1
|
Natural and synthetic compounds for glioma treatment based on ROS-mediated strategy. Eur J Pharmacol 2023:175537. [PMID: 36871663 DOI: 10.1016/j.ejphar.2023.175537] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 01/08/2023] [Accepted: 01/23/2023] [Indexed: 03/06/2023]
Abstract
Glioma is the most frequent and most malignant tumor of the central nervous system (CNS),accounting for about 50% of all CNS tumor and approximately 80% of the malignant primary tumors in the CNS. Patients with glioma benefit from surgical resection, chemo- and radio-therapy. However these therapeutical strategies do not significantly improve the prognosis, nor increase survival rates owing to restricted drug contribution in the CNS and to the malignant characteristics of glioma. Reactive oxygen species (ROS) are important oxygen-containing molecules that regulate tumorigenesis and tumor progression. When ROS accumulates to cytotoxic levels, this can lead to anti-tumor effects. Multiple chemicals used as therapeutic strategies are based on this mechanism. They regulate intracellular ROS levels directly or indirectly, resulting in the inability of glioma cells to adapt to the damage induced by these substances. In the current review, we summarize the natural products, synthetic compounds and interdisciplinary techniques used for the treatment of glioma. Their possible molecular mechanisms are also presented. Some of them are also used as sensitizers: they modulate ROS levels to improve the outcomes of chemo- and radio-therapy. In addition, we summarize some new targets upstream or downstream of ROS to provide ideas for developing new anti-glioma therapies.
Collapse
|
2
|
Urade R, Chou CK, Chou HL, Chen BH, Wang TN, Tsai EM, Hung CT, Wu SJ, Chiu CC. Phthalate derivative DEHP disturbs the antiproliferative effect of camptothecin in human lung cancer cells by attenuating DNA damage and activating Akt/NF-κB signaling pathway. ENVIRONMENTAL TOXICOLOGY 2023; 38:332-342. [PMID: 36394428 DOI: 10.1002/tox.23686] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 06/16/2023]
Abstract
Plasticizers/phthalates play a facilitating role in the development of cancer and help the tumor to grow and metastasize. Camptothecin (CPT) and its derivatives are known to have anticancer properties of inhibiting cell growth, promoting cell apoptosis, and increasing autophagy. Therefore, in this study, we investigated whether the presence of di(2-ethylhexyl) phthalate (DEHP) could hinder apoptosis and autophagy caused by CPT in non-small cell lung cancer (NSCLC) cells. We found that DEHP interferes with CPT-induced apoptosis and autophagy and increases the prosurvival pathway by reducing the DNA damage marker γ-H2AX and activating the Akt and NF-κB pathways. Furthermore, we also confirmed that combining DEHP with 3-MA has additive effects in inhibiting autophagy and apoptosis in NSCLC cells. Taken together, our findings show that DEHP could affect CPT-induced anticancer treatment and provide evidence to show that DEHP induces chemoresistance in CPT-based chemotherapy.
Collapse
Affiliation(s)
- Ritesh Urade
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Chon-Kit Chou
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau, People's Republic of China
| | - Han-Lin Chou
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Bing-Hung Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tsu-Nai Wang
- Department of Public Health, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Eing-Mei Tsai
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- The Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chun-Tzu Hung
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shyh-Jong Wu
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chien-Chih Chiu
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
3
|
Chen W, Yang W, Zhang C, Liu T, Zhu J, Wang H, Li T, Jin A, Ding L, Xian J, Tian T, Pan B, Guo W, Wang B. Modulation of the p38 MAPK Pathway by Anisomycin Promotes Ferroptosis of Hepatocellular Carcinoma through Phosphorylation of H3S10. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6986445. [PMID: 36466092 PMCID: PMC9715334 DOI: 10.1155/2022/6986445] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/27/2022] [Accepted: 10/08/2022] [Indexed: 07/25/2023]
Abstract
Hepatocellular carcinoma (HCC) is a prevalent malignant tumor worldwide. Ferroptosis is emerging as an effective target for tumor treatment as it has been shown to potentiate cell death in some malignancies. However, it remains unclear whether histone phosphorylation events, an epigenetic mechanism that regulates transcriptional expression, are involved in ferroptosis. Our study found that supplementation with anisomycin, an agonist of p38 mitogen-activated protein kinase (MAPK), induced ferroptosis in HCC cells, and the phosphorylation of histone H3 on serine 10 (p-H3S10) was participated in anisomycin-induced ferroptosis. To investigate the anticancer effects of anisomycin-activated p38 MAPK in HCC, we analyzed cell viability, colony formation, cell death, and cell migration in Hep3B and HCCLM3 cells. The results showed that anisomycin could significantly suppress HCC cell colony formation and migration and induce HCC cell death. The hallmarks of ferroptosis, such as abnormal accumulation of iron and elevated levels of lipid peroxidation and malondialdehyde, were detected to confirm the ability of anisomycin to promote ferroptosis. Furthermore, coincubation with SB203580, an inhibitor of activated p38 MAPK, partially rescued anisomycin-induced ferroptosis. And the levels of p-p38 MAPK and p-H3S10 were successively increased by anisomycin treatment. The relationship between p-H3S10 and ferroptosis was revealed by ChIP sequencing. The reverse transcription PCR and immunofluorescence results showed that NCOA4 was upregulated both in mRNA and protein levels after anisomycin treatment. And by C11-BODIPY staining, we found that anisomycin-induced lipid reactive oxygen species was reduced after NCOA4 knockdown. In conclusion, the anisomycin-activated p38 MAPK promoted ferroptosis of HCC cells through H3S10 phosphorylation.
Collapse
Affiliation(s)
- Wei Chen
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenjing Yang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chunyan Zhang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
| | - Te Liu
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Zhu
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hao Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tong Li
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Anli Jin
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lin Ding
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jingrong Xian
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tongtong Tian
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Baishen Pan
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Guo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Shanghai Zhongshan Hospital, Fudan University, Shanghai, China
| | - Beili Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Shanghai Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Jadhao M, Tsai EM, Yang HC, Chen YF, Liang SS, Wang TN, Teng YN, Huang HW, Wang LF, Chiu CC. The Long-Term DEHP Exposure Confers Multidrug Resistance of Triple-Negative Breast Cancer Cells through ABC Transporters and Intracellular ROS. Antioxidants (Basel) 2021; 10:949. [PMID: 34208283 PMCID: PMC8230873 DOI: 10.3390/antiox10060949] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 12/18/2022] Open
Abstract
The characteristics of phthalates had been thought to be similar to endocrine disruptors, which increases cancer risk. The role of phthalates in acquired drug resistance remains unclear. In this study, we investigated the effect of di-(2-ethylhexyl) phthalate (DEHP) on acquired drug resistance in breast cancer. MCF7 and MDA-MB-231 breast cancer cells were exposed to long-term physiological concentration of DEHP for more than three months. Long-exposure DEHP permanently attenuated the anti-proliferative effect of doxorubicin with estrogen receptor-independent activity even after withdrawal of DEHP. Long term DEHP exposure significantly reduced ROS (O2-) level in MDA-MB-231 cells while increased in MCF7 cells. ATP-binding cassette (ABC) transporters possess a widely recognized mechanism of drug resistance and are considered a target for drug therapy. Upregulation of ABC family proteins, ABCB-1 and ABCC-1 observed in DEHP-exposed clones compared to doxorubicin-resistant (DoxR) and parental MDA-MB-231 cells. A viability assay showed enhanced multidrug resistance in DEHP-exposed clones against Dox, topotecan, and irinotecan. Inhibition of ABC transporters with tariquidar, enhanced drug cytotoxicity through increased drug accumulation reversing acquired multidrug resistance in MDA-MB-231 breast cancer cells. Tariquidar enhanced Dox cytotoxicity by increasing intracellular ROS production leading to caspase-3 mediated apoptosis. Activation of PI3K/Akt signaling enhanced proliferation and growth of DEHP-exposed MDA-MB-231 cells. Overall, long-term DEHP exposure resulted in acquired multidrug resistance by upregulating ABCB-1 and ABCC1; apart from proliferation PI3K/Akt may be responsible for acquired drug resistance through ABC transporter upregulation. Targeting ABCB1 and ABCC1 with tariquidar may be a promising strategy for reversing the acquired multidrug resistance of triple-negative breast cancer cells.
Collapse
Affiliation(s)
- Mahendra Jadhao
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan; or
| | - Eing-Mei Tsai
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- The Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ho-Chun Yang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.Y.); (S.-S.L.)
- Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung 804, Taiwan;
| | - Yih-Fung Chen
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Shih-Shin Liang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.Y.); (S.-S.L.)
| | - Tsu-Nai Wang
- Department of Public Health, College of Health Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Yen-Ni Teng
- Department of Biological Sciences and Technology, National University of Tainan, Tainan 700, Taiwan;
| | - Hurng-Wern Huang
- Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung 804, Taiwan;
| | - Li-Fang Wang
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan; or
| | - Chien-Chih Chiu
- The Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.Y.); (S.-S.L.)
- Center for Cancer Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| |
Collapse
|
5
|
Remizov YO, Kornev AA, Pevzner LM, Petrov ML, Boitsov VM, Stepakov AV. In Vitro Activity of Organochalcogen Compounds: I. Cytotoxic Effect of 4-(1,2,3-Thiadiazol-4-yl)furans Against K562 and HeLa Tumor Cell Lines. RUSS J GEN CHEM+ 2020. [DOI: 10.1134/s1070363220110328] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
6
|
Wang P, Jiang Z, Liu X, Yu K, Wang C, Li H, Zhong L. PI16 attenuates response to sorafenib and represents a predictive biomarker in hepatocellular carcinoma. Cancer Med 2020; 9:6972-6983. [PMID: 32779397 PMCID: PMC7541153 DOI: 10.1002/cam4.3331] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 06/05/2020] [Accepted: 06/25/2020] [Indexed: 01/07/2023] Open
Abstract
Sorafenib has become the only FDA‐approved first‐line therapy for advanced hepatocellular carcinoma (HCC) for more than 10 years, but there is still no validated predictive or prognostic marker. Peptidase inhibitor 16 (PI16) is a functionally unknown gene in cancer research. This study aimed to determine the exact function of PI16 in HCC and whether it can represent as a biomarker for sorafenib response. We found that PI16 was over expressed in HCC tissues vs paired normal tissues. PI16 knockdown sensitize HCC cells to sorafenib treatment both in vitro and in vivo, whereas ectopic PI16 expression produced the opposite effect. Mechanistically, PI16 could suppress p38 MAPK/caspase‐dependent apoptosis in this process, and p38 MAPK inhibitor reversed the sorafenib sensitive phenotype caused by PI16 inhibition. Clinically, immunohistochemistry was used to detect PI16 levels in resected patients with HCC prior to sorafenib treatment. We showed that high PI16 levels represented an independent risk factor for disease progression in patients treated with sorafenib. Patients with low PI16 showed significantly better progression free survival and overall survival after sorafenib therapy. In conclusion, PI16 attenuates response to sorafenib treatment in HCC, and may be a helpful prognostic biomarker of sorafenib treatment.
Collapse
Affiliation(s)
- Pusen Wang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhongyi Jiang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueni Liu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kanru Yu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunguang Wang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Li
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Zhong
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
A novel function of IMPA2, plays a tumor-promoting role in cervical cancer. Cell Death Dis 2020; 11:371. [PMID: 32409648 PMCID: PMC7224180 DOI: 10.1038/s41419-020-2507-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/10/2020] [Accepted: 04/14/2020] [Indexed: 12/19/2022]
Abstract
Discovery of genes and molecular mechanism involved in cervical cancer development would promote the prevention and treatment. By comparing gene expression profiles of cervical carcinoma in situ (CCIS) and adjacent normal tissues, we identified a potential cancer-promoting gene, IMPA2. This study aimed to elucidate the role of IMPA2 and underlying molecular mechanisms in cervical cancer progression. To do this expression of IMPA2 was compared between human cervical cancer and corresponding adjacent normal cervical tissues firstly. CCK-8 assay, clone formation assay, wound healing assay, transwell assay, and tumor formation in nude mice were performed to demonstrate the effect of IMPA2 in cervical cancer proliferation and metastasis. Further proteomic profiling and western blotting explored the molecular pathway involved in the IMPA2-regulating process. The results showed that IMPA2 gene expression was upregulated in cervical cancer. Consistently, silencing of IMPA2 suppressed tumor formation in BALB/c nude mice. Short hairpin RNA (shRNA)-mediated IMPA2 silencing significantly inhibited proliferation and colony-forming abilities of cervical cancer cells, while IMPA2 overexpression had little impact. Also, IMPA2 silencing suppressed cellular migration, but overexpression promoted migration. Proteomics analysis revealed the involvement of mitogen-activated protein kinase (MAPK) pathway in tumor-promoting action of IMPA2. Significantly, the inhibition of IMPA2 activated ERK phosphorylation, and its inhibitory effects can be restored by using selective ERK inhibitor, FR180204. In conclusion, IMPA2 acts as an oncogene in the proliferation and migration of cervical cancer. IMPA2 downregulated ERK phosphorylation to promote cervical cancer. These findings identify a new mechanism underlying cervical cancer and suggest a regulating effect of IMPA2 in MAPK signaling pathway.
Collapse
|
8
|
Jash M, De S, Pramanik S, Chowdhury C. Palladium(II)-Catalyzed Cascade Reactions of Ene–Ynes Tethered to Cyano/Aldehyde: Access to Naphtho[1,2-b]furans and Benzo[g]indoles. J Org Chem 2019; 84:8959-8975. [DOI: 10.1021/acs.joc.9b00861] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Moumita Jash
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
| | - Sukanya De
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
| | - Subhendu Pramanik
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
| | - Chinmay Chowdhury
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Kolkata 700032, India
| |
Collapse
|
9
|
Ye Q, Wang W, Hao C, Mao X. Agaropentaose protects SH-SY5Y cells against 6-hydroxydopamine-induced neurotoxicity through modulating NF-κB and p38MAPK signaling pathways. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.04.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
10
|
Yuan F, Liu B, Xu Y, Li Y, Sun Q, Xu P, Geng R, Den G, Yang J, Zhang S, Gao L, Liao J, Liu J, Yang X, Tan Y, Chen Q. TIPE3 is a regulator of cell apoptosis in glioblastoma. Cancer Lett 2019; 446:1-14. [DOI: 10.1016/j.canlet.2018.12.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 11/29/2018] [Accepted: 12/27/2018] [Indexed: 12/19/2022]
|
11
|
Chien CM, Yang JC, Wu PH, Wu CY, Chen GY, Wu YC, Chou CK, Tseng CH, Chen YL, Wang LF, Chiu CC. Phytochemical naphtho[1,2-b] furan-4,5‑dione induced topoisomerase II-mediated DNA damage response in human non-small-cell lung cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 54:109-119. [PMID: 30668360 DOI: 10.1016/j.phymed.2018.06.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/18/2018] [Accepted: 06/18/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Phytochemical naphtho[1,2-b] furan-4,5‑dione (NFD) presenting in Avicennia marina exert anti-cancer effects, but little is known regarding about DNA damage-mediated apoptosis in non-small-cell lung carcinoma (NSCLC). PURPOSE To examine whether NFD-induced apoptosis of NSCLC cells is correlated with the induction of DNA damage, and to investigate its underlying mechanism. STUDY DESIGN The anti-proliferative effects of NFD were assessed by MTS Assay Kit FACS assay, and in vivo nude mice xenograft assay. The DNA damage related proteins, the Bcl-2 family and pro-apoptotic factors were examined by immunofluorescence assay, q-PCR, and western blotting. The activity of NF-κB p65 in nuclear extracts was detected using a colorimetric DNA-binding ELISA assay. The inhibitory activity of topoisomerase II (TOPO II) was evaluated by molecular docking and TOPO II catalytic assay. RESULTS NFD exerted selective cytotoxicity against NSCLC H1299, H1437 and A549 cells rather than normal lung-embryonated cells MRC-5. Remarkably, we found that NFD activated the hull marker and modulator of DNA damage repairs such as γ-H2AX, ATM, ATR, CHK1, and CHK2 probably caused by the accumulation of intracellular reactive oxygen species (ROS) and inhibition of TOPO II activity. Furthermore, the suppression of transcription factor NF-κB by NFD resulted in significantly decreased levels of pro-survival proteins including Bcl-2 family Bcl-2, Bcl-xL and Mcl-1 and the endogenous inhibitors of apoptosis XIAP and survivin in H1299 cells. Moreover, the nude mice xenograft assay further validated the suppression of H1299 growth by NFD, which is the first report for evaluating the anti-cancer effect of NFD in vivo. CONCLUSION These findings provide a novel mechanism indicating the inhibition of TOPO II activity and NF-κB signaling by NFD, leading to DNA damage and apoptosis of NSCLC tumor cells.
Collapse
Affiliation(s)
- Ching-Ming Chien
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan; Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung, Taiwan
| | - Juan-Cheng Yang
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan; BioActive Lipid Research Center, BenQ Medical Center, Suzhou, Jiangsu Province, China; Research Center for Natural Products & Drug development, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Pin-Hsuan Wu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chang-Yi Wu
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Guan-Yu Chen
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan
| | - Yang-Chang Wu
- Research Center for Natural Products & Drug development, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chon-Kit Chou
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Hua Tseng
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yeh-Long Chen
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Li-Fang Wang
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan; Center of Excellence for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Cancer Center, Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
12
|
A Novel Aurora-A Inhibitor (MLN8237) Synergistically Enhances the Antitumor Activity of Sorafenib in Hepatocellular Carcinoma. MOLECULAR THERAPY-NUCLEIC ACIDS 2018; 13:176-188. [PMID: 30292139 PMCID: PMC6172479 DOI: 10.1016/j.omtn.2018.08.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/07/2018] [Accepted: 08/19/2018] [Indexed: 02/08/2023]
Abstract
Currently, sorafenib-based therapy is the standard treatment for advanced hepatocellular carcinoma (HCC), and there is a strong rationale for investigating its use in combination with other agents to achieve better therapeutic effects. Aurora-A, a member of a family of mitotic serine/threonine kinases, is frequently overexpressed in human cancers and therefore represents a target for therapy. Here, we investigated a novel Aurora-A inhibitor, MLN8237, together with sorafenib in HCC cells in vitro and in vivo, and elucidated the possible molecular mechanism. Here, it was found that MLN8237 was strongly synergistic with sorafenib in inhibition of HCC progression by altering cell growth, cell-cycle regulation, apoptosis, migration, invasion, and angiogenesis. Mechanism dissection suggests that the combination of MLN8237 and sorafenib led to significant inhibition of the activation of phospho-Akt (p-Akt) and phospho-p38 mitogen-activated protein kinase (p-p38 MAPK) and their downstream genes including CDK4, cyclinD1, and VEGFA. The activators of p-Akt and p-p38 MAPK signaling partially reversed the synergistic inhibitory effects of sorafenib and MLN8237 on HCC progression. Subsequent in vivo studies further confirmed the synergistic effects of sorafenib and MLN8237. Collectively, the newly developed sorafenib-MLN8237 combination may be a novel therapy to better inhibit HCC progression.
Collapse
|
13
|
Cai L, Li CM, Tang WJ, Liu MM, Chen WN, Qiu YY, Li R. Therapeutic Effect of Penta-acetyl Geniposide on Adjuvant-Induced Arthritis in Rats: Involvement of Inducing Synovial Apoptosis and Inhibiting NF-κB Signal Pathway. Inflammation 2018; 41:2184-2195. [DOI: 10.1007/s10753-018-0861-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
14
|
Li Z, Liu P, Zhang H, Zhao S, Jin Z, Li R, Guo Y, Wang X. Role of GABA B receptors and p38MAPK/NF-κB pathway in paclitaxel-induced apoptosis of hippocampal neurons. PHARMACEUTICAL BIOLOGY 2017; 55:2188-2195. [PMID: 29115173 PMCID: PMC6130610 DOI: 10.1080/13880209.2017.1392987] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Revised: 08/21/2017] [Accepted: 09/21/2017] [Indexed: 06/07/2023]
Abstract
CONTEXT The effects of the anticancer drug paclitaxel on learning and memory are rarely studied. OBJECTIVE This study investigated changes in GABAB receptor expression during paclitaxel-induced apoptosis of hippocampal neurons and the role of the p38MAPK/NF-κB pathway in this process. MATERIALS AND METHODS Hippocampal neurons isolated from neonatal Sprague-Dawley rats were divided into six groups: Control (C), SB (10 µL of 10-µmol/L SB203580), SN (53 µg/mL SN50), N (1 µmol/L paclitaxel), SB + N (10 µmol/L SB203580 + 1 µmol/L paclitaxel) and SN + N (53 µg/mL SN50 + 1 µmol/L paclitaxel). Cells in different groups were treated with corresponding agents for 24 h at 37 °C. The apoptosis rate and protein levels of GABAB1 receptors and NF-κB p65 were evaluated. Rat models of neuropathic pain was induced by paclitaxel and were divided into four groups such as N, B + N, SN + N and SN + B + N groups. Rats in the N group received intrathecal injections of normal saline solution. Rats in the B + N group received intrathecal injections of 10 μL baclofen (0.05 μg/μL). Rats in the SN + N and SN + B + N groups received intrathecal injections of SN50 and SN50 plus baclofen, respectively. Spatial learning and memory were evaluated in rat models based on the escape latency and the number of crossings over the platform and protein levels of GABAB1 receptors, NF-κB, IL-1β and TNFα were measured by immunohistochemistry assay and western blot. RESULTS The neuronal apoptosis rate was significantly increased in N (49.16 ± 3.12)%, SB + N (31.18 ± 3.02)% and SN + N (28.47 ± 3.75)% groups, accompanied by increased levels of GABAB1 receptors and NF-κB p65 (p < 0.05). The paclitaxel-treated rats demonstrated significantly increased latency (24.32 ± 2.94)s and decreased the crossings number (3.14 ± 0.63) after 15 d in the Morris water maze (p < 0.05). Immunohistochemistry assay showed that compared with the N group (GABAB1:9.0 ± 1.6, NF-κB p65:29.6 ± 2.4, IL-1β: 30.4 ± 3.4, TNFα: 31.0 ± 3.4), B + N, SN + N and SN + B + N groups evidently increased levels of GABAB1 receptor (B + N:SN + N:SN + B + N = 19.4 ± 2.1:20.8 ± 1.9:28.0 ± 1.9) but significantly decreased levels of NF-κB p65 (B + N:SN + N:SN + B + N = 21.2 ± 1.5:18.6 ± 2.1:12.6 ± 1.5), IL-1β (B + N:SN + N:SN + B + N = 22.0 ± 1.0:19.6 ± 1.8:14.6 ± 1.5) and TNF α (B + N:SN + N:SN + B + N = 23.0 ± 1.6:22.2 ± 0.8:16.6 ± 1.7). Similar findings were found in western blot analysis. DISCUSSIONS AND CONCLUSIONS Paclitaxel may reduce cognitive function in rats through the p38MAPK/NF-κB pathway and GABAB1 receptors.
Collapse
Affiliation(s)
- Zhao Li
- Department of Anesthesiology, The Third Hospital of HeBei Medical University, Shijiazhuang, HeBei Province, China
| | - Peng Liu
- Department of Anesthesiology, The Third Hospital of HeBei Medical University, Shijiazhuang, HeBei Province, China
| | - Hailin Zhang
- Department of Pharmacology, HeBei Medical University, Shijiazhuang, HeBei Province, China
| | - Shuang Zhao
- Department of Anesthesiology, The Third Hospital of HeBei Medical University, Shijiazhuang, HeBei Province, China
| | - Zi Jin
- Department of Anesthesiology, The Third Hospital of HeBei Medical University, Shijiazhuang, HeBei Province, China
| | - Rui Li
- Department of Anesthesiology, The Third Hospital of HeBei Medical University, Shijiazhuang, HeBei Province, China
| | - Yuexian Guo
- Department of Anesthesiology, The Third Hospital of HeBei Medical University, Shijiazhuang, HeBei Province, China
| | - Xiuli Wang
- Department of Anesthesiology, The Third Hospital of HeBei Medical University, Shijiazhuang, HeBei Province, China
| |
Collapse
|
15
|
The quinone-based derivative, HMNQ induces apoptotic and autophagic cell death by modulating reactive oxygen species in cancer cells. Oncotarget 2017; 8:99637-99648. [PMID: 29245930 PMCID: PMC5725121 DOI: 10.18632/oncotarget.21005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 08/28/2017] [Indexed: 12/17/2022] Open
Abstract
8-Hydroxy-2-methoxy-1,4-naphthoquinone (HMNQ), a natural compound isolated from the bark of Juglans sinensis Dode, displays cytotoxic activity against various human cancer cells. However, the molecular mechanism of the anticancer effect is unclear. In this study, we examined the cytotoxic mechanism of HMNQ at the molecular level in human cancer cells. Cells were treated with HMNQ in a dose- or time-dependent manner. HMNQ treatment inhibited cell viability, colony formation and cell migration, indicating that HMNQ induced cancer cell death. HMNQ-treated cells resulted in apoptotic cell death through PARP-1 cleavage, Bax upregulation and Bcl-2 downregulation. HMNQ was also observed to induce autophagy by upregulating Beclin-1 and LC3. Furthermore, HMNQ induced reactive oxygen species (ROS) production, which was attenuated by the ROS scavengers, NAC and GSH. Finally, HMNQ increased expression of JNK phosphorylation and the JNK inhibitor SP600125 rescued HMNQ-induced cell death, suggesting that the cytotoxicity of HMNQ is mediated by the JNK signaling pathway. Taken together, our findings show that HMNQ exhibits anticancer activity through induction of ROS-mediated apoptosis and autophagy in human cancer cells. These data suggest the potential value of HMNQ as a natural anticancer drug.
Collapse
|
16
|
Yang SC, Yen FL, Wang PW, Aljuffali IA, Weng YH, Tseng CH, Fang JY. Naphtho[1,2-b]furan-4,5-dione is a potent anti-MRSA agent against planktonic, biofilm and intracellular bacteria. Future Microbiol 2017; 12:1059-1073. [DOI: 10.2217/fmb-2017-0044] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Aim: Naphtho[1,2-b]furan-4,5-dione (N12D) and naphtho[2,3-b]furan-4,9-dione (N23D) are furanonaphthoquinone derivatives from natural resources. We examined the antimicrobial activity of N12D and N23D against drug-resistant Staphylococcus aureus. Materials & methods: Minimum inhibitory concentration, minimum bactericidal concentration, bacterial viability and agar diffusion assay were conducted against methicillin-resistant S. aureus (MRSA) and clinical isolates of vancomycin-resistant S. aureus. Results & conclusion: The minimum inhibitory concentration of N12D and N23D against MRSA was 4.9–9.8 and 39 μM, respectively. With regard to the agar diffusion test, the inhibition zone of the quinone compounds was threefold larger than that of oxacillin. N12D was found to inhibit MRSA biofilm thickness from 24 to 16 μm as observed by confocal microscopy. N12D showed a significant reduction of the intracellular MRSA burden without decreasing the macrophage viability. The antibacterial mechanisms of N12D may be bacterial wall/membrane damage and disturbance of gluconeogenesis and the tricarboxylic acid cycle.
Collapse
Affiliation(s)
- Shih-Chun Yang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan 333, Taiwan
| | - Feng-Lin Yen
- Department of Fragrance & Cosmetic Science, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Pei-Wen Wang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan
| | - Ibrahim A Aljuffali
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Yi-Han Weng
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan 333, Taiwan
| | - Chih-Hua Tseng
- Department of Fragrance & Cosmetic Science, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Research Center for Natural Products & Drug Development, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center for Infectious Disease & Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan 333, Taiwan
- Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Kweishan, Taoyuan 333, Taiwan
- Research Center for Food & Cosmetic Safety & Research Center for Chinese Herbal Medicine, Chang Gung University of Science & Technology, Taoyuan 333, Taiwan
- Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan 333, Taiwan
| |
Collapse
|
17
|
Fong Y, Wu CY, Chang KF, Chen BH, Chou WJ, Tseng CH, Chen YC, Wang HMD, Chen YL, Chiu CC. Dual roles of extracellular signal-regulated kinase (ERK) in quinoline compound BPIQ-induced apoptosis and anti-migration of human non-small cell lung cancer cells. Cancer Cell Int 2017; 17:37. [PMID: 28286419 PMCID: PMC5339964 DOI: 10.1186/s12935-017-0403-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 02/16/2017] [Indexed: 02/06/2023] Open
Abstract
Background 2,9-Bis[2-(pyrrolidin-1-yl)ethoxy]-6-{4-[2-(pyrrolidin-1-yl)ethoxy] phenyl}-11H-indeno[1,2-c]quinoline-11-one (BPIQ), is a synthetic quinoline analog. A previous study showed the anti-cancer potential of BPIQ through modulating mitochondrial-mediated apoptosis. However, the effect of BPIQ on cell migration, an index of cancer metastasis, has not yet been examined. Furthermore, among signal pathways involved in stresses, the members of the mitogen-activated protein kinase (MAPK) family are crucial for regulating the survival and migration of cells. In this study, the aim was to explore further the role of MAPK members, including JNK, p38 and extracellular signal-regulated kinase (ERK) in BPIQ-induced apoptosis and anti-migration of human non-small cell lung cancer (NSCLC) cells. Methods Western Blot assay was performed for detecting the activation of MAPK members in NSCLC H1299 cells following BPIQ administration. Cellular proliferation was determined using a trypan blue exclusion assay. Cellular apoptosis was detected using flow cytometer-based Annexin V/propidium iodide dual staining. Cellular migration was determined using wound-healing assay and Boyden’s chamber assay. Zymography assay was performed for examining MMP-2 and -9 activities. The assessment of MAPK inhibition was performed for further validating the role of JNK, p38, and ERK in BPIQ-induced growth inhibition, apoptosis, and migration of NSCLC cells. Results Western Blot assay showed that BPIQ treatment upregulates the phosphorylated levels of both MAPK proteins JNK and ERK. However, only ERK inhibitor rescues BPIQ-induced growth inhibition of NSCLC H1299 cells. The results of Annexin V assay further confirmed the pro-apoptotic role of ERK in BPIQ-induced cell death of H1299 cells. The results of wound healing and Boyden chamber assays showed that sub-IC50 (sub-lethal) concentrations of BPIQ cause a significant inhibition of migration in H1299 cells accompanied with downregulating the activity of MMP-2 and -9, the motility index of cancer cells. Inhibition of ERK significantly enhances BPIQ-induced anti-migration of H1299 cells. Conclusions Our results suggest ERK may play dual roles in BPIQ-induced apoptosis and anti-migration, and it would be worthwhile further developing strategies for treating chemoresistant lung cancers through modulating ERK activity. Electronic supplementary material The online version of this article (doi:10.1186/s12935-017-0403-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yao Fong
- Department of Thoracic Surgery, Chi-Mei Medical Center, Tainan, 710 Taiwan
| | - Chang-Yi Wu
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, 804 Taiwan.,Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807 Taiwan
| | - Kuo-Feng Chang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807 Taiwan
| | - Bing-Hung Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807 Taiwan.,The Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804 Taiwan
| | - Wan-Ju Chou
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807 Taiwan
| | - Chih-Hua Tseng
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung, 807 Taiwan
| | - Yen-Chun Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807 Taiwan
| | - Hui-Min David Wang
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung, 402 Taiwan
| | - Yeh-Long Chen
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, 807 Taiwan
| | - Chien-Chih Chiu
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, 804 Taiwan.,Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807 Taiwan.,Translational Research Center, Cancer Center and Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 807 Taiwan.,Research Center for Environment Medicine, Kaohsiung Medical University, Kaohsiung, 807 Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807 Taiwan
| |
Collapse
|
18
|
Tombul M, Bulut A, Türk M, Uçar B, Işılar Ö. Synthesis and biological activity of ferrocenyl furoyl derivatives. INORG NANO-MET CHEM 2017. [DOI: 10.1080/15533174.2016.1218510] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Mustafa Tombul
- Department of Chemistry, Faculty of Art and Science, Kırıkkale University, Kırıkkale, Turkey
| | - Adnan Bulut
- Department of Chemistry, Faculty of Art and Science, Kırıkkale University, Kırıkkale, Turkey
| | - Mustafa Türk
- Department of Bioengineering, Faculty of Engineering, Kırıkkale University, Kırıkkale, Turkey
| | - Büşra Uçar
- Department of Chemistry, Faculty of Art and Science, Kırıkkale University, Kırıkkale, Turkey
| | - Özer Işılar
- Department of Chemistry, Faculty of Art and Science, Kırıkkale University, Kırıkkale, Turkey
| |
Collapse
|
19
|
Bai ZT, Wu ZR, Xi LL, Li X, Chen P, Wang FQ, Meng WB, Zhou WC, Wu XA, Yao XJ, Zhang M. Inhibition of invasion by N-trans-feruloyloctopamine via AKT, p38MAPK and EMT related signals in hepatocellular carcinoma cells. Bioorg Med Chem Lett 2016; 27:989-993. [PMID: 28073674 DOI: 10.1016/j.bmcl.2016.12.073] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/20/2016] [Accepted: 12/28/2016] [Indexed: 01/04/2023]
Abstract
N-trans-feruloyloctopamine (FO) isolated from Garlic skin was identified as the primary antioxidant constituents, however, the effect of which on HCC invasion is still unclear. Herein, the FO was synthesized and its antitumor activities were evaluated in HCC cell lines. Cellular functional analyses have revealed that the reformed FO owns strong abilities of inhibiting cell proliferation and invasion in HCC cells. Molecular data have further showed that FO could significantly decrease the phosphorylation levels of Akt and p38 MAPK. In addition, the expression of Slug was inhibited and the level of E-cadherin increased. Molecular docking analysis indicates that the H-bond and hydrophobic interactions were critical for FO and E-cadherin binding, but FO did not seem to act directly on phosphorylated Akt and p38 MAPK. We have thus concluded that reformed FO inhibits cell invasion might be directly through EMT related signals (E-cadherin) and indirectly through PI3K/Akt, p38 MAPK signaling pathways. FO might be a promising drug in HCC treatment and prognosis.
Collapse
Affiliation(s)
- Zhong-Tian Bai
- The Second Department of General Surgery, The First Hospital of Lanzhou University, Key Laboratory of Biotherapy and Regenerative Medicine, Gansu Province, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, Gansu, China
| | - Zheng-Rong Wu
- School of Pharmacy, Lanzhou University, Lanzhou 730020, China
| | - Li-Li Xi
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Xun Li
- The Second Department of General Surgery, The First Hospital of Lanzhou University, Key Laboratory of Biotherapy and Regenerative Medicine, Gansu Province, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, Gansu, China.
| | - Peng Chen
- School of Pharmacy, Lanzhou University, Lanzhou 730020, China
| | - Fu-Qiang Wang
- The Second Department of General Surgery, The First Hospital of Lanzhou University, Key Laboratory of Biotherapy and Regenerative Medicine, Gansu Province, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, Gansu, China
| | - Wen-Bo Meng
- The Second Department of General Surgery, The First Hospital of Lanzhou University, Key Laboratory of Biotherapy and Regenerative Medicine, Gansu Province, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, Gansu, China
| | - Wen-Ce Zhou
- The Second Department of General Surgery, The First Hospital of Lanzhou University, Key Laboratory of Biotherapy and Regenerative Medicine, Gansu Province, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, Gansu, China
| | - Xin-An Wu
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Xiao-Jun Yao
- Department of Chemistry, Lanzhou University, Lanzhou 730000, China
| | - Min Zhang
- Gansu Provincial Hospital, Lanzhou 730000, China
| |
Collapse
|
20
|
Decreased expression of PBLD correlates with poor prognosis and functions as a tumor suppressor in human hepatocellular carcinoma. Oncotarget 2016; 7:524-37. [PMID: 26594798 PMCID: PMC4808015 DOI: 10.18632/oncotarget.6358] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 11/14/2015] [Indexed: 12/11/2022] Open
Abstract
Recent accumulating genomic and proteomic data suggested that decreased expression of phenazine biosynthesis-like domain-containing protein (PBLD) was frequently involved in hepatocellular carcinoma (HCC). However, there is lack of systematical investigation focusing on its expression pattern, clinical relevance, and biological function. Here, we found that PBLD was frequently decreased in HCC tissues relative to adjacent non-tumorigenic liver tissues. This decreased expression was significantly associated with poor tumor differentiation and advanced tumor stage. Kaplan–Meier analysis further showed that recurrence-free survival and overall survival were significantly worse among patients with low PBLD expression. Moreover, multivariate analyses revealed that PBLD was an independent predictor of OS and RFS. This prognostic value of PBLD was further validated in another independent cohort. We also found PBLD inhibited HCC cell growth and invasion in vitro and tumor growth in vivo. Furthermore, forced expression of PBLD influenced multiple downstream genes related to MAPK, NF-κB, EMT, and angiogenesis signaling pathways. PBLD deletion was an independent predictor of poor prognosis in patients with HCC. Elevated PBLD expression may reduce HCC cell growth and invasion via inactivation of several tumorigenesis-related signaling pathways.
Collapse
|
21
|
An Acetamide Derivative as a Camptothecin Sensitizer for Human Non-Small-Cell Lung Cancer Cells through Increased Oxidative Stress and JNK Activation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:9128102. [PMID: 27843533 PMCID: PMC5098095 DOI: 10.1155/2016/9128102] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 08/31/2016] [Indexed: 11/17/2022]
Abstract
In recent years, combination chemotherapy is a primary strategy for treating lung cancer; however, the issues of antagonism and side effects still limit its applications. The development of chemosensitizer aims to sensitize chemoresistant cancer cells to anticancer drugs and therefore improve the efficacy of chemotherapy. In this study, we examined whether N-[2-(morpholin-4-yl)phenyl]-2-{8-oxatricyclo[7.4.0.0,2,7]trideca-1(9),2(7),3,5,10,12-hexaen-4-yloxy}acetamide (NPOA), an acetamide derivative, sensitizes human non-small-cell lung cancer (NSCLC) H1299 cells towards camptothecin- (CPT-) induced apoptosis effects. Our results demonstrate that the combination of CPT and NPOA enhances anti-lung-cancer effect. The cytometer-based Annexin V/propidium iodide (PI) staining showed that CPT and NPOA cotreatment causes an increased population of apoptotic cells compared to CPT treatment alone. Moreover, Western blotting assay showed an enhancement of Bax expression and caspase cascade leading to cell death of H1299 cells. Besides, CPT and NPOA cotreatment-mediated disruption of mitochondrial membrane potential (MMP) in H1299 cells may function through increasing the activation of the stressed-associated c-Jun N-terminal kinase (JNK). These results showed that NPOA treatment sensitizes H1299 cells towards CPT-induced accumulation of cell cycle S phase and mitochondrial-mediated apoptosis through regulating endogenous ROS and JNK activation. Accordingly, NPOA could be a candidate chemosensitizer of CPT derivative agents such as irinotecan or topotecan in the future.
Collapse
|
22
|
Fong Y, Tang CC, Hu HT, Fang HY, Chen BH, Wu CY, Yuan SS, Wang HMD, Chen YC, Teng YN, Chiu CC. Inhibitory effect of trans-ferulic acid on proliferation and migration of human lung cancer cells accompanied with increased endogenous reactive oxygen species and β-catenin instability. Chin Med 2016; 11:45. [PMID: 27733866 PMCID: PMC5045596 DOI: 10.1186/s13020-016-0116-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 09/19/2016] [Indexed: 12/13/2022] Open
Abstract
Background Trans-ferulic (FA) acid exhibits antioxidant effects in vitro. However, the underlying mechanism of trans-FA activity in cellular physiology, especially cancer physiology, remains largely unknown. This study investigated the cellular physiological effects of trans-FA on the H1299 human lung cancer cell line. Methods The 2,2-diphenyl-1-picrylhydrazyl assay was used to determine free radical scavenging capability. Assessment of intracellular reactive oxygen species (ROS) was evaluated using oxidized 2ʹ,7ʹ-dichlorofluorescin diacetate and dihydroethidium staining. Trypan blue exclusion, colony formation, and anchorage-independent growth assays were used to determine cellular proliferation. Annexin V staining assay was used to assess cellular apoptosis by flow cytometry. Wound healing and Boyden’s well assays were used to detect the migration and invasion of cells. Gelatin zymography was used to detect matrix metalloproteinase (MMP-2 and MMP-9) activity. Western blotting was used to detect expression levels of various signaling pathway proteins. Results DPPH assay results indicated that trans-FA exerted potent antioxidant effects. However, trans-FA increased intracellular ROS levels, including hydrogen peroxide and superoxide anion, in H1299 cells. Trans-FA treatment inhibited cellular proliferation and induced moderate apoptotic cell death at the highest concentration used (0.6 mM). Furthermore, trans-FA moderately inhibited the migration of H1299 cells at the concentrations of 0.3 and 0.6 mM and attenuated MMP-2 and MMP-9 activity. Trans-FA caused the phosphorylation of β-catenin, resulting in proteasomal degradation of β-catenin. Conversely, trans-FA treatment increased the expression of pro-apoptotic factor Bax and decreased the expression of pro-survival factor survivin. Conclusion Various concentrations (0.06–0.6 mM) of trans-FA exert both anti-proliferation and anti-migration effects in the human lung cancer cell line H1299. Electronic supplementary material The online version of this article (doi:10.1186/s13020-016-0116-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yao Fong
- Department of Thoracic Surgery, Chi-Mei Medical Center, Tainan, 710 Taiwan
| | - Chia-Chun Tang
- Division of Chest, Ten Chan General Hospital, Chung-Li, 320 Taiwan, ROC
| | - Huei-Ting Hu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807 Taiwan
| | - Hsin-Yu Fang
- Department of Food Nutrition, Chung-Hwa University of Medical Technology, Tainan, 701 Taiwan
| | - Bing-Hung Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807 Taiwan.,The Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804 Taiwan
| | - Chang-Yi Wu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807 Taiwan.,Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, 804 Taiwan
| | - Shyng-Shiou Yuan
- Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung, 807 Taiwan
| | - Hui-Min David Wang
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung, 402 Taiwan
| | - Yen-Chun Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807 Taiwan
| | - Yen-Ni Teng
- Department of Biological Sciences and Technology, National University of Tainan, Tainan, 700 Taiwan
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807 Taiwan.,Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, 804 Taiwan.,Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung, 807 Taiwan.,Research Center for Environment Medicine, Kaohsiung Medical University, Kaohsiung, 807 Taiwan
| |
Collapse
|
23
|
Ramanathan D, Namitharan K, Pitchumani K. Copper(i)–Y zeolite catalyzed N-sulfonylketenimine mediated annulation of hydroxynaphthoquinones: syntheses of naphtho[2,1-b]furan-2,5-diones and benzo[de]chromene-2,6-diones. Chem Commun (Camb) 2016; 52:8436-9. [DOI: 10.1039/c6cc03571j] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
An efficient, one pot synthesis of novel naphtho[2,1-b]furan-2,5-diones and benzo[de]chromene-2,6-diones was reported using copper(i)–Y zeolite as catalyst.
Collapse
Affiliation(s)
| | | | - Kasi Pitchumani
- School of Chemistry
- Madurai Kamaraj University
- Madurai – 625 021
- India
- Centre for Green Chemistry Processes
| |
Collapse
|
24
|
Chiu CC, Chou HL, Chen BH, Chang KF, Tseng CH, Fong Y, Fu TF, Chang HW, Wu CY, Tsai EM, Lin SR, Chen YL. BPIQ, a novel synthetic quinoline derivative, inhibits growth and induces mitochondrial apoptosis of lung cancer cells in vitro and in zebrafish xenograft model. BMC Cancer 2015; 15:962. [PMID: 26672745 PMCID: PMC4682281 DOI: 10.1186/s12885-015-1970-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 12/01/2015] [Indexed: 11/30/2022] Open
Abstract
Background 2,9-Bis[2-(pyrrolidin-1-yl)ethoxy]-6-{4-[2-(pyrrolidin-1-yl)ethoxy] phenyl}-11H-indeno[1,2-c]quinolin-11-one (BPIQ) is a derivative from 6-arylindeno[1,2-c]quinoline. Our previous study showed the anti-cancer potential of BPIQ compared to its two analogues topotecan and irinotecan. In the study, the aim is to investigate the potency and the mechanism of BPIQ against lung cancer cells. Methods Both in vitro and zebrafish xenograft model were performed to examine the anti-lung cancer effect of BPIQ. Flow cytometer-based assays were performed for detecting apoptosis and cell cycle distribution. Western blot assay was used for detecting the changes of apoptotic and cell cycle-associated proteins. siRNA knockdown assay was performed for confirming the apoptotic role of Bim. Results Both in vitro and zebrafish xenograft model demonstrated the anti-lung cancer effect of BPIQ. BPIQ-induced proliferative inhibition of H1299 cells was achieved through the induction of G2/M-phase arrest and apoptosis. The results of Western blot showed that BPIQ-induced G2/M-phase arrest was associated with a marked decrease in the protein levels of cyclin B and cyclin-dependent kinase 1 (CDK1). The up-regulation of pro-apoptotic Bad, Bim and down-regulation of pro-survival XIAP and survivin was observed following BPIQ treatment. Conclusions BPIQ-induced anti-lung cancer is involved in mitochondrial apoptosis. BPIQ could be a promising anti-lung cancer drug for further applications. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1970-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan. .,Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan. .,Translational Research Center, Cancer Center, Department of Medical Research, and Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 807, Taiwan. .,Research Center for Environment Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan. .,Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung, Taiwan.
| | - Han-Lin Chou
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan. .,Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung, Taiwan.
| | - Bing-Hung Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan. .,Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung, Taiwan.
| | - Kuo-Feng Chang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| | - Chih-Hua Tseng
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung, 807, Taiwan. .,Research Center for Environment Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| | - Yao Fong
- Department of Thoracic Surgery, Chi-Mei Medical Center, Tainan, 710, Taiwan.
| | - Tzu-Fun Fu
- Department of Medical Laboratory Science and Biotechnology, School of Medicine, National Cheng Kung University, Tainan, 701, Taiwan.
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University; Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| | - Chang-Yi Wu
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan.
| | - Eing-Mei Tsai
- Research Center for Environment Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| | - Shinne-Ren Lin
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| | - Yeh-Long Chen
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| |
Collapse
|
25
|
Shi M, Ren X, Wang X, Wang H, Liu G, Yuan X, Zheng S, Yu L, Pan S, Song G, Guo Q, Li L, Zhang X, Zhang Z, Ding H, Jiang G. A novel combination of oridonin and valproic acid in enhancement of apoptosis induction of HL-60 leukemia cells. Int J Oncol 2015; 48:734-46. [PMID: 26676928 DOI: 10.3892/ijo.2015.3294] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 11/27/2015] [Indexed: 11/06/2022] Open
Abstract
Oridonin, obtained from the traditional Chinese herbal medicine rabdosia rubescens, exerts potent antitumor activities in cancer cells. Valproic acid (VPA), as a potent histone deacetylase inhibitor (HDACI), also plays an important role in inhibition of proliferation of tumor cells. However, there are no reports so far on the cooperation between oridonin and VPA for anti-leukemic effect. Therefore, in the present study, we undertook experiments to determine whether lower concentration of oridonin in conjunction with lower concentration of VPA would produce even more encouraging synergistic effect than each of them alone, and to clarify its molecular mechanism. The results demonstrated that the lower concentration of oridonin in combination with lower concentration of VPA synergistically inhibited the proliferation of HL-60 cells, and induced obvious caspase-dependent apoptosis through activation of the intrinsic apoptosis pathway, which is involved in the downregulation of Bcl-2/Bax ratio, release of cytochrome c to cytosol and caspase-9 activation, as well as through the extrinsic apoptosis pathway mediated by Fas/FasL and caspase-8 activation. In addition, MAPK signaling pathway was also involved in apoptosis induced by oridonin plus VPA. Furthermore, the combination treatment in vivo remarkably reduced the xenograft tumor size and triggered tumor cell apoptosis. Taken together, the novel combination of oridonin plus VPA exerted synergistic anti-proliferative and apoptosis-inducing effects on human myeloid leukemia cells, and may serve as a potential promising anti-leukemia strategy.
Collapse
Affiliation(s)
- Meiyan Shi
- Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Key Laboratory for Tumor Immunology and Chinese Medicine Immunology of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan University, Jinan, Shandong, P.R. China
| | - Xia Ren
- Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Key Laboratory for Tumor Immunology and Chinese Medicine Immunology of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan University, Jinan, Shandong, P.R. China
| | - Xidi Wang
- Laboratory Department, People's Hospital of Zhangqiu City, Zhangqiu, Shandong, P.R. China
| | - Hengxiao Wang
- Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Key Laboratory for Tumor Immunology and Chinese Medicine Immunology of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan University, Jinan, Shandong, P.R. China
| | - Guoqiang Liu
- Department of Hematology, Shengli Oilfield Central Hospital, Dongying, Shandong, P.R. China
| | - Xiaofen Yuan
- Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Key Laboratory for Tumor Immunology and Chinese Medicine Immunology of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan University, Jinan, Shandong, P.R. China
| | - Shubo Zheng
- General Surgery Department, People's Hospital of Wenshang County, Wenshang, Shandong, P.R. China
| | - Linchang Yu
- Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Key Laboratory for Tumor Immunology and Chinese Medicine Immunology of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan University, Jinan, Shandong, P.R. China
| | - Sufei Pan
- Department of Hematology, Shengli Oilfield Central Hospital, Dongying, Shandong, P.R. China
| | - Guanhua Song
- Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Key Laboratory for Tumor Immunology and Chinese Medicine Immunology of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan University, Jinan, Shandong, P.R. China
| | - Qiang Guo
- Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Key Laboratory for Tumor Immunology and Chinese Medicine Immunology of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan University, Jinan, Shandong, P.R. China
| | - Lianlian Li
- Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Key Laboratory for Tumor Immunology and Chinese Medicine Immunology of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan University, Jinan, Shandong, P.R. China
| | - Xiaoyu Zhang
- Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Key Laboratory for Tumor Immunology and Chinese Medicine Immunology of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan University, Jinan, Shandong, P.R. China
| | - Zhiyong Zhang
- Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Key Laboratory for Tumor Immunology and Chinese Medicine Immunology of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan University, Jinan, Shandong, P.R. China
| | - Huifang Ding
- Department of Hematology, Shengli Oilfield Central Hospital, Dongying, Shandong, P.R. China
| | - Guosheng Jiang
- Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Key Laboratory for Tumor Immunology and Chinese Medicine Immunology of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan University, Jinan, Shandong, P.R. China
| |
Collapse
|
26
|
2-Methoxy-1,4-naphthoquinone (MNQ) induces apoptosis of A549 lung adenocarcinoma cells via oxidation-triggered JNK and p38 MAPK signaling pathways. Life Sci 2015; 135:158-64. [DOI: 10.1016/j.lfs.2015.03.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 02/23/2015] [Accepted: 03/20/2015] [Indexed: 11/21/2022]
|
27
|
BubR1 Acts as a Promoter in Cellular Motility of Human Oral Squamous Cancer Cells through Regulating MMP-2 and MMP-9. Int J Mol Sci 2015; 16:15104-17. [PMID: 26151845 PMCID: PMC4519889 DOI: 10.3390/ijms160715104] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 06/10/2015] [Accepted: 06/15/2015] [Indexed: 11/16/2022] Open
Abstract
BubR1 is a critical component of spindle assembly checkpoint, ensuring proper chromatin segregation during mitosis. Recent studies showed that BubR1 was overexpressed in many cancer cells, including oral squamous cell carcinomas (OSCC). However, the effect of BubR1 on metastasis of OSCC remains unclear. This study aimed to unravel the role of BubR1 in the progression of OSCC and confirm the expression of BubR1 in a panel of malignant OSCC cell lines with different invasive abilities. The results of quantitative real-time PCR showed that the mRNA level of BubR1 was markedly increased in four OSCC cell lines, Ca9-22, HSC3, SCC9 and Cal-27 cells, compared to two normal cells, normal human oral keratinocytes (HOK) and human gingival fibroblasts (HGF). Moreover, the expression of BubR1 in these four OSCC cell lines was positively correlated with their motility. Immunofluorescence revealed that BubR1 was mostly localized in the cytosol of human gingival carcinoma Ca9-22 cells. BubR1 knockdown significantly decreased cellular invasion but slightly affect cellular proliferation on both Ca9-22 and Cal-27 cells. Consistently, the activities of metastasis-associated metalloproteinases MMP-2 and MMP-9 were attenuated in BubR1 knockdown Ca9-22 cells, suggesting the role of BubR1 in promotion of OSCC migration. Our present study defines an alternative pathway in promoting metastasis of OSCC cells, and the expression of BubR1 could be a prognostic index in OSCC patients.
Collapse
|
28
|
Role of mimic of manganese superoxide dismutase in proliferation and apoptosis of gastric carcinoma BGC-823 cells in vitro and in vivo. Int Immunopharmacol 2015; 26:277-85. [DOI: 10.1016/j.intimp.2015.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 03/30/2015] [Accepted: 04/01/2015] [Indexed: 11/23/2022]
|
29
|
Polypeptide Fraction from Arca subcrenata Induces Apoptosis and G2/M Phase Arrest in HeLa Cells via ROS-Mediated MAPKs Pathways. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:930249. [PMID: 26089952 PMCID: PMC4454769 DOI: 10.1155/2015/930249] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 04/30/2015] [Indexed: 01/06/2023]
Abstract
Arca subcrenata is documented in the literature of marine Traditional Chinese Medicine. Polypeptide fraction from A. subcrenata, coded as P2, was demonstrated to possess significant antitumor activity in our previous study. However, the underlying mechanism remains undefined. The present study was carried out to investigate the underlying antitumor mechanism of P2 in human cervical cancer HeLa cells by MTT, FCM, LSCM, and western blot assays. The results revealed that P2 significantly induced apoptosis of HeLa cells in a concentration- and time-dependent manner. High level of ROS was provoked by P2, which was in turn responsible for induction of apoptosis through activation of intrinsic mitochondrial pathway and JNK1/2, p38 MAPK pathways, as well as inhibition of ERK1/2 pathway, as evidenced by the abrogation of P2's effect on HeLa cells preincubated with the ROS scavenger NAC. P2 also was observed to display significant effect on G2/M phase arrest by downregulating the expression of cyclin B1/cdc2 complex and upregulating the expression of p21. These findings demonstrate that P2 induces apoptosis and G2/M phase arrest in HeLa cells through ROS-mediated MAPKs pathways, suggesting that P2 would be worth investigating as a promising agent within the scope of marine drugs for treatment of cervical cancer.
Collapse
|
30
|
Tseng CH, Tzeng CC, Chiu CC, Hsu CY, Chou CK, Chen YL. Discovery of 2-[2-(5-nitrofuran-2-yl)vinyl]quinoline derivatives as a novel type of antimetastatic agents. Bioorg Med Chem 2015; 23:141-8. [DOI: 10.1016/j.bmc.2014.11.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 11/10/2014] [Accepted: 11/11/2014] [Indexed: 12/12/2022]
|
31
|
Wang YH, Xu XJ, Li HL. Hepatoprotective effects of Mimic of Manganese superoxide dismutase against carbon tetrachloride-induced hepatic injury. Int Immunopharmacol 2014; 22:126-32. [DOI: 10.1016/j.intimp.2014.06.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 05/16/2014] [Accepted: 06/10/2014] [Indexed: 11/27/2022]
|
32
|
Ordoñez R, Carbajo-Pescador S, Prieto-Dominguez N, García-Palomo A, González-Gallego J, Mauriz JL. Inhibition of matrix metalloproteinase-9 and nuclear factor kappa B contribute to melatonin prevention of motility and invasiveness in HepG2 liver cancer cells. J Pineal Res 2014; 56:20-30. [PMID: 24117795 DOI: 10.1111/jpi.12092] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 08/30/2013] [Indexed: 12/30/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal human cancers worldwide because of its high incidence and its metastatic potential. Extracellular matrix degradation by matrix metalloproteinases (MMPs) has been connected with cancer cell invasion, and it has been suggested that inhibition of MMPs by synthetic and natural inhibitors may be of great importance in the HCC therapies. Melatonin, the main product of the pineal gland, exerts antiproliferative, proapoptotic, and antiangiogenic properties in HepG2 human hepatocellular cells, and exhibits anti-invasive and antimetastatic activities by suppressing the enzymatic activity of MMP-9 in different tumor types. However, the underlying mechanism of anti-invasive activity in HCC models has not been fully elucidated. Here, we demonstrate that 1 mm melatonin dosage reduced in IL-1β-induced HepG2 cells MMP-9 gelatinase activity and inhibited cell invasion and motility through downregulation of MMP-9 gene expression and upregulation of the MMP-9-specific inhibitor tissue inhibitor of metalloproteinases (TIMP)-1. No significant changes were observed in the expression and activity of MMP-2, the other proteinase implicated in matrix collagen degradation, and its tissue inhibitor, TIMP-2. Also, melatonin significantly suppressed IL-1β-induced nuclear factor-kappaB (NF-κB) translocation and transcriptional activity. In summary, we demonstrate that melatonin modulates motility and invasiveness of HepG2 cell in vitro through a molecular mechanism that involves TIMP-1 upregulation and attenuation of MMP-9 expression and activity via NF-κB signal pathway inhibition.
Collapse
Affiliation(s)
- Raquel Ordoñez
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), León, Spain; Institute of Biomedicine (IBIOMED), University of León, León, Spain
| | | | | | | | | | | |
Collapse
|
33
|
Wang ZS, Luo P, Dai SH, Liu ZB, Zheng XR, Chen T. Salvianolic acid B induces apoptosis in human glioma U87 cells through p38-mediated ROS generation. Cell Mol Neurobiol 2013; 33:921-8. [PMID: 23842993 DOI: 10.1007/s10571-013-9958-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Accepted: 06/27/2013] [Indexed: 10/26/2022]
Abstract
Salvianolic acid B (SalB), the main water-soluble bioactive compounds isolated from the traditional Chinese medical herb Danshen, has been shown to exert anti-cancer effect in several cancer cell lines. The aim of our study was to investigate the potential anti-cancer effect of SalB in human glioma U87 cells. We found that treatment with SalB significantly decreased cell viability of U87 cells in a dose- and time-dependent manner. SalB also enhanced the intracellular ROS generation and induced apoptotic cell death in U87 cells. Western blot analysis suggested that SalB increased the phosphorylation of p38 MAPK and p53 in a dose-dependent manner. Moreover, blocking p38 activation by specific inhibitor SB203580 or p38 specific siRNA partly reversed the anti-proliferative and pro-apoptotic effects, and ROS production induced by SalB treatment. The anti-tumor activity of SalB in vivo was also demonstrated in U87 xenograft glioma model. All of these findings extended the anti-cancer effect of SalB in human glioma cell lines, and suggested that these inhibitory effects of SalB on U87 glioma cell growth might be associated with p38 activation mediated ROS generation. Thus, SalB might be concerned as an effective and safe natural anticancer agent for glioma prevention and treatment.
Collapse
Affiliation(s)
- Zi-shu Wang
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, China,
| | | | | | | | | | | |
Collapse
|
34
|
Li HH, Su JH, Chiu CC, Lin JJ, Yang ZY, Hwang WI, Chen YK, Lo YH, Wu YJ. Proteomic investigation of the sinulariolide-treated melanoma cells A375: effects on the cell apoptosis through mitochondrial-related pathway and activation of caspase cascade. Mar Drugs 2013; 11:2625-42. [PMID: 23880933 PMCID: PMC3736442 DOI: 10.3390/md11072625] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 07/09/2013] [Accepted: 07/10/2013] [Indexed: 12/22/2022] Open
Abstract
Sinulariolide is an active compound isolated from the cultured soft coral Sinularia flexibilis. In this study, we investigated the effects of sinulariolide on A375 melanoma cell growth and protein expression. Sinulariolide suppressed the proliferation and migration of melanoma cells in a concentration-dependent manner and was found to induce both early and late apoptosis by flow cytometric analysis. Comparative proteomic analysis was conducted to investigate the effects of sinulariolide at the molecular level by comparison between the protein profiles of melanoma cells treated with sinulariolide and those without treatment. Two-dimensional gel electrophoresis (2-DE) master maps of control and treated A375 cells were generated by analysis with PDQuest software. Comparison between these maps showed up- and downregulation of 21 proteins, seven of which were upregulated and 14 were downregulated. The proteomics studies described here identify some proteins that are involved in mitochondrial dysfunction and apoptosis-associated proteins, including heat shock protein 60, heat shock protein beta-1, ubiquinol cytochrome c reductase complex core protein 1, isocitrate dehydrogenase (NAD) subunit alpha (down-regulated), and prohibitin (up-regulated), in A375 melanoma cells exposed to sinulariolide. Sinulariolide-induced apoptosis is relevant to mitochondrial-mediated apoptosis via caspase-dependent pathways, elucidated by the loss of mitochondrial membrane potential, release of cytochrome c, and activation of Bax, Bad and caspase-3/-9, as well as suppression of p-Bad, Bcl-xL and Bcl-2. Taken together, our results show that sinulariolide-induced apoptosis might be related to activation of the caspase cascade and mitochondria dysfunction pathways. Our results suggest that sinulariolide merits further evaluation as a chemotherapeutic agent for human melanoma.
Collapse
Affiliation(s)
- Hsing-Hui Li
- National Museum of Marine Biology and Aquarium, Pingtung 94450, Taiwan; E-Mails: (H.-H.L.); (J.-H.S.)
| | - Jui-Hsin Su
- National Museum of Marine Biology and Aquarium, Pingtung 94450, Taiwan; E-Mails: (H.-H.L.); (J.-H.S.)
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 80761, Taiwan; E-Mail:
| | - Jen-Jie Lin
- Graduate Institute of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 91202, Taiwan; E-Mail:
| | - Zih-Yan Yang
- Graduate Institute of Food Science, National Pingtung University of Science and Technology, Pingtung 91202, Taiwan; E-Mail:
| | - Wen-Ing Hwang
- Department of Food Science and Nutrition, Meiho University, Pingtung 91202, Taiwan; E-Mails: (W.-I.H.); (Y.-K.C.)
| | - Yu-Kuei Chen
- Department of Food Science and Nutrition, Meiho University, Pingtung 91202, Taiwan; E-Mails: (W.-I.H.); (Y.-K.C.)
| | - Yu-Hsuan Lo
- Excellence Biotech Co., Kaohsiung 80655, Taiwan; E-Mail:
| | - Yu-Jen Wu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 80761, Taiwan; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +886-8-7799821 (ext. 8613); Fax: +886-8-7797821
| |
Collapse
|
35
|
Inhibition of VEGF expression through blockade of Hif1α and STAT3 signalling mediates the anti-angiogenic effect of melatonin in HepG2 liver cancer cells. Br J Cancer 2013; 109:83-91. [PMID: 23756865 PMCID: PMC3708553 DOI: 10.1038/bjc.2013.285] [Citation(s) in RCA: 186] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 05/13/2013] [Accepted: 05/15/2013] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) growth relies on angiogenesis via vascular endothelial growth factor (VEGF) release. Hypoxia within tumour environment leads to intracellular stabilisation of hypoxia inducible factor 1 alpha (Hif1α) and signal transducer and activator of transcription (STAT3). Melatonin induces apoptosis in HCC, and shows anti-angiogenic features in several tumours. In this study, we used human HepG2 liver cancer cells as an in vitro model to investigate the anti-angiogenic effects of melatonin. METHODS HepG2 cells were treated with melatonin under normoxic or CoCl2-induced hypoxia. Gene expression was analysed by RT-qPCR and western blot. Melatonin-induced anti-angiogenic activity was confirmed by in vivo human umbilical vein endothelial cells (HUVECs) tube formation assay. Secreted VEGF was measured by ELISA. Immunofluorescence was performed to analyse Hif1α cellular localisation. Physical interaction between Hif1α and its co-activators was analysed by immunoprecipitation and chromatin immunoprecipitation (ChIP). RESULTS Melatonin at a pharmacological concentration (1 mM) decreases cellular and secreted VEGF levels, and prevents HUVECs tube formation under hypoxia, associated with a reduction in Hif1α protein expression, nuclear localisation, and transcriptional activity. While hypoxia increases phospho-STAT3, Hif1α, and CBP/p300 recruitment as a transcriptional complex within the VEGF promoter, melatonin 1 mM decreases their physical interaction. Melatonin and the selective STAT3 inhibitor Stattic show a synergic effect on Hif1α, STAT3, and VEGF expression. CONCLUSION Melatonin exerts an anti-angiogenic activity in HepG2 cells by interfering with the transcriptional activation of VEGF, via Hif1α and STAT3. Our results provide evidence to consider this indole as a powerful anti-angiogenic agent for HCC treatment.
Collapse
|
36
|
Fang HY, Wang HM, Chang KF, Hu HT, Hwang LJ, Fu TF, Lin YC, Chang WC, Chiu TP, Wen ZH, Fong Y, Chiu CC, Chen BH. Feruloyl-L-arabinose attenuates migration, invasion and production of reactive oxygen species in H1299 lung cancer cells. Food Chem Toxicol 2013; 58:459-66. [PMID: 23707472 DOI: 10.1016/j.fct.2013.05.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2011] [Revised: 05/10/2013] [Accepted: 05/13/2013] [Indexed: 12/15/2022]
Abstract
Ferulic acid (FA), a phenolic compound, is an abundant dietary antioxidant and exerts the mitogenic effect on cells. Recently, we isolated an active FA derivative, namely feruloyl-L-arabinose (FAA), from coba husk. The aim of this study was to investigate the effects of FAA on the proliferation, migration and invasion of H1299 human lung cancer cells. Our results showed a strong antioxidant potential of FAA. Additionally, FAA inhibited the migration and invasion ability, while causing a significant accumulation of G2/M-population, of H1299 tumor cells in a dose-dependent manner, whereas no significant change on cell proliferation was observed. Results from the wound healing assay revealed that cell migration ability was markedly inhibited by FAA treatments. Similarly, results of gelatin zymography study showed that FAA treatments significantly decreased the activities of matrix metalloproteinase (MMP)-2 and MMP-9, suggesting that FAA-mediated inhibition on migration and invasion of lung cancer cells may be achieved by the down-regulation of the MMPs activities. Taken together, our present work provides a new insight into the novel inhibitory function of FAA on cell migration in H1299 cells, suggesting its promising role in the chemoprevention of lung cancer.
Collapse
Affiliation(s)
- Hsin-Yu Fang
- Department of Food Nutrition, Chung-Hwa University of Medical Technology, Tainan 701, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Golden berry-derived 4β-hydroxywithanolide E for selectively killing oral cancer cells by generating ROS, DNA damage, and apoptotic pathways. PLoS One 2013; 8:e64739. [PMID: 23705007 PMCID: PMC3660349 DOI: 10.1371/journal.pone.0064739] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 04/17/2013] [Indexed: 01/01/2023] Open
Abstract
Background Most chemotherapeutic drugs for killing cancer cells are highly cytotoxic in normal cells, which limits their clinical applications. Therefore, a continuing challenge is identifying a drug that is hypersensitive to cancer cells but has minimal deleterious effects on healthy cells. The aims of this study were to evaluate the potential of 4β-hydroxywithanolide (4βHWE) for selectively killing cancer cells and to elucidate its related mechanisms. Methodology and Principal Findings Changes in survival, oxidative stress, DNA damage, and apoptosis signaling were compared between 4βHWE-treated oral cancer (Ca9-22) and normal fibroblast (HGF-1) cells. At 24 h and 48 h, the numbers of Ca9-22 cells were substantially decreased, but the numbers of HGF-1 cells were only slightly decreased. Additionally, the IC50 values for 4βHWE in the Ca9-22 cells were 3.6 and 1.9 µg/ml at 24 and 48 h, respectively. Time-dependent abnormal increases in ROS and dose-responsive mitochondrial depolarization can be exploited by using 4βHWE in chemotherapies for selectively killing cancer cells. Dose-dependent DNA damage measured by comet-nuclear extract assay and flow cytometry-based γ-H2AX/propidium iodide (PI) analysis showed relatively severer damage in the Ca9-22 cells. At both low and high concentrations, 4βHWE preferably perturbed the cell cycle in Ca9-22 cells by increasing the subG1 population and arrest of G1 or G2/M. Selective induction of apoptosis in Ca9-22 cells was further confirmed by Annexin V/PI assay, by preferential expression of phosphorylated ataxia-telangiectasia- and Rad3-related protein (p-ATR), and by cleavage of caspase 9, caspase 3, and poly ADP-ribose polymerase (PARP). Conclusions/Significance Together, the findings of this study, particularly the improved understanding of the selective killing mechanisms of 4βHWE, can be used to improve efficiency in killing oral cancer cells during chemoprevention and therapy.
Collapse
|
38
|
Sook SH, Lee HJ, Kim JH, Sohn EJ, Jung JH, Kim B, Kim JH, Jeong SJ, Kim SH. Reactive oxygen species-mediated activation of AMP-activated protein kinase and c-Jun N-terminal kinase plays a critical role in beta-sitosterol-induced apoptosis in multiple myeloma U266 cells. Phytother Res 2013; 28:387-94. [PMID: 23640957 DOI: 10.1002/ptr.4999] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 02/26/2013] [Accepted: 03/22/2013] [Indexed: 11/07/2022]
Abstract
Although beta-sitosterol has been well known to have anti-tumor activity in liver, lung, colon, stomach, breast and prostate cancers via cell cycle arrest and apoptosis induction, the underlying mechanism of anti-cancer effect of beta-sitosterol in multiple myeloma cells was never elucidated until now. Thus, in the present study, the role of reactive oxygen species (ROS) in association with AMP-activated protein kinase (AMPK) and c-Jun N-terminal kinase (JNK) pathways was demonstrated in beta-sitosterol-treated multiple myeloma U266 cells. Beta-sitosterol exerted cytotoxicity, increased sub-G1 apoptotic population and activated caspase-9 and -3, cleaved poly (ADP-ribose) polymerase (PARP) followed by decrease in mitochondrial potential in U266 cells. Beta-sitosterol promoted ROS production, activated AMPK, acetyl-CoA carboxylase (ACC) and JNK in U266 cells. Also, beta-sitosterol attenuated the phosphorylation of AKT, mammalian target of rapamycin and S6K, and the expression of cyclooxygenase-2 and VEGF in U266 cells. Conversely, AMPK inhibitor compound C and JNK inhibitor SP600125 suppressed apoptosis induced by beta-sitosterol in U266 cells. Furthermore, ROS scavenger N-acetyl L-cysteine attenuated beta-sitosterol-mediated sub-G1 accumulation, PARP cleavage, JNK and AMPK activation in U266 cells. Overall, these findings for the first time suggest that ROS-mediated activation of cancer metabolism-related genes such as AMPK and JNK plays an important role in beta-sitosterol-induced apoptosis in U266 multiple myeloma cells.
Collapse
Affiliation(s)
- Song Hyo Sook
- College of Korean Medicine, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul, 130-701, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Cardiotoxin III inhibits proliferation and migration of oral cancer cells through MAPK and MMP signaling. ScientificWorldJournal 2013; 2013:650946. [PMID: 23710144 PMCID: PMC3654281 DOI: 10.1155/2013/650946] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 03/12/2013] [Indexed: 11/17/2022] Open
Abstract
Cardiotoxin III (CTXIII), isolated from the snake venom of Formosan cobra Naja naja atra, has previously been found to induce apoptosis in many types of cancer. Early metastasis is typical for the progression of oral cancer. To modulate the cell migration behavior of oral cancer is one of the oral cancer therapies. In this study, the possible modulating effect of CTXIII on oral cancer migration is addressed. In the example of oral squamous carcinoma Ca9-22 cells, the cell viability was decreased by CTXIII treatment in a dose-responsive manner. In wound-healing assay, the cell migration of Ca9-22 cells was attenuated by CTXIII in a dose- and time-responsive manner. After CTXIII treatment, the MMP-2 and MMP-9 protein expressions were downregulated, and the phosphorylation of JNK and p38-MAPK was increased independent of ERK phosphorylation. In conclusion, CTXIII has antiproliferative and -migrating effects on oral cancer cells involving the p38-MAPK and MMP-2/-9 pathways.
Collapse
|
40
|
Ye L, Shi Z, Liu H, Yang X, Wang K. GdCl3 induced Hep G2 cell death through mitochondrial and external death pathways without significant elevation of ROS generation. Biol Trace Elem Res 2013; 151:148-55. [PMID: 23129526 DOI: 10.1007/s12011-012-9538-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2012] [Accepted: 10/22/2012] [Indexed: 01/13/2023]
Abstract
Gadolinium (Gd) compounds have important applications as MRI contrast and potential anticancer agents. The present study investigated the mechanisms of the proapoptotic effect of gadolinium chloride (GdCl(3)) on hepatoblastoma cell line (Hep G2) tumor cells. The experimental results indicated that GdCl(3) induced apoptosis of Hep G2 at high concentration and with long time incubation; however, unlike the actions on normal cell lines, GdCl(3) did not cause any oxidative stress on tumor cells. Cytochrome c (Cyt c) and apoptosis inducing factor release, Bax translocation, collapse of mitochondria membrane potential, caspase 3 and 8 activation, and Bid cleavage were observed along with a sustained activation of extracellular signal-regulated kinase (ERK) and c-Jun NH2 terminal kinase (JNK). Addition of ERK and JNK inhibitor attenuated the effect of GdCl(3) induced apoptosis and Cyt c release. All the results suggested a novel mechanism that GdCl(3) induced Hep G2 cell death through intrinsic and external death pathways without significant elevation of reactive oxygen species generation. The present work provided new insight to understand the mechanisms of the biological effects of GdCl(3) and implications for the development of anticancer Gd agents.
Collapse
Affiliation(s)
- Lihua Ye
- State Key Laboratories of Natural and Biomimetic Drugs, Peking University, Beijing, People's Republic of China
| | | | | | | | | |
Collapse
|
41
|
Carbajo-Pescador S, Steinmetz C, Kashyap A, Lorenz S, Mauriz JL, Heise M, Galle PR, González-Gallego J, Strand S. Melatonin induces transcriptional regulation of Bim by FoxO3a in HepG2 cells. Br J Cancer 2012; 108:442-9. [PMID: 23257900 PMCID: PMC3566813 DOI: 10.1038/bjc.2012.563] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background: Melatonin induces apoptosis in many different cancer cell lines, including hepatocellular carcinoma cells. However, the responsible pathways have not been clearly elucidated. A member of the forkhead transcription factors' family, FoxO3a, has been implicated in the expression of the proapoptotic protein Bim (a Bcl-2-interacting mediator of cell death). In this study, we used human HepG2 liver cancer cells as an in vitro model to investigate whether melatonin treatment induces Bim through regulation by the transcription factor FoxO3a. Methods: Cytotoxicity of melatonin was compared in HepG2 hepatoblastoma cells and primary human hepatocytes. Proapoptotic Bim expression was analysed by reverse transcriptase–polymerase chain reaction and western blot. Reporter gene assays and chromatin immunoprecipitation assays were performed to analyse whether FoxO3a transactivates the Bim promoter. Small interfering RNA (siRNA) was used to study the role of FoxO3a in Bim expression. Immunofluorescence was performed to analyse FoxO3a localisation in HepG2 cells. Results: Melatonin treatment induces apoptosis in HepG2 cells, but not in primary human hepatocytes. The proapoptotic effect was mediated by increased expression of the BH3-only protein Bim. During melatonin treatment, we observed increased transcriptional activity of the forkhead-responsive element and could demonstrate that FoxO3a binds to a specific sequence within the Bim promoter. Furthermore, melatonin reduced phosphorylation of FoxO3a at Thr32 and Ser253, and induced its increased nuclear localisation. Moreover, silencing experiments with FoxO3a siRNA prevented Bim upregulation. Conclusion: This study shows that melatonin can induce apoptosis in HepG2 hepatocarcinoma cells through the upregulation of proapoptotic Bim mediated by nuclear translocation and activation of the transcription factor FoxO3a.
Collapse
Affiliation(s)
- S Carbajo-Pescador
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd) and Institute of Biomedicine, University of León, León, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Neoh CA, Wang RYL, Din ZH, Su JH, Chen YK, Tsai FJ, Weng SH, Wu YJ. Induction of apoptosis by sinulariolide from soft coral through mitochondrial-related and p38MAPK pathways on human bladder carcinoma cells. Mar Drugs 2012; 10:2893-911. [PMID: 23249971 PMCID: PMC3528132 DOI: 10.3390/md10122893] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Revised: 11/30/2012] [Accepted: 12/12/2012] [Indexed: 11/16/2022] Open
Abstract
Sinulariolide, an isolated compound from the soft coral Sinularia flexibilis, possesses the anti-proliferative, anti-migratory and apoptosis-inducing activities against the TSGH bladder carcinoma cell. The anti-tumor effects of sinulariolide were determined by 3-(4,5-cimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay, cell migration assay and flow cytometry, respectively. Sinulariolide inhibited the growth and migration of bladder carcinoma cells in a dose-dependent manner, as well as induced both early and late apoptosis as determined by the flow cytometer. Also, the sinulariolide-induced apoptosis is related to the mitochondrial-mediated apoptosis via caspase-dependent pathways, elucidated by the loss of mitochondrial membrane potential, release of cytochrome C, activation of caspase-3/-9, Bax and Bad, as well as suppression of Bcl-2/Bcl-xL/Mcl-1. Detection of the PARP-1 cleaved product suggested the partial involvement of caspase-independent pathways. Moreover, inhibition of p38MAPK activity leads to the rescue of the cell cytotoxicity of sinulariolide-treated TSGH cells, indicating that the p38MAPK pathway is also involved in the sinulariolide-induced cell apoptosis. Altogether, these results suggest that sinulariolide induces apoptosis against bladder cancer cells through mitochondrial-related and p38MAPK pathways.
Collapse
Affiliation(s)
- Choo-Aun Neoh
- Department of Research, Pingtung Christian Hospital, Pingtung 90059, Taiwan; E-Mail:
| | - Robert Y.-L. Wang
- Department of Biomedical Sciences and Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan 33302, Taiwan; E-Mail:
| | - Zhong-Hao Din
- Graduate Institute of Applied Healthy and Biotechnology, Meiho University, Pingtung 91202, Taiwan; E-Mail:
| | - Jui-Hsin Su
- National Museum of Marine Biology and Aquarium, Pingtung 94446, Taiwan; E-Mail:
| | - Yu-Kuei Chen
- Department of Food Science and Nutrition, Meiho University, Pingtung 91202, Taiwan; E-Mail:
| | - Feng-Jen Tsai
- Department of Beauty Science, Meiho University, Pingtung 91202, Taiwan; E-Mail:
| | - Shun-Hsiang Weng
- Department of Hospitality Management, Meiho University, Pingtung 91202, Taiwan; E-Mail:
| | - Yu-Jen Wu
- Department of Beauty Science, Meiho University, Pingtung 91202, Taiwan; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +886-8-7799821 (ext. 8613); Fax: +886-8-7797821
| |
Collapse
|
43
|
Chun J, Joo EJ, Kang M, Kim YS. Platycodin D induces anoikis and caspase-mediated apoptosis via p38 MAPK in AGS human gastric cancer cells. J Cell Biochem 2012; 114:456-70. [DOI: 10.1002/jcb.24386] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 08/30/2012] [Indexed: 01/13/2023]
|
44
|
Su TR, Tsai FJ, Lin JJ, Huang HH, Chiu CC, Su JH, Yang YT, Chen JYF, Wong BS, Wu YJ. Induction of apoptosis by 11-dehydrosinulariolide via mitochondrial dysregulation and ER stress pathways in human melanoma cells. Mar Drugs 2012; 10:1883-1898. [PMID: 23015779 PMCID: PMC3447343 DOI: 10.3390/md10081883] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 08/06/2012] [Accepted: 08/14/2012] [Indexed: 02/07/2023] Open
Abstract
In this study the isolated compound 11-dehydrosinulariolide from soft coral Sinularia leptoclados possessed anti-proliferative, anti-migratory and apoptosis-inducing activities against A2058 melanoma cells. Anti-tumor effects of 11-dehydrosinulariolide were determined by MTT assay, cell migration assay and flow cytometry. Growth and migration of melanoma cells were dose-dependently inhibited by 2–8 μg/mL 11-dehydrosinulariolide. Flow cytometric data indicated that 11-dehydrosinulariolide induces both early and late apoptosis in melanoma cells. It was found that the apoptosis induced by 11-dehydrosinulariolide is relevant to mitochondrial-mediated apoptosis via caspase-dependent pathways, elucidated by loss of mitochondrial membrane potential (∆Ψm), release of cytochrome C, activation of caspase-3/-9 and Bax as well as suppression of Bcl-2/Bcl-xL. The cleavage of PARP-1 suggested partial involvement of caspase-independent pathways. Immunoblotting data displayed up-regulations of PERK/eIF2α/ATF4/CHOP and ATF6/CHOP coupling with elevation of ER stress chaperones GRP78, GRP94, calnexin, calreticulin and PDI, implicating the involvement of these factors in ER stress-mediated apoptosis induced by 11-dehydrosinulariolide. The abolishment of apoptotic events after pre-treatment with salubrinal indicated that ER stress-mediated apoptosis is also induced by 11-dehydrosinulariolide against melanoma cells. The data in this study suggest that 11-dehydrosinulariolide potentially induces apoptosis against melanoma cells via mitochondrial dysregulation and ER stress pathways.
Collapse
Affiliation(s)
- Tzu-Rong Su
- Antai Medical Care Cooperation Antai Tian-Sheng Memorial Hospital, Pingtung 92842, Taiwan; (T.-R.S.); (B.-S.W.)
| | - Feng-Jen Tsai
- Department of Beauty Science, Meiho University, Pingtung 91202, Taiwan; (F.-J.T.); (H.H.H.)
| | - Jen-Jie Lin
- Graduate Institute of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 91202, Taiwan;
| | - Han Hsiang Huang
- Department of Beauty Science, Meiho University, Pingtung 91202, Taiwan; (F.-J.T.); (H.H.H.)
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 80761, Taiwan; (C.-C.C.); (J.Y.-F.C.)
| | - Jui-Hsin Su
- National Museum of Marine Biology and Aquarium, Pingtung 94446, Taiwan;
| | - Ya-Ting Yang
- Chemistry Department, National Sun Yat-Sen University, No. 70, Lienhai Rd., Kaohsiung 80424, Taiwan;
| | - Jeff Yi-Fu Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 80761, Taiwan; (C.-C.C.); (J.Y.-F.C.)
| | - Bing-Sang Wong
- Antai Medical Care Cooperation Antai Tian-Sheng Memorial Hospital, Pingtung 92842, Taiwan; (T.-R.S.); (B.-S.W.)
| | - Yu-Jen Wu
- Department of Beauty Science, Meiho University, Pingtung 91202, Taiwan; (F.-J.T.); (H.H.H.)
- Author to whom correspondence should be addressed; or ; Tel.: +886-8-7799821 (ext. 8600); Fax: +886-8-7797821
| |
Collapse
|
45
|
Su TR, Lin JJ, Chiu CC, Chen JYF, Su JH, Cheng ZJ, Hwang WI, Huang HH, Wu YJ. Proteomic investigation of anti-tumor activities exerted by sinularin against A2058 melanoma cells. Electrophoresis 2012; 33:1139-52. [PMID: 22539317 DOI: 10.1002/elps.201100462] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The extracts from soft corals have been increasingly investigated for biomedical and therapeutic purposes. The aim of this study is to examine and analyze the anti-tumor effects of the genus Sinularia extract sinularin on A2058 melanoma cells using MTT assay, cell migration assay, wound healing assay, flow cytometric analysis, and proteomic analysis. Sinularin dose-dependently (1-5 μg/mL) inhibited melanoma cell proliferation while the treatment at identical concentrations suppressed cell migration. Sinularin dose-dependently enhanced apoptotic melanoma cells and caused tumor cell accumulation at G2/M phase, indicating that sinularin exerts apoptosis-induced and cell cycle-delayed activities in A2058 melanoma cells. Comparative proteomic analysis was conducted to investigate the effects of sinularin at the molecular level by comparison between the protein profiling of melanoma cells treated with sinularin and without the treatment. Thirty-five differential proteins (13 upregulated and 22 downregulated) concerning the treatment were identified by liquid chromatography-tandem mass spectrometry. Proteomic data and Western blot displayed the levels of several tumor inhibitory or apoptosis-associated proteins including annexin A1, voltage-dependent anion-selective channel protein 1 and prohibitin (upregulated), heat shock protein 60, heat shock protein beta-1, and peroxiredoxin-2 (downregulated) in A2058 melanoma cells exposed to sinularin. Increased expression of p53, cleaved-caspase-3, cleaved-caspase-8, cleaved-caspase-9, p21, and Bax and decreased expression of Bcl-2 in sinularin-treated melanoma cells suggest that the anti-tumor activities of sinularin against melanoma cells are particularly correlated with these pro-apoptotic factors. These data provide important information for the mechanisms of anti-tumor effects of sinularin on melanoma cells and may be helpful for drug development and progression monitoring of human melanoma.
Collapse
Affiliation(s)
- Tzu-Rong Su
- Antai Tian-Sheng Memorial Hospital, Pingtung, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Lin CL, Chen RF, Chen JYF, Chu YC, Wang HM, Chou HL, Chang WC, Fong Y, Chang WT, Wu CY, Chiu CC. Protective effect of caffeic acid on paclitaxel induced anti-proliferation and apoptosis of lung cancer cells involves NF-κB pathway. Int J Mol Sci 2012; 13:6236-6245. [PMID: 22754361 PMCID: PMC3382759 DOI: 10.3390/ijms13056236] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 05/07/2012] [Accepted: 05/14/2012] [Indexed: 01/19/2023] Open
Abstract
Caffeic acid (CA), a natural phenolic compound, is abundant in medicinal plants. CA possesses multiple biological effects such as anti-bacterial and anti-cancer growth. CA was also reported to induce fore stomach and kidney tumors in a mouse model. Here we used two human lung cancer cell lines, A549 and H1299, to clarify the role of CA in cancer cell proliferation. The growth assay showed that CA moderately promoted the proliferation of the lung cancer cells. Furthermore, pre-treatment of CA rescues the proliferation inhibition induced by a sub-IC50 dose of paclitaxel (PTX), an anticancer drug. Western blot showed that CA up-regulated the pro-survival proteins survivin and Bcl-2, the down-stream targets of NF-κB. This is consistent with the observation that CA induced nuclear translocation of NF-κB p65. Our study suggested that the pro-survival effect of CA on PTX-treated lung cancer cells is mediated through a NF-κB signaling pathway. This may provide mechanistic insights into the chemoresistance of cancer calls.
Collapse
Affiliation(s)
- Chien-Liang Lin
- Department of Life Science and Institute of Zoology, National Taiwan University, Taipei 106, Taiwan; E-Mails: (C.-L.L.); (R.-F.C.)
| | - Ruei-Feng Chen
- Department of Life Science and Institute of Zoology, National Taiwan University, Taipei 106, Taiwan; E-Mails: (C.-L.L.); (R.-F.C.)
| | - Jeff Yi-Fu Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; E-Mails: (J.Y.-F.C.); (Y.-C.C.); (H.-L.C.)
| | - Ying-Chieh Chu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; E-Mails: (J.Y.-F.C.); (Y.-C.C.); (H.-L.C.)
| | - Hui-Min Wang
- Department of Fragrance and Cosmetics Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan; E-Mail:
| | - Han-Lin Chou
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; E-Mails: (J.Y.-F.C.); (Y.-C.C.); (H.-L.C.)
| | - Wei-Chiao Chang
- Graduate Institute of Medical Genetics, Kaohsiung Medical University, Kaohsiung 807, Taiwan; E-Mail:
| | - Yao Fong
- Chest Surgery, Chi-Mei Foundation Medical Center, Yung Kang City, Tainan 901, Taiwan; E-Mail:
| | - Wen-Tsan Chang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; E-Mail:
- Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chang-Yi Wu
- Department of Biological Sciences, National Sun Yat-Sen University, 70 Lien Hai Road, Kaohsiung 804, Taiwan; E-Mail:
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; E-Mails: (J.Y.-F.C.); (Y.-C.C.); (H.-L.C.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +886-7-312-1101 (ext. 2368); Fax: +886-7-312-5339
| |
Collapse
|
47
|
Ye Y, Yin DT, Chen L, Zhou Q, Shen R, He G, Yan Q, Tong Z, Issekutz AC, Shapiro CL, Barsky SH, Lin H, Li JJ, Gao JX. Identification of Piwil2-like (PL2L) proteins that promote tumorigenesis. PLoS One 2010; 5:e13406. [PMID: 20975993 PMCID: PMC2958115 DOI: 10.1371/journal.pone.0013406] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Accepted: 09/09/2010] [Indexed: 11/19/2022] Open
Abstract
PIWIL2, a member of PIWI/AGO gene family, is expressed in the germline stem cells (GSCs) of testis for gametogenesis but not in adult somatic and stem cells. It has been implicated to play an important role in tumor development. We have previously reported that precancerous stem cells (pCSCs) constitutively express Piwil2 transcripts to promote their proliferation. Here we show that these transcripts de facto represent Piwil2-like (PL2L) proteins. We have identified several PL2L proteins including PL2L80, PL2L60, PL2L50 and PL2L40, using combined methods of Gene-Exon-Mapping Reverse Transcription Polymerase Chain Reaction (GEM RT-PCR), bioinformatics and a group of novel monoclonal antibodies. Among them, PL2L60 rather than Piwil2 and other PL2L proteins is predominantly expressed in various types of human and mouse tumor cells. It promotes tumor cell survival and proliferation in vitro through up-regulation of Stat3 and Bcl2 gene expressions, the cell cycle entry from G(0/1) into S-phase, and the nuclear expression of NF-κB, which contribute to the tumorigenicity of tumor cells in vivo. Consistently, PL2L proteins rather than Piwil2 are predominantly expressed in the cytoplasm or cytoplasm and nucleus of euchromatin-enriched tumor cells in human primary and metastatic cancers, such as breast and cervical cancers. Moreover, nuclear PL2L proteins are always co-expressed with nuclear NF-κB. These results reveal that PL2L60 can coordinate with NF-κB to promote tumorigenesis and might mediate a common pathway for tumor development without tissue restriction. The identification of PL2L proteins provides a novel insight into the mechanisms of cancer development as well as a novel bridge linking cancer diagnostics and anticancer drug development.
Collapse
Affiliation(s)
- Yin Ye
- Department of Pathology, Ohio State University Medical Center, Columbus, Ohio, United States of America
| | - De-Tao Yin
- Department of Pathology, Ohio State University Medical Center, Columbus, Ohio, United States of America
- Department of General Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Chen
- Department of Pathology, Ohio State University Medical Center, Columbus, Ohio, United States of America
| | - Quansheng Zhou
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital, Soochow University, Suzhou, China
| | - Rulong Shen
- Department of Pathology, Ohio State University Medical Center, Columbus, Ohio, United States of America
| | - Gang He
- Department of Pathology, Ohio State University Medical Center, Columbus, Ohio, United States of America
| | - Qingtao Yan
- Department of Pathology, Ohio State University Medical Center, Columbus, Ohio, United States of America
| | - Zhenyu Tong
- Department of Pathology, Ohio State University Medical Center, Columbus, Ohio, United States of America
| | - Andrew C. Issekutz
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Charles L. Shapiro
- Department of Internal Medicine, Ohio State University Medical Center, Columbus, Ohio, United States of America
- Comprehensive Cancer Center, Ohio State University Medical Center, Columbus, Ohio, United States of America
| | - Sanford H. Barsky
- Department of Pathology, Ohio State University Medical Center, Columbus, Ohio, United States of America
- Comprehensive Cancer Center, Ohio State University Medical Center, Columbus, Ohio, United States of America
| | - Haifan Lin
- Department of Cell Biology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Jian-Jian Li
- Department of Radiation Oncology, University of California Davis, Sacramento, California, United States of America
| | - Jian-Xin Gao
- Department of Pathology, Ohio State University Medical Center, Columbus, Ohio, United States of America
- Comprehensive Cancer Center, Ohio State University Medical Center, Columbus, Ohio, United States of America
| |
Collapse
|
48
|
Chen JYF, Hwang CC, Chen WY, Lee JC, Fu TF, Fang K, Chu YC, Huang YL, Lin JC, Tsai WH, Chang HW, Chen BH, Chiu CC. Additive effects of C(2)-ceramide on paclitaxel-induced premature senescence of human lung cancer cells. Life Sci 2010; 87:350-7. [PMID: 20624405 DOI: 10.1016/j.lfs.2010.06.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Revised: 06/09/2010] [Accepted: 06/25/2010] [Indexed: 01/13/2023]
Abstract
AIMS the aims of the study are to investigate the additive effect of exogenous short-carbon chain phospholipids, C(2)-ceramide, on an anti-cancer drug paclitaxel (PTX)-induced senescence of human non-small cell lung cancer (NSCLC) cells deficient in functional p53 and p16, and to examine whether mitogen-activated protein kinase (MAPK) plays a role in ceramide-sensitized senescence of NSCLC cells. MAIN METHODS to determine whether exogenous C(2)-ceramide renders lung cancer cells more sensitive to PTX treatment, techniques employing a flow cytometry-based cell cycle analysis and acidic β-galactosidase staining for senescent cells were used. Furthermore, to elucidate the role of MAPK proteins in modulating senescence, assays for protein levels of selective MAPKs and Bcl-2 family members, and detection of transcriptional levels senescence-associated genes were used in the study. KEY FINDINGS a sub-lethal dose of C(2)-ceramide sensitized the NSCLC H1299 cells to PTX treatment. The additive effects of C(2)-ceramide and PTX resulted in proliferative inhibition, G(2)-phase arrest of cell cycle, activation of p38 and eventually premature senescence. Importantly, neither p53, p21(waf1/cip1) nor p16(ink4) was shown to be involved in C(2)-ceramide-sensitized proliferative inhibition and senescence of H1299 cells by PTX in our study. SIGNIFICANCE our study demonstrates that the short-carbon chain C(2)-ceramide can effectively sensitize PTX-induced senescence of H1299 cells via both p21(waf1/cip1)- and p16(ink4)-independent pathways.
Collapse
Affiliation(s)
- Jeff Yi-Fu Chen
- Department of Biotechnology, Center of Excellence for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|