Network


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

Hotspot


Dive into the research topics where Wenqian Dong is active.

Publication


Featured researches published by Wenqian Dong.


Nature Communications | 2017

Blockade of IDO-kynurenine-AhR metabolic circuitry abrogates IFN-γ-induced immunologic dormancy of tumor-repopulating cells.

Yuying Liu; Xiaoyu Liang; Xiaonan Yin; Jiadi Lv; Ke Tang; Jingwei Ma; Tiantian Ji; Huafeng Zhang; Wenqian Dong; Xun Jin; Degao Chen; Yanchun Li; Songyan Zhang; Heidi Qunhui Xie; Bin Zhao; Tong Zhao; Jinzhi Lu; Zhuowei Hu; Xuetao Cao; F. Xiao-Feng Qin; Bo Huang

Interactions with the immune system may lead tumorigenic cells into dormancy. However, the underlying molecular mechanism is poorly understood. Using a 3D fibrin gel model, we show that IFN-γ induces tumour-repopulating cells (TRCs) to enter dormancy through an indolamine 2,3-dioxygenase 1 (IDO1)-kynurenine (Kyn)-aryl hydrocarbon receptor (AhR)-p27 dependent pathway. Mechanistically, IFN-γ signalling triggers differentiated tumour cell apoptosis via STAT1; however, when IDO1 and AhR are highly expressed as in TRCs, IFN-γ results in IDO1/AhR-dependent p27 induction that prevents STAT1 signalling, thus suppressing the process of cell death and activating the dormancy program. Blocking the IDO/AhR metabolic circuitry not only abrogates IFN-γ-induced dormancy but also results in enhanced repression of tumour growth by IFN-γ-induced apoptosis of TRCs both in vitro and in vivo. These data present a previously unrecognized mechanism of inducing TRC dormancy by IFN-γ, suggesting a potential effective cancer immunotherapeutic modality through the combination of IFN-γ and IDO/AhR inhibitors.


OncoImmunology | 2016

Tumor cell-derived microparticles polarize M2 tumor-associated macrophages for tumor progression.

Ruihua Ma; Tiantian Ji; Degao Chen; Wenqian Dong; Huafeng Zhang; Xiaonan Yin; Jingwei Ma; Xiaoyu Liang; Yi Zhang; Guan-Xin Shen; Xiaofeng Qin; Bo Huang

ABSTRACT Despite identification of macrophages in tumors (tumor-associated macrophages, TAM) as potential targets for cancer therapy, the origin and function of TAM in the context of malignancy remain poorly characterized. Here, we show that microparticles (MPs), as a by-product, released by tumor cells act as a general mechanism to mediate M2 polarization of TAM. Taking up tumor MPs by macrophages is a very efficient process, which in turn results in the polarization of macrophages into M2 type, not only leading to promoting tumor growth and metastasis but also facilitating cancer stem cell development. Moreover, we demonstrate that the underlying mechanism involves the activation of the cGAS/STING/TBK1/STAT6 pathway by tumor MPs. Finally, in addition to murine tumor MPs, we show that human counterparts also possess consistent effect on human M2 polarization. These findings provide new insights into a critical role of tumor MPs in remodeling of tumor microenvironment and better understanding of the communications between tumors and macrophages.


Nature Communications | 2018

Chloroquine modulates antitumor immune response by resetting tumor-associated macrophages toward M1 phenotype

Degao Chen; Jing Xie; Roland Fiskesund; Wenqian Dong; Xiaoyu Liang; Jiadi Lv; Xun Jin; Jinyan Liu; Siqi Mo; Tianzhen Zhang; Feiran Cheng; Yabo Zhou; Huafeng Zhang; Ke Tang; Jingwei Ma; Yuying Liu; Bo Huang

Resetting tumor-associated macrophages (TAMs) is a promising strategy to ameliorate the immunosuppressive tumor microenvironment and improve innate and adaptive antitumor immunity. Here we show that chloroquine (CQ), a proven anti-malarial drug, can function as an antitumor immune modulator that switches TAMs from M2 to tumor-killing M1 phenotype. Mechanistically, CQ increases macrophage lysosomal pH, causing Ca2+ release via the lysosomal Ca2+ channel mucolipin-1 (Mcoln1), which induces the activation of p38 and NF-κB, thus polarizing TAMs to M1 phenotype. In parallel, the released Ca2+ activates transcription factor EB (TFEB), which reprograms the metabolism of TAMs from oxidative phosphorylation to glycolysis. As a result, CQ-reset macrophages ameliorate tumor immune microenvironment by decreasing immunosuppressive infiltration of myeloid-derived suppressor cells and Treg cells, thus enhancing antitumor T-cell immunity. These data illuminate a previously unrecognized antitumor mechanism of CQ, suggesting a potential new macrophage-based tumor immunotherapeutic modality.Tumour-associated macrophages (TAMs) display an M2 phenotype that promote tumour immune escape. Here the authors show that Chloroquine (CQ), a lysosome inhibitor used against malaria, inhibits tumour growth by switching TAMs into an M1 tumor-killing phenotype by repolarizing macrophages metabolism.


Journal of Clinical Investigation | 2018

STAT3/p53 pathway activation disrupts IFN-β–induced dormancy in tumor-repopulating cells

Yuying Liu; Jiadi Lv; Jinyan Liu; Xiaoyu Liang; Xun Jin; Jing Xie; Le Zhang; Degao Chen; Roland Fiskesund; Ke Tang; Jingwei Ma; Huafeng Zhang; Wenqian Dong; Siqi Mo; Tianzhen Zhang; Feiran Cheng; Yabo Zhou; Qingzhu Jia; Bo Zhu; Yan Kong; Jun Guo; Haizeng Zhang; Zhuowei Hu; Xuetao Cao; F. Xiao-Feng Qin; Bo Huang

Dynamic interaction with the immune system profoundly regulates tumor cell dormancy. However, it is unclear how immunological cues trigger cancer cell–intrinsic signaling pathways for entering into dormancy. Here, we show that IFN-&bgr; treatment induced tumor-repopulating cells (TRC) to enter dormancy through an indolamine 2,3-dioxygenase/kynurenine/aryl hydrocarbon receptor/p27–dependent (IDO/Kyn/AhR/p27-dependent) pathway. Strategies to block this metabolic circuitry did not relieve dormancy, but led to apoptosis of dormant TRCs in murine and human melanoma models. Specifically, blocking AhR redirected IFN-&bgr; signaling to STAT3 phosphorylation through both tyrosine and serine sites, which subsequently facilitated STAT3 nuclear translocation and subsequent binding to the p53 promoter in the nucleus. Upregulation of p53 in turn disrupted the pentose phosphate pathway, leading to excessive ROS production and dormant TRC death. Additionally, in melanoma patients, high expression of IFN-&bgr; correlated with tumor cell dormancy. Identification of this mechanism for controlling TRC dormancy by IFN-&bgr; provides deeper insights into cancer-immune interaction and potential new cancer immunotherapeutic modalities.


Cancer Research | 2018

3D Fibrin Stiffness mediates Dormancy of Tumor-repopulating cells via a Cdc42-driven Tet2 Epigenetic Program

Yuying Liu; Jiadi Lv; Xiaoyu Liang; Xiaonan Yin; Le Zhang; Degao Chen; Xun Jin; Roland Fiskesund; Ke Tang; Jingwei Ma; Huafeng Zhang; Wenqian Dong; Siqi Mo; Tianzhen Zhang; Feiran Cheng; Yabo Zhou; Jing Xie; Ning Wang; Bo Huang

Dormancy is recognized as a critical biological event for tumorigenic cells surviving in an extremely harsh environment. Understanding the molecular process of dormancy can unlock novel approaches to tackle cancers. We recently reported that stem-like tumor-repopulating cells (TRC) sense mechanical signals and rapidly proliferate in a 90 Pa soft fibrin matrix. Here, we show that a stiff mechanical environment induces TRC dormancy via an epigenetic program initiated by translocation of Cdc42, a cytosolic regulator of mechanotransduction, into the nucleus, where it promotes transcription of hydroxymethylating enzyme Tet2. Tet2 epigenetically activated cell-cycle-inhibiting genes p21 and p27 to induce dormancy, but also caused downregulation of integrin β3 to maintain dormancy. This stiffness-mediated dormancy was recapitulated in mouse models for both murine and primary human melanoma TRCs. These data identify an epigenetic program directed by mechanics, which drives highly tumorigenic TRCs to enter dormancy in a stiff mechanical environment.Significance: A mechanics-directed epigenetic program enables tumor-repopulating cells to enter dormancy in a stiff mechanical environment. Cancer Res; 78(14); 3926-37. ©2018 AACR.


OncoImmunology | 2017

Oral delivery of tumor microparticle vaccines activates NOD2 signaling pathway in ileac epithelium rendering potent antitumor T cell immunity

Wenqian Dong; Huafeng Zhang; Xiaonan Yin; Yuying Liu; Degao Chen; Xiaoyu Liang; Xun Jin; Jiadi Lv; Jingwei Ma; Ke Tang; Zhuowei Hu; Xiaofeng Qin; Bo Huang

ABSTRACT Exploiting gut mucosal immunity to design new antitumor vaccination strategy remains unexplored. Tumor cell-derived microparticles (T-MP) are natural biomaterials that are capable of delivering tumor antigens and innate signals to dendritic cells (DC) for tumor-specific T cell immunity. Here, we show that T-MPs by oral vaccination route effectively access and activate mucosal epithelium, leading to subsequent antitumor T cell responses. Oral vaccination of T-MPs generated potent inhibitory effect against the growth of B16 melanoma and CT26 colon cancer in mice, which required both T cell and DC activation. T-MPs, once entering intestinal lumen, were mainly taken up by ileac intestinal epithelial cells (IEC), where T-MPs activated NOD2 and its downstream MAPK and NF-κB, leading to chemokine releasing, including CCL2, from IECs to attract CD103+ CD11c+ DCs. Furthermore, ileac IECs could transcytose T-MPs to the basolateral site, where T-MPs were captured by those DCs for cross-presentation of loaded antigen contents. Elucidating these molecular and cellular mechanisms highlights T-MPs as a novel antitumor oral vaccination strategy with great potential of clinical applications.


Nature Communications | 2018

Publisher Correction: Chloroquine modulates antitumor immune response by resetting tumor-associated macrophages toward M1 phenotype.

Degao Chen; Jing Xie; Roland Fiskesund; Wenqian Dong; Xiaoyu Liang; Jiadi Lv; Xun Jin; Jinyan Liu; Siqi Mo; Tianzhen Zhang; Feiran Cheng; Yabo Zhou; Huafeng Zhang; Ke Tang; Jingwei Ma; Yuying Liu; Bo Huang

In the originally published version of this Article, images in Fig. 5n were inadvertently replaced with duplicates of images in Fig. 5o during the production process. This has now been corrected in both the PDF and HTML versions of the Article.


Cancer immunology research | 2018

Circulating tumor microparticles promote lung metastasis by reprogramming inflammatory and mechanical niches via a macrophage-dependent pathway

Huafeng Zhang; Yuandong Yu; Li Zhou; Jingwei Ma; Ke Tang; Pingwei Xu; Tiantian Ji; Xiaoyu Liang; Jiadi Lv; Wenqian Dong; Tianzhen Zhang; Degao Chen; Jing Xie; Yuying Liu; Bo Huang

Lung macrophages are induced by tumor-derived microparticles to drive development of metastasis via mediators that promote immune, inflammatory, and mechanical reprogramming of the microenvironment. Elucidation of this pathway has implications for therapeutic prevention or treatment of lung metastasis. Despite the frequency of lung metastasis and its associated mortality, the mechanisms behind metastatic tumor cell survival and colonization in the lungs remain elusive. Here, we show that tumor cell–released microparticles (T-MPs) from the primary tumor site play a critical role in the metastatic process. The T-MPs remodeled the lung parenchyma via a macrophage-dependent pathway to create an altered inflammatory and mechanical response to tumor cell invasion. Mechanistically, we show that circulating T-MPs readily enter the lung parenchyma where they are taken up by local macrophages and induce CCL2 production. CCL2 recruits CD11b+Ly6Chigh inflammatory monocytes to the lungs where they mature into F4/80+CD11b+Ly6C− macrophages that not only produce IL6 but also trigger fibrin deposition. IL6 and the deposited fibrin facilitate the survival and growth of tumor-repopulating cells in the lungs by providing chemical and mechanical signals, respectively, thus setting the stage for lung metastasis. These data illustrate that T-MPs reprogram the lung microenvironment promoting metastasis. Cancer Immunol Res; 6(9); 1046–56. ©2018 AACR.


Biomaterials | 2016

Delivery of oncolytic adenovirus into the nucleus of tumorigenic cells by tumor microparticles for virotherapy

Li Ran; Xiaohua Tan; Yanchun Li; Huafeng Zhang; Ruihua Ma; Tiantian Ji; Wenqian Dong; Tong Tong; Yuying Liu; Degao Chen; Xiaonan Yin; Xiaoyu Liang; Ke Tang; Jingwei Ma; Yi Zhang; Xuetao Cao; Zhuowei Hu; Xiaofeng Qin; Bo Huang


Cancer Cell | 2018

Tumor-Repopulating Cells Induce PD-1 Expression in CD8+ T Cells by Transferring Kynurenine and AhR Activation

Yuying Liu; Xiaoyu Liang; Wenqian Dong; Yi Fang; Jiadi Lv; Tianzhen Zhang; Roland Fiskesund; Jing Xie; Jinyan Liu; Xiaonan Yin; Xun Jin; Degao Chen; Ke Tang; Jingwei Ma; Huafeng Zhang; Jing Yu; Jun Yan; Huaping Liang; Siqi Mo; Feiran Cheng; Yabo Zhou; Haizeng Zhang; Jing Wang; Jingnan Li; Yang Chen; Bing Cui; Zhuowei Hu; Xuetao Cao; F. Xiao-Feng Qin; Bo Huang

Collaboration


Dive into the Wenqian Dong's collaboration.

Top Co-Authors

Avatar

Degao Chen

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Jingwei Ma

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Xiaoyu Liang

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Huafeng Zhang

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Ke Tang

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Jiadi Lv

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Tiantian Ji

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Xuetao Cao

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Zhuowei Hu

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Jing Xie

Peking Union Medical College

View shared research outputs
Researchain Logo
Decentralizing Knowledge