Network


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

Hotspot


Dive into the research topics where Weiping Yuan is active.

Publication


Featured researches published by Weiping Yuan.


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.


Cell Stem Cell | 2012

Mouse Embryonic Head as a Site for Hematopoietic Stem Cell Development

Zhuan Li; Yu Lan; Wenyan He; Dongbo Chen; Jun Wang; Fan Zhou; Yu Wang; Huayan Sun; Xianda Chen; Chunhong Xu; Sha Li; Yakun Pang; Guangzhou Zhang; Liping Yang; Lingling Zhu; Ming Fan; Aijia Shang; Zhenyu Ju; Lingfei Luo; Yu-Qiang Ding; Wei Guo; Weiping Yuan; Xiao Yang; Bing Liu

In the mouse embryo, the aorta-gonad-mesonephros (AGM) region is considered to be the sole location for intraembryonic emergence of hematopoietic stem cells (HSCs). Here we report that, in parallel to the AGM region, the E10.5-E11.5 mouse head harbors bona fide HSCs, as defined by long-term, high-level, multilineage reconstitution and self-renewal capacity in adult recipients, before HSCs enter the circulation. The presence of hemogenesis in the midgestation head is indicated by the appearance of intravascular cluster cells and the blood-forming capacity of a sorted endothelial cell population. In addition, lineage tracing via an inducible VE-cadherin-Cre transgene demonstrates the hemogenic capacity of head endothelium. Most importantly, a spatially restricted lineage labeling system reveals the physiological contribution of cerebrovascular endothelium to postnatal HSCs and multilineage hematopoiesis. We conclude that the mouse embryonic head is a previously unappreciated site for HSC emergence within the developing embryo.


Nature | 2016

Tracing haematopoietic stem cell formation at single-cell resolution

Fan Zhou; Xianlong Li; Weili Wang; Ping Zhu; Jie Zhou; Wenyan He; Meng Ding; Fuyin Xiong; Xiaona Zheng; Zhuan Li; Yanli Ni; Xiaohuan Mu; Lu Wen; Tao Cheng; Yu Lan; Weiping Yuan; Fuchou Tang; Bing Liu

Haematopoietic stem cells (HSCs) are derived early from embryonic precursors, such as haemogenic endothelial cells and pre-haematopoietic stem cells (pre-HSCs), the molecular identity of which still remains elusive. Here we use potent surface markers to capture the nascent pre-HSCs at high purity, as rigorously validated by single-cell-initiated serial transplantation. Then we apply single-cell RNA sequencing to analyse endothelial cells, CD45− and CD45+ pre-HSCs in the aorta–gonad–mesonephros region, and HSCs in fetal liver. Pre-HSCs show unique features in transcriptional machinery, arterial signature, metabolism state, signalling pathway, and transcription factor network. Functionally, activation of mechanistic targets of rapamycin (mTOR) is shown to be indispensable for the emergence of HSCs but not haematopoietic progenitors. Transcriptome data-based functional analysis reveals remarkable heterogeneity in cell-cycle status of pre-HSCs. Finally, the core molecular signature of pre-HSCs is identified. Collectively, our work paves the way for dissection of complex molecular mechanisms regulating stepwise generation of HSCs in vivo, informing future efforts to engineer HSCs for clinical applications.


Nature Medicine | 2010

CIB1 is a Regulator of Pathological Cardiac Hypertrophy

Joerg Heineke; Mannix Auger-Messier; Robert N. Correll; Jian Xu; Matthew J. Benard; Weiping Yuan; Helmut Drexler; Leslie V. Parise; Jeffery D. Molkentin

Hypertrophic heart disease is a leading health problem in Western countries. Here we identified the small EF hand domain–containing protein Ca2+ and integrin–binding protein-1 (CIB1) in a screen for previously unknown regulators of cardiomyocyte hypertrophy. Yeast two-hybrid screening for CIB1-interacting partners identified a related EF hand domain–containing protein, calcineurin B, the regulatory subunit of the prohypertrophic protein phosphatase calcineurin. CIB1 localizes primarily to the sarcolemma in mouse and human myocardium, where it anchors calcineurin to control its activation in coordination with the L-type Ca2+ channel. CIB1 protein amounts and membrane association were enhanced in cardiac pathological hypertrophy, but not in physiological hypertrophy. Consistent with these observations, Cib1-deleted mice showed a marked reduction in myocardial hypertrophy, fibrosis, cardiac dysfunction and calcineurin–nuclear factor of activated T cells (NFAT) activity after pressure overload, whereas the degree of physiologic hypertrophy after swimming exercise was not altered. Transgenic mice with inducible and cardiac-specific overexpression of CIB1 showed enhanced cardiac hypertrophy in response to pressure overload or calcineurin signaling. Moreover, mice lacking Ppp3cb (encoding calcineurin A, β isozyme) showed no enhancement in cardiac hypertrophy associated with CIB1 overexpression. Thus, CIB1 functions as a previously undescribed regulator of cardiac hypertrophy through its ability to regulate the association of calcineurin with the sarcolemma and its activation.


Journal of Cell Biology | 2006

CIB1 is an endogenous inhibitor of agonist-induced integrin αIIbβ3 activation

Weiping Yuan; Tina M. Leisner; Andrew W. McFadden; Zhengyan Wang; Mark K. Larson; Shantres Clark; Christel Boudignon-Proudhon; Stephen C.-T. Lam; Leslie V. Parise

In response to agonist stimulation, the αIIbβ3 integrin on platelets is converted to an active conformation that binds fibrinogen and mediates platelet aggregation. This process contributes to both normal hemostasis and thrombosis. Activation of αIIbβ3 is believed to occur in part via engagement of the β3 cytoplasmic tail with talin; however, the role of the αIIb tail and its potential binding partners in regulating αIIbβ3 activation is less clear. We report that calcium and integrin binding protein 1 (CIB1), which interacts directly with the αIIb tail, is an endogenous inhibitor of αIIbβ3 activation; overexpression of CIB1 in megakaryocytes blocks agonist-induced αIIbβ3 activation, whereas reduction of endogenous CIB1 via RNA interference enhances activation. CIB1 appears to inhibit integrin activation by competing with talin for binding to αIIbβ3, thus providing a model for tightly controlled regulation of αIIbβ3 activation.


Molecular and Cellular Biology | 2006

CIB1 is essential for mouse spermatogenesis.

Weiping Yuan; Tina M. Leisner; Andrew W. McFadden; Shantres Clark; Sylvia Hiller; Nobuyo Maeda; Deborah A. O'Brien; Leslie V. Parise

ABSTRACT CIB1 is a 22-kDa calcium binding, regulatory protein with ∼50% homology to calmodulin and calcineurin B. CIB1 is widely expressed and binds to a number of effectors, such as integrin αIIb, PAK1, and polo-like kinases, in different tissues. However, the in vivo functions of CIB1 are not well understood. To elucidate the function of CIB1 in whole animals, we used homologous recombination in embryonic stem cells to generate Cib1−/− mice. Although Cib1−/− mice grow normally, the males are sterile due to disruption of the haploid phase of spermatogenesis. This is associated with reduced testis size and numbers of germ cells in seminiferous tubules, increased germ cell apoptosis, and the loss of elongated spermatids and sperm. Cib1−/− testes also show increased mRNA and protein expression of the cell cycle regulator Cdc2/Cdk1. In addition, mouse embryonic fibroblasts (MEFs) derived from Cib1−/− mice exhibit a much slower growth rate compared to Cib1+/+ MEFs, suggesting that CIB1 regulates the cell cycle, differentiation of spermatogenic germ cells, and/or differentiation of supporting Sertoli cells.


Cell Reports | 2015

Combined loss of Tet1 and Tet2 promotes B-cell, but not myeloid malignancies in mice

Zhigang Zhao; Li Chen; Meelad M. Dawlaty; Feng Pan; Ophelia Weeks; Yuan Zhou; Zeng Cao; Hui Shi; Jiapeng Wang; Li Lin; Shi Chen; Weiping Yuan; Zhaohui S. Qin; Hongyu Ni; Stephen D. Nimer; Feng Chun Yang; Rudolf Jaenisch; Peng Jin; Mingjiang Xu

TET1/2/3 are methylcytosine dioxygenases that regulate cytosine hydroxymethylation. Tet1/2 are abundantly expressed in HSC/HPCs and are implicated in hematological malignancies. Tet2 deletion in mice causes myeloid malignancies, while Tet1-null mice develop B cell lymphoma after an extended period of latency. Interestingly, TET1/2 are often concomitantly downregulated in acute B-lymphocytic leukemia. Here, we investigated the overlapping and non-redundant functions of Tet1/2 using Tet1/2 double-knockout (DKO) mice. DKO and Tet2(-/-) HSC/HPCs show overlapping and unique 5 hmC and 5 mC profiles. DKO mice exhibit strikingly decreased incidence and delayed onset of myeloid malignancies in comparison to Tet2(-/-) mice and in contrast develop lethal B cell malignancies. Transcriptome analysis of DKO tumors reveals expression changes in many genes dysregulated in human B cell malignancies, including LMO2, BCL6, and MYC. These results highlight the critical roles of TET1/2 individually and together in the pathogenesis of hematological malignancies.


Blood | 2015

Leukemic marrow infiltration reveals a novel role for Egr3 as a potent inhibitor of normal hematopoietic stem cell proliferation

Hui Cheng; Sha Hao; Yanfeng Liu; Yakun Pang; Shihui Ma; Fang Dong; Jing Xu; Guoguang Zheng; Shaoguang Li; Weiping Yuan; Tao Cheng

Cytopenias resulting from the impaired generation of normal blood cells from hematopoietic precursors are important contributors to morbidity and mortality in patients with leukemia. However, the process by which normal hematopoietic cells are overtaken by emerging leukemia cells and how different subsets of hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) are distinctly influenced during leukemic cell infiltration is poorly understood. To investigate these important questions, we used a robust nonirradiated mouse model of human MLL-AF9 leukemia to examine the suppression of HSCs and HPCs during leukemia cell expansion in vivo. Among all the hematopoietic subsets, long-term repopulating HSCs were the least reduced, whereas megakaryocytic-erythroid progenitors were the most significantly suppressed. Notably, nearly all of the HSCs were forced into a noncycling state in leukemic marrow at late stages, but their reconstitution potential appeared to be intact upon transplantation into nonleukemic hosts. Gene expression profiling and further functional validation revealed that Egr3 was a strong limiting factor for the proliferative potential of HSCs. Therefore, this study provides not only a molecular basis for the more tightened quiescence of HSCs in leukemia, but also a novel approach for defining functional regulators of HSCs in disease.


Circulation Research | 2007

CIB1 Regulates Endothelial Cells and Ischemia-Induced Pathological and Adaptive Angiogenesis

Mohamed A. Zayed; Weiping Yuan; Tina M. Leisner; Dan Chalothorn; Andrew W. McFadden; Michael D. Schaller; M. Elizabeth Hartnett; James E. Faber; Leslie V. Parise

Pathological angiogenesis contributes to various ocular, malignant, and inflammatory disorders, emphasizing the need to understand this process on a molecular level. CIB1 (calcium- and integrin-binding protein), a 22-kDa EF-hand–containing protein, modulates the activity of p21-activated kinase 1 in fibroblasts. Because p21-activated kinase 1 also contributes to endothelial cell function, we hypothesized that CIB1 may have a role in angiogenesis. We found that endothelial cells depleted of CIB1 by either short hairpin RNA or homologous recombination have reduced migration, proliferation, and tubule formation. Moreover, loss of CIB1 in these cells decreases p21-activated kinase 1 activation, downstream extracellular signal-regulated kinase 1/2 activation, and matrix metalloproteinase 2 expression, all of which are known to contribute to angiogenesis. Consistent with these findings, tissues derived from CIB1-deficient (CIB1−/−) mice have reduced growth factor–induced microvessel sprouting in ex vivo organ cultures and in vivo Matrigel plugs. Furthermore, in response to ischemia, CIB1−/− mice demonstrate decreased pathological retinal and adaptive hindlimb angiogenesis. Ischemic CIB1−/− hindlimbs also demonstrate increased tissue damage and significantly reduced p21-activated kinase 1 activation. These data therefore reveal a critical role for CIB1 in ischemia-induced pathological and adaptive angiogenesis.


Thrombosis and Haemostasis | 2008

Characterization of calcium- and integrin-binding protein 1 (CIB1) knockout platelets: Potential compensation by CIB family members

Jan C. DeNofrio; Weiping Yuan; Brenda R. Temple; Holly R. Gentry; Leslie V. Parise

Platelet aggregation requires activation of the alphaIIbbeta3 integrin, an event regulated by the integrin cytoplasmic tails. CIB1 binds to the cytoplasmic tail of the integrin alphaIIb subunit. Previous over-expression and knockdown studies in murine megakaryocytes demonstrated that CIB1 inhibits integrin alphaIIbbeta3 activation. Here we analyzed Cib1(-/-) mice to determine the function of CIB1 in platelets in vitro and in vivo. We found that although these mice had no overt platelet phenotype, mRNA level of CIB1 homolog CIB3 was increased in Cib1(-/-) megakaryocytes. In vitro binding experiments showed that recombinant CIB1, -2 and -3 bound specifically to an alphaIIb cytoplasmic tail peptide. Subsequent protein modeling experiments indicated that CIBs 1-3 each have a highly conserved hydrophobic binding pocket. Therefore, the potential exists for compensation for the loss of CIB1 by these CIB family members, thereby preventing pathologic thrombus formation in Cib1(-/-) mice.

Collaboration


Dive into the Weiping Yuan's collaboration.

Top Co-Authors

Avatar

Tao Cheng

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Hui Cheng

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xiaomin Wang

Academy of Medical Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jing Xu

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Yuan Zhou

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Weili Wang

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Xiaofan Zhu

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Yakun Pang

Peking Union Medical College

View shared research outputs
Researchain Logo
Decentralizing Knowledge