Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
Acta Pharmacol Sin ; 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38839936

RESUMO

Cardiac fibrosis is a pathological scarring process that impairs cardiac function. N-acetyltransferase 10 (Nat10) is recently identified as the key enzyme for the N4-acetylcytidine (ac4C) modification of mRNAs. In this study, we investigated the role of Nat10 in cardiac fibrosis following myocardial infarction (MI) and the related mechanisms. MI was induced in mice by ligation of the left anterior descending coronary artery; cardiac function was assessed with echocardiography. We showed that both the mRNA and protein expression levels of Nat10 were significantly increased in the infarct zone and border zone 4 weeks post-MI, and the expression of Nat10 in cardiac fibroblasts was significantly higher compared with that in cardiomyocytes after MI. Fibroblast-specific overexpression of Nat10 promoted collagen deposition and induced cardiac systolic dysfunction post-MI in mice. Conversely, fibroblast-specific knockout of Nat10 markedly relieved cardiac function impairment and extracellular matrix remodeling following MI. We then conducted ac4C-RNA binding protein immunoprecipitation-sequencing (RIP-seq) in cardiac fibroblasts transfected with Nat10 siRNA, and revealed that angiomotin-like 1 (Amotl1), an upstream regulator of the Hippo signaling pathway, was the target gene of Nat10. We demonstrated that Nat10-mediated ac4C modification of Amotl1 increased its mRNA stability and translation in neonatal cardiac fibroblasts, thereby increasing the interaction of Amotl1 with yes-associated protein 1 (Yap) and facilitating Yap translocation into the nucleus. Intriguingly, silencing of Amotl1 or Yap, as well as treatment with verteporfin, a selective and potent Yap inhibitor, attenuated the Nat10 overexpression-induced proliferation of cardiac fibroblasts and prevented their differentiation into myofibroblasts in vitro. In conclusion, this study highlights Nat10 as a crucial regulator of myocardial fibrosis following MI injury through ac4C modification of upstream activators within the Hippo/Yap signaling pathway.

2.
Int J Biol Macromol ; 267(Pt 2): 131657, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38636753

RESUMO

Porous starch (PS) is a modified starch with commendable biodegradable and adsorption properties. PS exhibits poor thermal stability, and the aqueous solution casting method is conventionally used for PS-activated packaging films. This approach limits the large-scale production of films and makes it difficult to play the functions of porous pores. In this study, PS was prepared by enzymatic digestion combined with freeze-drying and adsorbed with clove essential oil (CEO) after cross-linking with sodium trimetaphosphate. Subsequently, a novel PLA/PBAT/TPS/ScPS-CEO sustained release active packaging film was prepared by blending PLA, PBAT, TPS, and ScPS-CEO using industrial melt extrusion. Compared with PS, ScPS effectively slowed down the release of CEO from the film, with the maximum release of active substances at equilibrium increasing by approximately 100 %, which significantly enhanced the persistence of the antimicrobial and antioxidant properties. The polylactic acid/poly (butylene adipate-co-terephthalate)/thermoplastic starch/trimetaphosphate-crosslinked porous starch incorporated with clove essential oil (PLA/PBAT/TPS/ScPS-CEO) film could reduce the proteolysis, lipid oxidation and microbial growth of salmon, extending its shelf life by approximately 100 % at 4 °C. These results indicate that the ScPS can be used in fresh packaging material in practical applications.


Assuntos
Antioxidantes , Poliésteres , Amido , Amido/química , Poliésteres/química , Antioxidantes/química , Antioxidantes/farmacologia , Porosidade , Anti-Infecciosos/farmacologia , Anti-Infecciosos/química , Preparações de Ação Retardada/farmacologia , Embalagem de Alimentos/métodos , Temperatura
3.
Nat Commun ; 15(1): 2137, 2024 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-38459019

RESUMO

Translational control is crucial for protein production in various biological contexts. Here, we use Ribo-seq and RNA-seq to show that genes related to oxidative phosphorylation are translationally downregulated during heart regeneration. We find that Nat10 regulates the expression of Uqcr11 and Uqcrb mRNAs in mouse and human cardiomyocytes. In mice, overexpression of Nat10 in cardiomyocytes promotes cardiac regeneration and improves cardiac function after injury. Conversely, treating neonatal mice with Remodelin-a Nat10 pharmacological inhibitor-or genetically removing Nat10 from their cardiomyocytes both inhibit heart regeneration. Mechanistically, Nat10 suppresses the expression of Uqcr11 and Uqcrb independently of its ac4C enzyme activity. This suppression weakens mitochondrial respiration and enhances the glycolytic capacity of the cardiomyocytes, leading to metabolic reprogramming. We also observe that the expression of Nat10 is downregulated in the cardiomyocytes of P7 male pig hearts compared to P1 controls. The levels of Nat10 are also lower in female human failing hearts than non-failing hearts. We further identify the specific binding regions of Nat10, and validate the pro-proliferative effects of Nat10 in cardiomyocytes derived from human embryonic stem cells. Our findings indicate that Nat10 is an epigenetic regulator during heart regeneration and could potentially become a clinical target.


Assuntos
Miócitos Cardíacos , Processamento de Proteína Pós-Traducional , Animais , Feminino , Humanos , Masculino , Camundongos , Acetiltransferases/metabolismo , Miócitos Cardíacos/metabolismo , Acetiltransferases N-Terminal/metabolismo , RNA Mensageiro/metabolismo , Suínos
4.
Eur J Pharmacol ; 963: 176245, 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38052413

RESUMO

Mammalian heart is capable to regenerate almost completely early after birth through endogenous cardiomyocyte proliferation. However, this regenerative capacity diminishes gradually with growth and is nearly lost in adulthood. Cannabidiol (CBD) is a major component of cannabis and has various biological activities to regulate oxidative stress, fibrosis, inflammation, and cell death. The present study was conducted to investigate the pharmacological effects of CBD on heart regeneration in post-MI mice. MI models in adult mice were constructed via coronary artery ligation, which were administrated with or without CBD. Our results demonstrate that systemic administration (10 mg/kg) of CBD markedly increased cardiac regenerative ability, reduced infarct size, and restored cardiac function in MI mice. Consistently, in vitro study also showed that CBD was able to promote the proliferation of neonatal cardiomyocytes. Mechanistically, the expression of miR-143-3p related to cardiomyocyte proliferation was significantly down-regulated in CBD-treated cardiomyocytes, while the overexpression of miR-143-3p inhibited cardiomyocyte mitosis and eliminated CBD-induced cardiomyocyte proliferation. Moreover, CBD enhanced the expression of Yap and Ctnnd1, which were demonstrated as the target genes of miR-143-3p. Silencing of Yap and Ctnnd1 hindered the proliferative effects of CBD. We further revealed that inhibition of the cannabinoid receptor 2 impeded the regulatory effect of CBD on miR-143-3p and its downstream target Yap/Ctnnd1, which ultimately eliminated the pro-proliferative effect of CBD on neonatal and adult cardiomyocytes. Taken together, CBD promotes cardiomyocyte proliferation and heart regeneration after MI via miR-143-3p/Yap/Ctnnd1 signaling pathway, which provides a new strategy for cardiac repair in adult myocardium.


Assuntos
Canabidiol , MicroRNAs , Infarto do Miocárdio , Animais , Camundongos , Miócitos Cardíacos , Canabidiol/farmacologia , Canabidiol/uso terapêutico , Infarto do Miocárdio/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Proliferação de Células , Regeneração/fisiologia , Mamíferos/genética
6.
BMC Plant Biol ; 23(1): 118, 2023 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-36849930

RESUMO

BACKGROUND: Arbuscular mycorrhizal fungi (AMF) have a positive effect on drought tolerance of plants after establishing reciprocal resymbiosis with roots, while the underlying mechanism is not deciphered. Metabolomics can explain the mechanism of plant response to environmental stress by analyzing the changes of all small molecular weight metabolites. The purpose of this study was to use Ultra High Performance Liquid Chromatography Q Exactive Mass Spectrometer to analyze changes in root metabolites of walnut (Juglans regia) after inoculation with an arbuscular mycorrhizal fungus Diversispora spurca under well-watered (WW) and drought stress (DS). RESULTS: Sixty days of soil drought significantly inhibited root mycorrhizal colonization rate, shoot and root biomass production, and leaf water potential in walnut, while AMF inoculation significantly increased biomass production and leaf water potential, accompanied by a higher increase magnitude under DS versus under WW. A total of 3278 metabolites were identified. Under WW, AMF inoculation up-regulated 172 metabolites and down-regulated 61 metabolites, along with no changes in 1104 metabolites. However, under DS, AMF inoculation up-regulated 49 metabolites and down-regulated 116 metabolites, coupled with no changes in 1172 metabolites. Among them, juglone (a quinone found in walnuts) as the first ranked differential metabolite was up-regulated by AMF under WW but not under DS; 2,3,5-trihydroxy-5-7-dimethoxyflavanone as the first ranked differential metabolite was increased by AMF under DS but not under WW. The KEGG annotation showed a large number of metabolic pathways triggered by AMF, accompanied by different metabolic pathways under WW and DS. Among them, oxidative phosphorylation and phenylalanine metabolism and biosynthesis were triggered by AMF in response to WW and DS, where N-acetyl-L-phenylalanine was induced by AMF to increase under DS, while decreasing under WW. CONCLUSION: This study provides new insights into the metabolic mechanisms of mycorrhiza-enhanced drought tolerance in walnuts.


Assuntos
Juglans , Micorrizas , Secas , Metabolômica , Resistência à Seca
7.
Front Plant Sci ; 13: 1089420, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36523633

RESUMO

Arbuscular mycorrhizal fungi (AMF) have important roles in enhancing drought tolerance of host plants, but it is not clear whether and how AMF increase drought tolerance in walnut (Juglans regia). We hypothesized that AMF could activate antioxidant defense systems and heat shock transcription factors (Hsfs) transcription levels to alleviate oxidative damage caused by drought. The walnut variety 'Liaohe No. 1' was inoculated with Diversispora spurca and exposed to well-watered (WW, 75% of the maximum soil water capacity) and drought stress (DS, 50% of the maximum soil water capacity) for 6 weeks. Plant growth, antioxidant defense systems, and expressions of five JrHsfs in leaves were studied. Such drought treatment inhibited root mycorrhizal colonization, while plant growth performance was still improved by AMF inoculation. Mycorrhizal fungal inoculation triggered the increase in soluble protein, glutathione (GSH), ascorbic acid (ASC), and total ASC contents and ascorbic peroxidase and glutathione reductase activities, along with lower hydrogen peroxide (H2O2), superoxide anion radical (O2 •-), and malondialdehyde (MDA) levels, compared with non-inoculation under drought. Mycorrhizal plants also recorded higher peroxidase, catalase, and superoxide dismutase activities than non-mycorrhizal plants under drought. The expression of JrHsf03, JrHsf05, JrHsf20, JrHsf22, and JrHsf24 was up-regulated under WW by AMF, while the expression of JrHsf03, JrHsf22, and JrHsf24 were up-regulated only under drought by AMF. It is concluded that D. spurca induced low oxidative burst in drought-stressed walnut through activating antioxidant defense systems and part Hsfs expressions.

8.
Pharmacol Res ; 184: 106422, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36058431

RESUMO

Targeting cardiomyocyte plasticity has emerged as a new strategy for promoting heart repair after myocardial infarction. However, the precise mechanistic network underlying heart regeneration is not completely understood. As noncoding RNAs, circular RNAs (circRNAs) play essential roles in regulating cardiac physiology and pathology. The present study aimed to investigate the potential roles of circMdc1 in cardiac repair after injury and elucidate its underlying mechanisms. Here, we identified that circMdc1 levels were upregulated in postnatal mouse hearts but downregulated in the regenerative myocardium. The expression of circMdc1 in cardiomyocytes is sensitive to oxidative stress, which was attenuated by N-acetyl-cysteine. Enforced circMdc1 expression inhibited cardiomyocyte proliferation, while circMdc1 silencing led to cardiomyocyte cell cycle re-entry. In vivo, the cardiac-specific adeno-associated virus-mediated knockdown of circMdc1 promoted cardiac regeneration and heart repair accompanied by improved heart function. Conversely, circMdc1 overexpression blunted the regenerative capacity of neonatal hearts after apex resection. Moreover, circMdc1 was able to block the translation of its host gene Mdc1 specifically by binding to PABP, affecting DNA damage and the chromosome stability of cardiomyocytes. Furthermore, overexpression of Mdc1 caused damaged mouse hearts to regenerate and repair after myocardial infarction in vivo. Oxidative stress-sensitive circMdc1 plays an important role in cardiac regeneration and heart repair after injury by regulating DNA damage and chromosome stability in cardiomyocytes by blocking the translation of the host gene Mdc1.


Assuntos
Infarto do Miocárdio , Miócitos Cardíacos , Animais , Animais Recém-Nascidos , Ciclo Celular , Proteínas de Ciclo Celular/genética , Proliferação de Células , Instabilidade Cromossômica , Cisteína/metabolismo , Coração/fisiologia , Camundongos , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Oxidantes/metabolismo , RNA Circular/genética , Regeneração/fisiologia
9.
Int J Med Sci ; 19(8): 1254-1264, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35928721

RESUMO

Mammalian cardiomyocytes (CMs) maintain a low capacity for self-renewal in adulthood, therefore the induction of CMs cycle re-entry is an important approach to promote myocardial repair after injury. Recently, photobiomodulation (PBM) has been used to manipulate physiological activities of various tissues and organs by non-invasive means. Here, we demonstrate that conditioned PBM using light-emitting diodes with a wavelength of 630 nm (LED-Red) was capable of promoting the proliferation of neonatal CMs. Further studies showed that low-power LED-Red affected the expression of miR-877-3p and promoted the proliferation of CMs. In contrast, silencing of miR-877-3p partially abolished the pro-proliferative actions of LED-Red irradiation on CMs. Mechanistically, GADD45g was identified as a downstream target gene of miR-877-3p. Conditioned LED-Red irradiation also inhibited the expression of GADD45g in neonatal CMs. Moreover, GADD45g siRNA reversed the positive effect of LED-Red on the proliferation of neonatal CMs. Taken together, conditioned LED-Red irradiation increased miR-877-3p expression and promoted the proliferation of neonatal CMs by targeting GADD45g. This finding provides a new insight into the role of LED-Red irradiation in neonatal CMs biology and suggests its potential application in myocardial injury repair.


Assuntos
MicroRNAs , Miócitos Cardíacos , Animais , Proliferação de Células/genética , Mamíferos/genética , Mamíferos/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Miocárdio/metabolismo
10.
Int J Biol Sci ; 18(7): 2980-2993, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35541894

RESUMO

Photobiomodulation (PBM) has emerged as an alternative therapy involved in modulating a variety of biological effects. In this study, we verified whether PBM can affect cardiac physiological activity in mice through noninvasive irradiation using light-emitting diodes at a wavelength of 630 nm (LED-Red). We found that the PBM involved in regulating the repair of injured myocardium is wavelength-limited. LED-Red caused cardiomyocytes (CMs) that had exited the cell cycle to divide and proliferate again, and the cell proliferation ratio increased significantly with the accumulation of intracellular photopower. In addition, LED-Red promoted myocardial revascularization and myocardial regeneration, reduced the area of fibrosis in mice with myocardial infarction (MI), and thus improved cardiac contractile function. In regard to the mechanism, miRNA sequencing analysis showed that low-power LED-Red irradiation could induce differential changes in miRNAs in CMs. Among them, miR-136-5p was identified as a cardiac photo-sensitive miRNA and was obviously inhibited after stimulation, which produced a proliferation-promoting effect on CMs. Subsequent luciferase reporter assays confirmed the involvement of Ino80 as a binding target of miR-136-5p in the regulatory process of CM proliferation. Similarly, LED-Red irradiation elevated intracellular Ino80 expression. After knockdown of Ino80, the proliferation-promoting effect of LED-Red on CMs was inhibited. Collectively, this study demonstrates that LED-Red can promote CM proliferation by inhibiting cardiac photo-sensitive miRNA- miR-136-5p expression through targeting Ino80. The findings provided a new potential strategy for the treatment of ischemic cardiomyopathy (ICD).


Assuntos
Terapia com Luz de Baixa Intensidade , MicroRNAs , Infarto do Miocárdio , Animais , Apoptose , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/terapia , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo
11.
Front Cardiovasc Med ; 8: 753664, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34926608

RESUMO

Heart failure (HF) is the common consequences of various cardiovascular diseases, often leading to severe cardiac output deficits with a high morbidity and mortality. In recent years, light emitting diodes-based therapy (LEDT) has been widely used in multiple cardiac diseases, while its modulatory effects on cardiac function with HF still remain unclear. Therefore, the objective of this study was to investigate the effects of LED-Red irradiation on cardiac function in mice with HF and to reveal its mechanisms. In this study, we constructed a mouse model of HF. We found that LED-Red (630 nm) was an effective wavelength for the treatment of HF. Meanwhile, the application of LED-Red therapy to treat HF mice improved cardiac function, ameliorate heart morphology, reduced pulmonary edema, as well as inhibited collagen deposition. Moreover, LED-Red therapy attenuated the extent of perivascular fibrosis. Besides, LED-Red irradiation promoted calcium transients in cardiomyocytes as well as upregulated ATP synthesis, which may have positive implications for contractile function in mice with HF. Collectively, we identified that LED-Red exerts beneficial effects on cardiac function in HF mice possibly by promoting the synthesis of ATP.

12.
Molecules ; 26(19)2021 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-34641527

RESUMO

Toddalia asiatica (L.) Lam. (Rutaceae) has shown a broad spectrum of biological properties, such as anti-inflammatory, antioxidant, antimicrobial, anti-HIV, and anticancer properties. The present study is concerned with the separation of the main components with broad partition coefficients (KD values) from T. asiatica, using linear gradient high-speed counter-current chromatography (LGCCC) combined with an off-line two-dimensional (2D) mode. Similar to the binary gradient HPLC, the LGCCC mode is operated by the adjustment of the proportion between the mobile phase of 5:5:1:9 (v/v) (pump A) and 5:5:4.5:5.5 (v/v) (pump B) in an n-hexane/ethyl acetate/methanol/water solvent system. The off-line 2D-CCC mode was used in this study for the secondary separation of two similar KD value compounds with n-hexane/ethyl acetate/methanol/water (5:5:4:6, v/v). Notably, six coumarins, namely, tomentin (1), toddalolactone (2), 5,7,8-trimethoxycoumarin (3), mexoticin (4), isopimpinellin (5), and toddanone (6), were efficiently separated. The structures of the pure compounds were elucidated by spectral techniques and compared with the literature.


Assuntos
Cumarínicos/isolamento & purificação , Distribuição Contracorrente/métodos , Raízes de Plantas/química , Rutaceae/química , Cromatografia Líquida de Alta Pressão , Cumarínicos/química , Furocumarinas/isolamento & purificação , Estrutura Molecular , Solventes/química
13.
Mol Ther Nucleic Acids ; 26: 22-33, 2021 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-34513291

RESUMO

N6-methyladenosine (m6A), as the most abundant modification of mammalian messenger RNAs, is essential for tissue development and pathogenesis. However, the biological significance of m6A methylation in cardiac differentiation and development remains largely unknown. Here, we identify that the downregulation of m6A demethylase ALKBH5 is responsible for the increase of m6A methylation and cardiomyocyte fate determination of human embryonic stem cells (hESCs) from mesoderm cells (MESs). In contrast, ALKBH5 overexpression remarkably blocks cardiomyocyte differentiation of hESCs. Mechanistically, KDM5B and RBBP5, the components of H3K4 modifying enzyme complexes, are identified as downstream targets for ALKBH5 in cardiac-committed hESCs. Loss of function of ALKBH5 alters the expression of KDM5B and RBBP5 through impairing stability of their mRNAs, which in turn promotes the transcription of GATA4 by enhancing histone H3 Lys4 trimethylation (H3K4me3) at the promoter region of GATA4. Taken together, we reveal a previously unidentified role of m6A demethylase ALKBH5 in determining cardiac lineage commitment of hESCs.

14.
Oxid Med Cell Longev ; 2021: 9962922, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34336126

RESUMO

Myocardial infarction refers to myocardial necrosis caused by acute or persistent coronary ischemia and hypoxia. It is considered to be one of the significant crises threatening human health in the world. Following myocardial infarction, collagen gradually replaces the original tissue due to the loss of many cardiomyocytes, myocardial contractile function decreases, and myocardial fibrosis eventually leads to heart failure. Phototherapy is a new treatment which has shown superior efficacy on the nerve, skeletal muscle, skin, and other tissues. Likewise, there is growing evidence that phototherapy also has many positive effects on the heart. Therefore, this article introduces the progress of research on phototherapy as a new therapeutic strategy in the treatment of myocardial infarction. The wavelength of photobiomodulation in the treatment of myocardial infarction is specific, and the influence of light source power and light duration on the tissue presents a bell-shaped distribution. Under these conditions, phototherapy can promote ATP synthesis and angiogenesis, inhibit the inflammatory response, improve heart function, reduce infarct size, and protect myocardium. In addition, we summarized the molecular mechanisms of phototherapy. According to the location of photoreceptors, they can be divided into mitochondrial and nonmitochondrial parts.


Assuntos
Terapia com Luz de Baixa Intensidade/métodos , Infarto do Miocárdio/terapia , Animais , Humanos
15.
Pharmacol Res ; 174: 105845, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34428587

RESUMO

AIMS: N6-Methyladenosine (m6A), one of the important epigenitic modifications, is very commom in messenger RNAs (mRNAs) of eukaryotes, and has been involved in various diseases. However, the role of m6A modification in heart regeneration after injury remains unclear. The study was conducted to investigate whether targeting methyltransferase-like 3 (METTL3) could replenish the loss of cardiomyocytes (CMs) and improve cardiac function after myocardial infarction (MI). METHODS AND RESULTS: METTL3 knockout mouse line was generated. A series of functional experiments were carried out and the molecular mechanism was further explored. We identified that METTL3, a methyltransferase of m6A methylation, is upregulated in mouse hearts after birth, which is the opposite of the changes in CMs proliferation. Furthermore, both METTL3 heterozygous knockout mice and administration of METTL3 shRNA adenovirus in mice exhibited CMs cell cycle re-entered, infract size decreased and cardiac function improved after MI. Mechanically, the silencing of METTL3 promoted CMs proliferation by reducing primary miR-143 (pri-miR-143) m6A modificaiton, thereby inhibiting the pri-miR-143 into mature miR-143-3p. Moreover, we found that miR-143-3p has targeting effects on Yap and Ctnnd1 so as to regulate CMs proliferation. CONCLUSION: METTL3 deficiency contributes to heart regeneration after MI via METTL3-pri-miR-143-(miR-143)-Yap/Ctnnd1 axis. This study provides new insights into the significance of RNA m6A modification in heart regeneration.


Assuntos
Adenosina/metabolismo , Metiltransferases/metabolismo , Infarto do Miocárdio/metabolismo , Adenoviridae , Animais , Ciclo Celular , Coração , Humanos , Masculino , Metilação , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs , RNA Mensageiro , Regeneração , Transdução de Sinais , Transfecção , Regulação para Cima
16.
Front Pharmacol ; 12: 680349, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34248630

RESUMO

Cardiac hypertrophy is a common pathological process of various cardiovascular diseases, which is often accompanied with structural and electrical remodeling, and can even lead to sudden cardiac death. However, its molecular mechanism still remains largely unknown. Here, we induced cardiomyocyte hypertrophy by angiotensin II (Ang II), and found that miR-27a-3p and hypertrophy-related genes were up-regulated. Further studies showed that miR-27a-3p-inhibitor can alleviate myocardial hypertrophy and electrical remodeling. Moreover, luciferase assay confirmed that miR-27a-3p could regulate the expression of downstream Hoxa10 at the transcriptional level by targeting at its 3'UTR. At the same time, the protein expression of Hoxa10 was significantly reduced in Ang II-treated cardiomyocytes. Furthermore, overexpression of Hoxa10 can reverse myocardial hypertrophy and electrical remodeling induced by Ang II in cardiomyocytes. Finally, we found that Hoxa10 positively regulated the expression of potassium channel protein Kv4.3 which was down-regulated in hypertrophic cardiomyocytes. Taken together, our results revealed miR-27a-3p/Hoxa10/Kv4.3 axis as a new mechanism of Ang II-induced cardiomyocyte hypertrophy, which provided a new target for clinical prevention and treatment of cardiac hypertrophy and heart failure.

17.
Curr Drug Targets ; 22(16): 1832-1843, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33745431

RESUMO

ACE2 has long been known as an injury protective protein, which can protect against a variety of organ damage such as the heart, liver, kidney, and lung. Especially in cardiovascular diseases, as a negative regulator of RAAS, ACE2 is an extremely important protective factor that mainly plays a role by converting Ang II to Ang-(1-7). Nevertheless, with the recent outbreak of COVID-19, it is exposed that another identity of ACE2 is the entry receptor for SARS-CoV-2, which previously serves as the entry receptor for SARS. With the in-depth clinical research, it is found that the severity and susceptibility of COVID-19 are related to cardiovascular diseases, and SARS-CoV-2 binding to ACE2 receptor is also potentially associated with heart injury symptoms. Therefore, in this article, we mainly summarize the relationship between ACE2, COVID-19, and cardiovascular diseases/heart injury.


Assuntos
Enzima de Conversão de Angiotensina 2/metabolismo , COVID-19 , Doenças Cardiovasculares , Traumatismos Cardíacos , COVID-19/patologia , Doenças Cardiovasculares/virologia , Traumatismos Cardíacos/virologia , Humanos
18.
Int J Nanomedicine ; 16: 481-492, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33500618

RESUMO

BACKGROUND: Dihydromyricetin (DMY), a natural flavonoid, has reportedly antibacterial, antioxidant, anticancer and other properties. In the present study, DMY was used as a reducing agent and stabilizer to synthesize silver nanoparticles (AgNPs), and the optimal conditions for its synthesis were studied. The DMY-AgNPs were investigated for their DPPH scavenging properties and their potential against human pathogenic and food-borne bacteria viz. Escherichia coli (E. coli), and Salmonella. In addition, DMY-AgNPs also showed excellent inhibitory effects on cancer Hela, HepG2 and MDA-MB-231 cell lines. METHODS: The dihydromyricetin-mediated AgNPs (DMY-AgNPs) were characterized by ultraviolet-visible spectrophotometer (UV-Vis spectra), scanning electron microscopy (SEM), transmission electron microscopy (TEM), dynamic light scattering (DLS), Fourier-transform infrared spectroscopy (FTIR), X-ray photoelectron spectroscopy (XPS), and X-ray powder diffraction (XRD). Antioxidant activity of DMY-AgNPs was determined by 1.1-diphenyl-2-picrylhydrazyl (DPPH) scavenging. The antibacterial activity was determined by 96-well plate (AGAR) gradient dilution, while anticancer potential was determined by MTT assay. RESULTS: The results showed that the dispersion of AgNPs had the maximum UV-visible absorption at about 410 nm. The synthesized nanoparticles were almost spherical. FTIR was used to identify functional groups that may lead to the transformation of metal ions into nanoparticles. The results showed that the prepared AgNPs were coated with biological molecules in the extraction solution. The biosynthesized DMY-AgNPs exhibited good antioxidant properties, at various concentrations (0.01-0.1mg/mL), the free radical scavenging rate was about 56-92%. Furthermore, DMY-AgNPs possessed good antibacterial properties against Escherichia coli (E. coli), and Salmonella at room temperature. The minimum inhibitory concentrations (MIC) were 10-6 g/L, and 10-4 g/L, respectively. The bioactivity of DMY-mediated AgNPs was studied using MTT assay against Hela, HepG2 and MDA-MB-231 cancer cell lines, and all showed good inhibitory effects. CONCLUSION: The present study provides a green approach for the synthesis of DMY-AgNPs which exhibited stronger antioxidant, antibacterial and anticancer properties compared to the dihydromyricetin. DMY-AgNPs can serve as an economical, efficient, and effective antimicrobial material for its applications in food and pharmaceutical fields.


Assuntos
Antibacterianos/farmacologia , Antineoplásicos/farmacologia , Antioxidantes/farmacologia , Flavonóis/farmacologia , Nanopartículas Metálicas/química , Prata/farmacologia , Bactérias/efeitos dos fármacos , Linhagem Celular Tumoral , Escherichia coli/efeitos dos fármacos , Humanos , Nanopartículas Metálicas/ultraestrutura , Testes de Sensibilidade Microbiana , Espectrofotometria Ultravioleta , Difração de Raios X
19.
Theranostics ; 11(6): 3000-3016, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33456585

RESUMO

N6-methyladenosine (m6A) RNA modification, a dynamic and reversible process, is essential for tissue development and pathogenesis. However, the potential involvement of m6A in the regulation of cardiomyocyte (CM) proliferation and cardiac regeneration remains unclear. In this study, we aimed to investigate the essential role of m6A modification in heart regeneration during postnatal and adult injury. Methods and results: In this study, we identified the downregulation of m6A demethylase ALKBH5, an m6A "eraser" that is responsible for increased m6A methylation, in the heart after birth. Notably, ALKBH5 knockout mice exhibited decreased cardiac regenerative ability and heart function after neonatal apex resection. Conversely, forced expression of ALKBH5 via adeno-associated virus-9 (AAV9) delivery markedly reduced the infarct size, restored cardiac function and promoted CM proliferation after myocardial infarction in juvenile (7 days old) and adult (8-weeks old) mice. Mechanistically, ALKBH5-mediated m6A demethylation improved the mRNA stability of YTH N6-methyladenosine RNA-binding protein 1 (YTHDF1), thereby increasing its expression, which consequently promoted the translation of Yes-associated protein (YAP). The modulation of ALKBH5 and YTHDF1 expression in human induced pluripotent stem cell-derived cardiomyocytes consistently yielded similar results. Conclusion: Taken together, our findings highlight the vital role of the ALKBH5-m6A-YTHDF1-YAP axis in the regulation of CMs to re-enter the cell cycle. This finding suggests a novel potential therapeutic strategy for cardiac regeneration.


Assuntos
Homólogo AlkB 5 da RNA Desmetilase/genética , Proliferação de Células/genética , Coração/fisiologia , Miócitos Cardíacos/fisiologia , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Regeneração/genética , Animais , Células Cultivadas , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/genética , Infarto do Miocárdio/fisiopatologia
20.
Acta Pharmacol Sin ; 42(6): 921-931, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32839503

RESUMO

The neonatal heart possesses the ability to proliferate and the capacity to regenerate after injury; however, the mechanisms underlying these processes are not fully understood. Melatonin has been shown to protect the heart against myocardial injury through mitigating oxidative stress, reducing apoptosis, inhibiting mitochondrial fission, etc. In this study, we investigated whether melatonin regulated cardiomyocyte proliferation and promoted cardiac repair in mice with myocardial infarction (MI), which was induced by ligation of the left anterior descending coronary artery. We showed that melatonin administration significantly improved the cardiac functions accompanied by markedly enhanced cardiomyocyte proliferation in MI mice. In neonatal mouse cardiomyocytes, treatment with melatonin (1 µM) greatly suppressed miR-143-3p levels. Silencing of miR-143-3p stimulated cardiomyocytes to re-enter the cell cycle. On the contrary, overexpression of miR-143-3p inhibited the mitosis of cardiomyocytes and abrogated cardiomyocyte mitosis induced by exposure to melatonin. Moreover, Yap and Ctnnd1 were identified as the target genes of miR-143-3p. In cardiomyocytes, inhibition of miR-143-3p increased the protein expression of Yap and Ctnnd1. Melatonin treatment also enhanced Yap and Ctnnd1 protein levels. Furthermore, Yap siRNA and Ctnnd1 siRNA attenuated melatonin-induced cell cycle re-entry of cardiomyocytes. We showed that the effect of melatonin on cardiomyocyte proliferation and cardiac regeneration was impeded by the melatonin receptor inhibitor luzindole. Silencing miR-143-3p abrogated the inhibition of luzindole on cardiomyocyte proliferation. In addition, both MT1 and MT2 siRNA could cancel the beneficial effects of melatonin on cardiomyocyte proliferation. Collectively, the results suggest that melatonin induces cardiomyocyte proliferation and heart regeneration after MI by regulating the miR-143-3p/Yap/Ctnnd1 signaling pathway, providing a new therapeutic strategy for cardiac regeneration.


Assuntos
Proliferação de Células/efeitos dos fármacos , Melatonina/uso terapêutico , Infarto do Miocárdio/tratamento farmacológico , Miócitos Cardíacos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Animais Recém-Nascidos , Cateninas/metabolismo , Ciclo Celular/efeitos dos fármacos , Células Cultivadas , Coração/efeitos dos fármacos , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Receptor MT1 de Melatonina/metabolismo , Receptor MT2 de Melatonina/metabolismo , Regeneração/efeitos dos fármacos , Proteínas de Sinalização YAP , delta Catenina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA