Network


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

Hotspot


Dive into the research topics where Chia-Liang Lin is active.

Publication


Featured researches published by Chia-Liang Lin.


Oncotarget | 2015

Licochalcone A induces autophagy through PI3K/Akt/mTOR inactivation and autophagy suppression enhances Licochalcone A-induced apoptosis of human cervical cancer cells.

Jen-Pi Tsai; Chien-Hsing Lee; Tsung-Ho Ying; Chu-Liang Lin; Chia-Liang Lin; Jung-Tsung Hsueh; Yi-Hsien Hsieh

The use of dietary bioactive compounds in chemoprevention can potentially reverse, suppress, or even prevent cancer progression. However, the effects of licochalcone A (LicA) on apoptosis and autophagy in cervical cancer cells have not yet been clearly elucidated. In this study, LicA treatment was found to significantly induce the apoptotic and autophagic capacities of cervical cancer cells in vitro and in vivo. MTT assay results showed dose- and time-dependent cytotoxicity in four cervical cancer cell lines treated with LicA. We found that LicA induced mitochondria-dependent apoptosis in SiHa cells, with decreasing Bcl-2 expression. LicA also induced autophagy effects were examined by identifying accumulation of Atg5, Atg7, Atg12 and microtubule-associated protein 1 light chain 3 (LC3)-II. Treatment with autophagy-specific inhibitors (3-methyladenine and bafilomycin A1) enhanced LicA-induced apoptosis. In addition, we suggested the inhibition of phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of mTOR pathway by LicA. Furthermore, the inhibition of PI3K/Akt by LY294002/si-Akt or of mTOR by rapamycin augmented LicA-induced apoptosis and autophagy. Finally, the in vivo mice bearing a SiHa xenograft, LicA dosed at 10 or 20 mg/kg significantly inhibited tumor growth. Our findings demonstrate the chemotherapeutic potential of LicA for treatment of human cervical cancer.


Journal of Neuro-oncology | 2016

Inhibition of pentraxin 3 in glioma cells impairs proliferation and invasion in vitro and in vivo

Jai-Nien Tung; Chung-Po Ko; Shun-Fa Yang; Chun-Wen Cheng; Pei-Ni Chen; Chia-Yu Chang; Chia-Liang Lin; Te-Fang Yang; Yi-Hsien Hsieh; Kun-Chung Chen

Pentraxin 3 (PTX3) is an inflammatory molecule that is involved in immune responses, inflammation, and cancer. Recent evidence suggests that PTX3 plays a critical role in tumor progression; however, its impact on the biological function of gliomas remains unknown. In the present study, immunohistochemical staining showed that patients with high-grade gliomas exhibited increased expression levels of PTX3 compared to those with low-grade gliomas (P < 0.001). Furthermore, knockdown of PTX3 in GBM8401 cells inhibits proliferation, increases p21 protein levels, and decreases cyclin D1 protein levels, resulting in cell cycle arrest at the G0/G1 phase. In addition, knockdown of PTX3 significantly decreases GBM8401 cell migration and invasion through the downregulation of matrix metalloproteinase-1 and -2 (MMP-1 and MMP-2) expression. In a GBM8401 xenograft animal model, PTX3 knockdown decreases tumor growth in vivo. In conclusion, PTX3 plays an important role in glioma cell proliferation and invasion, and may thus serve as a novel potential therapeutic target in the treatment of gliomas.


Cellular Physiology and Biochemistry | 2018

Protodioscin Induces Apoptosis Through ROS-Mediated Endoplasmic Reticulum Stress via the JNK/p38 Activation Pathways in Human Cervical Cancer Cells

Chia-Liang Lin; Chien-Hsing Lee; Chien-Min Chen; Chun-Wen Cheng; Pei-Ni Chen; Tsung-Ho Ying; Yi-Hsien Hsieh

Background/Aims: Protodioscin (PD) is a steroidal saponin with anti-cancer effects on a number of cancer cells, but the anti-tumor effects and mechanism of action of PD on human cervical cancer cells is unclear. Methods: We determined cell viability using the MTT assay. Cell death, mitochondrial membrane potential (MMP), intracellular reactive oxygen species (ROS) generation, and endoplasmic reticulum (ER) stress were measured on a flow cytometry. Caspase activation, ER stress, and MMP-dependent apoptosis proteins in cervical cancer cells in response to PD were determined by Western blot analysis. The ability of ATF4 binding to ChIP promoter was measured using the ChIP assay. Results: We demonstrated that PD inhibits cell viability, causes a loss of mitochondrial function, and induces apoptosis, as evidenced by up-regulation of caspase-8, -3, -9, -PARP, and Bax activation, and down-regulation of Bcl-2 expression. PD was shown to induce ROS and the ER stress pathway, including GRP78, p-eIF-2α, ATF4, and CHOP. Pre-treatment with NAC, a ROS production inhibitor, significantly reduced ER stress and apoptosis-related proteins induced by PD. Transfection of GRP78/CHOP-siRNA effectively inhibited PD-induced ER stress-dependent apoptosis. Moreover, treatment with PD significantly increased p38 and JNK activation. Co-administration of a JNK inhibitor (SP600125) or p38 inhibitor (SB203580) abolished cell death and ER stress effects during PD treatment. In addition, PD induced the expression of nuclear ATF4 and CHOP, as well as the binding ability of ATF4 to the CHOP promoter. Conclusion: Our results suggest that PD is a promising therapeutic agent for the treatment of human cervical cancer.


Biochimica et Biophysica Acta | 2017

Norcantharidin induces mitochondrial-dependent apoptosis through Mcl-1 inhibition in human prostate cancer cells

Chu-Liang Lin; Chien-Min Chen; Chia-Liang Lin; Chun-Wen Cheng; Chien-Hsing Lee; Yi-Hsien Hsieh

Norcantharidin (NCTD) is the demethylated form of cantharidin that exhibits anticancer potential in many cancer cell types. Recent reports suggest that NCTD targeting ROS/AMPK and DNA replication signaling pathway could be an effective strategy for the treatment of PCa cells. However, supportive evidence is limited to the effect of NCTD that induction of apoptosis through suppression of the Mcl-1. Here, we show that NCTD induced PCa cell apoptosis and triggered caspase activation, which was associated with mitochondria dysfunction. Mechanistic investigations suggested that NCTD modulated the Akt signaling via increased nuclear translocation and interaction with the myeloid cell leukemia-1 (Mcl-1) promoter by FOXO4, resulting in an apoptotic effect. Moreover, miR-320d, which targets Mcl-1, was significantly upregulated after NCTD treatment. Overexpression of miR-320d by NCTD induced mitochondria dysfunction and apoptosis, which was notably attenuated with a miR-320d inhibitor. In vivo xenograft analysis revealed that NCTD significantly reduced tumor growth in mice with PC3 tumor xenografts. Taken together, our results provide new insights into the critical role of NCTD in suppressing Mcl-1 via epigenetic upregulation of miR-320d, resulting in PCa cell apoptosis.


Cellular Physiology and Biochemistry | 2017

Alpha-Mangostin Suppresses the Metastasis of Human Renal Carcinoma Cells by Targeting MEK/ERK Expression and MMP-9 Transcription Activity

Chien-Min Chen; Shu-Ching Hsieh; Chia-Liang Lin; Yu-Syun Lin; Jen-Pi Tsai; Yi-Hsien Hsieh

Background/Aims: α-mangostin has anti-carcinogenic effects against several cancers. We investigated the molecular mechanism of this compound on the metastasis of human renal carcinoma cells. Methods: Cell viability was measured using the MTT assay, and cell cycle distribution using flow cytometry. A Matrigel-based assay was used to measure in vitro cell migration and invasion. MAPK-related proteins and matrix metalloproteinase (MMP)-9 and MMP-2 expression were measured by western blotting, and MMP2/-9 activities were determined by gelatin zymography. RT-qPCR and a luciferase assay were used to examine the transcriptional activity of MMP-9. Results: α-mangostin inhibited the migration and invasion of RCC cells in a dose-dependent manner, but had no evident cytotoxic effects. Treatment of 786-O cells with α-mangostin inhibited activation of MEK and ERK. Treatment with a specific MEK inhibitor (U0126) enhanced the inhibitory effects of α-mangostin on cell migration and invasion, and the phosphorylation of ERK and MEK. Moreover, α-mangostin inhibited the expression of the MMP-9 mRNA levels as well as the activity of MMP-9 promoter, and these suppressive effects were further enhanced by U0126. Conclusions: Our results suggest that α-mangostin suppresses cell migration and invasion via MEK/ERK/MMP9 pathway, and might be a promising anti-metastatic agent against human renal cell carcinoma.


Environmental Toxicology | 2017

β-mangostin suppresses human hepatocellular carcinoma cell invasion through inhibition of MMP-2 and MMP-9 expression and activating the ERK and JNK pathways

Chien-Feng Huang; Ying-Hock Teng; Fung-Jou Lu; Wen-Hung Hsu; Chia-Liang Lin; Chia-Chen Hung; Jai-Nien Tung; Yi-Hsien Hsieh; Chung-Jung Liu

β‐mangostin is a dietary xanthone that has been reported to have the anticancer properties in some human cancer cell types. However, the antimetastatic effect and molecular mechanism of β‐mangostin action in human hepatocellular carcinoma (HCC) cells remain unknown. In this study, we found that β‐mangostin did not induce cytotoxicity in human HCC cells (SK‐Hep‐1, Huh‐7 and HA22T/VGH cells). β‐mangostin could inhibit migration and invasion of human HCC cells. Meanwhile, β‐mangostin significantly decreased the protein activities and expression of matrix metalloproteinase (MMP)‐2 and MMP‐9 via increasing the activation of MEK1/2, ERK1/2, MEK4 and JNK1/2 signaling pathways. Furthermore, using specific inhibitor for ERK1/2 (PD98059) and JNK1/2 (JNKII) significantly restored the expression of MMP‐2/‐9 and invasion by β‐mangostin treatment in Huh‐7 cells. In addition, β‐mangostin effectively restored the protein levels and transcription activity of MMP‐2 and MMP‐9 in siERK or siJNK‐transfected Huh‐7 cells, concomitantly with promotion on cell migration and invasion. Taken together, these findings are the first to demonstrate the antimetastatic activity of β‐mangostin against human HCC cells, which may act as a promising therapeutic agent for the treatment of HCC.


Environmental Toxicology | 2018

Synergistic antimetastatic effect of cotreatment with licochalcone A and sorafenib on human hepatocellular carcinoma cells through the inactivation of MKK4/JNK and uPA expression

Min-Hua Wu; Yi-Fang Chiu; Wen-Jun Wu; Pei-Lien Wu; Ching-Yi Lin; Chia-Liang Lin; Yi-Hsien Hsieh; Chung-Jung Liu

To improve the clinical outcome of tumor chemotherapy, more effective combination treatments against tumor metastasis and recurrence are required. Licochalcone A (LicA) is the root of Glycyrrhiza inflata and has been reported to possess anti‐inflammatory, antimicrobial, and antitumor effects. Sorafenib (Sor), a multikinase inhibitor, is used to treat patients with solid tumors such as advanced hepatocellular carcinoma (HCC). However, the synergistic effects of LicA and Sor on the metastasis of human HCC cells have not been reported. We found that LicA and Sor did not have cytotoxic effects or arrest growth in human SK‐Hep‐1 and Huh‐7 cells. In addition, treatment with LicA or Sor alone inhibited migration and invasion in human SK‐Hep‐1 and Huh‐7 HCC cells. Furthermore, cotreatment with LicA and Sor synergistically inhibited the migration and invasion of HCC cells and significantly inhibited uPA protein expression. Notably, cotreatment of LicA and Sor synergistically and significantly downregulated MKK4‐JNK expression. Through tail vein injection in nude mice, the aforementioned cotreatment synergistically suppressed SK‐Hep‐1 cell‐mediated lung metastasis. These findings first revealed the synergistic effects of LicA and Sor cotreatment against human HCC cells, further suggesting that beneficial effects on tumor regression could be confirmed through prospective clinical trials.


Environmental Toxicology | 2018

Nimbolide induced apoptosis by activating ERK-mediated inhibition of c-IAP1 expression in human hepatocellular carcinoma cells

Kuan-Chun Hsueh; Chia-Liang Lin; Jai-Nien Tung; Shun-Fa Yang; Yi-Hsien Hsieh

Nimbolide is one of the major compounds from the leaves and flowers of the neem tree and exhibits antitumor properties on various cancer cells. However, no report has shown that nimbolide induces apoptosis in vitro and in vivo in human hepatocellular carcinoma cells. Our results indicated that it inhibited cell growth in Huh‐7 and PLC/PRF/5 cells. We also found that nimbolide induced cell death through the induction of G2/M phase arrest and mitochondrial dysfunction, accompanied by the increased expression of cleaved caspase‐7, caspase‐9, caspase‐3, caspase‐PARP, and Bax and decreased expression of Mcl‐1 and Bcl‐2. A human apoptosis antibody array analysis demonstrated that inhibition of the apoptosis family proteins (XIAP, c‐IAP1, and c‐IAP2) was one of the major targets of nimbolide. Additionally, nimbolide sustained activation of ERK expression. Moreover, pretreatment with U0126 (MEK inhibitor) markedly abolished nimbolide‐inhibited cell viability, induced cell apoptosis, ERK phosphorylation, cleaved caspase‐9, caspase‐3, cleaved‐PARP activation, and increased c‐IAP1 expression in Huh‐7 cells. An in vivo study showed that nimbolide significantly reduced Huh‐7 tumor growth and weight in a xenograft mouse model. This study indicated the antitumor potential of nimbolide in human hepatocellular carcinoma cells.


International Journal of Molecular Sciences | 2017

Praeruptorin A Inhibits Human Cervical Cancer Cell Growth and Invasion by Suppressing MMP-2 Expression and ERK1/2 Signaling

Min-Hua Wu; Chia-Liang Lin; Hui-Ling Chiou; Shun-Fa Yang; Ching-Yi Lin; Chung-Jung Liu; Yi-Hsien Hsieh

Praeruptorin A (PA) is a pyranocumarin present in the dried root of Peucedanumpraeruptorum Dunn that has anticancer effects against several types of cells. However, the effect of PA on human cervical cancer cells is unknown. Our results indicate that PA significantly inhibited cell proliferation, colony formation, migration, invasion, and wound closure of HeLa and SiHa cells, induced cell cycle arrest at G0/G1 phase, upregulated Rb, p16, p21 and p27 proteins and downregulated cyclin D1 and S-phase kinase-associated protein 2 (Skp2) proteins. PA also significantly reduced expression of matrix metalloproteinase-2 (MMP-2) and increased expression of tissue inhibitor of metalloproteinase-2 (TIMP-2). In addition, PA suppressed ERK1/2 activation and increased the effect of PD98059 (a specific MEK1/2 inhibitor) in downregulation of MMP-2 and upregulation of TIMP-2. PA treatment inhibited the effect of 12-O-tetradecanoylphorbol-13-acetate (TPA) on upregulation of ERK1/2 activation, MMP-2 expression, cellular migration, and invasion of HeLa cells. Our findings are the first to demonstrate the activity of PA against cervical cancer cells, and suggest this agent has promise as a therapeutic agent in treatment of human cervical cancer.


Tumor Biology | 2015

Induction of cell cycle arrest, DNA damage, and apoptosis by nimbolide in human renal cell carcinoma cells

Yi-Hsien Hsieh; Chien-Hsing Lee; Hsiao-Yun Chen; Shu-Ching Hsieh; Chia-Liang Lin; Jen-Pi Tsai

Collaboration


Dive into the Chia-Liang Lin's collaboration.

Top Co-Authors

Avatar

Yi-Hsien Hsieh

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Chun-Wen Cheng

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Jen-Pi Tsai

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Shu-Ching Hsieh

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Shun-Fa Yang

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Chu-Liang Lin

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Chung-Jung Liu

Kaohsiung Medical University

View shared research outputs
Top Co-Authors

Avatar

Chung-Po Ko

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Hui-Ling Chiou

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Kun-Chung Chen

Chung Shan Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge