Network


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

Hotspot


Dive into the research topics where Xueyan Duan is active.

Publication


Featured researches published by Xueyan Duan.


Cell | 2016

PPM1A Functions as a Smad Phosphatase to Terminate TGFβ Signaling

Xia Lin; Xueyan Duan; Yao Yun Liang; Ying Su; Katharine H. Wrighton; Jianyin Long; Min Hu; Candi M. Davis; Jinrong Wang; F. Charles Brunicardi; Yigong Shi; Ye-Guang Chen; Anming Meng; Xin-Hua Feng

TGFbeta signaling controls diverse normal developmental processes and pathogenesis of diseases including cancer and autoimmune and fibrotic diseases. TGFbeta responses are generally mediated through transcriptional functions of Smads. A key step in TGFbeta signaling is ligand-induced phosphorylation of receptor-activated Smads (R-Smads) catalyzed by the TGFbeta type I receptor kinase. However, the potential of Smad dephosphorylation as a regulatory mechanism of TGFbeta signaling and the identity of Smad-specific phosphatases remain elusive. Using a functional genomic approach, we have identified PPM1A/PP2Calpha as a bona fide Smad phosphatase. PPM1A dephosphorylates and promotes nuclear export of TGFbeta-activated Smad2/3. Ectopic expression of PPM1A abolishes TGFbeta-induced antiproliferative and transcriptional responses, whereas depletion of PPM1A enhances TGFbeta signaling in mammalian cells. Smad-antagonizing activity of PPM1A is also observed during Nodal-dependent early embryogenesis in zebrafish. This work demonstrates that PPM1A/PP2Calpha, through dephosphorylation of Smad2/3, plays a critical role in terminating TGFbeta signaling.


Journal of Biological Chemistry | 2006

Protein Serine/Threonine Phosphatase PPM1A Dephosphorylates Smad1 in the Bone Morphogenetic Protein Signaling Pathway

Xueyan Duan; Yao-Yun Liang; Xin-Hua Feng; Xia Lin

Bone morphogenetic proteins (BMPs) are secreted polypeptides belonging to the transforming growth factor-β (TGF-β) superfamily that activates a broad range of biological responses in the metazoan organism. The BMP-initiated signaling pathway is under tight control by processes including regulation of the ligands, the receptors, and the key downstream intracellular effector Smads. A critical point of control in BMP signaling is the phosphorylation of Smad1, Smad5, and Smad8 in their C-terminal SXS motif. Although such phosphorylation, which is mediated by the type I BMP receptor kinases in response to BMP stimulation, is well characterized, biochemical mechanisms underlying Smad dephosphorylation remain to be elucidated. In this study, we have found that PPM1A, a metal ion-dependent protein serine/threonine phosphatase, physically interacts with and dephosphorylates Smad1 both in vitro and in vivo. Functionally, overexpression of PPM1A abolishes BMP-induced transcriptional responses, whereas RNA interference-mediated knockdown of PPM1A enhances BMP signaling. Collectively, our study suggests that PPM1A plays an important role in controlling BMP signaling through catalyzing Smad dephosphorylation.


Journal of Biological Chemistry | 2014

C-terminal Domain (CTD) Small Phosphatase-like 2 Modulates the Canonical Bone Morphogenetic Protein (BMP) Signaling and Mesenchymal Differentiation via Smad Dephosphorylation

Yulan Zhao; Mu Xiao; Baoguo Sun; Zhengmao Zhang; Tao Shen; Xueyan Duan; Paul B. Yu; Xin-Hua Feng; Xia Lin

Background: Dephosphorylation of R-Smads in the nucleus shuts off TGF-β superfamily signaling. Results: SCP4 specifically dephosphorylates BMP-activated Smad1/5/8, but not TGF-β-activated Smad2/3, and ectopic expression of SCP4 inhibits BMP signaling, whereas SCP4 depletion enhances BMP signaling. Conclusion: SCP4 is a nuclear phosphatase terminating BMP signaling. Significance: Identification of SCP4 may suggest its physiological functions in BMP-induced cellular processes and relevant diseases. The bone morphogenetic protein (BMP) signaling pathway regulates a wide range of cellular responses in metazoans. A key step in the canonical BMP signaling is the phosphorylation and activation of transcription factors Smad1, Smad5, and Smad8 (collectively Smad1/5/8) by the type I BMP receptors. We previously identified PPM1A as a phosphatase toward dephosphorylation of all receptor-regulated Smads (R-Smads), including Smad1/5/8. Here we report another nuclear phosphatase named SCP4/CTDSPL2, belonging to the FCP/SCP family, as a novel Smad phosphatase in the nucleus. SCP4 physically interacts with and specifically dephosphorylates Smad1/5/8, and as a result attenuates BMP-induced transcriptional responses. Knockdown of SCP4 in multipotent mesenchymal C2C12 cells leads to increased expression of BMP target genes and consequently promotes BMP-induced osteogenic differentiation. Collectively, our results demonstrate that SCP4, as a Smad phosphatase, plays a critical role in BMP-induced signaling and cellular functions.


Cancer Research | 2014

SCP Phosphatases Suppress Renal Cell Carcinoma by Stabilizing PML and Inhibiting mTOR/HIF Signaling

Yu-Ching Lin; Li Ting Lu; Hsin Yi Chen; Xueyan Duan; Xia Lin; Xin-Hua Feng; Ming Jer Tang; Ruey-Hwa Chen

The tumor-suppressor protein promyelocytic leukemia (PML) is aberrantly degraded in multiple types of human cancers through mechanisms that are incompletely understood. Here, we show that the phosphatase SCP1 and its isoforms SCP2/3 dephosphorylate PML at S518, thereby blocking PML ubiquitination and degradation mediated by the prolyl isomerase Pin1 and the ubiquitin ligase KLHL20. Clinically, SCP1 and SCP3 are downregulated in clear cell renal cell carcinoma (ccRCC) and these events correlated with PMLS518 phosphorylation, PML turnover, and high-grade tumors. Restoring SCP1-mediated PML stabilization not only inhibited malignant features of ccRCC, including proliferation, migration, invasion, tumor growth, and tumor angiogenesis, but also suppressed the mTOR-HIF pathway. Furthermore, blocking PML degradation in ccRCC by SCP1 overexpression or Pin1 inhibition enhanced the tumor-suppressive effects of the mTOR inhibitor temsirolimus. Taken together, our results define a novel pathway of PML degradation in ccRCC that involves SCP downregulation, revealing contributions of this pathway to ccRCC progression and offering a mechanistic rationale for combination therapies that jointly target PML degradation and mTOR inhibition for ccRCC treatment.


Cell Research | 2014

Specific control of BMP signaling and mesenchymal differentiation by cytoplasmic phosphatase PPM1H

Tao Shen; Chuang Sun; Zhengmao Zhang; Ningyi Xu; Xueyan Duan; Xin-Hua Feng; Xia Lin

Bone morphogenetic proteins (BMPs) belong to the TGF-β superfamily of structurally related signaling proteins that regulate a wide array of cellular functions. The key step in BMP signal transduction is the BMP receptor-mediated phosphorylation of transcription factors Smad1, 5, and 8 (collectively Smad1/5/8), which leads to the subsequent activation of BMP-induced gene transcription in the nucleus. In this study, we describe the identification and characterization of PPM1H as a novel cytoplasm-localized Smad1/5/8-specific phosphatase. PPM1H directly interacts with Smad1/5/8 through its Smad-binding domain, and dephosphorylates phospho-Smad1/5/8 (P-Smad1/5/8) in the cytoplasm. Ectopic expression of PPM1H attenuates BMP signaling, whereas loss of PPM1H activity or expression greatly enhances BMP-dependent gene regulation and mesenchymal differentiation. In conclusion, this study suggests that PPM1H acts as a gatekeeper to prevent excessive BMP signaling through dephosphorylation and subsequent nuclear exclusion of P-Smad1/5/8 proteins.


Methods of Molecular Biology | 2010

Coupling of dephosphorylation and nuclear export of Smads in TGF-β signaling

Fangyan Dai; Xueyan Duan; Yao Yun Liang; Xia Lin; Xin-Hua Feng

In eukaryotes, regulation of signaling mediators/effectors in the nucleus is one of the principal mechanisms that govern duration and strength of signaling. Smads are a family of structurally related intracellular proteins that serve as signaling effectors for transforming growth factor beta (TGF-beta) and TGF-beta-related proteins. Accumulating evidence demonstrates that Smads possess intrinsic nucleocytoplasmic shuttling capacity, which enables them to transmit TGF-beta signals from cell membrane to nucleus. We recently identified two important steps in the termination of nuclear Smad signaling. The first step is initiated by a serine/threonine phosphatase PPM1A that dephosphorylates Smad2/3 in the nucleus, thereby shutting down signaling capacity of phosphorylated Smad2/3. The second step involves nuclear export of dephosphorylated Smad2/3 with the aid of nuclear protein RanBP3 to terminate Smad signaling. This chapter introduces methods for examining nuclear export of Smad2/3 in TGF-beta signaling.


Journal of Surgical Research | 2011

Control Of Aneuploidy By Novel Histone H3 Phosphatase H3P

X. Lin; Zhengmao Zhang; Xueyan Duan; Xin-Hua Feng


Journal of Surgical Research | 2010

Role of SMAD Phosphatase SMP4 in Organ Development

Xin-Hua Feng; B. Chen; Zhengmao Zhang; Xueyan Duan; M. Chu; X. Lin


Journal of Surgical Research | 2010

Identification of POA as a Novel Phosphatase of Akt Kinase in a Genome-Wide Functional Genomic Screen

Xueyan Duan; Tao Shen; X. Gao; J. Zhang; Xin-Hua Feng; X. Lin


Journal of Surgical Research | 2009

45: Cytoplasmic Phosphatase PPM1B Controls Outputs of TGFβ Signal Transduction

Xueyan Duan; Yao-Yun Liang; F.C. Brunicardi; X. Lin

Collaboration


Dive into the Xueyan Duan's collaboration.

Top Co-Authors

Avatar

Xin-Hua Feng

Life Sciences Institute

View shared research outputs
Top Co-Authors

Avatar

X. Lin

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Xia Lin

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Tao Shen

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Zhengmao Zhang

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Yao-Yun Liang

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

B. Chen

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

F.C. Brunicardi

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jinrong Wang

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge