Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 79(3): 425-442.e7, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32615088

RESUMEN

Double-strand breaks (DSBs) are the most deleterious DNA lesions, which, if left unrepaired, may lead to genome instability or cell death. Here, we report that, in response to DSBs, the RNA methyltransferase METTL3 is activated by ATM-mediated phosphorylation at S43. Phosphorylated METTL3 is then localized to DNA damage sites, where it methylates the N6 position of adenosine (m6A) in DNA damage-associated RNAs, which recruits the m6A reader protein YTHDC1 for protection. In this way, the METTL3-m6A-YTHDC1 axis modulates accumulation of DNA-RNA hybrids at DSBs sites, which then recruit RAD51 and BRCA1 for homologous recombination (HR)-mediated repair. METTL3-deficient cells display defective HR, accumulation of unrepaired DSBs, and genome instability. Accordingly, depletion of METTL3 significantly enhances the sensitivity of cancer cells and murine xenografts to DNA damage-based therapy. These findings uncover the function of METTL3 and YTHDC1 in HR-mediated DSB repair, which may have implications for cancer therapy.


Asunto(s)
Adenosina/análogos & derivados , Neoplasias de Cabeza y Cuello/genética , Metiltransferasas/genética , Proteínas del Tejido Nervioso/genética , Factores de Empalme de ARN/genética , Reparación del ADN por Recombinación/efectos de los fármacos , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Adenosina/metabolismo , Animales , Antibióticos Antineoplásicos/farmacología , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Bleomicina/farmacología , Línea Celular Tumoral , ADN/genética , ADN/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Células HEK293 , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/mortalidad , Neoplasias de Cabeza y Cuello/patología , Humanos , Metiltransferasas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas del Tejido Nervioso/metabolismo , Hibridación de Ácido Nucleico , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoblastos/patología , Fosforilación , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Factores de Empalme de ARN/metabolismo , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Ribonucleasa H/genética , Ribonucleasa H/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/mortalidad , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
2.
FASEB J ; 37(12): e23284, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37905981

RESUMEN

Cellular apoptosis is a central mechanism leveraged by chemotherapy to treat human cancers. 5-Methylcytosine (m5C) modifications installed on both DNA and mRNA are documented to regulate apoptosis independently. However, the interplay or crosstalk between them in cellular apoptosis has not yet been explored. Here, we reported that promoter methylation by DNMT1 coordinated with mRNA methylation by NSun2 to regulate osteosarcoma cell apoptosis. DNMT1 was induced during osteosarcoma cell apoptosis triggered by chemotherapeutic drugs, whereas NSun2 expression was suppressed. DNMT1 was found to repress NSun2 expression by methylating the NSun2 promoter. Moreover, DNMT1 and NSun2 regulate the anti-apoptotic genes AXL, NOTCH2, and YAP1 through DNA and mRNA methylation, respectively. Upon exposure to cisplatin or doxorubicin, DNMT1 elevation drastically reduced the expression of these anti-apoptotic genes via enhanced promoter methylation coupled with NSun2 ablation-mediated attenuation of mRNA methylation, thus rendering osteosarcoma cells to apoptosis. Collectively, our findings establish crosstalk of importance between DNA and RNA cytosine methylations in determining osteosarcoma resistance to apoptosis during chemotherapy, shedding new light on future treatment of osteosarcoma, and adding additional layers to the control of gene expression at different epigenetic levels.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Humanos , Metilación , ARN Mensajero/genética , Citosina , ADN , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/genética , Apoptosis/genética
3.
Nucleic Acids Res ; 49(5): 2848-2858, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33589924

RESUMEN

The ubiquitous RNA-binding protein HuR (ELAVL1) promotes telomerase activity by associating with the telomerase noncoding RNA TERC. However, the role of the neural-specific members HuB, HuC, and HuD (ELAVL2-4) in telomerase activity is unknown. Here, we report that HuB and HuD, but not HuC, repress telomerase activity in human neuroblastoma cells. By associating with AU-rich sequences in TERC, HuB and HuD repressed the assembly of the TERT-TERC core complex. Furthermore, HuB and HuD competed with HuR for binding to TERC and antagonized the function of HuR that was previously shown to enhance telomerase activity to promote cell growth. Our findings reveal a novel mechanism controlling telomerase activity in human neuroblastoma cells that involves a competition between HuR and the related, neural-specific proteins HuB and HuD.


Asunto(s)
Proteína 1 Similar a ELAV/metabolismo , Proteína 2 Similar a ELAV/metabolismo , Proteína 4 Similar a ELAV/metabolismo , ARN/metabolismo , Telomerasa/metabolismo , Línea Celular Tumoral , Senescencia Celular , Proteína 1 Similar a ELAV/antagonistas & inhibidores , Humanos
4.
J Biol Chem ; 296: 100080, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33199370

RESUMEN

Post-transcriptional regulation of mRNA translation and stability is primarily achieved by RNA-binding proteins, which are of increasing importance for heart function. Furthermore, G-quadruplex (G4) and G4 resolvase activity are involved in a variety of biological processes. However, the role of G4 resolvase activity in heart function remains unknown. The present study aims to investigate the role of RNA helicase associated with adenylate- and uridylate-rich element (RHAU), an RNA-binding protein with G4 resolvase activity in postnatal heart function through deletion of Rhau in the cardiomyocytes of postnatal mice. RHAU-deficient mice displayed progressive pathological remodeling leading to heart failure and mortality and impaired neonatal heart regeneration. RHAU ablation reduced the protein levels but enhanced mRNA levels of Yap1 and Hexim1 that are important regulators for heart development and postnatal heart function. Furthermore, RHAU was found to associate with both the 5' and 3' UTRs of these genes to destabilize mRNA and enhance translation. Thus, we have demonstrated the important functions of RHAU in the dual regulation of mRNA translation and stability, which is vital for heart physiology.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , ARN Mensajero/metabolismo , Recombinasas/metabolismo , Regiones no Traducidas 3'/genética , Regiones no Traducidas 3'/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Sitios de Unión , Western Blotting , Línea Celular , Biología Computacional , ARN Helicasas DEAD-box/genética , Ecocardiografía , Células HEK293 , Humanos , Ratones , Biosíntesis de Proteínas/genética , Biosíntesis de Proteínas/fisiología , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , RNA-Seq , Recombinasas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Señalizadoras YAP
5.
Cancer Sci ; 113(4): 1154-1167, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35178834

RESUMEN

The AU-rich binding factor 1 (AUF1) is one of the well known adenylate-uridylate-rich element (ARE)-specific RNA-binding proteins (ARE-BPs) for which dysregulation has been reported in various human cancers. However, the involvement of AUF1 in the initiation and progression of hepatocellular carcinoma (HCC) is still elusive. In this study, we aimed at exploring the clinical significance, function, and mechanism of the abnormal expression of AUF1 in HCC. Using a bioinformatics analysis of The Cancer Genome Atlas (TCGA) and Liver Cancer Institute (LCI) database, we identified that AUF1 was abnormally highly expressed in HCC tissues and that the high expression of AUF1 was correlated with poor prognosis in patients with HCC. We also confirmed the increased AUF1 expression and its prognostic value in our HBV-related HCC cohorts. AUF1 overexpression in hepatoma cells promoted cell proliferation and increased the resistance of hepatoma cells toward doxorubicin, whereas knockdown of AUF1 exerted the opposite effects. Mechanistically, we demonstrated that AKR1B10 was a critical target of AUF1 and was essential for sustaining the AUF1-induced proliferation and drug resistance of hepatoma cells. AUF1 increased AKR1B10 expression by binding to the 3'UTR region of AKR1B10 mRNA and stabilizing AKR1B10 mRNA. Additionally, we demonstrated that E2F1 enhanced AUF1 expression in HCC at the transcription level. Our study revealed a novel role of AUF1 in promoting the development and drug resistance of HCC via the post-transcriptional regulation of AKR1B10 expression. The E2F1/AUF1/AKR1B10 axis can serve as a potential therapeutic target in HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Aldo-Ceto Reductasas/genética , Aldo-Ceto Reductasas/metabolismo , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Resistencia a Medicamentos , Factor de Transcripción E2F1/metabolismo , Ribonucleoproteína Nuclear Heterogénea D0 , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , ARN Mensajero/genética , Regulación hacia Arriba
6.
Cell Mol Life Sci ; 78(4): 1709-1727, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32734582

RESUMEN

Abdominal aortic aneurysm (AAA) is characterized by inflammatory cell infiltration and aggravated by hyperhomocysteinemia (HHcy). It is unknown whether the homocysteine (Hcy)-activated RNA methyltransferase NOP2/Sun domain family member 2 (NSun2) is associated with AAA. Here, we found that NSun2 deficiency significantly attenuated elastase-induced and HHcy-aggravated murine AAA with decreased T cell infiltration in the vessel walls. T cell labeling and adoptive transfer experiments confirmed that NSun2 deficiency inhibited the chemotaxis of vessels to T cells. RNA sequencing of endothelial cells showed that Hcy induced the accumulation of various metabolic enzymes of the phospholipid PC-LPC-LPA metabolic pathway, especially autotaxin (ATX). In the elastase-induced mouse model of AAA, ATX was specifically expressed in the endothelium and the plasma ATX concentration was upregulated and even higher in the HHcy group, which were decreased dramatically by NSun2 knockdown. In vitro Transwell experiments showed that ATX dose-dependently promoted T cell migration. HHcy may upregulate endothelial ATX expression and secretion and in turn recruit T cells into the vessel walls to induce vascular inflammation and consequently accelerate the pathogenesis of AAA. Mechanistically, secreted ATX interacted with T cells by binding to integrin α4, which subsequently activated downstream FAK/Src-RhoA signaling pathways and then induced T cell chemokinesis and adhesion. ATX overexpression in the vessel walls reversed the inhibited development of AAA in the NSun2-deficient mice. Therefore, NSun2 mediates the development of HHcy-aggravated AAA primarily by increasing endothelial ATX expression, secretion and T cell migration, which is a novel mechanism for HHcy-aggravated vascular inflammation and pathogenesis of AAA.


Asunto(s)
Aneurisma de la Aorta Abdominal/genética , Hiperhomocisteinemia/genética , Inflamación/genética , Metiltransferasas/genética , Hidrolasas Diéster Fosfóricas/genética , Animales , Aneurisma de la Aorta Abdominal/complicaciones , Aneurisma de la Aorta Abdominal/inmunología , Aneurisma de la Aorta Abdominal/patología , Movimiento Celular/genética , Modelos Animales de Enfermedad , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Regulación de la Expresión Génica/genética , Humanos , Hiperhomocisteinemia/complicaciones , Hiperhomocisteinemia/patología , Inflamación/complicaciones , Inflamación/patología , Ratones , Transducción de Señal/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo
7.
Circ Res ; 125(7): 707-719, 2019 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-31412728

RESUMEN

RATIONALE: PGC1α (peroxisome proliferator-activated receptor gamma coactivator 1α) represents an attractive target interfering bioenergetics and mitochondrial homeostasis, yet multiple attempts have failed to upregulate PGC1α expression as a therapy, for instance, causing cardiomyopathy. OBJECTIVE: To determine whether a fine-tuning of PGC1α expression is essential for cardiac homeostasis in a context-dependent manner. METHODS AND RESULTS: Moderate cardiac-specific PGC1α overexpression through a ROSA26 locus knock-in strategy was utilized in WT (wild type) mice and in G3Terc-/- (third generation of telomerase deficient; hereafter as G3) mouse model, respectively. Ultrastructure, mitochondrial stress, echocardiographic, and a variety of biological approaches were applied to assess mitochondrial physiology and cardiac function. While WT mice showed a relatively consistent PGC1α expression from 3 to 12 months old, age-matched G3 mice exhibited declined PGC1α expression and compromised mitochondrial function. Cardiac-specific overexpression of PGC1α (PGC1αOE) promoted mitochondrial and cardiac function in 3-month-old WT mice but accelerated cardiac aging and significantly shortened life span in 12-month-old WT mice because of increased mitochondrial damage and reactive oxygen species insult. In contrast, cardiac-specific PGC1α knock in in G3 (G3 PGC1αOE) mice restored mitochondrial homeostasis and attenuated senescence-associated secretory phenotypes, thereby preserving cardiac performance with age and extending health span. Mechanistically, age-dependent defect in mitophagy is associated with accumulation of damaged mitochondria that leads to cardiac impairment and premature death in 12-month-old WT PGC1αOE mice. In the context of telomere dysfunction, PGC1α induction replenished energy supply through restoring the compromised mitochondrial biogenesis and thus is beneficial to old G3 heart. CONCLUSIONS: Fine-tuning the expression of PGC1α is crucial for the cardiac homeostasis because the balance between mitochondrial biogenesis and clearance is vital for regulating mitochondrial function and homeostasis. These results reinforce the importance of carefully evaluating the PGC1α-boosting strategies in a context-dependent manner to facilitate clinical translation of novel cardioprotective therapies.


Asunto(s)
Longevidad , Miocitos Cardíacos/metabolismo , Biogénesis de Organelos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Animales , Células Cultivadas , Femenino , Homeostasis , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias Cardíacas/metabolismo , Miocitos Cardíacos/fisiología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Especies Reactivas de Oxígeno/metabolismo , Telomerasa/genética , Telomerasa/metabolismo
8.
J Clin Lab Anal ; 35(4): e23724, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33543804

RESUMEN

BACKGROUND: This article is to explore changes in levels of coagulation parameters in different trimesters among healthy pregnant women in China. METHODS: A total of 760 eligible women were enrolled (first-trimester group: n = 183, second-trimester group: n = 183, third-trimester group: n = 263, non-pregnant group: n = 131). Seven parameters including prothrombin time (PT), activated partial thromboplastin time (APTT), thrombin time (TT), fibrinogen (FIB), D-dimer (DD), fibrinogen degradation products (FDP), and antithrombin III (ATIII), of all participants were collected. The non-parametric 2.5th-97.5th percentiles reference intervals were calculated for each parameter. RESULTS: The reference intervals for FIB, PT, APTT, TT, FDP, DD, and ATIII at first trimester were 2.11-4.32 g/L, 10.90-13.85 s, 24.60-39.28 s, 12.95-15.88 s, 0.04-2.55 µg/mL, 0.03-1.15 µg/mL, and 75.57%-125.31%, respectively. The reference intervals at second trimester were 2.31-4.77 g/L, 9.70-12.64 s, 24.16-35.43 s, 12.95-15.88 s, 0.15-7.40 µg/mL, 0.08-2.13 µg/mL, and 74.35%-119.28%, respectively. For the third-trimester, the intervals were 2.39-4.96 g/L, 9.20-11.95 s, 23.90-35.51 s, 13.41-18.00 s, 0.55-13.43 µg/mL, 0.15-3.60 µg/mL, and 71.61%-118.29%, respectively. The third-trimester group showed decreased PT, APTT, and ATIII and increased FIB, TT, DD and FDP as compared with the other groups. CONCLUSION: In this study, level changes of coagulation parameters in different trimesters were observed. And the ranges for coagulation parameters were presented, which may provide some reference for clinicians to more accurately monitor the coagulation and fibrinolytic system in pregnant women.


Asunto(s)
Pueblo Asiatico , Coagulación Sanguínea , Trimestres del Embarazo/sangre , Mujeres Embarazadas , Adulto , Femenino , Humanos , Embarazo , Valores de Referencia
9.
Int J Mol Sci ; 21(11)2020 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-32527012

RESUMEN

Serine-threonine kinase receptor-associated protein (STRAP) functions as a regulator of both TGF-ß and p53 signaling that participates in the regulation of cell proliferation and cell death in response to various stresses. Here, we demonstrate that STRAP acetylation plays an important role in p53-mediated cell cycle arrest and apoptosis. STRAP is acetylated at lysines 147, 148, and 156 by the acetyltransferases CREB-binding protein (CBP) and that the acetylation is reversed by the deacetylase sirtuin7 (SIRT7). Hypo- or hyperacetylation mutations of STRAP at lysines 147, 148, and 156 (3KR or 3KQ) influence its activation and stabilization of p53. Moreover, following 5-fluorouracil (5-FU) treatment, STRAP is mobilized from the cytoplasm to the nucleus and promotes STRAP acetylation. Our finding on the regulation of STRAP links p53 with SIRT7 influencing p53 activity and stability.


Asunto(s)
Proteínas de Unión al ARN/metabolismo , Sirtuinas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Acetilación/efectos de los fármacos , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Fluorouracilo/farmacología , Células HCT116 , Humanos , Lisina/metabolismo , Estabilidad Proteica , Proteínas de Unión al ARN/genética , Sirtuinas/genética , Proteína p53 Supresora de Tumor/genética
10.
Am J Physiol Cell Physiol ; 317(4): C762-C775, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31365297

RESUMEN

Compelling evidence indicates that epigenetic regulations orchestrate dynamic macrophage polarization. N6-methyladenosine (m6A) methylation is the most abundant epigenetic modification of mammalian mRNA, but its role in macrophage polarization is still completely unknown. Here, we show that the m6A-catalytic enzyme methyltransferase like 3 (METTL3) is specifically upregulated following the M1 polarization of mouse macrophages. Furthermore, METTL3 knockdown through siRNA transfection markedly inhibited M1, but enhanced M2, macrophage polarization. Conversely, its overexpression via plasmid transfection greatly facilitated M1, but attenuated M2, macrophage polarization. Further methylated RNA immunoprecipitation and in vitro m6A methylation assays suggested that METTL3 directly methylates mRNA encoding signal transducer and activator of transcription 1 (STAT1), a master transcription factor controlling M1 macrophage polarization, at its coding sequence and 3'-untranslated regions. In addition, METTL3-mediated STAT1 mRNA methylation significantly increased mRNA stability and subsequently upregulated STAT1 expression. In conclusion, METTL3 drives M1 macrophage polarization by directly methylating STAT1 mRNA, potentially serving as an anti-inflammatory target.


Asunto(s)
Adenosina/análogos & derivados , Antiinflamatorios/farmacología , Macrófagos/efectos de los fármacos , Metiltransferasas/efectos de los fármacos , Adenosina/farmacología , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Activación de Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Metilación/efectos de los fármacos , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Factor de Transcripción STAT1/efectos de los fármacos
11.
Haematologica ; 104(10): 1984-1994, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30819915

RESUMEN

Transmembrane protein 30A (Tmem30a) is the ß-subunit of P4-ATPases which function as flippase that transports aminophospholipids such as phosphatidylserine from the outer to the inner leaflets of the plasma membrane to maintain asymmetric distribution of phospholipids. It has been documented that deficiency of Tmem30a led to exposure of phosphatidylserine. However, the role of Tmem30a in vivo remains largely unknown. Here we found that Vav-Cre-driven conditional deletion of Tmem30a in hematopoietic cells led to embryonic lethality due to severe anemia by embryonic day 16.5. The numbers of erythroid colonies and erythroid cells were decreased in the Tmem30a deficient fetal liver. This was accompanied by increased apoptosis of erythroid cells. Confocal microscopy analysis revealed an increase of localization of erythropoietin receptor to areas of membrane raft microdomains in response to erythropoietin stimulation in Ter119-erythroid progenitors, which was impaired in Tmem30a deficient cells. Moreover, erythropoietin receptor (EPOR)-mediated activation of the STAT5 pathway was significantly reduced in Tmem30a deficient fetal liver cells. Consistently, knockdown of TMEM30A in human CD34+ cells also impaired erythropoiesis. Our findings demonstrate that Tmem30a plays a critical role in erythropoiesis by regulating the EPOR signaling pathway through the formation of membrane rafts in erythroid cells.


Asunto(s)
Eritropoyesis , Feto/embriología , Hematopoyesis Extramedular , Células Madre Hematopoyéticas/enzimología , Hígado/embriología , Proteínas de la Membrana/deficiencia , Animales , Feto/citología , Células Madre Hematopoyéticas/citología , Hígado/citología , Microdominios de Membrana/enzimología , Microdominios de Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Transducción de Señal
12.
Circ Res ; 118(6): 944-56, 2016 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-26838785

RESUMEN

RATIONALE: Vascular endothelial inflammation, including the expression of intercellular adhesion molecule 1 (ICAM-1), is a key event in vascular diseases. However, the mechanisms underlying the regulation of ICAM-1 are largely unknown. OBJECTIVE: To investigate the mechanisms on the regulation of ICAM-1 by NOP2/Sun domain family, member 2 (NSun2)-mediated mRNA methylation and the impact of NSun2-ICAM-1 regulatory process in vascular inflammation and allograft arteriosclerosis. METHODS AND RESULTS: By using in vitro, in cells, and in vivo methylation assays, we showed that the tRNA methyltransferase NSun2 methylated the ICAM-1 mRNA. Methylation by NSun2 promoted the translation of ICAM-1, thereby increasing the adhesion of leukocytes to endothelial cells. Tumor necrosis factor-α or homocysteine activated the methyltransferase activity of NSun2 by repressing the phosphorylation of NSun2 by Aurora-B. The levels of ICAM-1 induction and of leukocyte adhesion to vascular endothelium observed with homocysteine treatment in wild-type rats were markedly decreased in NSun2(-/-) rats. In a rat model of aortic allograft, the lack of donor NSun2 impaired the formation of allograft arteriosclerosis. CONCLUSIONS: NSun2 upregulates the expression of ICAM-1 by methylating ICAM-1 mRNA. This regulatory process impacts on vascular inflammation and allograft arteriosclerosis.


Asunto(s)
Endotelio Vascular/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Metiltransferasas/deficiencia , ARN Mensajero/metabolismo , Animales , Aorta Torácica/metabolismo , Aorta Torácica/patología , Adhesión Celular/fisiología , Endotelio Vascular/patología , Técnicas de Inactivación de Genes , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación/metabolismo , Inflamación/patología , Masculino , Metilación , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas
13.
J Cell Biochem ; 118(9): 2587-2598, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28247949

RESUMEN

N6-methyladenosine (m6A) and m5C methylation are two major types of RNA methylation, but the impact of joint modifications on the same mRNA is unknown. Here, we show that in p21 3'UTR, NSUN2 catalyzes m5C modification and METTL3/METTL14 catalyzes m6A modification. Interestingly, methylation at m6A by METTL3/METTL14 facilitates the methylation of m5C by NSUN2, and vice versa. NSUN2-mediated m5C and METTL3/METTL14-mediated m6A methylation synergistically enhance p21 expression at the translational level, leading to elevated expression of p21 in oxidative stress-induced cellular senescence. Our findings on p21 mRNA methylation and expression reveal that joint m6A and m5C modification of the same RNA may influence each other, coordinately affecting protein expression patterns. J. Cell. Biochem. 118: 2587-2598, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Adenosina/análogos & derivados , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Metiltransferasas/metabolismo , Biosíntesis de Proteínas , Regiones no Traducidas 3' , Adenosina/genética , Adenosina/metabolismo , Senescencia Celular/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Regulación de la Expresión Génica , Células HeLa , Humanos , Metilación , Metiltransferasas/genética , Estrés Oxidativo/genética
14.
Biochem Biophys Res Commun ; 493(1): 94-99, 2017 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-28919411

RESUMEN

Interleukin-17A (IL-17A) has been proven to participate in the process of various autoimmune diseases. The elevation of plasma homocysteine (Hcy), known as hyperhomocysteinemia (HHcy), is related to various chronic inflammatory diseases. Though HHcy-induced upregulation of IL-17A expression in T lymphocytes has been examined, the way in which IL-17A is regulated remains unclear. In this study, western blotting assays showed that Hcy (100 µM) upregulated NOP2/Sun domain family, member 2 (NSun2) expression in rat T lymphocytes. HHcy-induced upregulation of IL-17A observed in plasma of wild-type rats was markedly decreased in NSun2-/- rats in vivo. Mechanistically, by using in vitro methylation assays and high-performance liquid chromatography-mass spectrum (HPLC-MS) analysis, we showed that the tRNA methyltransferase NSun2 methylated the IL-17A mRNA in an m5C pattern. The results from bisulfite sequencing indicated that NSun2 methylated IL-17A mRNA at cytosine C466 in vitro and in vivo. Furthermore, we analyzed the activity of pGL3-derived reporters bearing IL-17A mRNA fragments and found that methylation by NSun2 promoted the translation of IL-17A. In conclusion, NSun2 mediates HHcy-induced upregulation of IL-17A expression by methylating IL-17A mRNA and promoting its translation in T lymphocytes.


Asunto(s)
Homocisteína/metabolismo , Interleucina-17/metabolismo , Metiltransferasas/genética , Metiltransferasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Linfocitos T/metabolismo , Animales , Células Cultivadas , Regulación de la Expresión Génica/fisiología , Metilación , Ratas , Regulación hacia Arriba/fisiología
15.
J Biol Chem ; 290(44): 26627-37, 2015 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-26354435

RESUMEN

Proteinase activated-receptor 2 (PAR2) participates in cancer metastasis promoted by serine proteinases. The current study aimed to test the molecular mechanism by which PAR2 promotes cancer cell migration. In different cancer cells, activation of PAR2 by activating peptide (PAR2-AP) dramatically increased cell migration, whereas knock down of PAR2 inhibited cellular motility. The PAR2 activation also repressed miR-125b expression while miR-125b mimic successfully blocked PAR2-induced cell migration. Moreover, Grb associated-binding protein 2 (Gab2) was identified as a novel target gene of miR-125b and it mediated PAR2-induced cell migration. The correlation of PAR2 with miR-125b and Gab2 was further supported by the findings obtained from human colorectal carcinoma specimens. Remarkably, knock down of NOP2/Sun domain family, member 2 (NSun2), a RNA methyltransferase, blocked the reduction in miR-125b induced by PAR2. Furthermore, PAR2 activation increased the level of N(6)-methyladenosine (m(6)A)-containing pre-miR-125b in NSun2-dependent manner. Taken together, our results demonstrated that miR-125b mediates PAR2-induced cancer cell migration by targeting Gab2 and that NSun2-dependent RNA methylation contributes to the down-regulation of miR-125b by PAR2 signaling. These findings suggest a novel epigenetic mechanism by which microenvironment regulates cancer cell migration by altering miRNA expression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , ARN Neoplásico/metabolismo , Receptor PAR-2/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenosina/análogos & derivados , Adenosina/metabolismo , Sitios de Unión , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Epigénesis Genética , Células HCT116 , Células HT29 , Humanos , Metilación/efectos de los fármacos , Metiltransferasas/antagonistas & inhibidores , Metiltransferasas/genética , Metiltransferasas/metabolismo , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Oligopéptidos/farmacología , Oligorribonucleótidos/genética , Oligorribonucleótidos/metabolismo , Unión Proteica , ARN Neoplásico/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptor PAR-2/metabolismo , Transducción de Señal , Microambiente Tumoral/genética
16.
J Biol Chem ; 290(2): 926-40, 2015 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-25391651

RESUMEN

ERBB4, one of four ErbB receptor tyrosine kinase family members, plays an important role in the etiology and progression of lung cancer. In this study, we found that the ERBB4 protein levels were consistently up-regulated in lung cancer tissues, whereas the mRNA levels varied randomly, suggesting that a post-transcriptional mechanism was involved in regulating ERBB4 expression. Because microRNAs are powerful post-transcriptional regulators of gene expression, we used bioinformatic analyses to search for microRNAs that can potentially target ERBB4. We identified specific targeting sites for miR-193a-3p in the 3'-UTR of ERBB4. We further identified an inverse correlation between miR-193a-3p levels and ERBB4 protein levels, but not mRNA levels, in lung cancer tissue samples. By overexpressing or knocking down miR-193a-3p in lung cancer cells, we experimentally confirmed that miR-193a-3p directly recognizes the 3'-UTR of the ERBB4 transcript and regulates ERBB4 expression. Furthermore, the biological consequences of the targeting of ERBB4 by miR-193a-3p were examined in vitro via cell proliferation, invasion, and apoptosis assays and in vivo using a mouse xenograft tumor model. We demonstrated that the repression of ERBB4 by miR-193a-3p suppressed proliferation and invasion and promoted apoptosis in lung cancer cells and that miR-193a-3p exerted an anti-tumor effect by negatively regulating ERBB4 in xenograft mice. Taken together, our findings provide the first clues regarding the role of miR-193a-3p as a tumor suppressor in lung cancer through the inhibition of ERBB4 translation.


Asunto(s)
Apoptosis/genética , Neoplasias Pulmonares/genética , MicroARNs/genética , Receptor ErbB-4/biosíntesis , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/patología , Ratones , Biosíntesis de Proteínas , Receptor ErbB-4/genética , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Biochem Biophys Res Commun ; 478(2): 1006-13, 2016 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-27498005

RESUMEN

Aging-related vascular dysfunction contributes to cardiovascular morbidity and mortality. Cartilage oligomeric matrix protein (COMP), a vascular extracellular matrix protein, has been described as a negative regulatory factor for the vascular aging-related processes including atherosclerosis and vascular calcification. However, whether COMP is implicated in the process of vascular aging remains unclear. Here, we identified a novel function of COMP in preventing vascular aging and vascular smooth muscle cells (VSMCs) senescence. Firstly, vascular COMP expression was decreased in three different senescence-accelerated mouse models and was also declining with age. COMP(-/-) mice displayed elevated senescence-associated markers expression, including p53, p21 and p16, in the aortas compared with their wild type (WT) littermates. In accordance, COMP deficiency induced aging-related vascular dysfunction as evidenced by the significantly reduced phenylephrine-induced contraction and increased vascular stiffness as evaluated by pulse wave velocity. The aortic wall of COMP(-/-) mice was susceptible to senescence by displaying senescence-associated ß-galactosidase (SA ß-gal) activity induced by periadventitial application of CaCl2 to the abdominal aorta. In vitro, COMP knockdown by small interfering (si) RNA led to the elevation of p53, p21 and p16 as well as SA ß-gal activity in VSMCs after H2O2 stimulation. VSMCs isolated from COMP(-/-) mice showed elevated senescence-associated markers expression and supplement of COMP adenovirus to COMP-deficient VSMCs greatly rescued cellular senescence. Taken together, these findings revealed the essential role of COMP in retarding the development of vascular aging and VSMC senescence.


Asunto(s)
Envejecimiento/metabolismo , Vasos Sanguíneos/metabolismo , Proteína de la Matriz Oligomérica del Cartílago/metabolismo , Senescencia Celular , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/metabolismo , Animales , Aorta/metabolismo , Vasos Sanguíneos/fisiopatología , Proteína de la Matriz Oligomérica del Cartílago/deficiencia , Ratones Endogámicos C57BL , Modelos Animales , Ratas
18.
J Biol Chem ; 289(6): 3217-30, 2014 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-24344134

RESUMEN

The tumor suppressor PTEN plays a critical role in the regulation of multiple cellular processes that include survival, cell cycle, proliferation, and apoptosis. PTEN is frequently mutated or deleted in various human cancer cells to promote tumorigenesis. PTEN is regulated by SUMOylation, but the SUMO E3 ligase involved in the SUMOylation of PTEN remains unclear. Here, we demonstrated that PIASxα is a SUMO E3 ligase for PTEN. PIASxα physically interacted with PTEN both in vitro and in vivo. Their interaction depended on the integrity of phosphatase and C2 domains of PTEN and the region of PIASxα comprising residues 134-347. PIASxα enhanced PTEN protein stability by reducing PTEN ubiquitination, whereas the mutation of PTEN SUMO1 conjugation sites neutralized the effect of PIASxα on PTEN protein half-life. Functionally, PIASxα, as a potential tumor suppressor, negatively regulated the PI3K-Akt pathway through stabilizing PTEN protein. Overexpression of PIASxα led to G0/G1 cell cycle arrest, thus triggering cell proliferation inhibition and tumor suppression, whereas PIASxα knockdown or deficiency in catalytic activity abolished the inhibition. Together our studies suggest that PIASxα is a novel SUMO E3 ligase for PTEN, and it positively regulates PTEN protein level in tumor suppression.


Asunto(s)
Fosfohidrolasa PTEN/metabolismo , Proteínas Inhibidoras de STAT Activados/metabolismo , Proteína SUMO-1/metabolismo , Sumoilación/fisiología , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/fisiología , Células HeLa , Humanos , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Inhibidoras de STAT Activados/genética , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fase de Descanso del Ciclo Celular/fisiología , Proteína SUMO-1/genética , Proteínas Supresoras de Tumor/genética , Ubiquitina-Proteína Ligasas/genética
19.
Nat Metab ; 6(6): 1161-1177, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38698281

RESUMEN

Diabetic cardiomyopathy is characterized by myocardial lipid accumulation and cardiac dysfunction. Bile acid metabolism is known to play a crucial role in cardiovascular and metabolic diseases. Takeda G-protein-coupled receptor 5 (TGR5), a major bile acid receptor, has been implicated in metabolic regulation and myocardial protection. However, the precise involvement of the bile acid-TGR5 pathway in maintaining cardiometabolic homeostasis remains unclear. Here we show decreased plasma bile acid levels in both male and female participants with diabetic myocardial injury. Additionally, we observe increased myocardial lipid accumulation and cardiac dysfunction in cardiomyocyte-specific TGR5-deleted mice (both male and female) subjected to a high-fat diet and streptozotocin treatment or bred on the diabetic db/db genetic background. Further investigation reveals that TGR5 deletion enhances cardiac fatty acid uptake, resulting in lipid accumulation. Mechanistically, TGR5 deletion promotes localization of CD36 on the plasma membrane through the upregulation of CD36 palmitoylation mediated by the palmitoyl acyltransferase DHHC4. Our findings indicate that the TGR5-DHHC4 pathway regulates cardiac fatty acid uptake, which highlights the therapeutic potential of targeting TGR5 in the management of diabetic cardiomyopathy.


Asunto(s)
Cardiomiopatías Diabéticas , Ácidos Grasos , Receptores Acoplados a Proteínas G , Animales , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Cardiomiopatías Diabéticas/metabolismo , Ratones , Masculino , Femenino , Ácidos Grasos/metabolismo , Humanos , Ratones Noqueados , Ácidos y Sales Biliares/metabolismo , Dieta Alta en Grasa , Antígenos CD36/metabolismo , Antígenos CD36/genética , Miocardio/metabolismo , Metabolismo de los Lípidos , Miocitos Cardíacos/metabolismo , Diabetes Mellitus Experimental/metabolismo
20.
Cell Rep ; 43(5): 114238, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38748875

RESUMEN

Triacylglyceride (TAG) synthesis in the small intestine determines the absorption of dietary fat, but the underlying mechanisms remain to be further studied. Here, we report that the RNA-binding protein HuR (ELAVL1) promotes TAG synthesis in the small intestine. HuR associates with the 3' UTR of Dgat2 mRNA and intron 1 of Mgat2 pre-mRNA. Association of HuR with Dgat2 3' UTR stabilizes Dgat2 mRNA, while association of HuR with intron 1 of Mgat2 pre-mRNA promotes the processing of Mgat2 pre-mRNA. Intestinal epithelium-specific HuR knockout reduces the expression of DGAT2 and MGAT2, thereby reducing the dietary fat absorption through TAG synthesis and mitigating high-fat-diet (HFD)-induced non-alcoholic fatty liver disease (NAFLD) and obesity. Our findings highlight a critical role of HuR in promoting dietary fat absorption.


Asunto(s)
Dieta Alta en Grasa , Proteína 1 Similar a ELAV , Absorción Intestinal , Triglicéridos , Triglicéridos/metabolismo , Triglicéridos/biosíntesis , Animales , Proteína 1 Similar a ELAV/metabolismo , Proteína 1 Similar a ELAV/genética , Ratones , Dieta Alta en Grasa/efectos adversos , Humanos , Ratones Endogámicos C57BL , Masculino , Diacilglicerol O-Acetiltransferasa/metabolismo , Diacilglicerol O-Acetiltransferasa/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Enfermedad del Hígado Graso no Alcohólico/genética , Obesidad/metabolismo , Obesidad/genética , ARN Mensajero/metabolismo , ARN Mensajero/genética , Grasas de la Dieta/metabolismo , Grasas de la Dieta/farmacología , Ratones Noqueados , Regiones no Traducidas 3'/genética , Aciltransferasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA