappa B/deficiencia , Factor 3 Regulador del Interferón/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas
5.
Basic Res Cardiol ; 117(1): 10, 2022 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-35247074

RESUMEN

The role of long non-coding RNA (lncRNA) in endogenous cardiac regeneration remains largely elusive. The mammalian cardiomyocyte is capable of regeneration for a brief period after birth. This fact allows the exploration of the roles of critical lncRNAs in the regulation of cardiac regeneration. Through a cardiac regeneration model by apical resection (AR) of the left ventricle in neonatal mice, we identified an lncRNA named natriuretic peptide A antisense RNA 1 (NPPA-AS1), which negatively regulated cardiomyocyte proliferation. In neonates, NPPA-AS1 deletion did not affect heart development, but was sufficient to prolong the postnatal window of regeneration after AR. In adult mice, NPPA-AS1 deletion improved cardiac function and reduced infarct size after myocardial infarction (MI), associated with a significant improvement in cardiomyocyte proliferation. Further analysis showed that NPPA-AS1 interacted with DNA repair-related molecule splicing factor, proline- and glutamine-rich (SFPQ). A heteromer of SFPQ and non-POU domain-containing octamer-binding protein (NONO) was required for double-strand DNA break repair, but NPPA-AS1 was competitively bound with SFPQ due to the overlapped binding sites of SFPQ and NONO. NPPA-AS1 deletion promoted the binding of SFPQ-NONO heteromer, decreased DNA damage, and activated cardiomyocyte cell cycle re-entry. Together, loss of NPPA-AS1 promoted cardiomyocyte proliferation by stabilizing SFPQ-NONO heteromer-induced DNA repair and exerted a therapeutic effect against MI in adult mice. Consequently, NPPA-AS1 may be a novel target for stimulating cardiac regeneration to treat MI.


Asunto(s)
Infarto del Miocardio , ARN Largo no Codificante , Animales , Factor Natriurético Atrial , Proliferación Celular , Reparación del ADN , Proteínas de Unión al ADN , Mamíferos , Ratones , Infarto del Miocardio/genética , Miocitos Cardíacos , Procainamida/análogos & derivados , ARN Largo no Codificante/genética , Proteínas de Unión al ARN , Regeneración
6.
BMC Cardiovasc Disord ; 22(1): 568, 2022 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-36572851

RESUMEN

BACKGROUND: GRACE risk score models are capable of predicting all-cause mortality of non-ST elevation myocardial infarction (NSTEMI) patients. However, its utility for evaluating major adverse cardiovascular events (MACE) in NSTEMI patients with multivessel disease (MVD) remains unclear. METHODS AND RESULTS: This study was designed as a retrospective cohort study that recruited patients with NSTEMI and multivessel disease between September 2013 and December 2018 in Daping Hospital, Chongqing, China. The primary outcome was a composite outcome that included all-cause mortality, recurrent angina, non-fatal myocardial infarction, coronary re-vascularization, and non-fatal strokes. Of the 827 patients with NSTEMI, 32 did not complete follow-up and 430 were excluded because of single-vessel disease. The remaining 365 NSTEMI patients with MVD had a median follow-up of 3.0 (IQR 2.6-3.3) years, 78 patients experienced outcomes. The GRACE risk score predicted the MACE (hazard ratio 1.014, 95% CI 1.006-1.021, P < 0.001). The GRACE risk score performed well in predicting all-cause mortality (c-statistic 0.72, 95% CI 0.59-0.85, P = 0.001) in MVD but was less powerful in predicting MACE (c-statistic 0.69, 95% CI 0.62-0.75, P < 0.001). When combining the GRACE risk score with the SYNTAX score, and blood urea nitrogen for predicting all-cause mortality and MACE events, the c-statistic value increased to 0.82 and 0.81 (P < 0.001). CONCLUSION: In NSTEMI patients with MVD, the GRACE score showed an acceptable predictive value for all-cause mortality, but it was less powerful in predicting MACE. Blood urea nitrogen may be valuable in assessing long-term cardiovascular events in patients with MVD.


Asunto(s)
Infarto del Miocardio , Infarto del Miocardio sin Elevación del ST , Humanos , Pronóstico , Infarto del Miocardio sin Elevación del ST/diagnóstico , Infarto del Miocardio sin Elevación del ST/terapia , Estudios Retrospectivos , Medición de Riesgo , Factores de Riesgo
7.
Eur Heart J ; 42(14): 1415-1430, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33280021

RESUMEN

AIMS: G protein-coupled receptor kinase 4 (GRK4) has been reported to play an important role in hypertension, but little is known about its role in cardiomyocytes and myocardial infarction (MI). The goal of present study is to explore the role of GRK4 in the pathogenesis and progression of MI. METHODS AND RESULTS: We studied the expression and distribution pattern of GRK4 in mouse heart after MI. GRK4 A486V transgenic mice, inducible cardiomyocyte-specific GRK4 knockout mice, were generated and subjected to MI with their control mice. Cardiac infarction, cardiac function, cardiomyocyte apoptosis, autophagic activity, and HDAC4 phosphorylation were assessed. The mRNA and protein levels of GRK4 in the heart were increased after MI. Transgenic mice with the overexpression of human GRK4 wild type (WT) or human GRK4 A486V variant had increased cardiac infarction, exaggerated cardiac dysfunction and remodelling. In contrast, the MI-induced cardiac dysfunction and remodelling were ameliorated in cardiomyocyte-specific GRK4 knockout mice. GRK4 overexpression in cardiomyocytes aggravated apoptosis, repressed autophagy, and decreased beclin-1 expression, which were partially rescued by the autophagy agonist rapamycin. MI also induced the nuclear translocation of GRK4, which inhibited autophagy by increasing HDAC4 phosphorylation and decreasing its binding to the beclin-1 promoter. HDAC4 S632A mutation partially restored the GRK4-induced inhibition of autophagy. MI caused greater impairment of cardiac function in patients carrying the GRK4 A486V variant than in WT carriers. CONCLUSION: GRK4 increases cardiomyocyte injury during MI by inhibiting autophagy and promoting cardiomyocyte apoptosis. These effects are mediated by the phosphorylation of HDAC4 and a decrease in beclin-1 expression.


Asunto(s)
Quinasa 4 del Receptor Acoplado a Proteína-G/fisiología , Infarto del Miocardio , Miocitos Cardíacos , Animales , Apoptosis , Autofagia , Beclina-1 , Histona Desacetilasas , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Remodelación Ventricular
8.
Int Heart J ; 63(5): 814-820, 2022 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-36104232

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is an emerging driver of cardiac arrhythmias. However, the relationship between NAFLD and malignant arrhythmia in non-ST-segment elevation myocardial infarction (NSTEMI) patients is still unclear.In this study, 358 NSTEMI inpatients were enrolled. They all received 24-hour Holter monitoring after percutaneous coronary intervention. All inpatients were divided into two groups: the non-NAFLD group (236 cases, 65.9%) and the NAFLD group (122 cases, 34.1%). Compared with the non-NAFLD group, the NAFLD group had a significantly higher incidence of PVCs/hour > 5 (premature ventricular complexes, 32.0% versus 9.3%, P < 0.001), ventricular tachycardia (VT, 22.1% versus 5.9%, P < 0.001), and sinus arrest (SA, 7.4% versus 1.3%, P = 0.002). We found that NAFLD was closely associated with the occurrence of VT [unadjusted odds ratio (OR) 4.507, 95% confidence interval (CI) 2.263-8.974, P < 0.001] and SA (OR 6.186, 95%CI 1.643-23.291, P = 0.007). After adjusting for age, sex, body mass index, and other confounding factors, the above differences were still statistically significant (VT: OR 4.808, 95%CI 2.254-10.253, P < 0.001; SA: OR 9.589, 95%CI 2.027-45.367, P = 0.004).NAFLD is associated with the occurrence of VT and SA in NSTEMI patients. It indicates that NAFLD might be a risk factor for malignant arrhythmias in post-NSTEMI patients.


Asunto(s)
Paro Cardíaco , Infarto del Miocardio sin Elevación del ST , Enfermedad del Hígado Graso no Alcohólico , Intervención Coronaria Percutánea , Infarto del Miocardio con Elevación del ST , Taquicardia Ventricular , Complejos Prematuros Ventriculares , Paro Cardíaco/complicaciones , Humanos , Infarto del Miocardio sin Elevación del ST/complicaciones , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Intervención Coronaria Percutánea/efectos adversos , Factores de Riesgo , Infarto del Miocardio con Elevación del ST/complicaciones , Taquicardia Ventricular/complicaciones , Taquicardia Ventricular/etiología , Complejos Prematuros Ventriculares/etiología
9.
Acta Pharmacol Sin ; 40(10): 1314-1321, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31061533

RESUMEN

Irisin, a myokine, is cleaved from the extracellular portion of fibronectin domain-containing 5 protein in skeletal muscle and myocardium and secreted into circulation as a hormone during exercise. Irisin has been found to exert protective effects against lung and heart injuries. However, whether irisin influences myocardial infarction (MI) remains unclear. In this study we investigated the therapeutic effects of irisin in an acute MI model and its underlying mechanisms. Adult C57BL/6 mice were subjected to ligation of the left anterior descending coronary artery and treated with irisin for 2 weeks after MI. Cardiac function was assessed using echocardiography. We found that irisin administration significantly alleviated MI-induced cardiac dysfunction and ventricular dilation at 4 weeks post-MI. Irisin significantly reduced infarct size and fibrosis in post-MI hearts. Irisin administration significantly increased angiogenesis in the infarct border zone and decreased cardiomyocyte apoptosis, but did not influence cardiomyocyte proliferation. In human umbilical vein endothelial cells (HUVEC), irisin significantly increased the phosphorylation of ERK, and promoted the migration of HUVEC detected in wound-healing and transwell chamber migration assay. The effects of irisin were blocked by the ERK inhibitor U0126. In conclusion, irisin improves cardiac function and reduces infarct size in post-MI mouse heart. The therapeutic effect is associated with its pro-angiogenic function through activating ERK signaling pathway.


Asunto(s)
Fibronectinas/metabolismo , Infarto del Miocardio/metabolismo , Neovascularización Patológica/metabolismo , Animales , Apoptosis/efectos de los fármacos , Butadienos/farmacología , Movimiento Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Fibronectinas/antagonistas & inhibidores , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/patología , Neovascularización Patológica/patología , Nitrilos/farmacología , Proteínas Recombinantes/metabolismo
10.
J Card Surg ; 34(11): 1208-1214, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31441553

RESUMEN

BACKGROUND: Proximal aortic dilatation is frequently associated with aortic valve pathology. The treatment of mild to moderate proximal aortic dilatation (maximal diameter: 40-50 mm) at the time of aortic valve replacement (AVR) is still controversial. We retrospectively analyzed the fate and progression of the proximal dilated aorta after isolated AVR in tricuspid aortic valve (TAV) patients, to determine if ascending aortic replacement (AAR) is recommended at the time of the initial AVR. METHODS: The review of our hospital database revealed a subgroup of 127 TAV disease patients with mild to moderate ascending aortic dilatation, who underwent isolated AVR (group I, n = 68) or AVR combined AAR (group II, n = 59) from January 2000 to December 2013. Follow-up was obtained through a telephone interview/outpatient interview. Adverse aortic events were defined as aortic dissection/ rupture, or diameter of proximal aorta ≥55 mm, or re-do aortic surgery contributable to the dilated aorta during follow-up. RESULTS: There were no differences in age, gender, heart function, hypertension, diabetes, smoking, chronic renal failure, and atrium fibrillation between two groups except for the maximum aortic diameter (group I 43.91 ± 2.0 vs group II 45.20 ± 2.63, P < .05). The cross-clamp time and cardiopulmonary bypass time was significantly less in group I than that in group II, owing to the replacement of the proximal aorta. A total of 126 patients were discharged home successfully, with 0.79% hospital mortality. There was no significant difference of hospital mortality and morbidity between the two groups. Follow-up was successfully obtained in 106 patients (84.13%). Mean follow-up time was (9.60 ± 3.47) years. The overall survival at 10-year follow-up was 72.46% ± 6.42% in group I versus 74.55% ± 6.87% in group II ( P = .73). The freedom from adverse aortic events at 10-year was 89.59% ± 4.02% in group I versus 96.88% ± 3.07% in group II ( P = .09). No significant difference in survival rate and freedom from adverse aortic events can be obtained between the two groups. CONCLUSION: Progression of proximal aorta leading to adverse aortic events after isolated AVR in TAV patients is infrequent. AVR alone is acceptable and reasonable in patients with mild to moderate proximal aortic dilatation if connective tissue disorders are not present.


Asunto(s)
Válvula Aórtica/cirugía , Implantación de Prótesis de Válvulas Cardíacas , Válvula Tricúspide/cirugía , Válvula Aórtica/patología , Dilatación Patológica , Humanos
11.
J Mol Cell Cardiol ; 123: 1-12, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30153439

RESUMEN

The IκB kinase (IKK) complex plays a well-documented role in cancer and immune system. This function has been widely attributed to its role as the master regulator of the NF-κB family. Particularly, IKKɑ, a member of IKK complex, is reported to have various regulating effects in inflammatory and malignant diseases. However, its role as well as its mechanism of function in macrophages following myocardial ischemia and reperfusion (I/R) injury remains unexplored. In vivo, sham or I/R operations were performed on macrophage-specific IKKɑ knockout (mIKKɑ-/-) mice and their IKKɑflox/flox littermates. We ligated the left anterior descending (LAD) coronary artery of I/R groups simulating ischemia for 30 min, followed by a reperfusion period of 3 days and 7 days, respectively. The hearts of mIKKɑ-/- mice exhibited significantly increased inflammation and macrophage aggregation as compared to their IKKɑflox/flox littermates. Moreover, in the mIKKɑ-/- group subjected to I/R macrophages had a tendency to polarize to M1 phenotype. In vitro, we stimulated RAW264.7 cells with Lipopolysaccharides (LPS) after infection by the lentivirus, either knocking-down or overexpressing IKKɑ. We discovered that a deficiency of IKKɑ in RAW264.7 caused increased expression of pro-inflammatory markers compared to normal RAW264.7 after LPS stimulation. Inversely, pro-inflammatory factors were inhibited with IKKɑ overexpression. Mechanistically, IKKɑ directly combined with RelB to regulate macrophage polarization. Furthermore, IKKɑ regulated MEK1/2-ERK1/2 and downstream p65 signaling cascades after LPS stimulation. Overall, our data reveals that IKKɑ is a novel mediator protecting against the development of myocardial I/R injury via negative regulation of macrophage polarization to M1 phenotype. Thus, IKKɑ may serve as a valuable therapeutic target for the treatment of myocardial I/R injury.


Asunto(s)
Quinasa I-kappa B/metabolismo , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Daño por Reperfusión Miocárdica/etiología , Daño por Reperfusión Miocárdica/metabolismo , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ecocardiografía , Expresión Génica , Quinasa I-kappa B/genética , Inmunohistoquímica , Inmunofenotipificación , Mediadores de Inflamación/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Daño por Reperfusión Miocárdica/diagnóstico , Miocardio/metabolismo , FN-kappa B/metabolismo , Células RAW 264.7 , Transducción de Señal
12.
Circulation ; 136(9): 834-848, 2017 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-28642276

RESUMEN

BACKGROUND: Adult mammalian hearts have a limited ability to generate new cardiomyocytes. Proliferation of existing adult cardiomyocytes (ACMs) is a potential source of new cardiomyocytes. Understanding the fundamental biology of ACM proliferation could be of great clinical significance for treating myocardial infarction (MI). We aim to understand the process and regulation of ACM proliferation and its role in new cardiomyocyte formation of post-MI mouse hearts. METHODS: ß-Actin-green fluorescent protein transgenic mice and fate-mapping Myh6-MerCreMer-tdTomato/lacZ mice were used to trace the fate of ACMs. In a coculture system with neonatal rat ventricular myocytes, ACM proliferation was documented with clear evidence of cytokinesis observed with time-lapse imaging. Cardiomyocyte proliferation in the adult mouse post-MI heart was detected by cell cycle markers and 5-ethynyl-2-deoxyuridine incorporation analysis. Echocardiography was used to measure cardiac function, and histology was performed to determine infarction size. RESULTS: In vitro, mononucleated and bi/multinucleated ACMs were able to proliferate at a similar rate (7.0%) in the coculture. Dedifferentiation proceeded ACM proliferation, which was followed by redifferentiation. Redifferentiation was essential to endow the daughter cells with cardiomyocyte contractile function. Intercellular propagation of Ca2+ from contracting neonatal rat ventricular myocytes into ACM daughter cells was required to activate the Ca2+-dependent calcineurin-nuclear factor of activated T-cell signaling pathway to induce ACM redifferentiation. The properties of neonatal rat ventricular myocyte Ca2+ transients influenced the rate of ACM redifferentiation. Hypoxia impaired the function of gap junctions by dephosphorylating its component protein connexin 43, the major mediator of intercellular Ca2+ propagation between cardiomyocytes, thereby impairing ACM redifferentiation. In vivo, ACM proliferation was found primarily in the MI border zone. An ischemia-resistant connexin 43 mutant enhanced the redifferentiation of ACM-derived new cardiomyocytes after MI and improved cardiac function. CONCLUSIONS: Mature ACMs can reenter the cell cycle and form new cardiomyocytes through a 3-step process: dedifferentiation, proliferation, and redifferentiation. Intercellular Ca2+ signal from neighboring functioning cardiomyocytes through gap junctions induces the redifferentiation process. This novel mechanism contributes to new cardiomyocyte formation in post-MI hearts in mammals.


Asunto(s)
Infarto del Miocardio/patología , Miocitos Cardíacos/citología , Animales , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Conexina 43/antagonistas & inhibidores , Conexina 43/genética , Conexina 43/metabolismo , Citocinesis , Ecocardiografía , Uniones Comunicantes/metabolismo , Corazón/diagnóstico por imagen , Humanos , Ratones , Ratones Transgénicos , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Interferencia de ARN , Ratas , Transducción de Señal , Troponina I/metabolismo
13.
Clin Sci (Lond) ; 131(24): 2919-2932, 2017 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-29162747

RESUMEN

After myocardial infarction (MI), the heart is difficult to repair because of great loss of cardiomyoctyes and lack of cardiac regeneration. Novel drug candidates that aim at reducing pathological remodeling and stimulating cardiac regeneration are highly desirable. In the present study, we identified if and how a novel porcupine inhibitor CGX1321 influenced MI and cardiac regeneration. Permanent ligation of left anterior descending (LAD) coronary artery was performed in mice to induce MI injury. Cardiac function was measured by echocardiography, infarct size was examined by TTC staining. Fibrosis was evaluated with Masson's trichrome staining and vimentin staining. As a result, CGX1321 administration blocked the secretion of Wnt proteins, and inhibited both canonical and non-canonical Wnt signaling pathways. CGX1321 improved cardiac function, reduced myocardial infarct size, and fibrosis of post-MI hearts. CGX1321 significantly increased newly formed cardiomyocytes in infarct border zone of post-MI hearts, evidenced by the increased EdU+ cardiomyocytes. Meanwhile, CGX1321 increased Ki67+ and phosphohistone H3 (PH3+) cardiomyocytes in culture, indicating enhanced cardiomyocyte proliferation. The mRNA microarray showed that CGX1321 up-regulated cell cycle regulating genes such as Ccnb1 and Ccne1 CGX1321 did not alter YAP protein phosphorylation and nuclear translocation in cardiomyocytes. In conclusion, porcupine inhibitor CGX1321 reduces MI injury by limiting fibrosis and promoting regeneration. It promotes cardiomyocyte proliferation by stimulating cell cycle regulating genes with a Hippo/YAP-independent pathway.


Asunto(s)
Aciltransferasas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Proteínas de la Membrana/antagonistas & inhibidores , Infarto del Miocardio/tratamiento farmacológico , Miocardio/enzimología , Miocitos Cardíacos/efectos de los fármacos , Regeneración/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Aciltransferasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Fibrosis , Antígeno Ki-67/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Infarto del Miocardio/enzimología , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/patología , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Fosfoproteínas/metabolismo , Fosforilación , Ratas , Factores de Tiempo , Regulación hacia Arriba , Proteínas Wnt/metabolismo , Proteína Wnt-5a/metabolismo , Proteínas Señalizadoras YAP
14.
Tumour Biol ; 2016 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-27658778

RESUMEN

A disintegrin and metalloproteinase (ADAM) family members are known to process the target membrane-bound molecules through the quick induction of their protease activities under interaction with other molecules, which have diverse roles in tissue morphogenesis and pathophysiological remodeling. Among these, ADAM17 is a membrane-bound protease that sheds the extracellular domain of various receptors or its ligands from the cell membrane and subsequently activates downstream signaling transduction pathways. Importantly, breast cancer remains a mainspring of cancer-induced death in women, and numerous regulatory pathways have been implicated in the formation of breast cancer. Substantial evidence has demonstrated that an obvious increased in ADAM17 cell surface expression has been discovered in breast cancer and was shown to be associated with mammary tumorigenesis, invasiveness, and drug resistance. Over the last decades, it has received more than its share of attention that ADAM17 plays a potential role in breast cancer, including cell proliferation, invasion, angiogenesis, apoptosis, and trastuzumab resistance. In our review, we discuss the mechanisms through which ADAM17 acts on breast cancer tumorigenesis and progression. Thus, this will provide further impetus for exploiting ADAM17 as a new target for breast cancer treatment.

15.
J Pathol ; 236(3): 360-72, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25726944

RESUMEN

Growth/differentiation factor 1 (GDF1) is a secreted glycoprotein of the transforming growth factor-ß (TGF-ß) superfamily that mediates cell differentiation events during embryonic development. GDF1 is expressed in several tissues, including the heart. However, the functional role of GDF1 in myocardial infarction (MI)-induced cardiac remodelling and dysfunction is not known. Here, we performed gain-of-function and loss-of-function studies using cardiac-specific GDF1 transgenic (TG) and knockout (KO) mice to determine the role of GDF1 in the pathogenesis of functional and architectural cardiac remodelling after MI, which was induced by surgical left anterior descending coronary artery ligation. Our results demonstrate that overexpression of GDF1 in the heart causes a significant decrease in MI-derived mortality post-MI and leads to attenuated infarct size expansion, left ventricular (LV) dilatation, and cardiac dysfunction at 1 week and 4 weeks after MI injury. Compared with control animals, cardiomyocyte apoptosis, inflammation, hypertrophy, and interstitial fibrosis were all remarkably reduced in the GDF1-TG mice following MI. In contrast, GDF1 deficiency greatly exacerbated the pathological cardiac remodelling response after infarction. Further analysis of the in vitro and in vivo signalling events indicated that the beneficial role of GDF1 in MI-induced cardiac dysfunction and LV remodelling was associated with the inhibition of non-canonical (MEK-ERK1/2) and canonical (Smad) signalling cascades. Overall, our data reveal that GDF1 in the heart is a novel mediator that protects against the development of post-infarction cardiac remodelling via negative regulation of the MEK-ERK1/2 and Smad signalling pathways. Thus, GDF1 may serve as a valuable therapeutic target for the treatment of MI.


Asunto(s)
Regulación de la Expresión Génica , Factor 1 de Diferenciación de Crecimiento/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Infarto del Miocardio/fisiopatología , Remodelación Ventricular , Animales , Apoptosis , Fibrosis , Factor 1 de Diferenciación de Crecimiento/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fenotipo , Proteínas Smad/genética , Proteínas Smad/metabolismo , Organismos Libres de Patógenos Específicos , Regulación hacia Arriba
16.
J Cell Physiol ; 230(9): 2128-41, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25656550

RESUMEN

Mitochondrial (mt) dysfunction and oxidative stress are involved in the pathogenesis of ischemia/reperfusion (I/R)-injury. Lycopene, a lipophilic antioxidant found mainly in tomatoes and in other vegetables and fruits, can protect mtDNA against oxidative damage. However, the role of mtDNA in myocardial I/R-injury is unclear. In the present study, we aimed to determine if and how lycopene protects cardiomyocytes from I/R-injury. In both in vitro and in vivo studies, I/R-injury increased mt 8-hydroxyguanine (8-OHdG) content, decreased mtDNA content and mtDNA transcription levels, and caused mitochondrial dysfunction in cardiomyocytes. These effects of I/R injury on cardiomycoytes were blocked by pre-treatment with lycopene. MtDNA depletion alone was sufficient to induce cardiomyocyte death. I/R-injury decreased the protein level of a key activator of mt transcription, mitochondrial transcription factor A (Tfam), which was blocked by lycopene. The protective effect of lycopene on mtDNA was associated with a reduction in mitochondrial ROS production and stabilization of Tfam. In conclusion, lycopene protects cardiomyocytes from the oxidative damage of mtDNA induced by I/R-injury.


Asunto(s)
Carotenoides/administración & dosificación , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Miocitos Cardíacos/patología , Estrés Oxidativo/efectos de los fármacos , Animales , Antioxidantes/administración & dosificación , Daño del ADN/efectos de los fármacos , ADN Mitocondrial/efectos de los fármacos , ADN Mitocondrial/genética , Licopeno , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/efectos de los fármacos , Oxidación-Reducción , Ratas
17.
Biochem Biophys Res Commun ; 467(4): 841-6, 2015 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-26474702

RESUMEN

Farnesoid X receptor (FXR) is a member of the nuclear receptor superfamily, which plays important roles in bile acids/lipid homeostasis and inflammation. Monocyte chemoattractant protein-1 (MCP-1) contributes to macrophage infiltration into body tissues during inflammation. Here we investigated whether FXR can regulate MCP-1 expression in murine macrophage. FXR activation down regulate MCP-1 mRNA and protein levels in ANA-1 and Raw264.7 cells. Luciferase reporter assay, Gel shift and Chromatin immunoprecipitation assays have revealed that the activated FXR bind to the FXR element located in -738 bp âˆ¼  -723 bp in MCP-1 promoter. These results suggested that FXR may serve as a novel target for regulating MCP-1 levels for the inflammation related diseases therapies.


Asunto(s)
Quimiocina CCL2/metabolismo , Macrófagos/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Sitios de Unión , Línea Celular/efectos de los fármacos , Quimiocina CCL2/genética , Ácido Quenodesoxicólico/farmacología , Regulación hacia Abajo , Macrófagos/efectos de los fármacos , Ratones , Regiones Promotoras Genéticas , Receptores Citoplasmáticos y Nucleares/genética
18.
Basic Res Cardiol ; 110(6): 56, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26463597

RESUMEN

Caspase activation and recruitment domain 3 (CARD3) is a caspase recruitment domain (CARD)-containing serine/threonine kinase and plays a pivotal role in apoptosis, immunity, tissue development and proliferation. To date, the causal relationship between CARD3 and myocardial infarction (MI) remains largely unexplored. This study aimed to identify the functional significance of CARD3 in the regulation of cardiac remodelling after MI and the underlying mechanisms of its effects. The levels of CARD3 expression were up-regulated in failing human and mouse post-infarction hearts. In addition, CARD3-knockout (KO) mice and transgenic mice overexpressing CARD3 in the heart were then generated and subjected to MI. Compared with wild-type (WT) control mice, CARD3-KO mice developed smaller infarct sizes, improved survival rates, and preserved left ventricle (LV) function after MI. Significantly, CARD3-KO hearts had less cardiomyocyte apoptosis and inflammatory cell infiltration in the infarct border zone. Attenuated LV remodelling was also observed in the KO hearts following MI, with reduced cardiac hypertrophy and fibrosis. Conversely, CARD3 overexpression resulted in the opposite MI-induced phenotype. Similar results were observed in ex vivo-cultured neonatal rat cardiomyocytes exposed to hypoxia. Mechanistically, we discovered that the CARD3-mediated detrimental effects of MI were associated with the activation of the NF-κB and p38 signalling cascades. Taken together, these data demonstrate that CARD3 serves as a novel positive modulator of ventricular remodelling after MI via the regulation of the NF-κB and p38 signalling. Thus, CARD3 may be a promising therapeutic target for the treatment of heart failure after MI.


Asunto(s)
Infarto del Miocardio/metabolismo , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Remodelación Ventricular , Animales , Apoptosis , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Inflamación/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones Endogámicos C57BL , Ratones Transgénicos , Infarto del Miocardio/mortalidad , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , FN-kappa B/metabolismo , Ratas Sprague-Dawley , Regulación hacia Arriba
19.
Basic Res Cardiol ; 110(3): 25, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25840773

RESUMEN

Dickkopf-3 (DKK3) is a secreted glycoprotein of the Dickkopf family (DKK1-4) that modulates Wnt signalling. DKK3 has been reported to regulate cell development, proliferation, apoptosis, and immune response. However, the functional role of DKK3 in cardiac remodelling after myocardial infarction (MI) has not yet been elucidated. This study aimed to explore the functional significance of DKK3 in the regulation of post-MI remodelling and its underlying mechanisms. MI was induced by surgical left anterior descending coronary artery ligation in transgenic mice expressing cardiac-specific DKK3 and DKK3 knockout (KO) mice as well as their non-transgenic and DKK3(+/+) littermates. Our results demonstrated that after MI, mice with DKK3 deficiency had increased mortality, greater infarct size, and exacerbated left ventricular (LV) dysfunction. Significantly, at 1 week post-MI, the hearts of DKK3-KO mice exhibited increased apoptosis, inflammation, and LV remodelling compared with the hearts of their DKK3(+/+) littermates. Conversely, DKK3 overexpression led to the opposite phenotype after infarction. Similar results were observed in cultured neonatal rat cardiomyocytes exposed to hypoxia in vitro. Mechanistically, DKK3 promotes cardioprotection by interrupting the ASK1-JNK/p38 signalling cascades. In conclusion, our results indicate that DKK3 protects against the development of MI-induced cardiac remodelling via negative regulation of the ASK1-JNK/p38 signalling pathway. Thus, our study suggests that DKK3 may represent a potential therapeutic target for the treatment of heart failure after MI.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Remodelación Ventricular/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Apoptosis/fisiología , Western Blotting , Quimiocinas , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Humanos , Etiquetado Corte-Fin in Situ , Ratones , Ratones Noqueados , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/fisiología
20.
Clin Sci (Lond) ; 129(3): 259-69, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25783200

RESUMEN

We set out to investigate whether and how SRY (sex-determining region, Y) DNAs in plasma EVs (extracellular vesicles) is involved in the pathogenesis of atherosclerosis. PCR and gene sequencing found the SRY gene fragment in plasma EVs from male, but not female, patients; EVs from male patients with CAD (coronary artery disease) had a higher SRY GCN (gene copy number) than healthy subjects. Additional studies found that leucocytes, the major source of plasma EVs, had higher SRY GCN and mRNA and protein expression in male CAD patients than controls. After incubation with EVs from SRY-transfected HEK (human embryonic kidney)-293 cells, monocytes (THP-1) and HUVECs (human umbilical vein endothelial cells), which do not endogenously express SRY protein, were found to express newly synthesized SRY protein. This resulted in an increase in the adherence factors CD11-a in THP-1 cells and ICAM-1 (intercellular adhesion molecule 1) in HUVECs. EMSA showed that SRY protein increased the promoter activity of CD11-a in THP-1 cells and ICAM-1 in HUVECs. There was an increase in THP-1 cells adherent to HUVECs after incubation with SRY-EVs. SRY DNAs transferred from EVs have pathophysiological significance in vivo; injection of SRY EVs into ApoE-/- (apolipoprotein-knockout) mice accelerated atherosclerosis. The SRY gene in plasma EVs transferred to vascular endothelial cells may play an important role in the pathogenesis of atherosclerosis; this mechanism provides a new approach to the understanding of inheritable CAD in men.


Asunto(s)
Aterosclerosis/genética , Aterosclerosis/metabolismo , Células Endoteliales/metabolismo , Leucocitos/metabolismo , Proteína de la Región Y Determinante del Sexo/metabolismo , Animales , Adhesión Celular , Células Cultivadas , Células Endoteliales/citología , Endotelio Vascular/metabolismo , Espacio Extracelular/metabolismo , Humanos , Leucocitos/citología , Masculino , Ratones , Proteína de la Región Y Determinante del Sexo/genética , Molécula 1 de Adhesión Celular Vascular/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA