Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Toxicology ; 494: 153587, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37406984

ABSTRACT

In recent years, the role and mechanism of long non-coding RNA (LncRNA) in cardiovascular diseases have received increasing attention. The chemotherapy agent, doxorubicin (DOX), is one of the most effective drugs for various cancers, but its efficacy is limited by its cardiotoxicity. Therefore, further exploration is required for the molecular mechanism of DOX-induced cardiotoxicity. This study intended to investigate the role of LncRNA Non-coding RNA activated by DNA damage (NORAD) in DOX-induced cardiotoxicity, for which we adopted the AC16 human cardiomyocyte cell line for the exploration. The results showed that LncRNA NORAD knockdown could increase DOX-induced cardiomyocyte apoptosis and mitochondrial ROS level. LncRNA NORAD overexpression obtained reverse results, which further validated its role in DOX-induced cardiomyocyte apoptosis and mitochondrial ROS level. Moreover, cardiotoxicity was induced in both LncRNA NORAD-knockout and wild-type mice with DOX, showing that gene knockout aggravated pathologic lesions in the myocardial tissues of mice. Taken together, LncRNA NORAD affected DOX-induced cardiotoxicity via mitochondrial apoptosis, fission (PUM-MFF), and autophagy (p53-Parkin) pathways both in vivo and in vitro.


Subject(s)
Cardiotoxicity , RNA, Long Noncoding , Mice , Humans , Animals , Cardiotoxicity/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Reactive Oxygen Species/metabolism , Doxorubicin/toxicity , Myocytes, Cardiac , Apoptosis
2.
Aging (Albany NY) ; 15(15): 7513-7532, 2023 07 29.
Article in English | MEDLINE | ID: mdl-37517088

ABSTRACT

Long noncoding RNAs (lncRNAs) play important roles in the development of age-related macular degeneration (AMD). However, the effect of long non-coding RNA activated by DNA damage (NORAD) on AMD remains unknown. This study aimed to investigate the effect of NORAD on RPE cell senescence and degeneration. Irradiated adult retinal pigment epithelial cell line-19 (ARPE-19) and sodium iodate-treated mice were used as in vitro and in vivo AMD models. Results showed that irradiation-induced AMD characteristics of ARPE-19 and NORAD-knockdown aggravated cell cycle arrest in the G2/M phase, cell apoptosis and cell senescence along with the increased expression of phosphorylated P53 (p-P53) and P21. AMD factors C3, ICAM-1, APP, APOE, and VEGF-A were also increased by NORAD-knockdown. Moreover, NORAD-knockdown increased irradiation-induced reduction of mitochondrial homeostasis factors, (i.e., TFAM and POLG) and mitochondrial respiratory chain complex genes (i.e., ND1 and ND5) along with mitochondrial reactive oxygen species (ROS). We also identified a strong interaction of NORAD and PGC-1α and sirtuin 1 (SIRT1) in ARPE-19; that is, NORAD knockdown increases the acetylation of PGC-1α. In NORAD knockout mice, NORAD-knockout accelerated the sodium iodate-reduced retinal thickness reduction, function impairment and loss of retinal pigment in the fundus. Therefore, NORAD-knockdown accelerates retinal cell senescence, apoptosis, and AMD markers via PGC-1α acetylation, mitochondrial ROS, and the p-P53-P21signaling pathway, in which NORAD-mediated effect on PGC-1α acetylation might occur through the direct interaction with PGC-1α and SIRT1.


Subject(s)
Macular Degeneration , RNA, Long Noncoding , Mice , Animals , Reactive Oxygen Species/metabolism , Oxidative Stress , RNA, Long Noncoding/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Macular Degeneration/genetics , Macular Degeneration/metabolism , Retinal Pigments/metabolism , Retinal Pigments/pharmacology , Retinal Pigment Epithelium
3.
Toxicol Lett ; 377: 29-37, 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36739041

ABSTRACT

Recombinant human endostatin (rh-endostatin) is an anti-angiogenic drug, which is used for the treatment of advanced non-small-cell lung cancer (NSCLC) and other cancers. However, its side effects, especially the cardiotoxicity with unclear mechanisms limit its wide application in clinical practice. In this study, human cardiomyocyte cell line AC16 and human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) treated with different doses of rh-endostatin were used to analyze its effect on cardiac cell toxicity. The results revealed that rh-endostatin dose-dependently enhanced cardiomyocyte apoptosis through Apaf-1 apoptotic factor and apoptosis-related proteins such as p53. rh-endostatin-induced changes of mitochondrial function and mitophagy were involved in rh-endostatin-mediated cardiac cell toxicity. Rh-endostatin-induced cardiotoxicity was further verified in vivo in mice. Interestingly, Rh-endostatin-induced cardiotoxicity was inhibited by dihydromyricetin (DHM) both in cultured cells in vitro and in mouse hearts in vivo. The study provides new inside into rh-endostatin-induced cardiotoxicity and identified a novel potential medication DHM to overcome the serious adverse effect.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Drug-Related Side Effects and Adverse Reactions , Induced Pluripotent Stem Cells , Lung Neoplasms , Humans , Mice , Animals , Myocytes, Cardiac , Endostatins/toxicity , Endostatins/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cardiotoxicity , Lung Neoplasms/metabolism , Mice, Inbred C57BL , Drug-Related Side Effects and Adverse Reactions/metabolism
4.
Stroke Vasc Neurol ; 8(3): 181-192, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36241224

ABSTRACT

BACKGROUND: Ischaemic stroke and other cardiovascular illnesses are characterised by abnormalities in the processes of thrombosis and haemostasis, which rely on platelet activity. In platelets, a wide variety of microRNAs (long non-coding RNA, lncRNAs) is found. Due to the absence of nuclear DNA in platelets, lncRNAs may serve as critical post-transcriptional regulators of platelet activities. However, research into the roles of lncRNAs in platelets is limited. OBJECTIVE: The purpose of this study is to learn more about the molecular mechanism by which MALAT1 affects platelet activity and thrombus formation. METHODS/RESULTS: The CD34+ megakaryocytes used in this research as an in vitro model for human megakaryocytes and platelets. Cell adhesion and spreading are enhanced in the absence and presence of agonists in CD34+ megakaryocytes subjected to MALAT1 knockdown (KD). The adhesion and activity of platelet-like particles produced by MALAT1 KD cells are significantly enhanced at rest and after thrombin activation. Thrombus development on a collagen matrix is also greatly enhanced in the microfluidic whole-blood perfusion model: platelets lacking MALAT1 exhibit elevated accumulation, distributing area and activity. In addition, MALAT1-deficient mice bleed less and form a stable occlusive thrombus more quickly than wild-type mice. PTEN and PDK1 regulated the activity of MALAT1 in platelets to carry out its PI3k/Akt/GSK-3ß signalling pathway-related function. CONCLUSION: The suppression of MALAT1 expression significantly increases platelet adhesion, spreading, platelet activity, and thrombus formation. lncRNAs may constitute a unique class of platelet function modulators.


Subject(s)
Brain Ischemia , RNA, Long Noncoding , Stroke , Thrombosis , Animals , Humans , Mice , Blood Platelets/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Platelet Aggregation , Brain Ischemia/metabolism , Disease Models, Animal , Stroke/metabolism , Thrombosis/genetics
5.
Arch Physiol Biochem ; 129(1): 198-203, 2023 Feb.
Article in English | MEDLINE | ID: mdl-32907416

ABSTRACT

BACKGROUND: We investigated the effects and mechanism of swimming on platelet function in mice fed with a high-fat diet. MATERIAL AND METHODS: Mice were randomly divided into the control group (NC), high-fat group (HF), and high-fat diet combined with swimming group (FE). The FE group swam for 60 min a day, 5 days a week, for 8 weeks. RESULTS: Compared with the NC group, the HF group had significant weight gain, dyslipidemia, abbreviated bleeding time after tail breakage, increased clot retraction, increased platelet aggregation rate, increased spread of platelets on fibrinogen, and increased pAKT level in platelets. Compared with the HF group, the FE group had lower body weight, improved dyslipidemia, prolonged bleeding time, reduced clot retraction, reduced platelet aggregation rate, decreased spread of platelets on fibrinogen, and decreased pAKT level in platelets. CONCLUSIONS: By inhibiting the level of pAKT in platelets, swimming improves platelet dysfunction in mice fed with a high-fat diet.


Subject(s)
Diet, High-Fat , Dyslipidemias , Mice , Animals , Diet, High-Fat/adverse effects , Swimming , Blood Platelets , Fibrinogen , Platelet Aggregation
6.
Cells ; 11(14)2022 07 21.
Article in English | MEDLINE | ID: mdl-35883699

ABSTRACT

Recently, long noncoding RNAs (lncRNAs) have been key regulators for both mRNAs and proteins in nucleated cells. However, the expression profiles of lncRNAs in non-nucleated cells such as platelets are currently unclear. In this study, we determined the expression profiles of lncRNAs in human platelets. We found that 6109 lncRNAs were expressed in human platelets. Interestingly, 338 lncRNAs were differentially expressed in hyperreactive and hyporeactive platelets. Bioinformatics' analysis revealed that these aberrantly expressed lncRNAs might be related to platelet activity and other platelet functions. To provide a proof of concept, we measured the expression levels of PARLncRNA-1, a down-regulated lncRNA of hyperreactive platelets, in platelets from 12 patients with acute myocardial infarction and their controls. We found that the lncRNA was also significantly down-regulated in platelets from patients, which was partially reversed by treatment with aspirin a known antiplatelet drug. LncRNAs may represent a novel class of modulators for platelet functions.


Subject(s)
RNA, Long Noncoding , Gene Expression Profiling , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics
7.
Stroke Vasc Neurol ; 7(6): 500-509, 2022 12.
Article in English | MEDLINE | ID: mdl-35725243

ABSTRACT

BACKGROUND: Postsynaptic density protein-95 (PSD95) plays an important role in cerebral ischaemia injury, but its mechanism needs further research. This study aimed to explore the role of PSD95 in (Ang-(1-7))-Mas-mediated cerebral ischaemia protection and its regulatory mechanism. METHODS: Oxygen-glucose deprivation (OGD) neuron and rat middle cerebral artery occlusion (MCAO) models were used as in vitro and in vivo models, respectively. TAT-MAS9C was used to disrupt the interaction between PSD95 and Mas. The recombinant PSD95 adenovirus (Ad-PSD95) was used to overexpress PSD95 in neurons. RESULTS: Results showed that in OGD neurons, Ang-(1-7) could promote cell viability; reduce cell apoptosis; reduce the cell membrane localisation of Mas; upregulate the expression levels of pAKT, bcl-2 and I-κB; and downregulate the expression levels of Bax, pI-κB, tumour necrosis factor alpha and interleukin-1ß. TAT-MAS9C could enhance the aforementioned effects of Ang-(1-7). However, the PSD95 overexpression inhibited the aforementioned effects of Ang-(1-7). In the MCAO rat model, the 2,3,5-triphenyltetrazolium chloride (TTC) staining showed that Ang-(1-7) reduced the infarct volume. The Morris water maze test showed that the number of crossings over the platform area in the Ang-(1-7) group was significantly increased. TAT-MAS9C could promote the protective effect of Ang-(1-7). CONCLUSIONS: Results suggested that PSD95 alleviated the activation of AKT and the inhibition of nuclear factor kappa B signalling pathway mediated by the Ang-(1-7)-Mas complex, thereby reducing neuronal activity, increasing apoptosis and inhibiting the Ang-(1-7)-Mas-mediated cerebral ischaemia protection.


Subject(s)
Brain Injuries , Brain Ischemia , Rats , Animals , Disks Large Homolog 4 Protein/metabolism , Brain Ischemia/metabolism , Signal Transduction , Apoptosis , Infarction, Middle Cerebral Artery/pathology
8.
Diabetes Metab Syndr Obes ; 14: 4351-4360, 2021.
Article in English | MEDLINE | ID: mdl-34737591

ABSTRACT

PURPOSE: To investigate resistance to diet-induced obesity (DIO) and monosodium glutamate (MSG)-induced obesity as well as the underlying mechanisms. METHODS: Newborn mice were used to construct DIO and MSG-induced obesity models. Obesity indices, such as body weight, body length, Lee index, body temperature, food intake, fat weight, and leptin level, were examined. Mice that did not exhibit obesity were defined as the obesity-resistant group. The morphological changes of white adipose tissue were observed by hematoxylin and eosin staining, and expression levels of PR domain containing 16 (Prdm16) and uncoupling protein-1 (Ucp-1) in white adipose tissue were measured by Western blot. RESULTS: Obesity-resistant mice fed a high-fat diet showed resistance beginning at week 5 along with lower weights and lengths than those in the obesity group from weeks 5 to 12. MSG-induced obesity-resistant mice showed features consistent with resistance to obesity from week 1 along with higher body lengths relative to the obesity group; however, the weight difference was not significant until week 10, when body weights decreased significantly in obesity-resistant mice. The Lee index was lower in obesity-resistant mice than in the obesity group and the normal group, further suggesting obesity resistance. Additionally, obesity-resistant mice showed higher levels of leptin, whereas obese mice induced by a high-fat diet showed leptin resistance. Furthermore, Prdm16 and Ucp-1 levels were both downregulated in the obesity group and upregulated in obesity-resistant mice, showing that white fat browning was highest in obesity-resistant mice. CONCLUSION: The phenotypes of mice with DIO and MSG-induced obesity differed. Obesity resistance might be related to Prdm16 and Ucp-1-mediated white adipocyte browning.

9.
Int J Mol Med ; 48(4)2021 10.
Article in English | MEDLINE | ID: mdl-34368855

ABSTRACT

Heme oxygenase­1 (HO­1) has been reported to be upregulated following renal ischemia­reperfusion injury (IRI) and plays a key cytoprotective role; however, the underlying molecular mechanisms of its protective effects remain poorly understood. In the present study, in order to further elucidate the molecular mechanisms underlying the cytoprotective role of HO­1 in renal IRI, HO­1+/+ and HO­1+/­ mice were subjected to renal ischemia and subsequent reperfusion followed by the analysis of blood urea nitrogen (BUN) and serum creatinine (SCr) levels, the severity of histological changes, HO­1 and vascular cell adhesion molecule­1 (VCAM­1) protein expression, the mRNA expression of inflammatory factors and the effects of VCAM­1 blockade. The results of the present study demonstrated that the upregulated expression levels of VCAM­1 in HO­1+/­ mice during IRI increased the extent of renal tissue damage and activated the inflammatory response. These effects were subsequently reversed following infusion with an anti­VCAM­1 antibody. In addition, the upregulated expression of VCAM­1 in mouse glomerulus vascular endothelial cells isolated from HO­1+/­ mice increased the adhesion and migration of neutrophils, effects which were also reversed upon incubation with an anti­VCAM­1 antibody. These results indicated that HO­1 knockdown may upregulate the expression of VCAM­1 during renal IRI, resulting in increased neutrophil recruitment and the activation of the inflammatory response, thereby exacerbating renal IRI. The present study thus highlights the regulatory mechanisms of HO­1 in renal IRI and provides a potential target for the clinical treatment of IRI following renal transplantation.


Subject(s)
Heme Oxygenase-1/genetics , Kidney Diseases/genetics , Membrane Proteins/genetics , Neutrophils/immunology , Reperfusion Injury/genetics , Up-Regulation/genetics , Vascular Cell Adhesion Molecule-1/genetics , Animals , Blood Urea Nitrogen , Creatinine/blood , Creatinine/immunology , Heme Oxygenase-1/immunology , Inflammation/blood , Inflammation/genetics , Inflammation/immunology , Kidney/immunology , Kidney/pathology , Kidney Diseases/immunology , Male , Membrane Proteins/immunology , Mice , Reperfusion Injury/immunology , Up-Regulation/immunology , Vascular Cell Adhesion Molecule-1/immunology
10.
Front Pharmacol ; 12: 673621, 2021.
Article in English | MEDLINE | ID: mdl-34149424

ABSTRACT

Mammalian cardiomyocytes exit the cell cycle during the perinatal period, and although cardiomyocytes differentiated from human induced-pluripotent stem cells (hiPSC-CMs) are phenotypically immature, their intrinsic cell-cycle activity remains limited. Thus, neither endogenous cardiomyocytes nor the small number of transplanted hiPSC-CMs that are engrafted by infarcted hearts can remuscularize the myocardial scar. microRNAs are key regulators of cardiomyocyte proliferation, and when adeno-associated viruses coding for microRNA-199a (miR-199a) expression were injected directly into infarcted pig hearts, measures of cardiac function and fibrosis significantly improved, but the treatment was also associated with lethal arrhythmia. For the studies reported here, the same vector (AAV6-miR-199a) was transduced into hiPSC-CMs, and the cells were subsequently evaluated in a mouse model of myocardial infarction. AAV6-mediated miR-199a overexpression increased proliferation in both cultured and transplanted hiPSC-CMs, and measures of left ventricular ejection fraction, fractional shortening, and scar size were significantly better in mice treated with miR-199a-overexpressing hiPSC-CMs than with hiPSC-CMs that had been transduced with a control vector. Furthermore, although this investigation was not designed to characterize the safety of transplanted AAV6-miR-199a-transduced hiPSC-CMs, there was no evidence of sudden death. Collectively, these results support future investigations of miR-199a-overexpressing hiPSC-CMs in large animals.

11.
Circulation ; 144(3): 210-228, 2021 07 20.
Article in English | MEDLINE | ID: mdl-33951921

ABSTRACT

BACKGROUND: Human induced pluripotent stem cells with normal (wild-type) or upregulated (overexpressed) levels of CCND2 (cyclin D2) expression were differentiated into cardiomyocytes (CCND2WTCMs or CCND2OECMs, respectively) and injected into infarcted pig hearts. METHODS: Acute myocardial infarction was induced by a 60-minute occlusion of the left anterior descending coronary artery. Immediately after reperfusion, CCND2WTCMs or CCND2OECMs (3×107 cells each) or an equivalent volume of the delivery vehicle was injected around the infarct border zone area. RESULTS: The number of the engrafted CCND2OECMs exceeded that of the engrafted CCND2WTCMs from 6- to 8-fold, rising from 1 week to 4 weeks after implantation. In contrast to the treatment with the CCND2WTCMs or the delivery vehicle, the administration of CCND2OECM was associated with significantly improved left ventricular function, as revealed by magnetic resonance imaging. This correlated with reduction of infarct size, fibrosis, ventricular hypertrophy, and cardiomyocyte apoptosis, and increase of vascular density and arterial density, as per histologic analysis of the treated hearts. Expression of cell proliferation markers (eg, Ki67, phosphorylated histone 3, and Aurora B kinase) was also significantly upregulated in the recipient cardiomyocytes from the CCND2OECM-treated than from the CCND2WTCM-treated pigs. The cell proliferation rate and the hypoxia tolerance measured in cultured human induced pluripotent stem cell cardiomyocytes were significantly greater after treatment with exosomes isolated from the CCND2OECMs (CCND2OEExos) than from the CCND2WTCMs (CCND2WTExos). As demonstrated by our study, CCND2OEExos can also promote the proliferation activity of postnatal rat and adult mouse cardiomyocytes. A bulk miRNA sequencing analysis of CCND2OEExos versus CCND2WTExos identified 206 and 91 miRNAs that were significantly upregulated and downregulated, respectively. Gene ontology enrichment analysis identified significant differences in the expression profiles of miRNAs from various functional categories and pathways, including miRNAs implicated in cell-cycle checkpoints (G2/M and G1/S transitions), or the mechanism of cytokinesis. CONCLUSIONS: We demonstrated that enhanced potency of CCND2OECMs promoted myocyte proliferation in both grafts and recipient tissue in a large mammal acute myocardial infarction model. These results suggest that CCND2OECMs transplantation may be a potential therapeutic strategy for the repair of infarcted hearts.


Subject(s)
Cell Differentiation/genetics , Cyclin D2/genetics , Gene Expression , Induced Pluripotent Stem Cells/cytology , Myocardial Infarction/therapy , Myocytes, Cardiac/metabolism , Stem Cell Transplantation , Animals , Biomarkers , Cell Culture Techniques , Cell Proliferation , Cell Separation , Cells, Cultured , Disease Models, Animal , Gene Knock-In Techniques , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Myocardial Infarction/diagnosis , Myocardial Infarction/etiology , Myocytes, Cardiac/cytology , Neovascularization, Physiologic/genetics , Recovery of Function , Swine , Treatment Outcome
12.
Front Bioeng Biotechnol ; 9: 599928, 2021.
Article in English | MEDLINE | ID: mdl-33842440

ABSTRACT

The billions of cardiomyocytes lost to acute myocardial infarction (MI) cannot be replaced by the limited regenerative capacity of adult mammalian hearts, and despite decades of research, there are still no clinically effective therapies for remuscularizing and restoring damaged myocardial tissue. Although the majority of the cardiac mass is composed of cardiomyocytes, cardiac fibroblasts (CFs) are one type of most numerous cells in the heart and the primary drivers of fibrosis, which prevents ventricular rupture immediately after MI but the fibrotic scar expansion and LV dilatation can eventually lead to heart failure. However, embryonic CFs produce cytokines that can activate proliferation in cultured cardiomyocytes, and the structural proteins produced by CFs may regulate cardiomyocyte cell-cycle activity by modulating the stiffness of the extracellular matrix (ECM). CFs can also be used to generate induced-pluripotent stem cells and induced cardiac progenitor cells, both of which can differentiate into cardiomyocytes and vascular cells, but cardiomyocytes appear to be more readily differentiated from iPSCs that have been reprogrammed from CFs than from other cell types. Furthermore, the results from recent studies suggest that cultured CFs, as well as the CFs present in infarcted hearts, can be reprogrammed directly into cardiomyocytes. This finding is very exciting as should we be able to successfully increase the efficiency of this reprogramming, we could remuscularize the injured ventricle and restore the LV function without need the transplantation of cells or cell products. This review summarizes the role of CFs in the innate response to MI and how their phenotypic plasticity and involvement in ECM production might be manipulated to improve cardiac performance in injured hearts.

13.
Front Genet ; 11: 571467, 2020.
Article in English | MEDLINE | ID: mdl-33193674

ABSTRACT

Platelets are derived from megakaryocytes and play an important role in blood coagulation. By using high throughput sequencing, we have found that the long non-coding RNA (lncRNA) nuclear paraspeckle assembly transcript 1 (NEAT1) is abundant in platelets (GEO ID: 200097348). However, little is known about its role in regulating megakaryocyte differentiation and platelet activity. This study aims to clarify the effect of NEAT1 on MEG-01 differentiation and platelet-like particle (PLP) activity. NEAT1 in MEG-01 cells was knocked down by siRNA transfection. The adhesion of MEG-01 and PLP to collagen-coated coverslips was observed under a fluorescence microscope. Flow cytometry was used to investigate cell apoptosis, cell cycle, the levels of D41/CD42b on MEG-01 cells and CD62P on PLPs. Quantitative real-time polymerase chain reaction was used to detect NEAT1 and IL-8 expression levels. Western blot was used to measure the protein levels of Bcl-2, Bax, cleaved caspase-3, and IL-8. RNA-binding protein immunoprecipitation was used to detect the interaction of NEAT1 and splicing factor proline/glutamine-rich (SFPQ). Results showed that NEAT1 knockdown decreased the adhesion ability of thrombin-stimulated MEG-01 and PLP. The expression of CD62P on PLPs and CD41/CD42b on MEG-01 cells was inhibited by NEAT1 knockdown. In addition, NEAT1 knockdown inhibited cell apoptosis with increased Bcl2/Bax ratio and decreased cleaved caspase-3, and reduced the percentage of cells in the G0/G1 phase. Meanwhile, NEAT1 knockdown inhibited the expression of IL-8. A strong interaction of NEAT1 and SFPQ, a transcriptional repressor of IL-8, was identified. NEAT1 knockdown reduced the interaction between SFPQ and NEAT1.The results suggest that lncRNA NEAT1 knockdown decreases MEG-01 differentiation, PLP activity, and IL-8 level. The results also indicate that the regulation of NEAT1 on IL-8 may be realized via a direct interaction between NEAT1 and SFPQ.

14.
PLoS One ; 15(7): e0232963, 2020.
Article in English | MEDLINE | ID: mdl-32730272

ABSTRACT

Mammalian cardiomyocytes exit the cell cycle shortly after birth. As a result, an occurrence of coronary occlusion-induced myocardial infarction often results in heart failure, postinfarction LV dilatation, or death, and represents one of the most significant public health morbidities worldwide. Interestingly however, the hearts of neonatal pigs have been shown to regenerate following an acute myocardial infarction (MI) occuring on postnatal day 1 (P1); a recovery period which is accompanied by an increased expression of markers for cell-cycle activity, and suggests that early postnatal myocardial regeneration may be driven in part by the MI-induced proliferation of pre-existing cardiomyocytes. In this study, we identified signaling pathways known to regulate the cell cycle, and determined of these, the pathways persistently upregulated in response to MI injury. We identified five pathways (mitogen associated protein kinase [MAPK], Hippo, cyclic [cAMP], Janus kinase/signal transducers and activators of transcription [JAK-STAT], and Ras) which were comprehensively upregulated in cardiac tissues collected on day 7 (P7) and/or P28 of the P1 injury hearts. Several of the initiating master regulators (e.g., CSF1/CSF1R, TGFB, and NPPA) and terminal effector molecules (e.g., ATF4, FOS, RELA/B, ITGB2, CCND1/2/3, PIM1, RAF1, MTOR, NKF1B) in these pathways were persistently upregulated at day 7 through day 28, suggesting there exists at least some degree of regenerative activity up to 4 weeks following MI at P1. Our observations provide a list of key regulators to be examined in future studies targeting cell-cycle activity as an avenue for myocardial regeneration.


Subject(s)
Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Animals , Animals, Newborn , Cyclic AMP/metabolism , MAP Kinase Signaling System , Myocardial Infarction/metabolism , Swine , Time Factors
15.
Aging (Albany NY) ; 12(7): 6385-6400, 2020 04 08.
Article in English | MEDLINE | ID: mdl-32267831

ABSTRACT

Long noncoding RNAs (lncRNAs) play important roles in the development of vascular diseases. However, the effect of lncRNA NORAD on atherosclerosis remains unknown. This study aimed to investigate the effect NORAD on endothelial cell injury and atherosclerosis. Ox-LDL-treated human umbilical vein endothelial cells (HUVECs) and high-fat-diet (HFD)-fed ApoE-/- mice were used as in vitro and in vivo models. Results showed that NORAD-knockdown induced cell cycle arrest in G0/G1 phase, aggravated ox-LDL-induced cell viability reduction, cell apoptosis, and cell senescence along with the increased expression of Bax, P53, P21 and cleaved caspase-3 and the decreased expression of Bcl-2. The effect of NORAD on cell viability was further verified via NORAD-overexpression. NORAD- knockdown increased ox-LDL-induced reactive oxygen species, malondialdehyde, p-IKBα expression levels and NF-κB nuclear translocation. Proinflammatory molecules ICAM, VCAM, and IL-8 were also increased by NORAD- knockdown. Additionally, we identified the strong interaction of NORAD and IL-8 transcription repressor SFPQ in HUVECs. In ApoE-/- mice, NORAD-knockdown increased the lipid disorder and atherosclerotic lesions. The results have suggested that lncRNA NORAD attenuates endothelial cell senescence, endothelial cell apoptosis, and atherosclerosis via NF-κB and p53-p21 signaling pathways and IL-8, in which NORAD-mediated effect on IL-8 might through the direct interaction with SFPQ.


Subject(s)
Atherosclerosis/genetics , Endothelial Cells/metabolism , Interleukin-8/metabolism , Lipoproteins, LDL/metabolism , RNA, Long Noncoding/metabolism , Animals , Apoptosis , Cell Cycle Checkpoints , Cellular Senescence , Down-Regulation , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells , Humans , Mice , NF-kappa B/metabolism , Oxidation-Reduction , Signal Transduction
16.
Biomolecules ; 10(2)2020 01 25.
Article in English | MEDLINE | ID: mdl-31991759

ABSTRACT

Heart failure (HF) is a deadly disease that is difficult to accurately diagnose. Circular RNAs (circRNAs) are a novel class of noncoding RNAs that might play important roles in many cardiovascular diseases. However, their role in HF remains unclear. CircRNA microarrays were performed on plasma samples obtained from three patients with HF and three healthy controls. The profiling results were validated by quantitative reverse transcription polymerase chain reaction. The diagnostic value of circRNAs for HF was evaluated by receiver operating characteristic (ROC) curves. The expression profiles indicated that 477 circRNAs were upregulated and 219 were downregulated in the plasma of patients with HF compared with healthy controls. Among the dysregulated circRNAs, hsa_circ_0112085 (p = 0.0032), hsa_circ_0062960 (p = 0.0006), hsa_circ_0053919 (p = 0.0074) and hsa_circ_0014010 (p = 0.025) showed significantly higher expression in patients with HF compared with healthy controls. The area under the ROC curve for hsa_circ_0062960 for HF diagnosis was 0.838 (p < 0.0001). Correlation analysis showed that the expression of hsa_circ_0062960 was highly correlated with B-type natriuretic peptide (BNP) serum levels. Some differential circRNAs were found to be related to platelet activity by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. The landscape of circRNA expression profiles may play a role in HF pathogenesis and improve our understanding of platelet function in HF. Moreover, hsa_circ_0062960 has potential as a novel diagnostic biomarker for HF.


Subject(s)
Gene Expression Regulation/genetics , Heart Failure/blood , RNA, Circular/blood , Aged , Female , Heart Failure/genetics , Heart Failure/pathology , Humans , Male , Microarray Analysis , Middle Aged , Natriuretic Peptide, Brain/blood , Natriuretic Peptide, Brain/genetics , Platelet Activation/genetics
17.
Biomolecules ; 9(11)2019 10 24.
Article in English | MEDLINE | ID: mdl-31652997

ABSTRACT

BACKGROUND: Disease-related injury in any organ triggers a complex cascade of cellular and molecular responses that culminate in tissue fibrosis, inflammation, and angiogenesis simultaneously. Multiple cell angiogenesis is an essential part of the tissue damage response, which is involved in fibrosis development. RC28-E is a novel recombinant dual decoy receptor lgG1 Fc-fusion protein that can block vascular endothelial growth factor (VEGFA), platelet-derived growth factor (PDGF), and fibroblast growth factor-2 (FGF-2) simultaneously. This protein has stepped into clinical trials (NCT03777254) for the treatment of pathological neovascularization-related diseases. Here, we report on the role of RC28-E during anti-fibrosis and its potential multitarget function in regulating fibrosis. METHODS: A bleomycin-induced pulmonary fibrosis C57BL/6 mouse model was established. Hematoxylin and eosin staining (HE) and Masson staining (Masson's) were performed to evaluate the pulmonary fibrosis based on the scoring from, Ashcroft score. Fibrosis related factors and inflammatory cytokines including HYP, α-SMA, procollagen, ICAM, IL-6, IL-1, and TNF-α were also determined at the protein and mRNA levels to characterize the fibrosis. Both mRNA and protein levels of VEGF, FGF, and transforming growth factor (TGF)-ß were detected by quantitative real-time PCR (qRT-PCR) and immunohistochemical (IHC) analysis, respectively. Pulmonary fibrosis and related cytokines were re-evaluated in vivo after 3 doses of RC28-E (5 mg/kg, 15 mg/kg, and 50 mg/kg, ip. Tiw × 9) in comparison with a mono-target antagonist treatment (VEGF or FGF blocking). RC28-E attenuated the activation of TGF-ß induced fibroblasts in vitro. Expression levels of α-SMA and collagen I, as well as proliferation and migration, were determined with the human skin fibroblast cell line Detroit 551 and primary murine pulmonary fibroblast cells. The mechanism of RC28-E via the TGF-ß/Smad pathway was also investigated. RESULTS: RC28-E exhibits significant anti-fibrosis effects on Idiopathic pulmonary fibrosis (IPF) in vivo. Moreover, TGF-ß induced fibroblast activation in vitro via the inhibition of the TGF-ß downstream Smad pathway, thus providing potential therapeutics for clinical disease-related fibrosis-like IPF as well as chemotherapy-induced fibrosis in cancer therapy.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Lung Injury/drug therapy , Pulmonary Fibrosis/drug therapy , Recombinant Fusion Proteins/therapeutic use , Angiogenesis Inhibitors/pharmacology , Animals , Bleomycin , Cell Line , Cell Survival/drug effects , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblasts/drug effects , Humans , Lung/drug effects , Lung/pathology , Lung Injury/chemically induced , Lung Injury/pathology , Male , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Recombinant Fusion Proteins/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors
18.
Fish Shellfish Immunol ; 82: 531-535, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30176335

ABSTRACT

Circular RNAs (CircRNAs) represent an important class of noncoding RNAs to regulate biological and development processes. In the present study, the cDNA library was constructed and sequenced using Illumina Hiseq 2000 platform to identify circRNAs in half-smooth tongue sole. In the sequencing library, we validated a conservatively expressed circRNA with 363 nucleotides on W chromosome (termed as Wcirc5284614), which was spliced with the exon3, exon4, and exon5 of CNOT6L. Reverse transcription polymerase chain reaction indicated that Wcirc5284614 was mainly expressed in female's immune tissues, especially in the head kidney and spleen of the female. Comparative analysis showed that circRNA was highly conservative in vertebrate. These circRNA profiles provide new valuable resources for further study on the function and mechanism of circRNAs in half-smooth tongue sole, especially in immune organs.


Subject(s)
Flounder/genetics , Flounder/immunology , Head Kidney/immunology , RNA/genetics , Spleen/immunology , Animals , Female , Gene Library , Head Kidney/metabolism , Male , RNA, Circular , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Sex Factors , Spleen/metabolism
19.
Acta Pharmacol Sin ; 39(5): 885-892, 2018 May.
Article in English | MEDLINE | ID: mdl-29698390

ABSTRACT

Contact inhibition and its disruption of vascular smooth muscle cells (VSMCs) are important cellular events in vascular diseases. But the underlying molecular mechanisms are unclear. In this study we investigated the roles of microRNAs (miRNAs) in the contact inhibition and its disruption of VSMCs and the molecular mechanisms involved. Rat VSMCs were seeded at 30% or 90% confluence. MiRNA expression profiles in contact-inhibited confluent VSMCs (90% confluence) and non-contact-inhibited low-density VSMCs (30% confluence) were determined. We found that multiple miRNAs were differentially expressed between the two groups. Among them, miR-145 was significantly increased in contact-inhibited VSMCs. Serum could disrupt the contact inhibition as shown by the elicited proliferation of confluent VSMCs. The contact inhibition disruption accompanied with a down-regulation of miR-145. Serum-induced contact inhibition disruption of VSMCs was blocked by overexpression of miR-145. Moreover, downregulation of miR-145 was sufficient to disrupt the contact inhibition of VSMCs. The downregulation of miR-145 in serum-induced contact inhibition disruption was related to the activation PI3-kinase/Akt pathway, which was blocked by the PI3-kinase inhibitor LY294002. KLF5, a target gene of miR-145, was identified to be involved in miR-145-mediated effect on VSMC contact inhibition disruption, as it could be inhibited by knockdown of KLF5. In summary, our results show that multiple miRNAs are differentially expressed in contact-inhibited VSMCs and in non-contact-inhibited VSMCs. Among them, miR-145 is a critical gene in contact inhibition and its disruption of VSMCs. PI3-kinase/Akt/miR-145/KLF5 is a critical signaling pathway in serum-induced contact inhibition disruption. Targeting of miRNAs related to the contact inhibition of VSMCs may represent a novel therapeutic approach for vascular diseases.


Subject(s)
Contact Inhibition/physiology , MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Animals , Cell Count , Cell Proliferation/physiology , Chromones/pharmacology , Down-Regulation , Kruppel-Like Transcription Factors/metabolism , Male , MicroRNAs/genetics , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Signal Transduction/physiology
20.
Nan Fang Yi Ke Da Xue Xue Bao ; 36(9): 1242-1246, 2016 08 20.
Article in Chinese | MEDLINE | ID: mdl-27687658

ABSTRACT

OBJECTIVE: To construct a lentivirus vector carrying wheat germ agglutinin (WGA) and evaluate its ability of tracing WGA in the brain of mice with ischemic brain injury. METHODS: WGA gene was inserted into the lentiviral vector Plvx IRES-ZsGreen1 using genetic engineering methods. 293T cells were transfected with the vector and 3 packaging plasmids (RPEV, PRRE, and VSVG) to obtain the recombinant lentivirus for infection of human adipose-derived stem cells (hADSCs). The infected hADSCs were injected into the damaged brain area by in situ injection in a mouse model of middle cerebral artery occlusion (MCAO) and the expression of GFP was traced. RESULTS: Immunofluorescence identification detected WGA protein expression in the infected hADSCs, which survived in the infarct area of mice with MCAO. CONCLUSION: Packaging WGA gene in lentivirus is a reliable approach to allow efficient neuroanatomical tracing of various cells.


Subject(s)
Adipose Tissue/cytology , Genetic Vectors , Stem Cells/cytology , Transfection , Wheat Germ Agglutinins/metabolism , Animals , HEK293 Cells , Humans , Lentivirus , Mice , Plasmids , Wheat Germ Agglutinins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...