Network


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

Hotspot


Dive into the research topics where Rui Su is active.

Publication


Featured researches published by Rui Su.


Cancer Cell | 2017

FTO Plays an Oncogenic Role in Acute Myeloid Leukemia as a N6-Methyladenosine RNA Demethylase.

Zejuan Li; Hengyou Weng; Rui Su; Xiaocheng Weng; Zhixiang Zuo; Chenying Li; Huilin Huang; Sigrid Nachtergaele; Lei Dong; Chao Hu; Xi Qin; Lichun Tang; Yungui Wang; Gia-Ming Hong; Hao Huang; Xiao Wang; Ping Chen; Sandeep Gurbuxani; Stephen Arnovitz; Yuanyuan Li; Shenglai Li; Jennifer Strong; Mary Beth Neilly; Richard A. Larson; Xi Jiang; Pumin Zhang; Jie Jin; Chuan He; Jianjun Chen

N6-Methyladenosine (m6A) represents the most prevalent internal modification in mammalian mRNAs. Despite its functional importance in various fundamental bioprocesses, the studies of m6A in cancer have been limited. Here we show that FTO, as an m6A demethylase, plays a critical oncogenic role in acute myeloid leukemia (AML). FTO is highly expressed in AMLs with t(11q23)/MLL rearrangements, t(15;17)/PML-RARA, FLT3-ITD, and/or NPM1 mutations. FTO enhances leukemic oncogene-mediated cell transformation and leukemogenesis, and inhibits all-trans-retinoic acid (ATRA)-induced AML cell differentiation, through regulating expression of targets such as ASB2 and RARA by reducing m6A levels in these mRNA transcripts. Collectively, our study demonstrates the functional importance of the m6A methylation and the corresponding proteins in cancer, and provides profound insights into leukemogenesis and drug response.


Cell Stem Cell | 2017

METTL14 Inhibits Hematopoietic Stem/Progenitor Differentiation and Promotes Leukemogenesis via mRNA m6A Modification

Hengyou Weng; Huilin Huang; Huizhe Wu; Xi Qin; Boxuan Simen Zhao; Lei Dong; Hailing Shi; Jennifer R. Skibbe; Chao Shen; Chao Hu; Yue Sheng; Yungui Wang; Mark Wunderlich; Bin Zhang; Louis C. Doré; Rui Su; Xiaolan Deng; Kyle Ferchen; Chenying Li; Miao Sun; Zhike Lu; Xi Jiang; Guido Marcucci; James C. Mulloy; Jianhua Yang; Zhijian Qian; Minjie Wei; Chuan He; Jianjun Chen

N6-methyladenosine (m6A), the most prevalent internal modification in eukaryotic messenger RNAs (mRNAs), plays critical roles in many bioprocesses. However, its functions in normal and malignant hematopoiesis remain elusive. Here, we report that METTL14, a key component of the m6A methyltransferase complex, is highly expressed in normal hematopoietic stem/progenitor cells (HSPCs) and acute myeloid leukemia (AML) cells carrying t(11q23), t(15;17), or t(8;21) and is downregulated during myeloid differentiation. Silencing of METTL14 promotes terminal myeloid differentiation of normal HSPCs and AML cells and inhibits AML cell survival/proliferation. METTL14 is required for development and maintenance of AML and self-renewal of leukemia stem/initiation cells (LSCs/LICs). Mechanistically, METTL14 exerts its oncogenic role by regulating its mRNA targets (e.g., MYB and MYC) through m6A modification, while the protein itself is negatively regulated by SPI1. Collectively, our results reveal the SPI1-METTL14-MYB/MYC signaling axis in myelopoiesis and leukemogenesis and highlight the critical roles of METTL14 and m6A modification in normal and malignant hematopoiesis.


Blood | 2015

Overexpression and knockout of miR-126 both promote leukemogenesis.

Zejuan Li; Ping Chen; Rui Su; Yuanyuan Li; Chao Hu; Yungui Wang; Stephen Arnovitz; Miao He; Sandeep Gurbuxani; Zhixiang Zuo; Abdel G. Elkahloun; Shenglai Li; Hengyou Weng; Hao Huang; Mary Beth Neilly; Shusheng Wang; Eric N. Olson; Richard A. Larson; Michelle M. Le Beau; Jiwang Zhang; Xi Jiang; Minjie Wei; Jie Jin; Paul Liu; Jianjun Chen

It is generally assumed that gain- and loss-of-function manipulations of a functionally important gene should lead to the opposite phenotypes. We show in this study that both overexpression and knockout of microRNA (miR)-126 surprisingly result in enhanced leukemogenesis in cooperation with the t(8;21) fusion genes AML1-ETO/RUNX1-RUNX1T1 and AML1-ETO9a (a potent oncogenic isoform of AML1-ETO). In accordance with our observation that increased expression of miR-126 is associated with unfavorable survival in patients with t(8;21) acute myeloid leukemia (AML), we show that miR-126 overexpression exhibits a stronger effect on long-term survival and progression of AML1-ETO9a-mediated leukemia stem cells/leukemia initiating cells (LSCs/LICs) in mice than does miR-126 knockout. Furthermore, miR-126 knockout substantially enhances responsiveness of leukemia cells to standard chemotherapy. Mechanistically, miR-126 overexpression activates genes that are highly expressed in LSCs/LICs and/or primitive hematopoietic stem/progenitor cells, likely through targeting ERRFI1 and SPRED1, whereas miR-126 knockout activates genes that are highly expressed in committed, more differentiated hematopoietic progenitor cells, presumably through inducing FZD7 expression. Our data demonstrate that miR-126 plays a critical but 2-faceted role in leukemia and thereby uncover a new layer of miRNA regulation in cancer. Moreover, because miR-126 depletion can sensitize AML cells to standard chemotherapy, our data also suggest that miR-126 represents a promising therapeutic target.


Scientific Reports | 2016

DNA Methylation mediated down-regulating of MicroRNA-33b and its role in gastric cancer

Haixin Yin; Peng Song; Rui Su; Gui-Hua Yang; Lei Dong; Min Luo; Bin Wang; Bei Gong; Changzheng Liu; Wei Song; Fang Wang; Yanni Ma; Jun-Wu Zhang; Weibin Wang; Jia Yu

The discovery of microRNAs (miRNAs) provides a new and powerful tool for studying the mechanism, diagnosis and treatment of human cancers. Currently, down-regulation of tumor suppressive miRNAs by CpG island hypermethylation is emerging as a common hallmark of cancer. Here, we reported that the down-regulation of miR-33b was associated with pM stage of gastric cancer (GC) patients. Ectopic expression of miR-33b in HGC-27 and MGC-803 cells inhibited cell proliferation, migration and invasion, which might be due to miR-33b targeting oncogene c-Myc. Moreover, enhanced methylation level of the CpG island upstream of miR-33b in GC patients with down-regulated miR-33b was confirmed by methylation-specific PCR (MSP) amplification. Furthermore, re-introduction of miR-33b significantly suppressed tumorigenesis of GC cells in the nude mice. In conclusion, miR-33b acts as a tumor suppressor and hypermethylation of the CpG island upstream of miR-33b is responsible for its down-regulation in gastric cancer.


Scientific Reports | 2015

ZFP36L1 promotes monocyte/macrophage differentiation by repressing CDK6.

Ming-Tai Chen; Lei Dong; Xin-Hua Zhang; Xiao-Lin Yin; Hong-Mei Ning; Chao Shen; Rui Su; Feng Li; Li Song; Yanni Ma; Fang Wang; Hua-Lu Zhao; Jia Yu; Jun-Wu Zhang

RNA binding proteins (RBPs)-mediated post-transcriptional control has been implicated in influencing various aspects of RNA metabolism and playing important roles in mammalian development and pathological diseases. However, the functions of specific RBPs and the molecular mechanisms through which they act in monocyte/macrophage differentiation remain to be determined. In this study, through bioinformatics analysis and experimental validation, we identify that ZFP36L1, a member of ZFP36 zinc finger protein family, exhibits significant decrease in acute myeloid leukemia (AML) patients compared with normal controls and remarkable time-course increase during monocyte/macrophage differentiation of PMA-induced THP-1 and HL-60 cells as well as induction culture of CD34+ hematopoietic stem/progenitor cells (HSPCs). Lentivirus-mediated gain and loss of function assays demonstrate that ZFP36L1 acts as a positive regulator to participate in monocyte/macrophage differentiation. Mechanistic investigation further reveals that ZFP36L1 binds to the CDK6 mRNA 3′untranslated region bearing adenine-uridine rich elements and negatively regulates the expression of CDK6 which is subsequently demonstrated to impede the in vitro monocyte/macrophage differentiation of CD34+ HSPCs. Collectively, our work unravels a ZFP36L1-mediated regulatory circuit through repressing CDK6 expression during monocyte/macrophage differentiation, which may also provide a therapeutic target for AML therapy.


Cancer Research | 2016

PBX3 and MEIS1 Cooperate in Hematopoietic Cells to Drive Acute Myeloid Leukemias Characterized by a Core Transcriptome of the MLL-Rearranged Disease

Zejuan Li; Ping Chen; Rui Su; Chao Hu; Yuanyuan Li; Abdel G. Elkahloun; Zhixiang Zuo; Sandeep Gurbuxani; Stephen Arnovitz; Hengyou Weng; Yungui Wang; Shenglai Li; Hao Huang; Mary Beth Neilly; Gang Greg Wang; Xi Jiang; Paul Liu; Jie Jin; Jianjun Chen

Overexpression of HOXA/MEIS1/PBX3 homeobox genes is the hallmark of mixed lineage leukemia (MLL)-rearranged acute myeloid leukemia (AML). HOXA9 and MEIS1 are considered to be the most critical targets of MLL fusions and their coexpression rapidly induces AML. MEIS1 and PBX3 are not individually able to transform cells and were therefore hypothesized to function as cofactors of HOXA9. However, in this study, we demonstrate that coexpression of PBX3 and MEIS1 (PBX3/MEIS1), without ectopic expression of a HOX gene, is sufficient for transformation of normal mouse hematopoietic stem/progenitor cells in vitro. Moreover, PBX3/MEIS1 overexpression also caused AML in vivo, with a leukemic latency similar to that caused by forced expression of MLL-AF9, the most common form of MLL fusions. Furthermore, gene expression profiling of hematopoietic cells demonstrated that PBX3/MEIS1 overexpression, but not HOXA9/MEIS1, HOXA9/PBX3, or HOXA9 overexpression, recapitulated the MLL-fusion-mediated core transcriptome, particularly upregulation of the endogenous Hoxa genes. Disruption of the binding between MEIS1 and PBX3 diminished PBX3/MEIS1-mediated cell transformation and HOX gene upregulation. Collectively, our studies strongly implicate the PBX3/MEIS1 interaction as a driver of cell transformation and leukemogenesis, and suggest that this axis may play a critical role in the regulation of the core transcriptional programs activated in MLL-rearranged and HOX-overexpressing AML. Therefore, targeting the MEIS1/PBX3 interaction may represent a promising therapeutic strategy to treat these AML subtypes.


Cell Research | 2018

RNA N 6 -methyladenosine modification in cancers: current status and perspectives

Xiaolan Deng; Rui Su; Hengyou Weng; Huilin Huang; Zejuan Li; Jianjun Chen

N6-methyladenosine (m6A), the most abundant internal modification in eukaryotic messenger RNAs (mRNAs), has been shown to play critical roles in various normal bioprocesses such as tissue development, stem cell self-renewal and differentiation, heat shock or DNA damage response, and maternal-to-zygotic transition. The m6A modification is deposited by the m6A methyltransferase complex (MTC; i.e., writer) composed of METTL3, METTL14 and WTAP, and probably also VIRMA and RBM15, and can be removed by m6A demethylases (i.e., erasers) such as FTO and ALKBH5. The fates of m6A-modified mRNAs rely on the functions of distinct proteins that recognize them (i.e., readers), which may affect the stability, splicing, and/or translation of target mRNAs. Given the functional importance of the m6A modification machinery in normal bioprocesses, it is not surprising that evidence is emerging that dysregulation of m6A modification and the associated proteins also contributes to the initiation, progression, and drug response of cancers. In this review, we focus on recent advances in the study of biological functions and the underlying molecular mechanisms of dysregulated m6A modification and the associated machinery in the pathogenesis and drug response of various types of cancers. In addition, we also discuss possible therapeutic interventions against the dysregulated m6A machinery to treat cancers.


Scientific Reports | 2017

ALOX5 exhibits anti-tumor and drug-sensitizing effects in MLL -rearranged leukemia

Yungui Wang; Jennifer R. Skibbe; Chao Hu; Lei Dong; Kyle Ferchen; Rui Su; Chenying Li; Hao Huang; Hengyou Weng; Huilin Huang; Xi Qin; Jie Jin; Jianjun Chen; Xi Jiang

MLL-rearranged acute myeloid leukemia (AML) remains a fatal disease with a high rate of relapse and therapeutic failure due to chemotherapy resistance. In analysis of our Affymetrix microarray profiling and chromatin immunoprecipitation (ChIP) assays, we found that ALOX5 is especially down-regulated in MLL-rearranged AML, via transcription repression mediated by Polycomb repressive complex 2 (PRC2). Colony forming/replating and bone marrow transplantation (BMT) assays showed that Alox5 exhibited a moderate anti-tumor effect both in vitro and in vivo. Strikingly, leukemic cells with Alox5 overexpression showed a significantly higher sensitivity to the standard chemotherapeutic agents, i.e., doxorubicin (DOX) and cytarabine (Ara-C). The drug-sensitizing role of Alox5 was further confirmed in human and murine MLL-rearranged AML cell models in vitro, as well as in the in vivo MLL-rearranged AML BMT model coupled with treatment of “5 + 3” (i.e. DOX plus Ara-C) regimen. Stat and K-Ras signaling pathways were negatively correlated with Alox5 overexpression in MLL-AF9-leukemic blast cells; inhibition of the above signaling pathways mimicked the drug-sensitizing effect of ALOX5 in AML cells. Collectively, our work shows that ALOX5 plays a moderate anti-tumor role and functions as a drug sensitizer, with a therapeutic potential, in MLL-rearranged AML.


Nature Communications | 2018

Author Correction: Targeted inhibition of STAT/TET1 axis as a therapeutic strategy for acute myeloid leukemia

Xi Jiang; Chao Hu; Kyle Ferchen; Ji Nie; Xiaolong Cui; Chih-Hong Chen; Liting Cheng; Zhixiang Zuo; William Seibel; Chunjiang He; Yixuan Tang; Jennifer R. Skibbe; Mark Wunderlich; William C. Reinhold; Lei Dong; Chao Shen; Stephen Arnovitz; Bryan Ulrich; Jiuwei Lu; Hengyou Weng; Rui Su; Huilin Huang; Yungui Wang; Chenying Li; Xi Qin; James C. Mulloy; Yi Zheng; Jiajie Diao; Jie Jin; Chong Li

The original version of this Article contained an error in the spelling of the author James C. Mulloy, which was incorrectly given as James Mulloy. This has now been corrected in both the PDF and HTML versions of the Article.


Cell | 2018

R-2HG Exhibits Anti-tumor Activity by Targeting FTO/m6A/MYC/CEBPA Signaling

Rui Su; Lei Dong; Chenying Li; Sigrid Nachtergaele; Mark Wunderlich; Ying Qing; Xiaolan Deng; Yungui Wang; Xiaocheng Weng; Chao Hu; Mengxia Yu; Jennifer R. Skibbe; Qing Dai; Dongling Zou; Tong Wu; Kangkang Yu; Hengyou Weng; Huilin Huang; Kyle Ferchen; Xi Qin; Bin Zhang; Jun Qi; Atsuo T. Sasaki; David R. Plas; James E. Bradner; Minjie Wei; Guido Marcucci; Xi Jiang; James C. Mulloy; Jie Jin

Collaboration


Dive into the Rui Su's collaboration.

Top Co-Authors

Avatar

Hengyou Weng

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Xi Jiang

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Huilin Huang

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jianjun Chen

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Lei Dong

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Xi Qin

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge