1
|
Han R, Li X, Gao X, Lv G. Cinnamaldehyde: Pharmacokinetics, anticancer properties and therapeutic potential (Review). Mol Med Rep 2024; 30:163. [PMID: 38994757 PMCID: PMC11267250 DOI: 10.3892/mmr.2024.13287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 06/21/2024] [Indexed: 07/13/2024] Open
Abstract
Cancer incidence is increasing globally, presenting a growing public health challenge. While anticancer drugs are crucial in treatment, their limitations, including poor targeting ability and high toxicity, hinder effectiveness and patient safety, requiring relentless scientific research and technological advancements to develop safer and more effective therapeutics. Cinnamaldehyde (CA), an active compound derived from the natural plant cinnamon, has garnered attention in pharmacological research due to its diverse therapeutic applications. CA has potential in treating a wide array of conditions, including cardiovascular diseases, diabetes, inflammatory disorders and various forms of cancer. The present review comprehensively summarizes the physicochemical and pharmacokinetic profiles of CA, and delves into the latest advancements in elucidating its potential mechanisms and targets across various cancer types. CA and its derivatives have antitumor effects, which encompass inhibiting cell proliferation, arresting the cell cycle, inducing apoptosis, limiting cell migration and invasion, and suppressing angiogenesis. Additionally, the present review explores targeted formulations of CA, laying a scientific foundation for further exploration of its implications in cancer prevention and treatment strategies.
Collapse
Affiliation(s)
- Ruxia Han
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Xueying Li
- School of Health, Binzhou Polytechnic, Binzhou, Shandong 256600, P.R. China
| | - Xinfu Gao
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
- Department of Pharmacy, Binzhou Medical University Hospital, Binzhou, Shandong 256603, P.R. China
| | - Guangyao Lv
- Department of Pharmacy, Binzhou Medical University Hospital, Binzhou, Shandong 256603, P.R. China
| |
Collapse
|
2
|
Huang JY, Lin YC, Chen HM, Lin JT, Kao SH. Adenine Combined with Cisplatin Promotes Anticancer Activity against Hepatocellular Cancer Cells through AMPK-Mediated p53/p21 and p38 MAPK Cascades. Pharmaceuticals (Basel) 2022; 15:ph15070795. [PMID: 35890094 PMCID: PMC9322617 DOI: 10.3390/ph15070795] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 12/24/2022] Open
Abstract
Cisplatin has been widely used in cancer treatments. Recent evidence indicates that adenine has potential anticancer activities against various types of cancers. However, the effects of the combination of adenine and cisplatin on hepatocellular carcinoma (HCC) cells remain sketchy. Here, our objective was to elucidate the anticancer activity of adenine in combination with cisplatin in HCC cells and its mechanistic pathways. Cell viability and cell cycle progression were assessed by the SRB assay and flow cytometry, respectively. Apoptosis was demonstrated by PI/annexin V staining and flow cytometric analysis. Protein expression, signaling cascade, and mRNA expression were analyzed by Western blotting and quantitative RT-PCR, respectively. Our results showed that adenine jointly potentiated the inhibitory effects of cisplatin on the cell viability of SK-Hep1 and Huh7 cells. Further investigation showed that adenine combined with cisplatin induced higher S phase arrest and apoptosis in HCC cells. Mechanically, adenine induced AMPK activation, reduced mTOR phosphorylation, and increased p53 and p21 levels. The combination of adenine and cisplatin synergistically reduced Bcl-2 and increased PUMA, cleaved caspase-3, and PARP in HCC cells. Adenine also upregulated the mRNA expression of p53, p21, PUMA, and PARP, while knockdown of AMPK reduced the increased expression of these genes. Furthermore, adenine also induced the activation of p38 MAPK through AMPK signaling, and the inhibition of p38 MAPK reduced the apoptosis of HCC cells with exposure to adenine combined with cisplatin. Collectively, these findings reveal that the combination of adenine and cisplatin synergistically enhances apoptosis of HCC cells, which may be attributed to the AMPK-mediated p53/p21 and p38 MAPK cascades. It suggests that adenine may be a potential adjuvant for the treatment of HCC in combination with cisplatin.
Collapse
Affiliation(s)
- Jhen-Yu Huang
- Institute of Medicine, College of Medicine, Chung Shan Medical University, Taichung City 402306, Taiwan;
| | - You-Cian Lin
- Cardiovascular Division, Surgical Department, China Medical University Hospital, Taichung City 404332, Taiwan;
- School of Medicine, College of Medicine, China Medical University, Taichung City 404332, Taiwan
| | - Han-Min Chen
- Institute of Applied Science and Engineering, Catholic Fu Jen University, New Taipei 242048, Taiwan;
| | - Jiun-Tsai Lin
- Energenesis Biomedical Co., Ltd., Taipei 114694, Taiwan;
| | - Shao-Hsuan Kao
- Institute of Medicine, College of Medicine, Chung Shan Medical University, Taichung City 402306, Taiwan;
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung City 402306, Taiwan
- Correspondence: ; Tel.: +886-4-24730022 (ext. 11681)
| |
Collapse
|
3
|
The Antitumor Effect of Cinnamaldehyde Derivative CB-PIC in Hepatocellular Carcinoma Cells via Inhibition of Pyruvate and STAT3 Signaling. Int J Mol Sci 2022; 23:ijms23126461. [PMID: 35742904 PMCID: PMC9223629 DOI: 10.3390/ijms23126461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 11/17/2022] Open
Abstract
Though cinnamaldehyde derivative (CB-PIC), a major compound of cinnamon, is known to have anticancer activity, its underlying mechanism is not fully understood. In the present study, the anticancer mechanism of CB-PIC was investigated in human hepatocellular carcinoma cells (HCCs) in association with signal transducer and activator of transcription 3 (STAT3) signaling. CB-PIC exerted cytotoxicity in HepG2 and Huh7 cells. CB-PIC increased the sub G1 population and attenuated the expression of pro-poly (ADP-ribose) polymerase (PARP) and pro-Caspase3 in HepG2 and Huh7 cells. Interestingly, CB-PIC significantly abrogated the expression of a glycolytic enzyme pyruvate kinase M2 (PKM2) in HepG2 cells more than in LNCaP, A549, and HCT-116 cells. Consistently, CB-PIC reduced the expression of hexokinase 2 (HK2) and PKM2, along with a reduced production of lactate in HepG2 and Huh7 cells. Notably, CB-PIC suppressed the phosphorylation of STAT3 in HepG2 and Huh7 cells and conversely STAT3 depletion enhanced the capacity of CB-PIC to suppress the expression of HK2, PKM2, and pro-caspase3 and to reduce the viability in Huh7 cells. Furthermore, CB-PIC activated the phosphorylation of AMPK and ERK and suppressed expression of IL-6 as STAT3-related genes in HepG2 and Huh7 cells. Conversely, pyruvate treatment reversed the inhibitory effect of CB-PIC on p-STAT3, HK2, PKM2, and pro-PARP in Huh7 cells. Overall, there findings suggest that CB-PIC exerts an apoptotic effect via inhibition of the Warburg effect mediated by p-STAT3 and pyruvate signaling.
Collapse
|
4
|
Singh M, Nicol AT, DelPozzo J, Wei J, Singh M, Nguyen T, Kobayashi S, Liang Q. Demystifying the Relationship Between Metformin, AMPK, and Doxorubicin Cardiotoxicity. Front Cardiovasc Med 2022; 9:839644. [PMID: 35141304 PMCID: PMC8818847 DOI: 10.3389/fcvm.2022.839644] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/04/2022] [Indexed: 12/12/2022] Open
Abstract
Doxorubicin (DOX) is an extremely effective and wide-spectrum anticancer drug, but its long-term use can lead to heart failure, which presents a serious problem to millions of cancer survivors who have been treated with DOX. Thus, identifying agents that can reduce DOX cardiotoxicity and concurrently enhance its antitumor efficacy would be of great clinical value. In this respect, the classical antidiabetic drug metformin (MET) has stood out, appearing to have both antitumor and cardioprotective properties. MET is proposed to achieve these beneficial effects through the activation of AMP-activated protein kinase (AMPK), an essential regulator of mitochondrial homeostasis and energy metabolism. AMPK itself has been shown to protect the heart and modulate tumor growth under certain conditions. However, the role and mechanism of the hypothesized MET-AMPK axis in DOX cardiotoxicity and antitumor efficacy remain to be firmly established by in vivo studies using tumor-bearing animal models and large-scale prospective clinical trials. This review summarizes currently available literature for or against a role of AMPK in MET-mediated protection against DOX cardiotoxicity. It also highlights the emerging evidence suggesting distinct roles of the AMPK subunit isoforms in mediating the functions of unique AMPK holoenzymes composed of different combinations of isoforms. Moreover, the review provides a perspective regarding future studies that may help fully elucidate the relationship between MET, AMPK and DOX cardiotoxicity.
Collapse
Affiliation(s)
- Manrose Singh
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY, United States
| | - Akito T. Nicol
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY, United States
| | - Jaclyn DelPozzo
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY, United States
| | - Jia Wei
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xian, China
| | - Mandeep Singh
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY, United States
| | - Tony Nguyen
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY, United States
| | - Satoru Kobayashi
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY, United States
| | - Qiangrong Liang
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY, United States
- *Correspondence: Qiangrong Liang
| |
Collapse
|
5
|
Huang CW, Lin YC, Hung CH, Chen HM, Lin JT, Wang CJ, Kao SH. Adenine Inhibits the Invasive Potential of DLD-1 Human Colorectal Cancer Cell via the AMPK/FAK Axis. Pharmaceuticals (Basel) 2021; 14:ph14090860. [PMID: 34577560 PMCID: PMC8469022 DOI: 10.3390/ph14090860] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/15/2021] [Accepted: 08/23/2021] [Indexed: 01/05/2023] Open
Abstract
Tumor metastasis is a major cause of death of patients with colorectal cancer (CRC). Our previous findings show that adenine has antiproliferation activity against tumor cells. However, whether adenine reduces the invasiveness of DLD-1 and SW480 CRC cells has not been thoroughly explored. In this study, we aimed to explore the effects of adenine on the invasion potential of DLD-1 cells. Our findings showed that adenine at concentrations of ≤200 μM did not influence the cell viability of DLD-1 and SW480 CRC cells. By contrast, adenine reduced the migratory potential of the CRC cells. Moreover, it decreased the invasion capacity of the CRC cells in a dose-dependent manner. We further observed that adenine downregulated the protein levels of tissue plasminogen activator, matrix metalloproteinase-9, Snail, TWIST, and vimentin, but upregulated the tissue inhibitor of metalloproteinase-1 expression in DLD-1 cells. Adenine decreased the integrin αV level and reduced the activation of integrin-associated signaling components, including focal adhesion kinase (FAK), paxillin, and Src in DLD-1 cells. Further observations showed that adenine induced AMP-activated protein kinase (AMPK) activation and inhibited mTOR phosphorylation in DLD-1 cells. The knockdown of AMPK restored the reduced integrin αV level and FAK/paxillin/Src signaling inhibited by adenine in DLD-1 cells. Collectively, these findings reveal that adenine reduces the invasion potential of DLD-1 cells through the AMPK/integrin/FAK axis, suggesting that adenine may have anti-metastatic potential in CRC cells.
Collapse
Affiliation(s)
- Chien-Wei Huang
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Armed Forces General Hospital, Kaohsiung 802301, Taiwan;
- Department of Nursing, Tajen University, Pingtung 907101, Taiwan
| | - You-Cian Lin
- Surgical Department Cardiovascular Division, China Medical University Hospital, Taichung 404332, Taiwan;
- School of Medicine, China Medical University, Taichung 404332, Taiwan
| | - Chia-Hung Hung
- Institute of Medicine, College of Medicine, Chung Shan Medical University, Taichung 402306, Taiwan; (C.-H.H.); (C.-J.W.)
| | - Han-Min Chen
- Institute of Applied Science and Engineering, Catholic Fu Jen University, New Taipei 242048, Taiwan;
| | - Jiun-Tsai Lin
- Energenesis Biomedical Co. Ltd., Taipei 114694, Taiwan;
| | - Chau-Jong Wang
- Institute of Medicine, College of Medicine, Chung Shan Medical University, Taichung 402306, Taiwan; (C.-H.H.); (C.-J.W.)
| | - Shao-Hsuan Kao
- Institute of Medicine, College of Medicine, Chung Shan Medical University, Taichung 402306, Taiwan; (C.-H.H.); (C.-J.W.)
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 402306, Taiwan
- Correspondence: ; Tel.: +886-4-247-30022 (ext. 11681)
| |
Collapse
|
6
|
Repositioning metformin and propranolol for colorectal and triple negative breast cancers treatment. Sci Rep 2021; 11:8091. [PMID: 33854147 PMCID: PMC8047046 DOI: 10.1038/s41598-021-87525-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 03/22/2021] [Indexed: 12/27/2022] Open
Abstract
Drug repositioning refers to new uses for existing drugs outside the scope of the original medical indications. This approach fastens the process of drug development allowing finding effective drugs with reduced side effects and lower costs. Colorectal cancer (CRC) is often diagnosed at advanced stages, when the probability of chemotherapy resistance is higher. Triple negative breast cancer (TNBC) is the most aggressive type of breast cancer, highly metastatic and difficult to treat. For both tumor types, available treatments are generally associated to severe side effects. In our work, we explored the effect of combining metformin and propranolol, two repositioned drugs, in both tumor types. We demonstrate that treatment affects viability, epithelial-mesenchymal transition and migratory potential of CRC cells as we described before for TNBC. We show that combined treatment affects different steps leading to metastasis in TNBC. Moreover, combined treatment is also effective preventing the development of 5-FU resistant CRC. Our data suggest that combination of metformin and propranolol could be useful as a putative adjuvant treatment for both TNBC and CRC and an alternative for chemo-resistant CRC, providing a low-cost alternative therapy without associated toxicity.
Collapse
|
7
|
Berkovic MC, Mikulic D, Bilic-Curcic I, Mrzljak A. How far along are we in revealing the connection between metformin and colorectal cancer? World J Gastroenterol 2021; 27:1362-1368. [PMID: 33911461 PMCID: PMC8047538 DOI: 10.3748/wjg.v27.i14.1362] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/14/2021] [Accepted: 03/17/2021] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is among the most prevalent cancers worldwide, and its prevention and reduction of incidence is imperative. The presence of diabetes has been associated with a 30% increased risk of CRC, likely through the mechanism of hyperinsulinemia, which promotes tumorigenesis via the insulin receptor in the epithelium or by insulin-like growth factor pathways, inflammation, or adipokines, inducing cancer cell proliferation and cancer spread. Metformin, the first-line agent in treating type 2 diabetes, has a chemopreventive role in CRC development. Additionally, preclinical studies suggest synergistic effects of metformin with oxaliplatin in inhibiting in vitro models of colon cancer. Although preclinical studies on the post diagnostic use of metformin were promising and suggested its synergistic effects with chemotherapy, the data on the possible effects of metformin after surgery and other CRC treatment in the clinical setting are less conclusive, and randomized controlled trials are still lacking.
Collapse
Affiliation(s)
- Maja Cigrovski Berkovic
- Department of Kinesiological Anthropology and Methodology, Faculty of Kinesiology, University of Zagreb, Zagreb 10000, Croatia
- Clinical Hospital Dubrava, Zagreb 10000, Croatia
| | - Danko Mikulic
- Department of Surgery, Merkur University Hospital, Zagreb 10000, Croatia
| | - Ines Bilic-Curcic
- Department of Pharmacology, Faculty of Medicine, University of J. J. Strossmayer Osijek, Osijek 31000, Croatia
- Clinical Hospital Center Osijek, Osijek 31000, Croatia
| | - Anna Mrzljak
- Department of Medicine, Merkur University Hospital, School of Medicine, Zagreb 10000, Croatia
| |
Collapse
|
8
|
Iqbal H, Menaa F, Khan NU, Razzaq A, Khan ZU, Ullah K, Kamal R, Sohail M, Thiripuranathar G, Uzair B, Rana NF, Khan BA, Menaa B. Two Promising Anti-Cancer Compounds, 2-Hydroxycinnaldehyde and 2-Benzoyloxycinnamaldehyde: Where do we stand? Comb Chem High Throughput Screen 2021; 25:808-818. [PMID: 33593253 DOI: 10.2174/1386207324666210216094428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/10/2021] [Accepted: 01/17/2021] [Indexed: 11/22/2022]
Abstract
Natural bioactive compounds with anti-carcinogenic activity are gaining tremendous interest in the field of oncology. Cinnamon, an aromatic condiment commonly used in tropical regions, appeared incredibly promising as adjuvant for cancer therapy. Indeed, its whole or active parts (e.g., bark, leaf) exhibited significant anti-carcinogenic activity, which is mainly due to two cinnamaldehyde derivatives, namely 2-hydroxycinnaldehyde (HCA) and 2-benzoyloxycinnamaldehyde (BCA). In addition to their anti-cancer activity, HCA and BCA exert immunomodulatory, anti-platelets, and anti-inflammatory activities. Highly reactive α,ß-unsaturated carbonyl pharmacophore, called Michael acceptor, contribute to their therapeutic effects. The molecular mechanisms, underlying their anti-tumoral and anti-metastatic effects are miscellaneous, strongly suggesting that these compounds are multi-targeting compounds. Nevertheless, unravelling the exact molecular mechanisms of HCA and BCA remain a challenging matter which is necessary for optimal controlled-drug targeting delivery, safety, and efficiency. Eventually, their poor pharmacological properties (e.g., systemic bioavailability and solubility) represent a limitation, and depend both on their administration route (e.g., per os, intravenously) and the nature of the formulation (e.g., free, smart nano-). This concise review focused on the potential of HCA and BCA as adjuvants in Cancer. We described their medicinal effects as well as provide an update about their molecular mechanisms reported either in-vitro, ex-vivo, or in animal models.
Collapse
Affiliation(s)
- Haroon Iqbal
- College of Pharmaceutical Sciences, Soochow University, Suzhou. China
| | - Farid Menaa
- Department of Oncology, California Innovations Corp., San Diego, CA. United States
| | - Naveed Ullah Khan
- College of Pharmaceutical Sciences, Soochow University, Suzhou. China
| | - Anam Razzaq
- College of Pharmaceutical Sciences, Soochow University, Suzhou. China
| | | | - Kifayat Ullah
- College of Pharmaceutical Sciences, Soochow University, Suzhou. China
| | - Robia Kamal
- College of Pharmaceutical Sciences, Soochow University, Suzhou. China
| | - Muhammad Sohail
- Department of Pharmacy, School of Pharmacy, Yantai University, Yantai. China
| | - Gobika Thiripuranathar
- Institute of Chemistry Ceylon, College of Chemical Sciences, Welikada, Rajagiriya. Sri Lanka
| | - Bushra Uzair
- Department of Bioinformatics and Biotechnology, Islamic International University, Islamabad. Pakistan
| | - Nosheen Fatima Rana
- Department of Biomedical Engineering & Sciences, School of Mechanical & Manufacturing Engineering, National University of Sciences & Technology, Islamabad. Pakistan
| | - Barkat Ali Khan
- Department of Pharmacy, Gomal University, D.I. Khan. Pakistan
| | - Bouzid Menaa
- Department of Oncology, California Innovations Corp., San Diego, CA. United States
| |
Collapse
|
9
|
Yoon YJ, Kwon BM. Cinnamomum cassia, apoptosis, STAT3 inactivation and reactive oxygen species in cancer studies. Cancer 2021. [DOI: 10.1016/b978-0-12-819547-5.00029-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
10
|
Ahn CR, Park J, Kim JE, Ahn KS, Kim YW, Jeong M, Kim HJ, Park SH, Baek SH. Cinnamaldehyde and Hyperthermia Co-Treatment Synergistically Induces ROS-Mediated Apoptosis in ACHN Renal Cell Carcinoma Cells. Biomedicines 2020; 8:biomedicines8090357. [PMID: 32957430 PMCID: PMC7555957 DOI: 10.3390/biomedicines8090357] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 09/15/2020] [Accepted: 09/15/2020] [Indexed: 12/13/2022] Open
Abstract
Renal cell carcinoma (RCC) represents the most common form of kidney cancer, which accounts for 3-5% newly diagnosed cancer cases. Since limited therapies are available for RCC, a search for new options is required. Therefore, in this study, we evaluated the combination effect of cinnamaldehyde (CNM) and hyperthermia treatment. CNM treatment combined with 43 °C hyperthermia synergistically increased cytotoxicity in RCC cell line ACHN cells. Through Western blot assays, we observed increased apoptosis signaling and decreased proliferation/metastasis signaling, along with a repressed heat shock protein 70 level. In flow cytometry analyses, CNM and hyperthermia combination clearly induced apoptosis and mitochondrial potential of ACHN cells, while arresting the cell cycle. Investigation of reactive oxygen species (ROS) suggested a significant increase of ROS generation by CNM and 43 °C hyperthermia co-treatment. We could verify that ROS is crucial in the apoptotic action of combination treatment with CNM and hyperthermia through further experiments regarding an ROS scavenger. Overall, we suggest CNM and hyperthermia combination treatment as an alternative option of anticancer strategies for RCC.
Collapse
Affiliation(s)
- Chae Ryeong Ahn
- College of Korean Medicine, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10326, Korea; (C.R.A.); (J.-E.K.); (Y.W.K.)
| | - Jinbong Park
- Department of Surgery, Beth Israel Deaconess Medical Center/Harvard Medical School, 330 Brookline Ave, Boston, MA 02215, USA;
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea;
| | - Jai-Eun Kim
- College of Korean Medicine, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10326, Korea; (C.R.A.); (J.-E.K.); (Y.W.K.)
| | - Kwang Seok Ahn
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea;
| | - Young Woo Kim
- College of Korean Medicine, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10326, Korea; (C.R.A.); (J.-E.K.); (Y.W.K.)
| | - Minjeong Jeong
- College of Korean Medicine, Woosuk University, 443 Samnye-ro, Samnye-eup, Wanju-gun, Jeollabuk-do 55338, Korea; (M.J.); (H.J.K.)
| | - Hong Jun Kim
- College of Korean Medicine, Woosuk University, 443 Samnye-ro, Samnye-eup, Wanju-gun, Jeollabuk-do 55338, Korea; (M.J.); (H.J.K.)
| | - Sun Hyang Park
- Department of Physiology, Research Institute for Endocrine Sciences, Medical School, Jeonbuk National University, 567 Baekje-daero, Dukjin-gu, Jeonju-si, Jeollabuk-do 54896, Korea;
| | - Seung Ho Baek
- College of Korean Medicine, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10326, Korea; (C.R.A.); (J.-E.K.); (Y.W.K.)
- Correspondence: ; Tel.: +82-31-961-5840
| |
Collapse
|
11
|
Corrigendum to "Activation of AMP-Activated Protein Kinase and Extracelluar Signal-Regulated Kinase Mediates CB-PIC-Induced Apoptosis in Hypoxic SW620 Colorectal Cancer Cells". EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:6289392. [PMID: 32774423 PMCID: PMC7400768 DOI: 10.1155/2020/6289392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 11/17/2022]
|
12
|
Kamarudin MNA, Sarker MMR, Zhou JR, Parhar I. Metformin in colorectal cancer: molecular mechanism, preclinical and clinical aspects. J Exp Clin Cancer Res 2019; 38:491. [PMID: 31831021 PMCID: PMC6909457 DOI: 10.1186/s13046-019-1495-2] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 11/28/2019] [Indexed: 02/07/2023] Open
Abstract
Growing evidence showed the increased prevalence of cancer incidents, particularly colorectal cancer, among type 2 diabetic mellitus patients. Antidiabetic medications such as, insulin, sulfonylureas, dipeptyl peptidase (DPP) 4 inhibitors and glucose-dependent insulinotropic peptide (GLP-1) analogues increased the additional risk of different cancers to diabetic patients. Conversely, metformin has drawn attention among physicians and researchers since its use as antidiabetic drug exhibited beneficial effect in the prevention and treatment of cancer in diabetic patients as well as an independent anticancer drug. This review aims to provide the comprehensive information on the use of metformin at preclinical and clinical stages among colorectal cancer patients. We highlight the efficacy of metformin as an anti-proliferative, chemopreventive, apoptosis inducing agent, adjuvant, and radio-chemosensitizer in various colorectal cancer models. This multifarious effects of metformin is largely attributed to its capability in modulating upstream and downstream molecular targets involved in apoptosis, autophagy, cell cycle, oxidative stress, inflammation, metabolic homeostasis, and epigenetic regulation. Moreover, the review highlights metformin intake and colorectal cancer risk based on different clinical and epidemiologic results from different gender and specific population background among diabetic and non-diabetic patients. The improved understanding of metformin as a potential chemotherapeutic drug or as neo-adjuvant will provide better information for it to be used globally as an affordable, well-tolerated, and effective anticancer agent for colorectal cancer.
Collapse
Affiliation(s)
- Muhamad Noor Alfarizal Kamarudin
- Brain Research Institute Monash Sunway (BRIMS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Selangor Malaysia
| | - Md. Moklesur Rahman Sarker
- Department of Pharmacy, State University of Bangladesh, 77 Satmasjid Road, Dhanmondi, Dhaka, 1205 Bangladesh
- Health Med Science Research Limited, 3/1 Block F, Lalmatia, Mohammadpur, Dhaka, 1207 Bangladesh
| | - Jin-Rong Zhou
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215 USA
| | - Ishwar Parhar
- Brain Research Institute Monash Sunway (BRIMS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Selangor Malaysia
| |
Collapse
|
13
|
Tan YJ, Lee YT, Petersen SH, Kaur G, Kono K, Tan SC, Majid AMSA, Oon CE. BZD9L1 sirtuin inhibitor as a potential adjuvant for sensitization of colorectal cancer cells to 5-fluorouracil. Ther Adv Med Oncol 2019; 11:1758835919878977. [PMID: 31632470 PMCID: PMC6767736 DOI: 10.1177/1758835919878977] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 09/06/2019] [Indexed: 02/06/2023] Open
Abstract
Background: This study aims to investigate the combination effect of a novel sirtuin
inhibitor (BZD9L1) with 5-fluorouracil (5-FU) and to determine its molecular
mechanism of action in colorectal cancer (CRC). Methods: BZD9L1 and 5-FU either as single treatment or in combination were tested
against CRC cells to evaluate synergism in cytotoxicity, senescence and
formation of micronucleus, cell cycle and apoptosis, as well as the
regulation of related molecular players. The effects of combined treatments
at different doses on stress and apoptosis, migration, invasion and cell
death mechanism were evaluated through two-dimensional and three-dimensional
cultures. In vivo studies include investigation on the
combination effects of BZD9L1 and 5-FU on colorectal tumour xenograft growth
and an evaluation of tumour proliferation and apoptosis using
immunohistochemistry. Results: Combination treatments exerted synergistic reduction on cell viability on HCT
116 cells but not on HT-29 cells. Combined treatments reduced survival,
induced cell cycle arrest, apoptosis, senescence and micronucleation in HCT
116 cells through modulation of multiple responsible molecular players and
apoptosis pathways, with no effect in epithelial mesenchymal transition
(EMT). Combination treatments regulated SIRT1 and SIRT2 protein expression
levels differently and changed SIRT2 protein localization. Combined
treatment reduced growth, migration, invasion and viability of HCT 116
spheroids through apoptosis, when compared with the single treatment. In
addition, combined treatment was found to reduce tumour growth in
vivo through reduction of tumour proliferation and necrosis
compared with the vehicle control group. This highlights the potential
therapeutic effects of BZD9L1 and 5-FU towards CRC. Conclusion: This study may pave the way for use of BZD9L1 as an adjuvant to 5-FU in
improving the therapeutic efficacy for the treatment of colorectal
cancer.
Collapse
Affiliation(s)
- Yi Jer Tan
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang, Malaysia
| | - Yeuan Ting Lee
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang, Malaysia
| | - Sven H Petersen
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Gurjeet Kaur
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang, Malaysia
| | - Koji Kono
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Soo Choon Tan
- USains Biomics Laboratory Testing Services Sdn. Bhd., Universiti Sains Malaysia, Penang, Malaysia
| | - Amin M S Abdul Majid
- EMAN Testing and Research Laboratories, Department of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang, Malaysia
| | - Chern Ein Oon
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang, 11800, Malaysia
| |
Collapse
|
14
|
Expression of miR‑542‑3p in osteosarcoma with miRNA microarray data, and its potential signaling pathways. Mol Med Rep 2018; 19:974-983. [PMID: 30569116 PMCID: PMC6323234 DOI: 10.3892/mmr.2018.9761] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 11/15/2018] [Indexed: 12/16/2022] Open
Abstract
Osteosarcoma (OS) is the most common pediatric primary bone tumor, with high malignancy rates and a poor prognosis following metastasis. At present, the role of microRNA (miR)-542-3p in OS remains to be elucidated. The purpose of the present study was to investigate the expression level of miR-542-3p in OS, and its potential molecular mechanisms, via a bioinformatics analysis. First, the expression of miR-542-3p in OS based on the continuous variables of the Gene Expression Omnibus database and PubMed was studied. Subsequently, the potential target genes of miR-542-3p were predicted using gene expression profiles and bioinformatics software. On the basis of the Database for Annotation, Visualization and Integrated Discovery, version 6.8, a study of gene ontology (GO) enrichment and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway knowledge base was conducted to explore the biological value of miR-542-3p in OS. Finally, the protein-protein interaction (PPI) network was completed using the STRING database. The expression of miR-542-3p in OS was revealed to be significantly higher compared with that in normal tissue. In total, 1,036 target genes of miR-542-3p were obtained. The results of the GO enrichment analysis revealed that the significant terms were ‘bone development’, ‘cell cycle arrest’ and ‘intracellular signal transduction’. The results of the KEGG analysis revealed the highlighted pathways that were targeted to miR-542-3p, including the sphingolipid signaling pathway (P=3.91×10−5), the phosphoinositide 3-kinase (PI3K)-AKT serine/threonine kinase (AKT) signaling pathway (P=3.17×10−5) and the insulin signaling pathway (P=1.04×10−5). The PPI network revealed eight hub genes: Ubiquitin-60S ribosomal protein L40, Ras-related C3 botulinum toxin substrate, mitogen-activated protein kinase 1, epidermal growth factor receptor, cystic fibrosis transmembrane conductance regulator, PI3K regulatory subunit 1, AKT1, and actin-related protein 2/3 complex subunit 1A, which may be the key target genes of miR-542-3p in OS. Taken together, these results have demonstrated that miR-542-3p was overexpressed in OS. The potential target genes and biological functions of miR-542-3p may provide novel insights into the differentially expressed genes that are involved in OS.
Collapse
|
15
|
Hong SH, Ismail IA, Kang SM, Han DC, Kwon BM. Cinnamaldehydes in Cancer Chemotherapy. Phytother Res 2016; 30:754-67. [PMID: 26890810 DOI: 10.1002/ptr.5592] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Revised: 01/15/2016] [Accepted: 01/22/2016] [Indexed: 12/16/2022]
Abstract
Cinnamaldehyde and cinnamaldehyde-derived compounds are candidates for the development of anticancer drugs that have received extensive research attention. In this review, we summarize recent findings detailing the positive and negative aspects of cinnamaldehyde and its derivatives as potential anticancer drug candidates. Furthermore, we describe the in vivo pharmacokinetics and metabolism of cinnamaldehydes. The oxidative and antioxidative properties of cinnamaldehydes, which contribute to their potential in chemotherapy, have also been discussed. Moreover, the mechanism(s) by which cinnamaldehydes induce apoptosis in cancer cells have been explored. In addition, evidence of the regulatory effects of cinnamaldehydes on cancer cell invasion and metastasis has been described. Finally, the application of cinnamaldehydes in treating various types of cancer, including breast, prostate, and colon cancers, has been discussed in detail. The effects of cinnamaldehydes on leukemia, hepatocellular carcinoma, and oral cancer have been summarized briefly. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Su-Hyung Hong
- Department of Oral Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, 700-412, Korea
| | - Ismail Ahmed Ismail
- Department of Oral Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, 700-412, Korea.,Laboratory of Molecular Cell Biology, Department of Zoology, Faculty of Science, Assiut University, Assiut, 71516, Egypt
| | - Sung-Min Kang
- Department of Oral Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, 700-412, Korea
| | - Dong Cho Han
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, University of Science and Technology in Korea, 125 Gwahakro Yoosunggu, Daejeon, 305-806, Korea
| | - Byoung-Mog Kwon
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, University of Science and Technology in Korea, 125 Gwahakro Yoosunggu, Daejeon, 305-806, Korea
| |
Collapse
|
16
|
Hsu CY, Lin CH, Lin JT, Cheng YF, Chen HM, Kao SH. Purine analogue ENERGI-F706 induces apoptosis of 786-O renal carcinoma cells via 5'-adenosine monophosphate-activated protein kinase activation. Mol Med Rep 2015; 12:4566-4571. [PMID: 26062651 DOI: 10.3892/mmr.2015.3906] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 05/01/2015] [Indexed: 11/06/2022] Open
Abstract
Purine compounds are known to activate 5'-adenosine monophosphate-activated protein kinase (AMPK), which has important roles in treatments for renal cell carcinoma. The present study was aimed to investigate the effects of the purine analogue ENERGI‑F706 on the human renal carcinoma cell line 786‑O and the underlying mechanisms. The results revealed that ENERGI‑F706 (0.2‑0.6 mg/ml) significantly decreased the cell viability to up to 36.4±2.4% of that of the control. Compared to 786‑O cells, ENERGI‑F706 exerted less suppressive effects on the viability of the human non‑tumorigenic renal cell line HK‑2. Flow cytometric analysis showed that ENERGI‑F706 contributed to cell cycle arrest at S‑phase and triggered apoptosis of 786‑O cells. Immunoblot analysis revealed that anti‑apoptotic B‑cell lymphoma 2 (Bcl‑2) levels were reduced and pro‑apoptotic Bcl‑2‑associated X protein levels were diminished. In addition, activation of caspase‑9, caspase‑3 and poly(adenosine diphosphate ribose) polymerase (PARP) was promoted in 786‑O cells in response to ENERGI‑F706. Effects of ENERGI‑F706 on AMPK cascades were investigated and the results showed that ENERGI‑F706 enhanced phosphorylation of AMPKα (T172) and p53 (S15), a downstream target of AMPK. In addition, the AMPK activation, p53 (S15) phosphorylation, reduction of Bcl‑2, cleavage of caspase‑3 and PARP as well as suppressed cell viability induced by ENERGI‑F706 were reversed in the presence of AMPK inhibitor compound C (dorsomorphin). In conclusion, the findings of the present study revealed that ENERGI‑F706 significantly suppressed the viability of 786‑O cells via induction of cell cycle arrest and apoptosis, attributing to AMPK and p53 activation and subsequent cell cycle regulatory and apoptotic signaling. It was therefore indicated that ENERGI‑F706 may be suitable for the treatment of renal cell carcinoma.
Collapse
Affiliation(s)
- Chao-Yu Hsu
- Division of Urology, Department of Surgery, Tungs' Taichung Metro Harbor Hospital, Taichung 435, Taiwan, R.O.C
| | - Chun-Hsiang Lin
- Institute of Biochemistry and Biotechnology, College of Medicine, Chung Shan Medical University, Taichung 402, Taiwan, R.O.C
| | - Jiun-Tsai Lin
- Institute of Applied Science and Engineering, Catholic Fu‑Jen University, New Taipei 242, Taiwan, R.O.C
| | - Yi-Fang Cheng
- Energenesis Biomedical Co. Ltd., New Taipei 235, Taiwan, R.O.C
| | - Han-Min Chen
- Institute of Applied Science and Engineering, Catholic Fu‑Jen University, New Taipei 242, Taiwan, R.O.C
| | - Shao-Hsuan Kao
- Institute of Biochemistry and Biotechnology, College of Medicine, Chung Shan Medical University, Taichung 402, Taiwan, R.O.C
| |
Collapse
|
17
|
Zheng F, Wu J, Zhao S, Luo Q, Tang Q, Yang L, Li L, Wu W, Hann SS. Baicalein increases the expression and reciprocal interplay of RUNX3 and FOXO3a through crosstalk of AMPKα and MEK/ERK1/2 signaling pathways in human non-small cell lung cancer cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:41. [PMID: 25948105 PMCID: PMC4457308 DOI: 10.1186/s13046-015-0160-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 04/20/2015] [Indexed: 12/19/2022]
Abstract
Background Baicalein, a natural flavonoid obtained from the Scutellaria baicalensis root, has been reported to inhibit growth of human lung cancer. However, the detailed mechanism underlying this has not been well elucidated. Methods Cell viability was measured using a 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assays. Apoptosis was detected by flow cytometry analysis and caspase 3/7 assays. The expression of RUNX3 and FOXO3a mRNA were measured by real time RT-PCR methods. Western blot analysis was performed to measure the phosphorylation and protein expression of AMP-activated protein kinase alpha (AMPKα) and extracellular signal-regulated kinase 1/2 (ERK1/2), runt-related transcription factor 3 (RUNX3) and forkhead box O3a (FOXO3a). Silencing of FOXO3a and RUNX3 were performed by small interfering RNA (siRNA) methods. Exogenous expression of FOXO3a or RUNX3 was carried out by electroporated transfection assays. Results We showed that baicalein significantly inhibited growth and induced apoptosis of non-small cell lung cancer (NSCLC) cells in a time- and dose-dependent manner. Baicalein induced RUNX3 and FOXO3a protein expression, and increased phosphorylation of AMPKα and ERK1/2. Moreover, the inhibitors of AMPK and MEK/ERK1/2 reversed the effect of baicalein on RUNX3 and FOXO3a protein expression. Interestingly, while compound C had little effect on blockade of baicalein-induced phosphorylation of ERK1/2, PD98059 significantly abrogated baicalein-induced phosphorylation of AMPKα. Intriguingly, while silencing of RUNX3 abolished the effect of baicalein on expression of FOXO3a and apoptosis, silencing of FOXO3a significantly attenuated baicalein-reduced cell proliferation. On the contrary, overexpression of FOXO3a restored the effect of baicalein on cell growth inhibition in cells silencing of endogenous FOXO3a gene and enhanced the effect of baicalein on RUNX3 protein expression. Finally, exogenous expression of RUNX3 increased FOXO3a protein and strengthened baicalein-induced phosphorylation of ERK1/2. Conclusion Collectively, our results show that baicalein inhibits growth and induces apoptosis of NSCLC cells through AMPKα- and MEK/ERK1/2-mediated increase and interaction of FOXO3a and RUNX3 protein. The crosstalk between AMPKα and MEK/ERK1/2 signaling pathways, and the reciprocal interplay of FOXO3a and RUNX3 converge on the overall response of baicalein. This study reveals a novel mechanism for regulating FOXO3a and RUNX3 signaling axis in response to baicalein and suggests a new strategy for NSCLC associated targeted therapy.
Collapse
Affiliation(s)
- Fang Zheng
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - Jingjing Wu
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - Shunyu Zhao
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - Qingmei Luo
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - Qing Tang
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - LiJun Yang
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - Liuning Li
- Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - WanYing Wu
- Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - Swei Sunny Hann
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China. .,Higher Education Mega Center, No. 55, Neihuan West Road, Panyu District, Guangzhou, Guangdong Province, 510006, People's Republic of China.
| |
Collapse
|
18
|
Zhao S, Wu J, Zheng F, Tang Q, Yang L, Li L, Wu W, Hann SS. β-elemene inhibited expression of DNA methyltransferase 1 through activation of ERK1/2 and AMPKα signalling pathways in human lung cancer cells: the role of Sp1. J Cell Mol Med 2015; 19:630-41. [PMID: 25598321 PMCID: PMC4369819 DOI: 10.1111/jcmm.12476] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 10/02/2014] [Indexed: 12/17/2022] Open
Abstract
β-elemene, a compound derived from Rhizoma zedoariae, is a promising new plant-derived drug with broad-spectrum anticancer activity. However, the underlying mechanism by which this agent inhibits human lung cancer cell growth has not been well elucidated. In this study, we showed that β-elemene inhibits human non-small cell lung carcinoma (NSCLC) cell growth, and increased phosphorylation of ERK1/2, Akt and AMPKα. Moreover, β-elemene inhibited expression of DNA methyltransferase 1 (DNMT1), which was not observed in the presence of the specific inhibitors of ERK (PD98059) or AMPK (compound C). Overexpression of DNMT1 reversed the effect of β-elemene on cell growth. Interestingly, metformin not only reversed the effect of β-elemene on phosphorylation of Akt but also strengthened the β-elemene-reduced DNMT1. In addition, β-elemene suppressed Sp1 protein expression, which was eliminated by either ERK1/2 or AMPK inhibitor. Conversely, overexpression of Sp1 antagonized the effect of β-elemene on DNMT1 protein expression and cell growth. Taken together, our results show that β-elemene inhibits NSCLC cell growth via ERK1/2- and AMPKα-mediated inhibition of transcription factor Sp1, followed by reduction in DNMT1 protein expression. Metformin augments the effect of β-elemene by blockade of Akt signalling and additively inhibition of DNMT1 protein expression. The reciprocal ERK1/2 and AMPKα signalling pathways contribute to the overall responses of β-elemene. This study reveals a potential novel mechanism by which β-elemene inhibits growth of NSCLC cells.
Collapse
Affiliation(s)
- ShunYu Zhao
- Laboratory of Tumor Biology, The second Clinical Medical Collage, University of Guangzhou Traditional Chinese MedicineGuangdong Province, China
| | - Jingjing Wu
- Laboratory of Tumor Biology, The second Clinical Medical Collage, University of Guangzhou Traditional Chinese MedicineGuangdong Province, China
| | - Fang Zheng
- Laboratory of Tumor Biology, The second Clinical Medical Collage, University of Guangzhou Traditional Chinese MedicineGuangdong Province, China
| | - Qing Tang
- Laboratory of Tumor Biology, The second Clinical Medical Collage, University of Guangzhou Traditional Chinese MedicineGuangdong Province, China
| | - LiJun Yang
- Laboratory of Tumor Biology, The second Clinical Medical Collage, University of Guangzhou Traditional Chinese MedicineGuangdong Province, China
| | - Liuning Li
- Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The second Clinical Medical Collage, University of Guangzhou Traditional Chinese MedicineGuangzhou, Guangdong Province, China
| | - WanYin Wu
- Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The second Clinical Medical Collage, University of Guangzhou Traditional Chinese MedicineGuangzhou, Guangdong Province, China
| | - Swei Sunny Hann
- Laboratory of Tumor Biology, The second Clinical Medical Collage, University of Guangzhou Traditional Chinese MedicineGuangdong Province, China
| |
Collapse
|
19
|
Shindo S, Hosokawa Y, Hosokawa I, Ozaki K, Matsuo T. Genipin inhibits MMP-1 and MMP-3 release from TNF-α-stimulated human periodontal ligament cells. Biochimie 2014; 107 Pt B:391-5. [DOI: 10.1016/j.biochi.2014.10.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 10/13/2014] [Indexed: 12/16/2022]
|
20
|
Chou CW, Cheng YW, Tsai CH. Phyllostachys edulis extract induces apoptosis signaling in osteosarcoma cells, associated with AMPK activation. DRUG DESIGN DEVELOPMENT AND THERAPY 2014; 8:1577-84. [PMID: 25284987 PMCID: PMC4181544 DOI: 10.2147/dddt.s69342] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Objective Bamboo is distributed worldwide, and its different parts are used as foods or as a traditional herb. Recently, antitumoral effects of bamboo extracts on several tumors have been increasingly reported; however, antitumoral activity of bamboo extracts on osteosarcoma remains unclear. In the present study, we investigated effects of an aqueous Phyllostachys edulis leaf extract (PEE) on osteosarcoma cells and the underlying mechanism of inhibition. Methods The growth of human osteosarcoma cell lines 143B and MG-63 and lung fibroblast MRC-5 cells was determined by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay. Apoptosis was demonstrated using TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling) assay and flow cytometric analysis. Phosphorylation and protein levels were determined by immunoblotting. Results After treatment with PEE, viability of 143B and MG-63 cells was dose-dependently reduced to 36.3%±1.6% of control values, which were similar to AICAR (5-aminoimidazole-4-carboxamide 1-β-D-ribofuranoside) treatments. In parallel, ratios of apoptotic cells and cells in the sub-G1 phase were significantly increased. Further investigation showed that PEE treatments led to activation of caspase cascades and changes of apoptotic mediators Bcl2, Bax, and p53. Consistently, our results revealed that PEE activated adenosine monophosphate-activated protein kinase (AMPK) signaling, and the AMPK activation was associated with the induction of apoptotic signaling. Conclusion Our results indicated that PEE suppressed the growth of 143B and MG-63 cells but moderately affected MRC-5 cells. PEE-induced apoptosis may attribute to AMPK activation and the following activation of apoptotic signaling cascades. These findings revealed that PEE possesses antitumoral activity on human osteosarcoma cells by manipulating AMPK signaling, suggesting that PEE alone or combined with regular antitumor drugs may be beneficial as osteosarcoma treatments.
Collapse
Affiliation(s)
- Chi-Wen Chou
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan ; Department of Orthopedics Surgery, National Taiwan University Hospital Yun-Lin Branch, Yun-Lin, Taiwan
| | - Ya-Wen Cheng
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chung-Hung Tsai
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|