Network


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

Hotspot


Dive into the research topics where Junqi He is active.

Publication


Featured researches published by Junqi He.


Endocrinology | 2011

Na(+)/H(+) Exchanger Regulatory Factor 1 (NHERF1) Is Required for the Estradiol-Dependent Increase of Phosphatase and Tensin Homolog (PTEN) Protein Expression

Longyan Yang; Ying Wang; Peng Chen; Jia Hu; Ying Xiong; Duiping Feng; Hua Liu; Hong Zhang; Hui Yang; Junqi He

Expression of phosphatase and tensin homologue deleted on chromosome 10 (PTEN) can be induced by estrogens at the posttranscriptional level. However, the molecular mechanism of the process is unclear. In this study, we found that the C terminus (CT) of PTEN is indispensable for 17-β-estradiol (E2)-increased PTEN expression. Therefore, we screened for PTEN-CT-associated proteins using a glutathione-S-transferase pull-down approach in combination with mass spectrometry-based proteomic analyses. Our experiments led to the identification of Na(+)/H(+) exchanger regulatory factor 1 (NHERF1) as a major PTEN-CT binding partner. The first postsynaptic density protein-95/Discslarge/zonula occludens-1 homology domain of NHERF1 and the last four amino acids of PTEN were found to be key determinants of this interaction. By associating with PTEN, NHERF1 could enhance PTEN protein expression by retention of PTEN turnover, as demonstrated by NHERF1 overexpression and small interfering RNA-mediated knockdown experiments, respectively. Furthermore, NHERF1 inhibited ubiquitination of the PTEN protein upon competition with binding of PTEN to neural precursor cell expressed, developmentally down-regulated 4, an ubiquitin E3 ligase. E2 strongly induced the expression of NHERF1 and PTEN only in estrogen receptor (ER)-positive cells but not in ER-negative cells. ICI182780, an ER-specific inhibitor, decreased the expression of both NHERF1 and PTEN, and ICI182780 pretreatment also retarded E2-increased PTEN expression in ER-MDA-MB-231 cells. In both ER-MDA-MB-231 and MCF-7 cells, E2 failed to increase PTEN expression when NHERF1 was knocked down. Taken together, these are the first results that present a possible mechanism for E2-increased PTEN expression. In this process, E2 first induces NHERF1 expression by activating the ER. Upon competition with neural precursor cell expressed, developmentally down-regulated 4, NHERF1 then interacts with PTEN to inhibit PTEN degradation, through an ubiquitination-dependent pathway. This in turn leads to the increase of PTEN expression at the protein level.


Amino Acids | 2010

EBP50 exerts tumor suppressor activity by promoting cell apoptosis and retarding extracellular signal-regulated kinase activity

Junfang Zheng; Licui Sun; Hua Liu; Yan Huang; Yang Li; Junqi He

The expression of Ezrin-radixin-moesin-binding phosphoprotein-50 (EBP50) and the intragenic mutation of the ebp50 gene have been reported to correlate with human breast cancer development, but the exact impacts on breast cancer development and its molecular mechanism are not fully understood. In this study, we investigate the potential function of EBP50 through over-expression in the breast cancer cell line, MDA-MB-231, which has low EBP50 protein expression levels. The effects of EBP50 over-expression on cellular proliferation, anchorage-independent growth and apoptosis were examined. In addition, the activity of extracellular signal-regulated kinase (ERK) was also determined. Our results show that a decrease of cellular proliferation and attenuation of colony-forming ability were evident in MDA-MB-231 cells stably transfected with an EBP50 expressing plasmid (EBP-231) when compared with control cells. There was also a statistically significant increase in spontaneous apoptosis in EBP-231 cells accompanied by an attenuation in ERK activity. Altogether, our results suggest that restoring EBP50 expression could suppress breast cancer cell proliferation by promoting cell apoptosis and inhibiting ERK activity, and that EBP50 may be a target for development of diagnostics and therapeutics in breast cancer.


Amino Acids | 2012

EBP50 inhibits EGF-induced breast cancer cell proliferation by blocking EGFR phosphorylation

Wenfang Yao; Duiping Feng; Weihua Bian; Longyan Yang; Yang Li; Zhiyu Yang; Ying Xiong; Junfang Zheng; Renyou Zhai; Junqi He

Ezrin-radixin-moesin-binding phosphoprotein-50 (EBP50) suppresses breast cancer cell proliferation, potentially through its regulatory effect on epidermal growth factor receptor (EGFR) signaling, although the mechanism by which this occurs remains unknown. Thus in our studies, we aimed to determine the effect of EBP50 expression on EGF-induced cell proliferation and activation of EGFR signaling in the breast cancer cell lines, MDA-MB-231 and MCF-7. In MDA-MB-231 cells, which express low levels of EBP50, EBP50 overexpression inhibited EGF-induced cell proliferation, ERK1/2 and AKT phosphorylation. In MCF-7 cells, which express high levels of EBP50, EBP50 knockdown promoted EGF-induced cell proliferation, ERK1/2 and AKT phosphorylation. Knockdown of EBP50 in EBP50-overexpressed MDA-MB-231 cells abrogated the inhibitory effect of EBP50 on EGF-stimulated ERK1/2 phosphorylation and restoration of EBP50 expression in EBP50-knockdown MCF-7 cells rescued the inhibition of EBP50 on EGF-stimulated ERK1/2 phosphorylation, further confirming that the activation of EGF-induced downstream molecules could be specifically inhibited by EBP50 expression. Since EGFR signaling was triggered by EGF ligands via EGFR phosphorylation, we further detected the phosphorylation status of EGFR in the presence or absence of EBP50 expression. Overexpression of EBP50 in MDA-MB-231 cells inhibited EGF-stimulated EGFR phosphorylation, whereas knockdown of EBP50 in MCF-7 cells enhanced EGF-stimulated EGFR phosphorylation. Meanwhile, total expression levels of EGFR were unaffected during EGF stimulation. Taken together, our data shows that EBP50 can suppress EGF-induced proliferation of breast cancer cells by inhibiting EGFR phosphorylation and blocking EGFR downstream signaling in breast cancer cells. These results provide further insight into the molecular mechanism by which EBP50 regulates the development and progression of breast cancer.


Journal of Neurochemistry | 2010

The PDZ domain protein CAL interacts with mGluR5a and modulates receptor expression

Shan Cheng; Jiuqin Zhang; Ping Zhu; Yanmei Ma; Ying Xiong; Licui Sun; Jianguo Xu; Hong Zhang; Junqi He

J. Neurochem. (2010) 112, 588–598.


FEBS Letters | 2008

A novel association of mGluR1a with the PDZ scaffold protein CAL modulates receptor activity

Jiuqin Zhang; Shan Cheng; Ying Xiong; Yanmei Ma; Dali Luo; Andreas Jeromin; Hong Zhang; Junqi He

MINT‐6797987, MINT‐6798009: NHERF‐2 (uniprotkb:Q15599) binds (MI:0407) to mGluR1a (uniprotkb:Q9R0W0) by protein array (MI:0089) MINT‐6798026, MINT‐6798048, MINT‐6798066: mGluR1a (uniprotkb:Q9R0W0) physically interacts (MI:0218) with CAL (uniprotkb:Q9HD26) by pull down (MI:0096) MINT‐6797953, MINT‐6797970: NHERF‐1 (uniprotkb:O14745) binds (MI:0407) to mGluR1a (uniprotkb:Q9R0W0) by protein array (MI:0089) MINT‐6797935: CAL (uniprotkb:Q9HD26) binds (MI:0407) to mGluR1a (uniprotkb:Q9R0W0) by protein array (MI:0089) MINT‐6798084: CAL (uniprotkb:Q9HD26) binds (MI:0407) to mGluR1a (uniprotkb:Q9R0W0) by filter binding (MI:0049) MINT‐6798134: mGluR1a (uniprotkb:Q9R0W0) physically interacts (MI:0218) with CAL (uniprotkb:Q9HD26) by anti tag coimmunoprecipitation (MI:0007) MINT‐6798158: CAL (uniprotkb:B4F775) physically interacts (MI:0218) with mGluR1a (uniprotkb:Q9R0W0) by anti bait coimmunoprecipitation (MI:0006) MINT‐6798233: CAL (uniprotkb:Q9HD26) colocalizes (MI:0403) with mGluR1a (uniprotkb:Q9R0W0) by fluorescence microscopy (MI:0416)


FEBS Letters | 2010

Beta-2 adrenergic receptor mediated ERK activation is regulated by interaction with MAGI-3.

Xiaomei Yang; Junfang Zheng; Ying Xiong; Hui Shen; Licui Sun; Yan Huang; Chaoyuan Sun; Yang Li; Junqi He

MINT‐7716320, MINT‐7716422, MINT‐7716502, MINT‐7716450, MINT‐7716470: beta2AR (uniprotkb:P07550) binds (MI:0407) to MAGI‐3 (uniprotkb:Q5TCQ9) by pull down (MI:0096)


Amino Acids | 2010

The β1-adrenergic receptor mediates extracellular signal-regulated kinase activation via Gαs

Junfang Zheng; Hui Shen; Ying Xiong; Xiaomei Yang; Junqi He

Abstractβ-Adrenergic receptors can activate extracellular signal-regulated kinases (ERKs) via different mechanisms. In this study, we investigated the molecular mechanism of β1-adrenergic receptor (β1AR)-mediated ERK activation in African green monkey kidney COS-7 cells. Treatment of cells with isoproterenol (ISO), a β1AR selective agonist, induced phosphorylation of ERK1/2 in a dose-dependent manner. ISO-stimulated ERK phosphorylation was not influenced by the Gβγ inhibitor, βAR kinase carboxyl terminal (βARKct) or by the Gi inhibitor, pertussis toxin (PTX), but it was clearly abolished via inhibition of protein kinase A (PKA) with H89, or of mitogen-activated protein kinase kinase (MEK1) with PD98059, revealing that the Gαs subunit is involved in ERK regulation through the PKA/MEK1 pathway. We also tested the effect of the adenylate cyclase activator forskolin on ERK activation, and the result was identical to that of ISO stimulation. Moreover, pretreatment with the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor AG1478 or with the Src tyrosine kinase inhibitor PP2 did not affect ERK activation. These observations suggest a mechanism of β1AR-mediated ERK activity that involves the Gαs subunit, but not EGFR or Src tyrosine kinase.


Oncotarget | 2015

MAGI3 negatively regulates Wnt/β-catenin signaling and suppresses malignant phenotypes of glioma cells.

Qian Ma; Ying Yang; Duiping Feng; Shuai Zheng; Ran Meng; Pengyan Fa; Chunjuan Zhao; Hua Liu; Ran Song; Tao Tao; Longyan Yang; Jie Dai; Songlin Wang; Wen Guo Jiang; Junqi He

Gliomas are the most common primary brain malignancies and are associated with a poor prognosis. Here, we showed that the PDZ domain-containing protein membrane-associated guanylate kinase inverted 3 (MAGI3) was downregulated at the both mRNA and protein levels in human glioma samples. MAGI3 inhibited proliferation, migration, and cell cycle progression of glioma cells in its overexpression and knockdown studies. By using GST pull-down and co-immunoprecipitation assays, we found that MAGI3 bound to β-catenin through its PDZ domains and the PDZ-binding motif of β-catenin. MAGI3 overexpression inhibited β-catenin transcriptional activity via its interaction with β-catenin. Consistently, MAGI3 overexpression in glioma cells C6 suppressed expression of β-catenin target genes including Cyclin D1 and Axin2, whereas MAGI3 knockdown in glioma cells U373 and LN229 enhanced their expression. MAGI3 overexpression decreased growth of C6 subcutaneous tumors in mice, and inhibited expression of β-catenin target genes in xenograft tumors. Furthermore, analysis based on the Gene Expression Omnibus (GEO) glioma dataset showed association of MAGI3 expression with overall survival and tumor grade. Finally, we demonstrated negative correlation between MAGI3 expression and activity of Wnt/β-catenin signaling through GSEA of three public glioma datasets and immunohistochemical staining of clinical glioma samples. Taken together, these results identify MAGI3 as a novel tumor suppressor and provide insight into the pathogenesis of glioma.


Molecules and Cells | 2013

EBP50 Phosphorylation by Cdc2/Cyclin B Kinase Affects Actin Cytoskeleton Reorganization and Regulates Functions of Human Breast Cancer Cell Line MDA-MB-231

Chaoyuan Sun; Junfang Zheng; Shan Cheng; Duiping Feng; Junqi He

The actin cytoskeleton plays an important role in cell shape determination, adhesion and cell cycle progression. Ezrinradixin-moesin (ERM)-binding phosphoprotein 50 (EBP50), also known as Na+-H+ exchanger regulatory factor 1 (NHERF1), associates with actin cytoskeleton and is related to cell cycle progression. Its Ser279 and Ser301 residues are phosphorylated by cyclin-dependent kinase 2 (cdc2)/cyclin B during the mitosis phase. However, the biological significance of EBP50 phosphorylation mediated by cdc2/cyclin B is not clear. In the present study, MDA-MB-231 cells with low levels of endogenous EBP50 protein were stably transfected with constructs of EBP50 wild type (WT), phosphodeficient (serine 279 and serine 301 mutated to alanine-S279A/S301A) or phospho-mimetic (serine 279 and serine 301 mutated to aspartic acid-S279D/S301D) mutants. Subsequently, multiple phenotypes of these cells were characterized. Failure of cdc2/cyclin B-mediated EBP50 phosphorylation in cells expressing S279A/S301A (AA cells) significantly increased F-actin content, enhanced the adherence of cells to the extracellular matrix, altered cell morphology and caused defects in cytokinesis, as reflected in the formation of giant cells with heteroploid DNA and multinucleation or giant nuclei. Furthermore, knockdown of EBP50 expression in AA cells rescued cell defects such as the cytokinesis failure and abnormal cell morphology. EBP50 S279A/ S301A had a weaker binding affinity with actin than EBP50 S279D/S301D, which might explain the increase of F-actin content in the AA cells. The present results suggest that cdc2/cyclin B-mediated EBP50 phosphorylation may play a role in the regulation of various cell functions by affecting actin cytoskeleton reorganization.


The FASEB Journal | 2016

NHERF1 regulates actin cytoskeleton organization through modulation of α-actinin-4 stability

Licui Sun; Junfang Zheng; Qiqi Wang; Ran Song; Hua Liu; Ran Meng; Tao Tao; Yang Si; Wenguo Jiang; Junqi He

The actin cytoskeleton is composed of a highly dynamic network of filamentous proteins, yet the molecular mechanism that regulates its organization and remodeling remains elusive. In this study, Na+/H+ exchanger regulatory factor (NHERF)‐1 loss‐of‐function and gain‐of‐function experiments reveal that polymerized actin cytoskeleton (F‐actin) in HeLa cells is disorganized by NHERF1, whereas actin protein expression levels exhibit no detectable change. To elucidate the molecular mechanism underlying actin cytoskeleton disorganization by NHERF1, a combined 2‐dimensional electrophoresis‐matrix‐assisted laser desorption/ionization‐time of flight mass spectrometry approach was used to screen for proteins regulated by NHERF1 in HeLa cells. α‐Actinin‐4, an actin cross‐linking protein, was identified. Glutathione S‐transferase pull‐down and coimmunoprecipitation studies showed the α‐actinin‐4 carboxyl‐terminal region specifically interacted with the NHERF1 postsynaptic density 95/disc‐large/zona occludens‐1 domain. The NHERF1/α‐actinin‐4 interaction increased α‐actinin‐4 ubiquitination and decreased its expression levels, resulting in actin cytoskeleton disassembly. Our study identified α‐actinin‐4 as a novel NHERF1 interaction partner and provided new insights into the regulatory mechanism of the actin cytoskeleton by NHERF1.—Sun, L., Zheng, J., Wang, Q., Song, R., Liu, H., Meng, R., Tao T., Si, Y., Jiang, W., He, J. NHERF1 regulates actin cytoskeleton organization through modulation of α‐actinin‐4 stability. FASEB J. 30, 578‐589 (2016). www.fasebj.org

Collaboration


Dive into the Junqi He's collaboration.

Top Co-Authors

Avatar

Junfang Zheng

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Shan Cheng

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Ying Xiong

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Hua Liu

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Duiping Feng

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Longyan Yang

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Qiqi Wang

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Ran Meng

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Tao Tao

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Songlin Wang

Capital Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge