Network


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

Hotspot


Dive into the research topics where Qianfei Wang is active.

Publication


Featured researches published by Qianfei Wang.


Nature Genetics | 2014

Identification of functional cooperative mutations of SETD2 in human acute leukemia

Xiaofan Zhu; Fuhong He; Huimin Zeng; Shaoping Ling; Aili Chen; Yaqin Wang; Xiaomei Yan; Wei Wei; Yakun Pang; Hui Cheng; Chunlan Hua; Yue Zhang; Yang X; Xin Lu; Lihua Cao; Lingtong Hao; Lili Dong; Wei Zou; Jun Wu; Xia Li; Si Zheng; Jin Yan; Jing Zhou; Lixia Zhang; Shuangli Mi; Xiaojuan Wang; Li Zhang; Yao Zou; Yumei Chen; Zhe Geng

Acute leukemia characterized by chromosomal rearrangements requires additional molecular disruptions to develop into full-blown malignancy, yet the cooperative mechanisms remain elusive. Using whole-genome sequencing of a pair of monozygotic twins discordant for MLL (also called KMT2A) gene–rearranged leukemia, we identified a transforming MLL-NRIP3 fusion gene and biallelic mutations in SETD2 (encoding a histone H3K36 methyltransferase). Moreover, loss-of-function point mutations in SETD2 were recurrent (6.2%) in 241 patients with acute leukemia and were associated with multiple major chromosomal aberrations. We observed a global loss of H3K36 trimethylation (H3K36me3) in leukemic blasts with mutations in SETD2. In the presence of a genetic lesion, downregulation of SETD2 contributed to both initiation and progression during leukemia development by promoting the self-renewal potential of leukemia stem cells. Therefore, our study provides compelling evidence for SETD2 as a new tumor suppressor. Disruption of the SETD2-H3K36me3 pathway is a distinct epigenetic mechanism for leukemia development.


Genes & Development | 2013

H3.3 actively marks enhancers and primes gene transcription via opening higher-ordered chromatin

Ping Chen; Jicheng Zhao; Yan Wang; Min Wang; Haizhen Long; Dan Liang; Li Huang; Zengqi Wen; Wei Li; Xia Li; Hongli Feng; Haiyong Zhao; Ping Zhu; Ming Li; Qianfei Wang; Guohong Li

The histone variants H3.3 and H2A.Z have recently emerged as two of the most important features in transcriptional regulation, the molecular mechanism of which still remains poorly understood. In this study, we investigated the regulation of H3.3 and H2A.Z on chromatin dynamics during transcriptional activation. Our in vitro biophysical and biochemical investigation showed that H2A.Z promoted chromatin compaction and repressed transcriptional activity. Surprisingly, with only four to five amino acid differences from the canonical H3, H3.3 greatly impaired higher-ordered chromatin folding and promoted gene activation, although it has no significant effect on the stability of mononucleosomes. We further demonstrated that H3.3 actively marks enhancers and determines the transcriptional potential of retinoid acid (RA)-regulated genes via creating an open chromatin signature that enables the binding of RAR/RXR. Additionally, the H3.3-dependent recruitment of H2A.Z on promoter regions resulted in compaction of chromatin to poise transcription, while RA induction results in the incorporation of H3.3 on promoter regions to activate transcription via counteracting H2A.Z-mediated chromatin compaction. Our results provide key insights into the mechanism of how histone variants H3.3 and H2A.Z function together to regulate gene transcription via the modulation of chromatin dynamics over the enhancer and promoter regions.


Blood | 2011

MLL fusion proteins preferentially regulate a subset of wild-type MLL target genes in the leukemic genome

Qianfei Wang; George Wu; Shuangli Mi; Fuhong He; Jun Wu; Jingfang Dong; Roger T. Luo; Ryan J. Mattison; Joseph J. Kaberlein; Shyam Prabhakar; Hongkai Ji; Michael J. Thirman

MLL encodes a histone methyltransferase that is critical in maintaining gene expression during embryonic development and hematopoiesis. 11q23 translocations result in the formation of chimeric MLL fusion proteins that act as potent drivers of acute leukemia. However, it remains unclear what portion of the leukemic genome is under the direct control of MLL fusions. By comparing patient-derived leukemic cell lines, we find that MLL fusion-bound genes are a small subset of that recognized by wild-type MLL. In an inducible MLL-ENL model, MLL fusion protein binding and changes in H3K79 methylation are limited to a specific portion of the genome, whereas wild-type MLL distributes to a much larger set of gene loci. Surprisingly, among 223 MLL-ENL-bound genes, only 12 demonstrate a significant increase in mRNA expression on induction of the fusion protein. In addition to Hoxa9 and Meis1, this includes Eya1 and Six1, which comprise a heterodimeric transcription factor important in several developmental pathways. We show that Eya1 has the capacity to immortalize hematopoietic progenitor cells in vitro and collaborates with Six1 in hematopoietic transformation assays. Altogether, our data suggest that MLL fusions contribute to the development of acute leukemia through direct activation of a small set of target genes.


Molecular and Cellular Biology | 2000

c-Myb binds to a sequence in the proximal region of the RAG-2 promoter and is essential for promoter activity in T-lineage cells.

Qianfei Wang; Josh Lauring; Mark S. Schlissel

ABSTRACT The RAG-2 gene encodes a component of the V(D)J recombinase which is essential for the assembly of antigen receptor genes in B and T lymphocytes. Previously, we reported that the transcription factor BSAP (PAX-5) regulates the murine RAG-2 promoter in B-cell lines. A partially overlapping but distinct region of the proximal RAG-2 promoter was also identified as an important element for promoter activity in T cells; however, the responsible factor was unknown. In this report, we present data demonstrating that c-Myb binds to a Myb consensus site within the proximal promoter and is critical for its activity in T-lineage cells. We show that c-Myb can transactivate a RAG-2 promoter-reporter construct in cotransfection assays and that this transactivation depends on the proximal promoter Myb consensus site. By using a chromatin immunoprecipitation (ChIP) strategy, fractionation of chromatin with anti-c-Myb antibody specifically enriched endogenous RAG-2 promoter DNA sequences. DNase I genomic footprinting revealed that the c-Myb site is occupied in a tissue-specific fashion in vivo. Furthermore, an integrated RAG-2 promoter construct with mutations at the c-Myb site was not enriched in the ChIP assay, while a wild-type integrated promoter construct was enriched. Finally, this lack of binding of c-Myb to a chromosomally integrated mutant RAG-2 promoter construct in vivo was associated with a striking decrease in promoter activity. We conclude that c-Myb regulates the RAG-2 promoter in T cells by binding to this consensus c-Myb binding site.


Phytomedicine | 2010

Selective fraction of Scutellaria baicalensis and its chemopreventive effects on MCF-7 human breast cancer cells

Chong-Zhi Wang; Xiao-Li Li; Qianfei Wang; Sangeeta R. Mehendale; Chun-Su Yuan

Based on our previous observation, the whole Scutellaria baicalensis extract (SbE) did not show significant breast cancer cell inhibitory effect. In this study, we isolated a baicalin-deprived-fraction (SbF1) of Scutellaria baicalensis, and baicalin-fraction (SbF3), and evaluated their anti-breast cancer properties using MCF-7 cells. The content of four flavonoids in extract/fractions were determined using high performance liquid chromatography. Analytical data showed that in SbF1, the major constituents are baicalein and wogonin, while SbF3 only contains baicalin. The antiproliferative effects of fractions and SbE were assayed using modified trichrome stain method. SbF1 showed significant antiproliferative effect. Treated with 100mug/ml of SbF1 for 72h inhibited MCF-7 cell growth by 81.6%, while in the same treatment concentration, SbF3 increased cell growth by 22.6%. SbF1 was recognized as an active fraction of SbE. The effects of four flavonoids in SbE, scutellarin, baicalin, baicalein and wogonin, were determined, and data showed that baicalein and wogonin significantly inhibited MCF-7 cell growth. In contrast, in certain concentrations, scutellarin and baicalin increased cancer cell growth. The effects of SbF1 on cell cycle and apoptosis were assayed using flow cytometry. SbF1 arrested MCF-7 cells in S- and G2/M-phases, and significantly increased induction of cell apoptosis. These combined phytochemical and biological data provide evidence for further chemopreventive studies of the baicalin-deprived SbE on breast cancer.


Blood Cells Molecules and Diseases | 2003

Regulation of granulocyte and monocyte differentiation by CCAAT/enhancer binding protein α

Alan D. Friedman; Jeffrey R. Keefer; Tanawan Kummalue; Huaitian Liu; Qianfei Wang; Rebecca Cleaves

CCAAT/enhancer binding protein alpha (C/EBPalpha)-ER induces 32Dcl3 neutrophilic differentiation and inhibits 32DPKCdelta maturation to macrophages in response to phorbol ester. In 32Dcl3 cells, C/EBPalpha-ER rapidly induces the PU.1 and C/EBPalpha RNAs even in the presence of cycloheximide, suggesting that these are direct C/EBPalpha genetic targets. C/EBPalpha strongly binds and modestly activates the murine PU.1 promoter via an evolutionarily conserved binding site. C/EBPalpha-ER variants incapable of binding DNA still slow G1 progression but do not induce differentiation. N-terminally truncated C/EBPalpha variants, including the p30 isoform expressed in a subset of AMLs, also retain the ability to slow 32D cl3 proliferation, whereas the C/EBPalpha(BRM2)-ER variant does not slow G1 progression, has a reduced capacity to induce early granulocytic markers, and does not induce terminal maturation. In 32DPKCdelta cells, C/EBPalpha-ER strongly inhibits endogenous or exogenous JunB induction, dependent upon the outer surface of the C/EBPalpha basic region, but does not inhibit c-Jun, PU.1, or C/EBPbeta expression. Exogenous JunB restores AP-1 DNA binding but does not overcome inhibition of monopoiesis by C/EBPalpha-ER. In summary, we propose that while C/EBPalpha is required for development of immature granulocyte-monocyte progenitors, C/EBPalpha subsequently inhibits monopoiesis, via inhibition of JunB express and via additional activities, and induces granulopoiesis, via induction of PU.1, C/EBPepsilon, and cell cycle arrest.


Oncogene | 2003

Cell cycle inhibition mediated by the outer surface of the C/EBPα basic region is required but not sufficient for granulopoiesis

Qianfei Wang; Rebecca Cleaves; Tanawan Kummalue; Claus Nerlov; Alan D. Friedman

CCAAT/enhancer binding protein α (C/EBPα) transactivates target genes dependent upon DNA binding via its basic region-leucine zipper domain and slows G1 progression by interaction with E2F, cdk2, or cdk4. E2F interacts with the non-DNA-binding surface of the C/EBPα basic region and C/EBPα residues 1–70 are required for repressing E2F targets, while cdk2 and cdk4 bind residues 177–191. C/EBPα-ER induces the 32D cl3 myeloblast cell line to differentiate to granulocytes. C/EBPα-ER variants incapable of binding DNA slowed G1, but did not induce early or late granulopoiesis, indicating that cell cycle inhibition as mediated by C/EBPα is not sufficient for differentiation. C/EBPα-ER variants lacking residues 11–70 or residues 11–70 and 178–200 both slowed the G1 to S transition. C/EBPα(GZ)-ER, containing the GCN4 rather than the C/EBPα leucine zipper, also slowed G1. In contrast, C/EBPα(BRM2)-ER, carrying mutations in the outer surface of the basic region required for interaction with E2F, did not slow G1. C/EBPα(BRM2)-ER induced early markers of granulopoiesis much less efficiently than C/EBPα-ER and did not direct terminal maturation. Inhibition of G1 progression using mimosine increased induction of late markers by G-CSF. Thus, both DNA binding and cell cycle arrest, mediated by opposite surfaces of the C/EBPα basic region, are required for granulopoiesis.


Oncogene | 2004

C/EBPαp30, a myeloid leukemia oncoprotein, limits G-CSF receptor expression but not terminal granulopoiesis via site-selective inhibition of C/EBP DNA binding

Rebecca Cleaves; Qianfei Wang; Alan D. Friedman

Heterozygous mutations of the CEBPA gene are present in 5% of acute myeloid leukemia (AML) cases and often lead to the expression of an N-terminally truncated, 30 kDa isoform, C/EBPαp30, from an internal translation start site. We have assessed the effect of C/EBPαp30 on granulopoiesis utilizing C/EBPαp30-ER, containing the estradiol receptor ligand-binding domain. In contrast to C/EBPα-ER, C/EBPαp30-ER did not induce 32Dcl3 myeloid cell differentiation in IL-3. However, both isoforms, when expressed at high levels, were capable of inhibiting E2F activity in 32Dcl3 cells and of slowing their G1 to S progression. C/EBPαp30 repressed expression of the endogenous G-CSF receptor several-fold. To facilitate investigation of the effect of C/EBPαp30-ER on granulopoiesis downstream of G-CSF signalling, we coexpressed exogenous G-CSF receptor. C/EBPαp30-ER/GR cells expressed several granulocytic markers in G-CSF and demonstrated nuclear maturation. Rat C/EBPα-ER and C/EBPαp30-ER, expressed in 293T cells, bound the C/EBP site from the NE gene with similar affinity, as did human C/EBPα and C/EBPαp30. In contrast, C/EBPαp30 bound the C/EBP sites in the PU.1 or GR gene with 3–6-fold reduced affinity. Thus, the selective inhibition of GR expression by C/EBPαp30-ER is due in part to its variable affinity for C/EBP sites. Variation in affinity for selected cis elements among isoforms may affect the biology of basic region-leucine zipper (bZIP) proteins.


Genome Biology | 2007

Detection of weakly conserved ancestral mammalian regulatory sequences by primate comparisons

Qianfei Wang; Shyam Prabhakar; Sumita Chanan; Jan Fang Cheng; Edward M. Rubin; Dario Boffelli

BackgroundGenomic comparisons between human and distant, non-primate mammals are commonly used to identify cis-regulatory elements based on constrained sequence evolution. However, these methods fail to detect functional elements that are too weakly conserved among mammals to distinguish them from non-functional DNA.ResultsTo evaluate a strategy for large scale genome annotation that is complementary to the commonly used distal species comparisons, we explored the potential of deep intra-primate sequence comparisons. We sequenced the orthologs of 558 kb of human genomic sequence, covering multiple loci involved in cholesterol homeostasis, in 6 non-human primates. Our analysis identified six non-coding DNA elements displaying significant conservation among primates but undetectable in more distant comparisons. In vitro and in vivo tests revealed that at least three of these six elements have regulatory function. Notably, the mouse orthologs of these three functional human sequences had regulatory activity despite their lack of significant sequence conservation, indicating that they are ancestral mammalian cis-regulatory elements. These regulatory elements could be detected even in a smaller set of three primate species including human, rhesus and marmoset.ConclusionWe have demonstrated that intra-primate sequence comparisons can be used to identify functional modules in large genomic regions, including cis-regulatory elements that are not detectable through comparison with non-mammalian genomes. With the available human and rhesus genomes and that of marmoset, which is being actively sequenced, this strategy can be extended to the whole genome in the near future.


Oncogene | 2006

p78/MCRS1 forms a complex with centrosomal protein Nde1 and is essential for cell viability.

Yoshihiko Hirohashi; Qianfei Wang; Qingdu Liu; Xiulian Du; Hongtao Zhang; Noriyuki Sato; Mark I. Greene

The centrosome, an organelle that functions as the major microtubule-organizing center, plays an essential role in the formation of the mitotic spindle and guiding accurate chromosome segregation. Centrosome aberrations are frequently associated with various forms of human cancers and it is thought that defects in this organelle contribute to genomic instability and malignant transformation. We recently identified and characterized a centrosome-localized protein complex that is comprised of Su48 and Nde1. Disruption of the normal function of these proteins leads to abnormal cell division. To extend our understanding of how this protein complex operates, we sought to identify Nde1-interacting molecules by the yeast two-hybrid screening method. Here, we demonstrate that both Nde1 and Su48 can associate with p78/MCRS1, a protein implicated in cancer development. We found that, whereas the majority of p78 localizes to the nucleus as reported in earlier studies, a fraction of the p78 protein can be detected in the centrosome. Moreover, we determined that a region containing the forkhead-associated domain of p78 is involved in association with Nde1 and Su48, as well as in centrosomal localization. We also provide evidence that the association between p78 and Nde1 is regulated by phosphorylation on Nde1. Furthermore, abrogation of the endogenous p78 function by small interfering RNA knockdown causes cell death and a modest delay in mitosis. These results indicate that a subset of the p78 proteins comprises a component of the centrosome and that p78 is essential for cell viability.

Collaboration


Dive into the Qianfei Wang's collaboration.

Top Co-Authors

Avatar

Fuhong He

Beijing Institute of Genomics

View shared research outputs
Top Co-Authors

Avatar

Gang Huang

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Xiaomei Yan

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Tao Cheng

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Yue Zhang

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Aili Chen

Beijing Institute of Genomics

View shared research outputs
Top Co-Authors

Avatar

Zhijian Xiao

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Yoshihiro Hayashi

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Yunzhu Dong

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jiachen Bu

Beijing Institute of Genomics

View shared research outputs
Researchain Logo
Decentralizing Knowledge