Network


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

Hotspot


Dive into the research topics where Yuan Lin is active.

Publication


Featured researches published by Yuan Lin.


Molecular Nutrition & Food Research | 2015

Citrus nobiletin ameliorates experimental colitis by reducing inflammation and restoring impaired intestinal barrier function.

Yongjian Xiong; Dapeng Chen; Changchun Yu; Bochao Lv; Jinyong Peng; Jingyu Wang; Yuan Lin

SCOPE Inflammatory bowel disease is a chronic inflammatory disorder of the gastrointestinal tract. Citrus nobiletin can exert robust anti-inflammatory effects in vivo and in vitro. We evaluated the impact of nobiletin on the excessive inflammatory response and impaired barrier function in a rodent colitis model. METHODS AND RESULTS Colitis was established by infusion with 1 mL 2,4,6-trinitrobenzene sulfonic acid (TNBS) dissolved in ethanol (40% v/v) in rats at a 125 mg/kg dose. Caco-2 cell monolayer exposed to LPSs is used as a culture model for intestinal permeability measurements. Nobiletin decreased rat epithelial proinflammatory cytokines and mediators production. Nobiletin restored impaired barrier function in colitic rats and Caco-2 monolayer. Nobiletin decreased protein expressions of Akt, nuclear factor-kappa B (NF-κB), and myosin light chain kinase (MLCK) isolated from rat intestinal epithelial tissue and Caco-2 cell, respectively. PI3K inhibitor or siRNA targeting of either Akt or NF-κB mitigated the impact of nobiletin on MLCK expression and barrier function in Caco-2 monolayer, respectively. CONCLUSION Nobiletin exerted anti-inflammatory effects in TNBS-induced colitis through the downregulation of inducible nitric oxide synthase (iNOS) and cyclooxygenase 2 (COX-2) expression. Nobiletin restored barrier function, which had been damaged after TNBS administration, through the inhibition of the Akt-NF-κB-MLCK pathway.


Apoptosis | 2015

SZC017, a novel oleanolic acid derivative, induces apoptosis and autophagy in human breast cancer cells.

Lei Gao; Yan Wang; Zhen Xu; Xiaorui Li; Jingjun Wu; Shumin Liu; Peng Chu; Zhengwu Sun; Bin Sun; Yuan Lin; Jinyong Peng; Guozhu Han; Shisheng Wang; Zeyao Tang

Oleanolic acid (OA) and its derivatives such as 2-cyano-3,12-dioxoolean-1,9-dien-28-oic acid (CDDO), CDDO-Me, and CDDO-Im show potent anticancer function. In this study, we elucidated the anticancer effect of SZC017, a novel OA derivative and identified the mechanisms by which SZC017 induces MCF-7 cell death. We found that SZC017 effectively decreased the cell viability of these breast cancer cells, but was less toxic to MCF10A mammary epithelial cells. Mechanisms underlying the inhibition of cell viability are apoptosis, autophagy induction, and G0/G1 phase arrest. SZC017 treatment suppressed the levels of Akt, phosphorylated-Akt (p-Akt), p-IκBα, total p65, and total p-p65, in addition to p-p65 in both the cytoplasm and nucleus. Furthermore, the inhibition of p65 nuclear translocation was confirmed by immunofluorescence staining. Cell viability was increased after pretreatment with chloroquine, an inhibitor of autophagy, whereas the level of procaspase-3 was significantly decreased. A concentration-dependent increase in the intracellular reactive oxygen species (ROS) level was observed in both MCF-7 and MDA-MB-231 cells. Additionally, pretreatment with N-acetyl-l-cysteine (NAC), a ROS scavenger, increased cell viability and the expression of Akt and procaspase-3, but decreased the ratio of LC3-II/I. These data show that SZC017 is an effectively selective anticancer agent against breast cancer cells, highlighting the potential use of this derivative as a breast cancer therapeutic agent.


Vascular Pharmacology | 2017

Phosphocreatine protects endothelial cells from Methylglyoxal induced oxidative stress and apoptosis via the regulation of PI3K/Akt/eNOS and NF-κB pathway.

Peng Chu; Guozhu Han; Anil Ahsan; Zhengwu Sun; Shumin Liu; Zonghui Zhang; Bin Sun; Yanlin Song; Yuan Lin; Jinyong Peng; Zeyao Tang

Methylglyoxal (MGO), an active metabolite of glucose, can cause cellular injury which has an affinity for the progression of diabetes-associated atherosclerosis. Phosphocreatine (PCr) is a well-known high-energy phosphate compound. However, its protective effects and mechanism in the formation of a diabetes-associated atherosclerosis have not been clarified. In the present study, we investigated whether PCr could prevent MGO-induced apoptosis in human umbilical vascular endothelial cells (HUVECs) and explored the possible mechanisms. Cells were pre-treated with PCr and then stimulated with MGO. Cell morphology, cytotoxicity and apoptosis were assessed by light microscopy, MTT assay, and Annexin V-FITC respectively. Apoptotic-related proteins were evaluated by Western blotting. Reactive oxygen species (ROS) generation, intracellular calcium and mitochondrial membrane potential (MMP) were measured with fluorescent probes. Our results showed that PCr dose-dependently prevented MGO associated HUVEC cytotoxicity and suppressed MGO activated ROS generation as well as apoptotic biochemical changes such as lactate dehydrogenase, malondialdehyde leakage, loss of MMP, decreased Bcl-2/Bax protein ratio, levels of caspase-3 and 9. In addition, the antiapoptotic effect of PCr enhanced p-Akt/Akt protein ratio, NO synthase (eNOS) activation, NO production and cGMP levels and also was partially suppressed by a PI3K inhibitor (LY294002). Furthermore, PCr also inhibited MGO-induced transcriptional activity of Nuclear factor kappa B (NFκB). In conclusion, our data described that PCr exerts an antiapoptotic effect in HUVECs exposed to oxidative stress by MGO through the mitochondrial pathway and the modulation of PI3K/Akt/eNOS and NF-κB signaling pathway. Thus, it might be a candidate therapeutic agent for diabetic-associated cardiovascular diseases.


Journal of Pharmacy and Pharmacology | 2013

Effects of berberine on rat jejunal motility

Dapeng Chen; Yongjian Xiong; Bochao Lv; Fangfei Liu; Li Wang; Zeyao Tang; Yuan Lin

The aim of the study was to evaluate berberine‐induced bidirectional regulation on the contractility of jejunum.


Apoptosis | 2017

EGCG protects against homocysteine-induced human umbilical vein endothelial cells apoptosis by modulating mitochondrial-dependent apoptotic signaling and PI3K/Akt/eNOS signaling pathways

Shumin Liu; Zhengwu Sun; Peng Chu; Hailong Li; Anil Ahsan; Ziru Zhou; Zonghui Zhang; Bin Sun; Jingjun Wu; Yalin Xi; Guozhu Han; Yuan Lin; Jinyong Peng; Zeyao Tang

Homocysteine (Hcy) induced vascular endothelial injury leads to the progression of endothelial dysfunction in atherosclerosis. Epigallocatechin gallate (EGCG), a natural dietary antioxidant, has been applied to protect against atherosclerosis. However, the underlying protective mechanism of EGCG has not been clarified. The present study investigated the mechanism of EGCG protected against Hcy-induced human umbilical vein endothelial cells (HUVECs) apoptosis. Methyl thiazolyl tetrazolium assay (MTT), transmission electron microscope, fluorescent staining, flow cytometry, western blot were used in this study. The study has demonstrated that EGCG suppressed Hcy-induced endothelial cell morphological changes and reactive oxygen species (ROS) generation. Moreover, EGCG dose-dependently prevented Hcy-induced HUVECs cytotoxicity and apoptotic biochemical changes such as reducing mitochondrial membrane potential (MMP), decreasing Bcl-2/Bax protein ratio and activating caspase-9 and 3. In addition, EGCG enhanced the protein ratio of p-Akt/Akt, endothelial nitric oxide synthase (eNOS) activation and nitric oxide (NO) formation in injured cells. In conclusion, the present study shows that EGCG prevents Hcy-induced HUVECs apoptosis via modulating mitochondrial apoptotic and PI3K/Akt/eNOS signaling pathways. Furthermore, the results indicate that EGCG is likely to represent a potential therapeutic strategy for atherosclerosis associated with Hyperhomocysteinemia (HHcy).


International Immunopharmacology | 2016

Anticancer effect of SZC015 on lung cancer cells through ROS-dependent apoptosis and autophagy induction mechanisms in vitro.

Bin Sun; Lei Gao; Anil Ahsan; Peng Chu; Yanlin Song; Hailong Li; Zonghui Zhang; Yuan Lin; Jinyong Peng; Zhicheng Song; Shisheng Wang; Zeyao Tang

Oleanolic acid (OA) and its several derivatives possess various pharmacological activities, such as antitumor and anti-inflammation. In present study, anticancer effect of SZC015, an OA derivative, and its underlying mechanisms were investigated. We demonstrated that cell viability was significantly decreased in SZC015-treated lung cancer cells, but has less cytotoxicity in human bronchial epithelial cell line. Further investigation verified that apoptosis and autophagy induction and G0/G1 phase arrest were observed in SZC015-treated H322 cells. Mechanically, the level of Akt, p-Akt, p-IκBα, and total p65, the p-p65 in the cytoplasm and nucleus were suppressed by SZC015 in H322 cells, respectively. Inhibition of p65 nuclear translocation was also confirmed by immunofluorescence staining. In addition, co-treatment with chloroquine, an autophagy inhibitor, significantly inhibited SZC015-induced autophagy and enhanced SZC015-induced apoptotic cell death. Intracellular ROS was increased in a concentration-dependent manner, which could be prevented by N-Acetyl l-Cysteine, an ROS scavenger. Moreover, the level of Akt and procaspase-3 were increased, while the ratio of LC3 II/I was decreased. Taken together, our study demonstrates that the inhibitory effect of SZC015 against H322 cells is mediated by excessive ROS generation that could suppress Akt/NF-κB signaling pathway, which thereby leads to apoptotic and autophagic cell death.


Apoptosis | 2016

Erratum to: Phosphocreatine protects against LPS-induced human umbilical vein endothelial cell apoptosis by regulating mitochondrial oxidative phosphorylation.

Zhengwu Sun; Xiaoyan Lan; Anil Ahsan; Yalin Xi; Shumin Liu; Zonghui Zhang; Peng Chu; Yushu Song; Fengyuan Piao; Jinyong Peng; Yuan Lin; Guozhu Han; Zeyao Tang

The original version of this article unfortunately contained a mistake. The arrow marks in Fig. 5 were incorrect. It is now corrected with this erratum. The correct version of Fig. 5 is given below. The authors apologise for this error and the inconvenience it has caused to the readers.


Molecular Nutrition & Food Research | 2015

Capsaicin alleviates abnormal intestinal motility through regulation of enteric motor neurons and MLCK activity: Relevance to intestinal motility disorders

Dapeng Chen; Yongjian Xiong; Yuan Lin; Zeyao Tang; Jingyu Wang; Li Wang; Jihong Yao

SCOPE Capsaicin is an active component of chili peppers, having diverse effects. However, the effects of capsaicin on intestinal motility are still controversial. The present study aimed to investigate the effects of capsaicin on intestinal motility disorder and uncover related mechanisms. MATERIALS AND RESULTS A rat model with intestinal motility disorder was established in vitro through adding different stimuli into tissue bath; in vivo using constipation and diarrhea model, respectively. Capsaicin exerted dual effects on intestinal motility, i.e. the relaxation and contraction of jejunum induced by corresponding stimulus were, respectively, regulated to be normal contraction by capsaicin. The mechanisms underlined capsaicin-induced dual effects were investigated using Western blotting, qRT-PCR, and whole-cell patch clamp, respectively. Results showed that cholinergic excitatory nerves, adrenergic nerves, and neurons containing nitric oxide synthase, which are the main muscle motor neurons in enteric nervous system (ENS), are involved in capsaicin-induced dual effects. The competition for regulation of Ca(2+) influx by capsaicin induced the interaction with components of the ENS. Capsaicin significantly increased myosin light chain kinase (MLCK) expression and myosin phosphorylation extent in jejunal segments of constipation-prominent rats and significantly decreased MLCK expression and myosin phosphorylation extent in jejunal segments of diarrhea-prominent rats. CONCLUSION In summary, capsaicin alleviates abnormal intestinal motility through regulating enteric motor neurons and MLCK activity, which is beneficial for the treatment of gastrointestinal motility disorders.


Digestive Diseases and Sciences | 2014

Epithelial MLCK and Smooth Muscle MLCK May Play Different Roles in the Development of Inflammatory Bowel Disease

Dapeng Chen; Yuan Lin; Yongjian Xiong

In the manuscript entitled ‘‘Claudin-2 Regulates Colorectal Inflammation via Myosin Light Chain Kinase-Dependent Signaling,’’ Nishida et al. [1] found that myosin light-chain kinase (MLCK) protein content in the entire mucosal layer of the colon tissue isolated from TNF-a-treated claudin-2 (-/-) mice was increased compared with that in the claudin-2 (?/?) mice. They concluded that claudin-2 could regulate colon inflammation via MLCK-dependent signaling pathway and claudin-2 as well as MLCK was the potential therapeutic targets for treating inflammatory bowel disease. Nishida et al. did not clarify whether epithelial MLCK (long MLCK, 220 kDa) or smooth muscle MLCK (short MLCK, 130 kDa) was observed in their study. It is well known that up-regulation of epithelial MLCK is involved in the defects of intestinal barrier function, which is critical to the initiation and progression of inflammatory bowel disease [2, 3]. The relationship between TNF-a-induced enhancement of epithelial MLCK expression and barrier dysfunction in inflammatory bowel disease is extensively studied; epithelial MLCK expression in different stages of inflammation may vary because the degree of inflammation and epithelial damage are different at different times [4]. Though claudin-2 up-regulation is prevented by epithelial MLCK inhibition [2], there is still an incomplete understanding of the causal relationship between the epithelial MLCK expression and claudin-2 expression in inflammatory bowel disease. Smooth muscle MLCK is transcribed from the same gene as epithelial MLCK, and the gene is the mylk 1 gene [5]. Smooth muscle MLCK plays an important role in the regulation of smooth muscle contraction, and variation of smooth muscle MLCK content leads to dysmotility [6]. The motility disorders secondarily cause abnormal growth of the intestinal flora, and the induced disturbance of this flora aggravates the pathogenesis of intestinal inflammation [7]. Based on the above information, it is critical to reveal that the epithelial and smooth muscle MLCK contents in different tissue layers and in different stages of intestinal inflammation can be observed. Furthermore, anti-MLCK antibody cross-reacts with both the short MLCK and long MLCK [5], and there should be two bands in the Western blotting image for determination of MLCK, which represent epithelial MLCK (220 kDa) and smooth muscle MLCK (130 kDa), respectively. Thanks very much to Digestive Diseases and Sciences for giving us the chance to discuss the hot topic regarding treating inflammatory bowel disease; thanks too for the contribution by Nishida et al.


Acta Pharmacologica Sinica | 2017

Geniposide ameliorates TNBS-induced experimental colitis in rats via reducing inflammatory cytokine release and restoring impaired intestinal barrier function

Bin Xu; Yanli Li; Ming Xu; Chang-chun Yu; Mengqiao Lian; Zeyao Tang; Chuan-xun Li; Yuan Lin

Geniposide is an iridoid glycosides purified from the fruit of Gardenia jasminoides Ellis, which is known to have antiinflammatory, anti-oxidative and anti-tumor activities. The present study aimed to investigate the effects of geniposide on experimental rat colitis and to reveal the related mechanisms. Experimental rat colitis was induced by rectal administration of a TNBS solution. The rats were treated with geniposide (25, 50 mg·kg−1·d−1, ig) or with sulfasalazine (SASP, 100 mg·kg−1·d−1, ig) as positive control for 14 consecutive days. A Caco-2 cell monolayer exposed to lipopolysaccharides (LPS) was used as an epithelial barrier dysfunction model. Transepithelial electrical resistance (TER) was measured to evaluate intestinal barrier function. In rats with TNBS-induced colitis, administration of geniposide or SASP significantly increased the TNBS-decreased body weight and ameliorated TNBS-induced experimental colitis and related symptoms. Geniposide or SASP suppressed inflammatory cytokine (TNF-α, IL-1β, and IL-6) release and neutrophil infiltration (myeloperoxidase activity) in the colon. In Caco-2 cells, geniposide (25–100 μg/mL) ameliorated LPS-induced endothelial barrier dysfunction via dose-dependently increasing transepithelial electrical resistance (TER). The results from both in vivo and in vitro studies revealed that geniposide down-regulated NF-κB, COX-2, iNOS and MLCK protein expression, up-regulated the expression of tight junction proteins (occludin and ZO-1), and facilitated AMPK phosphorylation. Both AMPK siRNA transfection and AMPK overexpression abrogated the geniposide-reduced MLCK protein expression, suggesting that geniposide ameliorated barrier dysfunction via AMPK-mediated inhibition of the MLCK pathway. In conclusion, geniposide ameliorated TNBS-induced experimental rat colitis by both reducing inflammation and modulating the disrupted epithelial barrier function via activating the AMPK signaling pathway.

Collaboration


Dive into the Yuan Lin's collaboration.

Top Co-Authors

Avatar

Dapeng Chen

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Zeyao Tang

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Yongjian Xiong

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Jinyong Peng

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Bochao Lv

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Li Wang

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Bin Sun

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Peng Chu

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Fangfei Liu

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Guozhu Han

Dalian Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge