Network


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

Hotspot


Dive into the research topics where Haiyan Xing is active.

Publication


Featured researches published by Haiyan Xing.


Blood | 2014

TBLR1 fuses to retinoid acid receptor α in a variant t(3;17)(q26;q21) translocation of acute promyelocytic leukemia.

Yirui Chen; Shouyun Li; Chunlin Zhou; Chengwen Li; Kun Ru; Qing Rao; Haiyan Xing; Zheng Tian; Kejing Tang; Yingchang Mi; Baohong Wang; Min Wang; Jianxiang Wang

The majority of acute promyelocytic leukemia (APL) cases are characterized by the PML-RARα fusion gene. Although the PML-RARα fusion gene can be detected in >98% of APL cases, RARα is also found to be fused with other partner genes, which are also related to all-trans retinoic acid (ATRA)-dependent transcriptional activity and cell differentiation. In this study, we identified a novel RARα fusion gene, TBLR1-RARα (GenBank KF589333), in a rare case of APL with a t(3;17)(q26;q21),t(7;17)(q11.2;q21) complex chromosomal rearrangement. To our knowledge, TBLR1-RARα is the 10th RARα chimeric gene that has been reported up to now. TBLR1-RARα contained the B-F domains of RARα and exhibited a distinct subcellular localization. It could form homodimers and also heterodimers with retinoid X receptor α. As a result, TBLR1-RARα exhibited diminished transcriptional activity by recruitment of more transcriptional corepressors compared with RARα. In the presence of pharmacologic doses of ATRA, TBLR1-RARα could be degraded, and its homodimerization was abrogated. Moreover, when treated with ATRA, TBLR1-RARα could mediate the dissociation and degradation of transcriptional corepressors, consequent transactivation of RARα target genes, and cell differentiation induction in a dose- and time-dependent manner.


Biochemical and Biophysical Research Communications | 2012

Metformin induces differentiation in acute promyelocytic leukemia by activating the MEK/ERK signaling pathway

Lei Huai; Cuicui Wang; Cuiping Zhang; Qihui Li; Yirui Chen; Yujiao Jia; Yan Li; Haiyan Xing; Zheng Tian; Qing Rao; Min Wang; Jianxiang Wang

Recent studies have shown that metformin, a widely used antidiabetic agent, may reduce the risk of cancer development. In this study, we investigated the antitumoral effect of metformin on both acute myeloid leukemia (AML) and acute promyelocytic leukemia (APL) cells. Metformin induced apoptosis with partial differentiation in an APL cell line, NB4, but only displayed a proapoptotic effect on several non-M3 AML cell lines. Further analysis revealed that a strong synergistic effect existed between metformin and all-trans retinoic acid (ATRA) during APL cell maturation and that metformin induced the hyperphosphorylation of extracellular signal-regulated kinase (ERK) in APL cells. U0126, a specific MEK/ERK activation inhibitor, abrogated metformin-induced differentiation. Finally, we found that metformin induced the degradation of the oncoproteins PML-RARα and c-Myc and activated caspase-3. In conclusion, these results suggest that metformin treatment may contribute to the enhancement of ATRA-induced differentiation in APL, which may deepen the understanding of APL maturation and thus provide insight for new therapy strategies.


Leukemia Research | 2009

siRNA-mediated down-regulation of iASPP promotes apoptosis induced by etoposide and daunorubicin in leukemia cells expressing wild-type p53

Hang Liu; Min Wang; Shiyong Diao; Qing Rao; Xinwei Zhang; Haiyan Xing; Jianxiang Wang

Oncoprotein inhibitory member of the ASPP family (iASPP) is a key inhibitor of tumor suppressor p53. Our previous study revealed that the expression of iASPP in acute leukemia (AL) patients was higher than that of normal control which implied that iASPP might play an important role in the pathogenesis and/or disease progression of AL. In this study, the iASPP expression was blocked by RNA interference (RNAi) in two leukemic cell lines, Nalm6 and K562, to explore the effects of iASPP on leukemia cells. The results indicated that down-regulation of endogenous iASPP increased p53-dependent apoptosis of leukemia cells. Thus, iASPP could be a molecular target in leukemia therapy.


Biochemical and Biophysical Research Communications | 2009

Overexpression of Rac1 in leukemia patients and its role in leukemia cell migration and growth

Jiying Wang; Qing Rao; Min Wang; Hui Wei; Haiyan Xing; Hang Liu; Yanzhong Wang; Kejing Tang; Leiwen Peng; Zheng Tian; Jianxiang Wang

Rac1 belongs to the Rho family that act as critical mediators of signaling pathways controlling cell migration and proliferation and contributes to the interactions of hematopoietic stem cells with their microenvironment. Alteration of Rac1 might result in unbalanced interactions and ultimately lead to leukemogenesis. In this study, we analyze the expression of Rac1 protein in leukemia patients and determine its role in the abnormal behaviours of leukemic cells. Rac1 protein is overexpressed in primary acute myeloid leukemia cells as compared to normal bone marrow mononuclear cells. siRNA-mediated silencing of Rac1 in leukemia cell lines induced inhibition of cell migration, proliferation, and colony formation. Additionally, blocking Rac1 activity by an inhibitor of Rac1-GTPase, NSC23766, suppressed cell migration and growth. We conclude that overexpression of Rac1 contributes to the accelerated migration and high proliferation potential of leukemia cells, which could be implicated in leukemia development and progression.


Molecular Oncology | 2013

Activation of Rac1 GTPase promotes leukemia cell chemotherapy resistance, quiescence and niche interaction

Jiying Wang; Pei Yu; Shuying Chen; Haiyan Xing; Yirui Chen; Min Wang; Kejing Tang; Zheng Tian; Qing Rao; Jianxiang Wang

Leukemia stem cells (LSCs) reside in bone marrow niche and receive important signals from the microenvironment that support self‐renewal, maintain quiescence and endow LSC with the ability of chemotherapy resistance. Rac1 belongs to the small GTP‐binding protein superfamily and is implicated in the interactions of hematopoietic progenitors and bone marrow niche. Our previous studies have shown that Rac1 is over‐expressed in leukemia patients and activation of Rac1 GTPase is closely associated with the efficient migration of leukemia cells. However, the potential functions for Rac1 GTPase in LSCs behaviors and in the residence of leukemia cells in niche remain unknown. In this study, by forced expression of a dominant‐negative form of Rac1 GTPase in a CD34+ myeloid leukemia cell line, as well as bone marrow cells from leukemia patients, we show that inactivation of Rac1 GTPase causes impaired migration and enhances chemotherapeutic sensitivity. Inactivation of Rac1 in leukemia cells also lead to a reduction in the frequency of cells in quiescent state and inhibition of homing to bone marrow niche. Gene expression analysis shows that inactivation of Rac1 down‐regulates the expression of several cell intrinsic cell cycle inhibitors such as p21, p27, and p57, as well as the extrinsic molecules that mediated the interaction of LSC with osteoblastic niche. Furthermore, we show that Rac1 mediated the localization in niche is further attributed to the maintenance of quiescence. Our results provide evidence for the critical role of Rac1 GTPase in leukemia cell chemotherapy resistance, quiescence maintenance and the interaction with bone marrow microenvironment.


Leukemia Research | 2014

N-Cadherin and Tie2 positive CD34 + CD38 − CD123 + leukemic stem cell populations can develop acute myeloid leukemia more effectively in NOD/SCID mice

Shaowei Qiu; Yujiao Jia; Haiyan Xing; Tengteng Yu; Jing Yu; Pei Yu; Kejing Tang; Zheng Tian; Huijun Wang; Yingchang Mi; Qing Rao; Min Wang; Jianxiang Wang

Emerging studies suggest that the population of malignant cells found in human acute myelogenous leukemia (AML) arises from a rare population of leukemic stem cells (LSCs). A lot of investigators have reported the identification of cell surface markers, such as CD123. Here, we report the identification of N-cadherin and Tie2 as LSCs markers. Inoculation of CD34(+)CD38(-)CD123(+)N-cadherin(+) and CD34(+)CD38(-)CD123(+) Tie2(+) population can induce leukemia in NOD/SCID mice. The leukemic blast cells from the primary leukemic mice could also induce leukemia in the secondary transplantation. These findings suggested that N-cadherin and Tie2 were the important markers that can assist in leukemia development.


Biochemical and Biophysical Research Communications | 2014

RPS27a promotes proliferation, regulates cell cycle progression and inhibits apoptosis of leukemia cells

Houcai Wang; Jing Yu; Lixia Zhang; Yuanyuan Xiong; Shuying Chen; Haiyan Xing; Zheng Tian; Kejing Tang; Hui Wei; Qing Rao; Min Wang; Jianxiang Wang

Ribosomal protein S27a (RPS27a) could perform extra-ribosomal functions besides imparting a role in ribosome biogenesis and post-translational modifications of proteins. The high expression level of RPS27a was reported in solid tumors, and we found that the expression level of RPS27a was up-regulated in advanced-phase chronic myeloid leukemia (CML) and acute leukemia (AL) patients. In this study, we explored the function of RPS27a in leukemia cells by using CML cell line K562 cells and its imatinib resistant cell line K562/G01 cells. It was observed that the expression level of RPS27a was high in K562 cells and even higher in K562/G01 cells. Further analysis revealed that RPS27a knockdown by shRNA in both K562 and K562G01 cells inhibited the cell viability, induced cell cycle arrest at S and G2/M phases and increased cell apoptosis induced by imatinib. Combination of shRNA with imatinib treatment could lead to more cleaved PARP and cleaved caspase-3 expression in RPS27a knockdown cells. Further, it was found that phospho-ERK(p-ERK) and BCL-2 were down-regulated and P21 up-regulated in RPS27a knockdown cells. In conclusion, RPS27a promotes proliferation, regulates cell cycle progression and inhibits apoptosis of leukemia cells. It appears that drugs targeting RPS27a combining with tyrosine kinase inhibitor (TKI) might represent a novel therapy strategy in TKI resistant CML patients.


The FASEB Journal | 2014

Oncogene iASPP enhances self-renewal of hematopoietic stem cells and facilitates their resistance to chemotherapy and irradiation

Yujiao Jia; Leiwen Peng; Qing Rao; Haiyan Xing; Lei Huai; Pei Yu; Yirui Chen; Cuicui Wang; Min Wang; Yingchang Mi; Jianxiang Wang

iASPP is a member of the apoptosis‐stimulating proteins of p53 (ASPP) family and negatively regulates the apoptotic function of p53. In a hematopoietic system, overexpression of iASPP results in blockage of apoptosis, which may play a role in regulating hematopoietic stem cell (HSC) numbers. To address this, we first analyzed the expression of iASPP in patients with acute leukemia (AL) and found it was highly expressed in patients with AL. We further established a transgenic mouse model in which human iASPP was specifically expressed in hematopoietic cells. Overexpression of iASPP led to an increase in the proportion of long‐term HSCs, short‐term HSCs, multipotent progenitors, and common myeloid progenitor. HSCs from iASPP transgenic mice had an advantage in long‐term reconstitution potential. In addition, the hematopoietic cells from iASPP transgenic mice exhibited a significantly lower level of p53 dependent apoptosis. After irradiation damage, hematopoietic cells of iASPP transgenic mice had a higher level of γ‐H2AX expression, which lasted for a longer time. These results provide the first evidence that the iASPP can increase HSC populations and reconstitution capacity. Interestingly, in response to cell damage stimuli, hematopoietic cells can be protected against apoptosis by iASPP; meanwhile these apoptosis‐resistant cells would have more mutation accumulation, which might be the potential risk for malignant transformation.—Jia, Y., Peng, L., Rao, Q., Xing, H., Huai, L., Yu, P., Chen, Y., Wang, C., Wang, M., Mi, Y., Wang, J. Oncogene iASPP enhances self‐renewal of hematopoietic stem cells and facilitates their resistance to chemotherapy and irradiation. FASEB J. 28, 2816–2827 (2014). www.fasebj.org


Cell Biology International | 2011

Low-expression of E-cadherin in leukaemia cells causes loss of homophilic adhesion and promotes cell growth.

Qing Rao; Ji‑Ying Wang; Jihong Meng; Kejing Tang; Yanzhong Wang; Min Wang; Haiyan Xing; Zheng Tian; Jianxiang Wang

E‐cadherin (epithelial cadherin) belongs to the calcium‐dependent adhesion molecule superfamily and is implicated in the interactions of haematopoietic progenitors and bone marrow stromal cells. Adhesion capacity to bone marrow stroma was impaired for leukaemia cells, suggesting that a breakdown of adhesive mechanisms governed by an adhesion molecule may exist in leukaemic microenvironment. We previously found that E‐cadherin was low expressed in primary acute leukaemia cells compared with normal bone marrow mononuclear cells. In this study, we investigate the functional importance of low E‐cadherin expression in leukaemia cell behaviours and investigate its effects in the abnormal interaction of leukaemic cells with stromal cells. After expression of E‐cadherin was restored by a demethylating agent in leukaemia cells, E‐cadherin‐specific adhesion was enhanced. Additionally, siRNA (small interfering RNA)‐mediated silencing of E‐cadherin in Raji cells resulted in a reduction of cell homophilic adhesion and enhancement of cell proliferation and colony formation. These results suggest that low expression of E‐cadherin contributes to the vigorous growth and transforming ability of leukaemic cells.


Oncotarget | 2016

Construction of a new anti-CD19 chimeric antigen receptor and the anti-leukemia function study of the transduced T cells

Na An; Zhongfei Tao; Saisai Li; Haiyan Xing; Kejing Tang; Zheng Tian; Qing Rao; Min Wang; Jianxiang Wang

Chimeric antigen receptor (CAR) transduced T cells have been used to efficiently kill the target tumor cells depending on the single chain variable fragment (scFv) against the specific tumor associated antigen. Here we show the high specific cytotoxicity of the CAR-T cells with very low effector to target cell (E:T) ratio owing to the CD19-scFv, which was constructed in our laboratory and proved to be highly effective in our previous study. Four plasmids containing three generation of CAR were constructed by cloning the CD19-CAR fragment into the lentiviral vector pCDH. CD3 positive T cells were successfully transduced and the CAR protein expression was confirmed by flow cytometry and Western blot. When cocultured with CD19 positive leukemia cell line Nalm-6 cells, CAR-T cells showed specific cytotoxicity: the percentage of target cells decreased to 0 in 24 hours; IL-2, IFN-γ and TNF-α produced in cocultured supernatants increased obviously; and the cytotoxicity reached more than 80%, still remarkable even when the E:T ratio was as low as 1:4. Dynamic change of cell interaction between CAR-T and leukemia cells was visually tracked by using living cells workstation for the first time. A NOD/SCID B-ALL murine model was established using Nalm-6 cells inoculation with a morbidity rate of 100%, and the survival time was prolonged statistically with CAR-T cell treatment. These data demonstrate that the CAR-T cells we prepared could be a promising treatment strategy for CD19 positive tumor diseases.

Collaboration


Dive into the Haiyan Xing's collaboration.

Top Co-Authors

Avatar

Min Wang

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Qing Rao

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Jianxiang Wang

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Kejing Tang

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Zheng Tian

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Yingchang Mi

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Shuang Liu

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Jing Yu

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Pei Yu

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Shouyun Li

Peking Union Medical College

View shared research outputs
Researchain Logo
Decentralizing Knowledge