Network


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

Hotspot


Dive into the research topics where Biao Fan is active.

Publication


Featured researches published by Biao Fan.


Journal of Clinical Investigation | 2012

Cholangiocarcinomas can originate from hepatocytes in mice.

Biao Fan; Yann Malato; Diego F. Calvisi; Syed Naqvi; Nataliya Razumilava; Silvia Ribback; Gregory J. Gores; Frank Dombrowski; Matthias Evert; Xin Chen; Holger Willenbring

Intrahepatic cholangiocarcinomas (ICCs) are primary liver tumors with a poor prognosis. The development of effective therapies has been hampered by a limited understanding of the biology of ICCs. Although ICCs exhibit heterogeneity in location, histology, and marker expression, they are currently thought to derive invariably from the cells lining the bile ducts, biliary epithelial cells (BECs), or liver progenitor cells (LPCs). Despite lack of experimental evidence establishing BECs or LPCs as the origin of ICCs, other liver cell types have not been considered. Here we show that ICCs can originate from fully differentiated hepatocytes. Using a mouse model of hepatocyte fate tracing, we found that activated NOTCH and AKT signaling cooperate to convert normal hepatocytes into biliary cells that act as precursors of rapidly progressing, lethal ICCs. Our findings suggest a previously overlooked mechanism of human ICC formation that may be targetable for anti-ICC therapy.


Gastroenterology | 2013

Yes-Associated Protein Up-regulates Jagged-1 and Activates the NOTCH Pathway in Human Hepatocellular Carcinoma

Darjus F. Tschaharganeh; Xin Chen; Philipp Latzko; Mona Malz; Matthias M. Gaida; Klaus Felix; Sara Ladu; Stephan Singer; Federico Pinna; Norbert Gretz; Carsten Sticht; Maria Lauda Tomasi; Salvatore Delogu; Matthias Evert; Biao Fan; Silvia Ribback; Lijie Jiang; Stefania Brozzetti; Frank Bergmann; Frank Dombrowski; Peter Schirmacher; Diego F. Calvisi; Kai Breuhahn

BACKGROUND & AIMS Cancer cells often lose contact inhibition to undergo anchorage-independent proliferation and become resistant to apoptosis by inactivating the Hippo signaling pathway, resulting in activation of the transcriptional co-activator yes-associated protein (YAP). However, the oncogenic mechanisms of YAP activity are unclear. METHODS By using cross-species analysis of expression data, the Notch ligand Jagged-1 (Jag-1) was identified as a downstream target of YAP in hepatocytes and hepatocellular carcinoma (HCC) cells. We analyzed the functions of YAP in HCC cells via overexpression and RNA silencing experiments. We used transgenic mice that overexpressed a constitutively activated form of YAP (YAP(S127A)), and measured protein levels in HCC, colorectal and pancreatic tumor samples from patients. RESULTS Human HCC cell lines and mouse hepatocytes that overexpress YAP(S127A) up-regulated Jag-1, leading to activation of the Notch pathway and increased proliferation. Induction of Jag-1, activation of Notch, and cell proliferation required binding of YAP to its transcriptional partner TEA domain family member 4 (TEAD4); TEAD4 binding required the Mst1/2 but not β-catenin signaling. Levels of YAP correlated with Jag-1 expression and Notch signaling in human tumor samples and correlated with shorter survival times of patients with HCC or colorectal cancer. CONCLUSIONS The transcriptional regulator YAP up-regulates Jag-1 to activate Notch signaling in HCC cells and mouse hepatocytes. YAP-dependent activity of Jag-1 and Notch correlate in human HCC and colorectal tumor samples with patient survival times, suggesting the use of YAP and Notch inhibitors as therapeutics for gastrointestinal cancer. Transcript profiling: microarray information was deposited at the Gene Expression Omnibus database (http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?token=jxepvsumwosqkve&acc=GSE35004).


Gastroenterology | 2014

Activation of β-Catenin and Yap1 in Human Hepatoblastoma and Induction of Hepatocarcinogenesis in Mice

Junyan Tao; Diego F. Calvisi; Sarangarajan Ranganathan; Antonio Cigliano; Lili Zhou; Sucha Singh; Lijie Jiang; Biao Fan; Luigi Terracciano; Sorin Armeanu–Ebinger; Silvia Ribback; Frank Dombrowski; Matthias Evert; Xin Chen; Satdarshan P.S. Monga

BACKGROUND & AIMS Aberrant activation of β-catenin and Yes-associated protein 1 (Yap1) signaling pathways have been associated with the development of multiple tumor types. Yap functions as a transcriptional coactivator by interacting with TEA domain DNA binding proteins. We investigated the interactions among these pathways during hepatic tumorigenesis. METHODS We used immunohistochemical analysis to determine expression of β-catenin and Yap1 in liver cancer specimens collected from patients in Europe and the United States, consisting of 104 hepatocellular carcinoma, 62 intrahepatic cholangiocarcinoma, and 94 hepatoblastoma samples. We assessed β-catenin and Yap1 signaling and interactions in hepatoblastoma cell lines ((HuH6, HepG2, HepT1, HC-AFW1, HepG2, and HC-AFW1); proteins were knocked down with small interfering RNAs, and effects on proliferation and cell death were measured. Sleeping beauty-mediated hydrodynamic transfection was used to overexpress constitutively active forms of β-catenin (ΔN90/β-catenin) and Yap1 (YapS127A) in livers of mice; tissues were collected, and histological and immunohistochemical analyses were performed. RESULTS We observed nuclear localization of β-catenin and Yap1 in 79% of hepatoblastoma samples but not in most hepatocellular carcinoma or intrahepatic cholangiocarcinoma samples. Yap1 and β-catenin coprecipitated in hepatoblastoma but not hepatocellular carcinoma cells. Small interfering RNA-mediated knockdown of Yap1 or β-catenin in hepatoblastoma cells reduced proliferation in an additive manner. Knockdown of Yap1 reduced its ability to coactivate transcription with β-catenin; β-catenin inhibitors inactivated Yap1. Overexpression of constitutively active forms of Yap1 and β-catenin in mouse liver led to rapid tumorigenesis, with 100% mortality by 11 weeks. Tumor cells expressed both proteins, and human hepatoblastoma cells expressed common targets of their 2 signaling pathways. Yap1 binding of TEA domain factors was required for tumorigenesis in mice. CONCLUSIONS β-catenin and the transcriptional regulator Yap1 interact physically and are activated in most human hepatoblastoma tissues; overexpression of activated forms of these proteins in livers of mice leads to rapid tumor development. Further analysis of these mice will allow further studies of these pathways in hepatoblastoma pathogenesis and could lead to the identification of new therapeutic targets.


PLOS ONE | 2012

Integration of DNA Copy Number Alterations and Transcriptional Expression Analysis in Human Gastric Cancer

Biao Fan; Somkid Dachrut; Ho Coral; Siu Tsan Yuen; Kent Man Chu; Simon Law; Lianhai Zhang; Jiafu Ji; Suet Yi Leung; Xin Chen

Background Genomic instability with frequent DNA copy number alterations is one of the key hallmarks of carcinogenesis. The chromosomal regions with frequent DNA copy number gain and loss in human gastric cancer are still poorly defined. It remains unknown how the DNA copy number variations contributes to the changes of gene expression profiles, especially on the global level. Principal Findings We analyzed DNA copy number alterations in 64 human gastric cancer samples and 8 gastric cancer cell lines using bacterial artificial chromosome (BAC) arrays based comparative genomic hybridization (aCGH). Statistical analysis was applied to correlate previously published gene expression data obtained from cDNA microarrays with corresponding DNA copy number variation data to identify candidate oncogenes and tumor suppressor genes. We found that gastric cancer samples showed recurrent DNA copy number variations, including gains at 5p, 8q, 20p, 20q, and losses at 4q, 9p, 18q, 21q. The most frequent regions of amplification were 20q12 (7/72), 20q12–20q13.1 (12/72), 20q13.1–20q13.2 (11/72) and 20q13.2–20q13.3 (6/72). The most frequent deleted region was 9p21 (8/72). Correlating gene expression array data with aCGH identified 321 candidate oncogenes, which were overexpressed and showed frequent DNA copy number gains; and 12 candidate tumor suppressor genes which were down-regulated and showed frequent DNA copy number losses in human gastric cancers. Three networks of significantly expressed genes in gastric cancer samples were identified by ingenuity pathway analysis. Conclusions This study provides insight into DNA copy number variations and their contribution to altered gene expression profiles during human gastric cancer development. It provides novel candidate driver oncogenes or tumor suppressor genes for human gastric cancer, useful pathway maps for the future understanding of the molecular pathogenesis of this malignancy, and the construction of new therapeutic targets.


BMC Cancer | 2012

Presence of S100A9-positive inflammatory cells in cancer tissues correlates with an early stage cancer and a better prognosis in patients with gastric cancer

Biao Fan; Lianhai Zhang; Yongning Jia; Xi-Yao Zhong; Liu Yr; Xiaojing Cheng; Xiaohong Wang; Xiaofang Xing; Ying Hu; Yingai Li; Hong Du; Wei Zhao; Zhao-Jian Niu; Ai-Ping Lu; Jiyou Li; Jiafu Ji

BackgroundS100A9 was originally discovered as a factor secreted by inflammatory cells. Recently, S100A9 was found to be associated with several human malignancies. The purpose of this study is to investigate S100A9 expression in gastric cancer and explore its role in cancer progression.MethodsS100A9 expression in gastric tissue samples from 177 gastric cancer patients was assessed by immunohistochemistry. The expression of its dimerization partner S100A8 and the S100A8/A9 heterodimer were also assessed by the same method. The effect of exogenous S100A9 on motility of gastric cancer cells AGS and BGC-823 was then investigated.ResultsS100A9 was specifically expressed by inflammatory cells such as macrophages and neutrophils in human gastric cancer and gastritis tissues. Statistical analysis showed that a high S100A9 cell count (> = 200) per 200x magnification microscopic field in cancer tissues was predictive of early stage gastric cancer. High S100A9-positive cell count was negatively correlated with lymph node metastasis (P = 0.009) and tumor invasion (P = 0.011). S100A9 was identified as an independent prognostic predictor of overall survival of patients with gastric cancer (P = 0.04). Patients with high S100A9 cell count were with favorable prognosis (P = 0.021). Further investigation found that S100A8 distribution in human gastric cancer tissues was similar to S100A9. However, the number of S100A8-positive cells did not positively correlate with patient survival. The inflammatory cells infiltrating cancer were S100A8/A9 negative, while those in gastritis were positive. Furthermore, exogenous S100A9 protein inhibited migration and invasion of gastric cancer cells.ConclusionsOur results suggested S100A9-positive inflammatory cells in gastric cancer tissues are associated with early stage of gastric cancer and good prognosis.


Hepatology | 2015

4EBP1/eIF4E and p70S6K/RPS6 axes play critical and distinct roles in hepatocarcinogenesis driven by AKT and N‐Ras proto‐oncogenes in mice

Chunmei Wang; Antonio Cigliano; Lijie Jiang; Xiaolei Li; Biao Fan; Maria G. Pilo; Yan Liu; Bing Gui; Marcella Sini; Jeffrey W. Smith; Frank Dombrowski; Diego F. Calvisi; Matthias Evert; Xin Chen

Concomitant expression of activated forms of v‐akt murine thymoma viral oncogene homolog (AKT) and Ras in mouse liver (AKT/Ras) leads to rapid tumor development through strong activation of the mammalian target of rapamycin complex 1 (mTORC1) pathway. mTORC1 functions by regulating p70S6K/ribosomal protein S6 (RPS6) and eukaryotic translation initiation factor 4E‐binding protein 1/ eukaryotic translation initiation factor 4E (4EBP1/eIF4E) cascades. How these cascades contribute to hepatocarcinogenesis remains unknown. Here, we show that inhibition of the RPS6 pathway by rapamycin effectively suppressed, whereas blockade of the 4EBP1/eIF4E cascade by 4EBP1A4, an unphosphorylatable form of 4EBP1, significantly delayed, AKT/Ras‐induced hepatocarcinogenesis. Combined treatment with rapamycin and 4EBP1A4 completely inhibited AKT/Ras hepatocarcinogenesis. This strong antineoplastic effect was successfully recapitulated by ablating regulatory associated protein of mTORC1, the major subunit of mTORC1, in AKT/Ras‐overexpressing livers. Furthermore, we demonstrate that overexpression of eIF4E, the proto‐oncogene whose activity is specifically inhibited by 4EBP1, resulted in hepatocellular carcinoma (HCC) development in cooperation with activated Ras. Mechanistically, we identified the ectonucleoside triphosphate diphosphohydrolase 5/ adenylate kinase 1/cytidine monophosphate kinase 1 axis and the mitochondrial biogenesis pathway as targets of the 4EBP1/eIF4E cascade in AKT/Ras and Ras/eIF4E livers as well as in human HCC cell lines and tissues. Conclusions: Complete inhibition of mTORC1 is required to suppress liver cancer development induced by AKT and Ras proto‐oncogenes in mice. The mTORC1 effectors, RPS6 and eIF4E, play distinct roles and are both necessary for AKT/Ras hepatocarcinogenesis. These new findings might open the way for innovative therapies against human HCC. (Hepatology 2015;61:200–213)


Tumor Biology | 2016

Trichostatin A potentiates TRAIL-induced antitumor effects via inhibition of ERK/FOXM1 pathway in gastric cancer

Lin Li; Biao Fan; Lianhai Zhang; Xiaofang Xing; Xiaojing Cheng; Xiaohong Wang; Ting Guo; Hong Du; Xian-Zi Wen; Jiafu Ji

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is an ideal apoptosis inducer and believed to have promise in cancer therapy, yet part of cancer cells exhibit resistance to TRAIL-mediated apoptosis. This necessitates the exploration of agents that resensitizes cancer cells to TRAIL. In our study, we found that Trichostatin A (TSA), an histone deacetylase (HDAC) inhibitor, augmented TRAIL-induced apoptosis in gastric cancer cells in a caspase-dependent manner. Besides, upregulation of DR5 and downregulation of anti-apoptotic proteins including XIAP, Mcl-1, Bcl-2 and Survivin also contributed to this synergism. Noticeably, TSA treatment inhibited Forkhead boxM1 (FOXM1), which expression level showed negative correlation with TRAIL sensitivity. Similarly, silencing of FOXM1 by small interfering RNA (siRNA) resensitized cancer cells to TRAIL and strengthened the TRAIL-augmenting effect of TSA. In addition, we demonstrated the depletion of FOXM1 was a consequence of the inactivation of ERK mediated by TSA. Collectively, it was first shown that TSA potentiated TRAIL sensitivity via ERK/FOXM1 pathway in gastric cancer cells. FOXM1 might serve as a biomarker for predicting sensitivity to TRAIL.


Oncology Reports | 2016

Paclitaxel enhances tumoricidal potential of TRAIL via inhibition of MAPK in resistant gastric cancer cells

Lin Li; Xian-Zi Wen; Zhaode Bu; Xiaojing Cheng; Xiaofang Xing; Xiaohong Wang; Lianhai Zhang; Ting Guo; Hong Du; Ying Hu; Biao Fan; Jiafu Ji

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) holds promise for cancer therapy due to its unique capacity to selectively trigger apoptosis in cancer cells. However, TRAIL therapy is greatly hampered by its resistance. A preclinical successful strategy is to identify combination treatments that sensitize resistant cancers to TRAIL. In the present study, we fully assessed TRAIL sensitivity in 9 gastric cancer cell lines. We found combined administration of paclitaxel (PTX) markedly enhanced TRAIL-induced apoptosis in resistant cancer cells both in vitro and in vivo. The sensitization to TRAIL was accompanied by activation of mitochondrial apoptotic pathway, upregulation of TRAIL receptors and downregulation of anti-apoptotic proteins including C-IAP1, C-IAP2, Livin and Mcl-1. Noticeably, we found PTX could suppress the activation of mitogen-activated protein kinases (MAPKs). Inhibition of MAPKs using specific inhibitors (ERK inhibitor U0126, JNK inhibitor SP600125 and P38 inhibitor SB202190) facilitated TRAIL-mediated apoptosis and cytotoxicity. Additionally, SP600125 upregulated TRAL receptors as well as downregulated C-IAP2 and Mcl-1 suggesting the anti-apoptotic role of JNK. Thus, PTX-induced suppression of MAPKs may contribute to restoring TRAIL senstitivity. Collectively, our comprehensive analyses gave new insight into the role of PTX on enhancing TRAIL sensitivity, and provided theoretical references on the development of combination treatment in TRAIL-resistant gastric cancer.


Hepatology | 2013

On the role of notch1 and adult hepatocytes in murine intrahepatic cholangiocarcinoma development

Matthias Evert; Frank Dombrowski; Biao Fan; Silvia Ribback; Xin Chen; Diego F. Calvisi

We write in reply to the letter of Dr. Y.F. Liaw. In our original article we included all the relevant studies mentioned in the metaanalyses of Sung et al. and Yang et al. The study of Benvegnu et al. was excluded because the main conclusion of the study was on hepatitis C-related cirrhosis. Concerning the two studies from Taiwan, Dr. Liaw misses our point about the conflicting conclusions. Briefly, the first study showed no reduction in the development of new cirrhosis after interferon treatment. The second study showed that interferon was effective in prevention of hepatocellular carcinoma (HCC) only in patients with preexisting cirrhosis at study recruitment. In the second study, presumably some of the treated patients with no cirrhosis on recruitment would have developed new cirrhosis if the first study was correct. This did indeed occur, mainly but not solely, in the “nonresponders” But interferon did not decrease HCC in this group of patients despite the shorter duration of the newly developed cirrhosis. This is the basic paradox of the studies. Dr. Liaw mentions the differences in the results obtained from two “studies” from Hong Kong with our published article on interferon treatment. However, he is citing two preliminary abstracts with incomplete patient follow-up (n 5 48 and 138). The database of our interferon studies was exhaustively reviewed. In the final article we included all interferon-treated patients (n 5 208). A control group (n 5 203), recruiting all patients who fulfilled the treatment criteria during the study period but were not treated, was added to demonstrate the effectiveness or otherwise of interferon treatment. Furthermore, Dr. Liaw erroneously quotes that the interferon-treated group was found to have a high risk for HCC. Our conclusion was that interferon treatment did not lower the risk of development of HCC, there being no statistical difference between the treated and untreated patients. Dr. Liaw also mentioned that our study mostly recruited immunotolerant patients, and therefore it was excluded from one metaanalysis. However, of the 278 patients in the four original trials, only 89 were in the children cohort. 14.3% of the adult population was cirrhotic. The hepatitis B e antigen (HBeAg) seroconversion rates of the treated population and the control group were comparable to each other as well as to other studies in the review. Dr. Liaw mentions that the proportion of patients with cirrhosis in his study of lamivudine in cirrhosis patients should be 60% and 65% for the lamivudine-treated group and control group, respectively. In our review article the respective proportions quoted were 31% and 39%. These were the proportions of patients with Ishak fibrosis score of 6; that is, patients with established, and therefore definite, cirrhosis. The proportions mentioned by Dr. Liaw included patients with Ishak fibrosis score 5, which were patients with incomplete cirrhosis. There is in fact debate as to whether an Ishak fibrosis score of 5 should or should not be included as “cirrhosis” under the more recent METAVIR system of fibrosis scoring. We therefore quoted only those with Ishak fibrosis score of 6. Finally, for the study showing entecavir is superior to lamivudine, it was published after we submitted and proof-read our review.


Journal of Hepatology | 2017

Pan-mTOR inhibitor MLN0128 is effective against intrahepatic cholangiocarcinoma in mice.

Shanshan Zhang; Xinhua Song; Dan Cao; Zhong Xu; Biao Fan; Li Che; Junjie Hu; Bin Chen; Mingjie Dong; Maria G. Pilo; Antonio Cigliano; Katja Evert; Silvia Ribback; Frank Dombrowski; Rosa Maria Pascale; Antonio Cossu; Gianpaolo Vidili; Alberto Porcu; Maria Maddalena Simile; Giovanni Mario Pes; Gianluigi Giannelli; John D. Gordan; Lixin Wei; Matthias Evert; Wenming Cong; Diego F. Calvisi; Xin Chen

BACKGROUND & AIMS Intrahepatic cholangiocarcinoma (ICC) is a lethal malignancy without effective treatment options. MLN0128, a second generation pan-mTOR inhibitor, shows efficacy for multiple tumor types. We evaluated the therapeutic potential of MLN0128 vs. gemcitabine/oxaliplatin in a novel ICC mouse model. METHODS We established a novel ICC mouse model via hydrodynamic transfection of activated forms of AKT (myr-AKT) and Yap (YapS127A) protooncogenes (that will be referred to as AKT/YapS127A). Genetic approaches were applied to study the requirement of mTORC1 and mTORC2 in mediating AKT/YapS127A driven tumorigenesis. Gemcitabine/oxaliplatin and MLN0128 were administered in AKT/YapS127A tumor-bearing mice to study their anti-tumor efficacy in vivo. Multiple human ICC cell lines were used for in vitro experiments. Hematoxylin and eosin staining, immunohistochemistry and immunoblotting were applied for the characterization and mechanistic study. RESULTS Co-expression of myr-AKT and YapS127A promoted ICC development in mice. Both mTORC1 and mTORC2 complexes were required for AKT/YapS127A ICC development. Gemcitabine/oxaliplatin had limited efficacy in treating late stage AKT/YapS127A ICC. In contrast, partial tumor regression was achieved when MLN0128 was applied in the late stage of AKT/YapS127A cholangiocarcinogenesis. Furthermore, when MLN0128 was administered in the early stage of AKT/YapS127A carcinogenesis, it led to disease stabilization. Mechanistically, MLN0128 efficiently inhibited AKT/mTOR signaling both in vivo and in vitro, inducing strong ICC cell apoptosis and only marginally affecting proliferation. CONCLUSIONS This study suggests that mTOR kinase inhibitors may be beneficial for the treatment of ICC, even in tumors that are resistant to standard of care chemotherapeutics, such as gemcitabine/oxaliplatin-based regimens, especially in the subset of tumors exhibiting activated AKT/mTOR cascade. Lay summary: We established a novel mouse model of intrahepatic cholangiocarcinoma (ICC). Using this new preclinical model, we evaluated the therapeutic potential of mTOR inhibitor MLN0128 vs. gemcitabine/oxaliplatin (the standard chemotherapy for ICC treatment). Our study shows the anti-neoplastic potential of MLN0128, suggesting that it may be superior to gemcitabine/oxaliplatin-based chemotherapy for the treatment of ICC, especially in the tumors exhibiting activated AKT/mTOR cascade.

Collaboration


Dive into the Biao Fan's collaboration.

Top Co-Authors

Avatar

Xin Chen

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Matthias Evert

University of Regensburg

View shared research outputs
Top Co-Authors

Avatar

Silvia Ribback

University of Greifswald

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Li Che

University of California

View shared research outputs
Top Co-Authors

Avatar

Lijie Jiang

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge