Network


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

Hotspot


Dive into the research topics where Wenyan He is active.

Publication


Featured researches published by Wenyan He.


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.


Journal of Genetics and Genomics | 2010

Characterization of OP9 as authentic mesenchymal stem cell line

Jiao Gao; Xin-Long Yan; Ren Li; Yi Liu; Wenyan He; Shengkun Sun; Yu Zhang; Bing Liu; Jiaxiang Xiong; Ning Mao

Mesenchymal stem cells (MSCs) are multipotent stem cells capable of differentiating into various cell types, including osteocytes, chondrocytes, adipocytes, myocytes, and tenocytes. However, the difficulty or failure in expanding the mouse MSCs in vitro greatly hampered important research in animal models. The OP9, a stromal cell line from mouse bone marrow, has hematopoietic supportive capacity. Here, we report that the OP9 has the immunophenotype (CD45(-), CD11b(-), FLK-1(-), CD31(-), CD34(-), CD44(+), CD29(+), Sca-1(+), CD86(-), and MHCII(-)) identical to canonical mouse MSCs. The expression of CD140a(+), CD140b(+), alpha-SMA(+) and Calponin(+) suggested the perivascular origin of OP9. Functionally, the OP9 had strong clonogenic ability and could be induced into osteocytes, chondrocytes and adipocytes. The lymphocyte transformation test (LTT) and mixed leukocyte reaction (MLR) showed that the OP9 could suppress T lymphocyte proliferation stimulated by nonspecific mitogens (PHA) or allogeneic lymphocytes (BALB/c T cells). Finally, the migration of OP9 could be efficiently induced by bFGF, IGF-1, IL-3, PDGF-BB, TGF-beta1 and TGF-beta3. In conclusion, the OP9 were bona fide MSCs, and such homogenous cell line will be helpful to delineate biological features of MSCs at the stem cell level.


Haematologica | 2010

Interleukin-3 promotes hemangioblast development in mouse aorta-gonad-mesonephros region

Wenyan He; Yu Lan; Hui-Yu Yao; Zhuan Li; Xiao-Yan Wang; Xiu-Sen Li; Jiyan Zhang; Yu Zhang; Bing Liu; Ning Mao

Background The hemangioblast is a bi-potential precursor cell with the capacity to differentiate into hematopoietic and vascular cells. In mouse E7.0–7.5 embryos, the hemangioblast can be identified by a clonal blast colony-forming cell (BL-CFC) assay or single cell OP9 co-culture. However, the ontogeny of the hemangioblast in mid-gestation embryos is poorly defined. Design and Methods The BL-CFC assay and the OP9 system were combined to illustrate the hemangioblast with lymphomyeloid and vascular potential in the mouse aorta-gonad-mesonephros region. The colony-forming assay, reverse transcriptase polymerase chain reaction analysis, immunostaining and flow cytometry were used to identify the hematopoietic potential, and Matrigel- or OP9-based methods were employed to evaluate endothelial progenitor activity. Results Functionally, the aorta-gonad-mesonephros-derived BL-CFC produced erythroid/myeloid progenitors, CD19+ B lymphocytes, and CD3+TCRβ+ T lymphocytes. Meanwhile, the BL-CFC-derived adherent cells generated CD31+ tube-like structures on OP9 stromal cells, validating the endothelial progenitor potential. The aorta-gonad-mesonephros-derived hemangioblast was greatly enriched in CD31+, endomucin+ and CD105+ subpopulations, which collectively pinpoints the endothelial layer as the main location. Interestingly, the BL-CFC was not detected in yolk sac, placenta, fetal liver or embryonic circulation. Screening of candidate cytokines revealed that interleukin-3 was remarkable in expanding the BL-CFC in a dose-dependent manner through the JAK2/STAT5 and MAPK/ERK pathways. Neutralizing interleukin-3 in the aorta-gonad-mesonephros region resulted in reduced numbers of BL-CFC, indicating the physiological requirement for this cytokine. Both hematopoietic and endothelial differentiation potential were significantly increased in interleukin-3-treated BL-CFC, suggesting a persistent positive influence. Intriguingly, interleukin-3 markedly amplified primitive erythroid and macrophage precursors in E7.5 embryos. Quantitative polymerase chain reaction analysis demonstrated declined Flk-1 and elevated Scl and von Willebrand factor transcription upon interleukin-3 stimulation, indicating accelerated hemangiopoiesis. Conclusions The hemangioblast with lymphomyeloid potential is one of the precursors of definitive hematopoiesis in the mouse aorta-gonad-mesonephros region. Interleukin-3 has a regulatory role with regards to both the number and capacity of the dual-potential hemangioblast.


Blood | 2014

Endothelial Smad4 restrains the transition to hematopoietic progenitors via suppression of ERK activation

Yu Lan; Wenyan He; Zhuan Li; Yu Wang; Jun Wang; Jiao Gao; Weili Wang; Tao Cheng; Bing Liu; Xiao Yang

In mouse mid-gestational embryos, definitive hematopoietic stem progenitor cells are derived directly from a very small proportion of the arterial endothelium. However, the physiological mechanisms restraining excessive endothelial-hematopoietic transition remain elusive. We show here that genetic deletion of Smad4 from the endothelium stage (using Tie2-Cre), but not from embryonic hematopoietic cells (using Vav-Cre), leads to a strikingly augmented emergence of intra-arterial hematopoietic clusters and an enhanced in vitro generation of hematopoietic progenitors, with no increase in the proliferation and survival of hematopoietic cluster cells. This finding indicates a temporally restricted negative effect of Smad4 on the endothelial to hematopoietic progenitor transition. Furthermore, the absence of endothelial Smad4 causes an increased expression of subaortic bone morphogenetic protein 4 and an activation of aortic extracellular signal-regulated kinase, thereby resulting in the excessive generation of blood cells. Collectively, our data for the first time identify a physiological suppressor that functions specifically during the transition of endothelial cells to hematopoietic progenitors and further suggest that endothelial Smad4 is a crucial modulator of the subaortic microenvironment that controls the hematopoietic fate of the aortic endothelium.


Developmental Dynamics | 2011

Migration of dorsal aorta mesenchymal stem cells induced by mouse embryonic circulation

Xin-Long Yan; Yu Lan; Xiao-Yan Wang; Wenyan He; Hui-Yu Yao; Dongbo Chen; Jiaxiang Xiong; Jiao Gao; Zhuan Li; Guan Yang; Xiu-Sen Li; Yuan-Lin Liu; Jiyan Zhang; Bing Liu; Ning Mao

Mesenchymal stem cells (MSCs) represent powerful tools for regenerative medicine for their differentiation and migration capacity. However, ontogeny and migration of MSCs in mammalian mid‐gestation conceptus is poorly understood. We identified canonical MSCs in the mouse embryonic day (E) 11.5 dorsal aorta (DA). They possessed homogenous immunophenotype (CD45−CD31−Flk‐1−CD44+CD29+), expressed perivascular markers (α‐SMA+NG2+PDGFRβ+PDGFRα+), and had tri‐lineage differentiation potential (osteoblasts, adipocytes, and chondrocytes). Of interest, MSCs were also detected in E12.5–E13.5 embryonic circulation, 24 hr later than in DA, suggesting migration like hematopoietic stem cells. Functionally, E12.5 embryonic blood could trigger efficient migration of DA‐MSCs through platelet‐derived growth factor (PDGF) receptor‐, transforming growth factor‐beta receptor‐, but not basic fibroblast growth factor receptor‐mediated signaling. Moreover, downstream JNK and AKT signaling pathway played important roles in embryonic blood‐ or PDGF‐mediated migration of DA‐derived MSCs. Taken together, these results revealed that clonal MSCs developed in the mouse DA. More importantly, the embryonic circulation, in addition to its conventional transporting roles, could modulate migration of MSC during early embryogenesis. Developmental Dynamics, 2011.


International Journal of Hematology | 2012

Characterization of hemangioblast in umbilical arteries of mid-gestation mouse embryos

Weixi Niu; He Huang; Liangyu Zhao; Zhuan Li; Wenyan He; Bing Liu; Longgui Li; Jiaxiang Xiong

Hemangioblasts are the common precursors of hematopoietic and vascular cells, and are characterized as blast colony-forming cells (BL-CFCs) in vitro. We previously identified BL-CFCs in the mouse aorta–gonads–mesonephros (AGM) region, but not yolk sac, placenta, circulation, or fetal liver. Here, we aim to determine whether BL-CFCs develop in the umbilical arteries (UA) that link the dorsal aorta (sub-region of AGM) and placenta. We find that the UA cells of E11.5 mouse embryos were capable of generating typical blast colonies. On replating, these colonies produced erythroid/myeloid progenitors and B220+ B lymphocytes in vitro, corroborating their definitive hematopoietic nature. They also generated CD31+ or endomucin+ tube-like structures on OP9 stromal cells, showing their endothelial potential. The proximal and distal regions of UA had equal numbers of BL-CFCs. To evaluate whether BL-CFCs can be autonomously maintained or expanded in UA or AGM, in vitro organ culture was performed. Interestingly, the BL-CFC pool in the AGM was significantly amplified, in striking contrast to a decrease in the UA. Taken together, our findings indicate that in addition to the AGM the UA serves as an important, but less supportive, niche for hemangioblast development.


Blood | 2008

Identification of mesenchymal stem cells in aorta-gonad-mesonephros and yolk sac of human embryos

Xiao-Yan Wang; Yu Lan; Wenyan He; Lei Zhang; Hui-Yu Yao; Chun-Mei Hou; Ying Tong; Yuan-Lin Liu; Guan Yang; Xiao-Dan Liu; Xiao Yang; Bing Liu; Ning Mao


Journal of Genetics and Genomics | 2013

Generation of Hematopoietic Stem Cells from Purified Embryonic Endothelial Cells by a Simple and Efficient Strategy

Zhuan Li; Fan Zhou; Dongbo Chen; Wenyan He; Yanli Ni; Lingfei Luo; Bing Liu


Experimental Hematology | 2015

Rictor is pivotal for the emergence and function of embryonic hematopoietic stem cells

Weili Wang; Wenyan He; Xiaohuan Mu; Jie Zhou; Yanli Ni; Yunze Zhao; Xiaomin Wang; Qianfei Wang; Tao Cheng; Bing Liu; Weiping Yuan

Collaboration


Dive into the Wenyan He's collaboration.

Top Co-Authors

Avatar

Bing Liu

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Zhuan Li

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Xiao Yang

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Dongbo Chen

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Fan Zhou

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Jiao Gao

Academy of Military Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Jiaxiang Xiong

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Tao Cheng

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

Weiping Yuan

Peking Union Medical College

View shared research outputs
Researchain Logo
Decentralizing Knowledge