Network


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

Hotspot


Dive into the research topics where Feng Ren is active.

Publication


Featured researches published by Feng Ren.


Cell Death and Disease | 2014

Peroxisome proliferator-activated receptor α activation attenuates the inflammatory response to protect the liver from acute failure by promoting the autophagy pathway

M Jiao; Feng Ren; Lin Zhou; X Zhang; Lei Zhang; T Wen; L Wei; Xin Wang; H Shi; L Bai; Shusen Zheng; Jian Zhang; Youhai H. Chen; Y Han; C Zhao; Z Duan

Peroxisome proliferator-activated receptor α (PPARα) has been reported to induce a potent anti-inflammatory response. Autophagy is a recently recognized rudimentary cellular response to inflammation and injury. The aim of the present study was to test the hypothesis that PPARα activation mediates autophagy to inhibit liver inflammation and protect against acute liver failure (ALF). PPARα expression during ALF and the impact of PPARα activation by Wy-14 643 on the hepatic immune response were studied in a D-galactosamine/lipopolysaccharide-induced mouse model. Autophagy was inhibited by 3-methyladenine or small interfering RNA (siRNA) against Atg7. In both the mouse model and human ALF subjects, PPARα was significantly downregulated in the injured liver. PPARα activation by pretreatment with Wy-14 643 protected against liver injury in mice. The protective effect of PPARα activation relied on the suppression of inflammatory mechanisms through the induction of autophagy. This hypothesis is supported by the following evidence: first, PPARα activation suppressed proinflammatory responses and inhibited phosphorylated NF-κBp65, phosphorylated JNK and phosphorylated ERK pathways in vivo. Second, protection by PPARα activation was due to the induction of autophagy because inhibition of autophagy by 3-methyladenine or Atg7 siRNA reversed liver protection and inflammation. Third, PPARα activation directly induced autophagy in primary macrophages in vitro, which protected cells from a lipopolysaccharide-induced proinflammatory response. Here, for the first time, we have demonstrated that PPARα-mediated induction of autophagy ameliorated liver injury in cases of ALF by attenuating inflammatory responses, indicating a potential therapeutic application for ALF treatment.


Journal of Cellular Biochemistry | 2013

Circulating histones exacerbate inflammation in mice with acute liver failure

Zongmei Wen; Yan Liu; Feng Li; Feng Ren; Dexi Chen; Xiuhui Li; Tao Wen

Circulating histones are a newly recognized mediator implicated in various inflammatory diseases. It is likely that the release of histones, from dying hepatocytes or inflammatory leukocytes, into the circulation initiates and amplifies inflammation during the course of acute liver failure (ALF). In this study, we investigated a putative pathogenic role of circulating histones in a murine model of ALF induced by D‐galactosamine (GalN) plus lipopolysaccharide (LPS). Hepatic function and histological indexes, myeloperoxidase (MPO) activity, hepatocyte apoptosis and the levels of circulating histone were measured in GalN/LPS‐treated mice. GalN/LPS caused severe liver damage and a notable increase in plasma concentration of circulating histones. To further assess the role of circulating histones in our model, we administered exogenous histones and anti‐histone H4 antibody. Notably, exogenous histones aggravated GalN/LPS‐induced hepatotoxicity, whereas anti‐histone antibody significantly protected mice. Circulating histones may serve as both a functional marker of ALF activity and as an inflammatory mediator contributing to the progression of ALF. Blockade of circulating histones shows potent protective effects, suggesting a potential therapeutic strategy for ALF. J. Cell. Biochem. 114: 2384–2391, 2013.


PLOS ONE | 2012

Inhibition of glycogen synthase kinase 3β ameliorates D-GalN/LPS-induced liver injury by reducing endoplasmic reticulum stress-triggered apoptosis.

Li-Yan Chen; Feng Ren; Haiyan Zhang; T. Wen; Zhengfu Piao; Li Zhou; Sujun Zheng; Jing Zhang; Yu Chen; Yuan-Ping Han; Zhong-Ping Duan; Ying-Ji Ma

Background Glycogen synthase kinase 3β(GSK3β) is a ubiquitous serine-threonine protein kinase that participates in numerous cellular processes and disease pathophysiology. We aimed to determine therapeutic potential of GSK3β inhibition and its mechanism in a well-characterized model of lipopolysaccharide (LPS)-induced model of acute liver failure (ALF). Methodology In a murine ALF model induced by D-GalN(700 mg/kg)/LPS(10 µg/kg), we analyzed GSK3β mechanisms using a specific chemical inhibitor, SB216763, and detected the role of endoplasmic reticulum stress (ERS). Mice were administered SB216763 at 2 h before or after D-GalN/LPS injection, respectively, and then sacrificed 6 h after D-GalN/LPS treatment to evaluate its prophylactic and therapeutic function. The lethality rate, liver damage, ERS, cytokine expression, MAP kinase, hepatocyte apoptosis and expression of TLR 4 were evaluated, respectively. Whether the inhibition of GSK3β activation protected hepatocyte from ERS-induced apoptosis was investigated in vitro. Principal Findings GSK3β became quickly activated (dephosphorylated) upon D-GalN/LPS exposure. Administration of SB216763 not only ameliorated liver injury, as evidenced by reduced transaminase levels, and well-preserved liver architecture, but also decreased lethality. Moreover, GSK3β inhibition resulted in down-regulation of pro-apoptotic proteins C/EBP–homologous protein(CHOP) and caspase-12, which are related to ERS. To further demonstrate the role of ERS, we found that GSK3β inhibition protected hepatocyte from ERS-induced cell death. GSK3β inhibition down-regulated the MAPK pathways, reduced expression of inflammatory cytokines and decreased expression of TLR4. Conclusions Our findings demonstrate the key function of GSK3β signaling in the pathophysiology of ALF, especially in regulating the ERS, and provide a rationale for targeting GSK3β as a potential therapeutic strategy to ameliorate ALF.


World Journal of Gastroenterology | 2014

Prognostic value of M30/M65 for outcome of hepatitis B virus-related acute-on-chronic liver failure.

Su-Jun Zheng; Shuang Liu; Mei Liu; Malcolm A McCrae; Jun-Feng Li; Yuan-Ping Han; Chunhui Xu; Feng Ren; Yu Chen; Zhongping Duan

AIM To determine the prognostic value of circulating indicators of cell death in acute-on-chronic liver failure (ACLF) patients with chronic hepatitis B virus (HBV) infection as the single etiology. METHODS Full length and caspase cleaved cytokeratin 18 (detected as M65 and M30 antigens) represent circulating indicators of necrosis and apoptosis. M65 and M30 were identified by enzyme-linked immunosorbent assay in 169 subjects including healthy controls (n = 33), patients with chronic hepatitis B (CHB, n = 55) and patients with ACLF (n = 81). According to the 3-mo survival period, ACLF patients were defined as having spontaneous recovery (n = 33) and non-spontaneous recovery which included deceased patients and those who required liver transplantation (n = 48). RESULTS Both biomarker levels significantly increased gradually as liver disease progressed (for M65: P < 0.001 for all; for M30: control vs CHB, P = 0.072; others: P < 0.001 for all). In contrast, the M30/M65 ratio was significantly higher in controls compared with CHB patients (P = 0.010) or ACLF patients (P < 0.001). In addition, the area under receiver operating characteristic curve (AUC) analysis demonstrated that both biomarkers had diagnostic value (AUC ≥ 0.80) in identifying ACLF from CHB patients. Interestingly, it is worth noting that the M30/M65 ratio was significantly different between spontaneous and non-spontaneous recovery in ACLF patients (P = 0.032). The prognostic value of the M30/M65 ratio was compared with the Model for End-Stage Liver Disease (MELD) and Child-Pugh scores at the 3-mo survival period, the AUC of the M30/M65 ratio was 0.66 with a sensitivity of 52.9% and the highest specificity of 92.6% (MELD:AUC = 0.71; sensitivity, 79.4%; specificity, 63.0%; Child-Pugh: AUC = 0.77; sensitivity, 61.8%; specificity, 88.9%). CONCLUSION M65 and M30 are strongly associated with liver disease severity. The M30/M65 ratio may be a potential prognostic marker for spontaneous recovery in patients with HBV-related ACLF.


International Journal of Infectious Diseases | 2015

Serum sphingomyelin has potential to reflect hepatic injury in chronic hepatitis B virus infection

Sujun Zheng; Feng Qu; Jun-Feng Li; Jing Zhao; Jing-Yun Zhang; Mei Liu; Feng Ren; Yu Chen; Jinlan Zhang; Zhongping Duan

OBJECTIVE To explore the relation between serum sphingolipids and hepatic injury in chronic HBV infection. METHODS A cohort of participants including 48 healthy persons, 103 chronic HBV-infected patients containing chronic hepatitis B (CHB) and HBV-related cirrhosis were included. High performance liquid chromatography coupled to tandem mass spectrometry (HPLC-MS/MS) was performed to detect serum sphingolipids. The serological indicators were detected and quantified. The valid liver biopsy specimens were acquired from twenty five CHB. RESULTS Twenty four serum sphingolipids were detected. There were eighteen sphingolipids showing significant differences between the healthy control and chronic HBV infection groups. In patients with chronic HBV infection, fourteen sphingolipids differed significantly between CHB and HBV-related cirrhosis. Among sphingolipids with a significant difference in both HBV infection vs healthy control and CHB vs cirrhosis, seven sphingolipids were independently related to the presence of cirrhosis. SM(d18:1/24:0), a sphingomyelin (SM) compound, was found to have a negative correlation with model for end-stage liver disease (MELD) score. Additionally, SM(d18:1/24:0) was demonstrated to have a correlation with inflammation grades by liver biopsy in CHB patients. CONCLUSIONS Serum sphingolipids have close relation with hepatic injury in chronic HBV infection, especially that SM(d18:1/24:0) might be a potential serum biomarker.


Inflammation | 2015

Endoplasmic Reticulum Stress-Activated Glycogen Synthase Kinase 3β Aggravates Liver Inflammation and Hepatotoxicity in Mice with Acute Liver Failure

Feng Ren; Li Zhou; Xiangying Zhang; Tao Wen; Hongbo Shi; Bangxiang Xie; Zhuo Li; Dexi Chen; Zheling Wang; Zhongping Duan

Endoplasmic reticulum stress (ER stress) has been increasingly recognized as an important mechanism in various liver diseases. However, its intrinsic physiological role in acute liver failure (ALF) remains largely undetermined. This study aimed to examine how ER stress orchestrates glycogen synthase kinase 3β (GSK3β) and inflammation to affect ALF. In a murine ALF model induced by d-galactosamine (d-GalN) and lipopolysaccharide (LPS), 4-phenylbutyric acid (4-PBA) is to be administered to relieve ER stress. The lethality rate, liver damage, cytokine expression, and the activity of GSK3β were evaluated. How to regulate LPS-induced inflammation and TNF-α-induced hepatocyte apoptosis by ER stress was investigated in vitro. In vivo, ER stress was triggered in the liver with the progression of mice ALF model. ER stress was essential for the development of ALF because ER stress inhibition by 4-PBA ameliorated the liver damage through decreasing liver inflammation and hepatocyte apoptosis. 4-PBA also decreased GSK3β activity in the livers of ALF mice. In vitro, ER stress induced by tunicamycin synergistically increased LPS-triggered pro-inflammatory cytokine induction and promoted the activation of nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK) pathway in bone marrow-derived macrophages; moreover, tunicamycin also cooperated with TNF-α to increase hepatocyte apoptosis. ER stress promoted LPS-triggered inflammation depending on GSK3β activation because inhibition of GSK3β by SB216763, the specific inhibitor of GSK3β, resulted in downregulation of pro-inflammatory genes. ER stress contributes to liver inflammation and hepatotoxicity in ALF, particularly by regulating GSK3β, and is therefore a potential therapeutic target for ALF.


Journal of Ethnopharmacology | 2017

Qingchangligan formula attenuates the inflammatory response to protect the liver from acute failure induced by d-galactosamine/lipopolysaccharide in mice

Xiangying Zhang; Jianbo Ding; Chunyan Gou; Tao Wen; Li Li; Xiaojun Wang; Huasheng Yang; Dan Liu; Jinli Lou; Dexi Chen; Feng Ren; Xiuhui Li

ETHNOPHARMACOLOGICAL RELEVANCE The Qingchangligan formula, a traditional Chinese medicine comprising five herbs, is useful for treatment of patients with liver failure; however, its protective and regulatory mechanisms remain elusive. AIM OF THE STUDY To test the hypothesis that the Qingchangligan formula protects mice against acute liver failure by inhibiting liver inflammation. MATERIALS AND METHODS Acute liver failure (ALF) was induced by intraperitoneal injection of D-GalN (700mg/kg) plus LPS (10μg/kg). The Qingchangligan formula was administered to mice in three doses of 50mg/kg (on day 1, day 2, and day 3) prior to D-GalN/LPS injection by intragastric administration. The mice in different groups were sacrificed at 6h after D-GalN/LPS injection, and liver samples and blood were collected for analysis. RESULTS Administration of the Qingchangligan formula not only ameliorated liver injury, as evidenced by reduced transaminase levels and well-preserved liver architecture, but also decreased the lethality in ALF mice. Moreover, in the ALF model, pretreatment with the Qingchangligan formula alleviated liver inflammation and decreased hepatocyte apoptosis. Further demonstrating the protective effects of the Qingchangligan formula, we found that pretreatment with the Qingchangligan formula reduced the expression of inflammatory cytokines by decreasing the expression of components of the mitogen-activated protein kinase (MAPK) pathway and promoting autophagy in vitro and in vivo. CONCLUSIONS Our findings demonstrated that the Qingchangligan formula exerts a protective effect against the pathophysiology of ALF, especially in regulating liver inflammation, and provide a rationale for using the Qingchangligan formula as a potential therapeutic strategy to ameliorate ALF.


International Journal of Infectious Diseases | 2014

Fibrosis progression in interferon treatment-naive Chinese plasma donors with chronic hepatitis C for 20 years: a cohort study

Jun-Feng Li; Shuang Liu; Feng Ren; Mei Liu; Hui-Li Wu; Yu Chen; Huaibin Zou; Li Bai; Ying Li; Su-Jun Zheng; Zhong-Ping Duan

OBJECTIVES To evaluate the progression of fibrosis and factors influencing this in interferon (IFN) treatment-naive Chinese plasma donors infected with hepatitis C virus (HCV) for approximately 20 years. METHODS From July 2010 to June 2011, we investigated 122 IFN treatment-naive chronic hepatitis C (CHC) patients infected by plasma donation in 1992-1995. Liver fibrosis stage and inflammation grade were evaluated by Metavir and Scheuer scoring systems, respectively. RESULTS One hundred and twenty patients underwent liver biopsy. Liver biopsy was not performed in one patient with cirrhosis due to ascites, and another patient was excluded because of an invalid biopsy specimen. Cirrhosis was observed in three patients (fibrosis stage F4 in two patients revealed by biopsy, and one patient with ascites confirmed by physical and Doppler ultrasound examination). Fibrosis stages F1 and F2 were present in 55 and 50 patients, respectively. The severity of liver inflammation was independently related to moderate to severe fibrosis (F ≥2). Older age and male sex showed an increasing tendency for more severe fibrosis (F3/F4) in the present cohort. CONCLUSIONS Based on histopathology results, the progression of fibrosis in patients with CHC infected by repeated plasma donation is slow after HCV infection of approximately 20 years. Liver inflammation is closely related to the development of moderate to severe liver fibrosis.


PLOS ONE | 2014

Plasma Sphingolipids as Potential Indicators of Hepatic Necroinflammation in Patients with Chronic Hepatitis C and Normal Alanine Aminotransferase Level

Junfeng Li; Feng Qu; Sujun Zheng; Jin-Yu Ren; Hui-Li Wu; Mei Liu; Hui Liu; Feng Ren; Yu Chen; Jinlan Zhang; Zhong-Ping Duan

Accurate estimation of hepatic necroinflammation caused by chronic hepatitis C (CHC) is crucial for prediction of prognosis and design of therapeutic strategy, which is particularly true for CHC patients with normal alanine aminotransferase (ALT) level. Recent studies have shown that sphingolipids have a close relationship with hepatitis C virus infection. The present study aimed to identify plasma sphingolipids related to hepatic necroinflammation. We included 120 treatment-naïve CHC patients and 64/120 had normal ALT levels (<40 U/L). CHC patients who underwent liver biopsies were subjected to Scheuer scoring analysis for scope of hepatic inflammation. Plasma sphingolipids were detected by high-performance liquid chromatography tandem mass spectrometry. Our results showed 44 plasma sphingolipids were detected altogether. Of all detected sphingolipids, hexosylceramide (HexCer) (d18∶1/22∶0) and HexCer (d18∶1/24∶0) showed a significant difference among G0/G1, G2, and G3/G4 (P<0.05). For identifying hepatic necroinflammation (G≥2), after adjusting other factors, the odds ratio (OR) of HexCer (d18∶1/22∶0) reached 1.01 (95% confidence interval [CI]: 1.00–1.02). Furthermore, the area under the curve (AUC) of HexCer (d18∶1/22∶0) was 0.7 (P = 0.01) and approached that of ALT (AUC = 0.78). However, in CHC patients with normal ALT, HexCer (d18∶1/22∶0) was an independent factor (OR: 1.02, 95% CI: 1.01–1.03) to identify the hepatic necroinflammation (G≥2). HexCer (d18∶1/22∶0) not only showed the largest AUC (0.78, P = 0.001), but also exhibited the highest specificity of all indicators. These results indicate that plasma HexCer (d18∶1/22∶0) is a potential indicator to distinguish hepatic necroinflammation in CHC patients. For CHC with normal ALT, the ability of HexCer (d18∶1/22∶0) to distinguish hepatic necroinflammation might be superior to conventional serum indicators.


Molecular Medicine Reports | 2016

Kaempferol induces apoptosis in HepG2 cells via activation of the endoplasmic reticulum stress pathway

Haiqing Guo; Feng Ren; Li Zhang; Xiangying Zhang; Rongrong Yang; Bangxiang Xie; Zhuo Li; Zhongjie Hu; Zhongping Duan; Jing Zhang

Kaempferol is a flavonoid compound that has gained importance due to its antitumor properties; however, the underlying mechanisms remain to be fully understood. The present study aimed to investigate the molecular mechanisms of the antitumor function of kaempferol in HepG2 hepatocellular carcinoma cells. Kaempferol was determined to reduce cell viability, increase lactate dehydrogenase activity and induce apoptosis in a concentration‑ and time‑dependent manner in HepG2 cells. Additionally, kaempferol‑induced apoptosis possibly acts via the endoplasmic reticulum (ER) stress pathway, due to the significant increase in the protein expression levels of glucose‑regulated protein 78, glucose‑regulated protein 94, protein kinase R‑like ER kinase, inositol‑requiring enzyme 1α, partial activating transcription factor 6 cleavage, caspase‑4, C/EBP homologous protein (CHOP) and cleaved caspase‑3. The pro‑apoptotic activity of kaempferol was determined to be due to induction of the ER stress‑CHOP pathway, as: i) ER stress was blocked by 4‑phenyl butyric acid (4‑PBA) pretreatment and knockdown of CHOP with small interfering RNA, which resulted in alleviation of kaempferol‑induced HepG2 cell apoptosis; and ii) transfection with plasmid overexpressing CHOP reversed the protective effect of 4‑PBA in kaempferol‑induced HepG2 cells and increased the apoptotic rate. Thus, kaempferol promoted HepG2 cell apoptosis via induction of the ER stress‑CHOP signaling pathway. These observations indicate that kaempferol may be used as a potential chemopreventive treatment strategy for patients with hepatocellular carcinoma.

Collaboration


Dive into the Feng Ren's collaboration.

Top Co-Authors

Avatar

Zhongping Duan

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Yu Chen

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Sujun Zheng

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Dexi Chen

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Hongbo Shi

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Mei Liu

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Jun-Feng Li

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Li Bai

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiangying Zhang

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiaohui Zhang

Capital Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge