Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Yuhua Fan is active.

Publication


Featured researches published by Yuhua Fan.


PLOS ONE | 2013

Ischemic Postconditioning-Mediated miRNA-21 Protects against Cardiac ischemia/reperfusion Injury via PTEN/Akt Pathway

Yingfeng Tu; Lin Wan; Yuhua Fan; Kezheng Wang; Lihong Bu; Tao Huang; Zhen Cheng; Baozhong Shen

Background Ischemic postconditioning (IPost) protects the reperfused heart from infarction which has drawn much attention recently. However, studies to date have rarely investigated the role of microRNAs (miRNAs) in IPost. The aims of this study were to investigate whether miR-21 is involved in the protective effect of IPost against myocardial ischemia-reperfusion (I/R) injury and disclose the potential molecular mechanisms involved. Methods and Results We found that miR-21 was remarkably up-regulated in mouse hearts after IPost. To determine the protective role of IPost-induced miR-21 up-regulation, the mice were divided into the following four groups: I/R group; I/R+IPost group (I/R mice treated with IPost); Antagomir-21+IPost+I/R group (I/R mice treated with anagomir-21 and IPost); Scramble+IPost+I/R group (I/R mice treated with scramble and IPost). The results showed IPost could reduce I/R injury-induced infarct size of the left ventricle, improve cardiac function, and prevent myocardial apoptosis, while knockdown of miR-21 with antagomir-21 could reverse these protective effects of IPost against mouse I/R injury. Furthermore, we confirmed that miR-21 plays a protective role in myocardial apoptosis through PTEN/Akt signaling pathway, which was abrogated by the PI3K inhibitor LY294002. The protective effect of miR-21 on myocardial apoptosis was further revealed in mouse hearts after IPost treatment in vivo. Conclusions Our data clearly demonstrate that miR-21 is involved in IPost-mediated cardiac protection against I/R injury and dysfunction through the PTEN/Akt signaling pathway in vivo. Identifying the beneficial roles of IPost-regulated miRNAs in cardiac protection, which may be a rational target selection for ischemic cardioprotection.


International Journal of Cardiology | 2013

Upregulation of microRNA-1 and microRNA-133 contributes to arsenic-induced cardiac electrical remodeling.

Hongli Shan; Yong Zhang; Benzhi Cai; Xi Chen; Yuhua Fan; Lili Yang; Xichuang Chen; Haihai Liang; Ying Zhang; Xiaohui Song; Chaoqian Xu; Yanjie Lu; Baofeng Yang; Zhimin Du

BACKGROUND A large body of evidence showed that arsenic trioxide (As2O3), a front-line drug for the treatment of acute promyelocytic leukemia, induced abnormal cardiac QT prolongation, which hampers its clinical use. The molecular mechanisms for this cardiotoxicity remained unclear. This study aimed to elucidate whether microRNAs (miRs) participate in As2O3-induced QT prolongation. METHODS A guinea pig model of As2O3-induced QT prolongation was established by intravenous injection with As2O3. Real-time PCR and Western blot were employed to determine the expression alterations of miRs and mRNAs, and their corresponding proteins. RESULTS The QT interval and QRS complex were significantly prolonged in a dose-dependent fashion after 7-day administration of As2O3. As2O3 induced a significant upregulation of the muscle-specific miR-1 and miR-133, as well as their transactivator serum response factor. As2O3 depressed the protein levels of ether-a-go-go related gene (ERG) and Kir2.1, the K(+) channel subunits responsible for delayed rectifier K(+) current IKr and inward rectifier K(+) current IK1, respectively. In vivo transfer of miR-133 by direct intramuscular injection prolonged QTc interval and increased mortality rate, along with depression of ERG protein and IKr in guinea pig hearts. Similarly, forced expression of miR-1 widened QTc interval and QRS complex and increased mortality rate, accompanied by downregulation of Kir2.1 protein and IK1. Application of antisense inhibitors to knockdown miR-1 and miR-133 abolished the cardiac electrical disorders caused by As2O3. CONCLUSIONS Deregulation of miR-133 and miR-1 underlies As2O3-induced cardiac electrical disorders and these miRs may serve as potential therapeutic targets for the handling of As2O3 cardiotoxicity.


PLOS ONE | 2012

M3 subtype of muscarinic acetylcholine receptor promotes cardioprotection via the suppression of miR-376b-5p.

Z.W. Pan; Yueping Guo; Hanping Qi; Kai Fan; Shu Wang; Hua Zhao; Yuhua Fan; Jing Xie; Feng Guo; Yunlong Hou; Ning Wang; Rong Huo; Yong Zhang; Yan Liu; Zhimin Du

The M3 subtype of muscarinic acetylcholine receptors (M3-mAChR) plays a protective role in myocardial ischemia and microRNAs (miRNAs) participate in many cardiac pathophysiological processes, including ischemia-induced cardiac injury. However, the role of miRNAs in M3-mAChR mediated cardioprotection remains unexplored. The present study was designed to identify miRNAs that are involved in cardioprotective effects of M3-mAChR against myocardial ischemia and elucidate the underlying mechanisms. We established rat model of myocardial ischemia and performed miRNA microarray analysis to identify miRNAs involved in the cardioprotection of M3-mAChR. In H9c2 cells, the viability, intracellular free Ca2+ concentration ([Ca2+]i), intracellular reactive oxygen species (ROS), miR-376b-5p expression level, brain derived neurophic factor (BDNF) and nuclear factor kappa-B (NF-κB) levels were measured. Our results demonstrated that M3-mAChR protected myocardial ischemia injury. Microarray analysis and qRT-PCR revealed that miR-376b-5p was significantly up-regulated in ischemic heart tissue and the M3-mAChRs agonist choline reversed its up-regulation. In vitro, miR-376b-5p promoted H2O2-induced H9c2 cell injuries measured by cells viability, [Ca2+]i and ROS. Western blot and luciferase assay identified BDNF as a direct target of miR-376b-5p. M3-mAChR activated NF-κB and thereby inhibited miR-376b-5p expression. Our data show that a novel M3-mAChR/NF-κB/miR-376b-5p/BDNF axis plays an important role in modulating cardioprotection. MiR-376b-5p promotes myocardial ischemia injury possibly by inhibiting BDNF expression and M3-mAChR provides cardioprotection at least partially mediated by the downregulation of miR-376b-5p through NF-κB. These findings provide new insight into the potential mechanism by which M3-mAChR provides cardioprotection against myocardial ischemia injury.


Scientific Reports | 2015

Overexpression of miRNA-497 inhibits tumor angiogenesis by targeting VEGFR2.

Yingfeng Tu; Li Liu; Dongliang Zhao; Youbin Liu; Xiaowei Ma; Yuhua Fan; Lin Wan; Tao Huang; Zhen Cheng; Baozhong Shen

Recent studies reported miR-497 exhibited inhibitory effects in various cancers. However, whether miR-497 is involved in inhibiting angiogenesis, which is critical for tumor growth and metastasis, is still unknown. The purpose of this study was to investigate the potential role of miR-497 in tumor angiogenesis. In this work, cell proliferation and apoptosis analyses were conducted to explore the potential function of miR-497 in HUVECs by using MTT and TUNEL assays. Western blotting (WB) was employed to validate the downstream targets of miR-497. Furthermore, in order to disclose the role of miR-497 on angiogenesis, VEGFR2-luc transgenic mice were treated with miR-497 mimic and applied to monitor tumor angiogenesis and growth by in vivo bioluminescent imaging (BLI). The results demonstrated that overexpression of miR-497 showed inhibitory effects on VEGFR2 activation and downstream Raf/MEK/ERK signal pathways in vitro and in vivo. Moreover, overexpression of miR-497 effectively induced HUVECs apoptosis by targeting VEGFR2 and downstream PI3K/AKT signaling pathway. Furthermore, miR-497 exhibited anti-angiogenesis and anti-tumor effects in the VEGFR2-luc breast tumor model proven by BLI, WB and immunohistochemistry analysis. In summary, miR-497 inhibits tumor angiogenesis and growth via targeting VEGFR2, indicating miR-497 can be explored as a potential drug candidate for cancer therapy.


Clinical and Experimental Pharmacology and Physiology | 2012

Activation of cardiac M3 muscarinic acetylcholine receptors has cardioprotective effects against ischaemia-induced arrhythmias.

Shu Wang; Hongmei Han; Yanan Jiang; Chao Wang; Haoxin Song; Z.W. Pan; Kai Fan; Juan Du; Yuhua Fan; Zhimin Du; Yan Liu

Increasing evidence indicates the important roles of M3 muscarinic acetylcholine receptors (M3 mAChR) in the regulation and maintenance of cardiac function and heart disease. In the present study, we investigated whether the M3 mAChR mediates the cardioprotection against ischaemia‐induced arrhythmias and the mechanisms involved. Myocardial ischaemia was established in Wistar rats by occlusion of the left anterior descending coronary artery. Rats were treated with choline chloride (an M3 mAChR agonist; 10 mg/kg, i.v.) 10 min before occlusion. In addition, 4‐diphenylacetoxy‐N‐methylpiperidine‐methiodide (4‐DAMP; 0.12 μg/kg, i.v.) was administered 5 min before choline in the 4‐DAMP‐treated group. Ischaemia‐induced arrhythmias were evaluated in each group for a period of 1 h after occlusion. After 24 h occlusion, protein and mRNA levels of L‐type Ca2+ channels and the Na+/Ca2+ exchanger (NCX) were determined. Ischaemia‐induced arrhythmias following coronary artery occlusion were diminished by choline and this effect was reversed in the 4‐DAMP‐treated group. In vitro, the effects of myocardial ischaemia were simulated by incubating isolated ventricular cardiomyocytes with Tyrodes solution (pH 6.8). Intracellular Ca2+ overload was confirmed and this was decreased by choline. Furthermore, choline reduced the L‐type Ca2+ current (ICa,L) compared with cardiomyocytes incubated in Tyrodes solution (pH 6.8) alone. Choline reduced the ‘ischaemia’‐induced upregulated expression of L‐type Ca2+ channels and NCX at both the protein and mRNA level. Based on these results, choline has an obvious protective effect against ischaemia‐induced arrhythmias that is mediated via activation of cardiac M3 mAChR, which reduces Ca2+ overload mediated by L‐type Ca2+ channels and the NCX.


PLOS ONE | 2014

Arsenic Trioxide and Resveratrol Show Synergistic Anti-Leukemia Activity and Neutralized Cardiotoxicity

Yuhua Fan; Meng Chen; Jia Meng; Lei Yu; Yingfeng Tu; Lin Wan; Kun Fang; Wenliang Zhu

Cardiotoxicity is an aggravating side effect of many clinical antineoplastic agents such as arsenic trioxide (As2O3), which is the first-line treatment for acute promyelocytic leukemia (APL). Clinically, drug combination strategies are widely applied for complex disease management. Here, an optimized, cardiac-friendly therapeutic strategy for APL was investigated using a combination of As2O3 and genistein or resveratrol. Potential combinations were explored with respect to their effects on mitochondrial membrane potential, reactive oxygen species, superoxide dismutase activity, autophagy, and apoptosis in both NB4 cells and neonatal rat left ventricular myocytes. All experiments consistently suggested that 5 µM resveratrol remarkably alleviates As2O3-induced cardiotoxicity. To achieve an equivalent effect, a 10-fold dosage of genistein was required, thus highlighting the dose advantage of resveratrol, as poor bioavailability is a common concern for its clinical application. Co-administration of resveratrol substantially amplified the anticancer effect of As2O3 in NB4 cells. Furthermore, resveratrol exacerbated oxidative stress, mitochondrial damage, and apoptosis, thereby reflecting its full range of synergism with As2O3. Addition of 5 µM resveratrol to the single drug formula of As2O3 also further increased the expression of LC3, a marker of cellular autophagy activity, indicating an involvement of autophagy-mediated tumor cell death in the synergistic action. Our results suggest a possible application of an As2O3 and resveratrol combination to treat APL in order to achieve superior therapeutics effects and prevent cardiotoxicity.


Cellular Physiology and Biochemistry | 2013

Genistein Ameliorates Adverse Cardiac Effects Induced by Arsenic Trioxide Through Preventing Cardiomyocytes Apoptosis

Yuhua Fan; Chen Wang; Yong Zhang; Pengzhou Hang; Yan Liu; Zhenwei Pan; Ning Wang; Zhimin Du

Background/Aims: Arsenic trioxide (As<sub>2</sub>O<sub>3</sub>) is a highly effective agent for treatment of acute promyelocytic leukemia (APL). However, consecutively administered As<sub>2</sub>O<sub>3</sub> induces serious adverse cardiac effects, including long QT syndrome (LQTs) and even sudden cardiac death. Previous studies have shown that genistein (Gen) exerts anti-oxidant, anti-inflammatory, and anti-apoptotic effects. The present study aimed to explore the potential protective effects of Gen on As<sub>2</sub>O<sub>3</sub>-induced adverse cardiac effects, and to elucidate the underlying molecular mechanisms. Methods: A rat model of As<sub>2</sub>O<sub>3</sub>-induced QT prolongation was generated by intravenous injection with As<sub>2</sub>O<sub>3</sub>. Surface electrocardiogram (ECG) and hemodynamics were employed to assess the LQTs and cardiac function. Intracellular calcium concentration ([Ca<sup>2+</sup>]<sub>i</sub>) and mitochondrial membrane potential (Δψm) were measured by confocal microscopy, and cardiomyocytes apoptosis were assessed by TUNEL assay. Western blot was applied to determine protein levels. Results: We found for the first time that treatment with Gen significantly reversed LQTs and dose-dependently improved As<sub>2</sub>O<sub>3</sub>-induced impairment of cardiac function. As<sub>2</sub>O<sub>3</sub> elevated [Ca<sup>2+</sup>]<sub>i</sub> and Gen mitigated this effect. Meanwhile, Gen significantly reversed As<sub>2</sub>O<sub>3</sub>-mediated cardiomyocytes apoptosis. Furthermore, Gen dose-dependently inhibited the phosphorylated JNK and p38-MAPK (pp38-MAPK), and blocked Δψm collapse, and further decreased cleaved caspase-3. Conclusion: Gen protects against the adverse cardiac effects of As<sub>2</sub>O<sub>3</sub> partly by mitigating cardiomyocytes apoptosis induced by As<sub>2</sub>O<sub>3</sub> through attenuating intracellular calcium overload and downregulating protein expression of p-JNK and pp38-MAPK to ameliorate the damage of Δψm leading to suppression of caspase-3 activation. Gen might be used as an adjunction therapy in APL patients receiving As<sub>2</sub>O<sub>3</sub> treatment to avoid, at least to minimize, the adverse cardiac effects of As<sub>2</sub>O<sub>3</sub>.


Cellular Physiology and Biochemistry | 2017

Resveratrol Protects Against Pulmonary Arterial Hypertension in Rats via Activation of Silent Information Regulator 1

Lei Yu; Yingfeng Tu; Xueling Jia; Kun Fang; Li Liu; Lin Wan; Chuanying Xiang; Yanan Wang; Xiangju Sun; Tianyou Liu; Dejun Yu; Weiwei Cao; Yinli Song; Yuhua Fan

Background/Objectives: The polyphenol resveratrol (Rev) has been found to exhibit various beneficial effects including prevention of pulmonary arterial hypertension (PAH). The present study was designed to investigate the action and potential mechanism of Rev on PAH, focusing on the role of SIRT1 (Silent Information Regulator 1) in apoptosis of pulmonary artery smooth muscle cells (PASMCs). Methods: PAH rats were established by exposure to hypoxia for 21 days. Rev and SRT1720 (a selective SIRT1 activator) were used to reverse PAH by gavaging rats. PASMCs were confronted with hypoxia for 24 h or 48 h and were then treated with Rev or SRT1720 in vitro. Western blot was performed to detect the protein expression of SIRT1. CCK-8 and scratch wound experiments were carried out to verify cell proliferation. In addition, the TUNEL positive assay and flow cytometry assay were used to measure PASMC apoptosis. Mitochondrial permeability transition (mPT) was identified by confocal microscopy. Right ventricular systolic pressure (RVSP) was determined with a Gould pressure transducer, and right ventricular hypertrophy (RVH) was determined by weighing the cardiac muscle. Results: We demonstrated that Rev could reverse the remodelling of the pulmonary vasculature, thus contributing to alleviating the severity of PAH. Down-regulation of SIRT1 was observed in PAH, but administration of Rev had no obvious effect on the protein expression of SIRT1. In addition, Rev could induce mitochondrial swelling and nuclear pyknosis, leading to small, dense, and dysmorphic mitochondria in rats exposed to hypoxia alone. Rev treatment inhibited PASMC proliferation in a dose-dependent manner in vitro. Incubation with SRT1720, a specific activator of SIRT1, significantly retarded PASMC proliferation and promoted PASMC apoptosis in vitro. The mechanism could be associated with inducing mPT damage in PASMCs. Rev and SRT1720 treatment mitigated RVSP and reduced RVH. Conclusion: Rev produced a beneficial effect partially by enhancing the activation of SIRT1, thus improving RVSP and reducing RVH. SIRT1 activation increased PASMC apoptosis by inducing mPT dysfunction, which might be a novel future strategy for the treatment of PAH.


Journal of the American Heart Association | 2016

Resveratrol Ameliorates Cardiac Hypertrophy by Down‐regulation of miR‐155 Through Activation of Breast Cancer Type 1 Susceptibility Protein

Yuhua Fan; Li Liu; Kun Fang; Tao Huang; Lin Wan; Youbin Liu; Sen Zhang; Dongxia Yan; Guangnan Li; Yanhui Gao; Yan-jie Lv; Yanjun Chen; Yingfeng Tu

Background The polyphenol resveratrol (Rev) has been reported to exhibit cardioprotective effects, such as inhibition of TAC (transverse aortic constriction) or isoprenaline (ISO)‐induced hypertrophy. MicroRNA‐155 (miR‐155) was found to be decreased in hypertrophic myocardium, which could be further reduced by pretreatment of Rev. The study was designed to investigate the molecular effects of miR‐155 on cardiac hypertrophy, focusing on the role of breast cancer type 1 susceptibility protein (BRCA1). Methods and Results We demonstrated that Rev alleviated severity of hypertrophic myocardium in a mice model of cardiac hypertrophy by TAC treatment. Down‐regulation of miR‐155 was observed in pressure overload– or ISO‐induced hypertrophic cardiomyoctyes. Interestingly, administration of Rev substantially attenuated miR‐155 level in cardiomyocytes. In agreement with its miR‐155 reducing effect, Rev relieved cardiac hypertrophy and restored cardiac function by activation of BRCA1 in cardiomyoctyes. Our results further revealed that forkhead box O3a (FoxO3a) was a miR‐155 target in the heart. And miR‐155 directly repressed FoxO3a, whose expression was mitigated in miR‐155 agomir and mimic treatment in vivo and in vitro. Conclusions We conclude that BRCA1 inactivation can increase expression of miR‐155, contributing to cardiac hypertrophy. And Rev produces their beneficial effects partially by down‐regulating miR‐155 expression, which might be a novel strategy for treatment of cardiac hypertrophy.


Cellular Physiology and Biochemistry | 2018

Multimodality Molecular Imaging of Cardiovascular Disease Based on Nanoprobes

Yingfeng Tu; Yao Sun; Yuhua Fan; Zhen Cheng; Bo Yu

Recently, multimodality molecular imaging has evolved into a fast-growing research field with goals of detecting and measuring biological processes in vivo non-invasively. Researchers have come to realize that the complementary abilities of different imaging modalities over single modality could provide more precisely information for the diagnosis of diseases. At present, nanoparticles-based multimodal imaging probes have received significant attention because of their ease of preparation and straightforward integration of each modality into one entity. More importantly, nanotechnology has an increasing impact on multimodality molecular imaging of cardiovascular diseases, such as atherosclerosis and vulnerable plaque, myocardial infarction, angiogenesis, apoptosis and so on. In this review, we briefly summarize that various nanoprobes are exploited for targeted molecular imaging of cardiovascular diseases, as well as associated multimodality imaging approaches and their applications in the diagnosis and treatment of cardiovascular diseases.

Collaboration


Dive into the Yuhua Fan's collaboration.

Top Co-Authors

Avatar

Yingfeng Tu

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Lin Wan

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Li Liu

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Tao Huang

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Zhimin Du

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Baozhong Shen

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Kun Fang

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Yan Liu

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Yong Zhang

Harbin Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge