Network


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

Hotspot


Dive into the research topics where Shun-Chang Wang is active.

Publication


Featured researches published by Shun-Chang Wang.


Molecular and Cellular Biology | 2012

Intermolecular Binding between TIFA-FHA and TIFA-pT Mediates Tumor Necrosis Factor Alpha Stimulation and NF-κB Activation

Chia-Chi Flora Huang; Jui-Hung Weng; Tong-You Wade Wei; Pei-Yu Gabriel Wu; Pang-Hung Hsu; Yu-Hou Chen; Shun-Chang Wang; Dongyan Qin; Chin Chun Hung; Shui-Tsung Chen; Andrew H.-J. Wang; John Y.-J. Shyy; Ming-Daw Tsai

ABSTRACT The forkhead-associated (FHA) domain recognizes phosphothreonine (pT) with high specificity and functional diversity. TIFA (TRAF-interacting protein with an FHA domain) is the smallest FHA-containing human protein. Its overexpression was previously suggested to provoke NF-κB activation, yet its exact roles in this signaling pathway and the underlying molecular mechanism remain unclear. Here we identify a novel threonine phosphorylation site on TIFA and show that this phosphorylated threonine (pT) binds with the FHA domain of TIFA, leading to TIFA oligomerization and TIFA-mediated NF-κB activation. Detailed analysis indicated that unphosphorylated TIFA exists as an intrinsic dimer and that the FHA-pT9 binding occurs between different dimers of TIFA. In addition, silencing of endogenous TIFA resulted in attenuation of tumor necrosis factor alpha (TNF-α)-mediated downstream signaling. We therefore propose that the TIFA FHA-pT9 binding provides a previously unidentified link between TNF-α stimulation and NF-κB activation. The intermolecular FHA-pT9 binding between dimers also represents a new mechanism for the FHA domain.


PLOS ONE | 2013

Phosphorylation of mRNA decapping protein Dcp1a by the ERK signaling pathway during early differentiation of 3T3-L1 preadipocytes.

Pei-Yu Chiang; Yu-Fang Shen; Yu-Lun Su; Ching-Han Kao; Nien-Yi Lin; Pang-Hung Hsu; Ming-Daw Tsai; Shun-Chang Wang; Geen-Dong Chang; Sheng-Chung Lee; Ching-Jin Chang

Background Turnover of mRNA is a critical step in the regulation of gene expression, and an important step in mRNA decay is removal of the 5′ cap. We previously demonstrated that the expression of some immediate early gene mRNAs is controlled by RNA stability during early differentiation of 3T3-L1 preadipocytes. Methodology/Principal Findings Here we show that the mouse decapping protein Dcp1a is phosphorylated via the ERK signaling pathway during early differentiation of preadipocytes. Mass spectrometry analysis and site-directed mutagenesis combined with a kinase assay identified ERK pathway–mediated dual phosphorylation at Ser 315 and Ser 319 of Dcp1a. To understand the functional effects of Dcp1a phosphorylation, we examined protein-protein interactions between Dcp1a and other decapping components with co-immunoprecipitation. Dcp1a interacted with Ddx6 and Edc3 through its proline-rich C-terminal extension, whereas the conserved EVH1 (enabled vasodilator-stimulated protein homology 1) domain in the N terminus of Dcp1a showed a stronger interaction with Dcp2. Once ERK signaling was activated, the interaction between Dcp1a and Ddx6, Edc3, or Edc4 was not affected by Dcp1a phosphorylation. Phosphorylated Dcp1a did, however, enhanced interaction with Dcp2. Protein complexes immunoprecipitated with the recombinant phosphomimetic Dcp1a(S315D/S319D) mutant contained more Dcp2 than did those immunoprecipitated with the nonphosphorylated Dcp1a(S315A/S319A) mutant. In addition, Dcp1a associated with AU-rich element (ARE)-containing mRNAs such as MAPK phosphatase-1 (MKP-1), whose mRNA stability was analyzed under the overexpression of Dcp1a constructs in the Dcp1a knockdown 3T3-L1 cells. Conclusions/Significance Our findings suggest that ERK-phosphorylated Dcp1a enhances its interaction with the decapping enzyme Dcp2 during early differentiation of 3T3-L1 cells.


Journal of Inflammation | 2015

Functional regulation of Zfp36l1 and Zfp36l2 in response to lipopolysaccharide in mouse RAW264.7 macrophages

Kuan-Ting Wang; Hsin-Hui Wang; Yan-Yun Wu; Yu-Lun Su; Pei-Yu Chiang; Nien-Yi Lin; Shun-Chang Wang; Geen-Dong Chang; Ching-Jin Chang

BackgroundThe tristetraprolin (TTP) family of mRNA-binding proteins contains three major members, Ttp, Zfp36l1, and Zfp36l2. Ttp down-regulates the stability of AU-rich element–containing mRNAs and functions as an anti-inflammation regulator.MethodsTo examine whether other TTP family proteins also play roles in the inflammatory response, their expression profiles and the possible mRNA targets were determined in the knockdown cells.ResultsTtp mRNA and protein were highly induced by lipopolysaccharide (LPS), whereas Zfp36l1 and Zfp36l2 mRNAs were down-regulated and their proteins were phosphorylated during early lipopolysaccharide stimulation. Biochemical and functional analyses exhibited that the decrease of Zfp36l2 mRNA was cross-regulated by Ttp. Knockdown of Zfp36l1 and Zfp36l2 increased the basal level of Mkp-1 mRNAs by prolonging its half-life. Increasing the expression of Mkp-1 inhibited the activation of p38 MAPK under lipopolysaccharide stimulation and down-regulated Tnfα, and Ttp mRNA. In addition, hyper-phosphorylation of Zfp36l1 might stabilize Mkp-1 expression by forming a complex with the adapter protein 14-3-3 and decreasing the interaction with deadenylase Caf1a.ConclusionsOur findings imply that the expression and phosphorylation of Zfp36l1 and Zfp36l2 may modulate the basal level of Mkp-1 mRNA to control p38 MAPK activity during lipopolysaccharide stimulation, which would affect the inflammatory mediators production. Zfp36l1 and Zfp36l2 are important regulators of the innate immune response.


International Journal of Biological Sciences | 2012

Differential Expression and Functional Analysis of the Tristetraprolin Family during Early Differentiation of 3T3-L1 Preadipocytes

Nien-Yi Lin; Tzi-Yang Lin; Wen-Hsuan Yang; Shun-Chang Wang; Kuan-Ting Wang; Yu-Lun Su; Yu-Wun Jiang; Geen-Dong Chang; Ching-Jin Chang

The tristetraprolin (TTP) family comprises zinc finger-containing AU-rich element (ARE)-binding proteins consisting of three major members: TTP, ZFP36L1, and ZFP36L2. The present study generated specific antibodies against each TTP member to evaluate its expression during differentiation of 3T3-L1 preadipocytes. In contrast to the inducible expression of TTP, results indicated constitutive expression of ZFP36L1 and ZFP36L2 in 3T3-L1 preadipocytes and their phosphorylation in response to differentiation signals. Physical RNA pull-down and functional luciferase assays revealed that ZFP36L1 and ZFP36L2 bound to the 3′ untranslated region (UTR) of MAPK phosphatase-1 (MKP-1) mRNA and downregulated Mkp-1 3′UTR-mediated luciferase activity. Mkp-1 is an immediate early gene for which the mRNA is transiently expressed in response to differentiation signals. The half-life of Mkp-1 mRNA was longer at 30 min of induction than at 1 h and 2 h of induction. Knockdown of TTP or ZFP36L2 increased the Mkp-1 mRNA half-life at 1 h of induction. Knockdown of ZFP36L1, but not ZFP36L2, increased Mkp-1 mRNA basal levels via mRNA stabilization and downregulated ERK activation. Differentiation induced phosphorylation of ZFP36L1 through ERK and AKT signals. Phosphorylated ZFP36L1 then interacted with 14-3-3, which might decrease its mRNA destabilizing activity. Inhibition of adipogenesis also occurred in ZFP36L1 and TTP knockdown cells. The findings indicate that the differential expression of TTP family members regulates immediate early gene expression and modulates adipogenesis.


International Journal of Biological Sciences | 2012

Drosophila eyes absent is a Novel mRNA Target of the Tristetraprolin (TTP) Protein DTIS11

Po-An Yeh; Wen-Hsuan Yang; Pei-Yu Chiang; Shun-Chang Wang; Mau-Sun Chang; Ching-Jin Chang

The Tristetraprolin (TTP) protein family includes four mammalian members (TTP, TIS11b, TIS11d, and ZFP36L3), but only one in Drosophila melanogaster (DTIS11). These proteins bind target mRNAs with AU-rich elements (AREs) via two C3H zinc finger domains and destabilize the mRNAs. We found that overexpression of mouse TIS11b or DTIS11 in the Drosophila retina dramatically reduced eye size, similar to the phenotype of eyes absent (eya) mutants. The eya transcript is one of many ARE-containing mRNAs in Drosophila. We showed that TIS11b reduced levels of eya mRNA in vivo. In addition, overexpression of Eya rescued the TIS11b overexpression phenotype. RNA pull-down and luciferase reporter analyses demonstrated that the DTIS11 RNA-binding domain is required for DTIS11 to bind the eya 3′ UTR and reduce levels of eya mRNA. Moreover, ectopic expression of DTIS11 in Drosophila S2 cells decreased levels of eya mRNA and reduced cell viability. Consistent with these results, TTP proteins overexpressed in MCF7 human breast cancer cells were associated with eya homologue 2 (EYA2) mRNA, and caused a decrease in EYA2 mRNA stability and cell viability. Our results suggest that eya mRNA is a target of TTP proteins, and that downregulation of EYA by TTP may lead to reduced cell viability in Drosophila and human cells.


RNA Biology | 2015

The RNA recognition motif of NIFK is required for rRNA maturation during cell cycle progression.

Wen-An Pan; Hsin-Yue Tsai; Shun-Chang Wang; Michael Hsiao; Pei-Yu Wu; Ming-Daw Tsai

Ribosome biogenesis governs protein synthesis. NIFK is transactivated by c-Myc, the key regulator of ribosome biogenesis. The biological function of human NIFK is not well established, except that it has been shown to interact with Ki67 and NPM1. Here we report that NIFK is required for cell cycle progression and participates in the ribosome biogenesis via its RNA recognition motif (RRM). We show that silencing of NIFK inhibits cell proliferation through a reversible p53-dependent G1 arrest, possibly by induction of the RPL5/RPL11-mediated nucleolar stress. Mechanistically it is the consequence of impaired maturation of 28S and 5.8S rRNA resulting from inefficient cleavage of internal transcribed spacer (ITS) 1, a critical step in the separation of pre-ribosome to small and large subunits. Complementation of NIFK silencing by mutants shows that RNA-binding ability of RRM is essential for the pre-rRNA processing and G1 progression. More specifically, we validate that the RRM of NIFK preferentially binds to the 5′-region of ITS2 rRNA likely in both sequence specific and secondary structure dependent manners. Our results show how NIFK is involved in cell cycle progression through RRM-dependent pre-rRNA maturation, which could enhance our understanding of the function of NIFK in cell proliferation, and potentially also cancer and ribosomopathies.


Biochemistry | 2017

Phospho-priming Confers Functionally Relevant Specificities for Rad53 Kinase Autophosphorylation

Eric S.-W. Chen; Jui-Hung Weng; Yu-Hou Chen; Shun-Chang Wang; Xiao-Xia Liu; Wei-Cheng Huang; Tsutomu Matsui; Yoshiaki Kawano; Jiahn-Haur Liao; Liang-Hin Lim; Yoshitaka Bessho; Kai-Fa Huang; Wen-Jin Wu; Ming-Daw Tsai

The vast majority of in vitro structural and functional studies of the activation mechanism of protein kinases use the kinase domain alone. Well-demonstrated effects of regulatory domains or allosteric factors are scarce for serine/threonine kinases. Here we use a site-specifically phosphorylated SCD1-FHA1-kinase three-domain construct of the serine/threonine kinase Rad53 to show the effect of phospho-priming, an in vivo regulatory mechanism, on the autophosphorylation intermediate and specificity. Unphosphorylated Rad53 is a flexible monomer in solution but is captured in an asymmetric enzyme:substrate complex in crystal with the two FHA domains separated from each other. Phospho-priming induces formation of a stable dimer via intermolecular pT-FHA binding in solution. Importantly, autophosphorylation of unprimed and phospho-primed Rad53 produced predominantly inactive pS350-Rad53 and active pT354-Rad53, respectively. The latter mechanism was also demonstrated in vivo. Our results show that, while Rad53 can display active conformations under various conditions, simulation of in vivo regulatory conditions confers functionally relevant autophosphorylation.


PLOS ONE | 2012

Tristetraprolin Inhibits Poly(A)-Tail Synthesis in Nuclear mRNA that Contains AU-Rich Elements by Interacting with Poly(A)-Binding Protein Nuclear 1

Yu-Lun Su; Shun-Chang Wang; Pei-Yu Chiang; Nien-Yi Lin; Yu-Fang Shen; Geen-Dong Chang; Ching-Jin Chang


Archive | 2012

Activation B κ Factor Alpha Stimulation and NF- and TIFA-pT Mediates Tumor Necrosis Intermolecular Binding between TIFA-FHA

Ming-Daw Tsai; Shui-Tsung Chen; Shun-Chang Wang; Dongyan Qin; Chin Chun Hung; Pei-Yu Gabriel Wu; Pang-Hung Hsu; Yu-Hou Chen; Chia-Chi Flora Huang; Jui-Hung Weng; Tong-You Wade


Archive | 2005

Automated Photometry and KBO Occultation Event Detection in the TAOS Project

Zhening Zhang; Charles R. Alcock; Federica B. Bianco; Yong-Ik Byun; Wan Ping Chen; Kem Holland Cook; Rahul Surendra Dave; Joseph Michael Giammarco; S. Ho Kee King; Tai Sing Lee; M. J. Lehner; Jack J. Lissauer; Simon L. Marshall; Imke de Pater; R. Porrata; John A. Rice; Megan E. Schwamb; Andrew H.-J. Wang; Shun-Chang Wang; Cathlyn Y. Wen

Collaboration


Dive into the Shun-Chang Wang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cathlyn Y. Wen

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wan Ping Chen

National Central University

View shared research outputs
Top Co-Authors

Avatar

Charles R. Alcock

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Imke de Pater

University of California

View shared research outputs
Top Co-Authors

Avatar

Jack J. Lissauer

State University of New York System

View shared research outputs
Top Co-Authors

Avatar

John A. Rice

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge