Network


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

Hotspot


Dive into the research topics where Jia-Ru Wu is active.

Publication


Featured researches published by Jia-Ru Wu.


Cancer and Metastasis Reviews | 2008

Signal cross talks for sustained MAPK activation and cell migration: the potential role of reactive oxygen species.

Wen-Sheng Wu; Jia-Ru Wu; Chi-Tan Hu

Signal transduction exerted by the microenvironment around the primary tumor locus may trigger tumor metastasis especially at the migration stage. Sustained mitogen activated protein kinase (MAPK) signaling involved in uncontrolled tumor cell migration rely on the cross talks between integrin, receptor tyrosine kinase (RTK) and protein kinase C (PKC). The molecular mechanisms for cross talking between these migration-related signal cascades leading to sustained cell migration are reviewed, focusing on the focal adhesion scaffold protein paxillin as the platform for signal integration. We proposed reactive oxygen species (ROS) as the critical signal messenger sustaining these signal cascades. For the cross talk of integrin with RTK, ROS may suppress paxillin-associated protein tyrosine phosphatase (PTP–PEST) relieving its negative regulatory effects. For the cross talk of integrin with PKC, PKC itself may phosphorylate integrin or paxillin-associated focal adhesion proteins to induce generation of ROS which may reactivate PKC. In the future, ROS will be validated as the promising therapeutic targets for prevention of tumor metastasis.


Molecular Cancer Research | 2006

Reactive Oxygen Species Mediated Sustained Activation of Protein Kinase C α and Extracellular Signal-Regulated Kinase for Migration of Human Hepatoma Cell Hepg2

Wen-Sheng Wu; Rong-Kung Tsai; Chung Hsing Chang; Sindy Wang; Jia-Ru Wu; Yu-Xun Chang

The tumor promoter 12-O-tetradecanoylphorbol-13-acetate (TPA) can trigger growth inhibition, epithelial-mesenchymal transition (EMT)–like cell scattering, and migration of hepatoma cells HepG2 in a protein kinase C-α (PKC-α)–dependent manner. Saikosaponin a, an ingredient of antitumorigenic Chinese herb Sho-Saiko-to, inhibited cell growth but did not induce EMT-like cell scattering and cell migration of HepG2. Saikosaponin a and TPA induced transient (for 30 minutes) and sustained (until 6 hours) phosphorylation of extracellular signal-regulated kinase (ERK), respectively. Generation of the reactive oxygen species (ROS) was induced by TPA, but not saikosaponin a, for 3 hours. As expected, scavengers of ROS, such as superoxide dismutase, catalase, and mannitol, and the thiol-containing antioxidant N-acetylcystein dramatically suppressed the TPA-triggered cell migration but not growth inhibition of HepG2. The generation of ROS induced by TPA was PKC, but not ERK, dependent. On the other hand, scavengers of ROS and N-acetylcystein also prevented PKC activation and ERK phosphorylation induced by TPA. On the transcriptional level, TPA can induce gene expression of integrins α5, α6, and β1 and reduce gene expression of E-cahedrin in a PKC- and ROS-dependent manner. In conclusion, ROS play a central role in mediating TPA-triggered sustained PKC and ERK signaling for regulation of gene expression of integrins and E-cahedrin that are responsible for EMT and migration of HepG2. (Mol Cancer Res 2006;4(10):747–58)


Journal of Biomedical Science | 2008

The transcriptional factor Snail simultaneously triggers cell cycle arrest and migration of human hepatoma HepG2

Chi-Tan Hu; Jia-Ru Wu; Tsu Yao Chang; Chuan-Chu Cheng; Wen-Sheng Wu

Snail was recently highlighted as a critical transcriptional factor for tumor metastasis. Real time RT/PCR and Western blot analysis demonstrated that Snail mRNA and protein, respectively, were induced by 12-O-tetradecanoylphorbol-13-acetate (TPA) in hepatoma cell HepG2. Blockade of gene expression of Snail by antisense oligodeoxynucleotide and/or siRNA technique can prevent not only the TPA-triggered EMT/cell migration and growth inhibition of HepG2 but also TPA-induced down-regulation of E-cadherin and up-regulation of p15(INK4b). Moreover, the TPA-triggered promoter activation of p15(INK4b) was also prevented. On the other hand, two of the HepG2 clone over-expressing Snail, namely S7 and S15, had a scattered fibroblastic morphology and acquired higher motility than parental HepG2. Also, the proportion of G0/G1 phase of S7 and S15 was higher than that of parental HepG2, consistent with the longer doubling time of both cells. Semiquantitative RT/PCR analysis demonstrated a greatly elevated gene expression of Snail accompanied with decreased E-cadherin and increased p15(INK4b) in both Snail-overexpressing cells. On the transcriptional level, p15(INK4b) promoter activity was 2.6-fold higher in S7 as compared with parental HepG2. Furthermore, electrophoretic mobility of DNA fragments encompassing proximal p15(INK4b) promoter can be retarded by incubation of nuclear extract of S7. Our results demonstrated that Snail play diverse trans-regulatory roles in HepG2. Notably, we suggested that Snail may upregulate p15(INK4b) gene expression by directly activating its promoter.


FEBS Journal | 2010

Snail associates with EGR‐1 and SP‐1 to upregulate transcriptional activation of p15INK4b

Chi-Tan Hu; Tsu-Yao Chang; Chuan-Chu Cheng; Chun-Shan Liu; Jia-Ru Wu; Ming-Che Li; Wen-Sheng Wu

Snail is a multifunctional transcriptional factor that has been described as a repressor in many different contexts. It is also proposed as an activator in a few cases relevant to tumor progression and cell‐cycle arrest. This study investigated the detailed mechanisms by which Snail upregulates gene expression of the CDK inhibitor p15INK4b in HepG2 induced by the tumor promoter tetradecanoyl phorbol acetate (TPA). Using deletion mapping, the TPA‐responsive element on the p15INK4b promoter was located between 77 and 228 bp upstream of the transcriptional initiation site, within which the putative binding regions of early growth response gene 1 (EGR‐1) and stimulatory protein 1 (SP‐1) were found. Gene expression of EGR‐1, Snail and SP‐1 can be induced by TPA within 0.5–6 h. In addition, basal levels of SP‐1, but not of the other two transcriptional factors, were observed. Blockade of TPA‐induced gene expression of Snail, EGR‐1 or SP‐1 suppressed activation of the p15–pro228 reporter plasmid harboring the TPA‐responsive element. More detailed deletion mapping and site‐directed mutagenesis further concluded that the overlapping EGR‐1/SP‐1‐binding site was required for TPA‐induced p15–pro228 activation. In an EMSA, a DNA–protein complex was elevated by TPA, which can be blocked by antibodies against EGR‐1, SP‐1 or Snail at 6 h. Immunoprecipitation/western blotting demonstrated that TPA could trigger the association of EGR‐1 with Snail or SP‐1. Furthermore, a double chromatin immunoprecipitation assay verified that EGR‐1 could form a complex with Snail or SP‐1 on the TPA‐responsive element after treatment with TPA for 2–6 h. Finally, we demonstrated a novel Snail‐target region which could be bound by Snail and was also required for TPA‐induced p15–pro228 activation. In conclusion, Snail associates with EGR‐1 and SP‐1 to mediate TPA‐induced transcriptional upregulation of p15INK4b in HepG2.


Cellular Signalling | 2013

PKC mediates fluctuant ERK-paxillin signaling for hepatocyte growth factor-induced migration of hepatoma cell HepG2

Chi-Tan Hu; Chuan-Chu Cheng; Siou-Mei Pan; Jia-Ru Wu; Wen-Sheng Wu

Hepatocyte growth factor (HGF) is critical for triggering metastasis of hepatocellular carcinoma cell (HCC). Extracellular signal-regulated kinase (ERK) mediates HGF-induced cell migration via focal adhesion signaling. Protein kinase C (PKC) is a negative regulator of ERK activation, however, both PKC and ERK were required for HGF-induced cell migration. To address this intriguing issue, the signal mechanisms for HGF-induced HepG2 cell migration were investigated in a long-term fashion. HGF-induced phosphorylations of ERK, Src (at Tyr 416) and paxillin (at Ser178 and Tyr31) were up and down for 3 times within 24h. HGF also induced fluctuant PKC activation and Rac degradation. Consistently, HGF induced intermittent actin polarization within 24h, which can be blocked by the inhibitors of PKC (Bisindolymaleimide) and ERK. Inhibitor studies revealed that ERK was required for HGF-induced paxillin phosphorylation at Ser178, whereas PKC and Rac-1 may suppress HGF-induced phosphorylation of ERK and paxillin (at Ser178) and upregulate phosphorylation of paxillin at Tyr31. Based on shRNA technique, PKCα and δ were responsible for suppressing HGF-induced phosphorylation of ERK and paxillin (at Ser178), whereas PKC ε and ζ were required for phosphorylation of paxillin at Tyr31. The HGF-induced fluctuant signaling is reminiscent of c-Met endocytosis. Using Concanavalin A, an inhibitor of endocytosis, we found that c-Met endocytosis was required for PKC to suppress ERK phosphorylation. Moreover, HGF-induced c-Met degradation was also fluctuant, which can be prevented by Bisindolymaleimide. In conclusion, PKC is critical for mediating HGF-induced fluctuant ERK-paxillin signaling during cell migration, probably via triggering endosomal degradation of c-Met.


Cellular Signalling | 2013

The role of endosomal signaling triggered by metastatic growth factors in tumor progression

Chi-Tan Hu; Jia-Ru Wu; Wen-Sheng Wu

Within tumor microenvironment, a lot of growth factors such as hepatocyte growth factor and epidermal growth factor may induce similar signal cascade downstream of receptor tyrosine kinase (RTK) and trigger tumor metastasis synergistically. In the past decades, the intimate relationship of RTK-mediated receptor endocytosis with signal transduction was well established. In general, most RTK undergoes clathrin-dependent endocytosis and/or clathrin-independent endocytosis. The internalized receptors may sustain the signaling within early endosome, recycling to plasma membrane for subsequent ligand engagement or sorting to late endosomes/lysosome for receptor degradation. Moreover, receptor endocytosis influences signal transduction in a temporal and spatial manner for periodical and polarized cellular processes such as cell migration. The endosomal signalings triggered by various metastatic factors are quite similar in some critical points, which are essential for triggering cell migration and tumor progression. There are common regulators for receptor endocytosis including dynamin, Rab4, Rab5, Rab11 and Cbl. Moreover, many critical regulators within the RTK signal pathway such as Grb2, p38, PKC and Src were also modulators of endocytosis. In the future, these may constitute a new category of targets for prevention of tumor metastasis.


Cellular Signalling | 2015

PKCε-mediated c-Met endosomal processing directs fluctuant c-Met-JNK-paxillin signaling for tumor progression of HepG2

Chi-Tan Hu; Chuan-Chu Cheng; Jia-Ru Wu; Siou-Mei Pan; Wen-Sheng Wu

Hepatocyte growth factor (HGF) induced c-Met signaling play critical roles in the progression of hepatocellular carcinoma (HCC). However, c-Met targeting approaches suffered resistance and side effect, thus identification of more suitable downstream targets is needed. Recently, we demonstrated HGF-induced fluctuant ERK/paxillin signaling within 24h. We further examined the underlying mechanisms for fluctuant c-Met/JNK/paxillin signal cascade within 12h. HGF-induced phosphorylation of c-Met, JNK, and paxillin (Ser178) shared a common fluctuation pattern characterized by an initial peak at 0.5h, a middle drop at 4h, and a later peak at 10h. Dynasore, the inhibitor of dynamin, suppressed HGF-induced c-Met internalization and phosphorylation of JNK and paxillin (Ser178) at 0.5h, indicating that endosome formation is required for initial signal enhancement. Further, depletion of PKCε not only enhanced HGF-induced phosphorylation of JNK and paxillin (Ser178) but also prevented c-Met degradation at 0.5h, suggesting that PKCε mediated c-Met degradation for signal declination. On the other hand, HGF induced colocalizations of both phosphorylated JNK and paxillin with the endosomal recycling protein GGA3 at 10h and depletion of GGA3 abolished membrane recycling of c-Met and phosphorylation of JNK/paxillin at the same time point. Interestingly, HGF induced GGA3 phosphorylation in a PKCε-dependent manner during 0.5-4h, which is associated with c-Met degradation in the same period. Finally, HGF-induced cell migration, invasion and intrahepatic metastasis of HepG2 were prevented by the inhibitors of endocytosis. Our results suggest that critical endosomal components are promising therapeutic targets for preventing HGF-induced progression of HCC.


PLOS ONE | 2015

Preclinical Trials for Prevention of Tumor Progression of Hepatocellular Carcinoma by LZ-8 Targeting c-Met Dependent and Independent Pathways

Jia-Ru Wu; Chi-Tan Hu; Ren-In You; Pei-ling Ma; Siou-Mei Pan; Ming-Che Lee; Wen-Sheng Wu

Hepatocellular carcinoma (HCC) is among the most lethal cancers. Mounting studies highlighted the essential role of the HGF/c-MET axis in driving HCC tumor progression. Therefore, c-Met is a potential therapeutic target for HCC. However, several concerns remain unresolved in c-Met targeting. First, the status of active c-Met in HCC must be screened to determine patients suitable for therapy. Second, resistance and side effects have been observed frequently when using conventional c-Met inhibitors. Thus, a preclinical system for screening the status of c-Met signaling and identifying efficient and safe anti-HCC agents is urgently required. In this study, immunohistochemical staining of phosphorylated c-Met (Tyr1234) on tissue sections indicated that HCCs with positive c-Met signaling accounted for approximately 46% in 26 cases. Second, many patient-derived HCC cell lines were established and characterized according to motility and c-Met signaling status. Moreover, LZ8, a medicinal peptide purified from the herb Lingzhi, featuring immunomodulatory and anticancer properties, was capable of suppressing cell migration and slightly reducing the survival rate of both c-Met positive and negative HCCs, HCC372, and HCC329, respectively. LZ8 also suppressed the intrahepatic metastasis of HCC329 in SCID mice. On the molecular level, LZ8 suppressed the expression of c-Met and phosphorylation of c-Met, ERK and AKT in HCC372, and suppressed the phosphorylation of JNK, ERK, and AKT in HCC329. According to receptor array screening, the major receptor tyrosine kinase activated in HCC329 was found to be the epidermal growth factor receptor (EGFR). Moreover, tyrosine-phosphorylated EGFR (the active EGFR) was greatly suppressed in HCC329 by LZ8 treatment. In addition, LZ8 blocked HGF-induced cell migration and c-Met-dependent signaling in HepG2. In summary, we designed a preclinical trial using LZ8 to prevent the tumor progression of patient-derived HCCs with c-Met-positive or -negative signaling.


Oncotarget | 2015

Hydrogen peroxide inducible clone-5 mediates reactive oxygen species signaling for hepatocellular carcinoma progression.

Jia-Ru Wu; Chi-Tan Hu; Ren-In You; Siou-Mei Pan; Chuan-Chu Cheng; Ming-Che Lee; Chao-Chuan Wu; Yao-Jen Chang; Shu-Chuan Lin; Chang–Shan Chen; Teng-Yi Lin; Wen-Sheng Wu

One of the signaling components involved in hepatocellular carcinoma (HCC) progression is the focal adhesion adaptor paxillin. Hydrogen peroxide inducible clone-5 (Hic-5), one of the paralogs of paxillin, exhibits many biological functions distinct from paxillin, but may cooperate with paxillin to trigger tumor progression. Screening of Hic-5 in 145 surgical HCCs demonstrated overexpression of Hic-5 correlated well with intra- and extra-hepatic metastasis. Hic-5 highly expressed in the patient derived HCCs with high motility such as HCC329 and HCC353 but not in the HCCs with low motility such as HCC340. Blockade of Hic-5 expression prevented constitutive migration of HCC329 and HCC353 and HGF-induced cell migration of HCC340. HCC329Hic-5(−), HCC353Hic-5(−), HCC372Hic-5(−), the HCCs stably depleted of Hic-5, exhibited reduced motility compared with each HCC expressing Scramble shRNA. Moreover, intra/extrahepatic metastasis of HCC329Hic-5(−) in SCID mice greatly decreased compared with HCC329Scramble. On the other hand, ectopic Hic-5 expression in HCC340 promoted its progression. Constitutive and HGF-induced Hic-5 expression in HCCs were suppressed by the reactive oxygen species (ROS) scavengers catalase and dithiotheritol and c-Jun N-terminal kinase (JNK) inhibitor SP600125. On the contrary, depletion of Hic-5 blocked constitutive and HGF-induced ROS generation and JNK phosphorylation in HCCs. Also, ectopic expression of Hic-5 enhanced ROS generation and JNK phosphorylation. These highlighted that Hic-5 plays a central role in the positive feedback ROS-JNK signal cascade. Finally, the Chinese herbal derived anti-HCC peptide LZ-8 suppressed constitutive Hic-5 expression and JNK phosphorylation. In conclusion, Hic-5 mediates ROS-JNK signaling and may serve as a therapeutic target for prevention of HCC progression.


Experimental Eye Research | 2008

Ethambutol induces PKC-dependent cytotoxic and antiproliferative effects on human retinal pigment cells.

Rong-Kung Tsai; Chung Hsing Chang; Chi Ming Hseu; Shen Ming Chang; Jia-Ru Wu; Hwei-Zu Wang; Wen Chen Wu; Wen-Sheng Wu

Ethambutol (EMB)-induced ocular side effects may involve the influence on functions of retinal pigment epithelium (RPE), in addition to EMB-induced optic neuropathy. To address this issue, the molecular and cellular effects of EMB on RPE including growth regulation, morphological responses, phagocytic activity, and the relevant signaling pathways were investigated. EMB (at optimal concentration 8.0mM) can trigger cell cycle arrest in both RPE50 and ARPE19 cells, accompanied by reduced DNA synthesis. EMB also induced cytoplasmic vacuole formation in both RPE cell lines. Under transmission electric microscope, the phagosomes were replaced by vacuoles and the number of microvilli was reduced in EMB-treated cells. Animal experiments also demonstrated the vacuole formation within RPE of the EMB-treated rats. On the other hand, by in vitro phagocytosis assay using rod outer segment (ROS) as the target, we found EMB suppressed phagocytosis in the cultured RPE, which is consistent with the decreased rhodopsin uptake in the RPE of the EMB-treated rats. Furthermore, inhibitor of protein kinase C but not MAPK, prevented the EMB-induced phenotypical changes. Using a non-radioactive PKC assay, we also demonstrated the PKC activity in both RPE cell lines can be induced by EMB. In conclusion, EMB may exert toxic effects in RPE including suppression of cell growth, formation of cytoplasmic vacuoles and reduction of phagocytic functions via PKC signal pathway.

Collaboration


Dive into the Jia-Ru Wu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chung Hsing Chang

Kaohsiung Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge