Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Med ; 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38806989

RESUMEN

Cardiac fibrosis caused by ventricular remodeling and dysfunction such as post-myocardial infarction (MI) can lead to heart failure. RNA N6-methyladenosine (m6A) methylation has been shown to play a pivotal role in the occurrence and development of many illnesses. In investigating the biological function of the m6A reader YTHDF1 in cardiac fibrosis, adeno-associated virus 9 was used to knock down or overexpress the YTHDF1 gene in mouse hearts, and MI surgery in vivo and transforming growth factor-ß (TGF-ß)-activated cardiac fibroblasts in vitro were performed to establish fibrosis models. Our results demonstrated that silencing YTHDF1 in mouse hearts can significantly restore impaired cardiac function and attenuate myocardial fibrosis, whereas YTHDF1 overexpression could further enhance cardiac dysfunction and aggravate the occurrence of ventricular pathological remodeling and fibrotic development. Mechanistically, zinc finger BED-type containing 6 mediated the transcriptional function of the YTHDF1 gene promoter. YTHDF1 augmented AXL translation and activated the TGF-ß-Smad2/3 signaling pathway, thereby aggravating the occurrence and development of cardiac dysfunction and myocardial fibrosis. Consistently, our data indicated that YTHDF1 was involved in activation, proliferation, and migration to participate in cardiac fibrosis in vitro. Our results revealed that YTHDF1 could serve as a potential therapeutic target for myocardial fibrosis.

2.
Adv Sci (Weinh) ; : e2308769, 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38810124

RESUMEN

Cardiac hypertrophy is a key factor driving heart failure (HF), yet its pathogenesis remains incompletely elucidated. Mettl1-catalyzed RNA N7-methylguanosine (m7G) modification has been implicated in ischemic cardiac injury and fibrosis. This study aims to elucidate the role of Mettl1 and the mechanism underlying non-ischemic cardiac hypertrophy and HF. It is found that Mettl1 is upregulated in human failing hearts and hypertrophic murine hearts following transverse aortic constriction (TAC) and Angiotensin II (Ang II) infusion. YY1 acts as a transcriptional factor for Mettl1 during cardiac hypertrophy. Mettl1 knockout alleviates cardiac hypertrophy and dysfunction upon pressure overload from TAC or Ang II stimulation. Conversely, cardiac-specific overexpression of Mettl1 results in cardiac remodeling. Mechanically, Mettl1 increases SRSF9 expression by inducing m7G modification of SRSF9 mRNA, facilitating alternative splicing and stabilization of NFATc4, thereby promoting cardiac hypertrophy. Moreover, the knockdown of SRSF9 protects against TAC- or Mettl1-induced cardiac hypertrophic phenotypes in vivo and in vitro. The study identifies Mettl1 as a crucial regulator of cardiac hypertrophy, providing a novel therapeutic target for HF.

3.
Redox Biol ; 72: 103145, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38583415

RESUMEN

Ferroptosis is a nonapoptotic form of regulated cell death that has been reported to play a central role in cardiac ischemia‒reperfusion (I/R) injury. N-acetyltransferase 10 (NAT10) contributes to cardiomyocyte apoptosis by functioning as an RNA ac4c acetyltransferase, but its role in cardiomyocyte ferroptosis during I/R injury has not been determined. This study aimed to elucidate the role of NAT10 in cardiac ferroptosis as well as the underlying mechanism. The mRNA and protein levels of NAT10 were increased in mouse hearts after I/R and in cardiomyocytes that were exposed to hypoxia/reoxygenation. P53 acted as an endogenous activator of NAT10 during I/R in a transcription-dependent manner. Cardiac overexpression of NAT10 caused cardiomyocyte ferroptosis to exacerbate I/R injury, while cardiomyocyte-specific knockout of NAT10 or pharmacological inhibition of NAT10 with Remodelin had the opposite effects. The inhibition of cardiomyocyte ferroptosis by Fer-1 exerted superior cardioprotective effects against the NAT10-induced exacerbation of post-I/R cardiac damage than the inhibition of apoptosis by emricasan. Mechanistically, NAT10 induced the ac4C modification of Mybbp1a, increasing its stability, which in turn activated p53 and subsequently repressed the transcription of the anti-ferroptotic gene SLC7A11. Moreover, knockdown of Mybbp1a partially abolished the detrimental effects of NAT10 overexpression on cardiomyocyte ferroptosis and cardiac I/R injury. Collectively, our study revealed that p53 and NAT10 interdependently cooperate to form a positive feedback loop that promotes cardiomyocyte ferroptosis to exacerbate cardiac I/R injury, suggesting that targeting the NAT10/Mybbp1a/p53 axis may be a novel approach for treating cardiac I/R.


Asunto(s)
Ferroptosis , Daño por Reperfusión Miocárdica , Miocitos Cardíacos , Proteína p53 Supresora de Tumor , Animales , Humanos , Masculino , Ratones , Acetiltransferasas/metabolismo , Acetiltransferasas/genética , Apoptosis , Modelos Animales de Enfermedad , Retroalimentación Fisiológica , Ferroptosis/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética
4.
Artículo en Inglés | MEDLINE | ID: mdl-37548549

RESUMEN

Aims: Myocardial ischemia-reperfusion (I/R) injury facilitates cardiomyocyte death and endangers human health. N6-methyladenosine (m6A) methylation plays a critical role in cardiovascular diseases. The m6A reader YTHDF2 identifies m6A-modified RNA and promotes target RNA degradation. Hence, we hypothesized that YTHDF2 affects I/R injury by regulating RNA stability. Results: Both messenger RNA (mRNA) and protein levels of YTHDF2 were upregulated in I/R mice and hypoxia-reoxygenation (H/R)-induced cardiomyocytes. Silencing endogenous YTHDF2 abrogated cardiac dysfunction and lowered the infarct size in I/R mice, and the forced expression of YTHDF2 aggravated these adverse pathological processes. Consistently, the protective effect of silencing YTHDF2 occurred in cardiomyocytes exposed to H/R and erastin. Further, RNA-Seq and RNA-binding protein immunoprecipitation (RIP) revealed that YTHDF2 recognized the m6A modification sites of the ferroptosis-related gene solute carrier family 7 member 11 (SLC7A11) mRNA to promote its degradation both in vivo and in vitro. Inhibition of SLC7A11 impaired cardiac function, increased infarct size, and the release of lactate dehydrogenase (LDH) in I/R mice after silencing YTHDF2. The beneficial effects of si-YTHDF2 on H/R injury were reversed by co-transfection with SLC7A11-specific siRNA (si-SLC7A11), which substantially exacerbated ferroptosis and the production of reactive oxygen species. Innovation and Conclusion: The cardioprotective effects of silencing YTHDF2 are accomplished by increasing SLC7A11 stability and expression, reducing ferroptosis, and providing novel potential therapeutic targets for treating ischemic cardiac diseases.

5.
Sci China Life Sci ; 66(12): 2786-2804, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37450238

RESUMEN

Ischemic heart failure (HF) remains a leading cause of morbidity and mortality. Maintaining homeostasis of cardiac function and preventing cardiac remodeling deterioration are critical to halting HF progression. Methyltransferase-like protein 13 (Mettl13) has been shown to regulate protein translation efficiency by acting as a protein lysine methyltransferase, but its role in cardiac pathology remains unexplored. This study aims to characterize the roles and mechanisms of Mettl13 in cardiac contractile function and HF. We found that Mettl13 was downregulated in the failing hearts of mice post-myocardial infarction (MI) and in a cellular model of oxidative stress. Cardiomyocyte-specific overexpression of Mettl13 mediated by AAV9-Mettl13 attenuated cardiac contractile dysfunction and fibrosis in response to MI, while silencing of Mettl13 impaired cardiac function in normal mice. Moreover, Mettl13 overexpression abrogated the reduction in cell shortening, Ca2+ transient amplitude and SERCA2a protein levels in the cardiomyocytes of adult mice with MI. Conversely, knockdown of Mettl13 impaired the contractility of cardiomyocytes, and decreased Ca2+ transient amplitude and SERCA2a protein expression in vivo and in vitro. Mechanistically, Mettl13 impaired the stability of c-Cbl by inducing lysine methylation of c-Cbl, which in turn inhibited ubiquitination-dependent degradation of SERCA2a. Furthermore, the inhibitory effects of knocking down Mettl13 on SERCA2a protein expression and Ca2+ transients were partially rescued by silencing c-Cbl in H2O2-treated cardiomyocytes. In conclusion, our study uncovers a novel mechanism that involves the Mettl13/c-Cbl/SERCA2a axis in regulating cardiac contractile function and remodeling, and identifies Mettl13 as a novel therapeutic target for ischemic HF.


Asunto(s)
Insuficiencia Cardíaca , Peróxido de Hidrógeno , Ratones , Animales , Peróxido de Hidrógeno/metabolismo , Insuficiencia Cardíaca/etiología , Miocitos Cardíacos/metabolismo , Ubiquitinación , Metiltransferasas/genética
6.
Signal Transduct Target Ther ; 8(1): 121, 2023 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-36967385

RESUMEN

Heart failure (HF) patients in general have a higher risk of developing cancer. Several animal studies have indicated that cardiac remodeling and HF remarkably accelerate tumor progression, highlighting a cause-and-effect relationship between these two disease entities. Targeting ferroptosis, a prevailing form of non-apoptotic cell death, has been considered a promising therapeutic strategy for human cancers. Exosomes critically contribute to proximal and distant organ-organ communications and play crucial roles in regulating diseases in a paracrine manner. However, whether exosomes control the sensitivity of cancer to ferroptosis via regulating the cardiomyocyte-tumor cell crosstalk in ischemic HF has not yet been explored. Here, we demonstrate that myocardial infarction (MI) decreased the sensitivity of cancer cells to the canonical ferroptosis activator erastin or imidazole ketone erastin in a mouse model of xenograft tumor. Post-MI plasma exosomes potently blunted the sensitivity of tumor cells to ferroptosis inducers both in vitro in mouse Lewis lung carcinoma cell line LLC and osteosarcoma cell line K7M2 and in vivo with xenograft tumorigenesis model. The expression of miR-22-3p in cardiomyocytes and plasma-exosomes was significantly upregulated in the failing hearts of mice with chronic MI and of HF patients as well. Incubation of tumor cells with the exosomes isolated from post-MI mouse plasma or overexpression of miR-22-3p alone abrogated erastin-induced ferroptotic cell death in vitro. Cardiomyocyte-enriched miR-22-3p was packaged in exosomes and transferred into tumor cells. Inhibition of cardiomyocyte-specific miR-22-3p by AAV9 sponge increased the sensitivity of cancer cells to ferroptosis. ACSL4, a pro-ferroptotic gene, was experimentally established as a target of miR-22-3p in tumor cells. Taken together, our findings uncovered for the first time that MI suppresses erastin-induced ferroptosis through releasing miR-22-3p-enriched exosomes derived from cardiomyocytes. Therefore, targeting exosome-mediated cardiomyocyte/tumor pathological communication may offer a novel approach for the ferroptosis-based antitumor therapy.


Asunto(s)
Exosomas , Ferroptosis , Insuficiencia Cardíaca , MicroARNs , Infarto del Miocardio , Neoplasias , Humanos , Ratones , Animales , Miocitos Cardíacos/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Ferroptosis/genética , Exosomas/metabolismo , Infarto del Miocardio/genética , Neoplasias/metabolismo , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología
7.
Transl Res ; 257: 30-42, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36775059

RESUMEN

Cardiac fibrosis is a common pathological change in the development of heart disease. Circular RNA (circRNA) has been shown to be related to the occurrence and development of various cardiovascular diseases. This study aimed to evaluate the effects and potential mechanisms of circHelz in cardiac fibrosis. Knockdown of circHelz alleviated cardiac fibrosis and myocardial fibroblast activation induced by myocardial infarction (MI) or angiotensin II (AngII) in vivo and transforming growth factor-ß (TGF-ß) in vitro. Overexpression of circHelz exacerbated cell proliferation and differentiation. Mechanistically, nuclear factor of activated T cells, cytoplasmic 2 (NFATc2) was found to act as a transcriptional activator to upregulate the expression of circHelz. The increased circHelz was demonstrated to bind to Yes-associated protein (YAP) and facilitate its localization in the nucleus to promote cell proliferation and growth. Moreover, silencing YAP1 reversed the detrimental effects caused by circHelz in vitro, as indicated by the observed decreases in cell viability, fibrotic marker expression levels, proliferation and migration. Collectively, the protective effect of circHelz knockdown against cardiac fibrosis injury is accomplished by inhibiting the nuclear translocation of YAP1. Thus, circHelz may be a novel target for the prevention and treatment of cardiovascular disease.


Asunto(s)
Infarto del Miocardio , ARN Circular , Humanos , ARN Circular/genética , ARN Circular/metabolismo , Miocardio/patología , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Fibrosis , Diferenciación Celular , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Fibroblastos/patología , Factor de Crecimiento Transformador beta1/metabolismo
8.
J Mol Cell Cardiol ; 158: 128-139, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34043986

RESUMEN

Myocardial infarction (MI)-induced the activation of NLRP3 inflammasome has been well known to aggravate myocardial injury and cardiac dysfunction by causing inflammation and pyroptosis in the heart. Circular RNAs (circRNAs) have been demonstrated to play critical roles in cardiovascular diseases. However, the functions and mechanisms of circRNAs in modulating cardiac inflammatory response and cardiomyocyte pyroptosis remain largely unknown. We revealed that circHelz, a novel circRNA transcribed from the helicase with zinc finger (Helz) gene, was significantly upregulated in both the ischemic myocardium of MI mouse and neonatal mouse ventricular cardiomyocytes (NMVCs) exposed to hypoxia. Overexpression of circHelz caused cardiomyocyte injury in NMVCs by activating the NLRP3 inflammasome and inducing pyroptosis, while circHelz silencing reduced these effects induced by hypoxia. Furthermore, knockdown of circHelz remarkably attenuated NLRP3 expression, decreased myocardial infarct size, pyroptosis, inflammation, and increased cardiac function in vivo after MI. Overexpression of miR-133a-3p in cardiomyocytes greatly prevented pyroptosis in the presence of hypoxia or circHelz by targeting NLRP3 in NMVCs. Mechanistically, circHelz functioned as an endogenous sponge for miR-133a-3p via suppressing its activity. Overall, our results demonstrate that circHelz causes myocardial injury by triggering the NLRP3 inflammasome-mediated pro-inflammatory response and subsequent pyroptosis in cardiomyocytes by inhibiting miR-133a-3p function. Therefore, interfering with circHelz/miR-133a-3p/NLRP3 axis might be a promising therapeutic approach for ischemic cardiac diseases.


Asunto(s)
Silenciador del Gen , Inflamasomas/metabolismo , MicroARNs/metabolismo , Infarto del Miocardio/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , ARN Helicasas/genética , ARN Circular/metabolismo , Transducción de Señal/genética , Animales , Animales Recién Nacidos , Hipoxia de la Célula , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Infarto del Miocardio/genética , Miocitos Cardíacos/metabolismo , Piroptosis/genética , ARN Circular/genética , Transfección , Regulación hacia Arriba
9.
Front Cell Dev Biol ; 9: 762853, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35004673

RESUMEN

N6-methyladenosine (m6A) methylation in RNA is a dynamic and reversible modification regulated by methyltransferases and demethylases, which has been reported to participate in many pathological processes of various diseases, including cardiac disorders. This study was designed to investigate an m6A writer Mettl14 on cardiac ischemia-reperfusion (I/R) injury and uncover the underlying mechanism. The m6A and Mettl14 protein levels were increased in I/R hearts and neonatal mouse cardiomyocytes upon oxidative stress. Mettl14 knockout (Mettl14+/-) mice showed pronounced increases in cardiac infarct size and LDH release and aggravation in cardiac dysfunction post-I/R. Conversely, adenovirus-mediated overexpression of Mettl14 markedly reduced infarct size and apoptosis and improved cardiac function during I/R injury. Silencing of Mettl14 alone significantly caused a decrease in cell viability and an increase in LDH release and further exacerbated these effects in the presence of H2O2, while overexpression of Mettl14 ameliorated cardiomyocyte injury in vitro. Mettl14 resulted in enhanced levels of Wnt1 m6A modification and Wnt1 protein but not its transcript level. Furthermore, Mettl14 overexpression blocked I/R-induced downregulation of Wnt1 and ß-catenin proteins, whereas Mettl14+/- hearts exhibited the opposite results. Knockdown of Wnt1 abrogated Mettl14-mediated upregulation of ß-catenin and protection against injury upon H2O2. Our study demonstrates that Mettl14 attenuates cardiac I/R injury by activating Wnt/ß-catenin in an m6A-dependent manner, providing a novel therapeutic target for ischemic heart disease.

10.
Int Immunopharmacol ; 80: 106116, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31978804

RESUMEN

The myocardial inflammatory response is a consequence of myocardial infarction (MI), which may deteriorate cardiac remodeling and lead to dysfunction in the heart post-MI. Dectin-1 is a c-type lectin, which has been shown to regulate innate immune responses to pathogens. However, the role of Dectin-1 in the heart diseases remains largely unknown. In this study, we aimed to investigate the effects of Dectin-1 on cardiac remodeling post-MI. We found that cardiac Dectin-1 mRNA and protein expressions were significantly elevated in C57BL/6 mice after MI. In vitro, hypoxia induced cardiomyocyte injury in parallel with increased Dectin-1 protein expression. Knockdown of Dectin-1 remarkably attenuated cardiomyocyte death under hypoxia and lipopolysaccharide (LPS) stimulation. In vivo administration of adeno-associated virus serotype 9 mediated silencing of Dectin-1, which significantly decreased cardiac fibrosis, dilatation, and improved cardiac function in the mice post-MI. At the molecular level, downregulation of Dectin-1 dramatically suppressed up-regulation of nuclear factor-κB (NF-κB), nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3), and the inflammatory genes involved in fibrogenesis and cardiac remodeling after MI. Furthermore, treatment with BAY11-7082, an inhibitor of NF-κB, repressed the activation of NF-κB, and attenuated LPS induced elevation of NLRP3 and cell death in cardiomyocytes. Collectively, upregulation of Dectin-1 in cardiomyocytes post-MI contributes to cardiac remodeling and cardiac dysfunction at least partially by activating NF-κB and NLRP3. This study identified Dectin-1 as a promising therapeutic target for ischemic heart disease.


Asunto(s)
Lectinas Tipo C/metabolismo , Infarto del Miocardio/inmunología , Transducción de Señal/inmunología , Remodelación Ventricular/inmunología , Animales , Modelos Animales de Enfermedad , Regulación hacia Abajo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Lectinas Tipo C/genética , Lipopolisacáridos/inmunología , Masculino , Ratones , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/patología , Miocardio/citología , Miocardio/inmunología , Miocardio/patología , Miocitos Cardíacos , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Nitrilos/farmacología , Nitrilos/uso terapéutico , Cultivo Primario de Células , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Sulfonas/farmacología , Sulfonas/uso terapéutico , Regulación hacia Arriba/inmunología , Remodelación Ventricular/efectos de los fármacos , Remodelación Ventricular/genética
11.
Guang Pu Xue Yu Guang Pu Fen Xi ; 29(1): 102-5, 2009 Jan.
Artículo en Chino | MEDLINE | ID: mdl-19385215

RESUMEN

Fat, protein and water were determined by visible and NIR transmittance spectroscopy in chilled pork. After preprocessed by multiplicative scatter correction (MSC), the quantitative analysis models were developed based on the original, first derivative and second derivative spectra by using partial least squares (PLS) at the temperatures of 0-4 degrees C and 20 degrees C, respectively. By comparing the correlation coefficient (r), RMSEC, and SEP, we found that the first derivative model was the best, and the performance for 0-4 degrees C was better than that for 20 degrees C. At 0-4 degrees C and 20 degrees C, the correlation coefficients were 0.950 and 0.924 for fat, 0.713 and 0.455 for protein and 0.944 and 0.914 for water respectively, SEP values were 2.41 and 2.95 for fat, 5.44 and 4.25 for protein, and 2.37 and 2.38 for water respectively. The results showed that the visible and NIR analysis could measure the fat and water contents in chilled pork well, but was bad for protein, and this was caused by processing line of chilled pork. What's more, the spectrum offset was found in the original spectra at about 770 nm to be about 10 nm.


Asunto(s)
Grasas/análisis , Carne/análisis , Espectroscopía Infrarroja Corta/métodos , Agua/análisis , Animales , Porcinos
12.
Guang Pu Xue Yu Guang Pu Fen Xi ; 28(11): 2536-9, 2008 Nov.
Artículo en Chino | MEDLINE | ID: mdl-19271484

RESUMEN

The research was to detect soluble solids content (SSC) of pear online by near infrared transmission spectrum. The movement speed of pear was 0.5 m x s(-1) the power of light source was 300 W, and semi-transmission was used to collect the spectrum of pears. The total experiment samples were 187 pears, with a calibration set of 147 pears and a validation set of 40 pears. Partial least squares (PLS) and principal component regression (PCR) technique were used to develop the calibration model for online detection. Spectral ranges of 550-700 nm, 700-850 nm, 550-850 nm were used to establish the calibration models, and it was found that the model with 550-850 nm was better than others whether for PLS or for PCR. Also, the models based on different pretreatment methods such as Savitzky-Golay smooth, first derivative, second derivative and so on were compared, and the result showed that the five point Savitzky-Golay smooth could increase S/N ratio and improve performance of the model, whereas first derivative and second derivative could do little to improve performance of the model. The best model had the satisfactory calibration and prediction abilities, with the correlation coefficient (Rc) = 0.948 8, root mean square error of calibration (RMSEC) = 0.236 and root mean square error of validation (RMSEP) = 0.548. The result in this study shows that the detection of SSC of pear online is feasible.


Asunto(s)
Pyrus/química , Espectroscopía Infrarroja Corta/métodos , Análisis de los Mínimos Cuadrados , Análisis de Regresión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...