Network


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

Hotspot


Dive into the research topics where Qiu-Xiong Lin is active.

Publication


Featured researches published by Qiu-Xiong Lin.


Scientific Reports | 2017

CircRNA_000203 enhances the expression of fibrosis-associated genes by derepressing targets of miR-26b-5p, Col1a2 and CTGF, in cardiac fibroblasts

Chun-Mei Tang; Ming Zhang; Lei Huang; Zhi-Qin Hu; Jie-Ning Zhu; Zhen Xiao; Zhuo Zhang; Qiu-Xiong Lin; Xi-Long Zheng; Min Yang; Shu-Lin Wu; Jian-Ding Cheng; Zhi-Xin Shan

Circular RNAs (circRNAs) participate in regulating gene expression in diverse biological and pathological processes. The present study aimed to investigate the mechanism underlying the modulation of circRNA_000203 on expressions of fibrosis-associated genes in cardiac fibroblasts. CircRNA_000203 was shown upregulated in the diabetic mouse myocardium and in Ang-II-induced mouse cardiac fibroblasts. Enforced-expression of circRNA_000203 could increase expressions of Col1a2, Col3a1 and α-SMA in mouse cardiac fibroblasts. RNA pull-down and RT-qPCR assay indicated that circRNA_000203 could specifically sponge miR-26b-5p. Dual luciferase reporter assay revealed that miR-26b-5p interacted with 3′UTRs of Col1a2 and CTGF, and circ_000203 could block the interactions of miR-26b-5p and 3′UTRs of Col1a2 and CTGF. Transfection of miR-26b-5p could post-transcriptionaly inhibit expressions of Col1a2 and CTGF, accompanied with the suppressions of Col3a1 and α-SMA in cardiac fibroblasts. Additionally, over-expression of circRNA_000203 could eliminate the anti-fibrosis effect of miR-26b-5p in cardiac fibroblasts. Together, our results reveal that suppressing the function of miR-26b-5p contributes to the pro-fibrosis effect of circRNA_000203 in cardiac fibroblasts.


British Journal of Pharmacology | 2008

Enhanced angiotensin converting enzyme 2 regulates the insulin/Akt signalling pathway by blockade of macrophage migration inhibitory factor expression

Jiu-Chang Zhong; Xi-Yong Yu; Qiu-Xiong Lin; Xiao-Hong Li; Huang Xz; Xiao Dz; Shu-Guang Lin

Macrophage migration inhibitory factor (MIF) is now known to be a pro‐inflammatory cytokine associated with insulin resistance. Our aim was to investigate whether angiotensin converting enzyme 2 (ACE2) could modulate the expression of MIF and the insulin/Akt‐endothelial nitric oxide (NO) synthase (eNOS) signalling in a human endothelial cell line (EAhy926).


Molecular Biology Reports | 2009

An efficient method to enhance gene silencing by using precursor microRNA designed small hairpin RNAs

Zhi-Xin Shan; Qiu-Xiong Lin; Chunyu Deng; Xiao-Hong Li; Wei Huang; Honghong Tan; Yong-Heng Fu; Min Yang; Xi-Yong Yu

Gene silencing can be mediated by small interfering RNA (siRNA) and microRNA (miRNA). To investigate the potential application of using a precursor microRNA (pre-miRNA) backbone for gene silencing, we studied the inhibition efficiency of exogenous GFP and endogenous GAPDH by conventional shRNA- and pre-miRNA-designed hairpins, respectively. In this study, the conventional shRNA-, pre-miRNA-30-, and pre-miRNA-155-designed hairpins targeting either GFP or GAPDH were transfected into the HEK293 cells that were mediated by the pSilencer-4.1-neo vector, which carries a modified RNA polymerase II-type CMV promoter. Comparisons with conventional GFP shRNA showed that GFP levels were reduced markedly by pre-miRNA-30- and pre-miRNA-155-designed GFP shRNAs by fluorescence microscopy. The consistent results from semi-quantitative RT-PCR and Western blot analysis revealed that pre-miRNA-30- and pre-miRNA-155-designed GFP shRNAs could suppress GFP expression significantly. As for endogenous GAPDH, the results from semi-quantitative RT-PCR and Western blot analysis showed that pre-miRNA-30- and pre-miRNA-155-designed GAPDH shRNAs could suppress GAPDH expression even more efficiently than conventional GAPDH shRNA. Together, this study confirmed the efficiency of gene silencing mediated by pre-miRNA-30- and pre-miRNA-155-designed shRNAs, demonstrating that pre-miRNA-designed hairpins are a good strategy for gene silencing.


Journal of Molecular and Cellular Cardiology | 2009

Involvement of Src in L-type Ca2+ channel depression induced by macrophage migration inhibitory factor in atrial myocytes.

Fang Rao; Chun-Yu Deng; Shu-Lin Wu; Ding‐Zhang Xiao; Xi-Yong Yu; Su-Juan Kuang; Qiu-Xiong Lin; Zhi-Xin Shan

Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine that controls inflammatory processes, and inflammation is known to play an important role in the pathogenesis of atrial fibrillation (AF). The present study sought to investigate whether MIF expression is responsible for the changes in L-type Ca2+ currents (I(Ca,L)) seen in AF. Whole-cell voltage-clamp recordings and biochemical assays were used to study the regulation and expression of I(Ca,L) in human atrial myocytes and in HL-1 cells. Basal I(Ca,L) was reduced in AF compared to sinus rhythm (SR) controls, mRNA and protein levels of the pore-forming alpha1C subunit of L-type Ca2+ channel (LCC alpha1C) were also decreased, while MIF expression levels were increased in AF. Levels of Src and activated Src (p-Src Y416) were higher in AF than in SR. Treatment of atrial myocytes from a patient with SR with human recombinant MIF (rMIF) (40 nM, 1 h) was found to depress I(Ca,L) amplitudes, while mouse rMIF (20 or 40 nM, 24 h) suppressed peak I(Ca,L) in HL-1 cells by approximately 69% and approximately 83% in a concentration-dependent manner. Mouse rMIF impaired the time-dependent recovery from inactivation of I(Ca,L) and down-regulated LCC alpha1C subunit levels. The depression of I(Ca,L) and decrease of LCC protein levels induced by rMIF were prevented by the Src inhibitors genistein and PP1. These results implicate MIF in the electrical remodeling that accompanies AF, probably by decreasing I(Ca,L) amplitudes through impairment of channel function, down-regulation of LCC alpha1C subunit levels, and the activation of c-Src kinases in atrial myocytes.


Clinical Pharmacology & Therapeutics | 2007

Evidence for Coexistence of Three β‐Adrenoceptor Subtypes in Human Peripheral Lymphocytes

Xi-Yong Yu; Shu-Guang Lin; Xiao-mei Wang; Yuan Liu; Bin Zhang; Qiu-Xiong Lin; Min Yang; Shu-Feng Zhou

Peripheral circulating lymphocytes are easily accessible cells for investigating changes in β‐adrenergic receptors (ADRBs) in humans, but previous reports indicate that these cells only express ADRB2. This study aimed to investigate whether ADRB1 and ADRB3 were expressed in peripheral lymphocytes and the changes of ADRBs in congestive heart failure. Our study demonstrates that ADRB1, ADRB2, and ADRB3 coexist in human peripheral lymphocytes, with differential binding property and expression level. Patients with congestive heart failure had significantly decreased total ADRB density and mRNA levels of ADRB1 and ADRB2 genes, but not ADRB3, compared with healthy subjects. The levels of mRNA of ADRB1 and ADRB2 in peripheral lymphocytes from patients with congestive heart disease were significantly increased after drug treatment. Our study, for the first time, indicates that human peripheral lymphocytes coexpress ADRB1, ADRB2, and ADRB3, which has important implications for precisely predicting clinical response to drug therapy in congestive heart failure.


Molecular Medicine Reports | 2013

Silencing of desmoplakin decreases connexin43/Nav1.5 expression and sodium current in HL‑1 cardiomyocytes

Qianhuan Zhang; Chunyu Deng; Fang Rao; Rohan M. Modi; Jiening Zhu; Xiao‐Ying Liu; Liping Mai; Honghong Tan; Xi-Yong Yu; Qiu-Xiong Lin; Ding‐Zhang Xiao; Su-Juan Kuang; Shu-Lin Wu

Desmosomes and gap junctions are situated in the intercalated disks of cardiac muscle and maintain the integrity of mechanical coupling and electrical impulse conduction between cells. The desmosomal plakin protein, desmoplakin (DSP), also plays a crucial role in the stability of these interconnected components as well as gap junction connexin proteins. In addition to cell‑to‑cell junctions, other molecules, including voltage‑gated sodium channels (Nav1.5) are present in the intercalated disk and support the contraction of cardiac muscle. Mutations in genes encoding desmosome proteins may result in fatal arrhythmias, including arrhythmogenic right ventricular cardiomyopathy (ARVC). Therefore, the aim of the present study was to determine whether the presence of DSP is necessary for the normal function and localization of gap junction protein connexin43 (Cx43) and Nav1.5. To examine this hypothesis, RNA interference was utilized to knock down the expression of DSP in HL‑1 cells and the content, distribution and function of Cx43 and Nav1.5 was assessed. Western blotting and flow cytometry experiments revealed that Cx43 and Nav1.5 expression decreased following DSP silencing. In addition, immunofluorescence studies demonstrated that a loss of DSP expression led to an abnormal distribution of Cx43 and Nav1.5, while scrape‑loading dye/transfer revealed a decrease in dye transfer in DSP siRNA‑treated cells. The sodium current was also recorded by the whole‑cell patch clamp technique. The results indicated that DSP suppression decreased sodium current and slowed conduction velocity in cultured cells. The present study indicates that impaired mechanical coupling largely affects electrical synchrony, further uncovering the pathogenesis of ARVC.


European Journal of Pharmacology | 2009

Pharmacological effects of carvedilol on T-type calcium current in murine HL-1 cells

Chunyu Deng; Fang Rao; Shu-Lin Wu; Su-Juan Kuang; Xiaoying Liu; Zhiling Zhou; Zhi-Xin Shan; Qiu-Xiong Lin; Weimin Qian; Min Yang; Qingshan Geng; Zhang Y; Xi-Yong Yu; Shu-Guang Lin

Carvedilol is widely used in the treatment of cardiovascular diseases including atrial fibrillation. T-type Ca(2+) channels have been recognized recently in the mechanisms underlying atrial arrhythmias. However, it is unclear whether carvedilol may affect the T-type Ca(2+) channel. The present study evaluated the pharmacological effects of carvedilol on T-type calcium current (I(Ca,T)) in the murine HL-1 cell line. I(Ca)(,T) was recorded by the whole-cell patch-clamp technique. Calcium transient was monitored by the fluorescent dye Fluo-4/AM and confocal laser scanning microscopy. Carvedilol reversibly inhibited I(Ca)(,T) in a concentration-dependent manner, with an IC50 of 2.1 microM. 3 microM carvedilol was found to decrease the peak I(Ca)(,T) amplitude at -20 mV from 20.1+/-1.8pA/pF to 10.9+/-2.1pA/pF. Carvedilol significantly shifted the steady-state inactivation curve of I(Ca)(,T) towards more negative potential by 12.8 mV, while the activation curve was not significantly altered. Carvedilol delayed recovery from inactivation of I(Ca)(,T), time constant (tau) was 112.4+/-3.5 ms in control and 270.1+/-4.7 ms in carvedilol. Carvedilol-induced inhibition rate in I(Ca)(,T) was enhanced with the increase in stimuli frequency, the inhibitory rate was 23.2+/-4.1% at 0.2 Hz and 47.2+/-0.6% at 2 Hz. Carvedilol still produced the significant decrease in the amplitude of I(Ca)(,T) in the presence of H-89, PKA inhibitor. Carvedilol significantly inhibited the amplitude of the calcium transient in a concentration-dependent manner. These findings indicate that carvedilol inhibits I(Ca)(,T) in atrial cells by mechanisms involving preferential interaction with the inactivated state of T-type Ca(2+) channel.


Scientific Reports | 2016

Myocyte-specific enhancer factor 2C: a novel target gene of miR-214-3p in suppressing angiotensin II-induced cardiomyocyte hypertrophy.

Chun-Mei Tang; Fangzhou Liu; Jie-Ning Zhu; Yong-Heng Fu; Qiu-Xiong Lin; Chun-Yu Deng; Zhi-Qin Hu; Hui Yang; Xi-Long Zheng; Jian-Ding Cheng; Shu-Lin Wu; Zhi-Xin Shan

The role of microRNA-214-3p (miR-214-3p) in cardiac hypertrophy was not well illustrated. The present study aimed to investigate the expression and potential target of miR-214-3p in angiotensin II (Ang-II)-induced mouse cardiac hypertrophy. In mice with either Ang-II infusion or transverse aortic constriction (TAC) model, miR-214-3p expression was markedly decreased in the hypertrophic myocardium. Down-regulation of miR-214-3p was observed in the myocardium of patients with cardiac hypertrophy. Expression of miR-214-3p was upregulated in Ang-II-induced hypertrophic neonatal mouse ventricular cardiomyocytes. Cardiac hypertrophy was attenuated in Ang-II-infused mice by tail vein injection of miR-214-3p. Moreover, miR-214-3p inhibited the expression of atrial natriuretic peptide (ANP) and β-myosin heavy chain (MHC) in Ang-II-treated mouse cardiomyocytes in vitro. Myocyte-specific enhancer factor 2C (MEF2C), which was increased in Ang-II-induced hypertrophic mouse myocardium and cardiomyocytes, was identified as a target gene of miR-214-3p. Functionally, miR-214-3p mimic, consistent with MEF2C siRNA, inhibited cell size increase and protein expression of ANP and β-MHC in Ang-II-treated mouse cardiomyocytes. The NF-κB signal pathway was verified to mediate Ang-II-induced miR-214-3p expression in cardiomyocytes. Taken together, our results revealed that MEF2C is a novel target of miR-214-3p, and attenuation of miR-214-3p expression may contribute to MEF2Cexpressionin cardiac hypertrophy.


European Journal of Pharmacology | 2017

The enhancement of TXA2 receptors-mediated contractile response in intrarenal artery dysfunction in type 2 diabetic mice

Su-Juan Kuang; Jie-Sheng Qian; Hui Yang; Fang Rao; Xiao-Yan Chen; Meng-Zhen Zhang; Zhi-Xin Shan; Qiu-Xiong Lin; Yu-Mei Xue; Shu-Lin Wu; Li Jiang; Chunbo Chen; Chunyu Deng

&NA; Thromboxane A2 (TXA2) has been implicated in the pathogenesis of diabetic vascular complications, although the underlying mechanism remains unclear. The present study investigated the alterations in TXA2 receptor signal transduction in type 2 diabetic renal arteries. The contraction of renal arterial rings in control (db/m+) mice and type 2 diabetic (db/db) mice was measured by a Multi Myograph System. Intracellular calcium concentration ([Ca2+]i) in vascular smooth muscle cells was measured by Fluo‐4/AM dye and confocal laser scanning microscopy. Quantitative real‐time PCR and Western blot analysis were used to determine gene and protein expression levels, respectively. A stable TXA2 mimic U46619 caused markedly stronger dose‐dependent contractions in the renal arteries of db/db mice than in those of db/m+ mice. This response was completely blocked by a TXA2 receptor antagonist GR32191 and significantly inhibited by U73122. U46619‐induced vasoconstriction was increased in the presence of nifedipine in db/db mice compared with that in db/m+ mice, whereas the response to U46619 did not differ between the two groups in the presence of SKF96365. Sarcoplasmic reticulum Ca2+ release‐mediated and CaCl2‐induced contractions did not differ between the two groups. In db/db mice, store‐operated Ca2+(SOC) entry‐mediated contraction in the renal arteries and SOC entry‐mediated Ca2+ influx in smooth muscle cells were significantly increased. And the gene and protein expressions of TXA2 receptors, Orai1 and Stim1 were upregulated in the diabetic renal arteries. Therefore the enhancement of U46619‐induced contraction was mediated by the upregulation of TXA2 receptors and downstream signaling in the diabetic renal arteries.


Scientific Reports | 2017

Activation of miR-34a-5p/Sirt1/p66shc pathway contributes to doxorubicin-induced cardiotoxicity

Jie-Ning Zhu; Yong-Heng Fu; Zhi-Qin Hu; Wen-Yu Li; Chun-Mei Tang; Hong-Wen Fei; Hui Yang; Qiu-Xiong Lin; Deming Gou; Shu-Lin Wu; Zhi-Xin Shan

The molecular mechanisms underlying anthracyclines-induced cardiotoxicity have not been well elucidated. MiRNAs were revealed dysregulated in the myocardium and plasma of rats received Dox treatment. MicroRNA-34a-5p (miR-34a-5p) was verified increased in the myocardium and plasma of Dox-treated rats, but was reversed in rats received Dox plus DEX treatments. Human miR-34a-5p was also observed increased in the plasma of patients with diffuse large B-cell lymphoma after 9- and 16-week epirubicin therapy. Up-regulation of miR-34a-5p was observed in Dox-induced rat cardiomyocyte H9c2 cells. MiR-34a-5p could augment Bax expression, but inhibited Bcl-2 expression, along with the increases of the activated caspase-3 and mitochondrial potentials in H9C2 cells. MiR-34a-5p was verified to modulate Sirt1 expression post-transcriptionally. In parallel to Sirt1 siRNA, miR-34a-5p could enhance p66shc expression, accompanied by increases of Bax and the activated caspase-3 and a decrease of Bcl-2 in H9c2 cells. Moreover, enforced expression of Sirt1 alleviated Dox-induced apoptosis of H9c2 cells, with suppressing levels of p66shc, Bax, the activated caspase-3 and miR-34a-5p, and enhancing Bcl-2 expression. Therefore, miR-34a-5p enhances cardiomyocyte apoptosis by targeting Sirt1, activation of miR-34a-5p/Sirt1/p66shc pathway contributes to Dox-induced cardiotoxicity, and blockage of this pathway represents a potential cardioprotective effect against anthracyclines.

Collaboration


Dive into the Qiu-Xiong Lin's collaboration.

Top Co-Authors

Avatar

Zhi-Xin Shan

Cardiovascular Institute of the South

View shared research outputs
Top Co-Authors

Avatar

Shu-Lin Wu

Cardiovascular Institute of the South

View shared research outputs
Top Co-Authors

Avatar

Chunyu Deng

Cardiovascular Institute of the South

View shared research outputs
Top Co-Authors

Avatar

Fang Rao

Cardiovascular Institute of the South

View shared research outputs
Top Co-Authors

Avatar

Min Yang

Cardiovascular Institute of the South

View shared research outputs
Top Co-Authors

Avatar

Su-Juan Kuang

Cardiovascular Institute of the South

View shared research outputs
Top Co-Authors

Avatar

Chun-Mei Tang

Cardiovascular Institute of the South

View shared research outputs
Top Co-Authors

Avatar

Hui Yang

Cardiovascular Institute of the South

View shared research outputs
Top Co-Authors

Avatar

Jie-Ning Zhu

Cardiovascular Institute of the South

View shared research outputs
Top Co-Authors

Avatar

Jiu-Chang Zhong

Cardiovascular Institute of the South

View shared research outputs
Researchain Logo
Decentralizing Knowledge