logo logo
miRNA-27a Transcription Activated by c-Fos Regulates Myocardial Ischemia-Reperfusion Injury by Targeting ATAD3a. Bao Yandong,Qiao Ying,Yu Hang,Zhang Zeying,Yang Huimin,Xin Xin,Chen Yuqiong,Guo Yuxuan,Wu Nan,Jia Dalin Oxidative medicine and cellular longevity MicroRNA-27a (miR-27a) has been implicated in myocardial ischemia-reperfusion injury (MIRI), but the underlying mechanism is not well understood. This study is aimed at determining the role of miR-27a in MIRI and at investigating upstream molecules that regulate miR-27a expression and its downstream target genes. miR-27a expression was significantly upregulated in myocardia exposed to ischemia/reperfusion (I/R) and cardiomyocytes exposed to hypoxia/reoxygenation (H/R). c-Fos could regulate miR-27a expression by binding to its promoter region. Moreover, overexpression of miR-27a led to a decrease in cell viability, an increase in LDH and CK-MB secretion, and an increase in apoptosis rates. In contrast, suppression of miR-27a expression resulted in the opposite effects. ATPase family AAA-domain-containing protein 3A (ATAD3a) was identified as a target of miR-27a. miR-27a regulated the translocation of apoptosis-inducing factor (AIF) from the mitochondria to the nucleus and H/R-induced apoptosis via the regulation of ATAD3a. It was found that inhibiting miR-27a in vivo by injecting a miR-27a sponge could ameliorate MIRI in an isolated rat heart model. In conclusion, our study demonstrated that c-Fos functions as an upstream regulator of miR-27a and that miR-27a regulates the translocation of AIF from the mitochondria to the nucleus by targeting ATAD3a, thereby contributing to MIRI. These findings provide new insight into the role of the c-Fos/miR-27a/ATAD3a axis in MIRI. 10.1155/2021/2514947
MicroRNA-27a regulates angiotensin II-induced vascular smooth muscle cell proliferation and migration by targeting α-smooth muscle-actin in vitro. Xu Miao-Miao,Deng Hao-Yuan,Li Hui-Hua Biochemical and biophysical research communications Angiotensin II (Ang II) modulates VSMCs phenotypic switch that play a critical role in the cardiovascular diseases. MicroRNA-27a (miR-27a) has been proven to be involved in regulating vascular remodeling; however, the functional role of miR-27a in VSMCs in response to Ang II stimulation need to be elucidated. Cell proliferation and migration were measured by Cell counting kit-8 (CCK-8), BrdU incorporation and scratch wound assay in VSMCs transfected with miR-27a or its inhibitor. The target of miR-27a was confirmed using bioinformatics analysis and luciferase reporter assay. Ang II treatment time-dependently increased proliferation and migration of VSMCs accompanied with downregulation of α-smooth muscle-actin (α-SMA) and upregulation of miR-27a expression. Moreover, knockdown of miR-27a in VSMCs significantly attenuated Ang II-induced cell proliferation and migration, whereas this effect was aggravated by overexpression of miR-27a. A potential mechanistic analysis revealed that miR-27a directly targeted α-SMA, which mediated miR-27a-induced cell proliferation and migration. In conclusion, these results indicate that miR-27a acts as a novel regulator of Ang II-induced proliferation and migration by directly targeting α-SMA expression in VSMCs in vitro, and may be a potential therapeutic target for treating vascular diseases. 10.1016/j.bbrc.2019.01.047
Effect of microRNA-27a-5p on apoptosis and inflammatory response of pancreatic acinar cells in acute pancreatitis by targeting PTEN. Kong Lei,Wu Qinghua,Zhao Liangchao,Ye Jinhua,Li Nengping,Yang Huali Journal of cellular biochemistry To investigate the apoptosis and inflammatory response of microRNA-27a-5p (miR-27a-5p) in pancreatic acinar cells of acute pancreatitis (AP) and its related mechanisms. Rat pancreatic acinar cell line AR42J was treated with caerulein (10nmol/L) to construct an acute pancreatitis cell model. Quantitative real-time polymerase chain reaction was performed to measure the expression of miR-27a-5p; The miR-27a-5p mimic was transfected into cell, and the apoptosis rate of the cells was detected by flow cytometry; The levels of TNF-α, IL-1, and IL-6 in the culture supernatant were determined by enzyme-linked immunosorbent assay; TargetScans database predicted and dual luciferase reporter gene assay verified the relationship between miR-27a-5p and the phosphatase and tensin homolog deleted on chromosome 10 (PTEN); The recovery experiment explored the apoptosis and the effects of inflammatory responses. The expression of miR-27a-5p decreased gradually (P < 0.05) and the expression of PTEN increased gradually (P < 0.05) with the prolongation of acting time. Upregulation of miR-27a-5p significantly promoted cell apoptosis (P < 0.05) and inhibited inflammatory response (P < 0.05); The TargetScans database predicted that the 3'UTR of PTEN contains a base complementary to the miR-27a-5p seed region. Cotransfection of wild-type vector (PTEN-WT) with miR-27a-5p mimic or miR-27a-5p inhibitor significantly affected the relative activity of luciferase (P < 0.05), and no significant impact was observed in mutant PTEN-MUT. Compared with miR-27a-5p + pcDNA group, transfection of miR-27a-5p mimic and pcDNA-PTEN significantly increased the expression of PTEN (P < 0.05), decreased the apoptotic rate (P < 0.05), and increased the inflammatory response (P < 0.05). miR-27a-5p induced apoptosis and inhibited the inflammatory response of pancreatic acinar cells in AP by targeting PTEN. 10.1002/jcb.28855
MiRNA-27a promotes the proliferation and invasion of human gastric cancer MGC803 cells by targeting via Wnt/β-catenin signaling pathway. Wu Fang,Li Jun,Guo Ni,Wang Xiao-Hui,Liao Yu-Qian American journal of cancer research This study aims to elucidate the effects of microRNA-27a (miR-27a) on the proliferation and invasion of gastric cancer (GC) cells by targeting SFRP1 via Wnt/β-catenin signaling pathway. GC and normal adjacent tissues were collected from 273 GC patients. Human gastric cancer cell line (MGC803) and normal human gastric mucosal cell line (GES-1) were cultured. The miR-27a mRNA expression was analyzed using Quantitative real-time polymerase chain reaction (qRT-PCR). Immunohistochemistry (IHC) test was used to detect miR-27a and SFRP1 protein expressions. After transfection, cells were divided into five groups: the negative control (NC) group, the miR-27a inhibitor group, the miR-27a mimics group, the miR-27a inhibitor + siRNA group and the miR-27a mimics + SFRP1 overexpression group. Western blotting was conducted to test SFRP1 and Wnt/β-catenin protein expression. Analysis for the target gene of miR-27a was performed using Luciferase assay. Cell proliferation, migration and invasion were determined by CCK8 and Transwell assay. The dual-luciferase reporter assay system was applied to analyze the effects of miR-27a on Wnt/β-catenin signaling pathway. In GC tissue and cell line, miR-27a protein and mRNA expressions were up-regulated, and SFRP1 protein and mRNA expressions were down-regulated. Luciferase assay indicated that miR-27a might target and regulate its expressions. When miR-27a was down-regulated, SFRP1 was up-regulated, and β-catenin, Wnt, p-β-catenin, and p-Wnt were significantly down-regulated. Compared with the NC group, the proliferation, migration and invasion of GC cells were remarkably increased in the miR-27a group, but these were declined in the miR-27a mimics + SFRP1 overexpression group. The proliferation, migration and invasion of GC cells were elevated in the miR-27a inhibitor + SFRP1 siRNA group compared with the miR-27a inhibitor group. These results showed that miR-27a was highly expressed in GC tissues and cells, and it might promote cell proliferation, migration and invasion by targeting via the activation of Wnt/β-catenin signaling pathway.
Pancreatic cancer cell-derived exosomal microRNA-27a promotes angiogenesis of human microvascular endothelial cells in pancreatic cancer via BTG2. Journal of cellular and molecular medicine Pancreatic cancer (PC) remains a primary cause of cancer-related deaths worldwide. Existing literature has highlighted the oncogenic role of microRNA-27a (miR-27a) in multiple cancers. Hence, the current study aimed to clarify the potential therapeutic role of PC cell-derived exosomal miR-27a in human microvascular endothelial cell (HMVEC) angiogenesis in PC. Initially, differentially expressed genes (DEGs) and miRs related to PC were identified by microarray analysis. Microarray analysis provided data predicting the interaction between miR-27a and BTG2 in PC, which was further verified by the elevation or depletion of miR-27a. Next, the expression of miR-27a and BTG2 in the PC tissues was quantified. HMVECs were exposed to exosomes derived from PC cell line PANC-1 to investigate the effects associated with PC cell-derived exosomes carrying miR-27a on HMVEC proliferation, invasion and angiogenesis. Finally, the effect of miR-27a on tumorigenesis and microvessel density (MVD) was analysed after xenograft tumour inoculation in nude mice. Our results revealed that miR-27a was highly expressed, while BTG2 was poorly expressed in both PC tissues and cell lines. miR-27a targeted BTG2. Moreover, miR-27a silencing inhibited PC cell proliferation and invasion, and promoted apoptosis through the elevation of BTG2. The in vitro assays revealed that PC cell-derived exosomes carrying miR-27a stimulated HMVEC proliferation, invasion and angiogenesis, while this effect was reversed in the HMVECs cultured with medium containing GW4869-treated PANC-1 cells. Furthermore, in vivo experiment revealed that miR-27a knockdown suppressed tumorigenesis and MVD. Taken together, cell-derived exosomes carrying miR-27a promotes HMVEC angiogenesis via BTG2 in PC. 10.1111/jcmm.14766
miR-27a-3p targeting RXRα promotes colorectal cancer progression by activating Wnt/β-catenin pathway. Liang Jiangtao,Tang Jianming,Shi Huijuan,Li Hui,Zhen Tiantian,Duan Jing,Kang Lili,Zhang Fenfen,Dong Yu,Han Anjia Oncotarget This study aimed to elucidate how miR-27a-3p modulates the Wnt/β-catenin signaling pathway to promote colorectal cancer (CRC) progression. Our results showed that the expression of miR-27a-3p was up-regulated in CRC and closely associated with histological differentiation, clinical stage, distant metastasis and CRC patients' survival. miR-27a-3p mimic suppressed apoptosis and promoted proliferation, migration, invasion of CRC cells and . Whereas miR-27a-3p inhibitor promoted apoptosis and suppressed proliferation, migration, invasion of CRC cells and . Furthermore, RXRα was the target gene of miR-27a-3p in CRC. miR-27a-3p expression negatively correlated with RXRα expression in CRC tissues. The underlining mechanism study showed that miR-27a-3p/RXRα/Wnt/β-catenin signaling pathway is involved in CRC progression. In conclusion, our findings first demonstrate that miR-27a-3p is a prognostic and/or potential therapeutic biomarker for CRC patients and RXRα as miR-27a-3p targeting gene plays an important role in activation of the Wnt/β-catenin pathway during CRC progression. 10.18632/oncotarget.19635
MicroRNA-27a mediates the Wnt/β-catenin pathway to affect the myocardial fibrosis in rats with chronic heart failure. Cardiovascular therapeutics OBJECTIVE:To clarify the influence of MicroRNA-27a (miR-27a)-mediated Wnt/β-catenin pathway on the myocardial fibrosis in rats with chronic heart failure (CHF). METHODS:The CHF rat models were constructed and randomly divided into four groups (Sham, Model, AntagomiR-27a, and NC antagomiR-27a groups). Echocardiography was used to test the cardiac function indexes, HE (haematoxylin-eosin) staining to observe the pathological injury of myocardium, Masson staining to analyze the collagen volume fraction (CVF), and qRT-PCR (quantitative real-time PCR) and Western blotting to detect the expressions of miR-27a and Wnt/β-catenin pathway-related proteins. Besides, cardiomyocytes were isolated and transfected with miR-27a mimic or miR-27a inhibitor to detect the expressions of Wnt/β-catenin pathway. RESULTS:The CHF rats were significantly increased in LVESD (left ventricular end systolic diameter) and LVEDD (left ventricular end diastolic diameter), and clearly reduced in FS (fractional shortening) and EF (left ventricular ejection fraction) (all P < 0.05). Moreover, LVWI (left ventricular mass index) and CVF (Collagen Volume Fraction), type I and type III collagen, and the ratio of type I/III collagen, as well as the expression of miR-27a, TGF-β1 and p-Smad2/3, β-catenin, p-GSK-3β and α-SMA were also elevated (all P < 0.05). Additionally, the CHF rats treated with AntagomiR-27a were improved in these indexes, and the expression of miR-27a and Wnt/β-catenin pathway was significantly inhibited (all P < 0.05). Furthermore, cardiomyocytes transfected with miR-27a inhibitor significantly decreased the levels of miR-27a and Wnt/β-catenin pathway (all P < 0.05). CONCLUSION:Down-regulation of miR-27a may inhibit the Wnt/β-catenin pathway to reduce the deposition of myocardial collagen, prevent myocardial fibrosis and improve cardiac function. This article is protected by copyright. All rights reserved. 10.1111/1755-5922.12468
Paclitaxel alleviated liver injury of septic mice by alleviating inflammatory response via microRNA-27a/TAB3/NF-κB signaling pathway. Yang Qiu,Zhang Dongshan,Li Ya,Li Yongquan,Li Yinpeng Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie Excessive inflammatory response and apoptosis play an important role in the sepsis-induced liver injury. Paclitaxel, a diterpene alkaloid of Taxus brevifolia, is widely used as an anti-tumor drug and shows protective effects on acute lung and kidney injury. However, whether it has a protective effect against sepsis-induced liver injury has not been reported. The objective of this study was to investigate the protective effects of paclitaxel in septic liver injury in mice and associated molecular mechanisms. Our results showed that paclitaxel treatment improved LPS-induced liver injury, as evidenced by the reduced aminotransferase activity, histological scores and apoptosis in the liver tissues. This was accompanied by the alleviating of inflammation and oxidative stress, such as decreased levels of tumor necrosis factor alpha (TNF-α), interleukin-1 (IL-6) interleukin-1β (IL-1β) and malondialdehyde (MDA) and increased levels of superoxide dismutase (SOD) and glutathione peroxidase (GSH-px) in serum and liver tissues. Subsequent microarray and qRT-PCR analysis further showed that miR-27a was significantly decreased in mice with sepsis, which was recovered by paclitaxel pretreatment. Antagomir-miR-27a suppressed the therapeutic effects of paclitaxel in mice liver injury model via promoting inflammatory response. Of note, TAB3, which participated in the activation of the NF-κB signaling pathway, was identified as a direct target of miR-27 by luciferase reporter gene assays. Then, we revealed a reverse relationship between miR-27a expression levels and TAB3 mRNA levels in liver tissues from septic mice. Furthermore, paclitaxel treatment significantly decreased the expression of NF-κB p65, but increased inhibitor of nuclear factor-κB-α (IκBα) protein levels in septic mice, suggesting the inactivation of NF-κB signaling pathway. Notably, the inhibitory effects of paclitaxel on NF-κB signaling pathway were reversed by antagomir-miR-27a. Our data indicated that paclitaxel significantly attenuated septic induced liver injury through reducing inflammatory response via miR-27a/TAB3/NF-κB signaling pathway. 10.1016/j.biopha.2017.11.003
miR-27a-3p protects against blood-brain barrier disruption and brain injury after intracerebral hemorrhage by targeting endothelial aquaporin-11. Xi Tianyang,Jin Feng,Zhu Ying,Wang Jialu,Tang Ling,Wang Yanzhe,Liebeskind David S,Scalzo Fabien,He Zhiyi The Journal of biological chemistry Previous studies have reported that miR-27a-3p is down-regulated in the serum of patients with intracerebral hemorrhage (ICH), but the implication of miR-27a-3p down-regulation in post-ICH complications remains elusive. Here we verified miR-27a-3p levels in the serum of ICH patients by real-time PCR and observed that miR-27a-3p is also significantly reduced in the serum of these patients. We then further investigated the effect of miR-27a-3p on post-ICH complications by intraventricular administration of a miR-27a-3p mimic in rats with collagenase-induced ICH. We found that the hemorrhage markedly reduced miR-27a-3p levels in the hematoma, perihematomal tissue, and serum and that intracerebroventricular administration of the miR-27a-3p mimic alleviated behavioral deficits 24 h after ICH. Moreover, ICH-induced brain edema, vascular leakage, and leukocyte infiltration were also attenuated by this mimic. Of note, miR-27a-3p mimic treatment also inhibited neuronal apoptosis and microglia activation in the perihematomal zone. We further observed that the miR-27a-3p mimic suppressed the up-regulation of aquaporin-11 (AQP11) in the perihematomal area and in rat brain microvascular endothelial cells (BMECs). Moreover, miR-27a-3p down-regulation increased BMEC monolayer permeability and impaired BMEC proliferation and migration. In conclusion, miR-27a-3p down-regulation contributes to brain edema, blood-brain barrier disruption, neuron loss, and neurological deficits following ICH. We conclude that application of exogenous miR-27a-3p may protect against post-ICH complications by targeting AQP11 in the capillary endothelial cells of the brain. 10.1074/jbc.RA118.001858
Oxidative stress-induced miR-27a targets the redox gene nuclear factor erythroid 2-related factor 2 in diabetic embryopathy. Zhao Yang,Dong Daoyin,Reece E Albert,Wang Ashley R,Yang Peixin American journal of obstetrics and gynecology BACKGROUND:Maternal diabetes induces neural tube defects, and oxidative stress is a causal factor for maternal diabetes-induced neural tube defects. The redox gene nuclear factor erythroid 2-related factor 2 is the master regulator of the cellular antioxidant system. OBJECTIVE:In this study, we aimed to determine whether maternal diabetes inhibits nuclear factor erythroid 2-related factor 2 expression and nuclear factor erythroid 2-related factor 2-controlled antioxidant genes through the redox-sensitive miR-27a. STUDY DESIGN:We used a well-established type 1 diabetic embryopathy mouse model induced by streptozotocin for our in vivo studies. Embryos at embryonic day 8.5 were harvested for analysis of nuclear factor erythroid 2-related factor 2, nuclear factor erythroid 2-related factor 2-controlled antioxidant genes, and miR-27a expression. To determine if mitigating oxidative stress inhibits the increase of miR-27a and the decrease of nuclear factor erythroid 2-related factor 2 expression, we induced diabetic embryopathy in superoxide dismutase 2 (mitochondrial-associated antioxidant gene)-overexpressing mice. This model exhibits reduced mitochondria reactive oxygen species even in the presence of hyperglycemia. To investigate the causal relationship between miR-27a and nuclear factor erythroid 2-related factor 2 in vitro, we examined C17.2 neural stem cells under normal and high-glucose conditions. RESULTS:We observed that the messenger RNA and protein levels of nuclear factor erythroid 2-related factor 2 were significantly decreased in embryos on embryonic day 8.5 from diabetic dams compared to those from nondiabetic dams. High-glucose also significantly decreased nuclear factor erythroid 2-related factor 2 expression in a dose- and time-dependent manner in cultured neural stem cells. Our data revealed that miR-27a was up-regulated in embryos on embryonic day 8.5 exposed to diabetes, and that high glucose increased miR-27a levels in a dose- and time-dependent manner in cultured neural stem cells. In addition, we found that a miR-27a inhibitor abrogated the inhibitory effect of high glucose on nuclear factor erythroid 2-related factor 2 expression, and a miR-27a mimic suppressed nuclear factor erythroid 2-related factor 2 expression in cultured neural stem cells. Furthermore, our data indicated that the nuclear factor erythroid 2-related factor 2-controlled antioxidant enzymes glutamate-cysteine ligase catalytic subunit, glutamate-cysteine ligase modifier subunit, and glutathione S-transferase A1 were down-regulated by maternal diabetes in embryos on embryonic day 8.5 and high glucose in cultured neural stem cells. Inhibiting miR-27a restored expression of glutamate-cysteine ligase catalytic subunit, glutamate-cysteine ligase modifier subunit, and glutathione S-transferase A1. Overexpressing superoxide dismutase 2 reversed the maternal diabetes-induced increase of miR-27a and suppression of nuclear factor erythroid 2-related factor 2 and nuclear factor erythroid 2-related factor 2-controlled antioxidant enzymes. CONCLUSION:Our study demonstrates that maternal diabetes-induced oxidative stress increases miR-27a, which, in turn, suppresses nuclear factor erythroid 2-related factor 2 and its responsive antioxidant enzymes, resulting in diabetic embryopathy. 10.1016/j.ajog.2017.10.040
An analysis of the global expression of microRNAs in an experimental model of physiological left ventricular hypertrophy. Martinelli Nidiane C,Cohen Carolina R,Santos Kátia G,Castro Mauro A,Biolo Andréia,Frick Luzia,Silvello Daiane,Lopes Amanda,Schneider Stéfanie,Andrades Michael E,Clausell Nadine,Matte Ursula,Rohde Luis E PloS one BACKGROUND:MicroRNAs (miRs) are a class of small non-coding RNAs that regulate gene expression. Studies of transgenic mouse models have indicated that deregulation of a single miR can induce pathological cardiac hypertrophy and cardiac failure. The roles of miRs in the genesis of physiological left ventricular hypertrophy (LVH), however, are not well understood. OBJECTIVE:To evaluate the global miR expression in an experimental model of exercise-induced LVH. METHODS:Male Balb/c mice were divided into sedentary (SED) and exercise (EXE) groups. Voluntary exercise was performed on an odometer-monitored metal wheels for 35 days. Various tests were performed after 7 and 35 days of training, including a transthoracic echocardiography, a maximal exercise test, a miR microarray (miRBase v.16) and qRT-PCR analysis. RESULTS:The ratio between the left ventricular weight and body weight was increased by 7% in the EXE group at day 7 (p<0.01) and by 11% at day 35 of training (p<0.001). After 7 days of training, the microarray identified 35 miRs that were differentially expressed between the two groups: 20 were up-regulated and 15 were down-regulated in the EXE group compared with the SED group (p = 0.01). At day 35 of training, 25 miRs were differentially expressed: 15 were up-regulated and 10 were decreased in the EXE animals compared with the SED animals (p<0.01). The qRT-PCR analysis demonstrated an increase in miR-150 levels after 35 days and a decrease in miR-26b, miR-27a and miR-143 after 7 days of voluntary exercise. CONCLUSIONS:We have identified new miRs that can modulate physiological cardiac hypertrophy, particularly miR-26b, -150, -27a and -143. Our data also indicate that previously established regulatory gene pathways involved in pathological LVH are not changed in physiological LVH. 10.1371/journal.pone.0093271
The transcardiac gradient of cardio-microRNAs in the failing heart. Marques Francine Z,Vizi Donna,Khammy Ouda,Mariani Justin A,Kaye David M European journal of heart failure AIMS:Differential microRNA expression in peripheral blood has been observed in patients with heart failure, suggesting their value as potential biomarkers and likely contributors to disease mechanisms. In the present study, we aimed to evaluate the transcardiac gradient of 84 cardio-microRNAs in healthy and failing hearts to determine which microRNAs are released or absorbed by the myocardium in heart failure. METHODS AND RESULTS:Eight healthy volunteers and nine patients with congestive heart failure were included. Arterial and coronary sinus blood samples were collected, and microRNAs were extracted. The expression of microRNAs was analysed using real-time PCR by the miScript miRNA PCR Array Human Cardiovascular Disease. In coronary sinus samples, the microRNAs miR-16-5p, miR-27a-3p, miR-27b-3p, miR-29b-3p, miR-29c-3p, miR-30e-5p, miR-92a-3p, miR-125b-5p, miR-140-5p, miR-195-5p, miR-424-5p, and miR-451a were significantly down-regulated, and let-7a-5p, let-7c-5p, let-7e-5p, miR-23b-3p, miR-107, miR-155-5p, miR-181a-5p, miR-181b-5p and miR-320a were up-regulated in heart failure. Left ventricular filling pressure was negatively correlated with miR-195, miR-16, miR-29b-3p, miR-29c-3p, miR-451a, and miR-92a-3p. The failing heart released let-7b-5p, let-7c-5p, let-7e-5p, miR-122-5p, and miR-21-5p, and absorbed miR-16-5p, miR-17-5p, miR-27a-3p, miR-30a-5p, miR-30d-5p, miR-30e-5p, miR-130a-3p, miR-140-5p, miR-199a-5p, and miR-451a. In silico analyses suggest that the transcardiac gradient of microRNAs in heart failure may target pathways related to heart disease. CONCLUSION:We determined the transcardiac gradient of cardio-microRNAs in failing hearts, which supports the use of these microRNAs as potential biomarkers. The microRNAs described here may have a role in the pathophysiology of heart failure as they might be involved in pathways related to disease progression, including fibrosis. 10.1002/ejhf.517
A phenotypic screen to identify hypertrophy-modulating microRNAs in primary cardiomyocytes. Jentzsch Claudia,Leierseder Simon,Loyer Xavier,Flohrschütz Isabell,Sassi Yassine,Hartmann Dorothee,Thum Thomas,Laggerbauer Bernhard,Engelhardt Stefan Journal of molecular and cellular cardiology MicroRNAs (miRNAs) are small non-coding RNAs that control expression of complementary target mRNAs. A growing number of miRNAs has been implicated in the pathogenesis of cardiac diseases, mostly based not on functional data, but on the observation that they are dysregulated in diseased myocardium. Consequently, our knowledge regarding a potential cardiac role of the majority of miRNAs is limited. Here, we report the development of an assay format that allows the simultaneous analysis of several hundred molecules with regard to their phenotypic effect on primary rat cardiomyocytes. Using automated microscopy and an edge detection algorithm, this assay achieved high reproducibility and a robust assessment of cardiomyocyte size as a key parameter. Screening a library of synthetic miRNAs revealed several miRNAs previously not recognized as pro- or anti-hypertrophic. Out of these, we selected nine miRNAs and confirmed the pro-hypertrophic potential of miR-22, miR-30c, miR-30d, miR-212, miR-365 and the anti-hypertrophic potential of miR-27a, miR-27b and miR-133a. Quantitative analysis of the expression level of pro-hypertrophic miRNAs in primary cardiomyocytes indicated a rather low level of correlation of the phenotypic effects of individual miRNAs and their expression level. This assay allows the automated determination of cell size in primary cardiomyocytes and permitted the identification of a set of miRNAs capable of regulating cardiomyocyte hypertrophy. Elucidating their mechanism of action should provide insight into mechanisms underlying the cardiomyocyte hypertrophic response. This article is part of a Special Issue entitled 'Possible Editorial'. 10.1016/j.yjmcc.2011.07.010
A panel of 4 microRNAs facilitates the prediction of left ventricular contractility after acute myocardial infarction. Devaux Yvan,Vausort Melanie,McCann Gerry P,Kelly Dominic,Collignon Olivier,Ng Leong L,Wagner Daniel R,Squire Iain B PloS one BACKGROUND:Prediction of clinical outcome after acute myocardial infarction (AMI) is challenging and would benefit from new biomarkers. We investigated the prognostic value of 4 circulating microRNAs (miRNAs) after AMI. METHODS:We enrolled 150 patients after AMI. Blood samples were obtained at discharge for determination of N-terminal pro-brain natriuretic peptide (Nt-proBNP) and levels of miR-16, miR-27a, miR-101 and miR-150. Patients were assessed by echocardiography at 6 months follow-up and the wall motion index score (WMIS) was used as an indicator of left ventricular (LV) contractility. We assessed the added predictive value of miRNAs against a multi-parameter clinical model including Nt-proBNP. RESULTS:Patients with anterior AMI and elevated Nt-proBNP levels at discharge from the hospital were at high risk of subsequent impaired LV contractility (follow-up WMIS>1.2, n = 71). A combination of the 4 miRNAs (miR-16/27a/101/150) improved the prediction of LV contractility based on clinical variables (P = 0.005). Patients with low levels of miR-150 (odds ratio [95% confidence interval] 0.08 [0.01-0.48]) or miR-101 (0.19 [0.04-0.97]) and elevated levels of miR-16 (15.9 [2.63-95.91]) or miR-27a (4.18 [1.36-12.83]) were at high risk of impaired LV contractility. The 4 miRNA panel reclassified a significant proportion of patients with a net reclassification improvement of 66% (P = 0.00005) and an integrated discrimination improvement of 0.08 (P = 0.001). CONCLUSION:Our results indicate that panels of miRNAs may aid in prognostication of outcome after AMI. 10.1371/journal.pone.0070644
Aerobic exercise training promotes physiological cardiac remodeling involving a set of microRNAs. Fernandes Tiago,Baraúna Valério G,Negrão Carlos E,Phillips M Ian,Oliveira Edilamar M American journal of physiology. Heart and circulatory physiology Left ventricular (LV) hypertrophy is an important physiological compensatory mechanism in response to chronic increase in hemodynamic overload. There are two different forms of LV hypertrophy, one physiological and another pathological. Aerobic exercise induces beneficial physiological LV remodeling. The molecular/cellular mechanisms for this effect are not totally known, and here we review various mechanisms including the role of microRNA (miRNA). Studies in the heart, have identified antihypertrophic miRNA-1, -133, -26, -9, -98, -29, -378, and -145 and prohypertrophic miRNA-143, -103, -130a, -146a, -21, -210, -221, -222, -27a/b, -199a/b, -208, -195, -499, -34a/b/c, -497, -23a, and -15a/b. Four miRNAs are recognized as cardiac-specific: miRNA-1, -133a/b, -208a/b, and -499 and called myomiRs. In our studies we have shown that miRNAs respond to swimming aerobic exercise by 1) decreasing cardiac fibrosis through miRNA-29 increasing and inhibiting collagen, 2) increasing angiogenesis through miRNA-126 by inhibiting negative regulators of the VEGF pathway, and 3) modulating the renin-angiotensin system through the miRNAs-27a/b and -143. Exercise training also increases cardiomyocyte growth and survival by swimming-regulated miRNA-1, -21, -27a/b, -29a/c, -30e, -99b, -100, -124, -126, -133a/b, -143, -144, -145, -208a, and -222 and running-regulated miRNA-1, -26, -27a, -133, -143, -150, and -222, which influence genes associated with the heart remodeling and angiogenesis. We conclude that there is a potential role of these miRNAs in promoting cardioprotective effects on physiological growth. 10.1152/ajpheart.00899.2014
Circulating miR-29a, among other up-regulated microRNAs, is the only biomarker for both hypertrophy and fibrosis in patients with hypertrophic cardiomyopathy. Roncarati Roberta,Viviani Anselmi Chiara,Losi Maria Angela,Papa Laura,Cavarretta Elena,Da Costa Martins Paula,Contaldi Carla,Saccani Jotti Gloria,Franzone Anna,Galastri Laura,Latronico Michael V G,Imbriaco Massimo,Esposito Giovanni,De Windt Leon,Betocchi Sandro,Condorelli Gianluigi Journal of the American College of Cardiology OBJECTIVES:The purpose of this paper was to determine whether microRNAs (miRNAs) involved in myocardial remodeling were differentially expressed in the blood of hypertrophic cardiomyopathy (HCM) patients, and whether circulating miRNAs correlated with the degree of left ventricular hypertrophy and fibrosis. BACKGROUND:miRNAs-small, noncoding ribonucleic acids (RNAs) that regulate gene expression by inhibiting RNA translation-modulate cellular function. Myocardial miRNAs modulate processes such as cardiomyocyte (CM) hypertrophy, excitation-contraction coupling, and apoptosis; non-CM-specific miRNAs regulate myocardial vascularization and fibrosis. Recently, the possibility that circulating miRNAs may be biomarkers of cardiovascular disease has been raised. METHODS:Forty-one HCM patients were characterized with conventional transthoracic echocardiography and cardiac magnetic resonance. Peripheral plasma levels of 21 miRNAs were assessed by quantitative real-time polymerase chain reaction and were compared with levels in a control group of 41 age- and sex-matched blood donors. RESULTS:Twelve miRNAs (miR-27a, -199a-5p, -26a, -145, -133a, -143, -199a-3p, -126-3p, -29a, -155, -30a, and -21) were significantly increased in HCM plasma. However, only 3 miRNAs (miR-199a-5p, -27a, and -29a) correlated with hypertrophy; more importantly, only miR-29a correlated also with fibrosis. CONCLUSIONS:Our data suggest that cardiac remodeling associated with HCM determines a significant release of miRNAs into the bloodstream: the circulating levels of both cardiac- and non-cardiac-specific miRNAs are significantly increased in the plasma of HCM patients. However, correlation with left ventricular hypertrophy parameters holds true for only a few miRNAs (i.e., miR-199a-5p, -27a, and -29a), whereas only miR-29a is significantly associated with both hypertrophy and fibrosis, identifying it as a potential biomarker for myocardial remodeling assessment in HCM. 10.1016/j.jacc.2013.09.041
Renin-sensitive microRNAs correlate with atherosclerosis plaque progression. Deiuliis J,Mihai G,Zhang J,Taslim C,Varghese J J,Maiseyeu A,Huang K,Rajagopalan S Journal of human hypertension Recent trials with inhibition of the renin-angiotensin-aldosterone system (RAAS) in patients with established atherosclerosis have been equivocal. MicroRNAs (miRs) are known to affect multiple pathways relevant to atherosclerosis, including RAAS. We postulated that the use of a direct renin antagonist would result in differential regulation of miRs. We examined monocyte miR expression before and after treatment with renin antagonist, Aliskiren, in patients with established cardiovascular disease as part of a prospective, single-center, randomized, double-blind and placebo-controlled clinical trial (NCT01417104). After screening, patients (mean age 62±3 years) were randomized to placebo or Aliskiren. Three-dimensional dark-blood magnetic resonance imaging assessment of atherosclerosis in the thoracic and abdominal aorta was conducted at baseline and at study completion (19-36 weeks). MiR expression arrays were performed on RNA from peripheral blood mononuclear cells collected at baseline and 12 weeks following randomization to placebo or Aliskiren and showed that hsa-miR-106b-5p, 27a-3p and 18b-5p were significantly downregulated with Aliskiren. Baseline expression of these miRs positively correlated with normalized total wall volume in subjects taking Aliskiren (miR-106b, R=0.62; miR-27a, R=0.63; miR-18b, R=0.77; P<0.05). Hsa-miR-106b-5p, 27a-3p and 18b-5p may represent pathway-specific adaptations to renin inhibition relevant to atherosclerosis. 10.1038/jhh.2013.97
MicroRNA-27a regulates cardiomyocytic apoptosis during cardioplegia-induced cardiac arrest by targeting interleukin 10-related pathways. Yeh Chi-Hsiao,Chen Tzu-Ping,Wang Yao-Chang,Lin Yu-Min,Fang Shu-Wen Shock (Augusta, Ga.) In this study, experiments were designed to determine whether microRNAs (miRNAs) play a role in the regulation or modulation of cardiomyocytic reactions under cardioplegia-induced cardiac arrest during cardiopulmonary bypass. MicroRNAs play powerful and unexpected roles in numerous cardiovascular diseases. MicroRNA-based therapeutics may provide a unique opportunity to translate this knowledge into the clinical setting. Sprague-Dawley rats (10 per group) were randomly divided into three groups: control, perfusion, and arrest groups. In the perfusion group, isolated hearts were perfused with oxygenated physiologic buffered solution for 3 h using a Langendorff apparatus. In the arrest group, cold crystalloid cardioplegia solution was used to induce and maintain cardiac arrest for 1 h; hearts were reperfused for 2 h with warm oxygenated phosphate-buffered saline solution. Cardiac miRNAs and protein expression patterns were detected using miRNA arrays and two-dimensional fluorescence difference gel electrophoresis followed by matrix-assisted laser desorption/ionization-time of flight mass spectrometry. Of 103 different miRNAs and 222 different proteins expressed in the three groups, miRNA-27a was the one considered to be related to the regulation of cardiomyocyte apoptosis by targeting the interleukin 10 pathway. Transfection of H9c2 cardiomyocytes with pre-miRNA-27a, which significantly decreased the mRNA and protein levels of interleukin 10 and increased expression of nuclear factor κB and its downstream cytokines during hypoxia/reperfusion injury, could activate caspase 3 and apoptosis. Our study demonstrated the altered expression of miRNAs in cardiomyocytes during cardioplegia-induced cardiac arrest. The involvement of miRNAs in cardiomyocytic apoptosis adds another level of complexity to gene regulation, which could open up novel avenues for cardiac protection strategies during cardiac surgery. 10.1097/SHK.0b013e318271f944
MicroRNA-23a/b and microRNA-27a/b suppress Apaf-1 protein and alleviate hypoxia-induced neuronal apoptosis. Chen Q,Xu J,Li L,Li H,Mao S,Zhang F,Zen K,Zhang C-Y,Zhang Q Cell death & disease Expression of apoptotic protease activating factor-1 (Apaf-1) gradually decreases during brain development, and this decrease is likely responsible for the decreased sensitivity of brain tissue to apoptosis. However, the mechanism by which Apaf-1 expression is decreased remains elusive. In the present study, we found that four microRNAs (miR-23a/b and miR-27a/b) of miR-23a-27a-24 and miR-23b-27b-24 clusters play key roles in modulating the expression of Apaf-1. First, we found that miR-23a/b and miR-27a/b suppressed the expression of Apaf-1 in vitro. Interestingly, the expression of the miR-23-27-24 clusters in the mouse cortex gradually increased in a manner that was inversely correlated with the pattern of Apaf-1 expression. Second, hypoxic injuries during fetal distress caused reduced expression of the miR-23b and miR-27b that was inversely correlated with an elevation of Apaf-1 expression during neuronal apoptosis. Third, we made neuronal-specific transgenic mice and found that overexpressing the miR-23b and miR-27b in mouse neurons inhibited the neuronal apoptosis induced by intrauterine hypoxia. In conclusion, our results demonstrate, in central neural system, that miR-23a/b and miR-27a/b are endogenous inhibitory factors of Apaf-1 expression and regulate the sensitivity of neurons to apoptosis. Our findings may also have implications for the potential target role of microRNAs in the treatment of neuronal apoptosis-related diseases. 10.1038/cddis.2014.92
Association of miR-196a2, miR-27a, and miR-499 polymorphisms with isolated congenital heart disease in a Chinese population. Yu K,Ji Y,Wang H,Xuan Q K,Li B B,Xiao J J,Sun W,Kong X Q Genetics and molecular research : GMR We hypothesized that single nucleotide polymorphisms (SNPs) in certain microRNAs contribute to congenital heart disease (CHD) phenotypes. Five hundred and seventy-three subjects were enrolled in this study. DNA extracted from peripheral blood cells was used for SNP genotyping of miR-196a2 (rs11614913), miR-27a (rs11671784, rs895819), and miR-499 (rs3746444). Allele and genotype association analyses were performed to evaluate the correlation between certain microRNA SNPs and three phenotypes of isolated CHD: atrial septal defect (ASD), ventricular septal defect (VSD), and patent ductus arteriosus (PDA). All the participants carried a homozygous CC variant of miR-27a (rs11671784). The homozygous CC variant of miR-196a2 (rs11614913, T>C) was negatively associated with ASD compared with the wild-type TT variant (OR = 0.379, 95%CI = 0.209-0.686, P = 0.001). The miR-196a2 C allele was negatively associated with ASD compared with the T allele (OR = 0.646, 95%CI = 0.491-0.849, P = 0.002). The statistically significant results were further confirmed by dominant and recessive model assays. SNPs of miR-27a (rs895819, T>C) and miR-499 (rs3746444, A>G) showed diverse association with ASD, VSD, or PDA, but the differences were not statistically significant. The rs11614913 (T>C) SNP of miR-196a2 is associated with ASD, and the homozygous CC variant and the C allele are protective factors associated with ASD. The homozygous CC variant and the C allele of the rs11614913 (T>C) SNP of miR-196a2 are associated with a significantly reduced risk of ASD. 10.4238/gmr15048929
miR-23a, miR-24 and miR-27a protect differentiating ESCs from BMP4-induced apoptosis. Musto A,Navarra A,Vocca A,Gargiulo A,Minopoli G,Romano S,Romano M F,Russo T,Parisi S Cell death and differentiation Numerous studies have indicated that BMP4 signaling is involved in the regulation of the early steps of development. In mouse embryonic stem cells (ESCs), BMP4 is crucial to sustain pluripotency and blocks differentiation towards neural fate. Here, through a systematic analysis of miRNAs in ESCs, we establish that BMP4 signaling regulates miR-23a, 27a and 24-2, through the recruitment of phospho-Smads at the promoter of the gene encoding this miRNA cluster. Suppression of miR-23a/b, 27a/b and 24 does not affect self-renewal or pluripotency, but induces an evident change of ESC differentiation, with a significant increase of the cells undergoing apoptosis after the transition from ESCs to epiblast stem cells (EpiSCs). BMP4 has been previously reported to cause apoptosis during ESC differentiation. By blocking BMP4 signaling, we completely prevent the apoptosis induced by suppression of the miRs. This suggests that the effects of miR suppression are the result of enhanced BMP4 signaling. This hypothesis is further supported by the observation that Smad5, the transcription factor downstream of the BMP4 receptor, is targeted by the miRNAs of the 23a and 23b clusters. Altogether, our results highlight the existence of a regulatory loop, involving Smad5 and the miR-23a clusters, that modulates the apoptotic response of ESCs to BMP4. 10.1038/cdd.2014.198
MicroRNA-27a regulates beta cardiac myosin heavy chain gene expression by targeting thyroid hormone receptor beta1 in neonatal rat ventricular myocytes. Nishi Hitoo,Ono Koh,Horie Takahiro,Nagao Kazuya,Kinoshita Minako,Kuwabara Yasuhide,Watanabe Shin,Takaya Tomohide,Tamaki Yodo,Takanabe-Mori Rieko,Wada Hiromichi,Hasegawa Koji,Iwanaga Yoshitaka,Kawamura Teruhisa,Kita Toru,Kimura Takeshi Molecular and cellular biology MicroRNAs (miRNAs), small noncoding RNAs, are negative regulators of gene expression and play important roles in gene regulation in the heart. To examine the role of miRNAs in the expression of the two isoforms of the cardiac myosin heavy chain (MHC) gene, α- and β-MHC, which regulate cardiac contractility, endogenous miRNAs were downregulated in neonatal rat ventricular myocytes (NRVMs) using lentivirus-mediated small interfering RNA (siRNA) against Dicer, an essential enzyme for miRNA biosynthesis, and MHC expression levels were examined. As a result, Dicer siRNA could downregulate endogenous miRNAs simultaneously and the β-MHC gene but not α-MHC, which implied that specific miRNAs could upregulate the β-MHC gene. Among 19 selected miRNAs, miR-27a was found to most strongly upregulate the β-MHC gene but not α-MHC. Moreover, β-MHC protein was downregulated by silencing of endogenous miR-27a. Through a bioinformatics screening using TargetScan, we identified thyroid hormone receptor β1 (TRβ1), which negatively regulates β-MHC transcription, as a target of miR-27a. Moreover, miR-27a was demonstrated to modulate β-MHC gene regulation via thyroid hormone signaling and to be upregulated during the differentiation of mouse embryonic stem (ES) cells or in hypertrophic hearts in association with β-MHC gene upregulation. These findings suggested that miR-27a regulates β-MHC gene expression by targeting TRβ1 in cardiomyocytes. 10.1128/MCB.00581-10
Downregulation of miR-23a and miR-27a following experimental traumatic brain injury induces neuronal cell death through activation of proapoptotic Bcl-2 proteins. Sabirzhanov Boris,Zhao Zaorui,Stoica Bogdan A,Loane David J,Wu Junfang,Borroto Carlos,Dorsey Susan G,Faden Alan I The Journal of neuroscience : the official journal of the Society for Neuroscience MicroRNAs (miRs) are small noncoding RNAs that negatively regulate gene expression at the post-transcriptional level. To identify miRs that may regulate neuronal cell death after experimental traumatic brain injury (TBI), we profiled miR expression changes during the first several days after controlled cortical impact (CCI) in mice. miR-23a and miR-27a were rapidly downregulated in the injured cortex in the first hour after TBI. These changes coincided with increased expression of the proapoptotic Bcl-2 family members Noxa, Puma, and Bax. In an etoposide-induced in vitro model of apoptosis in primary cortical neurons, miR-23a and miR-27a were markedly downregulated as early as 1 h after exposure, before the upregulation of proapoptotic Bcl-2 family molecules. Administration of miR-23a and miR-27a mimics attenuated etoposide-induced changes in Noxa, Puma, and Bax, reduced downstream markers of caspase-dependent (cytochrome c release and caspase activation) and caspase-independent (apoptosis-inducing factor release) pathways, and limited neuronal cell death. In contrast, miRs hairpin inhibitors enhanced etoposide-induced neuronal apoptosis and caspase activation. Importantly, administration of miR-23a and miR-27a mimics significantly reduced activation of Puma, Noxa, and Bax as well as attenuated markers of caspase-dependent and -independent apoptosis after TBI. Furthermore, miR-23a and miR-27a mimics significantly attenuated cortical lesion volume and neuronal cell loss in the hippocampus after TBI. These findings indicate that post-traumatic decreases in miR-23a and miR-27a contribute to neuronal cell death after TBI by upregulating proapoptotic Bcl-2 family members, thus providing a novel therapeutic target. 10.1523/JNEUROSCI.1260-14.2014
MiR-27a ameliorates inflammatory damage to the blood-spinal cord barrier after spinal cord ischemia: reperfusion injury in rats by downregulating TICAM-2 of the TLR4 signaling pathway. Li Xiao-Qian,Lv Huang-Wei,Wang Zhi-Lin,Tan Wen-Fei,Fang Bo,Ma Hong Journal of neuroinflammation BACKGROUND:Spinal cord ischemia reperfusion (IR) injury causes inflammation and subsequently increases blood-spinal cord barrier leakage and Toll-like receptor 4 (TLR4) pathway activation. MicroRNAs (miRs) effectively regulate numerous target mRNAs during ischemia. However, their roles during IR injury are poorly understood. We investigated miRs involvement, particularly miR-27a, in TLR4 pathway-mediated inflammatory responses after IR. METHOD:We used a genomics approach to examine changed miRs of rats that had undergone 14 minutes of ischemia, followed by 24 or 72 hours of reperfusion. Quantitative RT-PCR was used to identify and confirm the miRs involved in regulating TLR4 pathway activation. We scanned miR databases for potential miR targets and confirmed these targets by quantitative RT-PCR. The miR mimic and anti-miR oligonucleotides (AMOs) were intrathecally injected at 12-hour intervals beginning three days before the ischemia. The effects of miRs on the TLR4 pathway and downstream cytokines were analyzed by PCR, western blotting, and ELISA. Double immunofluorescence staining was perfumed to determine the relationship between the targets and TLR4. Blood-spinal cord barrier (BSCB) permeability was examined using Evans blue (EB) dye. RESULTS:A microarray analysis revealed that at 24 hours post-injury, three miRs were upregulated (>2.0 fold) and 15 miRs were downregulated (<0.5 fold), and at 72 hours, four miRs were upregulated and 14 were downregulated compared to their levels in sham-operated controls. We focused on miR-27a, which is predicted to contain sequences complementary to the 3'-untranslated region (UTR) of Toll-like receptor adaptor molecule 2 (TICAM-2). Double immunostaining indicated that TLR4 activation correlated with changes in TICAM-2 expression. Compared to the rats in the IR and negative control groups, intrathecal infusion of the miR-27a mimic attenuated IR-induced TLR4 activation and inflammatory damage to the BSCB, which was shown as decreased EB extravasation and lower levels of nuclear factor kappa-B (NF-κB) and lnterleukin (IL)-1β at 24 and 72 hours after reperfusion, whereas pretreatment with miR-27a AMO aggravated these injuries. CONCLUSIONS:We present the first evidence that miRs play an important role in spinal cord IR injury. We identified TICAM-2 as a novel target of miR-27a. miR-27a upregulation attenuates IR-induced inflammatory damage to the BSCB by negatively regulating TICAM-2 of the TLR4 signaling pathway and inhibiting the NF-κB/IL-1β pathway. These results provide new therapeutic targets for IR injury treatment. 10.1186/s12974-015-0246-3
MiR-27a regulates apoptosis in nucleus pulposus cells by targeting PI3K. Liu Gang,Cao Peng,Chen Huajiang,Yuan Wen,Wang Jianxi,Tang Xianye PloS one The precise role of apoptosis in the pathogenesis of intervertebral disc degeneration (IDD) remains to be elucidated. We analyzed degenerative nucleus pulposus (NP) cells and found that the expression of miR-27a was increased. The overexpression of miR-27a was further verified using real-time RT-PCR. Bioinformatics target prediction identified phosphoinositide-3 kinases (PI3K) as putative targets of miR-27a. Furthermore, miR-27a inhibited PI3K expression by directly targeting their 3'-UTRs, and this inhibition was abolished by mutation of the miR-27a binding sites. Various cellular processes including cell growth, proliferation, migration and adhesion are regulated by activation of the PI3K/AKT signaling pathway, and nucleus pulposus cells are known to strongly express the phosphorylated survival protein AKT. Our results identify PI3K as a novel target of miR-27a. Upregulation of miR-27a thus targets PI3K, initiating apoptosis of nucleus pulposus cells. This present study revealed that downregulated miR-27a might develop a novel intervention for IDD treatment through the prevention of apoptosis in Nucleus pulposus Cells. 10.1371/journal.pone.0075251
miR-27a-5p Attenuates Hypoxia-induced Rat Cardiomyocyte Injury by Inhibiting . Zhang Jinwei,Qiu Wanling,Ma Jideng,Wang Yujie,Hu Zihui,Long Keren,Wang Xun,Jin Long,Tang Qianzi,Tang Guoqing,Zhu Li,Li Xuewei,Shuai Surong,Li Mingzhou International journal of molecular sciences Acute myocardial infarction (AMI) is an ischemic heart disease with high mortality worldwide. AMI triggers a hypoxic microenvironment and induces extensive myocardial injury, including autophagy and apoptosis. MiRNAs, which are a class of posttranscriptional regulators, have been shown to be involved in the development of ischemic heart diseases. We have previously reported that hypoxia significantly alters the miRNA transcriptome in rat cardiomyoblast cells (H9c2), including miR-27a-5p. In the present study, we further investigated the potential function of miR-27a-5p in the cardiomyocyte response to hypoxia, and showed that miR-27a-5p expression was downregulated in the H9c2 cells at different hypoxia-exposed timepoints and the myocardium of a rat AMI model. Follow-up experiments revealed that miR-27a-5p attenuated hypoxia-induced cardiomyocyte injury by regulating autophagy and apoptosis via , which partly elucidated the anti-hypoxic injury effects of miR-27a-5p. Taken together, this study shows that miR-27a-5p has a cardioprotective effect on hypoxia-induced H9c2 cell injury, suggesting it may be a novel target for the treatment of hypoxia-related heart diseases. 10.3390/ijms20102418
miRNA‑27a promotes the proliferation and inhibits apoptosis of human pancreatic cancer cells by Wnt/β-catenin pathway. Cui Zhigang,Liu Geng,Kong Di Oncology reports A specific expression of miRNA in pancreatic cancer renders it the novel diagnostic marker of pancreatic cancer. Therefore, we investigated how the anticancer effect of miRNA‑27a suppressed cell growth and induced apoptosis of human pancreatic cancer cells. We upregulated miRNA‑27a expression in PANC-1 cells using miRNA‑27a mimic, which demonstrated that induction of cell growth and suppression of apoptosis of human pancreatic cancer cells were observed. However, anti‑miRNA‑27a inhibited cell growth and apoptosis in pancreatic cancer cells. The downregulation of miRNA‑27a suppressed Wnt/β-catenin pathway. The inhibition of Wnt/β-catenin pathway increased the anticancer effects of anti‑miRNA‑27a on human pancreatic cancer cells. Taken together, miRNA‑27a promotes the proliferation and inhibits apoptosis of human pancreatic cancer cells via Wnt/β-catenin pathway. 10.3892/or.2017.6120
HIF-1α-induced miR-23a∼27a∼24 cluster promotes colorectal cancer progression via reprogramming metabolism. Jin Fangfang,Yang Rong,Wei Yao,Wang Dong,Zhu Yanan,Wang Xiaohua,Lu Yousheng,Wang Yanbo,Zen Ke,Li Limin Cancer letters Tumor cells switch metabolic profile from oxidative phosphorylation to glycolysis in a hypoxic environment for survival and proliferation. The mechanisms governing this metabolic switch, however, remain incompletely understood. Here, we show that three miRNAs in the miR-23a∼27a∼24 cluster, miR-23a, miR-27a and miR-24, are the most upregulated miRNA cluster in colorectal cancer (CRC) under hypoxia. Gain- and loss-of-function assays, a human glucose metabolism array and gene pathway analyses confirm that HIF-1α-induced miR-23a∼27a∼24 cluster collectively regulate glucose metabolic network through regulating various metabolic pathways and targeting multiple tricarboxylic acid cycle (TCA)-related genes. In specific, miR-24/VHL/HIF-1α in CRC form a double-negative feedback loop, which in turn, promotes the cellular transition to the 'high HIF-1α/miR-24 and low VHL' state and facilitates cell survival. Our findings reveal that the miR-23a∼27a∼24 cluster is critical regulator switching CRC metabolism from oxidative phosphorylation to glycolysis, and controlling their expression can suppress colorectal cancer progression. 10.1016/j.canlet.2018.10.025
Suppression of microRNA‑27a protects against liver ischemia/reperfusion injury by targeting PPARγ and inhibiting endoplasmic reticulum stress. Chi Xiaobin,Jiang Yi,Chen Yongbiao,Yang Fang,Cai Qiucheng,Pan Fan,Lv Lizhi,Zhang Xiaojin Molecular medicine reports Liver ischemia‑reperfusion (I/R) injury is an important clinical issue related to liver transplantation. Recent studies suggest that microRNAs are implicated in various biological and pathological processes, including liver I/R injury. This study aimed to investigate the role and potential mechanism of miR‑27a during liver I/R injury. A liver I/R model was induced via 60 min of ischemia and reperfusion for 6 h in rats. Cells were transfected with miR‑27a mimics or the miR‑27a inhibitor to examine the effect of miR‑27a on liver I/R. Apoptotic cells were detected by flow cytometry and TUNEL staining. The expression of miR‑27a was measured by real‑time PCR. The expression of peroxisome proliferator‑activated receptor γ (PPARγ); gastrin‑releasing peptide 78 (GRP78) and C/EBP homologous protein (CHOP) were detected by western blot analysis. The results showed that miR‑27a was significantly upregulated during I/R injury in vivo and in vitro. In addition, miR‑27a inhibitors attenuated hypoxia/reoxygenation (H/R)‑induced oxidative stress, endoplasmic reticulum stress (ERS) and apoptosis in AML12 cells. By contrast, miR‑27a mimics promoted hypoxia/reoxygenation‑induced ERS, and apoptosis. Furthermore, PPARγ was identified as a target gene of miR‑27a using bioinformatic analysis and a dual‑luciferase reporter assay. Knockdown of PPARγ significantly abrogated the inhibitory effect of miR‑27a inhibitors on the ERS pathway. Moreover, the miR‑27a antagomir attenuated liver I/R injury in rats, a finding manifested by reduced ALT/AST, hepatocyte apoptosis, oxidative stress and inhibition of the ERS pathway. Taken together, these findings demonstrate that suppression of miR‑27a protects against liver I/R injury by targeting PPARγ and by inhibiting the ERS pathway. 10.3892/mmr.2019.10645
Arbutin protects HK-2 cells against high glucose-induced apoptosis and autophagy by up-regulating microRNA-27a. Lv Lina,Zhang Jing,Tian Fengqun,Li Xia,Li Dandan,Yu Xiulian Artificial cells, nanomedicine, and biotechnology Arbutin (ARB) has been widely used in skin pigmentation disorders. Nevertheless, the involvements of ARB in diabetic nephropathy (DN) are still unknown. We investigated the functions of ARB in high glucose (HG)-induced cell apoptosis and autophagy in HK-2 cells. Cell viability was examined through CCK-8 in HK-2 cells after disposal with 45 mM glucose and ARB (10-50 μM). Flow cytometry and western blot tested cell apoptosis and the related protein levels in HK-2 cells after 45 mM glucose and 50 μM ARB administration. RT-qPCR delved microRNA (miR)-27a expression in HG and ARB co-treated HK-2 cells. Effect of miR-27a on ARB affected cell apoptosis and autophagy was investigated after miR-27a inhibitor transfection. JNK and mTOR pathways were finally assessed by western blot. ARB alleviated HG-induced cell apoptosis, autophagy and regulated the related protein levels in HK-2 cells. MiR-27a expression was reduced in HG-treated cells, but was accelerated in HG and ARB co-treated HK-2 cells with the increased concentration. Inhibition of miR-27a apparently abolished the outcomes of ARB in HG-induced HK-2 cells apoptosis and autophagy. Besides, ARB blocked JNK and mTOR pathways by regulating miR-27a. The findings demonstrated that ARB alleviated apoptosis and autophagy in HG-treated HK-2 cells by regulating miR-27a/JNK/mTOR axis. 10.1080/21691401.2019.1640231
Inhibition of miR-27a suppresses the inflammatory response via the p38/MAPK pathway in intervertebral disc cells. Cao Zhenguo,Chen Liang Experimental and therapeutic medicine The current study aimed to investigate the role of miR-27a in intervertebral disc degeneration (IDD) and to examine the underlying mechanisms. Quantitative polymerase chain reaction (qPCR) was performed to detect the expression level of miR-27a in the nucleus pulposus (NP) tissues of patients with IDD, and the results revealed an increasing expression of miR-27a in IDD compared with the control. To further investigate the role of miR-27a in IDD, a stable human NP cell line with low miR-27a expression was generated by transfecting cells with a lentiviral antigomiR-27a inhibitor. In addition, a human NP cell inflammation model was established by lipopolysaccharide (LPS; 10 µM) stimulation. The miR-27a expression in NP cells was determined by qPCR, while the expression of its target proteins; p-p38 and nuclear factor (NF-κB) was measured by western blot analysis. Furthermore, the mRNA and protein expression levels of proinflammatory factors, including interleukin (IL)-1β, IL-6 and tumor necrosis factor-α (TNF-α), were also evaluated by qPCR and ELISA, respectively. The current results confirmed that miR-27a was significantly upregulated in IDD. , downregulation of miR-27a in LPS-stimulated NP cells by transfection with the miR-27a inhibitor resulted in suppression of p-p38 and NF-κB expression levels. Furthermore, the production of the proinflammatory factors IL-1β, IL-6 and TNF-α was significantly reduced in LPS-stimulated NP cells with downregulated miR-27a. In conclusion, miR-27a may function as a promoter in IDD development, while inhibition of miR-27a may suppress proinflammatory factors released by intervertebral disc cells by regulating the p38/mitogen-activated protein kinase (MAPK) signaling pathway. 10.3892/etm.2017.5053
MiR-27a promotes insulin resistance and mediates glucose metabolism by targeting PPAR-γ-mediated PI3K/AKT signaling. Chen Tianbao,Zhang Yi,Liu Yilan,Zhu Dexiao,Yu Jing,Li Guoqian,Sun Zhichun,Wang Wanru,Jiang Hongwei,Hong Zhenzhen Aging This study aimed to establish a high-fat diet (HFD)-fed obese mouse model and a cell culture model of insulin resistance (IR) in mature 3T3-L1 adipocytes. A dual-luciferase reporter assay (DLRA) was confirmed interaction between miR-27a and the 3'-untranslated region (UTR) of Peroxisome proliferator-activated receptor (PPAR)-γ. The inhibition of PPAR-γ expression by microRNA (miR)-27a in IR cells at both the protein and mRNA levels was confirmed by a mechanistic investigation. Moreover, the 3'-UTR of PPAR-γ was found to be a direct target of miR-27a, based on the DLRA. Furthermore, antagomiR-27a upregulated the activation of PI3K/Akt signaling and glucose transporter type 4 (GLUT4) expression at the protein and mRNA levels. Additionally, the PPAR inhibitor T0070907 repressed the insulin sensitivity upregulated by antagomiR-27a, which was accompanied by the inhibition of PPAR-γ expression and increased levels of AKT phosphorylation and GLUT4. The PI3K inhibitor wortmannin reduced miR-27a-induced increases in AKT phosphorylation, glucose uptake, and GLUT4. miR-27a is considered to be involved in the PPAR-γ-PI3K/AKT-GLUT4 signaling axis, thus leading to increased glucose uptake and decreased IR in HFD-fed mice and 3T3-L1 adipocytes. Therefore, miR-27a is a novel target for the treatment of IR in obesity and diabetes. 10.18632/aging.102263
Salidroside mitigates hydrogen peroxide-induced injury by enhancement of microRNA-27a in human trabecular meshwork cells. Zhao Jun,Du Xiujuan,Wang Meng,Yang Peiyao,Zhang Juanmei Artificial cells, nanomedicine, and biotechnology Salidroside (Sal) exerted widely pharmacological effects in multitudinous diseases had been certified. The actual study clarified the protective activity of Sal in HO-injured human trabecular meshwork (HTM) cells. HTM cells were disposed with HO to construct an oxidative damage model in vitro. Then, Sal was utilized to administrate HTM cells, and cell viability, apoptosis, apoptosis-interrelated proteins and ROS production were appraised using CCK-8, flow cytometry, western blot and DCFH-DA staining. MiR-27a inhibitor and its control were transfected into HTM cells, and the influences of miR-27a inhibition in HTM cells stimulated with HO and Sal were detected. PI3K/AKT and Wnt/β-catenin pathways were ultimately investigated to uncover the underlying mechanism. We found that HO evoked HTM cells oxidative damage, as evidenced by repressing cell viability, inducing apoptosis, activating cleaved-caspase-3/-9 expression and increasing ROS production. Sal significantly lightened HO-evoked oxidative damage in HTM cells. Additionally, miR-27a was up-regulated by Sal, and miR-27a suppression significantly reversed the protective effect of Sal on HO-injured HTM cells. Finally, Sal activated PI3K/AKT and Wnt/β-catenin pathways through enhancement of miR-27a in HO-injured HTM cells. In conclusion, these discoveries suggested that Sal could protect HTM cells against HO-evoked oxidative damage by activating PI3K/AKT and Wnt/β-catenin pathways through enhancement of miR-27a. Highlights HO evokes HTM cells oxidative damage; Sal relieves HO-induced oxidative damage in HTM cells; Sal enhances miR-27a expression in HO-injured HTM cells; Repressed miR-27a reverses the protective impacts of Sal on HO-injured HTM cells; Sal activates PI3K/AKT and Wnt/β-catenin pathways by increasing miR-27a. 10.1080/21691401.2019.1608222
MiR-27a targets sFRP1 in hFOB cells to regulate proliferation, apoptosis and differentiation. Guo Donggeng,Li Qiuxia,Lv Qing,Wei Qiujing,Cao Shuangyan,Gu Jieruo PloS one MicroRNAs (miRNAs) play a key role in the regulation of almost all the physiological and pathological processes, including bone metabolism. Recent studies have suggested that miR-27 might play a key role in osteoblast differentiation and bone formation. Increasing evidence indicates that the canonical Wnt signaling pathway contributes to different stages of bone formation. In this study, we identify miR-27a can promote osteoblast differentiation by repressing a new target, secreted frizzled-related proteins 1 (sFRP1) expression at the transcriptional level. Here, 21 candidate targets of miR-27a involved in canonical Wnt/β-catenin signaling were predicted, and a significant decrease in sFRP1 luciferase activity was observed both in 293T and MG63 cells co-transfected with the matched luciferase reporter constructs and miR-27a mimic. Furthermore, the presence of exogenous miR-27a significantly decreased sFRP1 mRNA and protein expression in hFOB1.19 cells during both proliferation and osteogenic differentiation. The over-expression of miR-27a or knockdown sFRP1 significantly increased the percentage of apoptotic hFOBs, the percentage of cells in the G2-M phase of the cell cycle and the expression of key osteoblastic markers, including ALP, SPP1, RUNX2 and ALP activity. Over-expression of miR-27a or knockdown of endogenous sFRP1 led to an accumulation of β-catenin in hFOBs. In the present study, we demonstrate that miR-27a induced gene silencing effect is a vital mechanism contributing to bone metabolism in hFOB cells in vitro, which is partly affected by the post-transcriptional regulation of sFRP1, during osteoblast proliferation, apoptosis and differentiation. 10.1371/journal.pone.0091354
MiR-27a-5p regulates apoptosis of liver ischemia-reperfusion injury in mice by targeting Bach1. Xing Yu,Li Jing,Li Shi-Peng,Xi Jiri,Ma Ning,Liu Lei,Wang Jin-Shan,Cai Jin-Zhen Journal of cellular biochemistry Ischemia-reperfusion (I/R) injury causes cellular dysfunction and a series of immune or apoptotic reactions. Bach1 is a mammalian transcription factor that represses Hmox1, which encodes heme oxygenase-1 (HO-1) that can degrade heme into free iron, carbon monoxide, and biliverdin, to play an important role in antioxidant, anti-inflammatory, and antiapoptotic activities. MicroRNAs (miRNAs) can be found in a variety of eukaryotic cells and viruses, a class of noncoding small RNAs that are encoded by endogenous genes. The aims of this study were to determine whether miR-27a-5p targets Bach1 and regulates cellular death; the dual-luciferase reporter assay was used to detect this and the results showed that miR-27a-5p significantly decreased the luciferase activity of the Bach1 3'-untranslated region. MiR-27a-5p was increased in mice during hepatic I/R and Bach1 was decreased. By transfecting the AML12 cells with the mimic, inhibitor miR-27a-5p in hypoxia/reoxygenation (H/R) models showed that overexpression of miR-27a-5p decreased Bach1 messenger RNA, upregulated HO-1 expression, and promoted antiapoptotic Bcl-2 and downregulated proapoptotic caspase-3 gene expression. In contrast, the miR-27a-5p inhibitor yielded the opposite results. Meanwhile, transfection with Bach1 small interference RNA obviously upregulated the protein levels of HO-1 and resulted in an increase in Bcl-2 and a decrease in caspase-3 protein levels. Thus, we can conclude that miR-27a-5p is relevant to liver I/R injury and overexpression of miR-27a-5p may alleviate apoptosis in H/R injury by targeting Bach1 in vitro. 10.1002/jcb.27383
rhTNFR:Fc increases Nrf2 expression via miR-27a mediation to protect myocardium against sepsis injury. Xue Wei-Liang,Bai Xiaoyan,Zhang Ling Biochemical and biophysical research communications OBJECTIVE:Sepsis is a whole-body inflammation disease and can result in septic shock and multiple organ failure. The previous study demonstrated that miR-27a plays a critical role in inflammation regulation. Here, we investigated that effect and its possible mechanism of rhTNFR:Fc on sepsis treatment. METHODS:LPS induced sepsis mice model was established. 10 mg/kg rhTNFR:Fc was used to treat sepsis mice by intravenous injection. RESULTS:RhTNFR:Fc improved cardiac function of sepsis mice, and markedly decreased miR-27a but increased Nrf2 expression level of myocardium in LPS treated mice. In H9C2 cells, rhTNFR:Fc also increased Nrf2 expression, elevated cell viability and decreased cell apoptosis. However, the effects were reversed by miR-27a mimic. In addition, miR-27a mimic reduced the activity of Nrf2 3'UTR while miR-27a inhibitor elevated enhanced its level. CONCLUSION:rhTNFR:Fc activated Nrf2 pathway to protect myocardium against LPS-induced sepsis injury via miR-27a regulation. 10.1016/j.bbrc.2015.07.051
Curcumin inhibits cell viability, migration, and invasion of thymic carcinoma cells via downregulation of microRNA-27a. Han Zhifeng,Zhang Jingzhe,Zhang Kun,Zhao Yinghao Phytotherapy research : PTR Curcumin (CUR) is a kind of polyphenolic compound and widely used in the treatment of diseases. However, the involvement of CUR in thymic carcinoma remains unknown. The object of our research is to clarify the role of CUR and related regulatory mechanism in thymic carcinoma cells. After treatment with CUR for 24 hr, cell viability, apoptosis, migration, and invasion of TC1889 cells were measured. Real-time polymerase chain reaction was executed to examine the expression of microRNA-27a (miR-27a) in thymic carcinoma tissues and TC1889 cells. After miR-27a mimic transfection, whether miR-27a is involved in CUR-modulated cell behaviors was measured. Finally, western blot was utilized to detect mTOR and Notch 1 pathways-linked proteins. CUR restrained cell viability and increased cell apoptosis of TC1889 cells. In addition, cell migration and invasion were restrained by CUR. Meanwhile, miR-27a expression was positively regulated in thymic carcinoma tissues and downregulated by CUR in TC1889 cells. Overexpressed miR-27a reversed the CUR-induced reduction of growth, migration, and invasion in TC1889 cells. Furthermore, CUR blocked mTOR and Notch 1 pathways via downregulating miR-27a. We demonstrated that CUR blocked mTOR and Notch 1 pathways via downregulating miR-27a, thereby suppressing cell growth, migration, and invasion of thymic carcinoma cells. 10.1002/ptr.6629
MicroRNA-27a promotes proliferation and suppresses apoptosis by targeting PLK2 in laryngeal carcinoma. Tian Yuan,Fu Shuang,Qiu Guang-Bin,Xu Zhen-Ming,Liu Ning,Zhang Xiao-Wen,Chen Sheng,Wang Ye,Sun Kai-Lai,Fu Wei-Neng BMC cancer BACKGROUND:miRNA-27a has been confirmed as an important regulator in carcinogenesis and other pathological processes. Whether and how it plays a role in the laryngeal carcinoma is unknown. METHODS:Mature miRNA-27a expression in laryngeal cancer was detected by qRT-PCR. Gain-of-function studies using mature miR-27a were performed to investigate cell proliferation and apoptosis in the Hep2 cells. In silico database analysis and luciferase reporter assay were applied to predict and validate the direct target, respectively. Loss-of-function assays were performed to investigate the functional significance of the miR-27a target gene. qRT-PCR and Western blot were used to evaluate mRNA and protein levels of the target, respectively. RESULTS:miR-27a was significantly up-regulated in the laryngeal tumor tissues compared to the adjacent non-tumor tissues. In silico database analysis result revealed that PLK2 is a potential target of miR-27a. luciferase reporter assay result showed the direct inhibition of miR-27a on PLK2-3'UTR. In the cases with miR-27a up-regulation, PLK2 protein expression level was significantly lower in cancer tissues than that in the adjacent non-tumor tissues, which showed a negative correlation with miR-27a expression level. Both miR-27a and knockdown of PLK2 caused the increase of the cell viability and colony formation and inhibition of the late apoptosis in the Hep2 cell lines. Moreover, miR-27a but not PLK2 also repressed the early apoptosis in the Hep2 cells. Additionally, no alteration of the Hep2 cell cycle induced by miR-27a was detected. CONCLUSIONS:miR-27a acts as an oncogene in laryngeal squamous cell carcinoma through down-regulation of PLK2 and may provide a novel clue into the potential mechanism of LSCC oncogenesis or serve as a useful biomarker in diagnosis and therapy in laryngeal cancer. 10.1186/1471-2407-14-678
Bilobalide protects H9c2 cell from oxygen-glucose-deprivation-caused damage through upregulation of miR-27a. Artificial cells, nanomedicine, and biotechnology Myocardial ischemia is a troublesome disease. Bilobalide possesses multiple biological functions. We researched the consequents of bilobalide in OGD-irritated H9c2 cells. OGD-stimulated H9c2 cells were treated by bilobalide, and/or transfected with miR-27a inhibitor or negative control. Use CCK-8 and flow cytometry to test cell activity and apoptosis, respectively. Luciferase activity experiment was to test targeting link between miR-27a and Tmub1. Levels of cell-cycle and apoptosis relative proteins and phosphorylation of PI3K/AKT and Wnt/β-catenin related proteins were detected through western blot. OGD stimulation reduced cell activity and negatively regulated the expression of CDK4, CDK6 and CyclinD1. Cell apoptosis was increased and its related proteins were affected by OGD. Bilobalide administration reversed all the results above caused by OGD. OGD negatively regulated miR-27a while bilobalide upregulated miR-27a. miR-27a's target gene was Tmub1. The protection consequents of bilobalide were suppressed when cells were transfected with a miR-27a inhibitor that cell activity was reduced and apoptosis was raised. Attenuation in the phosphorylation level of PI3K, AKT and β-catenin by OGD was reversed by bilobalide, whereas there were opposite results after transfected with miR-27a inhibitor. Bilobalide relieved OGD-caused H9c2 cell damage, raising cell activity and attenuating apoptosis via upregulating miR-27a and activating of PI3K/AKT and Wnt/β-catenin signal pathway. Highlights Bilobalide alleviates OGD-induced H9c2 cell injury. Bilobalide upregulates miR-27a expression in OGD-stimulated H9c2 cells. Bilobalide alleviates cell injury by upregulation of miR-27a. Bilobalide actuates PI3K/AKT and Wnt/β-catenin pathways. 10.1080/21691401.2019.1640708
MicroRNA-27a regulates hepatic lipid metabolism and alleviates NAFLD via repressing FAS and SCD1. Zhang Meiyuan,Sun Weilan,Zhou Minghao,Tang Yan Scientific reports MicroRNAs are implicated as crucial mediators in metabolic diseases including obesity, diabetes, and non-alcoholic fatty liver diseases (NAFLD). Here, we show miR-27a attenuated hepatic de novo lipogenesis and alleviated obesity-initiated NAFLD through inhibiting Fasn and Scd1 in liver. Hepatic levels of miR-27a were significantly augmented in HFD-fed and ob/ob mice. Further studies demonstrated that miR-27a directly interacted with 3' untranslated region (3'-UTR) of hepatic Fasn and Scd1 mRNAs and reduced their expression levels in mice. Adenovirus-mediated overexpression of miR-27a robustly blocked sodium oleate-induced triglyceride (TG) accumulation in mouse primary hepatocytes and reduced liver TG contents in mice via repressing hepatic lipogenesis. Furthermore, ectopic expression of hepatic miR-27a impaired lipid contents of livers and attenuated NAFLD development through suppressing lipogenesis in HCD-fed and ob/ob mice. Together, our results reveal a critical role of miR-27a in lipid homeostasis of liver and pathogenesis of NAFLD. 10.1038/s41598-017-15141-x
Neuroprotective effects of miR-27a against traumatic brain injury via suppressing FoxO3a-mediated neuronal autophagy. Sun Liqian,Zhao Manman,Wang Yan,Liu Aihua,Lv Ming,Li Youxiang,Yang Xinjian,Wu Zhongxue Biochemical and biophysical research communications MicroRNA-27a (miR-27a) has been reported to be a brain-specific miRNA and aberrantly expressed in the brain suffered from traumatic brain injury (TBI). The present study is designed to investigate the potential role and molecular mechanism of miR-27a in the pathogenesis of TBI. The level of miR-27a in brain was manipulated by intracerebroventricular injection of lentiviral-encoding miR-27a before TBI induction. Real-time PCR was used to detected miR-27a and Forkhead box O3a (FoxO3a) levels in the hippocampus. Then, we evaluated the impact of miR-27a overexpression on neurological function, brain edema, lesion volume and neuronal autophagy after TBI. The blinding of miR-27a to the 3'UTR of FoxO3a mRNA and its effects on FoxO3a translation were analyzed by luciferase reporter assay and Western blot. The downregulation of miR-27a and the increase in FoxO3a level were observed in the hippocampus post-TBI. Overexpression of miR-27a significantly attenuated neurological deficits and brain injury, especially suppressed autophagic activation after TBI. Furthermore, we identified that miR-27a directly targeted the FoxO3a 3'UTR region to reduced FoxO3a protein expression. Knockdown of FoxO3a significantly reversed high levels of autophagy-related genes induced by TBI. Taken together, Overexpression of miR-27a may protect against brain injury via suppressing FoxO3a-mediated neuronal autophagy following TBI. 10.1016/j.bbrc.2016.12.001
miR-27a is up regulated and promotes inflammatory response in sepsis. Wang Zhongchuan,Ruan Zhengshang,Mao Yanfei,Dong Wenwen,Zhang Yunqian,Yin Na,Jiang Lai Cellular immunology MicroRNAs (miRNAs) are short, non-coding RNAs that regulate the expression of multiple target genes. Dysregulation of miRNAs is common in sepsis. Through microRNA microarray and qRT-PCR we found that the levels of miR-27a, miR-153 and miR-143 are up regulated, while let-7a, miR-218 and miR-129-5p are down regulated in lungs of septic mice. Knocking down of miR-27a down regulates expression levels of TNF-α and IL-6 significantly via reducing the phosphorylation level of NF-κB p65 and inhibiting its DNA binding activity. Furthermore, neutralisation of miR-27a up regulates PPARγ level, down regulates TNF-α expression, relieves pulmonary inflammation and promotes survival of septic mice, which demonstrates that miR-27a plays an important role in regulating inflammatory response in sepsis and provides a potential target for clinical sepsis research and treatment. 10.1016/j.cellimm.2014.06.006
Mir-27a promotes apoptosis of cochlear sensory epithelium in Cx26 knockout mice. Wang Yunfeng,Lin Chen,He Yingzi,Li Ao,Ni Wenli,Sun Shan,Gu Xiaodong,Li Jian,Li Huawei Frontiers in bioscience (Landmark edition) To investigate the underlying molecular mechanism for connexin 26 (Cx26) knockout-induced apoptosis, we performed TUNEL assays to detect apoptosis in the cochlear sensory epithelium in Cx26 knockout mice. We also compared the miRNA expression profiles of Cx26 knockout and wild-type mice using microarray technology and bioinformatic analyses. Real-time PCR, luciferase reporter gene assays, and scala media microinjections were performed to identify the effect of a specific miRNA and its targets. The results showed that apoptosis increased in the cochlear sensory epithelium of Cx26 knockout mice. The abnormal expression of mir-27a and sgk1 in Cx26 knockout mice was verified with real-time PCR. Luciferase reporter gene assays showed that overexpression of mir-27a significantly decreased sgk1 reporter gene activity; an inhibitor of mir-27a blocked the effect. Mir-27a lentivirus also inhibited sgk1 expression in cultured cochlear tissue. Mir-27a shRNA treatment inhibited Cx26 knockout-induced apoptosis in the cochlear sensory epithelium of mice and increased the expression of sgk1 mRNA. Thus, mir-27a was identified as an apoptotic molecule that participates in Cx26 knockout-induced apoptosis in the cochlear sensory epithelium of mice by downregulating sgk1 expression. 10.2741/4393
Secreted Monocyte miR-27a, via Mesenteric Arterial Mas Receptor-eNOS Pathway, Causes Hypertension. Zou Xue,Wang Jialiang,Chen Caiyu,Tan Xiaorong,Huang Yu,Jose Pedro A,Yang Jian,Zeng Chunyu American journal of hypertension BACKGROUND:Essential hypertension is associated with increased plasma concentrations of extracellular vesicles (EVs). We aimed to determine the role of monocyte miR-27a in EVs on arterial Mas receptor expression, and its involvement in the pathogenesis of hypertension. METHODS:THP-1 cells were transfected with miR-27a mimic and miR-27a inhibitor, and EVs were collected. Mas receptor expression and endothelial nitric oxide synthase (eNOS) phosphorylation were determined by immunoblotting. Sprague-Dawley (SD) rats received EVs via tail-vein injection. Blood pressure (BP) was measured with the tail-cuff method. The vasodilatory response of mesenteric arteries was measured using a small vessel myograph. RESULTS:EVs from THP-1 cells increased rat BP by impairing Ang-(1-7)-mediated vasodilation in mesenteric arteries, which was further exaggerated by EVs from lipopolysaccharides-treated THP-1 cells. As the receptor and key signaling of Ang-(1-7), next experiments found that Mas receptor expression and eNOS phosphorylation were decreased in mesenteric arteries from EVs-treated SD rats. Screening studies found miR-27a in EVs may be involved in this process. Through transfection with miR-27a inhibitor or miR-27a mimic, we found that miR-27a downregulates Mas receptor expression in endothelial cells. Injection of EVs from miR-27a-transfected HEK-293 cells decreased Mas receptor and eNOS phosphorylation in mesenteric arteries, impaired Ang-(1-7)-mediated vasodilation and increased BP. Earlier effects were reversed using cells with downregulation of miR-27 in EVs. CONCLUSIONS:Monocyte miR-27a in EVs decreases Mas receptor expression and eNOS phosphorylation in endothelium, impairs Ang-(1-7)-mediated vasodilation, and causes hypertension. Understanding the contributions of EVs in the pathogenesis of hypertension may facilitate their use as a diagnostic biomarker. 10.1093/ajh/hpz112
Extracellular vesicular MicroRNA-27a* contributes to cardiac hypertrophy in chronic heart failure. Tian Changhai,Hu Guoku,Gao Lie,Hackfort Bryan T,Zucker Irving H Journal of molecular and cellular cardiology Under stress, the heart undergoes extensive remodeling resulting in cardiac fibrosis and hypertrophy, ultimately contributing to chronic heart failure (CHF). Alterations in microRNA levels are associated with dysfunctional gene expression profiles involved in the pathogenesis of heart failure. We previously showed that myocardial infarction-induced microRNA-enriched extracellular vesicles (EVs) contribute to the reduction in antioxidant enzymes by targeting Nrf2 signaling in CHF. MicroRNA-27a (miRNA-27a) is the predominant microRNA contained in cardiac fibroblast-derived EVs contributing to oxidative stress along with hypertrophic gene expression in cardiomyocytes. In the present study, we observed that miRNA-27a passenger strand (miRNA-27a*) was markedly upregulated in the non-infarcted area of the left ventricle of rats with CHF and encapsulated into EVs and secreted into the circulation. Bioinformatic analysis revealed that PDZ and LIM domain 5 (PDLIM5) is one of the major targets of miRNA-27a*, playing a major role in cardiac structure and function, and potentially contributing to the progression of cardiac hypertrophy. Our in vivo data demonstrate that PDLIM5 is down-regulated in the progression of heart failure, accompanied with the upregulation of hypertrophic genes and consistent with alterations in miRNA-27a*. Moreover, exogenous administration of miRNA27a* mimics inhibit PDLIM5 translation in cardiomyocytes whereas a miRNA27a* inhibitor enhanced PDLIM5 expression. Importantly, we confirmed that infarcted hearts have higher abundance of miRNA-27a* in EVs compared to normal hearts and further demonstrated that cultured cardiac fibroblasts secrete miRNA27a*-enriched EVs into the extracellular space in response to Angiotensin II stimulation, which inhibited PDLIM5 translation, leading to cardiomyocyte hypertrophic gene expression. In vivo studies suggest that the administration of a miRNA-27a* inhibitor in CHF rats partially blocks endogenous miR-27a* expression, prevents hypertrophic gene expression and improves myocardial contractility. These findings suggest that cardiac fibroblast-secretion of miRNA27a*-enriched EVs may act as a paracrine signaling mediator of cardiac hypertrophy that has potential as a novel therapeutic target. 10.1016/j.yjmcc.2020.04.032
Loss of microRNA-27a induces cardiac dysfunction through activating FoxO1. Qin X-D,Liu L European review for medical and pharmacological sciences OBJECTIVE:To elucidate how microRNA-27a and FoxO1 regulate cardiac dysfunction in mice. MATERIALS AND METHODS:Expression levels of ANP, BNP, β-MHC, α-SMA, Fn1, and Periostin in myocardial tissues of 2-month-old and 8-month-old microRNA-27a-KO mice and age-matched wild-type mice were determined by quantitative Real Time-Polymerase Chain Reaction (qRT-PCR) and Western blot. Dual-luciferase reporter gene assay was conducted in H9C2 cells to verify the binding condition between microRNA-27a and FoxO1. By transfection of microRNA-27a mimics or inhibitor, FoxO1 expression in H9C2 cells was determined at the mRNA and protein levels. HW/BW [ratio of heart weight (mg) and body weight (mg)], HW/TL [ratio of heart weight (mg) and tibial length (mm)], LVPWDT [left ventricular posterior wall diastolic thickness (mm)], LVEDD [left ventricular end-diastolic dimension (mm)], and FS (fractional shortening) in mice treated with or without FoxO1 inhibitor AS1842856 were accessed through echocardiography. RESULTS:MicroRNA-27a-KO mice had larger LVEDD, HW/BW, and HW/TL, but lower FS and LVPWDT than those of age-matched wild-type mice. Besides, higher levels of ANP, BNP, β-MHC, α-SMA, Fn1, and Periostin were observed in myocardial tissues of microRNA-27a-KO mice compared with those of age-matched wild-type mice. Dual-luciferase reporter gene assay revealed lower luciferase activity in H9C2 cells co-transfected with microRNA-27a mimics and wild-type FoxO1 than that of controls. The expression level of FoxO1 was negatively regulated by microRNA-27a in H9C2 cells at the mRNA and protein levels. After AS1842856 injection, HW/BW, HW/TL, and LVEDD in microRNA-27a-KO mice markedly decreased, whereas FS and LVPWDT elevated. By comparison, AS1842856 injection did not influence cardiac development in wild-type mice. CONCLUSIONS:MicroRNA-27a knockout could induce cardiac dysfunction in mice through upregulating FoxO1 expression. 10.26355/eurrev_201907_18340
[Effect of microRNA-27a-3p on proliferation, apoptosis and cell cycle of hepatoma cells]. Yang Z F,Yang Y,Zhang R L,Jia C L,Li Z P,Wang W R,Zhang H,Li S S,Bao Y X Zhonghua gan zang bing za zhi = Zhonghua ganzangbing zazhi = Chinese journal of hepatology To investigate the effect of miR-27a-3p on proliferation, apoptosis and cell cycle of hepatoma cells. A quantitative real-time polymerase chain reaction (qPCR) was used to detect differential expression of miR-27a-3p in normal hepatic epithelial cells (L02) and hepatoma cells (HepG2 and PLC). Cell experiment was divided into four groups: HepG2 overexpression cells, Mi-27a-3p overexpression group (Mi-27a) and negative control group (Mi-Con); PLC knockdown cells, Mi-27a-3p knockdown group (Mi-inhibitor-27a) and negative control group (Mi-inhibitor-Con). The expression of microRNA-27a-3p in each group after transfection was detected by qPCR analysis. MTT assay was used to detect the cell proliferation. Flow cytometry was used to detect the apoptosis and cell cycle. One-way ANOVA was used for multiple comparisons, and t-test was used to compare two groups. qPCR results showed that the expression levels of miR-27a-3p in L02, HepG2 and PLC increased sequentially, and the relative expression levels were 1.07 ± 0.04, 4.81 ± 0.64 and 11.31 ± 0.92, respectively ( < 0.05). MTT assay showed that the cell viability of HepG2 cells transfected with miR-27a-3p overexpression plasmid was significantly decreased compared with the negative control group ( < 0.05). The apoptosis assay showed that the apoptosis rate of miR-27a-3p overexpression group was higher than the negative control group ( < 0.05). The cell cycle results showed that the proportion of S phase cells in the miR-27a-3p overexpression cell group was significantly lower than the negative control group ( < 0.05). Furthermore, microRNA-27a-3p knockdown validation in PLC cells showed that MTT, apoptosis and cell cycle tests results were opposite to the results of HepG2 overexpression cells, and the differences were statistically significant ( < 0.05). miR-27a-3p can significantly inhibit the proliferation of hepatoma cells, promote cell apoptosis, alter the cell cycle distribution, and may become a potential target in hepatocellular carcinoma therapy. 10.3760/cma.j.issn.1007-3418.2019.03.006
CircNR3C1 inhibits proliferation of bladder cancer cells by sponging miR-27a-3p and downregulating cyclin D1 expression. Zheng Fuxin,Wang Miao,Li Yawei,Huang Chao,Tao Dan,Xie Fei,Zhang Hui,Sun Jiayin,Zhang Chuanhua,Gu Chaohui,Wang Zhendi,Jiang Guosong Cancer letters Accumulating evidences suggest that circular RNAs play vital roles in human cancers. Previously, we found that circHIPK3 suppressed invasion of bladder cancer cells via sponging miR-558 and downregulating heparanase expression. In this study, we discovered that a circular RNA derived from NR3C1 (circNR3C1) was downregulated in bladder cancer tissues and cell lines according to RNA-Seq data and qRT-PCR analysis. Functionally, we found that overexpression of circNR3C1 could significantly inhibit cell cycle progression and proliferation of bladder cancer cells in vitro, as well as suppress tumor growth in vivo. Mechanistically, we demonstrated that circNR3C1 possessed four targeting sites of miR-27a-3p and could effectively sponge miR-27a-3p to suppress the expression of cyclin D1. Furthermore, we revealed that miR-27a-3p functioned as an oncogene through interacting with 5'UTR of cyclin D1 to enhance its expression, which led to promote cell cycle progression and proliferation in bladder cancer cells. Conclusively, our findings further confirm the hypothesis that circRNAs function as "microRNA sponges", and our data suggest that circNR3C1 and miR-27a-3p would be potential therapeutic targets for bladder cancer treatment. 10.1016/j.canlet.2019.06.018
miR-27a-3p regulates proliferation and apoptosis of colon cancer cells by potentially targeting BTG1. Su Chang,Huang Dong-Ping,Liu Jian-Wen,Liu Wei-Yan,Cao Yi-Ou Oncology letters microRNA (miR/miRNA)-27a-3p has been reported to be abnormally expressed in various types of cancer, including colorectal cancer (CRC). B-cell translocation gene 1 (BTG1) has also been implicated with CRC. However, the association between miR-27a-3p and BTG1 in CRC, to the best of our knowledge, has not been investigated. In order to assess whether miR-27a-3p is associated with CRC, reverse transcription-quantitative PCR was performed on 20 paired CRC and paracancerous tissues for miRNA analysis. For the screening and validation of miR-27a-3p expression in colon cancer, several colon cancer cell lines (HCT-116, HCT8, SW480, HT29, LOVO and Caco2) and the normal colorectal epithelial cell line NCM460 were examined. The highest expression levels of miR-27a-3p were detected in the HCT-116, which was selected for further experimentation. The HCT-116 cells were divided into control, miR-27a-3p mimic and inhibitor groups, and cell proliferation was tested using an MTT assay. Additionally, miR-27a-3p inhibitor/mimic or BTG1 plasmid were transfected into the HCT-116 cells, and flow cytometry was performed to analyze cell cycle distributions. TUNEL analysis was performed to detect apoptosis. Protein levels of factors in the downstream signaling pathway mediated by miR-27a-3p [ERK/mitogen-activated extracellular signal-regulated kinase (MEK)] were detected. miR-27a-3p was revealed to be overexpressed in human CRC tissues and colon cancer cell lines. Knockdown of miR-27a-3p suppressed proliferation of HCT-116 cells and apoptosis was increased. It further markedly upregulated expression levels of BTG1 and inhibited activation of proteins of the ERK/MEK signaling pathway. In addition, overexpression of BTG1 in HCT-116 cells triggered G/S phase cell cycle arrest and increased apoptosis via the ERK/MEK signaling pathway. In conclusion, the present study demonstrated that the effects of miR-27a-3p on colon cancer cell proliferation and apoptosis were similar to those of the tumor suppressor gene BTG1. The miR-27a-3p/BTG1 axis may have potential implications for diagnostic and therapeutic approaches in CRC. 10.3892/ol.2019.10629
CircRNA cRAPGEF5 inhibits the growth and metastasis of renal cell carcinoma via the miR-27a-3p/TXNIP pathway. Chen Qiong,Liu Tao,Bao Yi,Zhao Tangliang,Wang Jie,Wang Hui,Wang Anbang,Gan Xinxin,Wu Zhenjie,Wang Linhui Cancer letters Circular RNAs (circRNAs) are reported to act as important regulators in cancers. CircRNA RAPGEF5 (cRAPGEF5) is derived from exons 2-6 of the RAPGEF5 gene and may promote papillary thyroid cancer progression. However, the role of cRAPGEF5 in renal cell carcinoma (RCC) remains unclear. In this study, we found cRAPGEF5 to be significantly downregulated in RCC tissues. Among 245 RCC cases, cRAPGEF5 downregulation correlated positively with aggressive clinical characteristics and independently predicted poor overall survival and recurrence-free survival. Functional assays demonstrated that cRAPGEF5 suppresses RCC proliferation and migration in vitro and in vivo. Mechanistically, RNA Immunoprecipitation and circRNA in vivo precipitation assays showed that cRAPGEF5 functions as a sponge of oncogenic miR-27a-3p, which targets the suppressor gene TXNIP. Interactions between miR-27a-3p and cRAPGEF5 or TXNIP were confirmed by dual-luciferase reporter assays. In conclusion, cRAPGEF5 plays a role in suppressing RCC via the miR-27a-3p/TXNIP pathway and may serve as a promising prognostic biomarker and novel therapeutic target for RCC patients. 10.1016/j.canlet.2019.10.017
Mesenchymal Stem Cell-Derived Extracellular Vesicles Alleviate Acute Lung Injury Via Transfer of miR-27a-3p. Wang Jiangmei,Huang Ruoqiong,Xu Qi,Zheng Guoping,Qiu Guanguan,Ge Menghua,Shu Qiang,Xu Jianguo Critical care medicine OBJECTIVES:The goal of this study was to determine the role of microRNA transfer in mediating the effects of mesenchymal stem cell-derived extracellular vesicles in acute lung injury. DESIGN:Experimental cell and animal studies. SETTING:University-based research laboratory. SUBJECTS:THP-1 monocytes, bone marrow-derived macrophages, and C57BL/6 mice. INTERVENTIONS:To determine the microRNA transfer in vitro, mesenchymal stem cells and mesenchymal stem cell-derived extracellular vesicles were cultured with THP-1 cells and bone marrow-derived macrophages and then assayed for microRNA expression in the target cells. To examine the role of microRNA transfer in vivo, mesenchymal stem cell-derived extracellular vesicles were administered to mice with lipopolysaccharide-induced lung injury. MEASUREMENTS AND MAIN RESULTS:Mesenchymal stem cell-derived extracellular vesicles were efficiently taken up by macrophages in vitro and in vivo. miR-27a-3p was one of the most highly expressed microRNAs in THP-1 cells in microarray analysis and was transferred from mesenchymal stem cells and mesenchymal stem cell-derived extracellular vesicles to THP-1/bone marrow-derived macrophages. Mesenchymal stem cell-derived extracellular vesicles promoted M2 polarization in bone marrow-derived macrophages, which was inhibited by lentiviral anti-miR-27a-3p transduction. Mesenchymal stem cell-derived extracellular vesicles administered systemically and intratracheally were as effective as mesenchymal stem cells in alleviating acute lung injury, elevating miR-27a-3p levels in alveolar macrophages, and promoting M2 macrophage polarization. Treatment of mesenchymal stem cell-derived extracellular vesicles concurrently decreased alveolar macrophage expression of nuclear factor kappa B subunit 1, a target of miR-27a-3p. Lentiviral transduction of mesenchymal stem cells with anti-miR-27a-3p or knockdown of miR-27a-3p in vivo abolished the effects of mesenchymal stem cell-derived extracellular vesicles on acute lung injury and M2 macrophage polarization. CONCLUSIONS:Mesenchymal stem cell-derived extracellular vesicles mitigate acute lung injury at least partially via transferring miR-27a-3p to alveolar macrophages. miR-27a-3p acts to target NFKB1 and is a crucial regulator of M2 macrophage polarization. 10.1097/CCM.0000000000004315
Involvement of miR-27a-3p in diabetic nephropathy via affecting renal fibrosis, mitochondrial dysfunction, and endoplasmic reticulum stress. Wu Lina,Wang Qingzhu,Guo Feng,Ma Xiaojun,Wang Jiao,Zhao Yanyan,Yan Yushan,Qin Guijun Journal of cellular physiology Diabetic nephropathy (DN) is acknowledged as a serious chronic complication of diabetes mellitus. Nevertheless, its pathogenesis is complicated and unclear. Thus, in this study, the role of miR-27a-3p-prohibitin/TMBIM6 signaling axis in the progression of DN was elucidated. Type 2 diabetic db/db mice and high glucose (HG)-challenged HK-2 cells were used as in vivo and in vitro models. Our results showed that miR-27a-3p was upregulated and prohibitin or transmembrane BAX inhibitor motif containing 6 (TMBIM6) was downregulated in the kidney tissues of db/db mice and HG-treated HK-2 cells. Silencing miR-27a-3p enhanced the expression of prohibitin and TMBIM6 in the kidney tissues and HK-2 cells. Inhibition of miR-27a-3p improved functional injury, as evidenced by decreased blood glucose, urinary albumin, serum creatinine, and blood urea nitrogen levels. MiR-27a-3p silencing ameliorated renal fibrosis, reflected by reduced profibrogenic genes (e.g., transforming growth factor β1, fibronectin, collagen I and III, and α-smooth muscle actin). Furthermore, inhibition of miR-27a-3p relieved mitochondrial dysfunction in the kidney of db/db mice, including upregulation of mitochondrial membrane potential, complex I and III activities, adenosine triphosphate, and mitochondrial cytochrome C, as well as suppressing reactive oxygen species production. In addition, miR-27a-3p silencing attenuated endoplasmic reticulum (ER) stress, reflected by reduced expression of p-IRE1α, p-eIF2α, XBP1s, and CHOP. Mechanically, we identified prohibitin and TMBIM6 as direct targets of miR-27a-3p. Inhibition of miR-27a-3p protected HG-treated HK-2 cells from apoptosis, extracellular matrix accumulation, mitochondrial dysfunction, and ER stress by regulating prohibitin or TMBIM6. Taken together, we reveal that miR-27a-3p-prohibitin/TMBIM6 signaling axis regulates the progression of DN, which can be a potential therapeutic target. 10.1002/jcp.29951
LncRNA NEAT1 regulated inflammation and apoptosis in a rat model of sepsis-induced acute kidney injury via MiR-27a-3p/TAB3 axis. Bioscience, biotechnology, and biochemistry This study explored the mechanism of NEAT1 in sepsis-induced AKI rats. Cecal ligation punctures (CLP)-induced AKI rats were injected with siRNA-NEAT1 lentivirus. Kidney histopathology and apoptosis were evaluated via hematoxylin-eosin and TUNEL staining, respectively. ELISA determined the levels of Blood urea nitrogen (BUN), serum creatinine (SCr), neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule-1 (KIM-1), TNF-α, Interleukin (IL)-1β, and IL-6. Colorimetry measured malondialdehyde (MDA), superoxide dismutase (SOD) activities. qPCR analyzed NEAT1, miR-27a-3p, TAB3, Bcl-2, and Bax expressions. siNEAT1 reversed the promotive effect of CLP on kidney histopathological injury, and BUN, SCr, NGAL, KIM-1, TNF-α, IL-1β, IL-6, MDA, and Bax levels and apoptosis, but raised CLP-downregulated SOD and Bcl-2 levels. NEAT1 sponged miR-27a-3p which targeted TAB3. siNEAT1 upregulated miR-27a-3p and downregulated TAB3 expression. TAB3 overexpression reversed the inhibitory effect of siNEAT1 on the LPS-induced apoptosis of HK-2 cells. siNEAT1 alleviated sepsis-induced AKI in rats and LPS-induced sepsis of cells via miR-27a-3p/TAB3 axis. 10.1080/09168451.2020.1792760
TUG1 Represses Apoptosis, Autophagy, and Inflammatory Response by Regulating miR-27a-3p/SLIT2 in Lipopolysaccharide-Treated Vascular Endothelial Cells. Dong Yuanyuan,Fan Gongchun,Li Yanhong,Zhou Qin The Journal of surgical research BACKGROUND:The dysfunction of vascular endothelial cells is associated with sepsis development. Long noncoding RNAs take part in the regulation of vascular endothelial cell function. This study aimed to explore the role and mechanism of long noncoding RNA taurine-upregulated gene 1 (TUG1) in lipopolysaccharide (LPS)-induced endothelial cell injury. METHODS:LPS-treated human umbilical vein endothelial cells (HUVECs) were used as a model of sepsis in vitro. Quantitative real-time polymerase chain reaction was performed to detect the expression of TUG1, microRNA-27a-3p (miR-27a-3p) and slit guidance ligand 2 (SLIT2) messenger RNA. Western blot was conducted to measure the protein levels of SLIT2 as well as those involved in apoptosis, autophagy, and inflammatory response. Flow cytometry was used to detect cell apoptotic rate. The targets of TUG1 and miR-27a-3p were predicted via starBase (http://starbase.sysu.edu.cn/index.php). Dual-luciferase reporter, RNA immunoprecipitation, and pull-down assays were carried out to validate the target correlation between miR-27a-3p and TUG1/SLIT2. RESULTS:TUG1 expression was decreased after the treatment of LPS in HUVECs. Overexpression of TUG1 decreased LPS-induced apoptosis, autophagy, and inflammatory response. TUG1 was a sponge of miR-27a-3p. Upregulation of miR-27a-3p reversed the suppressive effect of TUG1 overexpression on LPS-induced apoptosis, autophagy, and inflammatory response. SLIT2 was a target of miR-27a-3p. Knockdown of miR-27a-3p could inhibit LPS-induced injury by increasing SLIT2 in HUVECs. TUG1 could enhance SLIT2 expression by competitively sponging miR-27a-3p. CONCLUSIONS:TUG1 could repress cell apoptosis, autophagy, and inflammatory response in LPS-treated HUVECs by sponging miR-27a-3p to target SLIT2, providing a potential target for the treatment of sepsis. 10.1016/j.jss.2020.05.102
MiR-27a-3p Targeting GSK3β Promotes Triple-Negative Breast Cancer Proliferation and Migration Through Wnt/β-Catenin Pathway. Cancer management and research BACKGROUND:Dysregulation of microRNAs (miRNAs) was found to play crucial roles in varieties of cancers, which affect tumor proliferation and migration. MiR-27a-3p has been identified as a tumor-related miRNA in liver cancer, lung cancer, and colorectal cancer. However, the function of miR-27a-3p in triple-negative breast cancer (TNBC) and its possible molecular mechanisms have still not been elucidated. METHODS:QRT-PCR technique was used to detect the expression of miR-27a-3p in TNBC and normal breast cell lines or the effects of miR-27a-3p knockdown and overexpression in TNBC cell lines. Proliferation and migration were measured by CCK-8 method, colony formation, wound healing, and Transwell assays, respectively. Furthermore, we used a dual-luciferase reporter gene assay and Western blot analysis to identify GSK3β as a target of miR-27a-3p. RESULTS:In this study, we found that miR-27a-3p expression was significantly elevated in TNBC cell lines. Database analysis suggested that TNBC patients with a high expression of miR-27a-3p have poorer overall survival possibilities. Overexpression of miR-27a-3p promotes TNBC cells proliferation, colony formation, and cell migration in vitro. Nevertheless, dual-luciferase reporter result showed that miR-27a-3p directly targeted the 3'-UTR regions of GSK3β mRNA and negatively regulated its expression. Lastly, we demonstrated that miR-27a-3p inactivates Wnt/β-catenin signaling pathway via targeting GSK3β. CONCLUSION:These results indicate that expression of miR-27a-3p was highly expressed in TNBC and promoted tumor progression through attenuating GSK3β and may have a potential molecular-targeted strategy for TNBC therapy. 10.2147/CMAR.S255419
MicroRNA-27a-3p directly targets FosB to regulate cell proliferation, apoptosis, and inflammation responses in immunoglobulin a nephropathy. Liao Yu,Wang Ziyan,Wang Lixin,Lin Yanzhao,Ye Ziyi,Zeng Xufang,Wei Fangning Biochemical and biophysical research communications Immunoglobulin A nephropathy (IgAN) constitutes the most common primary glomerulonephritis worldwide; however, the exact pathogenesis of IgAN is unknown. Previous genome-wide analysis of microRNA (miRNA) expression in the kidney has confirmed that miRNAs are closely related to the pathological changes of IgAN. Accordingly, in this study we found that miR-27a-3p is upregulated in IgAN kidney tissues in addition to human podocytes and tubule epithelial HK2 but not mesangial cells. Methylthiazolyldiphenyl-tetrazolium bromide (MTT), flow cytometry, real-time polymerase chain reaction, western blot, and enzyme-linked immunosorbent assays were used to verify the regulatory effects of miR-27a-3p and its inhibition on cell proliferation, apoptosis, and release of inflammatory factors in podocytes and HK2 cells. The target genes of miR-27a-3p were predicted using bioinformatics software; the identity of FosB as a target gene of miR-27a-3p was confirmed by luciferase report assay and western blot. Overall, our findings demonstrated that miR-27a-3p regulates cell apoptosis, cell proliferation, and the release of inflammatory cytokines of human podocytes and HK2 cells by directly targeting FosB. Our results therefore suggested that miR-27a-3p might be associated with the pathophysiology of IgAN and may represent a potential target for further studies related to IgAN mechanism or therapeutics. 10.1016/j.bbrc.2020.06.115
Overexpression of Linc 4930556M19Rik Suppresses High Glucose-Triggered Podocyte Apoptosis, Fibrosis and Inflammation via the miR-27a-3p/Metalloproteinase 3 (TIMP3) Axis in Diabetic Nephropathy. Fan Hong,Zhang Weiwei Medical science monitor : international medical journal of experimental and clinical research BACKGROUND Long non-coding RNAs (lncRNAs) play vital roles in development of diabetic nephropathy (DN). The goal of our study was to investigate the functional roles of long intergenic noncoding RNA (lincRNA) 4930556M19Rik in DN. MATERIAL AND METHODS A DN cell model was constructed by exposing podocytes to high glucose (HG). A subcellular fraction assay was used to determine the level of 4930556M19Rik in the nucleus and cytoplasm of podocytes. Quantitative real-time polymerase chain reaction was used to evaluate expression of 4930556M19Rik and miR-27a-3p. Western blot assay was used to assessed levels of fibrosis-related proteins, podocin, and tissue inhibitor of metalloproteinase 3 (TIMP3). Flow cytometry analysis was performed to analyze cell apoptosis. Enzyme linked immunosorbent assay was used to examine secretion of inflammatory cytokines. Dual-luciferase reporter, RIP, and RNA pull-down assays were used to verify the relationship between miR-27a-3p and 4930556M19Rik or TIMP3. RESULTS 4930556M19Rik was significantly decreased in HG-stimulated podocytes and mainly enriched in the cytoplasm of podocytes. Elevation of 4930556M19Rik hampered HG-induced cell apoptosis, fibrosis, and inflammatory in podocytes. 4930556M19Rik sponged miR-27a-3p to negatively modulate miR-27a-3p expression. MiR-27a-3p overexpression reversed the impact of 4930556M19Rik mediated cell progression in HG-induced podocytes. Moreover, TIMP3 was the target for miR-27a-3p and miR-27a-3p inhibition slowed podocyte injury by targeting TIMP3. CONCLUSIONS 4930556M19Rik overexpression slowed HG-induced podocyte injury by downregulating miR-27a-3p and upregulating TIMP3. 10.12659/MSM.925361
MiR-27a-3p overexpression mitigates inflammation and apoptosis of lipopolysaccharides-induced alveolar epithelial cells by targeting FOXO3 and suppressing the activation of NAPDH/ROS. Shang Jian,Wang Lei,Tan Lili,Pan Ren,Wu Dan,Xia Yanfei,Xu Peng Biochemical and biophysical research communications BACKGROUND:Acute respiratory distress syndrome (ARDS) is multiple inflammatory injury lung disease. MiR-27a-3p alleviates lung injury, whether miR-27a-3p could affect the lung inflammation is not clear. Therefore, we established the lipopolysaccharides (LPS)-induced alveolar epithelial cell model to simulate ARDS inflammation in vitro to investigate the effect of miR-27a-3p in ARDS. METHODS:After LPS-induced alveolar epithelial cell model was established and FOXO3 was proved to be targeted by miR-27a-3p, the miR-27a-3p mimic, inhibitor, or FOXO3-overexpression plasmids were transfected into the cells. The effects of miR-27a-3p and FOXO3 on cell viability and apoptosis were then evaluated. The levels of apoptosis-/inflammation-related factors, miR-27a-3p, and FOXO3 were further analyzed. Also, the activities of reactive oxygen species (ROS) and nicotinamide adenine dinucleotide phosphate (NAPDH) in cells were examined. RESULTS:MiR-27a-3p was down-regulated in LPS-induced alveolar epithelial cells. The decreased-cell viability of the LPS-induced cells was increased by miR-27a-3p mimic while inhibited by FOXO3. The enhanced-apoptosis, and up-regulated Bax and C caspase-3 were reduced by miR-27a-3p mimic while inhibited by FOXO3; the down-regulated Bcl-2 of the LPS-induced cells was increased by miR-27a-3p mimic while inhibited by FOXO3. The up-regulated IL-6, IL-8, ROS, and NAPDH in the LPS-induced cells were reduced by miR-27a-3p mimic while inhibited by FOXO3. Besides, FOXO3 reversed the effect of miR-27a-3p mimic on the LPS-induced cells. CONCLUSION:MiR-27a-3p targeted FOXO3 to mitigated inflammation and apoptosis of LPS-induced alveolar epithelial cells via suppressing NAPDH/ROS activation. 10.1016/j.bbrc.2020.07.126
Circ_0010729 regulates hypoxia-induced cardiomyocyte injuries by activating TRAF5 via sponging miR-27a-3p. Lei Dazhou,Wang Yan,Zhang Luochao,Wang Zhifang Life sciences Ischemic cardiomyopathy is a severe cardiovascular disease with high mortality. Circular RNAs (circRNAs) are widely regulated in diverse human diseases, including Ischemic cardiomyopathy. This study aimed to investigate a novel functional mechanism of circRNA circ_0010729 in hypoxia-induced cardiomyocyte injuries. Human cardiomyocytes (AC16) were exposed to hypoxia to mimic ischemic cardiomyopathy in vitro. Cell viability, apoptosis/necrosis and glycolysis progress, were determined using 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay, flow cytometry assay and glycolysis stress test, respectively. Cell apoptosis was also assessed by the activity of cleaved caspase-3/7. The levels of glycolysis-related proteins and tumor necrosis factor receptor-associated factor 5 (TRAF5) were examined by western blot. The expression of circ_0010729 and miR-27a-3p was measured by real-time quantitative reverse transcription-polymerase chain reaction (qRT-PCR). The prediction about the targeted relationship was verified by dual-luciferase reporter assay, RNA immunoprecipitation (RIP) assay and RNA pull-down assay. As a result, hypoxia treatment inhibited cell viability, induced cell apoptosis and blocked glycolysis, however, these injuries were alleviated by circ_0010729 knockdown. MiR-27a-3p was targeted by circ_0010729, and miR-27a-3p inhibition reversed the role of circ_0010729 knockdown, leading to the deterioration of cell injuries. Further, TRAF5 was a target of miR-27a-3p, and circ_0010729 upregulated the expression of TRAF5 by sponging miR-27a-3p. MiR-27a-3p restoration enhanced cell viability, depleted cell apoptosis and promoted glycolysis of hypoxia-induced AC16 cells, while these effects were abolished by TRAF5 overexpression. In conclusion, circ_0010729 knockdown alleviated hypoxia-induced AC16 cell injuries by mediating the miR-27a-3p/TRAF5 axis. 10.1016/j.lfs.2020.118511
Dexmedetomidine inhibits inflammatory response and autophagy through the circLrp1b/miR-27a-3p/Dram2 pathway in a rat model of traumatic brain injury. Li Hengchang,Lu Chengxiang,Yao Wenfei,Xu Lixin,Zhou Jun,Zheng Bin Aging Circular RNAs (circRNAs) have a regulatory function on inflammation and autophagy, of which rno-circRNA_010705 (circLrp1b) appears to be significantly up-regulated following traumatic brain injury (TBI). Dexmedetomidine (DEX) shows improvement effects in TBI by inhibiting NLRP3/caspase-1. However, whether circLrp1b plays critical roles in DEX-mediated TBI attenuation and the underlying mechanisms remain unclear. After TBI was established in rats by controlled cortical impact (CCI) to cause brain trauma, they received an intracerebroventricular injection of lentiviral vector, followed by intraperitoneal injection of DEX. Administration of DEX ameliorated autophagy in rats following TBI, accompanied by up-regulated circLrp1b and Dram2 and down-regulated miR-27a-3p. DEX promoted the effects of circLrp1b in attenuating TBI-induced neurologic impairment, autophagy, and inflammation, which was significantly reversed by inhibition of miR-27a-3p or Dram2 overexpression. Mechanistically, northern blot and luciferase reporter assays indicated that circLrp1b up-regulated Dram2 expression by functioning as a sponge for miR-27a-3p to promote autophagy involved in TBI, which was reversed by DEX treatment. Collectively, this study demonstrated that DEX inhibits inflammatory response and autophagy involved in TBI through inactivation of the circLrp1b/miR-27a-3p/Dram2 signaling pathway. 10.18632/aging.103975
Circular RNA circ_HECTD1 regulates cell injury after cerebral infarction by miR-27a-3p/FSTL1 axis. Cell cycle (Georgetown, Tex.) Cerebral infarction is a common cerebrovascular disease caused by neural cell injury, with high mortality worldwide. Circular RNAs HECT domain E3 ubiquitin-protein ligase 1 (circ_HECTD1) has been reported to be related to the oxygen-glucose deprivation/reperfusion (OGD/R)-caused neuronal damage in cerebral ischemia. This study is designed to explore the role and mechanism of circ_HECTD1 in OGD/R-induced cell injury in cerebral ischemia. Circ_HECTD1, microRNA-27a-3p (miR-27a-3p), and Follistatin-like 1 (FSTL1) level were detected by real-time quantitative polymerase chain reaction (RT-qPCR). The localization of circ_HECTD1 was analyzed by subcellular fractionation assay. Cell proliferative ability and apoptosis were assessed by 5-ethynyl-2'-deoxyuridine (EdU), 3-(4, 5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide (MTT), and flow cytometry assays. The protein levels of proliferating cell nuclear antigen (PCNA), B-cell lymphoma-2 (Bcl-2), Bcl-2 related X protein (Bax), Cleaved poly-ADP-ribose polymerase (PARP), and FSTL1 were examined by western blot assay. The binding relationship between miR-27a-3p and circ_HECTD1 or FSTL1 was predicted by starbase 3.0 then verified by a dual-luciferase reporter assay. Circ_HECTD1 and FSTL1 were highly expressed, and miR-27a-3p was decreased in OGD/R-treated HT22 cells. Moreover, circ_HECTD1 knockdown could boost cell proliferative ability and repress apoptosis in OGD/R-triggered HT22 cells . Mechanical analysis discovered that circ_HECTD1 could regulate FSTL1 expression by sponging miR-27a-3p. Circ_HECTD1 deficiency could mitigate OGD/R-induced HT22 cell damage by modulating the miR-27a-3p/FSTL1 axis, providing a promising therapeutic target for cerebral infarction treatment. 10.1080/15384101.2021.1909885
MiR-27a-3p suppresses cerebral ischemia-reperfusion injury by targeting FOXO1. Li Wenyu,Zhu Qiongbin,Xu Xiaoyan,Hu Xingyue Aging Cerebral ischemia-reperfusion (CI/R) injury is a serious complication when treating patients experiencing ischemic stroke. Although the microRNA miR-27a-3p reportedly participates in ischemia/reperfusion (I/R) injury, its actions in CI/R remain unclear. To mimic CI/R , HT22 cells were subjected to oxygen glucose deprivation/reoxygenation (OGD/R). The results indicate that OGD inhibited growth and induced apoptosis among HT22 cells. The apoptosis was accompanied by increases in activated caspases 3 and 9 and decreases in Bcl-2. Oxidative stress was also increased, as indicated by increases in ROS and malondialdehyde and decreases in glutathione and superoxide dismutase. In addition, OGD induced G1 arrest in HT22 cells with corresponding upregulation of FOXO1 and p27 Kip1, suggesting the cell cycle arrest was mediated by FOXO1/p27 Kip1 signaling. Notably, FOXO1 was found to be the direct target of miR-27a-3p in HT22 cells. MiR-27a-3p was downregulated in OGD/R-treated HT22 cells, and miR-27a-3p mimics partially or entirely reversed all of the effects of OGD. Moreover, miR-27a-3p agomir significantly alleviated the symptoms of CI/R in a rat model of CI/R. Thus, MiR-27a-3p appears to suppress CI/R injury by targeting FOXO1. 10.18632/aging.202866
Biliverdin Protects Against Cerebral Ischemia/Reperfusion Injury by Regulating the miR-27a-3p/Rgs1 Axis. Li Junjie,Peng Lijia,Bai Wenya,Peng Peihua,Chen Wendong,Yang Wei,Shao Jianlin Neuropsychiatric disease and treatment Background:We have previously demonstrated that biliverdin has neuroprotective effects that ameliorate cerebral ischemia/reperfusion (I/R) injury in rats. However, the underlying mechanism is unknown. This study aimed at elucidating on the modulatory role of miR-27a-3p on Rgs1 as a mechanism by which biliverdin affects cerebral I/R injury. Methods:Middle cerebral artery occlusion/reperfusion (MCAO/R) was used to establish I/R rat models while oxygen glucose deprivation/reoxygenation (OGD/R) was used to induce hippocampal neurons to establish I/R models in vitro. Infarct volume was assessed by TTC staining. Apoptotic analyses of ischemic cortical neurons and cells were performed by TUNEL staining and flow cytometry, respectively. Cell viability was assessed by the CCK-8 assay while the target of miR-27a-3p was determined by double luciferase reporter assay. Relative expression levels of miR-27a-3p and Rgs1 (in vivo and in vitro) as well as markers of inflammation and apoptosis (in vitro) were detected by RT-qPCR. Then, Elisa and western blot were used to assess protein expression levels of inflammatory and apoptotic markers in vitro. Results:Biliverdin suppressed inflammation and apoptosis in hippocampal neurons upon OGD/R, and reduced cerebral infarction volume as well as apoptosis in the MCAO/R rat model. Furthermore, biliverdin upregulated miR-27a-3p and downregulated hippocampal neuron Rgs1 after OGD/R as well as in rat brain tissues after cerebral I/R. Bioinformatic analysis revealed an miR-27a-3p docking site in the 3'-UTR region of Rgs1. Luciferase reporter assays showed that Rgs1 is an miR-27a-3p target. Moreover, miR-27a-3p upregulation inhibited OGD/R-triggered inflammation and suppressed neuronal apoptosis. Rgs1 knockdown suppressed OGD/R-triggered inflammation and decreased neuronal apoptosis while miR-27a-3p downregulation reversed the protective effect of Rgs1 knockdown. Moreover, miR-27a-3p overexpression and Rgs1 silencing suppressed NF-κB (p65) expression. Conclusion:Biliverdin protects against cerebral I/R injury by regulating the miR-27a-3p/Rgs1 axis, thereby inhibiting inflammation and apoptosis. 10.2147/NDT.S300773
Linc-KIAA1737-2 promoted LPS-induced HK-2 cell apoptosis by regulating miR-27a-3p/TLR4/NF-κB axis. Hu Ming,Wei Jing,Yang Liu,Xu Jianhua,He Zhaofeng,Li Haiyuan,Ning Chao,Lu Shijun Journal of bioenergetics and biomembranes Inflammation and renal cell apoptosis participate in sepsis-induced acute kidney injury. Previous research found the upregulation of long non-coding RNA Linc-KIAA1737-2 in hypoxia- or inflammation-challenged human proximal tubular epithelial cells, but its role in sepsis-induced acute kidney injury is underexplored. In this research, we found that Linc-KIAA1737-2 could be upregulated in HK-2 human proximal tubular epithelial cells by LPS treatment, and knock-down of this lncRNA significantly attenuated LPS-induced apoptosis in HK-2 cells, while its overexpression showed opposite effect. MiR-27a-3p was confirmed to interact with Linc-KIAA1737-2 in HK-2 cells by RNA pull-down and dual-luciferase assay. MiR-27a-3p mimic transfection significantly attenuated LPS-induced HK-2 cell apoptosis by downregulating the protein levels of TLR4 and NF-κB, which was overturned by overexpression of Linc-KIAA1737-2. Our results suggested that Linc-KIAA1737-2 could promote LPS-induced apoptosis in HK-2 cells, and presumably sepsis-induced acute kidney injury, by regulating the miR-27a-3p/TLR4/NF-κB axis. 10.1007/s10863-021-09897-1
A novel identified circ-ANKHD1 targets the miR-27a-3p/SFRP1 signaling pathway and modulates the apoptosis of granulosa cells. Li Xiaoyan,Gao Fenglei,Fan Yushan,Xie Shefeng,Li Chengde,Meng Li,Li Li,Zhang Shouquan,Wei Hengxi Environmental science and pollution research international The specific expression profile and function of circular RNAs (circRNAs) in mammalian ovarian follicles, especially during the atresia process, are unclear. In this study, we verified and explored the expression and function of circ-ANKHD1 in granulosa cells. Our results showed that abundance of circ-ANKHD1 was significantly lower in the granulosa cells than that of ANKHD1. The expression of ANKHD1 was highest in the granulosa cells from follicles with a diameter of 5-6 mm and lowest in that with a diameter of 3-4 mm. Furthermore, the expression level of circ-ANKHD1 in the ovarian tissue of 1-day-old piglets was significantly higher than that of 17-month-old multiparous sows. The luciferase reporter assay showed the potential interaction between circ-ANKHD1 and miR-27a-3p/miR-142-5p. Furthermore, circ-ANKHD1 overexpression up-regulated SFRP1 expression, while miR-27a-3p overexpression suppressed SFRP1 expression in granulosa cells. Circ-ANKHD1 overexpression significantly decreased the cell apoptotic rates of the granulosa cells and repressed the cell population at G0/G1 and S phases but increased cell population at G2/M phase. Finally, circ-ANKHD1 overexpression increased the mRNA expression levels of Bcl-2 and cyclin D1 in the granulosa cells, while there are no effects on the mRNA expression levels of caspase-3, p53, Bax, and proliferating cell nuclear antigen. In conclusion, our study for the first time identified a novel circRNA, circ-ANKHD1 that may be associated with the biological functions of granulosa cells. Circ-ANKHD1 may promote the granulosa cell proliferation, but attenuate apoptosis, and these effects may be associated with modulation of miR-27a-3p/SFRP1. 10.1007/s11356-021-14699-4
MiR-27a-3p/Hoxa10 Axis Regulates Angiotensin II-Induced Cardiomyocyte Hypertrophy by Targeting Kv4.3 Expression. Cao Xuefeng,Zhang Zheng,Wang Yu,Shan Weichao,Wang Ruiting,Mao Shufang,Ding Shi,Pang Chong,Li Baoqun,Zhou Jian,Guo Xiaoyan,Guo Na,Li Cui,Liang Jing,Ma Wenya,Liu Yu,Zhao Liang Frontiers in pharmacology Cardiac hypertrophy is a common pathological process of various cardiovascular diseases, which is often accompanied with structural and electrical remodeling, and can even lead to sudden cardiac death. However, its molecular mechanism still remains largely unknown. Here, we induced cardiomyocyte hypertrophy by angiotensin II (Ang II), and found that miR-27a-3p and hypertrophy-related genes were up-regulated. Further studies showed that miR-27a-3p-inhibitor can alleviate myocardial hypertrophy and electrical remodeling. Moreover, luciferase assay confirmed that miR-27a-3p could regulate the expression of downstream at the transcriptional level by targeting at its 3'UTR. At the same time, the protein expression of Hoxa10 was significantly reduced in Ang II-treated cardiomyocytes. Furthermore, overexpression of can reverse myocardial hypertrophy and electrical remodeling induced by Ang II in cardiomyocytes. Finally, we found that Hoxa10 positively regulated the expression of potassium channel protein Kv4.3 which was down-regulated in hypertrophic cardiomyocytes. Taken together, our results revealed miR-27a-3p/Hoxa10/Kv4.3 axis as a new mechanism of Ang II-induced cardiomyocyte hypertrophy, which provided a new target for clinical prevention and treatment of cardiac hypertrophy and heart failure. 10.3389/fphar.2021.680349
HPV16 E6 enhances the radiosensitivity in HPV-positive human head and neck squamous cell carcinoma by regulating the miR-27a-3p/SMG1 axis. Long Dan,Xu Li,Deng Zeyi,Guo Dandan,Zhang Yangchun,Liu Zhaohui,Zhang Chunlin Infectious agents and cancer BACKGROUND:Head and neck squamous cell carcinoma (HNSCC) is the 6th most common malignant cancer type worldwide. Radiosensitivity has been shown to be significantly increased in patients with human papillomavirus (HPV)-positive HNSCC compared with HPV-negative patients. However, the clinical significance of HPV and its regulatory mechanisms in HNSCC are largely unknown. The aim of our study was to explore the regulatory mechanism of miR-27a-3p in the radiosensitivity of HPV-positive HNSCC cells. METHODS:E6-overexpressing and E6-knockdown HNSCC cell lines were generated and the transfection efficiencies were evaluated by quantitative real-time PCR (RT-qPCR) and western blotting. The expression of miR-27a-3p and DiGeorge syndrome critical region 8 (DGCR8) was examined by RT-qPCR after transfection with E6 overexpressing plasmid or E6 siRNA. The effects of miR-27a-3p on the radiosensitivity of HNSCC cells were explored by a colony formation and TUNEL staining assays. Bioinformatic tools and luciferase reporter assays were used to identify that SMG1 is the direct target of miR-27a-3p. Furthermore, the effect of E6 overexpression on the regulation of the miR-27a-3p/SMG1 axis was investigated. RESULTS:In our study, we found overexpression of HPV E6 upregulated the expression of DGCR8 and miR-27a-3p in HNSCC cells. We next confirmed that DGCR8 positively regulated the expression of miR-27a-3p in HNSCC cells. The luciferase reporter gene results verified that miR-27a-3p targeted the 3'UTR of SMG1 mRNA. MiR-27a-3p mimics transfection resulted in a decrease in SMG1 expression and miR-27a-3p inhibitor transfection increased SMG1 expression. Apoptotic activity of HNSCC cells was significantly increased in miR-27a-3p mimics HNSCC cells compared with control HNSCC cells. After treatment with 4 Gy irradiation, UM-SCC47 cells transfected with miR-27a-3p inhibitor or SMG1 overexpressing plasmid formed more colonies than the corresponding control cells. Furthermore, the rescue experiments demonstrated that HPV16 E6 improved the radiosensitivity of HNSCC cells by targeting miR-27a-3p/SMG1. CONCLUSION:Our study demonstrated that HPV16 E6 activated the DGCR8/miR-27a-3p/SMG1 axis to enhance the radiosensitivity. Our findings might provide a novel therapeutic target to improve the response of HNSCC to radiotherapy. 10.1186/s13027-021-00397-w
Sevoflurane inhibits the apoptosis of hypoxia/reoxygenation-induced cardiomyocytes via regulating miR-27a-3p-mediated autophagy. Zhang Yang,Zhan Biming,Hu Ying,Chen Shibiao,Zhang Qin The Journal of pharmacy and pharmacology INTRODUCTION:Sevoflurane (Sevo) prevents hypoxia/reoxygenation (H/R)-induced cardiomyocytes apoptosis. MiR-27a-3p expression is up-regulated in Sevo-treated hippocampal neurons. OBJECTIVE:This study explored whether the effect of Sevo on cardiomyocytes was mediated by miR-27a-3p. METHODS:The cardiomyocytes were cultured under H/R condition or pre-treated with Sevo, and further transfected with miR-27a-3p inhibitor or treated with an autophagy inhibitor 3-methyladenine (3-MA). Then the cell morphology was observed under an optical microscope. The cell viability and apoptosis were measured by MTT and flow cytometry. Expressions of miR-27a-3p, apoptosis-related, and autophagy-related factors were determined by western blot or RT-qPCR. KEY FINDINGS:Sevo improved the abnormal morphology, promoted the cell viability and the expressions of Bcl-2 and miR-27a-3p, but reduced the apoptosis and Bax and C-caspase-3 levels of H/R-induced cardiomyocytes. MiR-27a-3p inhibitor had an effect opposite to Sevo on the cardiomyocytes and further counteracted the effect of Sevo on the H/R-induced cardiomyocytes. Downregulation miR-27a-3p increased the expression of Beclin 1 and the ratio of LC3B-II to LC3B-I in H/R-induced cardiomyocytes. Furthermore, 3-MA had an opposite effect to miR-27a-3p inhibitor and further counteracted the effect of the miR-27a-3p inhibitor on H/R-induced cardiomyocytes. CONCLUSION:Sevo inhibited the apoptosis of H/R-induced cardiomyocytes via regulating miR-27a-3p-mediated autophagy. 10.1093/jpp/rgab111
MiR-27a-3p enhances the cisplatin sensitivity in hepatocellular carcinoma cells through inhibiting PI3K/Akt pathway. Yang Ying,Yang Zhifang,Zhang Ruili,Jia Chunli,Mao Rui,Mahati Shaya,Zhang Yuefen,Wu Ge,Sun Yan Na,Jia Xiao Yan,Aimudula Ainiwaer,Zhang Hua,Bao Yongxing Bioscience reports MicroRNAs (miRNAs) play an important role in drug resistance, and it is reported that miR-27a-3p regulated the sensitivity of cisplatin in breast cancer, lung cancer and ovarian cancer. However, the relationship between miR-27a-3p and chemosensitivity of cisplatin in hepatocellular carcinoma (HCC) was unclear, especially the underlying mechanism was unknown. In the present study, we analyzed miR-27a-3p expression levels in 372 tumor tissues and 49 adjacent tissues in HCC samples from TCGA database, and found that the miR-27a-3p was down-regulated in HCC tissues. The level of miR-27a-3p was associated with metastasis, Child-Pugh grade and race. MiR-27a-3p was regarded as a favorable prognosis indicator for HCC patients. Then, miR-27a-3p was overexpressed in HepG2 cell, and was knocked down in PLC cell. Next, we conducted a series of in vitro assays, including MTT, apoptosis and cell cycle assays to observe the biological changes. Further, inhibitor rate and apoptosis rate were detected with pre- and post-cisplatin treatment in HCC. The results showed that overexpression of miR-27a-3p repressed the cell viability, promoted apoptosis and increased the percentage of cells in G0/G1 phase. Importantly, overexpression of miR-27a-3p significantly increased the inhibitor rate and apoptosis rate with cisplatin intervention. Besides, we found that miR-27a-3p added cisplatin sensitivity potentially through regulating PI3K/Akt signaling pathway. Taken together, miR-27a-3p acted as a tumor suppressor gene in HCC cells, and it could be useful for modulating cisplatin sensitivity in chemotherapy. 10.1042/BSR20192007
Signature of circulating microRNAs in patients with acute heart failure. Ovchinnikova Ekaterina S,Schmitter Daniela,Vegter Eline L,Ter Maaten Jozine M,Valente Mattia A E,Liu Licette C Y,van der Harst Pim,Pinto Yigal M,de Boer Rudolf A,Meyer Sven,Teerlink John R,O'Connor Christopher M,Metra Marco,Davison Beth A,Bloomfield Daniel M,Cotter Gadi,Cleland John G,Mebazaa Alexandre,Laribi Said,Givertz Michael M,Ponikowski Piotr,van der Meer Peter,van Veldhuisen Dirk J,Voors Adriaan A,Berezikov Eugene European journal of heart failure AIMS:Our aim was to identify circulating microRNAs (miRNAs) associated with acute heart failure (AHF). METHODS AND RESULTS:Plasma miRNA profiling included 137 patients with AHF from 3 different cohorts, 20 with chronic heart failure (CHF), 8 with acute exacerbation of COPD, and 41 healthy controls. Levels of circulating miRNAs were measured using quantitative reverse transcription-polymerase chain reaction (qRT-PCR). Plasma levels of miRNAs in patients with AHF were decreased compared with CHF patients or healthy subjects, whereas no significant changes were observed between acute COPD patients and controls. Fifteen miRNAs found in the discovery phase to differ most significantly between healthy controls and patients with AHF were further investigated in an extended cohort of 100 AHF patients at admission and a separate cohort of 18 AHF patients at different time points. Out of these 15 miRNAs, 12 could be confirmed in an additional AHF validation cohort and 7 passed the Bonferroni correction threshold (miR-18a-5p, miR-26b-5p, miR-27a-3p, miR-30e-5p, miR-106a-5p, miR-199a-3p, and miR-652-3p, all P < 0.00005). A further drop in miRNA levels within 48 h after AHF admission was associated with an increased risk of 180-day mortality in a subset of the identified miRNAs. CONCLUSIONS:Declining levels of circulating miRNAs were associated with increasing acuity of heart failure. Early in-hospital decreasing miRNA levels were predictive for mortality in a subset of miRNAs in patients with AHF. The discovered miRNA panel may serve as a launch-pad for molecular pathway studies to identify new pharmacological targets and miRNA-based therapies. 10.1002/ejhf.332
Effect of Green and Brown Propolis Extracts on the Expression Levels of microRNAs, mRNAs and Proteins, Related to Oxidative Stress and Inflammation. Nutrients A large body of evidence highlights that propolis exerts many biological functions that can be ascribed to its antioxidant and anti-inflammatory components, including different polyphenol classes. Nevertheless, the molecular mechanisms are yet unknown. The aim of this study is to investigate the mechanisms at the basis of propolis anti-inflammatory and antioxidant activities. The effects of two brown and green propolis extracts-chemically characterized by RP-HPLC-PDA-ESI-MSn-on the expression levels of miRNAs associated with inflammatory responses (miR-19a-3p and miR-203a-3p) and oxidative stress (miR-27a-3p and miR-17-3p), were determined in human keratinocyte HaCat cell lines, treated with non-cytotoxic concentrations. The results showed that brown propolis, whose major polyphenolic components are flavonoids, induced changes in the expression levels of all miRNAs, and was more active than green propolis (whose main polyphenolic components are hydroxycinnamic acid derivatives) which caused changes only in the expression levels of miR-19a-3p and miR-27a-3p. In addition, only brown propolis was able to modify (1) the expression levels of mRNAs, the target of the reported miRNAs, which code for Tumor Necrosis Factor-α (TNF-α), Nuclear Factor, Erythroid 2 Like 2 (NFE2L2) and Glutathione Peroxidase 2 (GPX2), and (2) the protein levels of TNF-α and NFE2L2. In conclusion, brown and green propolis, which showed different metabolite profiles, exert their biological functions through different mechanisms of action. 10.3390/nu9101090
LncRNA NEAT1 promotes hypoxia-induced renal tubular epithelial apoptosis through downregulating miR-27a-3p. Jiang Xinxin,Li Daoting,Shen Wei,Shen Xiaogang,Liu Yueming Journal of cellular biochemistry Acute kidney injury (AKI) is a common kidney disorder that affects public health and the incidence of AKI. Sepsis, acute ischemia or hypoxia is the main reason for the occurrence of AKI. Recently, noncoding RNA that include microRNA and long noncoding RNA (lncRNAs) were reported to play important roles in AKI as well as have the potential to serve as a biomarker or therapeutic target for the development of the diagnostic and prognostic strategies of AKI. In the current study, we aimed to investigate the expression and biological function of lncRNA nuclear enriched abundant transcript 1 (NEAT1) in ischemia-induced AKI in patients' sample and in vitro. The expressions of NEAT1 and miR-27a-3p in ischemia/reperfusion-induced AKI patients were examined by quantitative reverse transcription polymerase chain reaction. Cell injury was induced by treatment of human kidney tubular cells (HK-2) with CoCl . After treatment, the influences of NEAT1 and miR-27a-3p on the cell apoptosis in the CoCl -stimulated HK-2 were tested by flow cytometry. The flow analysis results showed that the expression of NEAT1 was markedly higher in the ischemia-induced AKI patients compared with normal control. Moreover, repression the expression of NEAT1 decreased CoCl -induced injury in HK-2. The expression of miR-27a-3p was negatively regulated by NEAT1. Inhibition the expression of NEAT1 attenuated overexpression of miR-27a-3p enhanced CoCl -induced injury. In summary, an ischemia-induced injury may be enhanced by a high level of NEAT1 through targeting miR-27a-3p. Thus, NEAT1 has the potential to be explored as a biomarker for diagnosis and target for therapeutic strategies in ischemia-induced AKI. 10.1002/jcb.28909
EIF4A3-Induced circ-BNIP3 Aggravated Hypoxia-Induced Injury of H9c2 Cells by Targeting miR-27a-3p/BNIP3. Li Yansong,Ren Shuhong,Xia Jingwen,Wei Yong,Xi Yinhua Molecular therapy. Nucleic acids Acute myocardial infarction (AMI) results from long-term diminished blood supply diminishment (ischemia) to the heart, and the main reason for ischemia is hypoxia. BCL2 interaction protein 3 (BNIP3) can be upregulated by hypoxia and participates in the mediation of hypoxia-activated apoptosis in cardiac myocyte death. The purpose of this study was to interrogate the mechanism of BNIP3 in hypoxia-activated cardiac myocyte injury. Cell viability and apoptosis were evaluated by Cell counting kit 8 (CCK-8), 5-ethynyl-2'-deoxyuridine (EdU), TdT-mediated dUTP Nick-End Labeling (TUNEL), and caspase-3 activity assays. Molecular interactions were assessed by RNA immunoprecipitation (RIP) and pull-down assays. Gene levels were assessed via quantitative real-time PCR and western blot. BNIP3 expression was upregulated by hypoxia in H9c2 cells. We found that circ-BNIP3 (hsa_circ_0005972), whose annotated gene was BNIP3, was induced by hypoxia and positively regulated BNIP3 expression. Knockdown of BNIP3 or circ-BNIP3 reversed the effect of hypoxia in attenuating H9c2 cell viability and inducing apoptosis. circ-BNIP3 sponged miRNA-27a-3p (miR-27a-3p) to upregulate BNIP3 expression. Moreover, eukaryotic translation initiation factor 4A3 (EIF4A3) bound with the upstream region of the circ-BNIP3 mRNA transcript and induced circ-BNIP3 expression in H9c2 cells. EIF4A3-induced circ-BNIP3 aggravated hypoxia-caused injury of H9c2 cells through targeting miR-27a-3p/BNIP3 pathway, indicating circ-BNIP3 as a new target for relieving hypoxia-induced injury of cardiac myocytes. 10.1016/j.omtn.2019.11.017
Long Noncoding RNA SOX2-OT Exacerbates Hypoxia-Induced Cardiomyocytes Injury by Regulating miR-27a-3p/TGFR1 Axis. Cardiovascular therapeutics BACKGROUND:Myocardial infarction (MI) was a severe cardiovascular disease resulted from acute, persistent hypoxia, or ischemia condition. Additionally, MI generally led to heart failure, even sudden death. A multitude of research studies proposed that long noncoding RNAs (lncRNAs) frequently participated in the regulation of heart diseases. The specific function and molecular mechanism of SOX2-OT in MI remained unclear. . The current research was aimed to explore the role of SOX2-OT in MI. METHODS:Bioinformatics analysis (DIANA tools and Targetscan) and a wide range of experiments (CCK-8, flow cytometry, RT-qPCR, luciferase reporter, RIP, caspase-3 activity, trans-well, and western blot assays) were adopted to investigate the function and mechanism of SOX2-OT. RESULTS:We discovered that hypoxia treatment decreased cell viability but increased cell apoptosis. Besides, lncRNA SOX2-OT expression was upregulated in hypoxic HCMs. Hereafter, we confirmed that SOX2-OT could negatively regulate miR-27a-3p levels by directly binding with miR-27a-3p, and miR-27a-3p also could negatively regulate SOX2-OT levels. Furthermore, knockdown of SOX2-OT promoted cell proliferation, migration, and invasion, but limited cell apoptosis. However, these effects were reversed by anti-miR-27a-5p. Besides, we verified that miR-27a-3p binding with the 3'UTR of TGFBR1 and SOX2-OT regulated TGFR1 level by collaborating with miR-27a-3p in HCMs. Eventually, rescue assays validated that the influence of SOX2-OT silence or miR-27a-3p overexpression on cellular processes in cardiomyocytes injury was counteracted by TGFBR1 overexpression. CONCLUSIONS:Long noncoding RNA SOX2-OT exacerbated hypoxia-induced cardiomyocytes injury by regulating miR-27a-3p/TGFR1 axis, which may provide a novel insight for heart failure treatment. 10.1155/2020/2016259
miRNA-27a-3p and miRNA-222-3p as Novel Modulators of Phosphodiesterase 3a (PDE3A) in Cerebral Microvascular Endothelial Cells. Yasmeen S,Kaur S,Mirza A H,Brodin B,Pociot F,Kruuse C Molecular neurobiology Endothelial dysfunction is a key element in cerebral small vessel disease (CSVD), which may cause stroke and cognitive decline. Cyclic nucleotide signaling modulates endothelial function. The cyclic adenosine monophosphate-degrading enzyme phosphodiesterase 3 (PDE3) is an important treatment target which may be modulated by microRNAs (miRNAs) important for regulating gene expression. We aimed to identify PDE3-targeting miRNAs to highlight potential therapeutic targets for endothelial dysfunction and CSVD. PDE3-targeting miRNAs were identified by in silico analysis (TargetScan, miRWalk, miRanda, and RNA22). The identified miRNAs were ranked on the basis of TargetScan context scores and their expression (log2 read counts) in a human brain endothelial cell line (hCMEC/D3) described recently. miRNAs were subjected to co-expression meta-analysis (CoMeTa) to create miRNA clusters. The pathways targeted by the miRNAs were assigned functional annotations via the KEGG pathway and COOL. hCMEC/D3 cells were transfected with miRNA mimics miR-27a-3p and miR-222-3p, and the effect on PDE3A protein expression was analyzed by Western blotting. Only PDE3A is expressed in hCMEC/D3 cells. The in silico prediction identified 67 PDE3A-related miRNAs, of which 49 were expressed in hCMEC/D3 cells. Further analysis of the top two miRNA clusters (miR-221/miR-222 and miR-27a/miR-27b/miR-128) indicated a potential link to pathways relevant to cerebral and vascular integrity and repair. hCMEC/D3 cells transfected with miR-27a-3p and miR-222-3p mimics had reduced relative expression of PDE3A protein. PDE3A-related miRNAs miR-221/miR-222 and miR-27a/miR-27b/miR-128 are potentially linked to pathways essential for immune regulation as well as cerebral and vascular integrity/function. Furthermore, relative PDE3A protein expression was reduced by miR27a-3p and miR-222-3p. 10.1007/s12035-018-1446-5
MicroRNA-27a-3p Downregulation Inhibits Inflammatory Response and Hippocampal Neuronal Cell Apoptosis by Upregulating Mitogen-Activated Protein Kinase 4 (MAP2K4) Expression in Epilepsy: In Vivo and In Vitro Studies. Lu Jun,Zhou Nina,Yang Ping,Deng Lanqiuzi,Liu Ganzhe Medical science monitor : international medical journal of experimental and clinical research BACKGROUND This study aimed to discover the effect and mechanism of microRNA-27a-3p (miR-27a-3p) in epilepsy. MATERIAL AND METHODS To perform our investigation, in vivo and in vitro models of epilepsy were induced using kainic acid (KA). Expression of miR-27a-3p in the hippocampus of epileptic rats or normal rats or neuronal cells was detected using quantitative reverse transcription polymerase chain reaction (qRT-PCR). Racine score was used to assess seizures in epileptic rats. Cell viability and cell apoptosis were analyzed by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assay and flow cytometry. Enzyme-linked immunosorbent assay (ELISA) was performed to detect inflammatory factors expression. RESULTS Significantly higher expression of miR-27a-3p in the hippocampus of epileptic rats and in KA-induced neurons was observed. We found that miR-27a-3p inhibitor alleviated seizures in epileptic rats. miR-27a-3p inhibitor also inhibited apoptosis of hippocampal neurons in epileptic rats, promoted Bcl2 expression, and decreased Bax and Caspase3 expression. The results showed that miR-27a-3p inhibitor effectively reduced the expression levels of interleukin-1ß (IL-1ß), IL-6, and tumor necrosis factor-alpha (TNF-alpha) in hippocampal tissues of epileptic rats. Dual luciferase reporter assay showed that mitogen-activated protein kinase 4 (MAP2K4) was a direct target of miR-27a-3p. miR-27a-3p inhibitor significantly promoted the cell viability of KA-induced neurons, inhibited cell apoptosis, promoted the expression of Bcl-2, and decreased Bax and Caspase3 expression, and all these changes were abolished by MAP2K4-siRNA co-transfection. CONCLUSIONS Our preliminary findings indicated that miR-27a-3p inhibitor protected against epilepsy-induced inflammatory response and hippocampal neuronal apoptosis by targeting MAP2K4. 10.12659/MSM.916458
MicroRNA-27a-3p aggravates renal ischemia/reperfusion injury by promoting oxidative stress via targeting growth factor receptor-bound protein 2. Zhao X-R,Zhang Z,Gao M,Li L,Sun P-Y,Xu L-N,Qi Y,Yin L-H,Peng J-Y Pharmacological research Renal ischemia-reperfusion (RI/R) injury with high morbidity and mortality is one common clinical disease. Development of drug targets to treat the disorder is critical important. MiR-27a-3p plays important roles in regulating oxidative stress. However, its effects on RI/R injury have not been reported. In this paper, hypoxia/reoxygenation (H/R) models on NRK-52E and HK-2 cells, and RI/R model in C57BL/6 mice were established. The results showed that H/R in vitro decreased cell viability and increased ROS levels in cells, and RI/R caused renal injury and oxidative damage in mice. The expression levels of miR-27a-3p were up-regulated based on real-time PCR and FISH assays in model groups compared with control groups, which directly targeted Grb2 based on dual luciferase reporter assay and co-transfaction test. In addition, miR-27a- 3p markedly reduced Grb2 expression to down-regulate the expression levels of p-PI3K, p-AKT, Nrf2, HO-1, and up-regulate Keap1 expression in model groups. MiR-27a-3p mimics in vitro enhanced H/R-caused oxidative stress via increasing ROS levels and decreasing Grb2 expression to down-regulate PI3K-AKT signal. In contrary, miR-27a-3p inhibitor in vitro significantly reduced H/R-caused oxidative damage via decreasing ROS levels and increasing Grb2 expression to up-regulate PI3K-AKT signal. In vivo, miR-27a- 3p agomir exacerbated RI/R-caused renal damage by decreasing SOD level and increasing Cr, BUN, MDA levels via suppressing Grb2 expression to down-regulate PI3K- AKT signal. However, miR-27a -3p antagomir alleviated RI/R-caused oxidative damage via increasing Grb2 expression to up-regulate PI3k-AKT signal. Grb2siRNA in mice further enhanced RI/R-caused renal injury by increasing Cr, BUN, MDA levels and decreasing SOD level via inhibiting the expression levels of Grb2, Nrf2, HO-1, and increasing Keap1 expression. Our data showed that miR-27a-3p aggravated RI/R injury by promoting oxidative stress via targeting Grb2, which should be considered as one new drug target to treat RI/R injury. 10.1016/j.phrs.2020.104718
MicroRNA-27a-3p Reverses Adriamycin Resistance by Targeting BTG2 and Activating PI3K/Akt Pathway in Breast Cancer Cells. OncoTargets and therapy AIM:This study aimed to explore the regulative mechanisms of miR-27a-3p in chemo-resistance of breast cancer cells. MATERIALS AND METHODS:qRT-PCR was employed to determine miR-27a-3p expression in two breast cancer cell lines, MCF-7 and MCF-7/adriamycin-resistant cell line (MCF-7/ADR). The two cell lines were treated with miR-27a-3p mimics or inhibitors or corresponding negative control (NC), respectively. The changes were investigated by qRT-PCR, CCK-8 assay, Western blot (WB), colony formation assay, and flow cytometry assay. Moreover, luciferase reporter assay was analyzed to verify the downstream target gene of miR-27a-3p. Further investigation in the correlation between miR-27a-3p and BTG2 was launched by WB, flow cytometry assay, and CCK-8 assay. The expression of Akt and p-Akt was detected by WB. KEY FINDINGS:Significantly higher miR-27a-3p expression was confirmed in MCF-7/ADR as compared with sensitive cell line MCF-7 (<0.05). The down-regulation of miR-27a-3p in MCF-7/ADR enhanced the sensitivity of cancer cells to adriamycin treatment, decreased multidrug resistance gene 1/P-glycoprotein (MDR1/P-gp) expression, enhanced the apoptosis-related proteins expression, increased adriamycin-induced apoptosis, and inhibited cell proliferation as compared to NC groups (<0.05). The up-regulation of miR-27a-3p in MCF-7 showed the opposite results. BTG2 is identified as a direct target of miR-27a-3p and its down-regulation reversed ADR-resistance. BTG2 treatment exhibited inhibitory effect on PI3K/Akt pathway in MCF-7/ADR cells. SIGNIFICANCE:miR-27a-3p might be associated with resistance of breast cancer cells to adriamycin treatments, modulating cell proliferation and apoptosis by targeting BTG2 and promoting the PI3K/Akt pathway in breast cancer cells. miR-27a-3p/BTG2 axis might be a potential therapeutic target for clinical BC resistance. 10.2147/OTT.S256153
Dysregulated miR-27a-3p promotes nasopharyngeal carcinoma cell proliferation and migration by targeting Mapk10. Li Lihua,Luo Zhaohui Oncology reports miRNA-27a-3p is an important regulator of carcinogenesis and other pathological processes. However, its role in laryngeal carcinoma is still unknown. In our previous research, we found that miR-27a-3p expression was upregulated in nasopharyngeal carcinoma (NPC) using a microarray chip. In the present study, we identified miR-27a-3p as an endogenous promoter of metastatic invasion. The expression levels of miR-27a-3p were correlated with human metastatic progression outcomes and Kaplan-Meier survival. In silico database analyses revealed that Mapk10 is a potential target of miR-27a-3p, and luciferase reporter assay results revealed that miR-27a-3p directly inhibits the Mapk10 3' untranslated region (3'UTR). Real-time PCR and western blotting results ascertained that Mapk10 expression was regulated by miR‑27a-3p. In addition, miR‑27a-3p gain-of-function promoted cell proliferation, migration and invasion in 5-8 F NPC cells. These effects partially depended on Mapk10, and loss of miR‑27a-3p function had the opposite effects. 10.3892/or.2017.5544
Over-expressed miR-27a-3p inhibits inflammatory response to spinal cord injury by decreasing TLR4. Zhang P,Li L-Q,Zhang D,Shen Y European review for medical and pharmacological sciences OBJECTIVE:We investigate whether microRNA-27a-3p (miR-27a-3p) can inhibit the inflammatory response of spinal cord injury by negatively regulating toll-like receptor 4 (TLR4). PATIENTS AND METHODS:The quantitative Real-time polymerase chain reaction (qRT-PCR) assay was used to detect the expression of miR-27a-3p and TLR4 in serum samples from patients with spinal cord injury and in hydrogen peroxide-treated C8-B4 and C8-D1A cells. Dual luciferase reporter assays were used to detect targeted binding of TLR4 to miR-27a-3p. The protein expression of miR-27a-3p and TLR4 and the two inflammatory factors, tumor necrosis factor α (TNF-α) and interleukin 6 (IL-6), were all detected by Western blot. RESULTS:TLR4 expression was elevated and miR-27a-3p was decreased in serum samples from patients with spinal cord injury and in hydrogen peroxide-treated C8-D1A and C8-B4 cells. Dual luciferase reporter assays results demonstrated that miR-27a-3p can bind to TLR4. Up-regulation of miR-27a-3p can decrease the expression of TNF-α and IL-6 and can also reduce TLR4 expression. After overexpression of TLR4, the expression of TNF-α and IL-6 were increased. CONCLUSIONS:miR-27a-3p can inhibit the inflammatory response of spinal cord injury by negatively regulating TLR4. 10.26355/eurrev_201809_15800