Network


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

Hotspot


Dive into the research topics where Ching-Jin Chang is active.

Publication


Featured researches published by Ching-Jin Chang.


FEBS Journal | 2007

Regulation of tristetraprolin during differentiation of 3T3-L1 preadipocytes

Nien-Yi Lin; Chung-Tien Lin; Yu-Ling Chen; Ching-Jin Chang

Tristetraprolin is a zinc‐finger‐containing RNA‐binding protein. Tristetraprolin binds to AU‐rich elements of target mRNAs such as proto‐oncogenes, cytokines and growth factors, and then induces mRNA rapid degradation. It was observed as an immediate‐early gene that was induced in response to several kinds of stimulus, such as insulin and other growth factors and stimulators of innate immunity such as lipopolysaccharides. We observed that tristetraprolin was briefly expressed during a 1–8 h period after induction of differentiation in 3T3‐L1 preadipocytes. Detailed analysis showed that tristetraprolin mRNA expression was stimulated by fetal bovine serum and differentiation inducers, and was followed by rapid degradation. The 3′UTR of tristetraprolin mRNAs contain adenine‐ and uridine‐rich elements. Biochemical analyses using RNA pull‐down, RNA immunoprecipitation and gel shift experiments demonstrated that adenine‐ and uridine‐rich element‐binding proteins, HuR and tristetraprolin itself, were associated with tristetraprolin adenine‐ and uridine‐rich elements. Functional characterization confirmed that tristetraprolin negatively regulated its own expression. Thus, our results indicated that the tight autoregulation of tristetraprolin expression correlated with its critical functional role in 3T3‐L1 differentiation.


Journal of Biological Chemistry | 2013

Reversible Acetylation Regulates Salt-Inducible Kinase (SIK2) and Its Function in Autophagy

Fu-Chia Yang; Bertrand Chin-Ming Tan; Wei-Hao Chen; Lin Yh; Jing-Yi Huang; Hsin-Yun Chang; Sun Hy; Pang-Hung Hsu; Liou Gg; Shen J; Ching-Jin Chang; Chau-Chung Han; Tsai; Sheng-Chung Lee

Background: Salt-inducible kinase (SIK) 2 is an AMP-activated protein kinase family kinase that mediates hormonal and nutrient signaling but has no known link to cellular stress response. Results: p300/CBP and HDAC6 reciprocally regulates Lys-53 acetylation of SIK2, consequently impacting its activity and function in autophagosome maturation. Conclusion: SIK2 kinase activity, via a acetylation-based regulatory switch, contributes to autophagy progression. Significance: SIK2 may be linked to neurodegenerative or protein aggregate disorders. Salt-inducible kinase 2 (SIK2) is a serine/threonine protein kinase belonging to the AMP-activated protein kinase (AMPK) family. SIK2 has been shown to function in the insulin-signaling pathway during adipocyte differentiation and to modulate CREB-mediated gene expression in response to hormones and nutrients. However, molecular mechanisms underlying the regulation of SIK2 kinase activity remains largely elusive. Here we report a dynamic, post-translational regulation of its kinase activity that is coordinated by an acetylation-deaceytlation switch, p300/CBP-mediated Lys-53 acetylation inhibits SIK2 kinase activity, whereas HDAC6-mediated deacetylation restores the activity. Interestingly, overexpression of acetylation-mimetic mutant of SIK2 (SIK2-K53Q), but not the nonacetylatable K53R variant, resulted in accumulation of autophagosomes. Further consistent with a role in autophagy, knockdown of SIK2 abrogated autophagosome and lysosome fusion. Consequently, SIK2 and its kinase activity are indispensable for the removal of TDP-43Δ inclusion bodies. Our findings uncover SIK2 as a critical determinant in autophagy progression and further suggest a mechanism in which the interplay among kinase and deacetylase activities contributes to cellular protein pool homeostasis.


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.


Obesity | 2012

The Role of Nocturnin in Early Adipogenesis and Modulation of Systemic Insulin Resistance in Human

Siow-Wey Hee; Shu-Huei Tsai; Yi-Cheng Chang; Ching-Jin Chang; I‐Shing Yu; Po-Chu Lee; Wei-Jei Lee; Yun‐Chia Emily Chang; Lee-Ming Chuang

The deadenylase nocturnin (Noc, Ccrn4l) has been recently found to regulate lipid metabolism and to control preadipocyte differentiation. Here, we showed that among the five deadenylases tested, Noc and Pan2 exhibited a biphasic expression which is out of phase to each other during adipocyte differentiation of 3T3‐L1 cells. The expression levels of other deadenylases, including Parn, Ccr4, and Caf1, were relatively unchanged or reduced. The immediate early expressed Noc during 3T3‐L1 adipogenesis was involved in regulating mitotic clonal expansion (MCE) and cyclin D1 expression, as demonstrated in Noc‐silenced 3T3‐L1 cells and Noc−/− primary mouse embryonic fibroblasts (MEFs). Transcriptional profiling of Noc‐depleted 3T3‐L1 adipocytes revealed that most of the differentially expressed genes were related to cell growth and proliferation. In human adipose tissue, NOC mRNA level negatively associated with both fasting serum insulin and homeostasis model assessment of insulin resistance, and positively associated with both adiponectin mRNA levels and circulating adiponectin levels. Taken together, these results suggest the role of Noc in the modulation of early adipogenesis as well as systemic insulin sensitivity.


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.


PLOS ONE | 2017

A novel function of twins, B subunit of protein phosphatase 2A, in regulating actin polymerization

Po-An Yeh; Ching-Jin Chang

Actin is an important component of the cytoskeleton and its polymerization is delicately regulated by several kinases and phosphatases. Heterotrimeric protein phosphatase 2A (PP2A) is a potent phosphatase that is crucial for cell proliferation, apoptosis, tumorigenesis, signal transduction, cytoskeleton arrangement, and neurodegeneration. To facilitate these varied functions, different regulators determine the different targets of PP2A. Among these regulators of PP2A, the B subunits in particular may be involved in cytoskeleton arrangement. However, little is known about the role of PP2A in actin polymerization in vivo. Using sophisticated fly genetics, we demonstrated a novel function for the fly B subunit, twins, to promote actin polymerization in varied tissue types, suggesting a broad and conserved effect. Furthermore, our genetic data suggest that twins may act upstream of the actin-polymerized-proteins, Moesin and Myosin-light-chain, and downstream of Rho to promote actin polymerization. This work opens a new avenue for exploring the biological functions of a PP2A regulator, twins, in cytoskeleton regulation.


Biochemical and Biophysical Research Communications | 2006

Differential regulation of ARE-mediated TNFα and IL-1β mRNA stability by lipopolysaccharide in RAW264.7 cells

Yu-Ling Chen; Ya-Lin Huang; Nien-Yi Lin; Hui-Chen Chen; Wan-Chih Chiu; Ching-Jin Chang


Biochemical and Biophysical Research Communications | 2008

Modulation of immediate early gene expression by tristetraprolin in the differentiation of 3T3-L1 cells.

Nien-Yi Lin; Chung-Tien Lin; Ching-Jin Chang

Collaboration


Dive into the Ching-Jin Chang's collaboration.

Top Co-Authors

Avatar

Nien-Yi Lin

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yu-Lun Su

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Geen-Dong Chang

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Pei-Yu Chiang

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Sheng-Chung Lee

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Yu-Ling Chen

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Chung-Tien Lin

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Kuan-Ting Wang

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Mau-Sun Chang

Mackay Memorial Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge