Network


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

Hotspot


Dive into the research topics where Pengcheng Bu is active.

Publication


Featured researches published by Pengcheng Bu.


Nature Cell Biology | 2011

miR-34 miRNAs provide a barrier for somatic cell reprogramming

Yong Jin Choi; Chao-Po Lin; Jaclyn J. Ho; Xingyue He; Nobuhiro Okada; Pengcheng Bu; Yingchao Zhong; Sang Yong Kim; Margaux J. Bennett; Caifu Chen; Arzu Öztürk; Geoffrey G. Hicks; Greg J. Hannon; Lin He

Somatic reprogramming induced by defined transcription factors is a low-efficiency process that is enhanced by p53 deficiency. So far, p21 is the only p53 target shown to contribute to p53 repression of iPSC (induced pluripotent stem cell) generation, indicating that additional p53 targets may regulate this process. Here, we demonstrate that miR-34 microRNAs (miRNAs), particularly miR-34a, exhibit p53-dependent induction during reprogramming. Mir34a deficiency in mice significantly increased reprogramming efficiency and kinetics, with miR-34a and p21 cooperatively regulating somatic reprogramming downstream of p53. Unlike p53 deficiency, which enhances reprogramming at the expense of iPSC pluripotency, genetic ablation of Mir34a promoted iPSC generation without compromising self-renewal or differentiation. Suppression of reprogramming by miR-34a was due, at least in part, to repression of pluripotency genes, including Nanog, Sox2 and Mycn (also known as N-Myc). This post-transcriptional gene repression by miR-34a also regulated iPSC differentiation kinetics. miR-34b and c similarly repressed reprogramming; and all three miR-34 miRNAs acted cooperatively in this process. Taken together, our findings identified miR-34 miRNAs as p53 targets that play an essential role in restraining somatic reprogramming.


Molecular Cancer Therapeutics | 2006

Anti-CD146 monoclonal antibody AA98 inhibits angiogenesis via suppression of nuclear factor-κB activation

Pengcheng Bu; Lizeng Gao; Jie Zhuang; Jing Feng; Dongling Yang; Xiyun Yan

Our previous study showed that an anti-CD146 monoclonal antibody (mAb), AA98, which was raised against the vascular endothelial cells stimulated by a conditioned medium from hepatocarcinoma SMMC 7721 cells (SMMC 7721-CM), inhibited cell migration, angiogenesis, and tumor growth. However, the underlying mechanism was not elucidated. The objective of this study was to understand the mechanism by which mAb AA98 inhibits the endothelial cell migration and angiogenesis that is induced by SMMC 7721-CM. Using confocal imaging and biochemical studies, we found that SMMC 7721-CM induced nuclear factor κB (NF-κB) activation through the upstream p38 mitogen-activated protein kinase pathway, leading to the up-regulation of matrix metalloproteinase 9 and intercellular adhesion molecule 1 expression. Interestingly, all these activities stimulated by SMMC 7721-CM could be effectively inhibited by mAb AA98 in a dose- and time-dependent manner. Our data showed that the engagement of mAb AA98 with membrane protein CD146 inhibited p38 mitogen-activated protein kinase phosphorylation, suppressed NF-κB activation, and down-regulated matrix metalloproteinase 9 and intercellular adhesion molecule 1 expression, suggesting that the suppression of NF-κB is a critical point for the inhibitory function of mAb AA98 on endothelial cell migration, angiogenesis, and tumor metastasis. These results will provide clues for a better understanding of the mechanisms underlying tumor angiogenesis as well as antiangiogenesis therapy. [Mol Cancer Ther 2006;5(11):2872–8]


Cell Stem Cell | 2016

A miR-34a-Numb Feedforward Loop Triggered by Inflammation Regulates Asymmetric Stem Cell Division in Intestine and Colon Cancer.

Pengcheng Bu; Lihua Wang; Kai-Yuan Chen; Tara Srinivasan; Preetish Kadur Lakshminarasimha Murthy; Kuei-Ling Tung; Anastasia Kristine Varanko; Huanhuan Joyce Chen; Yiwei Ai; Sarah King; Steven M. Lipkin; Xiling Shen

Emerging evidence suggests that microRNAs can initiate asymmetric division, but whether microRNA and protein cell fate determinants coordinate with each other remains unclear. Here, we show that miR-34a directly suppresses Numb in early-stage colon cancer stem cells (CCSCs), forming an incoherent feedforward loop (IFFL) targeting Notch to separate stem and non-stem cell fates robustly. Perturbation of the IFFL leads to a new intermediate cell population with plastic and ambiguous identity. Lgr5+ mouse intestinal/colon stem cells (ISCs) predominantly undergo symmetric division but turn on asymmetric division to curb the number of ISCs when proinflammatory response causes excessive proliferation. Deletion of miR-34a inhibits asymmetric division and exacerbates Lgr5+ ISC proliferation under such stress. Collectively, our data indicate that microRNA and protein cell fate determinants coordinate to enhance robustness of cell fate decision, and they provide a safeguard mechanism against stem cell proliferation induced by inflammation or oncogenic mutation.


Journal of Clinical Investigation | 2012

Chemokine 25-induced signaling suppresses colon cancer invasion and metastasis.

Huanhuan Joyce Chen; Robert A. Edwards; Serena Tucci; Pengcheng Bu; Jeffrey W. Milsom; Sang Lee; Winfried Edelmann; Zeynep H. Gümüş; Xiling Shen; Steven M. Lipkin

Chemotactic cytokines (chemokines) can help regulate tumor cell invasion and metastasis. Here, we show that chemokine 25 (CCL25) and its cognate receptor chemokine receptor 9 (CCR9) inhibit colorectal cancer (CRC) invasion and metastasis. We found that CCR9 protein expression levels were highest in colon adenomas and progressively decreased in invasive and metastatic CRCs. CCR9 was expressed in both primary tumor cell cultures and colon-cancer-initiating cell (CCIC) lines derived from early-stage CRCs but not from metastatic CRC. CCL25 stimulated cell proliferation by activating AKT signaling. In vivo, systemically injected CCR9+ early-stage CCICs led to the formation of orthotopic gastrointestinal xenograft tumors. Blocking CCR9 signaling inhibited CRC tumor formation in the native gastrointestinal CCL25+ microenvironment, while increasing extraintestinal tumor incidence. NOTCH signaling, which promotes CRC metastasis, increased extraintestinal tumor frequency by stimulating CCR9 proteasomal degradation. Overall, these data indicate that CCL25 and CCR9 regulate CRC progression and invasion and further demonstrate an appropriate in vivo experimental system to study CRC progression in the native colon microenvironment.


Nature Communications | 2015

miR-1269 promotes metastasis and forms a positive feedback loop with TGF-β

Pengcheng Bu; Lihua Wang; Kai-Yuan Chen; Nikolai Rakhilin; Jian Sun; Closa A; Kuei-Ling Tung; Sarah King; Kristine Varanko A; Younong Xu; Huan Chen J; Zessin As; James B. Shealy; Cummings B; David S. Hsu; Steven M. Lipkin; Moreno; Zeynep H. Gümüş; Xiling Shen

As patient survival drops precipitously from early-stage cancers to late-stage and metastatic cancers, microRNAs that promote relapse and metastasis can serve as prognostic and predictive markers as well as therapeutic targets for chemoprevention. Here we show that miR-1269a promotes colorectal cancer (CRC) metastasis and forms a positive feedback loop with TGF-β signaling. miR-1269a is upregulated in late-stage CRCs, and long-term monitoring of 100 stage II CRC patients revealed that miR-1269a expression in their surgically removed primary tumors is strongly associated with risk of CRC relapse and metastasis. Consistent with clinical observations, miR-1269a significantly increases the ability of CRC cells to invade and metastasize in vivo. TGF-β activates miR-1269 via Sox4, while miR-1269a enhances TGF-β signaling by targeting Smad7 and HOXD10, hence forming a positive feedback loop. Our findings suggest that miR-1269a is a potential marker to inform adjuvant chemotherapy decisions for CRC patients and a potential therapeutic target to deter metastasis.


Nature Biotechnology | 2016

A recellularized human colon model identifies cancer driver genes

Huanhuan Joyce Chen; Zhubo Wei; Jian Sun; Asmita Bhattacharya; Rita E. Serda; Yuri Mackeyev; Steven A. Curley; Pengcheng Bu; Lihua Wang; Shuibing Chen; Leona Cohen-Gould; Emina Huang; Xiling Shen; Steven M. Lipkin; Neal G. Copeland; Nancy A. Jenkins; Michael L. Shuler

Refined cancer models are needed to bridge the gaps between cell line, animal and clinical research. Here we describe the engineering of an organotypic colon cancer model by recellularization of a native human matrix that contains cell-populated mucosa and an intact muscularis mucosa layer. This ex vivo system recapitulates the pathophysiological progression from APC-mutant neoplasia to submucosal invasive tumor. We used it to perform a Sleeping Beauty transposon mutagenesis screen to identify genes that cooperate with mutant APC in driving invasive neoplasia. We identified 38 candidate invasion-driver genes, 17 of which, including TCF7L2, TWIST2, MSH2, DCC, EPHB1 and EPHB2 have been previously implicated in colorectal cancer progression. Six invasion-driver genes that have not, to our knowledge, been previously described were validated in vitro using cell proliferation, migration and invasion assays and ex vivo using recellularized human colon. These results demonstrate the utility of our organoid model for studying cancer biology.


eLife | 2016

A long non-coding RNA targets microRNA miR-34a to regulate colon cancer stem cell asymmetric division

Lihua Wang; Pengcheng Bu; Yiwei Ai; Tara Srinivasan; Huanhuan Joyce Chen; Kun Xiang; Steven M. Lipkin; Xiling Shen

The roles of long non-coding RNAs (lncRNAs) in regulating cancer and stem cells are being increasingly appreciated. Its diverse mechanisms provide the regulatory network with a bigger repertoire to increase complexity. Here we report a novel LncRNA, Lnc34a, that is enriched in colon cancer stem cells (CCSCs) and initiates asymmetric division by directly targeting the microRNA miR-34a to cause its spatial imbalance. Lnc34a recruits Dnmt3a via PHB2 and HDAC1 to methylate and deacetylate the miR-34a promoter simultaneously, hence epigenetically silencing miR-34a expression independent of its upstream regulator, p53. Lnc34a levels affect CCSC self-renewal and colorectal cancer (CRC) growth in xenograft models. Lnc34a is upregulated in late-stage CRCs, contributing to epigenetic miR-34a silencing and CRC proliferation. The fact that lncRNA targets microRNA highlights the regulatory complexity of non-coding RNAs (ncRNAs), which occupy the bulk of the genome. DOI: http://dx.doi.org/10.7554/eLife.14620.001


Journal of Virological Methods | 2010

Influenza virus detection with pentabody-activated nanoparticles

Bin Mu; Xinglu Huang; Pengcheng Bu; Zhixue Cheng; Jing Feng; Dongling Yang; Changsheng Dong; Jianbing Zhang; Xiyun Yan

A nanoparticle-based immunoassay was developed for the rapid and sensitive detection of avian influenza virus (AIV). In this method, AIV-specific pentabody (pVHH3B) was conjugated to magnetic nanoparticles (MNPs) and used to capture AIV. Gold nanoparticles (GNPs), labelled with the anti-AIV mouse monoclonal antibody 3C8, were used as a detector. In the presence of target samples, the pentabody pVHH3B enriched AIV on the MNPs. Thereafter, mAb 3C8-labelled GNPs (GNPs-mAb3C8) bound to MNPs via AIV and were separated using a magnetic field. GNPs in the complex catalyzed the oxidation of hydroquinone to quinone, and the OD value of quinone was measured. The developed assay displayed substantial signal change after incubation in an AIV sample in a concentration-dependent manner. The detection limit was 10 ng/ml, which is 10 times more sensitive than conventional double-antibody sandwich enzyme-linked immunosorbent assay (DAS-ELISA). In conclusion, by combining MNPs and a novel pentabody pVHH3B, this study provided a sensitive influenza viral detection assay that has the potential to become a rapid, sensitive and inexpensive diagnostic tool for infectious diseases.


Cancer Research | 2016

NOTCH Signaling Regulates Asymmetric Cell Fate of Fast- and Slow-Cycling Colon Cancer Initiating Cells

Tara Srinivasan; Jewell Walters; Pengcheng Bu; Elaine Bich Than; Kuei Ling Tung; Kai Yuan Chen; Nicole C. Panarelli; Jeffrey W. Milsom; Leonard H. Augenlicht; Steven M. Lipkin; Xiling Shen

Colorectal cancer cells with stem-like properties, referred to as colon cancer-initiating cells (CCIC), have high tumorigenic potential. While CCIC can differentiate to promote cellular heterogeneity, it remains unclear whether CCIC within a tumor contain distinct subpopulations. Here, we describe the co-existence of fast- and slow-cycling CCIC, which can undergo asymmetric division to generate each other, highlighting CCIC plasticity and interconvertibility. Fast-cycling CCIC express markers, such as LGR5 and CD133, rely on MYC for their proliferation, whereas slow-cycling CCIC express markers, such as BMI1 and hTERT, are independent of MYC. NOTCH signaling promotes asymmetric cell fate, regulating the balance between these two populations. Overall, our results illuminate the basis for CCIC heterogeneity and plasticity by defining a direct interconversion mechanism between slow- and fast-cycling CCIC. Cancer Res; 76(11); 3411-21. ©2016 AACR.


Nature Biotechnology | 2015

Comprehensive models of human primary and metastatic colorectal tumors in immunodeficient and immunocompetent mice by chemokine targeting

Huanhuan Joyce Chen; Jian Sun; Zhiliang Huang; Harry Hou; Myra Arcilla; Nikolai Rakhilin; Daniel J. Joe; Jiahn Choi; Poornima Gadamsetty; Jeffrey W. Milsom; Govind Nandakumar; Randy S. Longman; Xi Kathy Zhou; Robert A. Edwards; Jonlin Chen; Kai Yuan Chen; Pengcheng Bu; Lihua Wang; Yitian Xu; Robert J. Munroe; Christian Abratte; Andrew D. Miller; Zeynep H. Gümüş; Michael L. Shuler; Nozomi Nishimura; Winfried Edelmann; Xiling Shen; Steven M. Lipkin

Current orthotopic xenograft models of human colorectal cancer (CRC) require surgery and do not robustly form metastases in the liver, the most common site clinically. CCR9 traffics lymphocytes to intestine and colorectum. We engineered use of the chemokine receptor CCR9 in CRC cell lines and patient-derived cells to create primary gastrointestinal (GI) tumors in immunodeficient mice by tail-vein injection rather than surgery. The tumors metastasize inducibly and robustly to the liver. Metastases have higher DKK4 and NOTCH signaling levels and are more chemoresistant than paired subcutaneous xenografts. Using this approach, we generated 17 chemokine-targeted mouse models (CTMMs) that recapitulate the majority of common human somatic CRC mutations. We also show that primary tumors can be modeled in immunocompetent mice by microinjecting CCR9-expressing cancer cell lines into early-stage mouse blastocysts, which induces central immune tolerance. We expect that CTMMs will facilitate investigation of the biology of CRC metastasis and drug screening.

Collaboration


Dive into the Pengcheng Bu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zeynep H. Gümüş

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Dongling Yang

Chinese Academy of Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge