Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
Cardiovasc Res ; 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39082269

ABSTRACT

AIMS: Circular RNAs are important players involving in a variety of physiological and pathological processes. However, their functions and mechanisms during myocardial ischemic injury and protection remain largely unknown. We recently found significant alterations of many circRNAs including circCHSY1 following myocardial ischemia/reperfusion (I/R) injury, whereas their exact functions are unclear. Here, we investigated roles of circCHSY1 in the acute myocardial I/R injury and the potential mechanisms involved. METHODS AND RESULTS: The expression of circCHSY1 was detected in cardiomyocytes from mouse, rat and human embryonic stem cells (hESC-CMs). It was further upregulated in mouse I/R (30 min/24 h) hearts, oxygen glucose deprivation/reperfused (OGD/R, 6 h/2 h) primary neonatal rat ventricular cardiomyocytes (NRCMs) and OGD/R (48 h/2 h) hESC-CMs. Adenovirus-mediated circCHSY1-overexpression significantly decreased infarct size and lactate dehydrogenase (LDH) release in mouse I/R hearts. Consistently, circCHSY1 overexpression reduced the LDH release in the OGD/R NRCMs and hESC-CMs, improved cell viability, and preserved mitochondrial function in the OGD/R NRCMs, whereas there were no significant differences in cell viability and LDH release between the OGD/R NRCMs with and without siRNA-mediated circCHSY1 knockdown. Mechanistically, circCHSY1 was detected to bind with miR-24-3p analyzed by dual luciferase assay and RNA pull-down assays. CircCHSY1 overexpression-mediated protective effects on cells and mitochondria in OGD/R NRCMs were reversed by the miR-24-3p mimic. Further, dual luciferase assay showed that miR-24-3p directly bound to heme oxygenase 1 (HO1) via its 3'UTR. The protein level of HO1 was downregulated by miR-24-3p mimic in OGD/R NRCMs but enhanced by the circCHSY1 overexpression in vitro and in vivo. Functionally, the HO1 knockdown by adenovirus in vivo and by siRNA in vitro eliminated cardioprotective effects of circCHSY1 overexpression. CONCLUSION: CircCHSY1 is upregulated following myocardial I/R injury. The higher level of circCHSY1 protects I/R hearts and cardiomyocytes. The protection of circCHSY1 is mediated through enhancement of the HO1 level, resulting in preserving mitochondrial homeostasis via targeting miR-24-3p in cardiomyocytes. These findings suggest circCHSY1 as a protective factor.

2.
Gene ; 898: 148129, 2024 Mar 10.
Article in English | MEDLINE | ID: mdl-38184021

ABSTRACT

Extracellular matrix (ECM) and vascular smooth muscle cells (VSMCs) are the main components of atherosclerosis (AS) plaque. VSMCs participate in plaque formation through phenotypic transformation. The complex interplay between ECM and VSMCs plays vital roles in the progression of AS throughout the disease. An in-depth investigation into the functions of ECM-related molecules in VSMC development might contribute to deciphering the complexity of AS pathogenesis. In this study, the roles and molecular mechanisms of the ECM-related molecule Fibulin-1 (FBLN1) in the development of AS and VSMCs were explored using RNA sequencing, bioinformatics analysis, and cell experiments. Furthermore, the expression of FBLN1, as determined by western blot analysis, immunohistochemistry, and real-time quantitative PCR, was significantly increased in AS vascular samples compared to normal vascular samples. Silencing the FBLN1 through AAV viral injection in mice revealed an improvement in AS. Functional analyses revealed that FBLN1 promoted VSMC proliferation, migration, and wound healing. Combined with RNA sequencing and TargetScan7.2 prediction data, 22 microRNAs (miRNAs) were found to have the potential for direct interaction with the FBLN1 3'UTR in VSMCs. Among these 22 miRNAs, it was demonstrated that microRNA-24-3p (miR-24-3p) could negatively regulate FBLN1 expression by directly binding to the FBLN1 3'UTR. Moreover, miR-24-3p inhibited cell proliferation, migration, and wound healing, and suppressed the expression of Ki67, matrix metalloproteinase-2 and -9 (MMP2/9) by targeting FBLN1 in VSMCs. Meanwhile, inhibition of FBLN1 expression could restrain VSMC phenotypic transformation. In conclusion, miR-24-3p inhibited VSMC proliferation and migration by targeting FBLN1. Additionally, multiple miRNAs with the potential to interact with the FBLN1 3'UTR were identified. These findings might deepen our understanding of ECM gene regulatory networks and the complex etiology of AS.


Subject(s)
Atherosclerosis , Calcium-Binding Proteins , MicroRNAs , Animals , Mice , 3' Untranslated Regions , Apolipoproteins E/genetics , Atherosclerosis/metabolism , Cell Movement/genetics , Cell Proliferation/genetics , Cells, Cultured , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism
3.
Mol Reprod Dev ; 89(9): 423-430, 2022 09.
Article in English | MEDLINE | ID: mdl-35818817

ABSTRACT

Preeclampsia (PE) is a pregnancy-related disease and is the leading cause of overall maternal mortality and morbidity. Our previous studies have shown that the serum and placental levels of retinol-binding protein 4 (RBP4) in PE are reduced. Our previous bioinformatics analysis predicted that RBP4 is a target of the microRNA miRNA-24-3p. In this study, our database analysis also indicated that RBP4 is a miR-24-3p target. Compared with that of the normal placenta, the expression level of RBP4 in human PE placenta was significantly reduced, and miR-24-3p was highly expressed. In HTR-8/SVneo cells, transfection of exogenous miR-24-3p reduced RBP4 expression. A dual-luciferase reporter assay validated RBP4 as a direct target of miR-24-3p, indicating that it directly binds to the 3'-untranslated region of RBP4. This binding was reversed by a mutation in the microRNA-binding site. Transwell invasion experiments and CCK8 assay showed that inhibitory effect of miR-24-3p reduced RBP4 mediated HTR-8/SVneo cell invasion and proliferation. These data provide a new overarching perspective on the physiological role played by miR-24-3p in regulating RBP4 during trophoblast dysfunction and PE development.


Subject(s)
MicroRNAs , Pre-Eclampsia , Retinol-Binding Proteins, Plasma , Trophoblasts , 3' Untranslated Regions/genetics , Cell Movement/genetics , Cell Proliferation , Female , Humans , Luciferases , MicroRNAs/genetics , MicroRNAs/metabolism , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Pregnancy , Retinol-Binding Proteins, Plasma/genetics , Retinol-Binding Proteins, Plasma/metabolism , Trophoblasts/metabolism
4.
Mol Reprod Dev ; 89(7): 271-280, 2022 07.
Article in English | MEDLINE | ID: mdl-35735229

ABSTRACT

Long noncoding RNA plasmacytoma variant translocation 1 (PVT1) is essential for the maintenance of normal functions of trophoblasts in preeclampsia (PE). This study aims to decipher the concrete mechanism of PVT1 with the microRNA-24-3p/Type-2 11ß-hydroxysteroid dehydrogenase (miR-24-3p/HSD11B2) axis in PE. PVT1, miR-24-3p, and HSD11B2 expression levels in normal placental tissues and PE placental tissues were defined. HTR-8/SVneo cells were transfected to determine the effects of PVT1, miR-24-3p, and HSD11B2 on the growth of HTR-8/SVneo cells. The relationships among PVT1/miR-24-3p/HSD11B2 in HTR-8/SVneo cells were identified. PVT1 and HSD11B2 were downregulated, while miR-24-3p was upregulated in the placenta of PE. Upregulated/downregulated PVT1 promoted/impeded the growth of human placental trophoblast (HTR-8/SVneo) cells in PE. Restored/knocked down miR-24-3p impeded/enhanced the growth of HTR-8/SVneo cells in PE. PVT1 inhibited miR-24-3p to mediate HSD11B2. PVT1 sponges miR-24-3p to regulate HSD11B2; thereby, the growth of placental trophoblasts is promoted in PE.


Subject(s)
MicroRNAs , Pre-Eclampsia , RNA, Long Noncoding , Trophoblasts , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Cell Movement/genetics , Cell Proliferation/genetics , Female , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Placenta/metabolism , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Pregnancy , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Trophoblasts/metabolism
5.
Cell Cycle ; 21(7): 674-684, 2022 04.
Article in English | MEDLINE | ID: mdl-35113004

ABSTRACT

Long non-coding RNAs (lncRNAs) are crucial drivers in the progression of human diseases such as myocardial infarction (MI). However, the impact of lncRNA MCM3AP antisense RNA 1 (MCM3AP-AS1) on MI remains unknown. This research was determined to explore the effect of MCM3AP-AS1 modulating microRNA-24-3p (miR-24-3p) and eukaryotic translation initiation factor 4 gamma 2 (EIF4G2) on MI. The rat MI models were constructed and, respectively, treated with altered MCM3AP-AS1, miR-24-3p or/and EIF4G2. Subsequently, the cardiac function, myocardial pathological injury, malondialdehyde content and superoxide dismutase activity were determined. The vascular endothelial cells (VECs) were isolated and treated severally, and then proliferation and migration of VECs were measured. MCM3AP-AS1, miR-24-3p, EIF4G2 and vascular endothelial growth factor (VEGF) expressions in myocardial tissues and VECs were assessed. MCM3AP-AS1 and EIF4G2 were upregulated while miR-24-3p and VEGF were downregulated in MI rat myocardial tissues. MCM3AP-AS1 silencing or miR-24-3p elevation improved cardiac function and myocardial pathological injury, suppressed malondialdehyde content, and also enhanced VEGF expression and superoxide dismutase activity in MI rats. In VECs, downregulated MCM3AP-AS1 or upregulated miR-24-3p accelerated cell proliferation and migration. These effects of miR-24-3p upregulation were reversed by overexpressed EIF4G2. Our study summarizes that reduced MCM3AP-AS1 elevates miR-24-3p to promote proliferation and migration of MI rat VECs by inhibiting EIF4G2.


Subject(s)
MicroRNAs , Myocardial Infarction , RNA, Long Noncoding , Acetyltransferases/genetics , Animals , Cell Proliferation/genetics , Endothelial Cells/metabolism , Intracellular Signaling Peptides and Proteins , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardial Infarction/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Rats , Vascular Endothelial Growth Factor A/genetics
6.
Mol Immunol ; 141: 309-317, 2022 01.
Article in English | MEDLINE | ID: mdl-34933177

ABSTRACT

OBJECTIVE: Some reports have suggested the involvement of microRNA-24-3p (miR-24-3p) in heart diseases. Here, the intention of this work was to unmask whether miR-24-3p from M2 macrophages-derived exosomes (M2-exo) could protect against myocardial injury after sepsis. METHODS: Mice model of sepsis was induced by intraperitoneal injection of lipopolysaccharide (LPS). miR-24-3p and tumor necrosis factor superfamily member 10 (Tnfsf10) expression levels were measured in the myocardial tissue of septic mice. M2-exo were isolated, in which miR-24-3p expression was altered. Then, septic mice were alone or in combination injected with the miR-24-3p-modified M2-exo or siRNA of Tnfsf10. Subsequently, cardiac function, apoptosis and serum inflammatory response were examined. RESULTS: miR-24-3p expression dropped while Tnfsf10 expression raised in the myocardial tissue of septic mice. M2-exo-derived miR-24-3p or deficiency of Tnfsf10 had cardioprotective effects on LPS-induced myocardial injury in mice through improving cardiac function and reducing cardiomyocyte apoptosis in the myocardial tissue and serum inflammation. A binding relation exhibited between miR-24-3p and Tnfsf10, and M2-exo-derived miR-24-3p alleviated LPS-induced myocardial injury by inhibiting Tnfsf10. CONCLUSION: Up-regulating miR-24-3p from M2-exo imposes cardioprotection against myocardial injury after sepsis through reducing Tnfsf10 expression.


Subject(s)
Exosomes/genetics , Macrophages/metabolism , MicroRNAs/genetics , Myocardium/metabolism , Sepsis/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics , Animals , Apoptosis/genetics , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Up-Regulation/genetics
7.
Mol Med ; 27(1): 107, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34521353

ABSTRACT

OBJECTIVES: Studies have widely explored in the filed of ischemic stroke (IS) with their focus on transcription factors. However, few studies have pivoted on sex determining region Y-box 2 (SOX2) in IS. Thus, this study is launched to figure out the mechanisms of SOX2 in IS. METHODS: Rat middle cerebral artery occlusion (MCAO) was established as a stroke model. MCAO rats were injected with depleted SOX2 or long non-coding RNA plasmacytoma variant translocation 1 (PVT1) to explore their roles in neurological deficits, cerebral water content, neuron survival, apoptosis and oxidative stress. The relationship among SOX2, PVT1, microRNA (miR)-24-3p and signal transducer and activator of transcription 3 (STAT3) was verified by a series of experiments. RESULTS: SOX2, PVT1 and STAT3 were highly expressed while miR-24-3p was poorly expressed in cerebral cortex tissues of MCAO rats. Depleted SOX2 or PVT1 alleviated brain injury in MCAO rats as reflected by neuronal apoptosis and oxidative stress restriction, brain water content reduction, and neurological deficit and neuron survival improvements. Overexpression of PVT1 functioned oppositely. Restored miR-24-3p abolished PVT1 overexpression-induced brain injury in MCAO rats. SOX2 directly promoted PVT1 expression and further increased STAT3 by sponging miR-24-3p. CONCLUSION: This study presents that depleting SOX2 improves IS via PVT1/miR-24-3p/STAT3 axis which may broaden our knowledge about the mechanisms of SOX2/PVT1/miR-24-3p/STAT3 axis and provide a reference of therapy for IS.


Subject(s)
Gene Expression Regulation , Ischemic Stroke/etiology , Ischemic Stroke/metabolism , MicroRNAs/genetics , RNA, Long Noncoding/genetics , SOXB1 Transcription Factors/metabolism , STAT3 Transcription Factor/metabolism , Animals , Biomarkers , Cell Line , Disease Susceptibility , Genes, Reporter , Humans , Immunohistochemistry , Ischemic Stroke/pathology , Male , Oxidative Stress , RNA Interference , Rats , SOXB1 Transcription Factors/genetics , Signal Transduction
8.
Mol Med Rep ; 23(6)2021 06.
Article in English | MEDLINE | ID: mdl-33880595

ABSTRACT

Abnormal circular RNAs (circRNAs) are associated with biological processes in cancer; however, the function of circRNAs remains largely unknown in non­small cell lung cancer (NSCLC). The present study aimed to investigate the role of hsa_circ_0058357 on the progression of NSCLC. Cell proliferation, migration and apoptosis were determined using Cell Counting Kit­8, Transwell and flow cytometry assays, respectively. Gene [circRNA and microRNA (miR)] and protein expression levels were determined via reverse transcription­quantitative PCR and immunoblotting. A luciferase assay was employed to detect the binding of miR­24­3p with AVL9 cell migration associated (AVL9), while a cancer xenograft model was established to evaluate cancer growth in vivo. The results demonstrated that hsa_circ_0058357 was highly expressed in human NSCLC tissues and NSCLC cells compared with para­cancerous tissues and human bronchial epithelial (HBE) cells, respectively. Knockdown of hsa_circ_0058357 significantly suppressed cell viability, migration and tumor growth, while it promoted apoptosis in NSCLC cells. As a competing endogenous RNA, hsa_circ_0058357 knockdown contributed to the increase of miR­24­3p expression in NSCLC cells. Of note, overexpression of miR­24­3p markedly abolished the exogenous hsa_circ_0058357­induced excessive proliferation, migration and apoptosis resistance of NSCLC cells. Mechanistically, as a signaling molecule in late secretory pathway, AVL9 was also expressed at a high level in NSCLC tissues and cells, which could be directly suppressed by miR­24­3p. In the tumor tissues, along with growth inhibition, hsa_circ_0058357 knockdown also mediated the elevation of miR­24­3p and the reduction of AVL9. Thus, it was suggested that hsa_circ_0058357 may be a crucial regulation factor in NSCLC by sponging hsa­miR­24­3p, leading to a decrease in miR­24­3p expression, and subsequent increase in AVL9 expression. Therefore, hsa_circ_0058357 may serve as a potential target for diagnosis and gene therapy for NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , MicroRNAs/metabolism , RNA, Circular/metabolism , Vesicular Transport Proteins/metabolism , Animals , Apoptosis , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Middle Aged , Signal Transduction , Vesicular Transport Proteins/genetics , Xenograft Model Antitumor Assays
9.
Exp Ther Med ; 21(4): 367, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33732340

ABSTRACT

Retinoblastoma (RB) is the most common primary intraocular cancer type that occurs during retinal development in childhood. Previous studies have reported that long non-coding RNAs (lncRNAs) are involved in the development of RB. Therefore, the aim of the present study was to investigate the effects and underlying regulatory mechanisms of nuclear paraspeckle assembly transcript 1 (NEAT1) in RB. The expression levels of NEAT1, microRNA (miR)-24-3p and leucine-rich-α-2-glycoprotein (LRG1) were detected using reverse transcription-quantitative PCR (RT-qPCR). Moreover, the protein expression levels of LRG1, matrix metalloproteinase 9, N-cadherin and E-cadherin were detected via western blotting. Furthermore, cell migration and invasion abilities were evaluated via Transwell assays. The targeting relationships between miR-24-3p and NEAT1 or LRG1 were predicted using online software and confirmed via dual-luciferase reporter assay. In the present study, NEAT1 and LRG1 were upregulated, and miR-24-3p was downregulated in RB tissues and cells compared with the corresponding healthy tissues and cells. Moreover, miR-24-3p was identified as a target of NEAT and LRG1 was demonstrated to be a direct target gene of miR-24-3p. Knockdown of NEAT1 or LRG1 significantly suppressed RB cell migration and invasion ability, while the effects were reversed by an miR-24-3p inhibitor. In addition, the downregulation of LRG1 caused by miR-24-3p was restored following the overexpression of NEAT1 in RB cells. It was also demonstrated that NEAT1 knockdown inhibited the epithelial-to-mesenchymal transition (EMT) pathway by inhibiting the expression of LRG via targeting miR-24-3p. In conclusion, the present results suggest that silencing of NEAT1 suppresses cell migration, invasion and the EMT process by downregulating LRG1 expression via sponging miR-24-3p in RB, thus indicating that NEAT1 may be a potential candidate for RB treatment.

10.
J Cell Mol Med ; 25(8): 3699-3713, 2021 04.
Article in English | MEDLINE | ID: mdl-33621425

ABSTRACT

MicroRNA-24-3p (miR-24-3p) has been implicated as a key promoter of chemotherapy resistance in numerous cancers. Meanwhile, cancer-associated fibroblasts (CAFs) can secret exosomes to transfer miRNAs, which mediate tumour development. However, little is known regarding the molecular mechanism of CAF-derived exosomal miR-24-3p in colon cancer (CC). Hence, this study intended to characterize the functional relevance of CAF-derived exosomal miR-24-3p in CC cell resistance to methotrexate (MTX). We identified differentially expressed HEPH, CDX2 and miR-24-3p in CC through bioinformatics analyses, and validated their expression in CC tissues and cells. The relationship among HEPH, CDX2 and miR-24-3p was verified using ChIP and dual-luciferase reporter gene assays. Exosomes were isolated from miR-24-3p inhibitor-treated CAFs (CAFs-exo/miR-24-3p inhibitor), which were used in combination with gain-of-function and loss-of-function experiments and MTX treatment. CCK-8, flow cytometry and colony formation assays were conducted to determine cell viability, apoptosis and colony formation, respectively. Based on the findings, CC tissues and cells presented with high expression of miR-24-3p and low expression of HEPH and CDX2. CDX2 was a target gene of miR-24-3p and could up-regulate HEPH. Under MTX treatment, overexpressed CDX2 or HEPH and down-regulated miR-24-3p reduced cell viability and colony formation and elevated cell apoptosis. Furthermore, miR-24-3p was transferred into CC cells via CAF-derived exosomes. CAF-derived exosomal miR-24-3p inhibitor diminished cell viability and colony formation and increased cell apoptosis in vitro and inhibited tumour growth in vivo under MTX treatment. Altogether, CAF-derived exosomal miR-24-3p accelerated resistance of CC cells to MTX by down-regulating CDX2/HEPH axis.


Subject(s)
CDX2 Transcription Factor/metabolism , Colonic Neoplasms/drug therapy , Drug Resistance, Neoplasm , Exosomes/genetics , Membrane Proteins/metabolism , Methotrexate/pharmacology , MicroRNAs/genetics , Aged , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , CDX2 Transcription Factor/genetics , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Proliferation , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Prognosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Exp Ther Med ; 19(4): 2467-2474, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32256723

ABSTRACT

Numerous reports have shown that dysfunction of vascular smooth muscle cells (VSMCs) serves a critical function in the development of cardiovascular disease, including coronary heart disease (CHD). microRNAs (miRNAs/miRs) have been reported to play important roles in regulating the function of VSMCs. The present study aimed to determine the role of miR-24-3p in VSMCs and to uncover the underlying mechanism. The expression of miR-24-3p in the peripheral blood samples of CHD patients was measured by reverse transcription-quantitative (RT-q)PCR. It was found that the level of miR-24-3p in the peripheral blood of patients with CHD was significantly upregulated compared with that in healthy controls. A dual luciferase reporter assay was performed to determine whether Bcl-2-like protein 11 (Bcl-2L11) was a target gene of miR-24-3p, and it was identified that Bcl-2L11 was a direct target of miR-24-3p. The mRNA level and protein expression of Bcl-2L11 in the peripheral blood of patients with CHD were measured by RT-qPCR and western blotting, respectively. The findings suggested that Bcl-2L11 was downregulated in the peripheral blood of patients with CHD. In addition, it was found that downregulation of miR-24-3p suppressed VSMC proliferation and promoted VSMC apoptosis, while the effects of the miR-24-3p inhibitor on cell viability and apoptosis were reversed by Bcl-2L11-small interfering (si)RNA. Additionally, downregulation of miR-24-3p increased the levels of Bcl-2L11, caspase-3 and Bax, and decreased Bcl-2 expression in VSMCs; these changes were abolished by Bcl-2L11-siRNA. In conclusion, the aforementioned results indicated that miR-24-3p was an important regulator in VSMC proliferation and apoptosis by targeting Bcl-2L11, which suggested that miR-24-3p might be a potential therapeutic target for the treatment of CHD.

12.
Biochem Biophys Res Commun ; 522(1): 47-52, 2020 01 29.
Article in English | MEDLINE | ID: mdl-31735332

ABSTRACT

Ischemia and reperfusion (I/R) injury is a common cause of hepatocyte injury and liver dysfunction during liver transplantation, but its mechanism is needed further explored. We aimed to investigate whether STING pathway activation is involved in the liver I/R and further determine the role of the microRNA(miR)-24-3p in liver I/R injury in mice. Our data showed that STING mRNA level was negatively related with miR-24-3p in livers of I/R-treated mice. Next, we identified that STING could be bound by miR-24-3p by bioinformatic and luciferase report assay. Moreover, downregulation of STING alleviated the protein expression of p-IRF3 and the serum level of inflammatory factor and aminotransferase in I/R mice model. Furthermore, transfection of I/R treated mice with exogenous miR-24-3p significantly inhibited the protein expression of STING and p-IRF3 in liver, and attenuated serum inflammatory cytokines release, as well as the dysfunction and apoptosis of liver in I/R model in vivo. This study suggests that miR-24-3p may ameliorate inflammatory response and cellular apoptosis in hepatic I/R process by targeting STING, which might be a potential therapeutic target for preventing liver I/R development and progression.


Subject(s)
Gene Expression Regulation , Liver/pathology , Membrane Proteins/metabolism , MicroRNAs/metabolism , Reperfusion Injury/pathology , Animals , Apoptosis , Disease Progression , Inflammation , Interferon Regulatory Factor-3/metabolism , Liver/metabolism , Liver Diseases/metabolism , Mice
13.
Oncol Lett ; 17(1): 365-371, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30655776

ABSTRACT

Hodgkin's lymphoma (HL) is a common hematologic tumor, and the incidence is increasing. At present, it is considered that miRNAs are closely related to HL. Substantial attention has been paid to the effects of miRNA on the pathophysiological process of HL. This study was focused on the potential role of miR-24-3p in HL by targeting DEDD. The reverse transcription-quantitative PCR (RT-qPCR) results demonstrated that miR-24-3p expression was highly elevated and DEDD expression reduced inversely in HL tissues compared to adjacent tissues. According to the results of CKK-8 assays, miR-24-3p was able to accelerate HL cell proliferation. In addition, the results of the Transwell assays also indicated that miR-24-3p promoted the invasion and migration abilities of HL cells. Moreover, the results demonstrated that miR-24-3p inhibited DEDD expression. Hence, the present study revealed that miR-24-3p could accelerate HL development through inhibiting DEDD.

14.
Eur J Immunol ; 43(11): 3028-37, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23934711

ABSTRACT

Plasma cells can survive for long periods and continuously secrete protective antibodies, but plasma cell production of autoantibodies or transformation to tumor cells is detrimental. Plasma cell survival depends on exogenous factors from the surrounding microenvironment, and largely unknown intracellular mediators that regulate cell homeostasis. Here we investigated the contribution of the microRNA 24-3p (miR-24-3p) to the survival of human plasma cells under the influence of IL-6 and SDF-1α (stromal cell derived factor 1), both of which are bone marrow survival niche mediators. Deep sequencing revealed a strong expression of miR-24-3p in primary B cells, plasma blasts, plasma cells, and in plasmacytoma cells. In vitro studies using primary cells and the plasmacytoma cell line RPMI-8226 revealed that (i) expression of miR-24-3p mediates plasma cell survival, (ii) miR-24-3p is upregulated by IL-6 and SDF-1α, (iii) IL-6 mediates cell survival under ER stress conditions via miR-24-3p expression, and (iv) IL-6-induced miR-24-3p expression depends on the activity of the MAP kinase Erk1/2. These results suggest a direct connection between an external survival signal and an intracellular microRNA in regulating plasma cell survival. miR-24-3p could therefore be a promising target for new therapeutic strategies for autoimmune and allergic diseases and for multiple myeloma.


Subject(s)
Interleukin-6/metabolism , MicroRNAs/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Plasma Cells/metabolism , B-Lymphocytes/metabolism , Base Sequence , Cell Line, Tumor , Cell Survival , Cellular Microenvironment/immunology , Chemokine CXCL12/metabolism , Endoplasmic Reticulum Stress/immunology , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/biosynthesis , Multiple Myeloma/genetics , Plasmacytoma/metabolism , Sequence Analysis, DNA , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL