Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Radiat Biol ; 99(10): 1639-1647, 2023.
Article in English | MEDLINE | ID: mdl-36867417

ABSTRACT

PURPOSE: Wireless communication has become an integral part of our lives. The growing number of antennas in our environment and the expanding use of mobile phones (MPs) are increasing the population's exposure to electromagnetic fields. The present study aimed to examine the potential impact of MPs radiofrequency electromagnetic fields (RF-EMF) exposure on the brainwaves of the resting electroencephalogram (EEG) in humans. MATERIALS AND METHODS: Twenty-one healthy volunteers were exposed to Global System for Mobile communications (GSM) signal at 900 MHz MP RF-EMF. The maximum specific absorption rate (SAR) of the MP averaged on 10 g tissue and 1 g tissue were measured at 0.49 W/kg, 0.70 W/kg, respectively. RESULTS: Results showed that while delta and beta rhythms of resting EEG were not affected, theta brainwaves were significantly modulated during exposure to RF-EMF related to MPs. For the first time, it was shown that this modulation is dependent on the eye condition, i.e. closed or open. CONCLUSIONS: This study strongly suggests that acute exposure to RF-EMF alters the EEG theta rhythm at rest. Long-term exposure studies are required to explore the effect of this disruption in high-risk or sensitive populations.


Subject(s)
Brain Waves , Cell Phone , Humans , Electroencephalography , Electromagnetic Fields/adverse effects , Radio Waves/adverse effects
2.
Acta Pharmacol Sin ; 43(6): 1383-1394, 2022 Jun.
Article in English | MEDLINE | ID: mdl-34493812

ABSTRACT

Myocardial ischemia-reperfusion (I/R) injury is a pathological process characterized by cardiomyocyte apoptosis, which leads to cardiac dysfunction. Increasing evidence shows that abnormal expression of long noncoding RNAs (lncRNAs) plays a crucial role in cardiovascular diseases. In this study we investigated the role of lncRNAs in myocardial I/R injury. Myocardial I/R injury was induced in mice by ligating left anterior descending coronary artery for 45 min followed by reperfusion for 24 h. We showed that lncRNA KnowTID_00006395, termed lncRNA-6395 was significantly upregulated in the infarct area of mouse hearts following I/R injury as well as in H2O2-treated neonatal mouse ventricular cardiomyocytes (NMVCs). Overexpression of lncRNA-6395 led to cell apoptosis and the expression change of apoptosis-related proteins in NMVCs, whereas knockdown of lncRNA-6395 attenuated H2O2-induced cell apoptosis. LncRNA-6395 knockout mice (lncRNA-6395+/-) displayed improved cardiac function, decreased plasma LDH activity and infarct size following I/R injury. We demonstrated that lncRNA-6395 directly bound to p53, and increased the abundance of p53 protein through inhibiting ubiquitination-mediated p53 degradation and thereby facilitated p53 translocation to the nucleus. More importantly, overexpression of p53 canceled the inhibitory effects of lncRNA-6395 knockdown on cardiomyocyte apoptosis, whereas knockdown of p53 counteracted the apoptotic effects of lncRNA-6395 in cardiomyocytes. Taken together, lncRNA-6395 as an endogenous pro-apoptotic factor, regulates cardiomyocyte apoptosis and myocardial I/R injury by inhibiting degradation and promoting sub-cellular translocation of p53.


Subject(s)
Myocardial Reperfusion Injury , RNA, Long Noncoding , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/pharmacology , Hydrogen Peroxide/pharmacology , Infarction/pathology , Mice , Mice, Knockout , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
3.
Basic Res Cardiol ; 116(1): 15, 2021 03 06.
Article in English | MEDLINE | ID: mdl-33675440

ABSTRACT

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.


Subject(s)
Cell Proliferation , Collagen/metabolism , Fibroblasts/metabolism , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , RNA, Long Noncoding/metabolism , Animals , Cells, Cultured , DNA-Binding Proteins/metabolism , Disease Models, Animal , Fibroblasts/pathology , Fibrosis , Male , Mice, Inbred C57BL , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , RNA Polymerase II/metabolism , RNA, Long Noncoding/genetics , RNA-Binding Proteins/metabolism , Transcription, Genetic
4.
Front Pharmacol ; 12: 631835, 2021.
Article in English | MEDLINE | ID: mdl-33664669

ABSTRACT

Purpose: Cardiomyocyte senescence is associated with a progressive decline in cardiac physiological function and the risk of cardiovascular events. lncRNA H19 (H19), a well-known long noncoding RNA (lncRNA), is involved in the pathophysiological process of multiple cardiovascular disease such as heart failure, cardiac ischemia and fibrosis. However, the role of H19 in cardiomyocyte senescence remains to be further explored. Methods: Senescence-associated ß-galactosidases (SA-ß-gal) staining was used to detect cardiomyocyte senescence. Western blot, qRT-PCR and luciferase reporter assay were employed to evaluate the role of H19 in cardiomyocyte senescence and its underling molecular mechanism. Results: H19 level was significantly increased in high glucose-induced senescence cardiomyocytes and aged mouse hearts. Overexpression of H19 enhanced the number of SA-ß-gal-positive cells, and the expression of senescence-related proteins p53 and p21, whereas H19 knockdown exerted the opposite effects. Mechanistically, H19 was demonstrated as a competing endogenous RNA (ceRNA) for microRNA-19a (miR-19a): H19 overexpression downregulated miR-19a level, while H19 knockdown upregulated miR-19a. The expression of SOSC1 was dramatically increased in senescence cardiomyocytes and aged mouse hearts. Further experiments identified SOCS1 as a downstream target of miR-19a. H19 upregulated SOCS1 expression and activated the p53/p21 pathway by targeting miR-19a, thus promoting the cardiomyocytes senescence. Conclusion: Our results show that H19 is a pro-senescence lncRNA in cardiomyocytes acting as a ceRNA to target the miR-19a/SOCS1/p53/p21 pathway. Our research reveals a molecular mechanism of cardiomyocyte senescence regulation and provides a novel target of the therapy for senescence-associated cardiac diseases.

5.
FASEB J ; 35(2): e21162, 2021 02.
Article in English | MEDLINE | ID: mdl-33150686

ABSTRACT

Cardiac fibrosis is characterized by the activation of cardiac fibroblasts and accumulation of extracellular matrix. METTL3, a component of methyltransferase complex, participates in multiple biological processes associated with mammalian development and disease progression. However, the role of METTL3 in cardiac fibrosis is still unknown. We performed fibroblasts activation with TGF-ß1 (20 ng/mL) in vitro and established in vivo mouse models with lentivirus to assess the effects of METTL3 on cardiac fibroblasts proliferation and collagen formation. Methylated RNA immunoprecipitation (MeRIP) was used to define the potential fibrosis-regulated gene. The expression level of METTL3 was increased in cardiac fibrotic tissue of mice with chronic myocardial infarction and cultured cardiac fibroblats (CFs) treated with TGF-ß1. Enforced expression of METTL3 promoted proliferation and fibroblast-to-myofibroblast transition and collagens accumulation, while silence of METTL3 did the opposite. Silence of METTL3 by lentivirus carrying METTL3 siRNA markedly alleviated cardiac fibrosis in MI mice. Transcriptome and N6-methyladenosine (m6 A) profiling analyses revealed that the expression and m6 A level of collagen-related genes were altered after silence of METTL3. METTL3-mediated m6 A modification is critical for the development of cardiac fibrosis, providing a molecular target for manipulating fibrosis and the associated cardiac diseases.


Subject(s)
Gene Silencing , Methyltransferases/genetics , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardium/pathology , Myofibroblasts/metabolism , Animals , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Collagen/genetics , Collagen/metabolism , Disease Models, Animal , Fibrosis , Male , Mice , Mice, Inbred C57BL , Myofibroblasts/drug effects , Signal Transduction/drug effects , Signal Transduction/genetics , Transcriptome , Transfection , Transforming Growth Factor beta1/pharmacology , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...