Network


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

Hotspot


Dive into the research topics where Daiming Fan is active.

Publication


Featured researches published by Daiming Fan.


Clinical Cancer Research | 2011

Yes-Associated Protein 1 Exhibits Oncogenic Property in Gastric Cancer and Its Nuclear Accumulation Associates with Poor Prognosis

Wei Kang; Joanna H.M. Tong; Anthony W.H. Chan; Tin-Lap Lee; Raymond Wai-Ming Lung; Patrick P.S. Leung; Ken K.Y. So; Kaichun Wu; Daiming Fan; Jun Yu; Joseph J.Y. Sung; Ka Fai To

Purpose: Yes-associated protein 1 (YAP1) is a multifunctional protein that can interact with different transcription factors to activate gene expression. The role of YAP1 in tumorigenesis is unclear. We aimed to investigate the functional role of YAP1 in tumorigenesis of gastric cancer. Experimental Design: YAP1 expresson in gastric adenocarcinoma was evaluated. The biological function was determined by proliferation assay, colony formation, cell invasion, and flow cytometric analysis through knocking down or ectopic expressing YAP1 in gastric cancer cell lines coupled with in vivo study. The possible downstream effectors of YAP1 were investigated by expression microarray. Results: YAP1 protein expression was upregulated in gastric cancer. Nuclear accumulation of YAP1 was associated with poor disease-specific survival (P = 0.021), especially in patients with early-stage diseases (P < 0.001). Knockdown YAP1 resulted in a significant reduction in proliferation, anchorage-dependent colony formation, cell invasion, and cell motility. Ectopic YAP1 expression promoted anchorage-independent colony formation, induced a more invasive phenotype, and accelerated cell growth both in vitro and in vivo. Microarray analysis highlighted the alteration of MAPK (mitogen-activated protein kinase) pathway by YAP1. We confirmed a constitutive activation of RAF/MEK/ERK (extracellular signal-regulated kinase) in YAP1-expressing MKN45 cells and further showed that YAP1 enhanced serum/epidermal growth factor–induced c-Fos expression in gastric cancer cells. Conclusions: Our findings supported that YAP1 exhibits oncogenic property in gastric cancer. We provided the first evidence that YAP1 exerted the oncogenic function by enhancing the capacity to activate the early-response gene pathway. YAP1 could be a prognostic biomarker and potential therapeutic target for gastric cancer. Clin Cancer Res; 17(8); 2130–9. ©2011 AACR.


Journal of Cellular Biochemistry | 2002

IGF-1 induces Pin1 expression in promoting cell cycle S-phase entry

Han You; Hongwu Zheng; Steven A. Murray; Qiang Yu; Takafumi Uchida; Daiming Fan; Zhi-Xiong Jim Xiao

Insulin‐like growth factor I (IGF‐1) is a well‐established mitogen to many different cell types and is implicated in progression of a number of human cancers, notably breast cancer. The prolyl isomerase Pin1 plays an important role in cell cycle regulation through its specific interaction with proteins that are phosphorylated at Ser/Thr‐Pro motifs. Pin1 knockout mice appear to have relatively normal development yet the Pin1−/−mouse embryo fibroblast (MEF) cells are defective in re‐entering cell cycle in response to serum stimulation after G0 arrest. Here, we report that Pin1−/− MEF cells display a delayed cell cycle S‐phase entry in response to IGF stimulation and that IGF‐1 induces Pin1 protein expression which correlates with the induction of cyclin D1 and RB phosphorylation in human breast cancer cells. The induction of Pin1 by IGF‐1 is mediated via the phosphatidylinositol 3‐kinase as well as the MAP kinase pathways. Treatment of PI3K inhibitor LY294002 and the MAP kinase inhibitor PD098059, but not p38 inhibitor SB203580, effectively blocks IGF‐1‐induced upregulation of Pin1, cyclin D1 and RB phosphorylation. Furthermore, we found that Cyclin D1 expression and RB phosphorylation are dramatically decreased in Pin1−/− MEF cells. Reintroducing a recombinant adenovirus encoding Pin1 into Pin1−/− MEF cells restores the expression of cyclin D1 and RB phosphorylation. Thus, these data suggest that the mitogenic function of IGF‐1 is at least partially linked to the induction of Pin1, which in turn stimulates cyclin D1 expression and RB phosphorylation, therefore contributing to G0/G1‐S transition. J. Cell. Biochem. 84: 211–216, 2002.


Cell Death and Disease | 2017

MiR-19a/b modulate the metastasis of gastric cancer cells by targeting the tumour suppressor MXD1

Qiong Wu; Z Yang; Y An; H Hu; J Yin; P Zhang; Yongzhan Nie; Kaichun Wu; Yongquan Shi; Daiming Fan

The microRNAs 19a and 19b, hereafter collectively referred to as miR-19a/b, were recognised to be the most important miRNAs in the oncomiRs—miR-17-92 cluster. However, the exact roles of miR-19a/b in cancers have not been elucidated. In the present study, miR-19a/b was found to be over-expressed in gastric cancer tissues and significantly associated with the patients’ metastasis of gastric cancer. Using gain or loss-of-function in in vitro and in vivo experiments, a pro-metastatic function of miR-19a/b was observed in gastric cancer. Furthermore, reporter gene assay and western blot showed that MXD1 is a direct target of miR-19a/b. Functional assays showed that not only MXD1 had an opposite effect to miR-19a/b in the regulation of gastric cancer cells, but also overexpression of MXD1 reduced both miR-19a/b and c-Myc levels, indicating a potential positive feedback loop among miR-19a/b, MXD1 and c-Myc. In conclusion, miR-17-92 cluster members miR-19a/b facilitated gastric cancer cell migration, invasion and metastasis through targeting the antagonist of c-Myc -- MXD1, implicating a novel mechanism for the malignant phenotypes of gastric cancer.


Journal of Cellular Biochemistry | 2012

Identification of cancer stem cells in vincristine preconditioned SGC7901 gastric cancer cell line

Zengfu Xue; Hui Yan; Juntang Li; Shuli Liang; Xiqiang Cai; Xiong Chen; Qiong Wu; Liucun Gao; Kaichun Wu; Yongzhan Nie; Daiming Fan

Cancer stem cells (CSCs), or tumor initiating cells, are a subpopulation of cancer cells with self‐renewal and differentiation properties. However, there has been no direct observation of the properties of gastric CSCs in vitro. Here we describe a vincristine (VCR)‐preconditioning approach to obtain cancer stem‐like cells (CSLCs) from the gastric cancer cell line SGC7901. The CSLCs displayed mesenchymal characteristics, including the up‐regulated mesenchymal markers Snail, Twist, and vimentin, and the down‐regulated epithelial marker E‐cadherin. Using a Matrigel‐based differentiation assay, CSLCs formed 2D tube‐like and 3D complex lumen‐like structures, which resembled differentiated gastric crypts. The characteristic of cellular differentiation was also found by transmission electron microscopy and up‐regulation of gastrointestinal genes CDX2 and SOX2. We further showed that CSLCs could self‐renew through significant asymmetric division compared with parent cells by tracing PKH‐26, BrdU, and EDU label‐retaining cells. In addition, these CSLCs also increased expression of CD44, CD90, and CXCR4 at the mRNA level, which was identified as novel targets. Furthermore, drug sensitivity assays and xenograft experiments demonstrated that the cells developed multi‐drug resistance (MDR) and significant tumorigenicity in vivo. In summary, gastric CSCs were identified from VCR‐preconditioned SGC7901 cell line, characterized by high tumorigenicity and the capacity for self‐renewal and differentiation. J. Cell. Biochem. 113: 302–312, 2012.


Journal of Cellular Biochemistry | 2004

Distinctive regulation and function of PI 3K/Akt and MAPKs in doxorubicin‐induced apoptosis of human lung adenocarcinoma cells

Yanqiu Zhao; Han You; Yu Yang; Lin Wei; Xin Zhang; Libo Yao; Daiming Fan; Qiang Yu

Regulation and function of PI 3K/Akt and mitogen‐activated protein kinases (MAPKs) in doxorubicin‐induced cell death were investigated in human lung adenocarcinoma cells. Doxorubicin induced dose‐dependent apoptosis of human lung adenocarcinoma NCI‐H522 cells. Prior to cell death, both Akt and the MAPK family members (MAPKs: ERK1/2, JNK, and p38) were activated in response to the drug treatment. The kinetics of the inductions for Akt and MAPKs are, however, distinct. The activation of Akt was rapid and transient, activated within 30 min of drug addition, then declined after 3 h, whereas the activations of three MAPKs occurred later, 4 h after addition of the drug and sustained until cell death occurred. Inhibition of PI 3K/Akt activation had no effect on MAPKs activation, suggesting that the two pathways are independently activated in response to the drug treatment. Inhibition of PI 3K/Akt and p38 accelerated and enhanced doxorubicin‐induced cell death. On the contrary, inhibition of ERK1/2 or JNK had no apparent effect on the cell death. Taken together, these results suggest that PI 3K/Akt and MAPKs signaling pathways are all activated, but with distinct mechanisms, in response to doxorubicin treatment. Activation of PI 3K/Akt and p38 modulates apoptotic signal pathways and inhibits doxorubicin‐induced cell death. These responses of tumor cells to cancer drug treatment may contribute to their drug resistance. Understanding of the mechanism and function of the responses will be beneficial for the development of novel therapeutic approaches for improvement of drug efficacy and circumvention of drug resistance.


Autophagy | 2010

Macroautophagy and ERK phosphorylation counteract the antiproliferative effect of proteasome inhibitor in gastric cancer cells.

William Ka Kei Wu; Chi Hin Cho; Lee Cw; Wu Yc; Yu L; Li Zj; Wong Cc; Li Ht; Lin Zhang; Ren Sx; Che Ct; Kaichun Wu; Daiming Fan; Jun Yu; Joseph J.Y. Sung

The ubiquitin-proteasome system and macroautophagy are two complementary pathways for protein degradation. Emerging evidence suggests that proteasome inhibition might be a promising approach for the treatment of cancer. In this study, we show that proteasome inhibitor MG-132 suppressed gastric cancer cell proliferation and induced macroautophagy. The induction of macroautophagy was evidenced by the formation of LC3+ autophagosomes and the accumulation of acidic vesicular organelles and autolysosomes and was accompanied by the suppression of mammalian target of rapamycin complex 1 activity. Abolition of macroautophagy by knockdown of Class III phosphatidylinositol-3 kinase Vps34 or ATG5/7 sensitized gastric cancer cells to the anti-proliferative effect of MG-132 by promoting G2/M cell cycle arrest. In addition, MG-132 increased ERK phosphorylation whose inhibition by MEK inhibitor significantly enhanced the anti-proliferative effect of proteasome inhibition. To conclude, this study demonstrates that macroautophagy and ERK phosphorylation serve as protective mechanisms to counteract the anti-proliferative effect of proteasome inhibition. This discovery may have implications for the application of proteasome-directed therapy for the treatment of cancer.


Cell Death and Disease | 2013

A set of microRNAs mediate direct conversion of human umbilical cord lining-derived mesenchymal stem cells into hepatocytes

L Cui; Yongquan Shi; X Zhou; Xin Wang; Jin Wang; Y Lan; M Wang; L Zheng; Hongliang Li; Qiong Wu; Jian Zhang; Daiming Fan; Y Han

In a previous study, we elucidated the specific microRNA (miRNA) profile of hepatic differentiation. In this study, we aimed to clarify the instructive role of six overexpressed miRNAs (miR-1246, miR-1290, miR-148a, miR-30a, miR-424 and miR-542-5p) during hepatic differentiation of human umbilical cord lining-derived mesenchymal stem cells (hMSCs) and to test whether overexpression of any of these miRNAs is sufficient to induce differentiation of the hMSCs into hepatocyte-like cells. Before hepatic differentiation, hMSCs were infected with a lentivirus containing a miRNA inhibitor sequence. We found that downregulation of any one of the six hepatic differentiation-specific miRNAs can inhibit HGF-induced hepatic differentiation including albumin expression and LDL uptake. Although overexpression of any one of the six miRNAs alone or liver-enriched miR-122 cannot initiate hepatic differentiation, ectopic overexpression of seven miRNAs (miR-1246, miR-1290, miR-148a, miR-30a, miR-424, miR-542-5p and miR-122) together can stimulate hMSC conversion into functionally mature induced hepatocytes (iHep). Additionally, after transplantation of the iHep cells into mice with CCL4-induced liver injury, we found that iHep not only can improve liver function but it also can restore injured livers. The findings from this study indicate that miRNAs have the capability of directly converting hMSCs to a hepatocyte phenotype in vitro.


Cancer Cell | 2017

CHD4 Has Oncogenic Functions in Initiating and Maintaining Epigenetic Suppression of Multiple Tumor Suppressor Genes

Limin Xia; Wenjie Huang; Marina Bellani; Michael M. Seidman; Kaichun Wu; Daiming Fan; Yongzhan Nie; Yi Cai; Yang W. Zhang; Li Rong Yu; Huili Li; Cynthia A. Zahnow; Wenbing Xie; Ray Whay Chiu Yen; Feyruz V. Rassool; Stephen B. Baylin

An oncogenic role for CHD4, a NuRD component, is defined for initiating and supporting tumor suppressor gene (TSG) silencing in human colorectal cancer. CHD4 recruits repressive chromatin proteins to sites of DNA damage repair, including DNA methyltransferases where it imposes de novo DNA methylation. At TSGs, CHD4 retention helps maintain DNA hypermethylation-associated transcriptional silencing. CHD4 is recruited by the excision repair protein OGG1 for oxidative damage to interact with the damage-induced base 8-hydroxydeoxyguanosine (8-OHdG), while ZMYND8 recruits it to double-strand breaks. CHD4 knockdown activates silenced TSGs, revealing their role for blunting colorectal cancer cell proliferation, invasion, and metastases. High CHD4 and 8-OHdG levels plus low expression of TSGs strongly correlates with early disease recurrence and decreased overall survival.


Autophagy | 2016

Chaperone-mediated autophagy regulates proliferation by targeting RND3 in gastric cancer

Jinfeng Zhou; Jianjun Yang; Xing Fan; Sijun Hu; Fenli Zhou; Jiaqiang Dong; Song Zhang; Yulong Shang; Xiaoming Jiang; Hao Guo; Ning Chen; Xiao Xiao; Jianqiu Sheng; Kaichun Wu; Yongzhan Nie; Daiming Fan

ABSTRACT LAMP2A is the key protein of chaperone-mediated autophagy (CMA), downregulation of LAMP2A leads to CMA blockade. CMA activation has been implicated in cancer growth, but the exact mechanisms are unclear. Elevated expression of LAMP2A was found in 8 kinds of tumors (n=747), suggesting that LAMP2A may have an important role in cancer progression. Unsurprisingly, LAMP2A knockdown in gastric cancer (GC) cells hindered proliferation, accompanied with altered expression of cell cycle-related proteins and accumulation of RND3/RhoE. Interactomic and KEGG analysis revealed that RND3 was a putative CMA substrate. Further study demonstrated that RND3 silencing could partly rescue the proliferation arrest induced by LAMP2A knockdown; RND3 was increased upon lysosome inhibition via both chemicals and LAMP2A-shRNA; Furthermore, RND3 could interact with CMA components HSPA8 and LAMP2A, and be engulfed by isolated lysosomes. Thus, constant degradation of RND3 by CMA is required to sustain rapid proliferation of GC cells. At last, the clinical significance of LAMP2A was explored in 593 gastric noncancerous lesions and 173 GC tissues, the results revealed that LAMP2A is a promising biomarker for GC early warning and prognosis of female GC patients.


Cell Death and Disease | 2017

miR-143 and miR-145 inhibit gastric cancer cell migration and metastasis by suppressing MYO6

Chao Lei; Feng Du; Lina Sun; Ting Li; Tingyu Li; Yali Min; Aiying Nie; Xin Wang; Lei Geng; Yuanyuan Lu; Xiaodi Zhao; Yongquan Shi; Daiming Fan

Metastasis is a major clinical obstacle responsible for the high mortality and poor prognosis of gastric cancer (GC). MicroRNAs (miRNAs) are critical mediators of metastasis that act by modulating their target genes. In this study, we found that miR-143 and miR-145 act via a common target gene, MYO6, to regulate the epithelial–mesenchymal transition (EMT) and inhibit metastasis. We determined that miR-143 and miR-145 were downregulated in GC, and the ectopic expression of miR-143 and/or miR-145 inhibited GC cell migration and metastasis. Furthermore, MYO6 was identified as a direct common target of miR-143 and miR-145 and was elevated in GC. Silencing of MYO6 resulted in a metastasis-suppressive activity similar to that of miR-143 and miR-145, while restoring MYO6 attenuated the anti-metastatic or anti-EMT effects caused by miR-143 and miR-145. Clinically, an inverse correlation was observed between miR-143/145 levels and MYO6 levels in GC tissues, and either miR-143/145 downregulation or MYO6 upregulation was associated with more malignant phenotypes in patients with GC. In conclusion, miR-143 and miR-145 suppress GC cell migration and metastasis by inhibiting MYO6 expression and the EMT, which provides a novel mechanism and promising therapeutic target for the treatment of GC metastasis.

Collaboration


Dive into the Daiming Fan's collaboration.

Top Co-Authors

Avatar

Kaichun Wu

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yongzhan Nie

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Guohong Han

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yongquan Shi

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Feng Du

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Jing Niu

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Limin Xia

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Wengang Guo

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiaodi Zhao

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Chuangye He

Fourth Military Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge