Network


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

Hotspot


Dive into the research topics where Yingqiu Xie is active.

Publication


Featured researches published by Yingqiu Xie.


Journal of Biological Chemistry | 2008

The 44-kDa Pim-1 Kinase Phosphorylates BCRP/ABCG2 and Thereby Promotes Its Multimerization and Drug-resistant Activity in Human Prostate Cancer Cells

Yingqiu Xie; Kexin Xu; Douglas E. Linn; Xi Yang; Zhiyong Guo; Hermela Shimelis; Takeo Nakanishi; Douglas D. Ross; Hegang Chen; Ladan Fazli; Martin Gleave; Yun Qiu

We previously showed that the 44-kDa serine/threonine kinase Pim-1 (Pim-1L) can protect prostate cancer cells from apoptosis induced by chemotherapeutic drugs (Xie, Y., Xu, K., Dai, B., Guo, Z., Jiang, T., Chen, H., and Qiu, Y. (2006) Oncogene 25, 70–78). To further explore the mechanisms of Pim-1L-mediated resistance to chemotherapeutic drugs in prostate cancer cells, we employed a yeast two-hybrid screening to identify cellular proteins that were associated with Pim-1L, and we found the ABC transporter BCRP/ABCG2 as one of the potential interacting partners of Pim-1L. We also showed that the expression level of Pim-1L and BCRP was up-regulated in mitoxantrone and docetaxel-resistant prostate cancer cell lines. Pim-1L was co-localized with BCRP on the plasma membrane and induced phosphorylation of BCRP at threonine 362. Knocking-down Pim-1L expression in the drug-resistant prostate cancer cells abolished multimer formation of endogenous BCRP and resensitized the resistant cells to chemotherapeutic drugs suggesting that BCRP phosphorylation induced by Pim-1L was essential for its functionality. This is further corroborated by our finding that the plasma membrane localization and drug-resistant activity of BCRP were compromised by T362A mutation. Our data suggest that Pim-1L may protect prostate cancer cells from apoptosis, at least in part, through regulation of transmembrane drug efflux pump. These findings may provide a potential therapeutic approach by disrupting Pim-1 signaling to reverse BCRP-mediated multidrug resistance.


Oncogene | 2006

The 44 kDa Pim-1 kinase directly interacts with tyrosine kinase Etk/BMX and protects human prostate cancer cells from apoptosis induced by chemotherapeutic drugs.

Yingqiu Xie; Kexin Xu; Bojie Dai; Zhiyong Guo; Tianyun Jiang; Hege Chen; Yun Qiu

Protein kinase Pim-1 has been implicated in the development of hematopoietic and prostatic malignancies. Here, we present the evidence that two isoforms, the 44 and 33 kDa Pim-1, are expressed in all human prostate cancer cell lines examined. The subcellular localization of human 44 kDa Pim-1 is primarily on the plasma membrane, while the 33 kDa isoform is present in both the cytosol and nucleus in PCA cells. The 44 kDa Pim-1 contains the proline-rich motif at the N-terminus and directly binds to the SH3 domain of tyrosine kinase Etk. Such interaction leads to the activation of Etk kinase activity possibly by competing with the tumor suppressor p53. This is corroborated by the fact that overexpression of the 44 kDa Pim-1 in prostate cancer cells confers the resistance to chemotherapeutic drugs. Our results suggest that these two isoforms of Pim-1 kinase may regulate distinct substrates and the 44 kDa Pim-1 may play a more prominent role in drug resistance in prostate cancer cells.


Oncogene | 2004

Synergism of cytoplasmic kinases in IL6-induced ligand-independent activation of androgen receptor in prostate cancer cells

Oekyung Kim; Tianyun Jiang; Yingqiu Xie; Zhiyong Guo; Hegang Chen; Yun Qiu

IL6 is a pleiotropic cytokine which has been implicated in ligand-independent activation of androgen receptor in prostate cancer cells. Here, we present the evidence that two cytoplasmic kinases Pim1 and Etk are involved in this process. We showed that Pim1 is expressed in all prostate cancer cell lines examined. Both the expression level and the kinase activity of Pim1 are regulated by IL6 in these cells. Furthermore, we showed that IL6 downstream tyrosine kinase Etk can induce tyrosine phosphorylation of Pim1 which is correlated with its kinase activity. Mutation of the conserved Tyrosine 218 in the activation loop results in reduced kinase activity of Pim1. Interestingly, Etk can also be activated by Pim1 when they are coexpressed in prostate cancer cells, suggesting a possible positive feedback loop between Etk and Pim1. It appears that both Pim1 and Etk are required for IL6-induced activation of androgen receptor-mediated transcription in prostate cancer cells because overexpression of the kinase-deficient form of either Pim1 or Etk dramatically blocks the IL6 effect. Coexpression of the two kinases together but neither one alone is sufficient to activate ARE-containing promoter. Taken together, our data suggest a synergism of Ser/Thr kinase Pim1 and tyrosine kinase Etk in IL6 signaling and provide new insights into ligand-independent activation of androgen receptor in prostate cancer cells.


Genes & Cancer | 2010

A Role for OCT4 in Tumor Initiation of Drug-Resistant Prostate Cancer Cells

Douglas E. Linn; Xi Yang; Feng Sun; Yingqiu Xie; Hege Chen; Hegang Chen; Saranya Chumsri; Angelika M. Burger; Yun Qiu

Drug resistance remains a clinical challenge in cancer treatment due to poor understanding of underlying mechanisms. We have established several drug-resistant prostate cancer cell lines by long-term culture in medium containing chemotherapeutic drugs. These resistant lines displayed a significant increase in side population cells due to overexpression of drug efflux pumps including ABCG2/BCRP and MDR1/Pgp. To uncover potential mechanisms underlying drug resistance, we performed microarray analysis to identify differentially expressed genes in 2 drug-resistant lines. We observed that POU5F1/OCT4, a transcription factor key to regulating pluripotency in embryonic stem cells, was upregulated in drug-resistant lines and accompanied by transcriptional activation of a set of its known target genes. Upregulation of OCT4 in drug-resistant cells was validated by RT-PCR and sequencing of PCR products as well as confirmation by Western blot and specific shRNA knockdown. Analysis of the regulatory region of POU5F1/OCT4 revealed a reduction of methylation in drug-resistant cell lines. Furthermore, these drug-resistant cells exhibited a significant increase in tumorigenicity in vivo. Subcutaneous inoculation of as few as 10 drug-resistant cells could initiate tumor formation in SCID mice, whereas no detectable tumors were observed from the parental line under similar conditions, suggesting that these drug-resistant cells may be enriched for tumor-initiating cells. Knocking down OCT4 expression by specific shRNAs attenuated growth of drug-resistant cells. Our data suggest that OCT4 re-expression in cancer cells may play an important role in carcinogenesis and provide one possible mechanism by which cancer cells acquire/maintain a drug-resistant phenotype.


Molecular Pharmacology | 2010

Pim-1 Kinase Protects P-Glycoprotein from Degradation and Enables Its Glycosylation and Cell Surface Expression

Yingqiu Xie; Mehmet Burcu; Douglas E. Linn; Yun Qiu; Maria R. Baer

The oncogenic serine/threonine kinase Pim-1 phosphorylates and activates the ATP-binding cassette transporter breast cancer resistance protein (ABCG2). The ABC transporter P-glycoprotein (Pgp; ABCB1) also contains a Pim-1 phosphorylation consensus sequence, and we hypothesized that Pim-1 also regulates Pgp. Pgp is exported from the endoplasmic reticulum (ER) as a 150-kDa species that is glycosylated to 170-kDa Pgp, translocates to the cell surface, and mediates drug efflux; alternatively, 150-kDa Pgp is cleaved to a 130-kDa proteolytic product by ER proteases or undergoes ubiquitination and proteasomal degradation. Pim-1 and Pgp interaction was studied in GST pull-down and phosphorylation in in vitro kinase assays. Pim-1 knockdown and inhibition effects on Pgp expression were studied by immunoblotting and flow cytometry and on Pgp stability by immunoblotting after cycloheximide treatment. Pim-1 directly interacted with and phosphorylated Pgp in intact cells and in vitro. Pim-1 knockdown or inhibition decreased cellular and cell surface 170-kDa Pgp, in association with both transient increase in 130-kDa Pgp and increased Pgp ubiquitination and proteasomal degradation. Pim-1 inhibition also decreased expression of 150-kDa Pgp in the presence of the glycosylation inhibitor 2-deoxy-d-glucose. Finally, Pim-1 inhibition sensitized Pgp-overexpressing cells to doxorubicin. Thus, Pim-1 regulates Pgp expression by protecting 150-kDa Pgp from proteolytic and proteasomal degradation and enabling Pgp glycosylation and cell surface translocation and thus Pgp-mediated drug efflux. Pim-1 inhibitors are entering clinical trials and may provide a novel approach to abrogating drug resistance.


Cancer Research | 2006

Tyrosine kinase Etk/BMX is up-regulated in human prostate cancer and its overexpression induces prostate intraepithelial neoplasia in mouse

Bojie Dai; Oekyung Kim; Yingqiu Xie; Zhiyong Guo; Kexin Xu; Bin Wang; Xiangtian Kong; Jonathan Melamed; Hegang Chen; Charles J. Bieberich; Alexander D. Borowsky; Hsing Jien Kung; Guo Wei; Michael C. Ostrowski; Angela Brodie; Yun Qiu

The nonreceptor tyrosine kinase Etk/BMX was originally identified from the human prostate xenograft CWR22. Here, we report that Etk is up-regulated in human prostate tumor specimens surveyed. Knocking down Etk expression by a specific small interfering RNA (siRNA) in prostate cancer cells attenuates cell proliferation, suggesting an essential role of Etk for prostate cancer cell survival and growth. Targeted expression of Etk in mouse prostate epithelium results in pathologic changes resembling human prostatic intraepithelial neoplasia, indicating that up-regulation of Etk may contribute to prostate cancer development. A marked increase of luminal epithelial cell proliferation was observed in the Etk transgenic prostate, which may be attributed in part to the elevated activity of Akt and signal transducers and activators of transcription 3 (STAT3). More interestingly, the expression level of acetyltransferase cyclic AMP-responsive element binding protein-binding protein (CBP) is also increased in the Etk transgenic prostate as well as in a prostate cancer cell line overexpressing Etk, concomitant with elevated histone 3 acetylation at lysine 18 (H3K18Ac). Down-modulation of Etk expression by a specific siRNA leads to a decrease of H3 acetylation in prostate cancer cell lines. Our data suggest that Etk may also modulate chromatin remodeling by regulating the activity of acetyltransferases, such as CBP. Given that Etk may exert its effects in prostate through modulation of multiple signaling pathways altered in human prostate cancer, the Etk transgenic mouse model may be a useful tool for studying the functions of Etk and identification of new molecular markers and drug targets relevant to human diseases.


Endocrine-related Cancer | 2010

Inhibition of the Rho GTPase, Rac1, decreases estrogen receptor levels and is a novel therapeutic strategy in breast cancer

Adena E. Rosenblatt; Maria I. Garcia; Leah S. Lyons; Yingqiu Xie; Carol A. Maiorino; Laurent Désiré; Joyce M. Slingerland; Kerry L. Burnstein

Rac1, a Rho GTPase, modulates diverse cellular processes and is hyperactive in some cancers. Estrogen receptor-alpha (ERα) in concert with intracellular signaling pathways regulates genes associated with cell proliferation, tumor development, and breast cancer cell survival. Therefore, we examined the possibility of Rac1 and ERα crosstalk in breast cancer cells. We found that Rac1 enhanced ERα transcriptional activity in breast cancer cells. Vav3, a Rho guanine nucleotide exchange factor that activates Rac1, was an upstream mediator, and P21/Cdc42/Rac1 activating kinase-1 (Pak-1) was a downstream effector of Rac1 enhancement of ERα activity. These results suggest that Rac1 may prove to be a therapeutic target. To test this hypothesis, we used a small molecule Rac inhibitor, EHT 1864, and found that EHT 1864 inhibited ERα transcriptional activity. Furthermore, EHT 1864 inhibited estrogen-induced cell proliferation in breast cancer cells and decreased tamoxifen-resistant breast cancer cell growth. EHT 1864 decreased activity of the promoter of the ERα gene resulting in down-regulation of ERα mRNA and protein levels. Therefore, ERα down-regulation by EHT 1864 is the likely mechanism of EHT 1864-mediated inhibition of ERα activity and estrogen-stimulated breast cancer cell proliferation. Since ERα plays a critical role in the pathogenesis of breast cancer and the Rac inhibitor EHT 1864 down-regulates ERα expression and breast cancer cell proliferation, further investigation of the therapeutic potential of Rac1 targeting in the treatment of breast cancer is warranted.


Plant Molecular Biology | 2003

Isolation and identification of a super strong plant promoter from cotton leaf curl Multan virus

Yingqiu Xie; Yule Liu; Meng Meng; Lei Chen; Zhen Zhu

The activity of the C1 and the V1 gene promoter of cotton leaf curl Multan virus (CLCuMV) was investigated in transgenic plants with the gus gene as a reporter gene. Quantitative GUS activity analysis of the transgenic plant leaves showed the average activity of the CLCuMV C1 gene promoter was 3- to 5-fold higher than that of the CaMV 35S promoter, with maximal expression being 10-fold higher. CLCuMV V1 gene promoter activity was only about 1/10th that of the CaMV 35S promoter in the absence of trans-activator C2. Histochemical GUS staining of the transgenic plants indicated that the CLCuMV C1 gene promoter was active in leaves, stems, roots and almost all reproductive organs. Functional analysis of promoter 5′-deletion series indicated that promoter activity of a 257 nucleotide fragment (−257 to the transcription initiation site) and a 241 nucleotide fragment (−241 to the transcription initiation site) were 5-fold and 2-fold stronger than that of the full-length CLCuMV C1 promoter respectively. These results demonstrate that the CLCuMV C1 promoter is a super-strong near-constitutive promoter in plants and has great application potential for plant genetic engineering studies.


PLOS ONE | 2013

Pim-1 Kinase Phosphorylates and Stabilizes 130 kDa FLT3 and Promotes Aberrant STAT5 Signaling in Acute Myeloid Leukemia with FLT3 Internal Tandem Duplication

Karthika Natarajan; Yingqiu Xie; Mehmet Burcu; Douglas E. Linn; Yun Qiu; Maria R. Baer

The type III receptor tyrosine kinase fms-like tyrosine kinase 3 (FLT3) is expressed on both normal hematopoietic stem cells and acute myeloid leukemia (AML) cells and regulates their proliferation. Internal tandem duplication (ITD) mutation of FLT3 is present in a third of AML cases, results in constitutive activation and aberrant signaling of FLT3, and is associated with adverse treatment outcomes. While wild-type (WT) FLT3 is predominantly a 150 kDa complex glycosylated cell surface protein, FLT3-ITD is partially retained in the endoplasmic reticulum as a 130 kDa underglycosylated species associated with the chaperones calnexin and heat shock protein (HSP) 90, and mediates aberrant STAT5 signaling, which upregulates the oncogenic serine/threonine kinase Pim-1. FLT3 contains a Pim-1 substrate consensus serine phosphorylation site, and we hypothesized that it might be a Pim-1 substrate. Pim-1 was indeed found to directly interact with and serine-phosphorylate FLT3. Pim-1 inhibition decreased the expression and half-life of 130 kDa FLT3, with partial abrogation by proteasome inhibition, in association with decreased FLT3 binding to calnexin and HSP90, and increased 150 kDa FLT3 expression and half-life, with abrogation by inhibition of glycosylation. These findings were consistent with Pim-1 stabilizing FLT3-ITD as a 130 kDa species associated with calnexin and HSP90 and inhibiting its glycosylation to form the 150 kDa species. Pim-1 knockdown effects were similar. Pim-1 inhibition also decreased phosphorylation of FLT3 at tyrosine 591 and of STAT5, and expression of Pim-1 itself, consistent with inhibition of the FLT3-ITD-STAT5 signaling pathway. Finally, Pim-1 inhibition synergized with FLT3 inhibition in inducing apoptosis of FLT3-ITD cells. This is, to our knowledge, the first demonstration of a role of Pim-1 in a positive feedback loop promoting aberrant signaling in malignant cells.


Journal of Biological Chemistry | 2012

Differential regulation of androgen receptor by PIM-1 kinases via phosphorylation-dependent recruitment of distinct ubiquitin E3 ligases

Douglas E. Linn; Xi Yang; Yingqiu Xie; Alan Alfano; Dhanraj Deshmukh; Xin Wang; Hermela Shimelis; Hegang Chen; Wei Li; Kexin Xu; Mingyuan Chen; Yun Qiu

Background: The androgen receptor (AR) plays a critical role in prostate cancer development and progression. Results: Oncogenic PIM-1 kinases directly interact with AR and induce AR phosphorylation at multiple residues. Conclusion: PIM-1 kinases differentially modulate AR activity via phosphorylation-dependent recruitment of distinct ubiquitin E3 ligases. Significance: Our findings provide new insights into mechanisms by which AR activity may be regulated in prostate cancer cells. Androgen receptor (AR) plays a pivotal role in prostate cancer. Regulation of AR transcriptional activity by post-translational modifications, such as phosphorylation by multiple kinases, is well documented. Here, we report that two PIM-1 kinase isoforms which are up-regulated during prostate cancer progression, namely PIM-1S and PIM-1L, modulate AR stability and transcriptional activity through differentially phosphorylating AR at serine 213 (Ser-213) and threonine 850 (Thr-850). Although both kinases are capable of interacting with and phosphorylating AR at Ser-213, only PIM-1L could phosphorylate Thr-850. We also showed that PIM-1S induced Ser-213 phosphorylation destabilizes AR by recruiting the ubiquitin E3 ligase Mdm2 and promotes AR degradation in a cell cycle-dependent manner, while PIM-1L-induced Thr-850 phosphorylation stabilizes AR by recruiting the ubiquitin E3 ligase RNF6 and promotes AR-mediated transcription under low-androgen conditions. Furthermore, both PIM-1 isoforms could promote prostate cancer cell growth under low-androgen conditions. Our data suggest that these kinases regulate AR stability and transcriptional activity through recruitment of different functional partners in a phosphorylation-dependent manner. As AR turnover has been previously shown to be critical for cell cycle progression in prostate cancer cells, PIM-1 kinase isoforms may promote prostate cancer cell growth, at least in part, through modulating AR activity via distinct mechanisms.

Collaboration


Dive into the Yingqiu Xie's collaboration.

Top Co-Authors

Avatar

Yun Qiu

University of Maryland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qing Yang

Meharry Medical College

View shared research outputs
Top Co-Authors

Avatar

Wenfu Lu

Meharry Medical College

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hegang Chen

University of Maryland

View shared research outputs
Top Co-Authors

Avatar

Kexin Xu

University of Maryland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shenji Liu

Meharry Medical College

View shared research outputs
Top Co-Authors

Avatar

Zhiyong Guo

University of Maryland

View shared research outputs
Researchain Logo
Decentralizing Knowledge