Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci China Life Sci ; 67(6): 1155-1169, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38811441

RESUMEN

CFIRL is a long noncoding RNA (lncRNA), we previously identified as the most significantly upregulated lncRNA in the failing hearts of patients with dilated cardiomyopathy (DCM). In this study, we determined the function of CFIRL and its role in DCM. Real-time polymerase chain reaction and in situ hybridization assays revealed that CFIRL was primarily localized in the nucleus of cardiac fibroblasts and robustly increased in failing hearts. Global knockdown or fibroblast-specific knockout of CFIRL attenuated transverse aortic constriction (TAC)-induced cardiac dysfunction and fibrosis in vivo. Overexpression of CFIRL in vitro promoted fibroblast proliferation and aggravated angiotensin II-induced differentiation to myofibroblasts. CFIRL knockdown attenuated these effects. Mechanistically, RNA pull-down assay and gene expression profiling revealed that CFIRL recruited ENO1, a newly identified noncanonical transcriptional factor, to activate IL-6 transcription. IL-6 exerted a paracrine effect on cardiomyocytes to promote cardiac hypertrophy, which can be prevented by CFIRL knockdown. These findings uncover the critical role of CFIRL, a fibroblast-associated lncRNA, in heart failure by facilitating crosstalk between fibroblasts and cardiomyocytes. CFIRL knockdown might be a potent strategy to prevent cardiac remodeling in heart failure, particularly in DCM.


Asunto(s)
Cardiomiopatía Dilatada , Fibroblastos , Fibrosis , Miocitos Cardíacos , ARN Largo no Codificante , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/metabolismo , Animales , Fibroblastos/metabolismo , Masculino , Humanos , Miocitos Cardíacos/metabolismo , Ratones , Proliferación Celular , Interleucina-6/metabolismo , Interleucina-6/genética , Ratones Endogámicos C57BL , Miocardio/metabolismo , Miocardio/patología , Miofibroblastos/metabolismo , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Diferenciación Celular , Técnicas de Silenciamiento del Gen
2.
Mol Ther ; 32(5): 1578-1594, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38475992

RESUMEN

Heart failure (HF) is manifested by transcriptional and posttranscriptional reprogramming of critical genes. Multiple studies have revealed that microRNAs could translocate into subcellular organelles such as the nucleus to modify gene expression. However, the functional property of subcellular Argonaute2 (AGO2), the core member of the microRNA machinery, has remained elusive in HF. AGO2 was found to be localized in both the cytoplasm and nucleus of cardiomyocytes, and robustly increased in the failing hearts of patients and animal models. We demonstrated that nuclear AGO2 rather than cytosolic AGO2 overexpression by recombinant adeno-associated virus (serotype 9) with cardiomyocyte-specific troponin T promoter exacerbated the cardiac dysfunction in transverse aortic constriction (TAC)-operated mice. Mechanistically, nuclear AGO2 activates the transcription of ANKRD1, encoding ankyrin repeat domain-containing protein 1 (ANKRD1), which also has a dual function in the cytoplasm as part of the I-band of the sarcomere and in the nucleus as a transcriptional cofactor. Overexpression of nuclear ANKRD1 recaptured some key features of cardiac remodeling by inducing pathological MYH7 activation, whereas cytosolic ANKRD1 seemed cardioprotective. For clinical practice, we found ivermectin, an antiparasite drug, and ANPep, an ANKRD1 nuclear location signal mimetic peptide, were able to prevent ANKRD1 nuclear import, resulting in the improvement of cardiac performance in TAC-induced HF.


Asunto(s)
Proteínas Argonautas , Modelos Animales de Enfermedad , Insuficiencia Cardíaca , Miocitos Cardíacos , Proteínas Represoras , Animales , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/patología , Ratones , Humanos , Proteínas Argonautas/metabolismo , Proteínas Argonautas/genética , Miocitos Cardíacos/metabolismo , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Remodelación Ventricular , Núcleo Celular/metabolismo , Proteínas Musculares/metabolismo , Proteínas Musculares/genética , Regulación de la Expresión Génica , Masculino , Dependovirus/genética , Transcripción Genética
3.
Circulation ; 149(14): 1102-1120, 2024 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-38126189

RESUMEN

BACKGROUND: Diabetes is associated with cardiovascular complications. microRNAs translocate into subcellular organelles to modify genes involved in diabetic cardiomyopathy. However, functional properties of subcellular AGO2 (Argonaute2), a core member of miRNA machinery, remain elusive. METHODS: We elucidated the function and mechanism of subcellular localized AGO2 on mouse models for diabetes and diabetic cardiomyopathy. Recombinant adeno-associated virus type 9 was used to deliver AGO2 to mice through the tail vein. Cardiac structure and functions were assessed by echocardiography and catheter manometer system. RESULTS: AGO2 was decreased in mitochondria of diabetic cardiomyocytes. Overexpression of mitochondrial AGO2 attenuated diabetes-induced cardiac dysfunction. AGO2 recruited TUFM, a mitochondria translation elongation factor, to activate translation of electron transport chain subunits and decrease reactive oxygen species. Malonylation, a posttranslational modification of AGO2, reduced the importing of AGO2 into mitochondria in diabetic cardiomyopathy. AGO2 malonylation was regulated by a cytoplasmic-localized short isoform of SIRT3 through a previously unknown demalonylase function. CONCLUSIONS: Our findings reveal that the SIRT3-AGO2-CYTB axis links glucotoxicity to cardiac electron transport chain imbalance, providing new mechanistic insights and the basis to develop mitochondria targeting therapies for diabetic cardiomyopathy.


Asunto(s)
Diabetes Mellitus , Cardiomiopatías Diabéticas , MicroARNs , Sirtuina 3 , Ratones , Animales , Sirtuina 3/genética , Genes Mitocondriales , Mitocondrias/genética , MicroARNs/genética , Miocitos Cardíacos/metabolismo , Diabetes Mellitus/metabolismo
4.
Mol Ther Nucleic Acids ; 32: 689-703, 2023 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-37215148

RESUMEN

Diabetes could directly induce cardiac injury, leading to cardiomyopathy. However, treatment strategies for diabetic cardiomyopathy remain limited. ZNF593-AS knockout and cardiomyocyte-specific transgenic mice were constructed. In addition, high-fat diet (HFD)-induced diabetic mouse model and db/db mice, another classic diabetic mouse model, were employed. ZNF593-AS was silenced using GapmeR, a modified antisense oligonucleotide, while overexpressed using a recombinant adeno-associated virus serotype 9-mediated gene delivery system. Transcriptome sequencing, RNA pull-down assays, and RNA immunoprecipitation assays were also performed to investigate the underlying mechanisms. ZNF593-AS expression was decreased in diabetic hearts. ZNF593-AS attenuated the palmitic acid-induced apoptosis of cardiomyocytes in vitro. In HFD-induced diabetic mice, ZNF593-AS deletion aggravated cardiac dysfunction and enhanced cardiac apoptosis and inflammation. In contrast, HFD-induced cardiac dysfunction was improved in ZNF593-AS transgenic mice. Consistently, ZNF593-AS exerted the same cardioprotective effects in db/db mice. Mechanistically, ZNF593-AS directly interacted with the functional domain of interferon regulatory factor 3 (IRF3), and suppressed fatty acid-induced phosphorylation and activation of IRF3, contributing to the amelioration of cardiac cell death and inflammation. In conclusion, our results identified the protective role of ZNF593-AS in diabetic cardiomyopathy, suggesting a novel potential therapeutic target.

5.
Mol Ther Nucleic Acids ; 31: 122-138, 2023 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-36618264

RESUMEN

Intensive glycemic control is insufficient for reducing the risk of heart failure among patients with diabetes mellitus (DM). While the "hyperglycemic memory" phenomenon is well documented, little is known about its underlying mechanisms. In this study, a type 1 DM model was established in C57BL/6 mice using streptozotocin (STZ). Leptin receptor-deficient (db/db) mice were used as a model of type 2 DM. A type 9 adeno-associated virus was used to overexpress or knock down miR-320 in vivo. Diastolic dysfunction was observed in the type 1 DM mice with elevated miR-320 expression. However, glycemic control using insulin failed to reverse diastolic dysfunction. miR-320 knockdown protected against STZ-induced diastolic dysfunction. Similar results were observed in the type 2 DM mice. In vitro, we found that miR-320 promoted CD36 expression, which in turn induced further miR-320 expression. CD36 was rapidly induced by hyperglycemia at protein level compared with the much slower induction of miR-320, suggesting a positive feedback loop of CD36/miR-320 with CD36 protein induction as the initial triggering event. In conclusion, in DM-induced cardiac injury, miR-320 and CD36 mutually enhance each other's expression, leading to a positive feedback loop and a sustained hyperlipidemic state in the heart.

6.
Mol Ther Nucleic Acids ; 27: 1127-1145, 2022 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-35251768

RESUMEN

Long noncoding RNAs (lncRNAs) play crucial roles in cardiovascular diseases. To date, only limited studies have reported the role of mitochondria-derived lncRNAs in heart failure (HF). In the current study, recombinant adeno-associated virus 9 was used to manipulate lncRNA cytb (lnccytb) expression in vivo. Fluorescence in situ hybridization (FISH) assay was used to determine the location of lnccytb, while microRNA (miRNA) sequencing and bioinformatics analyses were applied to identify the downstream targets. The competitive endogenous RNA (ceRNA) function of lnccytb was evaluated by biotin-coupled miRNA pull-down assays and luciferase reporter assays. Results showed that lnccytb expression was decreased in the heart of mice with transverse aortic constriction (TAC), as well as in the heart and plasma of patients with HF. FISH assay and absolute RNA quantification via real-time reverse transcription PCR suggested that the reduction of the lnccytb transcripts mainly occurred in the cytosol. Upregulation of cytosolic lnccytb attenuated cardiac dysfunction in TAC mice. Moreover, overexpression of cytosolic lnccytb in cardiomyocytes alleviated isoprenaline-induced reactive oxidative species (ROS) production and hypertrophy. Mechanistically, lnccytb acted as a ceRNA via sponging miR-103-3p, ultimately mitigating the suppression of PTEN by miR-103-3p. In summary, we demonstrated that the overexpression of cytosolic lnccytb could ameliorate HF.

7.
Front Med ; 16(1): 25-38, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34921674

RESUMEN

Cardiovascular diseases account for approximately 80% of deaths among individuals with diabetes mellitus, with diabetic cardiomyopathy as the major diabetic cardiovascular complication. Hyperglycemia is a symptom that abnormally activates multiple downstream pathways and contributes to cardiac hypertrophy, fibrosis, apoptosis, and other pathophysiological changes. Although glycemic control has long been at the center of diabetes therapy, multicenter randomized clinical studies have revealed that intensive glycemic control fails to reduce heart failure-associated hospitalization and mortality in patients with diabetes. This finding indicates that hyperglycemic stress persists in the cardiovascular system of patients with diabetes even if blood glucose level is tightly controlled to the normal level. This process is now referred to as hyperglycemic memory (HGM) phenomenon. We briefly reviewed herein the current advances that have been achieved in research on the underlying mechanisms of HGM in diabetic cardiomyopathy.


Asunto(s)
Enfermedades Cardiovasculares , Complicaciones de la Diabetes , Diabetes Mellitus , Cardiomiopatías Diabéticas , Hiperglucemia , Cardiomiopatías Diabéticas/etiología , Humanos , Hiperglucemia/complicaciones , Hiperglucemia/metabolismo , Estudios Multicéntricos como Asunto
8.
Med Rev (Berl) ; 2(2): 140-168, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37724243

RESUMEN

Cardiovascular diseases (CVDs) are the leading causes of death and disability worldwide, despite the wide diversity of molecular targets identified and the development of therapeutic methods. MicroRNAs (miRNAs) are a class of small (about 22 nucleotides) non-coding RNAs (ncRNAs) that negatively regulate gene expression at the post-transcriptional level in the cytoplasm and play complicated roles in different CVDs. While miRNA overexpression in one type of cell protects against heart disease, it promotes cardiac dysfunction in another type of cardiac cell. Moreover, recent studies have shown that, apart from cytosolic miRNAs, subcellular miRNAs such as mitochondria- and nucleus-localized miRNAs are dysregulated in CVDs. However, the functional properties of cellular- and subcellular-localized miRNAs have not been well characterized. In this review article, by carefully revisiting animal-based miRNA studies in CVDs, we will address the regulation and functional properties of miRNAs in various CVDs. Specifically, the cell-cell crosstalk and subcellular perspective of miRNAs are highlighted. We will provide the background for attractive molecular targets that might be useful in preventing the progression of CVDs and heart failure (HF) as well as insights for future studies.

9.
Signal Transduct Target Ther ; 6(1): 69, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33597502

RESUMEN

MicroRNAs (miRNAs) are aberrantly expressed in the pathophysiologic process of heart failure (HF). However, the functions of a certain miRNA in different cardiac cell types during HF are scarcely reported, which might be covered by the globe effects of it on the heart. In the current study, Langendorff system was applied to isolate cardiomyocytes (CMs) and cardiac fibroblasts (CFs) from transverse aortic constriction (TAC)-induced mice. Slight increase of miR-320 expression was observed in the whole heart tissue of TAC mice. Interestingly, miR-320 was significantly elevated in CMs but decreased in CFs from TAC mice at different time points. Then, recombinant adeno-associated virus 9 with cell-type-specific promoters were used to manipulate miR-320 expressions in vivo. Both in vitro and in vivo experiments showed the miR-320 overexpression in CMs exacerbated cardiac dysfunction, whereas overexpression of miR-320 in CFs alleviated cardiac fibrosis and hypertrophy. Mechanically, downstream signaling pathway analyses revealed that miR-320 might induce various effects via targeting PLEKHM3 and IFITM1 in CMs and CFs, respectively. Moreover, miR-320 mediated effects could be abolished by PLEKHM3 re-expression in CMs or IFITM1 re-expression in CFs. Interestingly, miR-320 treated CFs were able to indirectly affect CMs function, but not vice versa. Meanwhile, upstream signaling pathway analyses showed that miR-320 expression and decay rate were rigorously manipulated by Ago2, which was regulated by a cluster of cell-type-specific TFs distinctively expressed in CMs and CFs, respectively. Together, we demonstrated that miR-320 functioned differently in various cell types of the heart during the progression of HF.


Asunto(s)
Antígenos de Diferenciación/genética , Constricción Patológica/genética , Insuficiencia Cardíaca/genética , Péptidos y Proteínas de Señalización Intracelular/genética , MicroARNs/genética , Animales , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/terapia , Proteínas Argonautas/genética , Constricción Patológica/terapia , Dependovirus/genética , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación de la Expresión Génica , Corazón/fisiopatología , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/terapia , Humanos , Ratones , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología
10.
Circ Res ; 128(11): 1708-1723, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-33550812
11.
Aging (Albany NY) ; 12(21): 22019-22045, 2020 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-33186123

RESUMEN

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disorder worldwide. Multiple metabolic disorders, such as hyperlipidemia, hyperglycemia, insulin resistance and obesity, have been reportedly associated with NAFLD, but little is known about the detailed mechanisms. METHODS AND RESULTS: Here, we explored the effects of multiple metabolic disorders, especially hyperglycemia on lipid accumulation in liver using several well-established animal models. We found that liver lipid deposition was increased in both type 1 diabetes and high-fat diet (HFD) induced hyperlipidemia models, suggesting that either hyperglycemia or hyperlipidemia alone or together was able to trigger NAFLD. Moreover, we tested whether miR-320, a miRNA promoting lipid accumulation in heart revealed by our previous study, also participated in NAFLD. Though miR-320 treatment further increased liver lipid deposition in type 1 diabetes and HFD-feeding mice, it showed no effect in leptin-receptor deficient db/db mice. Interestingly, miR-320 affected different target genes in cytosol and nucleus, respectively, which collectively led to liver lipid overload. CONCLUSIONS: Our findings illustrated the complex roles of miRNAs in subcellular fractions including nucleus and cytoplasm, which may lead to new insights into the mechanisms and treatment strategies for NAFLD in the future.


Asunto(s)
Hiperglucemia/metabolismo , Hiperlipidemias/metabolismo , MicroARNs/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Animales , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Diabetes Mellitus Experimental , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Hiperglucemia/complicaciones , Hiperlipidemias/complicaciones , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD
12.
Mol Ther Nucleic Acids ; 19: 132-143, 2020 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-31837603

RESUMEN

In recent years, systematic analyses of the subcellular distribution of microRNAs (miRNAs) suggest that the majority of miRNAs are present in both nuclear and cytoplasmic compartments. However, the full extent of nuclear miRNA function in cardiomyocytes is currently unknown. Here, subcellular fractionation, followed by the miRNA microarray, revealed that most miRNAs were detectable in both nuclear and cytoplasmic fractions of cardiomyocytes. We employed miR-320 as an example to explore the function of nucleus-localized miRNAs, finding that CRISPR-Cas9-mediated Ago2 knockdown abolished miR-320-induced transcriptional remodeling. Furthermore, nuclear Ago2 re-expression restored the effects of miR-320 in the nucleus. Moreover, liquid chromatography-mass spectrometry (LC-MS) analysis revealed the association of nuclear Ago2 with transcription factors YLP motif-containing protein 1 (Ylpm1) and single-stranded DNA binding protein 1 (Ssbp1). Intersection of the data of transcriptome-sequencing (seq) with Ago2-chromatin immunoprecipitation (ChIP)-seq revealed that the binding of Ago2 with the target promoter DNA may require promoter RNAs. Specifically, Cep57 was upregulated, whereas Fscn2 was downregulated by miR-320, and a similar effect was also observed by knockdown of their promoter RNA, respectively. Chromatin isolation by RNA purification (ChIRP) analysis showed decreased binding of the Cep57 and Fscn2 promoter RNA on their promoter DNA by miR-320 overexpression.Our work provided a preliminary idea that promoter RNA transcripts act as "pioneers" to disrupt chromatin that permits Ago2/miR-320 complexes to target Cep57 or Fscn2 promoter DNA for transcriptional regulation. miRNAs are naturally located in both cytoplasm and nucleus; however, their pathophysiological functions are largely unknown. Our work provided a theoretical basis for developing nuclear miRNA-based therapeutics against various diseases in the future.

13.
Sci China Life Sci ; 63(5): 724-736, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-31664601

RESUMEN

Although numerous miRNAs have been discovered, their functions in the different subcellular organelles have remained obscure. In this study, we found that miR-665 was enriched in the nucleus of cardiomyocytes, and then investigated the underlying role of nuclear miR-665 in heart failure. RNA fluorescence in situ hybridization assays in human heart tissue sections and primary cardiomyocytes showed that miR-665 was localized in the nucleus of cardiomyocytes. Increased expression of nuclear miR-665 was observed not only in the cardiomyocytes isolated from the heart of mice treated in vivo by transverse aortic constriction (TAC), but also in phenylephrine (PE)-treated cultured cardiomyocytes in vitro. To further explore the role of miR-665 in heart failure, a type 9 recombinant adeno-associated virus (rAAV) system was employed to manipulate the expression of miR-665 in mice. Overexpression of miR-665 aggravated TAC-induced cardiac dysfunction, while down-expression of miR-665 showed opposite effects. Bioinformatic prediction and biological validation confirmed that the PTEN (phosphatase and tensin homolog) gene was one of the targets of miR-665 in the nucleus. Furthermore, restoring PTEN expression significantly eliminated the destructive effects of miR-665 over-expression in TAC-induced cardiac dysfunction. Our data showed that nuclear miR-665 aggravates heart failure via inhibiting PTEN expression, which provided a therapeutic approach for heart failure.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Tensinas/metabolismo , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Corazón , Humanos , Hibridación Fluorescente in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Miocitos Cardíacos/citología , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo
14.
Circ Res ; 125(12): 1106-1120, 2019 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-31638474

RESUMEN

RATIONALE: Diabetes mellitus is often associated with cardiovascular complications, which is the leading cause of morbidity and mortality among patients with diabetes mellitus, but little is known about the mechanism that connects diabetes mellitus to the development of cardiovascular dysfunction. OBJECTIVE: We aim to elucidate the mechanism underlying hyperglycemia-induced cardiac dysfunction on a well-established db/db mouse model for diabetes mellitus and diabetic complications that lead to heart failure. METHODS AND RESULTS: We first profiled the expression of microRNAs (miRNAs) by microarray and quantitative reverse transcription polymerase chain reaction on db/db mice and identified miR-320 as a key miRNA associated with the disease phenotype. We next established the clinical relevance of this finding by showing the upregulation of the same miRNA in the failing heart of patients with diabetes mellitus. We demonstrated the causal role of miR-320 in inducing diabetic cardiomyopathy, showing that miR-320 overexpression exacerbated while its inhibition improved the cardiac phenotype in db/db mice. Unexpectedly, we found that miR-320 acts as a small activating RNA in the nucleus at the level of transcription. By chromatin immunoprecipitation sequencing and chromatin immunoprecipitation quantitive polymerase chain reaction analysis of Ago2 (argonaute RISC catalytic component 2) and RNA polymerase II in response to miR-320 induction, we identified CD36 (fatty acid translocase) as a key target gene for this miRNA and showed that the induced expression of CD36 is responsible for increased fatty acid uptake, thereby causing lipotoxicity in the heart. CONCLUSIONS: These findings uncover a novel mechanism for diabetes mellitus-triggered cardiac dysfunction, provide an endogenous case for small activating RNA that has been demonstrated to date only with synthetic RNAs in transfected cells, and suggest a potential strategy to develop a miRNA-based therapy to treat diabetes mellitus-associated cardiovascular complications.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Cardiomiopatías Diabéticas/metabolismo , Ácidos Grasos/metabolismo , MicroARNs/biosíntesis , Miocitos Cardíacos/metabolismo , Activación Transcripcional/fisiología , Animales , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células Cultivadas , Diabetes Mellitus Experimental/genética , Cardiomiopatías Diabéticas/genética , Ácidos Grasos/genética , Humanos , Masculino , Ratones , MicroARNs/genética , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA