Network


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

Hotspot


Dive into the research topics where Chi Cheng Lu is active.

Publication


Featured researches published by Chi Cheng Lu.


Molecular Nutrition & Food Research | 2010

Chrysophanol induces necrosis through the production of ROS and alteration of ATP levels in J5 human liver cancer cells

Chi Cheng Lu; Jai Sing Yang; An Cheng Huang; Te Chun Hsia; Su Tze Chou; Chao Lin Kuo; Hsu Feng Lu; Tsung-Han Lee; Wellington Gibson Wood; Jing Gung Chung

Anthraquinone compounds have been shown to induce apoptosis in different cancer cell types. Effects of chrysophanol, an anthraquinone compound, on cancer cell death have not been well studied. The goal of this study was to examine if chrysophanol had cytotoxic effects and if such effects involved apoptosis or necrosis in J5 human liver cancer cells. Chrysophanol induced necrosis in J5 cells in a dose- and time-dependent manner. Non-apoptotic cell death was induced by chrysophanol in J5 cells and was characterized by caspase independence, delayed externalization of phosphatidylserine and plasma membrane disruption. Blockage of apoptotic induction by a general caspase inhibitor (z-VAD-fmk) failed to protect cells against chrysophanol-induced cell death. The levels of reactive oxygen species production and loss of mitochondrial membrane potential (DeltaPsi(m)) were also determined to assess the effects of chrysophanol. However, reductions in adenosine triphosphate levels and increases in lactate dehydrogenase activity indicated that chrysophanol stimulated necrotic cell death. In summary, human liver cancer cells treated with chrysophanol exhibited a cellular pattern associated with necrosis and not apoptosis.


Food and Chemical Toxicology | 2012

Antitumor effects of emodin on LS1034 human colon cancer cells in vitro and in vivo: Roles of apoptotic cell death and LS1034 tumor xenografts model

Yi Shih Ma; Shu Wen Weng; Meng Wei Lin; Chi Cheng Lu; Jo Hua Chiang; Jai Sing Yang; Kuang Chi Lai; Jing Pin Lin; Nou Ying Tang; Jaung Geng Lin; Jing Gung Chung

Emodin, an active natural anthraquinone derivative, is found in the roots and rhizomes of numerous Chinese medicinal herbs and exhibits anticancer effects on many types of human cancer cell lines. The aim of this study investigated that emodin induced apoptosis of human colon cancer cells (LS1034) in vitro and inhibited tumor nude mice xenografts bearing LS1034 in vivo. In in vitro study, emodin induced cell morphological changes, decreased the percentage of viability, induced G2/M phase arrest and increased ROS and Ca(2+) productions as well as loss of mitochondrial membrane potential (ΔΨ(m)) in LS1034 cells. Emodin-triggered apoptosis was also confirmed by DAPI staining and these effects are concentration-dependent. Western blot analysis indicated that the protein levels of cytochrome c, caspase-9 and the ratio of Bax/Bcl-2 were increased in LS1034 cells after emodin exposure. Emodin induced the productions of ROS and Ca(2+) release, and altered anti- and pro-apoptotic proteins, leading to mitochondrial dysfunction and activations of caspase-9 and caspase-3 for causing cell apoptosis. In in vivo study, emodin effectively suppressed tumor growth in tumor nude mice xenografts bearing LS1034. Overall, the potent in vitro and in vivo antitumor activities of emodin suggest that it might be developed for treatment of colon cancer in the future.


Leukemia Research | 2009

Rutin inhibits the proliferation of murine leukemia WEHI-3 cells in vivo and promotes immune response in vivo.

Jing Pin Lin; Jai Sing Yang; Chi Cheng Lu; Jo Hua Chiang; Chang Lin Wu; Jen Jyh Lin; Hui Lu Lin; Mei Due Yang; Kuo Ching Liu; Tsan Hung Chiu; Jing Gung Chung

Flavonoids are polyphenolic compounds found in various foods of plants. Rutin, one of the flavonoids, had been showed induced apoptosis in cancer cells. There is no available information to address rutin affects murine leukemia cells in vivo. In the present study, we are focused on the in vivo effects of rutin on leukemia WEHI-3 cells. The effects of rutin on WEHI-3 in BALB/c mice in vivo were also examined and the results indicated that rutin decreased the percentage of Mac-3 marker, indicating that the differentiation of the precursor of macrophage and T cells was inhibited. The weights of liver and spleen were decreased from rutin-treated groups compared to the control groups and the results indicated that rutin decreased the weight of these organs. One of the major characteristic of WEHI-3 leukemia is the enlarged spleen in murine after i.p. injection of WEHI-3 cells. After the pathological examination, the function of rutin was observed in the liver and spleen in the mice previously injected with WEHI-3 cells. Rutin promoted the activity of macrophage phagocytosis in cells which isolated from peritoneal (i.p.). Taken together, rutin can affect WEHI-3 cells in vivo.


Phytomedicine | 2009

Berberine inhibits human tongue squamous carcinoma cancer tumor growth in a murine xenograft model.

Yung Tsuan Ho; Jai Sing Yang; Chi Cheng Lu; Jo Hua Chiang; Tsai Chung Li; Jen Jyh Lin; Kuang Chi Lai; Ching Lung Liao; Jaung Geng Lin; Jing Gung Chung

Our primary studies showed that berberine induced apoptosis in human tongue cancer SCC-4 cells in vitro. But there is no report to show berberine inhibited SCC-4 cancer cells in vivo on a murine xenograft animal model. SCC-4 tumor cells were implanted into mice and groups of mice were treated with vehicle, berberine (10mg/kg of body weight) and doxorubicin (4mg/kg of body weight). The tested agents were injected once per four days intraperitoneally (i.p.), with treatment starting 4 weeks prior to cells inoculation. Treatment with 4mg/kg of doxorubicin or with 10mg/kg of berberine resulted in a reduction in tumor incidence. Tumor size in xenograft mice treated with 10mg/kg berberine was significantly smaller than that in the control group. Our findings indicated that berbeirne inhibits tumor growth in a xenograft animal model. Therefore, berberine may represent a tongue cancer preventive agent and can be used in clinic.


PLOS ONE | 2012

Novel quinazolinone MJ-29 triggers endoplasmic reticulum stress and intrinsic apoptosis in murine leukemia WEHI-3 cells and inhibits Leukemic mice

Chi Cheng Lu; Jai Sing Yang; Jo Hua Chiang; Mann-Jen Hour; Kuei Li Lin; Jen Jyh Lin; Wen Wen Huang; Minoru Tsuzuki; Tsung-Han Lee; Jing Gung Chung

The present study was to explore the biological responses of the newly compound, MJ-29 in murine myelomonocytic leukemia WEHI-3 cells in vitro and in vivo fates. We focused on the in vitro effects of MJ-29 on ER stress and mitochondria-dependent apoptotic death in WEHI-3 cells, and to hypothesize that MJ-29 might fully impair the orthotopic leukemic mice. Our results indicated that a concentration-dependent decrease of cell viability was shown in MJ-29-treated cells. DNA content was examined utilizing flow cytometry, whereas apoptotic populations were determined using annexin V/PI, DAPI staining and TUNEL assay. Increasing vital factors of mitochondrial dysfunction by MJ-29 were further investigated. Thus, MJ-29-provaked apoptosis of WEHI-3 cells is mediated through the intrinsic pathway. Importantly, intracellular Ca2+ release and ER stress-associated signaling also contributed to MJ-29-triggered cell apoptosis. We found that MJ-29 stimulated the protein levels of calpain 1, CHOP and p-eIF2α pathways in WEHI-3 cells. In in vivo experiments, intraperitoneal administration of MJ-29 significantly improved the total survival rate, enhanced body weight and attenuated enlarged spleen and liver tissues in leukemic mice. The infiltration of immature myeloblastic cells into splenic red pulp was reduced in MJ-29-treated leukemic mice. Moreover, MJ-29 increased the differentiations of T and B cells but decreased that of macrophages and monocytes. Additionally, MJ-29-stimulated immune responses might be involved in anti-leukemic activity in vivo. Based on these observations, MJ-29 suppresses WEHI-3 cells in vitro and in vivo, and it is proposed that this potent and selective agent could be a new chemotherapeutic candidate for anti-leukemia in the future.


International Journal of Oncology | 2012

Bufalin increases sensitivity to AKT/mTOR-induced autophagic cell death in SK-HEP-1 human hepatocellular carcinoma cells

Shih Chang Tsai; Jai Sing Yang; Shu Fen Peng; Chi Cheng Lu; Jo Hua Chiang; Jing Gung Chung; Meng Wei Lin; Jen Kun Lin; Sakae Amagaya; Cinderella Wai Shan Chung; Theng Thang Tung; Wen Wen Huang; Michael T. Tseng

Bufalin is the major component of Chan-Su (a traditional Chinese medicine, TCM) extracts from the venom of Bufo bufo gargarizan. In the present study, we investigated the pharmacological mechanisms of cell cycle arrest and autophagic cell death induced by bufalin in SK-HEP-1 human hepatocellular carcinoma cells in vitro. Bufalin inhibited cell survival by MTT assay and increased cell death by trypan blue exclusion assay in a concentration-dependent manner. In addition, bufalin induced G2/M phase arrest by reducing CDK1 activity. Bufalin triggered DNA fragmentation and apoptotic cell death in SK-HEP-1 cells by DNA gel electrophoresis, TUNEL and caspase-3 activity assay, while bufalin induced autophagic cell death by double-membrane vacuoles (transmission electron microscopy, TEM), acidic vesicular organelles (acridine orange staining) and cleavage of microtubule-associated protein 1 light chain 3 (LC3). Protein expression levels of cyclin A and B, CDK1, phospho-CDK1 (Thr161), Cdc25c, phospho-Cdc25c (Ser198), phospho-AKT (Thr308), phospho-AKT (Ser473), phospho‑mTOR (Ser2481) were downregulated. In contrast, protein expression levels of the Chk1, Wee1, LC3-II, Beclin-1, Atg 5, Atg 7 and Atg 12 were upregulated in SK-HEP-1 cells after bufalin treatment. Inhibition of autophagy by 3-methyladenine (an inhibitor of class III phosphatidylinositol-3 kinase; 3-MA) or bafilomycin A1 (an inhibitor of the vacuolar proton pump of lysosomes and endosomes) reduced the effect of bufalin on cell viability and enhanced the effect of bufalin on apoptosis. In conclusion, bufalin triggered autophagic cell death and G2/M phase arrest through the AKT/mTOR signaling pathway in SK-HEP-1 cells. Our findings showed that bufalin may be potentially efficacious in the treatment of human hepatocellular carcinoma.


International Journal of Oncology | 2013

Curcumin-loaded nanoparticles induce apoptotic cell death through regulation of the function of MDR1 and reactive oxygen species in cisplatin-resistant CAR human oral cancer cells

Pei Ying Chang; Shu Fen Peng; Chao Ying Lee; Chi Cheng Lu; Shih Chang Tsai; Tzong-Ming Shieh; Tian Shung Wu; Ming-Gene Tu; Michael Yuanchien Chen; Jai Sing Yang

Curcumin is a polyphenolic compound which possesses anticancer potential. It has been shown to induce cell death in a variety of cancer cells, however, its effect on CAL27‑cisplatin-resistant human oral cancer cells (CAR cells) has not been elucidated to date. The low water solubility of curcumin which leads to poor bioavailability, however, has been highlighted as a major limiting factor. In this study, we utilized water-soluble PLGA curcumin nanoparticles (Cur-NPs), and investigated the effects of Cur-NPs on CAR cells. The results showed Cur-NPs induced apoptosis in CAR cells but exhibited low cytotoxicity to normal human gingival fibroblasts (HGFs) and normal human oral keratinocytes (OKs). Cur-NPs triggered DNA concentration, fragmentation and subsequent apoptosis. Compared to untreated CAR cells, a more detectable amount of Calcein-AM accumulation was found inside the treated CAR cells. Cur-NPs suppressed the protein and mRNA expression levels of MDR1. Both the activity and the expression levels of caspase-3 and caspase-9 were elevated in the treated CAR cells. The Cur-NP-triggered apoptosis was blocked by specific inhibitors of pan-caspase (z-VAD-fmk), caspase-3 (z-DEVD-fmk), caspase-9 (z-LEHD-fmk) and antioxidant agent (N-acetylcysteine; NAC). Cur-NPs increased reactive oxygen species (ROS) production, upregulated the protein expression levels of cleaved caspase-3/caspase-9, cytochrome c, Apaf-1, AIF, Bax and downregulated the protein levels of Bcl-2. Our results suggest that Cur-NPs triggered the intrinsic apoptotic pathway through regulating the function of multiple drug resistance protein 1 (MDR1) and the production of reactive oxygen species (ROS) in CAR cells. Cur-NPs could be potentially efficacious in the treatment of cisplatin-resistant human oral cancer.


Leukemia Research | 2009

Benzyl isothiocyanate inhibits murine WEHI-3 leukemia cells in vitro and promotes phagocytosis in BALB/c mice in vivo

Mei Fen Tsou; Ching-Tien Peng; Mu Chin Shih; Jai Sing Yang; Chi Cheng Lu; Jo Hua Chiang; Chang Lin Wu; Jing Pin Lin; Chyi Lo; Ming Jen Fan; Jing Gung Chung

Many evidences have shown that dietary intake of cruciferous vegetables could protect against the risk of various types of malignancies. Benzyl isothiocyanate (BITC), one of the compounds from cruciferous vegetables, had shown induced cell cycle arrest and apoptosis in cancer cells. However, there is no available information to address that BITC affects murine leukemia cells in vitro and in vivo. Here, we investigated in vitro effects of BITC on murine leukemia WEHI-3 cells. BITC decreased the percentage of viable cells via G0/G1 arrest and apoptosis in WEHI-3 cells. BITC induced apoptosis through the dysfunction of mitochondria (decreased the levels of mitochondria membrane potential) and activation of caspase-3. Then we investigated in vivo effects of BITC on murine leukemia WEHI-3 cells and the results indicated that BITC decreased the weights of liver and spleen and it also decreased the percentage of CD11b and Mac-3 markers, indicating that the differentiation of the precursor of macrophage and B cells was inhibited. BITC promoted the activity of macrophage phagocytosis in cells which are isolated from PBMC and peritoneal (i.p.). Taken together, BITC can affect WEHI-3 cells in vitro and in vivo.


Phytotherapy Research | 2010

Curcumin inhibits human lung large cell carcinoma cancer tumour growth in a murine xenograft model

Chin Cheng Su; Jai Sing Yang; Chi Cheng Lu; Jo Hua Chiang; Chang Lin Wu; Jen Jyh Lin; Kuang Chi Lai; Te Chun Hsia; Hsu Feng Lu; Ming Jen Fan; Jing Gung Chung

Curcumin can decrease viable cells through the induction of apoptosis in human lung cancer NCI‐H460 cells in vitro. However, there are no reports that curcumin can inhibit cancer cells in vivo. In this study, NCI‐H460 lung tumour cells were implanted directly into nude mice and divided randomly into four groups to be treated with vehicle, curcumin (30 mg/kg of body weight), curcumin (45 mg/kg of body weight) and doxorubicin (8 mg/kg of body weight). Each agent was injected once every 4 days intraperitoneally (i.p.), with treatment starting 4 weeks after inoculation with the NCI‐H460 cells. Treatment with 30 mg/kg and 45 mg/kg of curcumin or with 8 mg/kg of doxorubicin resulted in a reduction in tumour incidence, size and weight compared with the control group. The findings indicate that curcumin can inhibit tumour growth in a NCI‐H460 xenograft animal model in vivo. Copyright


Phytotherapy Research | 2009

Quercetin Inhibited Murine Leukemia WEHI-3 Cells In Vivo and Promoted Immune Response

Chun Shu Yu; Kuang Chi Lai; Jai Sing Yang; Jo Hua Chiang; Chi Cheng Lu; Chang Lin Wu; Jing Pin Lin; Ching Lung Liao; Nou Ying Tang; W. Gibson Wood; Jing Gung Chung

Enhanced flavonoid consumption is closely related with a reduced cancer incidence as shown in epidemiological studies. Quercetin (3,5,7,3′,4′‐pentahydroxylflavone) is one of the active components of flavonoids which exist in natural plants, particularly in onions and fruits. It was reported that quercetin induced apoptosis in human cancer cell lines, including human leukemia HL‐60 cells, but there is no available information as to its effects on leukemia cells in vivo. The purpose of the present studies was to focus on the in vivo effects of quercetin on leukemia WEHI‐3 cells. The effects of quercetin on WEHI‐3 cells injected into BALB/c mice were examined. Quercetin decreased the percentage of Mac‐3 and CD11b markers, suggesting that the differentiation of the precursors of macrophages and T cells was inhibited. There was no effect on CD3 levels but increased CD19 levels. Quercetin decreased the weight of the spleen and liver compared with the olive oil treated animals. Quercetin stimulated macrophage phagocytosis of cells isolated from peritoneum. Quercetin also promoted natural killer cell activity. Based on pathological examination, an effect of quercetin was observed in the spleen of mice previously injected with WEHI‐3 cells. Apparently, quercetin affects WEHI‐3 cells in vivo. Copyright

Collaboration


Dive into the Chi Cheng Lu's collaboration.

Top Co-Authors

Avatar

Jo Hua Chiang

National Chung Hsing University

View shared research outputs
Top Co-Authors

Avatar

Hsu Feng Lu

Fu Jen Catholic University

View shared research outputs
Top Co-Authors

Avatar

Tsung-Han Lee

National Chung Hsing University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tian Shung Wu

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar

Sakae Amagaya

Nihon Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar

Yen Fang Wen

Industrial Technology Research Institute

View shared research outputs
Top Co-Authors

Avatar

Yih Jing Tang

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Minoru Tsuzuki

Nihon Pharmaceutical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge