Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Acta Pharmacol Sin ; 45(7): 1425-1437, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38839936

RESUMEN

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.


Asunto(s)
Fibrosis , Ratones Endogámicos C57BL , Infarto del Miocardio , Animales , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Ratones , Masculino , Proteínas Señalizadoras YAP/metabolismo , Fibroblastos/metabolismo , Citidina/análogos & derivados , Citidina/farmacología , Ratones Noqueados , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Acetiltransferasa E N-Terminal/metabolismo , Vía de Señalización Hippo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Células Cultivadas , Transducción de Señal , Acetiltransferasas N-Terminal/metabolismo , Miocardio/patología , Miocardio/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo
2.
Clin Chim Acta ; 558: 119672, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38621589

RESUMEN

BACKGROUND: Thromboxane metabolites could indirectly reflect platelet activation, among which 11-dehydro-thromboxane B2 (11dhTxB2) and 11-dehydro-2, 3-dinor thromboxane B2 (11dh23dinorTxB2) are two stable metabolites that are abundant in urine, and both are closely related to disease progression and drug use. However, most clinical application studies have focused on the single indicator of 11dhTxB2. We propose an LC-MS/MS method suitable for routine clinical screening with simultaneous determination of both metabolites and conduct preliminary studies in different populations. METHODS AND RESULTS: The thromboxane metabolites were extracted by liquid-liquid extraction and determined by LC-MS/MS. Reference intervals (RI) were established in 333 healthy adults and validated in 25 patients with coronary atherosclerosis (CA). This LC-MS/MS method was over a wide quantitative range (0.1-10 µmol/L), the imprecision and accuracy were 5.2 %-11 % and 89.3 %-106.5 %, and was suitable for clinical routine quantitative screening. The 95th percentile RI of unire 11dhTxB2 was 1220 (95 % CI: 1048, 1376) pg mg Cr -1, for 11dh23dinorTxB2, RI was 908 (95 % CI: 821, 1102) pg mg Cr -1. For the first time, we found a significant correlation between 11dhTxB2 and 11dh23dinorTxB2 in both healthy adults (r = 0.67, P < 0.001) and CA patients (r = 0.77, P < 0.001). CONCLUSION: The establishment of RI provides a reference for diseases related to platelet activation and the use of drugs, and the first discovery of the correlation between 11dhTxB2 and 11dh23dinorTxB2 in urine provides a new possibilitie for the diagnostic and prognostic of cardiovascular diseases.


Asunto(s)
Activación Plaquetaria , Espectrometría de Masas en Tándem , Tromboxano B2/análogos & derivados , Humanos , Masculino , Femenino , Adulto , Persona de Mediana Edad , Valores de Referencia , Tromboxanos/orina , Tromboxanos/metabolismo , Tromboxanos/sangre , Cromatografía Liquida , Anciano , Adulto Joven , Enfermedad de la Arteria Coronaria/orina , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/diagnóstico
3.
Anal Chem ; 95(30): 11449-11455, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37477343

RESUMEN

The blood-brain barrier (BBB) is essential for maintaining central nervous system (CNS) stability, and neuroinflammation may cause the dysfunction of the BBB. MicroRNA-146a (miR-146a) is closely associated with neuroinflammation, which showed significant upregulation in response to lipopolysaccharide (LPS) induction. Elucidating the relationship between LPS-induced miR-146a expression and the BBB could decipher the mechanism of many neurological diseases. Here, we constructed an in vitro microfluidic human-BBB (µF-hBBB) chip consisting of human umbilical vein vascular endothelial cells (HUVECs) and human astrocyte (HAs) cells. A tetrahedral DNA framework (TDF-3MB) nanoprobe was used to label miR-146a in HUVECs on µF-hBBB chips before and after LPS induction, and the study revealed a significant increase in miR-146a expression after LPS induction. We believe that such a µF-hBBB chip is a promising in vitro platform for further use in understanding CNS diseases.


Asunto(s)
Lipopolisacáridos , MicroARNs , Humanos , Lipopolisacáridos/farmacología , MicroARNs/genética , MicroARNs/metabolismo , Barrera Hematoencefálica/metabolismo , Enfermedades Neuroinflamatorias , Microfluídica , Inflamación/inducido químicamente , Inflamación/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , ADN
4.
Pharmacol Res ; 184: 106422, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36058431

RESUMEN

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.


Asunto(s)
Infarto del Miocardio , Miocitos Cardíacos , Animales , Animales Recién Nacidos , Ciclo Celular , Proteínas de Ciclo Celular/genética , Proliferación Celular , Inestabilidad Cromosómica , Cisteína/metabolismo , Corazón/fisiología , Ratones , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Oxidantes/metabolismo , ARN Circular/genética , Regeneración/fisiología
5.
Mater Today Bio ; 16: 100346, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35833198

RESUMEN

Circulating tumor cells (CTCs) have been recognized as a general biomarker for the early detection, diagnosis and therapy monitoring of cancer. Due to their extreme rarity in peripheral blood, the isolation and analysis of CTCs with high efficiency, high purity and high viability remains a tremendous technological challenge. Herein, we combined tetrahedral DNA framework (TDFs), herringbone channel (HB) chip, together with aptamer-triggered hybridization chain reaction (apt-HCR) to develop an efficient microfluidic system (T-µFS) for capture and release of simulated CTCs. The capture efficiency of MCF-7 â€‹cells was from 83.3% to 94.2% when the cell numbers ranged from 10 to 103 using our T-µFS in the whole blood. The release efficiency of the MCF-7 â€‹cells was 96.2% and the MCF-7 â€‹cell viability after release was 94.6% using our T-µFS in PBS buffer. Reculture and RT-qPCR studies showed that there was almost no damage by the capture and release treatment for the MCF-7 â€‹cells viability. These results revealed that our T-µFS could be developed as an integrated and automatic technical platform with great performance for multivalent capture and release of CTCs and have a wide application prospect for tumor liquid biopsy.

6.
Analyst ; 147(10): 2231-2237, 2022 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-35471423

RESUMEN

The accurate and effective imaging of tumor-related miRNA in living cells has been playing an increasingly important role in cancer imaging. However, due to the low miRNA content and complex intracellular microenvironment, the current imaging methods of miRNAs in living cells still have some limitations. In this work, we developed a designer nanoprobe of tetrahedral DNA framework (TDF) combined with MB (termed TDFM nanoprobe) for the efficient fluorescence imaging of tumor-related miRNA-214 in living cells. In cell-free experiments, we demonstrated that the TDFM nanoprobe has sensitive detection and good specificity by fluorescence measurements. Before the TDFM nanoprobe was used for intracellular miRNA-214 fluorescence imaging, we confirmed its intracellular stability and negligible cytotoxicity by a standard MTT assay. In intracellular imaging experiments, we observed the strong fluorescence signal exhibited by the cells incubated with the TDFM nanoprobe using confocal fluorescence microscopy, which indicated that the TDFM nanoprobe was suitable for detecting and imaging tumor-related miRNA-214 in living cells. Furthermore, under the optimal incubation conditions, we employed the TDFM nanoprobe to study differences in the expression levels of tumor-related miRNA-214 in human breast cancer cells (MCF-7) and human umbilical vein endothelial cells (HUVEC). The TDFM nanoprobe we designed shows great potential to be applied in the development of DNA nanodevices, providing an improved strategy for the fluorescence imaging of miRNAs in living cells.


Asunto(s)
MicroARNs , Neoplasias , ADN/genética , Células Endoteliales/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Imagen Molecular , Neoplasias/diagnóstico , Imagen Óptica , Microambiente Tumoral
7.
Front Cell Dev Biol ; 10: 835205, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35445023

RESUMEN

Stem cell transplantation (SCT) holds great promise for overcoming diseases by regenerating damaged cells, tissues and organs. The potential for self-renewal and differentiation is the key to SCT. RNA methylation, a dynamic and reversible epigenetic modification, is able to regulate the ability of stem cells to differentiate and regenerate. N 6-methyladenosine (m6A) is the richest form of RNA methylation in eukaryotes and is regulated by three classes of proteins: methyltransferase complexes, demethylase complexes and m6A binding proteins. Through the coordination of these proteins, RNA methylation precisely modulates the expression of important target genes by affecting mRNA stability, translation, selective splicing, processing and microRNA maturation. In this review, we summarize the most recent findings on the regulation of m6A modification in embryonic stem cells, induced pluripotent stem cells and adult stem cells, hoping to provide new insights into improving SCT technology.

8.
Nanomaterials (Basel) ; 12(4)2022 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-35214994

RESUMEN

Tumor-associated cell-free DNA (cfDNA) is a dynamic biomarker for genetic analysis, early diagnosis and clinical treatment of cancers. However, its detection has limitations because of its low abundance in blood or other complex bodily fluids. Herein, we developed an ultrasensitive cfDNA electrochemical biosensor (E-cfDNA sensor) based on tetrahedral DNA framework (TDF)-modified gold nanoparticles (Au NPs) with an interface for cfDNA detection. By accurately controlling the numbers of base pairs on each DNA framework, three types of TDFs were programmed: 26 base pairs of TDF; 17 base pairs of TDF; and 7 base pairs of TDF (TDF-26, TDF-16 and TDF-7, respectively). We also combined the TDF with hybridization chain reaction (HCR) to achieve signal amplification. Under optimal conditions, we detected the breast cancer susceptibility gene 1 (BRCA-1), a representative cfDNA closely related to breast cancer. An ultra-low detection limit of 1 aM with a linear range from 1 aM to 1 pM by TDF-26 was obtained, which was superior to the existing methods. Each type of TDF has excellent discrimination ability, which can distinguish single mismatch. More significantly, we also detected BRCA-1 in mimic serum samples, demonstrating that the E-cfDNA sensor has potential use in clinical research.

9.
Biosens Bioelectron ; 204: 114077, 2022 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-35180687

RESUMEN

In situ nondestructive bioanalysis of targets in nanoscale confined space, e.g. exosomes, poses a high challenge to analytical technologies, especially to molecular fluorescent probes, because it is required to enter the confined space to recognize the target, and maintain independent and stable signal output. The unexpected fluorescence quenching and fluorescence resonance energy transfer (FRET) caused by high-frequency Brownian motion and collision in confined space are the main limiting factors. Herein, we constructed a well-defined and programmable cubic DNA nanocage-based three-dimensional molecular beacon (ncMB), which successfully broke through the above dilemma, and realized the detection of miRNA in exosomes. Specifically, steric hindrance and electrostatic repulsion derived from the unique three-dimensional structure of ncMB result in a barrier between fluorescent probes, thus eliminating unexpected fluorescence quenching during single exosomal miRNA detection and unexpected FRET during dual exosomal miRNA detection. Benefiting from the excellent anti-fluorescence and anti-FRET performance of ncMB, compared with traditional molecular beacons (MB), the detected fluorescence signal in exosomes can be improved by an order of magnitude. Moreover, ncMB is proven to have powerful programmability and anti-interference capability. Overall, it is believed that the ncMB can eliminate the signal distortion that was usually associated with commonly used MB, especially in the confined space. The ncMB is considered as a powerful and versatile tool for accurate in situ signal output in exosomes and maybe other confined spaces.


Asunto(s)
Técnicas Biosensibles , Exosomas , MicroARNs , Técnicas Biosensibles/métodos , ADN/análisis , Exosomas/química , MicroARNs/análisis , MicroARNs/genética
10.
Mol Biol Rep ; 49(5): 3705-3712, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35122597

RESUMEN

BACKGROUND: Accurately detecting and quantifying tumor-related microRNAs (miRNAs) in living cells is of great value for early cancer diagnosis. Herein, we present poly-adenine (polyA)-mediated spherical nucleic acid (SNA) nanoprobes for intracellular miRNA imaging in living cells. METHODS AND RESULTS: polyA-mediated spherical nucleic acid (pASNA) nanoprobes consist of gold nanoparticles (AuNPs) anchored with fluorophore-labeled DNA molecules pre-hybridized with recognition sequences and polyA tails. The detection performance for miRNAs in vitro was studied to confirm the feasibility of pASNA nanoprobes for imaging live cell miRNAs. Before the pASNA nanoprobes were used for imaging intracellular miRNAs in MCF-7, HeLa, and LO2 cells, the stability and non-cytotoxicity were investigated using Dnase I and a standard colorimetric CCK8 assay. Flow cytometry, qRT-PCR analyses were conducted to confirm the different expression levels of miR-155 in live cells. Results showed that the pASNA nanoprobes had good detection sensitivity and specificity, excellent stability, and low toxicity. After incubating with pASNA nanoprobes, noticeable fluorescence signal enhancement could be clearly observed in MCF-7 and HeLa cells but not LO2 cells by confocal microscopy. Flow cytometry analysis and qRT-PCR indicated that MCF-7 and HeLa cells had higher miR-155 expression levels compared to LO2 cells. CONCLUSIONS: The pASNA nanoprobes we developed had good sensitivity and specificity, excellent nuclease stability and low toxicity, thus representing a new approach to exquisitely reveal the distribution of endogenous miRNAs in live cells.


Asunto(s)
Nanopartículas del Metal , MicroARNs , Ácidos Nucleicos , Oro , Células HeLa , Humanos , MicroARNs/análisis , MicroARNs/genética , Sondas de Ácido Nucleico , Imagen Óptica , Poli A
11.
Front Cardiovasc Med ; 8: 753664, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34926608

RESUMEN

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.
Pharmacol Res ; 174: 105845, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34428587

RESUMEN

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.


Asunto(s)
Adenosina/metabolismo , Metiltransferasas/metabolismo , Infarto del Miocardio/metabolismo , Adenoviridae , Animales , Ciclo Celular , Corazón , Humanos , Masculino , Metilación , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs , ARN Mensajero , Regeneración , Transducción de Señal , Transfección , Regulación hacia Arriba
13.
Curr Drug Targets ; 22(16): 1832-1843, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33745431

RESUMEN

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.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19 , Enfermedades Cardiovasculares , Lesiones Cardíacas , COVID-19/patología , Enfermedades Cardiovasculares/virología , Lesiones Cardíacas/virología , Humanos
14.
Theranostics ; 11(6): 3000-3016, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33456585

RESUMEN

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.


Asunto(s)
Desmetilasa de ARN, Homólogo 5 de AlkB/genética , Proliferación Celular/genética , Corazón/fisiología , Miocitos Cardíacos/fisiología , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Regeneración/genética , Animales , Células Cultivadas , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/genética , Infarto del Miocardio/fisiopatología
15.
Acta Pharmacol Sin ; 42(6): 921-931, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32839503

RESUMEN

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.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Melatonina/uso terapéutico , Infarto del Miocardio/tratamiento farmacológico , Miocitos Cardíacos/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Animales Recién Nacidos , Cateninas/metabolismo , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Corazón/efectos de los fármacos , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Receptor de Melatonina MT1/metabolismo , Receptor de Melatonina MT2/metabolismo , Regeneración/efectos de los fármacos , Proteínas Señalizadoras YAP , Catenina delta
16.
Mol Biomed ; 2(1): 34, 2021 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-35006441

RESUMEN

Cardiovascular diseases such as myocardial infarction (MI) is a major contributor to human mortality and morbidity. The mammalian adult heart almost loses its plasticity to appreciably regenerate new cardiomyocytes after injuries, such as MI and heart failure. The neonatal heart exhibits robust proliferative capacity when exposed to varying forms of myocardial damage. The ability of the neonatal heart to repair the injury and prevent pathological left ventricular remodeling leads to preserved or improved cardiac function. Therefore, promoting cardiomyocyte proliferation after injuries to reinitiate the process of cardiomyocyte regeneration, and suppress heart failure and other serious cardiovascular problems have become the primary goal of many researchers. Here, we review recent studies in this field and summarize the factors that act upon the proliferation of cardiomyocytes and cardiac repair after injury and discuss the new possibilities for potential clinical treatment strategies for cardiovascular diseases.

17.
Opt Lett ; 45(18): 5165-5168, 2020 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-32932479

RESUMEN

This Letter presents a single-layer, dual-frequency unit for generating orbital angular momentum (OAM) in the microwave range. The unit cell consists of a square frame and two concentric rings with branches. The developed units can produce multifunctional OAM with required OAM mode, beam number, and direction. To demonstrate this versatility, three reflectarrays operating at dual frequencies are designed, and one is fabricated and measured to validate the design. The reflectarray has the following advantages: high gain (15.4dBi at 10 GHz, 20.3dBi at 20 GHz), high aperture efficiency (13.53% at 10 GHz, 10.33% at 20 GHz), low divergence angle (7.5°at 10 GHz, 6° at 20 GHz), small size, and compactness in the form of a single-layer structure. The designed multifunctional reflectarray has potential applications in remote sensing, point-to-point communication, satellite communications, and others.

18.
Cell Death Differ ; 27(7): 2158-2175, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31969690

RESUMEN

Neonatal mammalian heart maintains a transient regeneration capacity after birth, whereas this regeneration ability gradually loses in the postnatal heart. Thus, the reactivation of cardiomyocyte proliferation is emerging as a key strategy for inducing heart regeneration in adults. We have reported that a highly conserved long noncoding RNA (lncRNA) LncDACH1 was overexpressed in the failing hearts. Here, we found that LncDACH1 was gradually upregulated in the postnatal hearts. Cardiac-specific overexpression of LncDACH1 (TG) in mice suppressed neonatal heart regeneration and worsened cardiac function after apical resection. Conversely, in vivo cardiac conditional knockout of LncDACH1 (CKO) and adenovirus-mediated silencing of endogenous LncDACH1 reactivated cardiomyocyte-proliferative potential and promoted heart regeneration after myocardial infarction (MI) in juvenile and adult mice. Mechanistically, LncDACH1 was found to directly bind to protein phosphatase 1 catalytic subunit alpha (PP1A), and in turn, limit its dephosphorylation activity. Consistently, PP1A siRNA or pharmacological blockers of PP1A abrogated cardiomyocyte mitosis induced by LncDACH1 silencing. Furthermore, LncDACH1 enhanced yes-associated protein 1 (YAP1) phosphorylation and reduced its nuclear translocation by binding PP1A. Verteporfin, a YAP1 inhibitor decreased LncDACH1 silencing-induced cardiomyocyte proliferation. In addition, targeting a conserved fragment of LncDACH1 caused cell cycle re-entry of human iPSC-derived cardiomyocytes. Collectively, LncDACH1 governs heart regeneration in postnatal and ischemic hearts via regulating PP1A/YAP1 signal, which confers a novel therapeutic strategy for ischemic heart diseases.


Asunto(s)
Infarto del Miocardio/genética , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Miocardio/patología , ARN Largo no Codificante/metabolismo , Regeneración , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenoviridae/metabolismo , Animales , Animales Recién Nacidos , Proliferación Celular , Secuencia Conservada , Pruebas de Función Cardíaca , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Infarto del Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fosforilación , Proteína Fosfatasa 1/metabolismo , ARN Largo no Codificante/genética , Transducción de Señal , Proteínas Señalizadoras YAP
19.
Bioinformatics ; 36(5): 1542-1552, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31591638

RESUMEN

MOTIVATION: Deep neural network (DNN) algorithms were utilized in predicting various biomedical phenotypes recently, and demonstrated very good prediction performances without selecting features. This study proposed a hypothesis that the DNN models may be further improved by feature selection algorithms. RESULTS: A comprehensive comparative study was carried out by evaluating 11 feature selection algorithms on three conventional DNN algorithms, i.e. convolution neural network (CNN), deep belief network (DBN) and recurrent neural network (RNN), and three recent DNNs, i.e. MobilenetV2, ShufflenetV2 and Squeezenet. Five binary classification methylomic datasets were chosen to calculate the prediction performances of CNN/DBN/RNN models using feature selected by the 11 feature selection algorithms. Seventeen binary classification transcriptome and two multi-class transcriptome datasets were also utilized to evaluate how the hypothesis may generalize to different data types. The experimental data supported our hypothesis that feature selection algorithms may improve DNN models, and the DBN models using features selected by SVM-RFE usually achieved the best prediction accuracies on the five methylomic datasets. AVAILABILITY AND IMPLEMENTATION: All the algorithms were implemented and tested under the programming environment Python version 3.6.6. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Biología Computacional , Redes Neurales de la Computación , Algoritmos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA