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










Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(3): e2310711121, 2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38190531

RESUMEN

Brown adipose tissue (BAT) is the main site of nonshivering thermogenesis which plays an important role in thermogenesis and energy metabolism. However, the regulatory factors that inhibit BAT activity remain largely unknown. Here, cardiotrophin-like cytokine factor 1 (CLCF1) is identified as a negative regulator of thermogenesis in BAT. Adenovirus-mediated overexpression of CLCF1 in BAT greatly impairs the thermogenic capacity of BAT and reduces the metabolic rate. Consistently, BAT-specific ablation of CLCF1 enhances the BAT function and energy expenditure under both thermoneutral and cold conditions. Mechanistically, adenylate cyclase 3 (ADCY3) is identified as a downstream target of CLCF1 to mediate its role in regulating thermogenesis. Furthermore, CLCF1 is identified to negatively regulate the PERK-ATF4 signaling axis to modulate the transcriptional activity of ADCY3, which activates the PKA substrate phosphorylation. Moreover, CLCF1 deletion in BAT protects the mice against diet-induced obesity by promoting BAT activation and further attenuating impaired glucose and lipid metabolism. Therefore, our results reveal the essential role of CLCF1 in regulating BAT thermogenesis and suggest that inhibiting CLCF1 signaling might be a potential therapeutic strategy for improving obesity-related metabolic disorders.


Asunto(s)
Tejido Adiposo Pardo , Metabolismo Energético , Animales , Ratones , Adenoviridae , Interleucinas , Obesidad/genética , Termogénesis/genética
2.
J Zhejiang Univ Sci B ; 24(8): 682-697, 2023 Aug 15.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-37551555

RESUMEN

Cardiac fibrosis is a cause of morbidity and mortality in people with heart disease. Anti-fibrosis treatment is a significant therapy for heart disease, but there is still no thorough understanding of fibrotic mechanisms. This study was carried out to ascertain the functions of cytokine receptor-like factor 1 (CRLF1) in cardiac fibrosis and clarify its regulatory mechanisms. We found that CRLF1 was expressed predominantly in cardiac fibroblasts. Its expression was up-regulated not only in a mouse heart fibrotic model induced by myocardial infarction, but also in mouse and human cardiac fibroblasts provoked by transforming growth factor-|ß1 (TGF|-|ß1). Gain- and loss-of-function experiments of CRLF1 were carried out in neonatal mice cardiac fibroblasts (NMCFs) with or without TGF-|ß1 stimulation. CRLF1 overexpression increased cell viability, collagen production, cell proliferation capacity, and myofibroblast transformation of NMCFs with or without TGF|-|ß1 stimulation, while silencing of CRLF1 had the opposite effects. An inhibitor of the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway and different inhibitors of TGF-|ß1 signaling cascades, comprising mothers against decapentaplegic homolog (SMAD)|-dependent and SMAD-independent pathways, were applied to investigate the mechanisms involved. CRLF1 exerted its functions by activating the ERK1/2 signaling pathway. Furthermore, the SMAD-dependent pathway, not the SMAD-independent pathway, was responsible for CRLF1 up-regulation in NMCFs treated with TGF-|ß1. In summary, activation of the TGF-|ß1/SMAD signaling pathway in cardiac fibrosis increased CRLF1 expression. CRLF1 then aggravated cardiac fibrosis by activating the ERK1/2 signaling pathway. CRLF1 could become a novel potential target for intervention and remedy of cardiac fibrosis.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Infarto del Miocardio , Receptores de Citocinas , Animales , Humanos , Ratones , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibrosis , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Infarto del Miocardio/metabolismo , Receptores de Citocinas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/farmacología
3.
J Biol Chem ; 298(6): 101946, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35447114

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is one of the most common chronic liver diseases worldwide. However, the molecular mechanisms that promote dysregulation of hepatic triglyceride metabolism and lead to NAFLD are poorly understood, and effective treatments are limited. Leukemia inhibitory factor (LIF) is a member of the interleukin-6 cytokine family and has been shown to regulate a variety of physiological processes, although its role in hepatic triglyceride metabolism remains unknown. In the present study, we measured circulating LIF levels by ELISA in 214 patients with biopsy-diagnosed NAFLD as well as 314 normal control patients. We further investigated the potential role and mechanism of LIF on hepatic lipid metabolism in obese mice. We found that circulating LIF levels correlated with the severity of liver steatosis. Patients with ballooning, fibrosis, lobular inflammation, and abnormally elevated liver injury markers alanine transaminase and aspartate aminotransferase also had higher levels of serum LIF than control patients. Furthermore, animal studies showed that white adipose tissue-derived LIF could ameliorate liver steatosis through activation of hepatic LIF receptor signaling pathways. Together, our results suggested that targeting LIF-LIF receptor signaling might be a promising strategy for treating NAFLD.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Animales , Humanos , Factor Inhibidor de Leucemia/sangre , Factor Inhibidor de Leucemia/metabolismo , Hígado/patología , Ratones , Ratones Obesos , Enfermedad del Hígado Graso no Alcohólico/sangre , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Triglicéridos/metabolismo
4.
Basic Res Cardiol ; 116(1): 15, 2021 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-33675440

RESUMEN

Long non-coding RNAs (lncRNAs) account for a large proportion of genomic transcripts and are critical regulators in various cardiac diseases. Though lncRNAs have been reported to participate in the process of diverse cardiac diseases, the contribution of lncRNAs in cardiac fibrosis remains to be fully elucidated. Here, we identified a novel anti-fibrotic lncRNA, SAIL (scaffold attachment factor B interacting lncRNA). SAIL was reduced in cardiac fibrotic tissue and activated cardiac fibroblasts. Gain- and loss-of-function studies showed that knockdown of SAIL promoted proliferation and collagen production of cardiac fibroblasts with or without TGF-ß1 (transforming growth factor beta1) treatment, while overexpression of SAIL did the opposite. In mouse cardiac fibrosis induced by myocardial infarction, knockdown of SAIL exacerbated, whereas overexpression of SAIL alleviated cardiac fibrosis. Mechanically, SAIL inhibited the fibrotic process by directly binding with SAFB via 23 conserved nucleotide sequences, which in turn blocked the access of SAFB to RNA pol II (RNA polymerase II) and reduced the transcription of fibrosis-related genes. Intriguingly, the human conserved fragment of SAIL (hSAIL) significantly suppressed the proliferation and collagen production of human cardiac fibroblasts. Our findings demonstrate that SAIL regulates cardiac fibrosis by regulating SAFB-mediated transcription of fibrotic related genes. Both SAIL and SAFB hold the potential to become novel therapeutic targets for cardiac fibrosis.


Asunto(s)
Proliferación Celular , Colágeno/metabolismo , Fibroblastos/metabolismo , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , ARN Largo no Codificante/metabolismo , Animales , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Fibroblastos/patología , Fibrosis , Masculino , Ratones Endogámicos C57BL , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Miocitos Cardíacos/patología , ARN Polimerasa II/metabolismo , ARN Largo no Codificante/genética , Proteínas de Unión al ARN/metabolismo , Transcripción Genética
5.
Life Sci ; 268: 119011, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33421522

RESUMEN

AIMS: Obesity is recognized as a risk factor for many metabolic disorders, particularly nonalcoholic fatty liver disease (NAFLD). However, the underlying mechanism is still poorly understood. Several lines of evidence indicate that microRNA (miRNA) is a key regulator of lipid metabolism. In this study, we investigated the role of miR-183-5p in the development of NAFLD. METHODS: The expression levels of miR-183-5p and B-cell translocation gene 1 (Btg1) were determined by quantitative real-time PCR and histological analysis in livers of obese mice and cell models induced with palmitic acid (PA), respectively. AML12 cells were treated with PA in the presence or absence of miR-183-5p mimics or inhibitor. Moreover, a Luciferase reporter assay was used to determine whether Btg1 is the direct target of miR-183-5p. Protein levels of BTG1 were estimated using western blotting. KEY FINDINGS: Expression of miR-183-5p was increased in the livers of three murine models and also in the AML12 cell model. Overexpression of miR-183-5p in the cell model and mice led to hepatic triglyceride (TG) accumulation and upregulation of lipogenic genes, whereas inhibition of miR-183-5p in the cell model improved hepatic TG accumulation. Mechanistically, we further identified Btg1 as a direct target gene of miR-183-5p. SIGNIFICANCE: Our findings revealed that miR-183-5p affected the regulation of hepatic TG homeostasis, which may provide a potential therapeutic target for hepatosteatosis.


Asunto(s)
Hígado/metabolismo , MicroARNs/genética , Proteínas de Neoplasias/genética , Obesidad/genética , Triglicéridos/metabolismo , Animales , Línea Celular , Regulación de la Expresión Génica , Hepatocitos/metabolismo , Metabolismo de los Lípidos/genética , Hígado/fisiología , Masculino , Ratones Endogámicos C57BL , Ratones Mutantes , Proteínas de Neoplasias/metabolismo , Obesidad/patología , Receptores de Leptina/genética , Triglicéridos/genética
6.
J Cell Mol Med ; 24(1): 737-746, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31749326

RESUMEN

Cardiomyocyte apoptosis is an important process occurred during cardiac ischaemia-reperfusion injury. Long non-coding RNAs (lncRNA) participate in the regulation of various cardiac diseases including ischaemic reperfusion (I/R) injury. In this study, we explored the potential role of lncRNA ACART (anti-cardiomyocyte apoptosis-related transcript) in cardiomyocyte injury and the underlying mechanism for the first time. We found that ACART was significantly down-regulated in cardiac tissue of mice subjected to I/R injury or cultured cardiomyocytes treated with hydrogen peroxide (H2 O2 ). Knockdown of ACART led to significant cardiomyocyte injury as indicated by reduced cell viability and increased apoptosis. In contrast, overexpression of ACART enhanced cell viability and reduced apoptosis of cardiomyocytes treated with H2 O2 . Meanwhile, ACART increased the expression of the B cell lymphoma 2 (Bcl-2) and suppressed the expression of Bcl-2-associated X (Bax) and cytochrome-C (Cyt-C). In addition, PPAR-γ was up-regulated by ACART and inhibition of PPAR-γ abolished the regulatory effects of ACART on cell apoptosis and the expression of Bcl-2, Bax and Cyt-C under H2 O2 treatment. However, the activation of PPAR-γ reversed the effects of ACART inhibition. The results demonstrate that ACART protects cardiomyocyte injury through modulating the expression of Bcl-2, Bax and Cyt-C, which is mediated by PPAR-γ activation. These findings provide a new understanding of the role of lncRNA ACART in regulation of cardiac I/R injury.


Asunto(s)
Apoptosis , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/citología , PPAR gamma/metabolismo , Sustancias Protectoras , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Largo no Codificante/genética , Animales , Supervivencia Celular , Masculino , Ratones , Ratones Endogámicos C57BL , Daño por Reperfusión Miocárdica/etiología , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/metabolismo , PPAR gamma/genética , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-bcl-2/genética
7.
Mol Ther Nucleic Acids ; 17: 297-309, 2019 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-31284127

RESUMEN

Ischemic cardiac disease is the leading cause of morbidity and mortality in the world. Despite the great efforts and progress in cardiac research, the current treatment of cardiac ischemia reperfusion injury (I/RI) is still far from being satisfactory. This study was performed to investigate the role of long non-coding RNA (lncRNA) H19 in regulating myocardial I/RI. We found that H19 expression was downregulated in the I/R hearts of mice and cardiomyocytes treated with H2O2. Overexpression of H19 alleviated myocardial I/RI of mice and cardiomyocyte injury induced by H2O2. We found that H19 functioned as a competing endogenous RNA of miR-877-3p, which decreased the expression of miR-877-3p through the base-pairing mechanism. In parallel, miR-877-3p was upregulated in H2O2-treated cardiomyocytes and mouse ischemia reperfusion (I/R) hearts. miR-877-3p exacerbated myocardial I/RI and cardiomyocyte apoptosis. We further established Bcl-2 as a downstream target of miR-877-3p. miR-877-3p inhibited the mRNA and protein expression of Bcl-2. Furthermore, H19 decreased the Bcl-2/Bax ratio at mRNA and protein levels, cytochrome c release, and activation of caspase-9 and caspase-3 in myocardial I/RI mice, which were canceled by miR-877-3p. In summary, the H19/miR-877-3p/Bcl-2 pathway is involved in regulation of mitochondrial apoptosis during myocardial I/RI, which provided new insight into molecular mechanisms underlying regulation of myocardial I/RI.

8.
J Mol Cell Cardiol ; 133: 188-198, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31220469

RESUMEN

Long noncoding RNAs (lncRNAs) are a class of novel molecular regulators in cardiac development and diseases. However, the role of specific lncRNAs in cardiac fibrosis remains to be fully explored. The aim of the present study was to investigate the effects and underlying mechanisms of lncRNA PCFL (pro-cardiac fibrotic lncRNA) on cardiac fibrosis after myocardial infarction (MI). Cardiac fibroblasts (CFs) with gain and loss of function of PCFL and mice with global knockout or overexpression of PCFL were used to explore the effects of PCFL on cardiac fibrosis. The data showed that PCFL was significantly increased in hearts of mice subjected to MI and CFs treated with transforming growth factor-ß1 (TGF-ß1). Overexpression of PCFL promoted collagen production and CF proliferation, while silencing PCFL exhibited the opposite effects. Compared with wild type MI mice, heterozygous knockout of PCFL (PCFL+/-) in mice significantly improved heart function and reduced cardiac fibrosis after MI. While overexpression of PCFL impaired cardiac function and aggravated MI-induced cardiac fibrosis. The mechanistic data demonstrated that PCFL functioned as a sponge of miR-378. Luciferase reporter assay confirmed the interaction of PCFL with miR-378. MiR-378 inhibited collagen production by suppressing its target gene, GRB2 (growth factor receptor bound protein 2). Knockdown of PCFL led to an increase of miR-378. Silencing of miR-378 reserved the inhibitory effects of PCFL knockdown on collagen production, cell proliferation and GRB2 expression. In conclusion, the study identifies a novel pro-fibrotic lncRNA, PCFL, and the mechanism involves the direct interaction of PCFL with miR-378, which in turn relieves the inhibition effect of miR-378 on GRB2 and promotes cardiac fibrosis.


Asunto(s)
Proteína Adaptadora GRB2/genética , MicroARNs/genética , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Interferencia de ARN , ARN Largo no Codificante/genética , Regiones no Traducidas 3' , Animales , Proliferación Celular , Colágeno/metabolismo , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibrosis , Regulación de la Expresión Génica , Silenciador del Gen , Heterocigoto , Masculino , Ratones , Ratones Noqueados , Infarto del Miocardio/metabolismo , Transducción de Señal
9.
Sci Rep ; 7: 42657, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28198439

RESUMEN

A long non-coding RNA (lncRNA), named myocardial infarction associated transcript (MIAT), has been documented to confer risk of myocardial infarction (MI). The aim of this study is to elucidate the pathophysiological role of MIAT in regulation of cardiac fibrosis. In a mouse model of MI, we found that MIAT was remarkably up-regulated, which was accompanied by cardiac interstitial fibrosis. MIAT up-regulation in MI was accompanied by deregulation of some fibrosis-related regulators: down-regulation of miR-24 and up-regulation of Furin and TGF-ß1. Most notably, knockdown of endogenous MIAT by its siRNA reduced cardiac fibrosis and improved cardiac function and restored the deregulated expression of the fibrosis-related regulators. In cardiac fibroblasts treated with serum or angiotensin II, similar up-regulation of MIAT and down-regulation of miR-24 were consistently observed. These changes promoted fibroblasts proliferation and collagen accumulation, whereas knockdown of MIAT by siRNA or overexpression of miR-24 with its mimic abrogated the fibrogenesis. Our study therefore has identified MIAT as the first pro-fibrotic lncRNA in heart and unraveled the role of MIAT in the pathogenesis of MI. These findings also promise that normalization of MIAT level may prove to be a therapeutic option for the treatment of MI-induced cardiac fibrosis and the associated cardiac dysfunction.


Asunto(s)
Infarto del Miocardio/genética , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocardio/patología , ARN Largo no Codificante/genética , Animales , Proliferación Celular , Supervivencia Celular , Colágeno/metabolismo , Modelos Animales de Enfermedad , Ecocardiografía , Fibroblastos/metabolismo , Fibrosis , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Pruebas de Función Cardíaca , Masculino , Ratones , MicroARNs , Infarto del Miocardio/diagnóstico , Infarto del Miocardio/fisiopatología , ARN Interferente Pequeño/genética
10.
Int J Biol Sci ; 13(1): 22-31, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28123343

RESUMEN

Background: MicroRNAs (miRNAs) have been emerged as important regulator in a multiple of cardiovascular disease, including arrhythmia, cardiac hypertrophy and fibrosis, and myocardial infarction. The aim of this study was to investigate whether miRNA let-7a has antihypertrophic effects in angiotensin II (AngII)-induced cardiac hypertrophy. Methods: Neonatal rat ventricular myocytes (NRVMs) were exposed to AngII for 36 h as a cellular model of hypertrophy; subcutaneous injection of AngII for 2 weeks was used to establish a mouse model of cardiac hypertrophy in vivo study. Cell surface area (CSA) was measured by immunofluorescence cytochemistry; expression of hypertrophy-related genes ANP, BNP, ß-MHC was detected by Real-time PCR; luciferase activity assay was performed to confirm the miRNA's binding site in the calmodulin (CaM) gene; CaM protein was detected by Western blot; the hypertrophy parameters were measured by echocardiographic assessment. Results: The expression of let-7a was decreased in AngII-induced cardiac hypertrophy in vitro and in vivo. Overexpression of let-7a attenuated AngII-induced increase of cell surface area and repressed the increased mRNA levels of ANP, BNP and ß-MHC. Dual-luciferase reporter assay showed that let-7a could bind to the 3'UTR of CaM 1 gene. Let-7a downregulated the expression of CaM protein. In vivo, let-7a produced inhibitory effects on cardiac hypertrophy, including the downregulation of cross-sectional area of cardiomyocytes in mouse heart, the reduction of IVSD and LVPWD, the suppression of hypertrophy marker genes ANP, BNP, ß-MHC mRNA level, and the downregulation of CaM protein level. Conclusions: let-7a possesses a prominent anti-hypertrophic property by targeting CaM genes. The findings provide new insight into molecular mechanism of cardiac hypertrophy.


Asunto(s)
Calmodulina/metabolismo , Cardiomegalia/genética , Cardiomegalia/metabolismo , MicroARNs/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Regiones no Traducidas 3'/genética , Angiotensina II/farmacología , Animales , Factor Natriurético Atrial/genética , Calmodulina/genética , Cardiomegalia/inducido químicamente , Células Cultivadas , MicroARNs/genética , Péptido Natriurético Encefálico/genética , ARN Mensajero/genética , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa
11.
Oxid Med Cell Longev ; 2016: 7524308, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27703600

RESUMEN

In the present study, we demonstrated that bone marrow mesenchymal stem cells (BMSCs) of the 3rd passage displayed the senescence-associated phenotypes characterized with increased activity of SA-ß-gal, altered autophagy, and increased G1 cell cycle arrest, ROS production, and expression of p53 and p21Cip1/Waf1 compared with BMSCs of the 1st passage. Cholesterol (CH) reduced the number of SA-ß-gal positive cells in a dose-dependent manner in aging BMSCs induced by H2O2 and the 3rd passage BMSCs. Moreover, CH inhibited the production of ROS and expression of p53 and p21Cip1/Waf1 in both cellular senescence models and decreased the percentage of BMSCs in G1 cell cycle in the 3rd passage BMSCs. CH prevented the increase in SA-ß-gal positive cells induced by RITA (reactivation of p53 and induction of tumor cell apoptosis, a p53 activator) or 3-MA (3-methyladenine, an autophagy inhibitor). Our results indicate that CH not only is a structural component of cell membrane but also functionally contributes to regulating cellular senescence by modulating cell cycle, autophagy, and the ROS/p53/p21Cip1/Waf1 signaling pathway.


Asunto(s)
Autofagia/efectos de los fármacos , Células de la Médula Ósea/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Colesterol/farmacología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Furanos/farmacología , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
12.
Biosci Rep ; 36(3)2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27129287

RESUMEN

The present study aimed to investigate whether long non-coding RNAs (lncRNAs) are involved in cardiac fibrogenesis induced by myocardial infarction (MI). The differentially expressed lncRNAs and mRNAs in peri-infarct region of mice 4 weeks after MI were selected for bioinformatic analysis including gene ontology (GO) enrichment, pathway and network analysis. Left ventricular tissue levels of lncRNAs and mRNAs were compared between MI and sham control mice, using a false discovery rate (FDR) of <5%. Out of 55000 lncRNAs detected, 263 were significantly up-regulated and 282 down-regulated. Out of 23000 mRNAs detected, 142 were significantly up-regulated and 67 down-regulated. Among the differentially expressed lncRNAs, 53 were up-regulated by ≥2.0-fold change and 37 down-regulated by ≤0.5-fold change. Nine up-regulated and five down-regulated lncRNAs were randomly selected for quantitative real-time PCR (qRT-PCR) verification. GO and pathway analyses revealed 173 correlated lncRNA-mRNA pairs for 57 differentially expressed lncRNAs and 20 differentially expressed genes which are related to the development of cardiac fibrosis. We identified TGF-ß3 as the top-ranked gene, a critical component of the transforming growth factor-ß (TGF-ß) and mitogen activated protein kinase (MAPK) signalling pathways in cardiac fibrosis. NONMMUT022554 was identified as the top-ranked lncRNA, positively correlated with six up-regulated genes, which are involved in the extracellular matrix (ECM)-receptor interactions and the phosphoinositid-3 kinase/protein kinase B (PI3K-Akt) signalling pathway. Our study has identified the expression signature of lncRNAs in cardiac fibrosis induced by MI and unravelled the possible involvement of the deregulated lncRNAs in cardiac fibrosis and the associated pathological processes.


Asunto(s)
Infarto del Miocardio/genética , ARN Largo no Codificante/biosíntesis , ARN Mensajero/biosíntesis , Factor de Crecimiento Transformador beta3/biosíntesis , Animales , Biología Computacional , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes/genética , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Humanos , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Infarto del Miocardio/patología , ARN Largo no Codificante/genética , ARN Mensajero/genética , Transducción de Señal , Factor de Crecimiento Transformador beta3/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...