Network


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

Hotspot


Dive into the research topics where Kefeng Dou is active.

Publication


Featured researches published by Kefeng Dou.


Cancer Letters | 2015

Circular RNA: A new star of noncoding RNAs

Shibin Qu; Xisheng Yang; Xiaolei Li; Jianlin Wang; Yuan Gao; Runze Shang; Wei Sun; Kefeng Dou; Haimin Li

Circular RNAs (circRNAs) are a novel type of RNA that, unlike linear RNAs, form a covalently closed continuous loop and are highly represented in the eukaryotic transcriptome. Recent studies have discovered thousands of endogenous circRNAs in mammalian cells. CircRNAs are largely generated from exonic or intronic sequences, and reverse complementary sequences or RNA-binding proteins (RBPs) are necessary for circRNA biogenesis. The majority of circRNAs are conserved across species, are stable and resistant to RNase R, and often exhibit tissue/developmental-stage-specific expression. Recent research has revealed that circRNAs can function as microRNA (miRNA) sponges, regulators of splicing and transcription, and modifiers of parental gene expression. Emerging evidence indicates that circRNAs might play important roles in atherosclerotic vascular disease risk, neurological disorders, prion diseases and cancer; exhibit aberrant expression in colorectal cancer (CRC) and pancreatic ductal adenocarcinoma (PDAC); and serve as diagnostic or predictive biomarkers of some diseases. Similar to miRNAs and long noncoding RNAs (lncRNAs), circRNAs are becoming a new research hotspot in the field of RNA and could be widely involved in the processes of life. Herein, we review the formation and properties of circRNAs, their functions, and their potential significance in disease.


Hepatology | 2014

c‐Myc‐mediated epigenetic silencing of MicroRNA‐101 contributes to dysregulation of multiple pathways in hepatocellular carcinoma

Lei Wang; Xiang Zhang; Lin-Tao Jia; Sijun Hu; Jing Zhao; Jiandong Yang; Weihong Wen; Zhe Wang; Tao Wang; Jun Zhao; Ruian Wang; Yan-Ling Meng; Yongzhan Nie; Kefeng Dou; Si-Yi Chen; Libo Yao; Daiming Fan; Rui Zhang; An-Gang Yang

The MYC oncogene is overexpressed in hepatocellular carcinoma (HCC) and has been associated with widespread microRNA (miRNA) repression; however, the underlying mechanisms are largely unknown. Here, we report that the c‐Myc oncogenic transcription factor physically interacts with enhancer of zeste homolog 2 (EZH2), a core enzymatic unit of polycomb repressive complex 2 (PRC2). Furthermore, miR‐101, an important tumor‐suppressive miRNA in human hepatocarcinomas, is epigenetically repressed by PRC2 complex in a c‐Myc‐mediated manner. miR‐101, in turn, inhibits the expression of two subunits of PRC2 (EZH2 and EED), thus creating a double‐negative feedback loop that regulates the process of hepatocarcinogenesis. Restoration of miR‐101 expression suppresses multiple malignant phenotypes of HCC cells by coordinate repression of a cohort of oncogenes, including STMN1, JUNB, and CXCR7, and further increases expression of endogenous miR‐101 by inhibition of PRC2 activation. In addition, co‐overexpression of c‐Myc and EZH2 in HCC samples was closely associated with lower expression of miR‐101 (P < 0.0001) and poorer prognosis of HCC patients (P < 0.01). Conclusions: c‐Myc collaborates with EZH2‐containing PRC2 complex in silencing tumor‐suppressive miRNAs during hepatocarcinogenesis and provides promising therapeutic candidates for human HCC. (Hepatology 2014;59:1850–1863)


Biomaterials | 2012

The differentiation of MSCs into functional hepatocyte-like cells in a liver biomatrix scaffold and their transplantation into liver-fibrotic mice.

Ru Ji; Ning Zhang; Nan You; Qiang Li; Weihui Liu; Nan Jiang; Jie Liu; Hongtao Zhang; Desheng Wang; Kaishan Tao; Kefeng Dou

Hepatocytes derived from mesenchymal stem cells (MSCs) hold great potential for cell-based therapies for liver diseases. The cell-based therapies are critically dependent on the hepatic differentiation of the MSCs with a high efficiency and on a considerable scale. Recent results have shown that decellularized organs provide a three-dimensional extracellular matrix for the lineage restriction of stem cell maturation. In this study, we compared the cell proliferation and hepatic differentiation of murine MSCs in a biomatrix scaffold from rat liver and in the presence and absence growth factors (GF) with a two-dimensional substrate. In the absence or presence of GF, the dynamic cultured scaffold (DCS) stimulated the MSCs to express endodermal and hepatocyte-specific genes and proteins associated with improved functions, and the cells exhibited the ultrastructural characteristics of mature hepatocytes. When transplanted into CCl(4)-injured mice, the cells pretreated with a combination of the DCS and GF exhibited increased survival, liver function, engraftment into the host liver and further hepatic differentiation. The paracrine effect of the transplanted cells on hepatic stellate cells and native hepatocytes played a key role in the treatment of the liver pathology. These studies define an effective method that facilitates the hepatic differentiation of MSCs exhibiting extensive functions and support further research into the use of a decellularized liver matrix as a bioscaffold for liver tissue engineering.


Cancer Science | 2010

Bmi‐1 is related to proliferation, survival and poor prognosis in pancreatic cancer

Wenjie Song; Kaishan Tao; Haimin Li; Chen Jin; Zhenshun Song; Jun Li; Hai Shi; Xiao Li; Zheng Dang; Kefeng Dou

B‐cell‐specific Moloney murine leukemia virus insertion site 1 (BMI1) is a member of the polycomb group of transcriptional repressors. Until now, its expression and functional significance in pancreatic carcinogenesis was unknown. In the present study, we demonstrated that expression of BMI1 was markedly up‐regulated in pancreatic cancer cell lines and surgically resected cancer specimens. In addition, BMI1 expression levels correlated positively with the presence of lymph node metastases and negatively with patient survival rates, suggesting a role for BMI1 in the progression of pancreatic cancer. Furthermore, stable down‐regulation of BMI1 suppressed cell growth, delayed the G1/S transition, and enhanced the susceptibility of different pancreatic cell lines to apoptosis following expression of a lentiviral‐mediated shRNA targeted for BMI1. Expression of the short‐hairpin RNA also correlated with the up‐regulation of p21 and Bax and the down‐regulation of cyclin D1, cyclin‐dependent kinase (CDK)‐2 and ‐4, Bcl‐2, and phospho‐Akt. Finally, growth suppression following BMI1 depletion was confirmed in a nude mouse model. In conclusion, our findings indicate that BMI1 plays an important role in the late progression of pancreatic cancer and may represent a novel therapeutic target for the treatment of pancreatic cancer. (Cancer Sci 2010)


Tumor Biology | 2012

MicroRNA-10b promotes migration and invasion through CADM1 in human hepatocellular carcinoma cells

Qing-jun Li; Liang Zhou; Fan Yang; Guo-xia Wang; Hang Zheng; Desheng Wang; Yong He; Kefeng Dou

MicroRNA-10b (miR-10b) was recently reported to be dysregulated in some types of cancer and to play a role in invasion and metastasis. However, effects and potential mechanisms of action of miR-10b in the metastasis of hepatocellular carcinoma (HCC) have not been explored. In this study, we confirmed that miR-10b is highly expressed in metastatic HCC tissues and in metastatic HCC cell lines by qRT-PCR. Moreover, patients with higher miR-10b expression had significantly poorer overall survival, and high miR-10b expression was an independent predictor of poor prognosis. Inhibition of miR-10b reduced cell migration and invasion in MHCC97H cells, whereas over-expression of miR-10b in HepG2 cells increased cell migration and invasion. Bioinformatics and luciferase reporter assays revealed that miR-10b binds the 3′-UTR of CADM1 mRNA and represses its translation. Western blot and qRT-PCR showed that CADM1 is inhibited by miR-10b over-expression. Silencing of CADM1 resulted in substantially increased cell motility and invasion similar to that observed with over-expression of miR-10b in HepG2 cells. These results suggest that miR-10b may positively regulate the invasion and metastasis of HCC through targeting CADM1.


Tumor Biology | 2017

Evaluation of sorafenib in Chinese unresectable hepatocellular carcinoma patients with prior surgery and portal vein tumor thrombosis: A subset analysis of GIDEON study data:

Sheng Long Ye; Xiaoping Chen; Jiamei Yang; Ping Bie; Shuijun Zhang; Fengyong Liu; Luming Liu; Jie Zhou; Kefeng Dou; Christina S.M. Yip; Xiaolei Yang

The purpose of this study was to examine the safety and efficacy of sorafenib in Chinese patients with unresectable hepatocellular carcinoma. Data of 338 Chinese patients from the Global Investigation of therapeutic DEcisions in hepatocellular carcinoma and Of its treatment with sorafeNib study database were included. Patients were divided into those who received and did not receive sorafenib prior to surgical resection and those with and without portal vein tumor thrombosis. In the non-surgery group, the median survival was 302 days (95% confidence interval: 244–371), and the median time from diagnosis to death was 428 days (95% confidence interval: 352–556); in the surgery group, half of the patients survived for 345 days and the median time from diagnosis to death was 1000 days (95% confidence interval: 750–2816). Median progression-free survival and median time to progression were not different between the two groups. Median overall survival was 360 days (95% confidence interval: 309–435) in the non–portal vein tumor thrombosis group and 240 days (95% confidence interval: 181–296) in the portal vein tumor thrombosis group; median time between hepatocellular carcinoma diagnosis and death was 750 days (95% confidence interval: 472–1000) and 420 days (95% confidence interval: 252–567), respectively, in the two groups. Median progression-free survival was 209 days (95% confidence interval: 166–264) for patients without portal vein tumor thrombosis and 154 days (95% confidence interval: 112–202) for patients with portal vein tumor thrombosis; median time to progression was 295 days (95% confidence interval: 209–463) and 221 days, respectively. Adverse events were generally comparable regardless of prior surgery and portal vein tumor thrombosis status. We thus conclude that earlier administration of sorafenib may result in improved outcomes in patients with unresectable hepatocellular carcinoma and portal vein tumor thrombosis.


International Journal of Oncology | 2013

MicroRNA-21 regulates the migration and invasion of a stem-like population in hepatocellular carcinoma

Liang Zhou; Zhaoxu Yang; Wenjie Song; Qing-jun Li; Fan Yang; Desheng Wang; Ning Zhang; Kefeng Dou

Due to invasion and intrahepatic metastasis, the prognosis for patients with hepatocellular carcinoma (HCC) is poor. However, the mechanisms underlying these processes of HCC remain unclear. Cancer stem cells may be involved in early systemic dissemination and metastasis formation and side population (SP) cells isolated from diverse cancer cells possess stem cell-like properties. However, the mechanisms involved in migration and invasion of cancer stem cells are not well understood. In this study, we identified and isolated populations of SP cells from HCC cell lines using flow cyto-metry. SP cells showed higher levels of migration and invasion capability. Higher expression of miR-21 was observed in SP cells. Silencing of miR-21 led to a reduction in the migration and invasion of these cells and overexpression of miR-21 can increase in cell migration and invasion. Overexpression of miR-21 did not cause degradation of PTEN or RECK or PDCD4 mRNA but drastically inhibited its protein expression. Consistent with these results, silencing miR-21 increased the levels of PTEN, RECK and PDCD4 protein, respectively. The role of silencing miR-21 was partially attenuated by silencing of PTEN or RECK or PDCD4 mRNA. The results of this study revealed the aberrant expression of miR-21 in SP cells and showed that miR-21 regulates the expression of multiple target proteins that are associated with tumor dissemination. MiR-21 is a pro-metastatic miRNA in SP cells and raises the possibility that therapy of HCC may be improved by pharmaceutical strategies directed towards miR-21.


PLOS ONE | 2012

Tolerance Induction by Exosomes from Immature Dendritic Cells and Rapamycin in a Mouse Cardiac Allograft Model

Xiao-xiao Li; Junjie Li; Jing-Yue Yang; Desheng Wang; Wei Zhao; Wenjie Song; Wei-Min Li; Jian Feng Wang; Wei Han; Zhuochao Zhang; Yong Yu; Da-Yong Cao; Kefeng Dou

Background Dendritic cells (DCs) release bioactive exosomes that play an important role in immune regulation. Because they express low levels of class I major histocompatibility complex (MHC) and co-stimulatory molecules, exosomes derived from donor immature DCs (imDex) prolong allograft survival by inhibiting T-cell activation. However, this effect is limited and does not induce immunological tolerance when imDex are administered alone. Thus, we tested the effect of combined treatment with donor imDex and low-dose rapamycin on inducing tolerance in a mouse cardiac transplantation model. Methods ImDex were obtained from the culture supernatant of immature DCs derived from donor mouse (C57BL/6) bone marrow and were injected with suboptimal doses of rapamycin into recipient mouse (BALB/c) before and after transplantation. The capacity of this treatment to induce immune tolerance was analyzed in vitro and in vivo using the mouse cardiac transplantation model. Results Donor imDex expressed moderate levels of MHC class II and low levels of MHC class I and co-stimulatory molecules, but neither imDex nor subtherapeutic rapamycin dose alone induced cardiac allograft tolerance. Combined treatment with imDex and rapamycin, however, led to donor specific cardiac allograft tolerance. This effect was accompanied by decreased anti-donor antigen cellular response and an increased percentage of spleen CD4+CD25+ T cells in recipients. Furthermore, this donor specific tolerance could be further transferred to naïve allograft recipients through injection of splenocytes, but not serum, from tolerant recipients. Conclusion Combined with immunosuppressive treatment, donor imDex can prolong cardiac allograft survival and induce donor specific allograft tolerance.


Hepatology | 2011

Canonical notch pathway protects hepatocytes from ischemia/reperfusion injury in mice by repressing reactive oxygen species production through JAK2/STAT3 signaling

Heng-Chao Yu; Hong-Yan Qin; Fei He; Lin Wang; Wei Fu; Dong Liu; Feng‐Cheng Guo; Liang Liang; Kefeng Dou; Hua Han

Hepatic ischemia/reperfusion (I/R) injury is initiated by reactive oxygen species (ROS) accumulated during the early reperfusion phase after ischemia, but cellular mechanisms controlling ROS production and scavenging have not been fully understood. In this study, we show that blocking Notch signal by knockout of the transcription factor RBP‐J or a pharmacological inhibitor led to aggravated hepatic I/R injury, as manifested by deteriorated liver function and increased apoptosis, necrosis, and inflammation, both in vitro and in vivo. Interruption of Notch signaling resulted in increased intracellular ROS in hepatocytes, and a ROS scavenger cured exacerbated hepatic I/R injury after Notch signaling blockade, suggesting that Notch signal deficiency aggravated I/R injury through increased ROS levels. Notch signal blockade resulted in down‐regulation of Hes5, leading to reduced formation of the Hes5‐STAT3 complex and hypophosphorylation of STAT3, which further attenuated manganese superoxide dismutase (MnSOD) expression and increased ROS and apoptosis. Indeed, overexpression of a constitutively active STAT3 rescued MnSOD expression and I/R injury–induced apoptosis in the absence of Notch signaling. Finally, forced Notch activation by ligand stimulation or Hes5 overexpression reduced intracellular ROS and protected hepatocytes from apoptosis after I/R injury through the activation of STAT3 and MnSOD expression.


International Journal of Biological Sciences | 2014

Long non-coding RNA URHC regulates cell proliferation and apoptosis via ZAK through the ERK/MAPK signaling pathway in hepatocellular carcinoma.

Wei-Hua Xu; Jian-Bin Zhang; Zheng Dang; Xiao Li; Ti Zhou; Jie Liu; Desheng Wang; Wen-Jie Song; Kefeng Dou

Long non-coding RNAs (lncRNAs) have previously been implicated in human disease states, especially cancer. Although the aberrant expression of lncRNAs has been observed in cancer, the biological functions and molecular mechanisms underlying aberrantly expressed lncRNAs in hepatocellular carcinoma (HCC) have not been widely established. In the present study, we investigated a novel lncRNA, termed URHC (up-regulated in hepatocellular carcinoma), and evaluated its role in the progression of HCC. Expression profiling using a lncRNA microarray revealed that URHC was highly expressed in 3 HCC cell lines compared to normal hepatocytes. Quantitative real-time polymerase chain reaction (qRT-PCR) analyses confirmed that URHC expression was increased in hepatoma cells and HCC tissues. Moreover, using qRT-PCR, we confirmed that URHC expression was up-regulated in 30 HCC cases (57.7%) and that its higher expression was correlated with poor overall survival. We further demonstrated that URHC inhibition reduced cell proliferation and promoted apoptosis. We hypothesize that URHC may function by regulating the sterile alpha motif and leucine zipper containing kinase AZK (ZAK) gene, which is located near URHC on the same chromosome. We found that ZAK mRNA levels were down-regulated in HCC tissues and the expression levels of ZAK were negatively correlated with those of URHC in the above HCC tissues. Next, we confirmed that URHC down-regulated ZAK, which is involved in URHC-mediated cell proliferation and apoptosis. Furthermore, ERK/MAPK pathway inactivation partially accounted for URHC-ZAK-induced cell growth and apoptosis. Thus, we concluded that high URHC expression can promote cell proliferation and inhibit apoptosis by repressing ZAK expression through inactivation of the ERK/MAPK pathway. These findings may provide a novel mechanism and therapeutic targets for the treatment of HCC.

Collaboration


Dive into the Kefeng Dou's collaboration.

Top Co-Authors

Avatar

Kaishan Tao

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Desheng Wang

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Liang Zhou

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Nan You

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Jianlin Wang

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Shuqiang Yue

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Zhenshun Song

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiao Li

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Xisheng Yang

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Wei Sun

Fourth Military Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge