1
|
Yang J, Chen D, He Q, Chen B, Pan Z, Zhang G, Li M, Li S, Xiao J, Wang H, Chen P, An Z. Arctiin alleviates knee osteoarthritis by suppressing chondrocyte oxidative stress induced by accumulated iron via AKT/NRF2/HO-1 signaling pathway. Sci Rep 2024; 14:31935. [PMID: 39738432 DOI: 10.1038/s41598-024-83383-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 12/13/2024] [Indexed: 01/02/2025] Open
Abstract
Iron overload (IO) was considered to be a risk factor for cartilage degradation in knee osteoarthritis (KOA) advancement. However, few drugs were found to improve cartilage degeneration by alleviating multiple cell death induced by the impaired iron level of the knee joints. We aimed to elucidate that Arctiin (ARC) plays a role in managing KOA caused by accumulated iron levels by restoring chondrocyte apoptosis and ferroptosis. Single-cell RNA sequencing analysis was used to discover the disparities in chondrocytes between KOA patients and non-KOA individuals. CCK-8 assay was performed to detect chondrocyte viability. Annexin V-FITC/PI staining determined the cell apoptosis rate. The fluorescence density reflected the iron content, ROS, lipid-ROS, and mitochondrial membrane potential. Q-RTPCR and Western Blotting were used to detect the expression levels of genes and proteins expression. Micro-CT and Safranin O-Fast Green staining were used to detect the phenotype of the knee joints. ARC increased cell viability and inhibited chondrocyte apoptosis. Further, ARC acts as an anti-ferroptosis effect by reducing the intracellular iron, ROS, and lipid-ROS content and restoring mitochondrial damage. Based on the results of scRNA-seq, we found that ARC can play a role by activating AKT/NRF2/HO-1 signaling pathway. In vivo, ARC can significantly improve the severity of KOA caused by IO. ARC alleviates oxidative stress in chondrocytes via the AKT/NRF2/HO-1 signaling pathway, suggesting the potential application of ARC in KOA.
Collapse
Affiliation(s)
- Junzheng Yang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Department of Orthopaedics, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, P. R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, P. R. China
| | - Delong Chen
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, P. R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, P. R. China
| | - Qi He
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, P. R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, P. R. China
| | - Baihao Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Department of Orthopaedics, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, P. R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, P. R. China
| | - Zhaofeng Pan
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, P. R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, P. R. China
| | - Gangyu Zhang
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Miao Li
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, P. R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, P. R. China
| | - Shaocong Li
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, P. R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, P. R. China
| | - Jiacong Xiao
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun Area, Guangzhou, P. R. China
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, P. R. China
| | - Haibin Wang
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, P. R. China.
| | - Peng Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Department of Orthopaedics, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, P.R. China.
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, P. R. China.
| | - Zhantian An
- Xinjiang Production and Construction Corps 13th division Red Star Hospital, Xinjiang Autonomous Region, 19 Qianjin East Road, Yizhou District, Xinjiang, P.R. China.
- The Affiliated Redstar Hospital of Shihezi University School of Medicine, Xinjiang Autonomous Region, 19 Qianjin East Road, Yizhou District, Xinjiang, P.R. China.
| |
Collapse
|
2
|
Tinivella A, Nwachukwu JC, Pinzi L, Dettori MA, Fabbri D, Carta P, Nettles KW, Rastelli G. Exploring Biological Targets of Magnolol and Honokiol and their Nature-Inspired Synthetic Derivatives: In Silico Identification and Experimental Validation of Estrogen Receptors. JOURNAL OF NATURAL PRODUCTS 2024. [PMID: 39526345 DOI: 10.1021/acs.jnatprod.4c00634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
In this work, we describe the results of a computational investigation aimed at identifying potential biological targets of honokiol, magnolol and a series of synthetic prodrug derivatives obtained through esterification of the free hydroxyl groups. The ligand-based and structure-based analyses revealed that these compounds potentially interact with several biological targets, some of which are known while others are new. Honokiol, magnolol, and three of the newly synthesized derivatives may bind to estrogen receptors ERα and ERβ. Biological testing confirmed that these compounds modulate estrogen-regulated transcriptional activity mediated by ERα or ERβ with potencies in the nanomolar range. In particular, magnolol and one of its derivatives (10) behaved as partial antagonists of ERα and ERβ, while compounds 8 and 11 behaved as partial agonists. These findings validate the computational predictions and shed light on the mechanism of action of these natural compounds, paving the way for further investigation in the context of targeted therapies.
Collapse
Affiliation(s)
- Annachiara Tinivella
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Jerome C Nwachukwu
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, Florida 33458, United States
| | - Luca Pinzi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | | | - Davide Fabbri
- Institute of Biomolecular Chemistry of CNR, Traversa La Crucca 3, 07100 Sassari, Italy
| | - Paola Carta
- Institute of Biomolecular Chemistry of CNR, Traversa La Crucca 3, 07100 Sassari, Italy
| | - Kendall W Nettles
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, Florida 33458, United States
| | - Giulio Rastelli
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| |
Collapse
|
3
|
Kundu M, Das S, Das CK, Kulkarni G, Das S, Dhara D, Mandal M. Magnolol induces cytotoxic autophagy in glioma by inhibiting PI3K/AKT/mTOR signaling. Exp Cell Res 2023; 424:113488. [PMID: 36736226 DOI: 10.1016/j.yexcr.2023.113488] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 12/18/2022] [Accepted: 01/18/2023] [Indexed: 02/04/2023]
Abstract
Glioma is difficult-to-treat because of its infiltrative nature and the presence of the blood-brain barrier. Temozolomide is the only FDA-approved drug for its management. Therefore, finding a novel chemotherapeutic agent for glioma is of utmost importance. Magnolol, a neolignan, has been known for its apoptotic role in glioma. In this work, we have explored a novel anti-glioma mechanism of Magnolol associated with its role in autophagy modulation. We found increased expression levels of Beclin-1, Atg5-Atg12, and LC3-II and lower p62 expression in Magnolol-treated glioma cells. PI3K/AKT/mTOR pathway proteins were also downregulated in Magnolol-treated glioma cells. Next, we treated the glioma cells with Insulin, a stimulator of PI3K/AKT/mTOR signaling, to confirm that Magnolol induced autophagy by inhibiting this pathway. Insulin reversed the effect on Magnolol-mediated autophagy induction. We also established the same in in vivo glioma model where Magnolol showed an anti-glioma effect by inducing autophagy. To confirm the cytotoxic effect of Magnolol-induced autophagy, we used Chloroquine, a late-stage autophagy inhibitor. Chloroquine efficiently reversed the anti-glioma effects of Magnolol both in vitro and in vivo. Our study revealed the cytotoxic effect of Magnolol-induced autophagy in glioma, which was not previously reported. Additionally, Magnolol showed no toxicity in non-cancerous cell lines as well as rat organs. Thus, we concluded that Magnolol is an excellent candidate for developing new therapeutic strategies for glioma management.
Collapse
Affiliation(s)
- Moumita Kundu
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| | - Subhayan Das
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| | - Chandan Kanta Das
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| | - Gaurav Kulkarni
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| | - Soumen Das
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| | - Dibakar Dhara
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, India.
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| |
Collapse
|
4
|
Lai YH, Liu WL, Lee TY, Kuo CW, Liu YR, Huang CY, Chen YH, Chen IL, Wu SH, Wang SC, Lee PY, Liu CC, Lo J, Chang YC, Kuo HF, Hsieh CC, Li CY, Liu PL. Magnolol regulates miR-200c-3p to inhibit epithelial-mesenchymal transition and retinoblastoma progression by modulating the ZEB1/E-cadherin axis in vitro and in vivo. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 110:154597. [PMID: 36603340 DOI: 10.1016/j.phymed.2022.154597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 11/21/2022] [Accepted: 12/10/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Retinoblastoma, the most common pediatric intraocular malignancy, can develop during embryogenesis, with most children being diagnosed at 3-4 years of age. Multimodal therapies are typically associated with high levels of cytotoxicity and side effects. Therefore, the development of novel treatments with minimal side effects is crucial. Magnolol has a significant anti-tumor effect on various cancers. However, its antitumor effect on retinoblastoma remains unclear. PURPOSE The study aimed to determine the effects of magnolol on the regulation of EMT, migration, invasion, and cancer progression in retinoblastoma and the modulation of miR-200c-3p expression and the Wnt/ zinc finger E-box binding homeobox 1 (ZEB1)/E-cadherin axis in vivo and in vitro. METHODS The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) (MTT) assay was used to evaluate magnolol-induced cell toxicity in the Y79 retinoblastoma cell line. Flow cytometry and immunostaining assays were performed to investigate the magnolol-regulated mitochondrial membrane potential and the intracellular and mitochondrial reactive oxygen species levels in Y79 retinoblastoma cells. Orthotopic and subcutaneous xenograft experiments were performed in eight-week-old male null mice to study retinoblastoma progression and metastasis. In situ hybridization and quantitative reverse transcription polymerase chain reaction (RT-qPCR) assays were performed to evaluate the level of the anti-cancer miRNA miR-200c-3p. The mRNA and protein levels of E-cadherin, β-catenin, α-smooth muscle actin (α-SMA), fibronectin-1, and ZEB1 were analyzed using RT-qPCR, immunoblot, immunocytochemistry, and immunohistochemistry assays in vitro and in vivo. RESULTS Magnolol increased E-cadherin levels and reduced the activation of the EMT signaling pathway, EMT, tumor growth, metastasis, and cancer progression in the Y79 retinoblastoma cell line as well as in the orthotopic and subcutaneous xenograft animal models. Furthermore, magnolol increased the expression of miR-200c-3p. Our results demonstrate that miRNA-200c-3p inhibits EMT progression through the Wnt16/β-catenin/ZEB1/E-cadherin axis, and the ZEB1 silencing response shows that miR-200c-3p regulates ZEB1-mediated EMT in retinoblastoma. CONCLUSION Magnolol has an antitumor effect by increasing E-cadherin and miRNA-200c-3p expression to regulate ZEB1-mediated EMT and cancer progression in retinoblastoma. The anti-tumor effect of magnolol by increasing E-cadherin and miRNA-200c-3p expression to regulate ZEB1-mediated EMT and cancer progression in retinoblastoma has been elucidated for the first time.
Collapse
Affiliation(s)
- Yu-Hung Lai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Ophthalmology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Wei-Lun Liu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan; Division of Critical Care Medicine, Department of Emergency and Critical Care Medicine, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City 24205, Taiwan
| | - Tsung-Ying Lee
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chung-Wen Kuo
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yu-Ru Liu
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chi-Yuan Huang
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yung-Hsiang Chen
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan; Department of Psychology, College of Medical and Health Science, Asia University, Taichung 41354, Taiwan
| | - I-Ling Chen
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Szu-Hui Wu
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Shu-Chi Wang
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Po-Yen Lee
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ching-Chih Liu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Ophthalmology, Chi Mei Medical Center, Tainan 71004, Taiwan
| | - Jung Lo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Yo-Chen Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Ophthalmology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Hsuan-Fu Kuo
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chong-Chao Hsieh
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Division of Cardiovascular Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan.
| | - Po-Len Liu
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
5
|
Naghashpour M, Dayer D, Karami H, Naghashpour M, Moghadam MT, Haeri SMJ, Suzuki K. Evaluating the Magnolol Anticancer Potential in MKN-45 Gastric Cancer Cells. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59020286. [PMID: 36837487 PMCID: PMC9963572 DOI: 10.3390/medicina59020286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/20/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023]
Abstract
Background and Objectives: Combination therapy improves the effect of chemotherapy on tumor cells. Magnolol, used in treating gastrointestinal disorders, has been shown to have anti-cancer properties. We investigated the synergistic effect of cisplatin and magnolol on the viability and maintenance of MKN-45 gastric cancer cells. Materials and Methods: The toxicity of magnolol and/or cisplatin was determined using the MTT technique. The trypan blue method was used to test magnolol and/or cisplatin's effect on MKN-45 cell growth. Crystal violet staining was used to assess the treated cells' tendency for colony formation. The expression of genes linked to apoptosis, cell cycle arrest, and cell migration was examined using the qPCR method. Results: According to MTT data, using magnolol and/or cisplatin significantly reduced cell viability. The ability of the treated cells to proliferate and form colonies was also reduced considerably. Magnolol and/or cisplatin treatment resulted in a considerable elevation in Bax expression. However, the level of Bcl2 expression was dramatically reduced. p21 and p53 expression levels were significantly increased in the treated cells, while MMP-9 expression was significantly reduced. Conclusions: These findings show that magnolol has a remarkable anti-tumor effect on MKN-45 cells. In combination with cisplatin, magnolol may be utilized to overcome cisplatin resistance in gastric cancer cells.
Collapse
Affiliation(s)
- Mahsa Naghashpour
- Department of Anatomical Sciences, Medical School, Arak University of Medical Sciences, Arak 38481-7-6341, Iran
| | - Dian Dayer
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 61357-15794, Iran
| | - Hadi Karami
- Department of Molecular Medicine and Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak 38481-7-6341, Iran
| | - Mahshid Naghashpour
- Department of Basic Medical Sciences, Faculty of Medicine, Abadan University of Medical Sciences, Abadan 6313833177, Iran
| | - Mahin Taheri Moghadam
- Department of Anatomical Science, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 61357-15753, Iran
| | - Seyed Mohammad Jafar Haeri
- Department of Anatomical Sciences, Medical School, Arak University of Medical Sciences, Arak 38481-7-6341, Iran
- Correspondence: (S.M.J.H.); (K.S.); Tel.: +98-9123276391 (S.M.J.H.)
| | - Katsuhiko Suzuki
- Faculty of Sport Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa 359-1192, Japan
- Correspondence: (S.M.J.H.); (K.S.); Tel.: +98-9123276391 (S.M.J.H.)
| |
Collapse
|
6
|
Wang X, Liu Q, Fu Y, Ding RB, Qi X, Zhou X, Sun Z, Bao J. Magnolol as a Potential Anticancer Agent: A Proposed Mechanistic Insight. Molecules 2022; 27:molecules27196441. [PMID: 36234977 PMCID: PMC9570903 DOI: 10.3390/molecules27196441] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/27/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer is a serious disease with high mortality and morbidity worldwide. Natural products have served as a major source for developing new anticancer drugs during recent decades. Magnolol, a representative natural phenolic lignan isolated from Magnolia officinali, has attracted considerable attention for its anticancer properties in recent years. Accumulating preclinical studies have demonstrated the tremendous therapeutic potential of magnolol via a wide range of pharmacological mechanisms against cancer. In this review, we summarized the latest advances in preclinical studies investigating anticancer properties of magnolol and described the important signaling pathways explaining its underlying mechanisms. Magnolol was capable of inhibiting cancer growth and metastasis against various cancer types. Magnolol exerted anticancer effects through inhibiting proliferation, inducing cell cycle arrest, provoking apoptosis, restraining migration and invasion, and suppressing angiogenesis. Multiple signaling pathways were also involved in the pharmacological actions of magnolol against cancer, such as PI3K/Akt/mTOR signaling, MAPK signaling and NF-κB signaling. Based on this existing evidence summarized in the review, we have conclusively confirmed magnolol had a multi-target anticancer effect against heterogeneous cancer disease. It is promising to develop magnolol as a drug candidate for cancer therapy in the future.
Collapse
Affiliation(s)
- Xiaofeng Wang
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou 570102, China
| | - Qingqing Liu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Yuanfeng Fu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Ren-Bo Ding
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Xingzhu Qi
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Xuejun Zhou
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou 570102, China
| | - Zhihua Sun
- State International Joint Research Center for Animal Health Breeding, Key Laboratory of Control and Prevention of Animal Disease of Xinjiang Production & Construction Corps, College of Animal Science and Technology, Shihezi University, Shihezi 832003, China
- Correspondence: (Z.S.); (J.B.)
| | - Jiaolin Bao
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
- Correspondence: (Z.S.); (J.B.)
| |
Collapse
|
7
|
A comprehensive insight into the antineoplastic activities and molecular mechanisms of deoxypodophyllotoxin: Recent trends, challenges, and future outlook. Eur J Pharmacol 2022; 928:175089. [PMID: 35688183 DOI: 10.1016/j.ejphar.2022.175089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 11/20/2022]
Abstract
Lignans constitute an important group of polyphenols, which have been demonstrated to potently suppress cancer cell proliferation. Numerous in vitro and in vivo studies indicate that deoxypodophyllotoxin as a natural lignan possesses potent anticancer activities against various types of human cancer. The purpose of current review is to provide the reader with the latest findings in understanding the anticancer effects and molecular mechanisms of deoxypodophyllotoxin. This review comprehensively describes the influence of deoxypodophyllotoxin on signaling cascades and molecular targets implicated in cancer cell proliferation and invasion. A number of various signaling molecules and pathways, including apoptosis, necroptosis, cell cycle, angiogenesis, vascular disruption, ROS, MMPs, glycolysis, and microtubules as well as NF-κB, PI3K/Akt/mTOR, and MAPK cascades have been reported to be responsible for the anticancer activities of deoxypodophyllotoxin. The results of present review suggest that the cyclolignan deoxypodophyllotoxin can be developed as a novel and potent anticancer agent, especially as an alternative option for treatment of resistant tumors to chemotherapy.
Collapse
|
8
|
Phan HTL, Nam YR, Kim HJ, Woo JH, NamKung W, Nam JH, Kim WK. In-vitro and in-vivo anti-allergic effects of magnolol on allergic rhinitis via inhibition of ORAI1 and ANO1 channels. JOURNAL OF ETHNOPHARMACOLOGY 2022; 289:115061. [PMID: 35114342 DOI: 10.1016/j.jep.2022.115061] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 10/15/2021] [Accepted: 01/28/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Flos Magnoliae (the dried flower buds of Magnolia biondii Pamp, FM) is a known herbal traditional medicine used for the symptomatic relief of nasal congestion and rhinorrhea caused by rhinitis and sinusitis. Magnolol, a neolignan from the magnolia family, is a secondary metabolite known to have anti-allergic and anti-inflammatory effects. However, the underlying mechanisms and therapeutic effect of magnolol in the treatment of allergic rhinitis (AR) remain elusive. AIMS OF THE STUDY Anoctamin 1 (ANO1), a calcium-activated anion channel, mediates mucus and electrolyte secretion in nasal airway epithelial cells, whereas calcium release-activated calcium channel protein 1 (ORAI1) participates in the activation of T-lymphocytes and mast cells. The aim of our study is to understand the mechanisms of action of magnolol against AR, i.e., whether it acts through the modulation of ANO1 and ORAI1 channels that are expressed in nasal epithelial cells and T-lymphocytes, respectively. MATERIALS AND METHODS Whole-cell patch clamp was used to record the activity of ORAI1 and ANO1 ion channels in ORAI1 or ANO1 overexpressed HEK293T cells, while the Ussing chamber apparatus was used to measure electrolyte transport via the epithelium, in Calu-3 cells cultured in an air-liquid interface. Additionally, calcium imaging of Jurkat T-lymphocytes was used to assess changes in the intracellular calcium concentration. Magnolol toxicity was assessed using the CCK-8 assay, and its effect on T-lymphocyte proliferation was measured by labeling human primary T-lymphocytes with carboxyfluorescein succinimidyl ester. Finally, OVA-induced Balb/c mice were employed to evaluate the effect of magnolol on nasal symptoms, as well as cytokine and eosinophil infiltration in AR. RESULTS Magnolol inhibits ORAI1 and ANO1 channels in a concentration-dependent manner. Magnolol (30 μM) inhibits anti-CD3 induced cellular proliferation and production of IL-2 via ORAI1 channels in T-lymphocytes. Further, ATP-induced electrolyte transport mediated by ANO1 channels is significantly inhibited by magnolol in IL-4 sensitized Calu-3 cells. Notably, 300 μM magnolol significantly attenuates cytokine and eosinophil infiltration, thus alleviating AR symptoms in mice OVA-induced AR. CONCLUSION Magnolol may be a promising therapeutic agent for the treatment and prevention of AR.
Collapse
Affiliation(s)
- Hong Thi Lam Phan
- Department of Physiology, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju, 38066, Republic of Korea; Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang, Gyeonggi-do, 10326, Republic of Korea
| | - Yu Ran Nam
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang, Gyeonggi-do, 10326, Republic of Korea
| | - Hyun Jong Kim
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang, Gyeonggi-do, 10326, Republic of Korea
| | - Joo Han Woo
- Department of Physiology, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju, 38066, Republic of Korea
| | - Wan NamKung
- Department of Integrated OMICS for Biomedical Science, WCU Program of Graduate School, Yonsei University, Seoul, 03722, Republic of Korea
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju, 38066, Republic of Korea; Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang, Gyeonggi-do, 10326, Republic of Korea.
| | - Woo Kyung Kim
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang, Gyeonggi-do, 10326, Republic of Korea; Department of Internal Medicine Graduate School of Medicine, Dongguk University, 27 Dongguk-ro, Ilsan Dong-gu, Goyang, Gyeonggi-do, 10326, Republic of Korea.
| |
Collapse
|
9
|
Tang Y, Wang L, Yi T, Xu J, Wang J, Qin JJ, Chen Q, Yip KM, Pan Y, Hong P, Lu Y, Shen HM, Chen HB. Synergistic effects of autophagy/mitophagy inhibitors and magnolol promote apoptosis and antitumor efficacy. Acta Pharm Sin B 2021; 11:3966-3982. [PMID: 35024319 PMCID: PMC8727919 DOI: 10.1016/j.apsb.2021.06.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/20/2021] [Accepted: 04/27/2021] [Indexed: 12/14/2022] Open
Abstract
Mitochondria as a signaling platform play crucial roles in deciding cell fate. Many classic anticancer agents are known to trigger cell death through induction of mitochondrial damage. Mitophagy, one selective autophagy, is the key mitochondrial quality control that effectively removes damaged mitochondria. However, the precise roles of mitophagy in tumorigenesis and anticancer agent treatment remain largely unclear. Here, we examined the functional implication of mitophagy in the anticancer properties of magnolol, a natural product isolated from herbal Magnolia officinalis. First, we found that magnolol induces mitochondrial depolarization, causes excessive mitochondrial fragmentation, and increases mitochondrial reactive oxygen species (mtROS). Second, magnolol induces PTEN-induced putative kinase protein 1 (PINK1)‒Parkin-mediated mitophagy through regulating two positive feedforward amplification loops. Third, magnolol triggers cancer cell death and inhibits neuroblastoma tumor growth via the intrinsic apoptosis pathway. Moreover, magnolol prolongs the survival time of tumor-bearing mice. Finally, inhibition of mitophagy by PINK1/Parkin knockdown or using inhibitors targeting different autophagy/mitophagy stages significantly promotes magnolol-induced cell death and enhances magnolol's anticancer efficacy, both in vitro and in vivo. Altogether, our study demonstrates that magnolol can induce autophagy/mitophagy and apoptosis, whereas blockage of autophagy/mitophagy remarkably enhances the anticancer efficacy of magnolol, suggesting that targeting mitophagy may be a promising strategy to overcome chemoresistance and improve anticancer therapy.
Collapse
Affiliation(s)
- Yancheng Tang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Liming Wang
- School of Biomedical Sciences, Hunan University, Changsha 410082, China
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Tao Yi
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Jun Xu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Jigang Wang
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- The First Affiliated Hospital of Southern University of Science and Technology, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen 518020, China
| | - Jiang-Jiang Qin
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Qilei Chen
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Ka-Man Yip
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Yihang Pan
- Department of Medical Research, Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Peng Hong
- Department of Medical Research, Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Yingying Lu
- Department of Medical Research, Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
- Department of Biomedical Science, City University of Hong Kong, Hong Kong SAR 999077, China
- Corresponding authors. Tel./fax: +852 93590902.
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- Corresponding authors. Tel./fax: +852 93590902.
| | - Hu-Biao Chen
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
- Corresponding authors. Tel./fax: +852 93590902.
| |
Collapse
|
10
|
Jatal R, Osman R, Mamdouh W, Awad GA. Lung targeted electrosprayed chitosan nanocomposite microparticles boost the cytotoxic activity of magnolol. CARBOHYDRATE POLYMER TECHNOLOGIES AND APPLICATIONS 2021. [DOI: 10.1016/j.carpta.2021.100169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
11
|
Lai YH, Lee PY, Lu CY, Liu YR, Wang SC, Liu CC, Chang YC, Chen YH, Su CC, Li CY, Liu PL. Thrombospondin 1-induced exosomal proteins attenuate hypoxia-induced paraptosis in corneal epithelial cells and promote wound healing. FASEB J 2021; 35:e21200. [PMID: 33341997 DOI: 10.1096/fj.202001106rrr] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 10/15/2020] [Accepted: 11/03/2020] [Indexed: 12/14/2022]
Abstract
Thrombospondin-1 (TSP1) is involved in corneal wound healing caused by chemical injury. Herein, we examined the effects of TSP1 on hypoxia-induced damages and wound-healing activity in human corneal epithelial (HCE) cells. Exosomal protein expression was determined using liquid chromatography-tandem mass spectrometry, and HCE cell migration and motility were examined through wound-healing assay and time-lapse microscopy. Reestablishment of cell junctions by TSP1 was assessed through confocal microscopy and 3D image reconstruction. Our results show that CoCl2 -induced hypoxia promoted HCE cell death by paraptosis. TSP1 protected these cells against paraptosis by attenuating mitochondrial membrane potential depletion, swelling and dilation of endoplasmic reticulum and mitochondria, and mitochondrial fission. Exosomes isolated from HCE cells treated with TSP1 contained wound healing-associated proteins that were taken up by HCE cells to promote tissue remodeling and repair. TSP1 protected HCE cells against hypoxia-induced damages and inhibited paraptosis progression by promoting cell migration, cell-cell adhesion, and extracellular matrix remodeling. These findings indicate that TSP1 ameliorates hypoxia-induced paraptosis in HCE cells and promotes wound healing and remodeling by regulating exosomal protein expression. TSP1 may, therefore, play important roles in the treatment of hypoxia-associated corneal diseases.
Collapse
Affiliation(s)
- Yu-Hung Lai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Ophthalmology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Po-Yen Lee
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chi-Yu Lu
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Ru Liu
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shu-Chi Wang
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Chih Liu
- Department of Ophthalmology, Chi Mei Medical Center, Tainan, Taiwan
| | - Yo-Chen Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Ophthalmology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yung-Hsiang Chen
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chia-Cheng Su
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Po-Len Liu
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| |
Collapse
|
12
|
Mottaghi S, Abbaszadeh H. Natural Lignans Honokiol and Magnolol as Potential Anticarcinogenic and Anticancer Agents. A Comprehensive Mechanistic Review. Nutr Cancer 2021; 74:761-778. [PMID: 34047218 DOI: 10.1080/01635581.2021.1931364] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Plant lignans constitute an important group of polyphenols, which have been demonstrated to significantly induce cancer cell death and suppress cancer cell proliferation with minimal toxicity against non-transformed cells. Numerous epidemiological studies have shown that the intake of lignans is associated with lower risk of several cancers. These natural compounds have the potential to inhibit carcinogenesis, tumor growth, and metastasis by targeting various signaling molecules and pathways. Growing evidence indicates that honokiol and magnolol as natural lignans possess potent anticancer activities against various types of human cancer. The aim of present review is to provide the reader with the newest findings in understanding the cellular and molecular mechanisms mediating anticancer effects of honokiol and magnolol. This review comprehensively elucidates the effects of honokiol and magnolol on the molecular targets and signal transduction pathways implicated in cancer cell proliferation and metastasis. The findings of current review indicate that honokiol and magnolol can be considered as promising carcinopreventive and anticancer agents.
Collapse
Affiliation(s)
- Sayeh Mottaghi
- Department of Pediatrics, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hassan Abbaszadeh
- Department of Pharmacology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
13
|
Mottaghi S, Abbaszadeh H. A comprehensive mechanistic insight into the dietary and estrogenic lignans, arctigenin and sesamin as potential anticarcinogenic and anticancer agents. Current status, challenges, and future perspectives. Crit Rev Food Sci Nutr 2021; 62:7301-7318. [PMID: 33905270 DOI: 10.1080/10408398.2021.1913568] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A large body of evidence indicates that lignans as polyphenolic compounds are beneficial against life-threatening diseases such as cancer. Plant lignans have the potential to induce cancer cell death and interfere with carcinogenesis, tumor growth, and metastasis. Epidemiological studies have revealed that the intake of lignans is inversely associated with the risk of several cancers. Moreover, numerous experimental studies demonstrate that natural lignans significantly suppress cancer cell proliferation with minimal toxicity against non-transformed cells. Dietary lignans arctigenin and sesamin have been found to have potent antiproliferative activities against various types of human cancer. The purpose of this review is to provide the reader with a deeper understanding of the cellular and molecular mechanisms underlying anticancer effects of arctigenin and sesamin. Our review comprehensively describes the effects of arctigenin and sesamin on the signaling pathways and related molecules involved in cancer cell proliferation and invasion. The findings of present review show that the dietary lignans arctigenin and sesamin seem to be promising carcinopreventive and anticancer agents. These natural lignans can be used as dietary supplements and pharmaceuticals for prevention and treatment of cancer.
Collapse
Affiliation(s)
- Sayeh Mottaghi
- Department of Pediatrics, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hassan Abbaszadeh
- Department of Pharmacology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
14
|
Chen Y, He X, Feng D, Li S. Compare the Effects of Magnolol on Gastric Cancer Cells Through c-Jun N-Terminal Kinase Signaling Pathway and Gold Magnetic. JOURNAL OF NANOSCIENCE AND NANOTECHNOLOGY 2021; 21:943-948. [PMID: 33183428 DOI: 10.1166/jnn.2021.18688] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
This article explores the effects and mechanisms of magnolol on the proliferation of gastric cancer cells as well as the apoptosis. First, 0 (control group), 20, 40, and 80 /x mol/L magnolol were observed on SGC-7901 cells for 24, 48, and 72 h. We use MTT method to measure the cell viability, and apoptosis and cells were detected by flow cytometry. Cell proliferation inhibition rate, apoptosis and cell cycle experiments showed that P-value < 0.05 means the difference is statistically significant. And the results which compare the control group, the 20, 40, and 80 /x mol/L show that honokiol had lower cell viability (P < 0.01), increased apoptotic rate (P < 0.01), and cell cycle stay in the G1 phase (P < 0.01), so we found that honokiol may suppress the proliferation of SGC-7901 cells and stimulate apoptosis by regulating cyclin and apoptosis-related proteins. With the development of nanomaterials synthesis technology and application in biomedicine, gold magnetic composite nanomaterials have unique properties, so they have been widely concerned in many applications. We combine gold and magnetic nanomaterials through other nanostructures to achieve the integration of diagnosis and treatment of tumors. We have synthesized two kinds of gold magnetic nanocomposites, GNR-PPy-FA nanocomposites. With the role of chemotherapy and heat and light therapy, GNR-PPy-FA nanocomposites have high light-to-heat conversion efficiency. Cell experiments verify the effect of chemotherapy and photothermal treatment of composite nanomaterials. After incubation with gold magnetic composite nanomaterials, the cell survival rate of tumor cells decreased to about 15%. In addition, both types of gold magnetic nanocomposites have the ability to dually target cancer cells, and the modification of folic acid and cancer cell membranes makes the material more biocompatible.
Collapse
Affiliation(s)
- Yu Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Xiuyun He
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Dagang Feng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Shijie Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| |
Collapse
|
15
|
Hsieh C, Li C, Hsu C, Chen H, Chen Y, Liu Y, Liu Y, Kuo H, Liu P. Mitochondrial protection by simvastatin against angiotensin II-mediated heart failure. Br J Pharmacol 2019; 176:3791-3804. [PMID: 31265743 PMCID: PMC6780047 DOI: 10.1111/bph.14781] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 06/04/2019] [Accepted: 06/06/2019] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND PURPOSE Mitochondrial dysfunction plays a role in the progression of cardiovascular diseases including heart failure. 3-Hydroxy-3-methylglutaryl-CoA reductase inhibitors (statins), which inhibit ROS synthesis, show cardioprotective effects in chronic heart failure. However, the beneficial role of statins in mitochondrial protection in heart failure remains unclear. EXPERIMENTAL APPROACH Rats were treated with angiotensin II (1.5 mg·kg-1 ·day-1 ) or co-administered simvastatin (oral, 10 mg·kg-1 ) for 14 days; and then administration was stopped for the following 14 days. Cardiac structure/function was examined by wheat germ agglutinin staining and echocardiography. Mitochondrial morphology and the numbers of lipid droplets, lysosomes, autophagosomes, and mitophagosomes were determined by transmission electron microscopy. Human cardiomyocytes were stimulated, and intracellular ROS and mitochondrial membrane potential (ΔΨm ) changes were measured by flow cytometry and JC-1 staining, respectively. Autophagy and mitophagy-related and mitochondria-regulated apoptotic proteins were identified by immunohistochemistry and western blotting. KEY RESULTS Simvastatin significantly reduced ROS production and attenuated the disruption of ΔΨm . Simvastatin induced the accumulation of lipid droplets to provide energy for maintaining mitochondrial function, promoted autophagy and mitophagy, and inhibited mitochondria-mediated apoptosis. These findings suggest that mitochondrial protection mediated by simvastatin plays a therapeutic role in heart failure prevention by modulating antioxidant status and promoting energy supplies for autophagy and mitophagy to inhibit mitochondrial damage and cardiomyocyte apoptosis. CONCLUSION AND IMPLICATIONS Mitochondria play a key role in mediating heart failure progression. Simvastatin attenuated heart failure, induced by angiotensin II, via mitochondrial protection and might provide a new therapy to prevent heart failure.
Collapse
Affiliation(s)
- Chong‐Chao Hsieh
- Graduate Institute of Clinical Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Division of Cardiovascular Surgery, Department of SurgeryKaohsiung Medical University HospitalKaohsiungTaiwan
| | - Chia‐Yang Li
- Graduate Institute of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Center for Infectious Disease and Cancer ResearchKaohsiung Medical UniversityKaohsiungTaiwan
| | - Chih‐Hsin Hsu
- Department of Internal MedicineCheng Kung University HospitalTainanTaiwan
| | - Hsiu‐Lin Chen
- Department of Respiratory Therapy, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
| | - Yung‐Hsiang Chen
- Graduate Institute of Integrated Medicine, College of Chinese MedicineChina Medical UniversityTaichungTaiwan
- Department of Psychology, College of Medical and Health ScienceAsia UniversityTaichungTaiwan
| | - Yu‐Peng Liu
- Graduate Institute of Clinical Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
| | - Yu‐Ru Liu
- Department of Respiratory Therapy, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
| | - Hsuan‐Fu Kuo
- Graduate Institute of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Department of Internal Medicine, Kaohsiung Municipal Ta‐Tung HospitalKaohsiung Medical UniversityKaohsiungTaiwan
| | - Po‐Len Liu
- Department of Respiratory Therapy, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Regenerative Medicine and Cell Therapy Research CenterKaohsiung Medical UniversityKaohsiungTaiwan
| |
Collapse
|
16
|
Lin TH, Chen SI, Su YC, Lin MC, Lin HJ, Huang ST. Conventional Western Treatment Combined With Chinese Herbal Medicine Alleviates the Progressive Risk of Lung Cancer in Patients With Chronic Obstructive Pulmonary Disease: A Nationwide Retrospective Cohort Study. Front Pharmacol 2019; 10:987. [PMID: 31572178 PMCID: PMC6753872 DOI: 10.3389/fphar.2019.00987] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 07/31/2019] [Indexed: 12/24/2022] Open
Abstract
Background and purpose: Lung cancer has high global incidence and mortality rates. Chronic obstructive pulmonary disease (COPD) is strongly associated with lung cancer and is an independent risk factor for lung cancer with or without smoking. Chinese herbal medicines (CHMs) are used to treat COPD. This study sought to determine whether CHM treatment effectively decreases the incidence of lung cancer in COPD patients receiving conventional Western medical treatment. Methods: Records obtained from the National Health Insurance Research Database (NHIRD) were used to identify 81,780 adults aged ≥18 years newly diagnosed with COPD in Taiwan between 2000 and 2010. Among them, 11,180 received CHMs after COPD diagnosis and 23,319 did not (non-CHM). After excluding patients with missing basic demographic information, each group consisted of 2,682 patients. Statistical methods analyzed the baseline characteristics for both groups and we performed a Cox proportional hazard regression analysis to examine the incidence of lung cancer. The cumulative incidence of lung cancer in COPD patients with or without CHM treatment was calculated by the Kaplan-Meier method. The association between herbs and formulas was examined by NodeXL to perform a network analysis of CHM. Results: COPD patients using CHM had a lower risk for lung cancer (adjusted hazards ratio [aHR] = 0.36, 95% confidence interval [CI] = 0.24–0.53, p < 0.001). Older age was associated with a higher risk of lung cancer: patients aged 40–59 years (aHR = 5.32, 95% CI = 2.19–12.94, p < 0.001) and those aged ≥60 years (aHR = 16.75, 95% CI = 7.54–37.23, p < 0.001) were at significantly greater risk compared with patients aged 18–39 years. CHM use was associated with a trend for a lower cumulative incidence of lung cancer compared with non-CHM use (p < 0.001). Among the 10 most commonly used single herbs and formulas used to decrease the risk of lung cancer in COPD patients, Fritillariae thunbergii was the most commonly used single herb and Xiao Qing Long Tang the most commonly used formula. Conclusion: The findings from this nationwide retrospective cohort study indicate that CHM as adjunctive therapy in COPD treatment regimens may reduce the risk of lung cancer in this vulnerable patient population.
Collapse
Affiliation(s)
- Tsai-Hui Lin
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Shu-I Chen
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Yuan-Chih Su
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan
| | - Mei-Chen Lin
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan
| | - Hung-Jen Lin
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan.,School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Sheng-Teng Huang
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan.,School of Chinese Medicine, China Medical University, Taichung, Taiwan.,An-Nan Hospital, China Medical University, Tainan, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung, Taiwan.,Research Center for Chinese Herbal Medicine, China Medical University, Taichung, Taiwan.,Cancer Research Center for Traditional Chinese Medicine, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
17
|
Cheng YC, Tsao MJ, Chiu CY, Kan PC, Chen Y. Magnolol Inhibits Human Glioblastoma Cell Migration by Regulating N-Cadherin. J Neuropathol Exp Neurol 2019; 77:426-436. [PMID: 29788114 DOI: 10.1093/jnen/nly021] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma is a primary malignant brain tumor with a poor prognosis. An effective treatment for glioblastoma is needed. Magnolol is a natural compound from Magnolia officinalis suggested to have antiproliferative activity. The aim of this research was to investigate the anticancer effects of magnolol in glioma, with an emphasis on migration and the underlying mechanism. Magnolol decreased the expression of focal adhesion-related proteins and inhibited LN229 and U87MG glioma cell migration. The levels of phosphorylated myosin light chain (p-MLC), phosphorylated myosin light chain kinase and myosin phosphatase target subunit 1 were reduced in response to magnolol treatment. In addition, immunostaining and membrane fractionation showed that the distribution of N-cadherin at the glioma cell membrane was decreased by magnolol. In an orthotropic xenograft animal model, magnolol treatment not only inhibited tumor progression but also reduced p-MLC and N-cadherin protein expression. In conclusion, magnolol reduces cell migration, potentially through regulating focal adhesions and N-cadherin in glioma cells. Magnolol is a potential candidate for glioma treatment.
Collapse
Affiliation(s)
- Yu-Chen Cheng
- Department of Biology and Anatomy, National Defense Medical Center, Taipei City, Taiwan
| | - Min-Jen Tsao
- Department of General Surgery, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Taiwan
| | - Chen-Yang Chiu
- Department of Biology and Anatomy, National Defense Medical Center, Taipei City, Taiwan
| | - Po-Chieh Kan
- Department of Biology and Anatomy, National Defense Medical Center, Taipei City, Taiwan
| | - Ying Chen
- Department of Biology and Anatomy, National Defense Medical Center, Taipei City, Taiwan
| |
Collapse
|
18
|
Zhou S, Wen H, Li H. Magnolol induces apoptosis in osteosarcoma cells via G0/G1 phase arrest and p53-mediated mitochondrial pathway. J Cell Biochem 2019; 120:17067-17079. [PMID: 31155771 DOI: 10.1002/jcb.28968] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 03/11/2019] [Accepted: 03/22/2019] [Indexed: 12/30/2022]
Abstract
Osteosarcoma is a highly invasive primary malignancy of bone. Magnolol is biologically active, which shows antitumor effects in a variety of cancer cell lines. However, it has not been elucidated magnolol's effects on human osteosarcoma cells (HOC). This study aimed to determine antitumor activity of magnolol and illustrate the molecular mechanism in HOC. Magnolol showed significant inhibition effect of growth on MG-63 and 143B cells and induced apoptosis and cell cycle arrest at G0/G1. In osteosarcoma cells, magnolol upregulated expressions of proapoptosis proteins and suppressed expressions of antiapoptosis proteins. Additionally, under the pretreatment of pifithrin-a (PFT-a, a p53 inhibitor), the magnolol-induced apoptosis was significantly reversed. The results above indicated that magnolol induces apoptosis in osteosarcoma cells may via G0/G1 phase arrest and p53-mediated mitochondrial pathway.
Collapse
Affiliation(s)
- Siqi Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Haiyan Wen
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Haohuan Li
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
19
|
Insights on the Multifunctional Activities of Magnolol. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1847130. [PMID: 31240205 PMCID: PMC6556366 DOI: 10.1155/2019/1847130] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/03/2019] [Accepted: 05/15/2019] [Indexed: 12/31/2022]
Abstract
Over years, various biological constituents are isolated from Traditional Chinese Medicine and confirmed to show multifunctional activities. Magnolol, a hydroxylated biphenyl natural compound isolated from Magnolia officinalis, has been extensively documented and shows a range of biological activities. Many signaling pathways include, but are not limited to, NF-κB/MAPK, Nrf2/HO-1, and PI3K/Akt pathways, which are implicated in the biological functions mediated by magnolol. Thus, magnolol is considered as a promising therapeutic agent for clinic research. However, the low water solubility, the low bioavailability, and the rapid metabolism of magnolol dramatically limit its clinical application. In this review, we will comprehensively discuss the last five-year progress of the biological activities of magnolol, including anti-inflammatory, antimicroorganism, antioxidative, anticancer, neuroprotective, cardiovascular protection, metabolism regulation, and ion-mediating activity.
Collapse
|
20
|
Luo H, Wu H, Yu X, Zhang X, Lu Y, Fan J, Tang L, Wang Z. A review of the phytochemistry and pharmacological activities of Magnoliae officinalis cortex. JOURNAL OF ETHNOPHARMACOLOGY 2019; 236:412-442. [PMID: 30818008 DOI: 10.1016/j.jep.2019.02.041] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 02/22/2019] [Accepted: 02/23/2019] [Indexed: 05/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Magnoliae Officinalis Cortex (the dried bark of Magnolia officinalis), a widely used traditional Chinese medicine, is also known as 'Houpo' (Chinese: ). Magnoliae Officinalis Cortex has a wide range of pharmacological effects and has been used to treat conditions such as abdominal distention, vomiting, diarrhea, food accumulation, Qi stagnation, constipation, phlegm and fluid retention and cough resulting from asthma. AIMS OF THE REVIEW The present paper reviews advances in research relating to the botany, ethnopharmacology, phytochemistry, pharmacology and toxicology of Magnoliae Officinalis Cortex. Prospects for future investigation and application of this herb are also discussed. MATERIALS AND METHODS Information on Magnoliae Officinalis Cortex was obtained from published materials, including ancient and modern books; PhD and MSc dissertations; monographs on medicinal plants; the pharmacopoeia of different countries and electronic databases, such as SCI finder, PubMed, Web of Science, ACS, Science Direct, Wiley, Springer, Taylor, AGRIS, Europe PMC, EBSCO host, CNKI, WanFang DATA, J-STAGE and Google Scholar. RESULTS More than 200 chemical compounds have been isolated from Magnoliae Officinalis Cortex, including lignans, phenylethanoid glycosides, phenolic glycosides, alkaloids, steroids and essential oils. The plant has been reported to have pharmacological effects on the digestive system, nervous system and cardiovascular and cerebrovascular systems, as well as antibacterial, anti-tumour, analgesic, anti-inflammatory and anti-oxidative effects. CONCLUSIONS Magnoliae Officinalis Cortex is an essential traditional Chinese medicine with pharmacological activities that mainly affect the digestive system, nervous system and cardiovascular and cerebrovascular systems. This review summarises its botany, ethnopharmacology, phytochemistry, pharmacology and toxicology. These information suggest that we should focus on the development of new drugs related to Magnoliae Officinalis Cortex, including specific constituents, so that Magnoliae Officinalis Cortex can exert greater therapeutic potential. Meanwhile, it is important to pay attention to the rational use of Magnolia resources, avoiding over-harvesting which could lead to lack of resources. We should also pursue research on Magnolia substitutes and develop resources such as Magnoliae Officinalis Flos and Magnolia Leaf.
Collapse
Affiliation(s)
- Hanyan Luo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing 100700, China
| | - Hongwei Wu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing 100700, China
| | - Xiankuo Yu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing 100700, China
| | - Xiao Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing 100700, China
| | - Yaqi Lu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing 100700, China
| | - Jianwei Fan
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Shandong 276006, China
| | - Liying Tang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing 100700, China.
| | - Zhuju Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing 100700, China.
| |
Collapse
|
21
|
Emran AA, Chinna Chowdary BR, Ahmed F, Hammerlindl H, Huefner A, Haass NK, Schuehly W, Schaider H. Magnolol induces cell death through PI3K/Akt-mediated epigenetic modifications boosting treatment of BRAF- and NRAS-mutant melanoma. Cancer Med 2019; 8:1186-1196. [PMID: 30793515 PMCID: PMC6434221 DOI: 10.1002/cam4.1978] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 11/26/2018] [Accepted: 12/23/2018] [Indexed: 01/08/2023] Open
Abstract
Most BRAF‐mutant melanoma patients experience a fulminate relapse after several months of treatment with BRAF/MEK inhibitors. To improve therapeutic efficacy, natural plant‐derived compounds might be considered as potent additives. Here, we show that magnolol, a constituent of Magnolia officinalis, induced G1 arrest, apoptosis and cell death in BRAF‐ and NRAS‐mutant melanoma cells at low concentration, with no effect in BRAF‐ and NRAS wild‐type melanoma cells and human keratinocytes. This was confirmed in a 3D spheroid model. The apoptosis‐inducing effect of magnolol was completely rescued by activating Akt suggesting a mechanism relying primarily on Akt signaling. Magnolol significantly downregulated the PI3K/Akt pathway which led to a global decrease of the active histone mark H3K4me3. Alongside, the repressive histone mark H3K9me3 was increased as a response to DNA damage. Magnolol‐induced alterations of histone modifications are reversible upon activation of the Akt pathway. Magnolol‐induced a synergistic effect in combination with either BRAF/MEK inhibitors dabrafenib/trametinib or docetaxel at a lower concentration than usually applied in melanoma patients. Combination of magnolol with targeted therapy or chemotherapy also led to analogous effects on histone marks, which was rescued by Akt pathway activation. Our study revealed a novel epigenetic mechanism of magnolol‐induced cell death in melanoma. Magnolol might therefore be a clinically useful addition to BRAF/MEK inhibitors with enhanced efficacy delaying or preventing disease recurrence.
Collapse
Affiliation(s)
- Abdullah Al Emran
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia.,Centenary Institute of Cancer Medicine and Cell Biology, Camperdown, New South Wales, Australia
| | - Brinda Reddy Chinna Chowdary
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Farzana Ahmed
- The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Heinz Hammerlindl
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Antje Huefner
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, University of Graz, Graz, Styria, Austria
| | - Nikolas K Haass
- Centenary Institute of Cancer Medicine and Cell Biology, Camperdown, New South Wales, Australia.,The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Wolfgang Schuehly
- Department of Pharmacognosy, Karl-Franzens University, Graz, Styria, Austria
| | - Helmut Schaider
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
22
|
Wang YD, Sun XJ, Yang WJ, Li J, Yin JJ. Magnolol exerts anticancer activity in hepatocellular carcinoma cells through regulating endoplasmic reticulum stress-mediated apoptotic signaling. Onco Targets Ther 2018; 11:5219-5226. [PMID: 30214227 PMCID: PMC6118277 DOI: 10.2147/ott.s168887] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Introduction Magnolol (Mag), a biologically active compound isolated from the root and stem bark of Magnolia officinalis, has been reported to induce apoptosis in several cancer cell lines in vitro. In the present study, we aimed to determine the anticancer effects of Mag on hepatocellular carcinoma (HCC) cells. Materials and methods The HepG2 cells were treated with varying concentrations of Mag (10, 20, and 30 μM) for 48 hours. The effects of Mag on the proliferation, migration, invasion, apoptosis and cell cycle progression of HepG2 cells were respectively detected by MTT assay, transwell assays, and flow cytometric analysis. A HepG2 cell-based tumor-bearing model was established to evaluate the effect of Mag on HCC tumor growth in vivo. The protein expression levels were determined by Western blot analysis. Results Our results showed that Mag inhibited the proliferation, migration, and invasion of HepG2 cells in vitro in a dose-dependent manner. In addition, Mag reduced the HCC tumor volume and weight in the mouse xenograft model. Subsequent studies showed that Mag induced apoptosis in HepG2 cells, accompanied by a loss in mitochondrial membrane potential, cytochrome c release, and induction of endoplasmic reticulum stress. Furthermore, inhibition of the endoplasmic reticulum stress by CHOP knockdown restored the effects of Mag in HepG2 cells. Conclusion The present study highlighted the possibility of using Mag as a novel therapeutic drug for HCC treatment.
Collapse
Affiliation(s)
- Ya-Dong Wang
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, People's Republic of China,
| | - Xue-Jun Sun
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, People's Republic of China
| | - Wei-Jun Yang
- Department of General Surgery, The First People's Hospital of Guiyang, Guiyang 550002, People's Republic of China
| | - Jing Li
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, People's Republic of China,
| | - Jia-Jun Yin
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, People's Republic of China,
| |
Collapse
|
23
|
Chiu YH, Hsu SH, Hsu HW, Huang KC, Liu W, Wu CY, Huang WP, Chen JYF, Chen BH, Chiu CC. Human non‑small cell lung cancer cells can be sensitized to camptothecin by modulating autophagy. Int J Oncol 2018; 53:1967-1979. [PMID: 30106130 PMCID: PMC6192723 DOI: 10.3892/ijo.2018.4523] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 06/01/2018] [Indexed: 12/18/2022] Open
Abstract
Lung cancer is a prevalent disease and is one of the leading causes of mortality worldwide. Despite the development of various anticancer drugs, the prognosis of lung cancer is relatively poor. Metastasis of lung cancer, as well as chemoresistance, is associated with a high mortality rate for patients with lung cancer. Camptothecin (CPT) is a well-known anticancer drug, which causes cancer cell apoptosis via the induction of DNA damage; however, the cytotoxicity of CPT easily reaches a plateau at a relatively high dose in lung cancer cells, thus limiting its efficacy. The present study demonstrated that CPT may induce autophagy in two human non‑small cell lung cancer cell lines, H1299 and H460. In addition, the results of a viability assay and Annexin V staining revealed that CPT-induced autophagy could protect lung cancer cells from programmed cell death. Conversely, the cytotoxicity of CPT was increased when autophagy was blocked by 3-methyladenine treatment. Furthermore, inhibition of autophagy enhanced the levels of CPT-induced DNA damage in the lung cancer cell lines. Accordingly, these findings suggested that autophagy exerts a protective role in CPT-treated lung cancer cells, and the combination of CPT with a specific inhibitor of autophagy may be considered a promising strategy for the future treatment of lung cancer.
Collapse
Affiliation(s)
- Yi-Han Chiu
- Department of Nursing, St. Mary's Junior College of Medicine, Nursing and Management, Yilan 266, Taiwan, R.O.C
| | - Shih-Hsien Hsu
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Hsiao-Wei Hsu
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Kuo-Chin Huang
- Holistic Education Center, Mackay Medical College, New Taipei City 252, Taiwan, R.O.C
| | - Wangta Liu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Chang-Yi Wu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Wei-Pang Huang
- Department of Life Science, National Taiwan University, Taipei 106, Taiwan, R.O.C
| | - Jeff Yi-Fu Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Bing-Hung Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Chien-Chih Chiu
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| |
Collapse
|
24
|
Magnolol: A Neolignan from the Magnolia Family for the Prevention and Treatment of Cancer. Int J Mol Sci 2018; 19:ijms19082362. [PMID: 30103472 PMCID: PMC6121321 DOI: 10.3390/ijms19082362] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 08/06/2018] [Accepted: 08/08/2018] [Indexed: 12/15/2022] Open
Abstract
The past few decades have witnessed widespread research to challenge carcinogenesis; however, it remains one of the most important health concerns with the worst prognosis and diagnosis. Increasing lines of evidence clearly show that the rate of cancer incidence will increase in future and will create global havoc, designating it as an epidemic. Conventional chemotherapeutics and treatment with synthetic disciplines are often associated with adverse side effects and development of chemoresistance. Thus, discovering novel economic and patient friendly drugs that are safe and efficacious is warranted. Several natural compounds have proved their potential against this dreadful disease so far. Magnolol is a hydroxylated biphenyl isolated from the root and stem bark of Magnolia tree. Magnolol can efficiently prevent or inhibit the growth of various cancers originating from different organs such as brain, breast, cervical, colon, liver, lung, prostate, skin, etc. Considering these perspectives, the current review primarily focuses on the fascinating role of magnolol against various types of cancers, and the source and chemistry of magnolol and the molecular mechanism underlying the targets of magnolol are discussed. This review proposes magnolol as a suitable candidate that can be appropriately designed and established into a potent anti-cancer drug.
Collapse
|
25
|
Ji L, Zhong B, Jiang X, Mao F, Liu G, Song B, Wang CY, Jiao Y, Wang JP, Xu ZB, Li X, Zhan B. Actein induces autophagy and apoptosis in human bladder cancer by potentiating ROS/JNK and inhibiting AKT pathways. Oncotarget 2017; 8:112498-112515. [PMID: 29348843 PMCID: PMC5762528 DOI: 10.18632/oncotarget.22274] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 09/20/2017] [Indexed: 02/06/2023] Open
Abstract
Human bladder cancer is a common genitourinary malignant cancer worldwide. However, new therapeutic strategies are required to overcome its stagnated survival rate. Triterpene glycoside Actein (ACT), extracted from the herb black cohosh, suppresses the growth of human breast cancer cells. Our study attempted to explore the role of ACT in human bladder cancer cell growth and to reveal the underlying molecular mechanisms. We found that ACT significantly impeded the bladder cancer cell proliferation via induction of G2/M cycle arrest. Additionally, ACT administration triggered autophagy and apoptosis in bladder cancer cells, proved by the autophagosome formation, LC3B-II accumulation, improved cleavage of Caspases/poly (ADP-ribose) polymerase (PARP). Furthermore, reduction of reactive oxygen species (ROS) and p-c-Jun N-terminal kinase (JNK) could markedly reverse ACT-induced autophagy and apoptosis. In contrast, AKT and mammalian target of rapamycin (mTOR) were greatly de-phosphorylated by ACT, while suppressing AKT and mTOR activity could enhance the effects of ACT on apoptosis and autophagy induction. In vivo, ACT reduced the tumor growth with little toxicity. Taken together, our findings indicated that ACT suppressed cell proliferation, induced autophagy and apoptosis through promoting ROS/JNK activation, and blunting AKT pathway in human bladder cancer, which indicated that ACT might be an effective candidate against human bladder cancer in future.
Collapse
Affiliation(s)
- Lu Ji
- Department of Urology, Huai’an First People's Hospital, Nanjing Medical University, Huai’an 223300, China
| | - Bing Zhong
- Department of Urology, Huai’an First People's Hospital, Nanjing Medical University, Huai’an 223300, China
| | - Xi Jiang
- Department of Urology, Huai’an First People's Hospital, Nanjing Medical University, Huai’an 223300, China
| | - Fei Mao
- Department of Urology, Huai’an First People's Hospital, Nanjing Medical University, Huai’an 223300, China
| | - Gang Liu
- Department of Orthopaedics, Huai’an First People's Hospital, Nanjing Medical University, Huai’an 223300, China
| | - Bin Song
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Cheng-Yuan Wang
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Yong Jiao
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Jiang-Ping Wang
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Zhi-Bin Xu
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Xing Li
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Bo Zhan
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| |
Collapse
|
26
|
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: 2.4] [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
|
27
|
Wang TH, Chan CW, Fang JY, Shih YM, Liu YW, Wang TCV, Chen CY. 2-O-Methylmagnolol upregulates the long non-coding RNA, GAS5, and enhances apoptosis in skin cancer cells. Cell Death Dis 2017; 8:e2638. [PMID: 28252643 PMCID: PMC5386561 DOI: 10.1038/cddis.2017.66] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/21/2017] [Accepted: 01/24/2017] [Indexed: 12/11/2022]
Abstract
Magnolol, a hydroxylated biphenol compound isolated from the bark of Magnolia officinalis, has been shown to exhibit anti-proliferative effect in various cancer cells, including skin cancer cells. Methoxylation of magnolol appears to improve its anti-inflammatory activity, yet the effect of this modification on the agent's antitumor activity remains unknown. In this work, we report that 2-O-methylmagnolol (MM1) displays improved antitumor activity against skin cancer cells compared to magnolol both in vitro and in vivo. The increased antitumor activity of MM1 appears to correlate with its increased ability to induce apoptosis. DNA microarray and network pathway analyses suggest that MM1 affects certain key factors involved in regulating apoptosis and programmed cell death. Interestingly, the level of the long non-coding (lnc) RNA of growth arrest-specific 5 (GAS5) was increased in MM1-treated cells, and inhibition of lncRNA GAS5 inhibited MM1-induced apoptosis. Conversely, overexpression of lncRNA GAS5 inhibited cell proliferation and promoted cell apoptosis in skin cancer cells. The expression of lncRNA GAS5 in the skin cancer tissues was found to be lower than that in the adjacent normal tissues in a majority of patients. Taken together, our findings suggest that MM1 has improved antitumor activity in skin cancer cells, and that this is due, at least in part, to the upregulation of lncRNA GAS5 and the enhancement of apoptosis.
Collapse
Affiliation(s)
- Tong-Hong Wang
- Graduate Institute of Health Industry Technology and Research Center for Industry of Human Ecology, College of Human Ecology, Chang Gung University of Science and Technology, Kwei-Shan, Tao-Yuan 333, Taiwan.,Tissue Bank, Chang Gung Memorial Hospital, Kwei-Shan, Tao-Yuan 333, Taiwan
| | - Chieh-Wen Chan
- Graduate Institute of Health Industry Technology and Research Center for Industry of Human Ecology, College of Human Ecology, Chang Gung University of Science and Technology, Kwei-Shan, Tao-Yuan 333, Taiwan
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kwei-Shan, Tao-Yuan 333, Taiwan.,Department of Anesthesiology, Chang Gung Memorial Hospital, Kwei-Shan, Tao-Yuan 333, Taiwan
| | - Ya-Min Shih
- Graduate Institute of Health Industry Technology and Research Center for Industry of Human Ecology, College of Human Ecology, Chang Gung University of Science and Technology, Kwei-Shan, Tao-Yuan 333, Taiwan
| | - Yi-Wen Liu
- Graduate Institute of Health Industry Technology and Research Center for Industry of Human Ecology, College of Human Ecology, Chang Gung University of Science and Technology, Kwei-Shan, Tao-Yuan 333, Taiwan
| | - Tzu-Chien V Wang
- Tissue Bank, Chang Gung Memorial Hospital, Kwei-Shan, Tao-Yuan 333, Taiwan.,Department of Molecular and Cellular Biology, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 333, Taiwan
| | - Chi-Yuan Chen
- Graduate Institute of Health Industry Technology and Research Center for Industry of Human Ecology, College of Human Ecology, Chang Gung University of Science and Technology, Kwei-Shan, Tao-Yuan 333, Taiwan.,Tissue Bank, Chang Gung Memorial Hospital, Kwei-Shan, Tao-Yuan 333, Taiwan
| |
Collapse
|
28
|
Novel histone deacetylase inhibitors derived from Magnolia officinalis significantly enhance TRAIL-induced apoptosis in non-small cell lung cancer. Pharmacol Res 2016; 111:113-125. [DOI: 10.1016/j.phrs.2016.05.028] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 05/24/2016] [Accepted: 05/25/2016] [Indexed: 12/30/2022]
|
29
|
Cheng S, Castillo V, Welty M, Eliaz I, Sliva D. Honokiol inhibits migration of renal cell carcinoma through activation of RhoA/ROCK/MLC signaling pathway. Int J Oncol 2016; 49:1525-1530. [DOI: 10.3892/ijo.2016.3663] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/02/2016] [Indexed: 11/06/2022] Open
|
30
|
Kuo HF, Liu PL, Chong IW, Liu YP, Chen YH, Ku PM, Li CY, Chen HH, Chiang HC, Wang CL, Chen HJ, Chen YC, Hsieh CC. Pigment Epithelium-Derived Factor Mediates Autophagy and Apoptosis in Myocardial Hypoxia/Reoxygenation Injury. PLoS One 2016; 11:e0156059. [PMID: 27219009 PMCID: PMC4878768 DOI: 10.1371/journal.pone.0156059] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 05/09/2016] [Indexed: 12/30/2022] Open
Abstract
Pigment epithelium-derived factor (PEDF) is a multifunctional protein that exhibits anti-angiogenic, antitumor, anti-inflammatory, antioxidative, anti-atherogenic, and cardioprotective properties. While it was recently shown that PEDF expression is inhibited under low oxygen conditions, the functional role of PEDF in response to hypoxia/reoxygenation (H/R) remains unclear. The goal of this study was to therefore investigate the influence of PEDF on myocardial H/R injury. For these analyses, PEDF-specific small interfering RNA-expressing and PEDF-expressing lentivirus (PEDF-LV) vectors were utilized to knockdown or stably overexpress PEDF, respectively, within human cardiomyocytes (HCM) in vitro. We noted that reactive oxygen species (ROS) play important roles in the induction of cell death pathways, including apoptosis and autophagy in ischemic hearts. Our findings demonstrate that overexpression of PEDF resulted in a significant reduction in ROS production and attenuation of mitochondrial membrane potential depletion under H/R conditions. Furthermore, PEDF inhibited the activation of a two-step apoptotic pathway in which caspase-dependent (caspase-9 and caspase-3) and caspase-independent (apoptosis inducing factor and endonuclease G), which in turn cleaves several crucial substrates including the DNA repair enzyme poly (ADP-ribose) polymerase. Meanwhile, overexpression of PEDF also promoted autophagy, a process that is typically activated in response to H/R. Therefore, these findings suggest that PEDF plays a critical role in preventing H/R injury by modulating anti-oxidant and anti-apoptotic factors and promoting autophagy.
Collapse
Affiliation(s)
- Hsuan-Fu Kuo
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 801, Taiwan
| | - Po-Len Liu
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Inn-Wen Chong
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Yu-Peng Liu
- Department of Genome Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yung-Hsiang Chen
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, 404, Taiwan
- Department of Psychology, College of Medical and Health Science, Asia University, Taichung, 413, Taiwan
| | - Po-Ming Ku
- Cardiovascular Center, Chi-Mei Hospital, Liouying, Tainan, 736, Taiwan
- Chia-Nan University of Pharmacy & Science, Tainan, 717, Taiwan
| | - Chia-Yang Li
- Department of Genome Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsiu-Hua Chen
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 801, Taiwan
| | - Hui-Ching Chiang
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 801, Taiwan
| | - Chiao-Lin Wang
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 801, Taiwan
| | - Huang-Jen Chen
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 801, Taiwan
| | - Yen-Chieh Chen
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 801, Taiwan
| | - Chong-Chao Hsieh
- Division of Cardiovascular Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
- * E-mail:
| |
Collapse
|
31
|
Li M, Zhang F, Wang X, Wu X, Zhang B, Zhang N, Wu W, Wang Z, Weng H, Liu S, Gao G, Mu J, Shu Y, Bao R, Cao Y, Lu J, Gu J, Zhu J, Liu Y. Magnolol inhibits growth of gallbladder cancer cells through the p53 pathway. Cancer Sci 2015; 106:1341-50. [PMID: 26250568 PMCID: PMC4638010 DOI: 10.1111/cas.12762] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 07/29/2015] [Accepted: 08/02/2015] [Indexed: 12/24/2022] Open
Abstract
Magnolol, the major active compound found in Magnolia officinalis has a wide range of clinical applications due to its anti-inflammation and anti-oxidation effects. This study investigated the effects of magnolol on the growth of human gallbladder carcinoma (GBC) cell lines. The results indicated that magnolol could significantly inhibit the growth of GBC cell lines in a dose- and time-dependent manner. Magnolol also blocked cell cycle progression at G0 /G1 phase and induced mitochondrial-related apoptosis by upregulating p53 and p21 protein levels and by downregulating cyclin D1, CDC25A, and Cdk2 protein levels. When cells were pretreated with a p53 inhibitor (pifithrin-a), followed by magnolol treatment, pifithrin-a blocked magnolol-induced apoptosis and G0 /G1 arrest. In vivo, magnolol suppressed tumor growth and activated the same mechanisms as were activated in vitro. In conclusion, our study is the first to report that magnolol has an inhibitory effect on the growth of GBC cells and that this compound may have potential as a novel therapeutic agent for the treatment of GBC.
Collapse
Affiliation(s)
- Maolan Li
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Laboratory of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Fei Zhang
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Laboratory of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Xu’an Wang
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Laboratory of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Xiangsong Wu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Laboratory of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Bingtai Zhang
- Department of General Surgery, Shanxi Medical University Second HospitalTaiyuan, China
| | - Ning Zhang
- Department of General Surgery, Shanxi Medical University Second HospitalTaiyuan, China
| | - Wenguang Wu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Laboratory of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Zheng Wang
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Laboratory of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Hao Weng
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Laboratory of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Shibo Liu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Laboratory of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Guofeng Gao
- Department of General Surgery, Shanxi Medical University Second HospitalTaiyuan, China
| | - Jiasheng Mu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Laboratory of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Yijun Shu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Laboratory of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Runfa Bao
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Laboratory of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Yang Cao
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Laboratory of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Jianhua Lu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Laboratory of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Jun Gu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Laboratory of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Jian Zhu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Laboratory of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Yingbin Liu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Laboratory of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong, University School of MedicineShanghai, China
- Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of MedicineShanghai, China
| |
Collapse
|
32
|
Japonicone A inhibits the growth of non-small cell lung cancer cells via mitochondria-mediated pathways. Tumour Biol 2015; 36:7473-82. [PMID: 25908173 DOI: 10.1007/s13277-015-3439-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 04/07/2015] [Indexed: 12/24/2022] Open
Abstract
Japonicone A, which is a natural product isolated from the aerial part of Inula japonica Thunb., has a wide range of clinical applications, including anti-inflammation and anti-oxidation. This study investigated the effects of japonicone A on the growth of non-small cell lung cancer (NSCLC) cell lines. The results showed that japonicone A significantly inhibited the growth of NSCLC cell lines in a dose- and time-dependent manner. This product also blocked cell cycle progression at S phase and induced mitochondrial-related apoptosis by upregulating Bax, cleaved caspase-9, cleaved caspase-3, and cleaved poly(ADP-ribose) polymerase (PARP) protein levels and by downregulating Bcl-2, cyclin D1, CDC25A, and CDK2 protein levels. In vivo, japonicone A suppressed tumor growth via the same mechanism as that observed in vitro. In conclusion, our study is the first to report that japonicone A has an inhibitory effect on the growth of NSCLC cells, indicating that japonicone A administration is a potential therapeutic approach for future NSCLC treatments.
Collapse
|
33
|
Lou C, Takahashi K, Irimura T, Saiki I, Hayakawa Y. Identification of Hirsutine as an anti-metastatic phytochemical by targeting NF-κB activation. Int J Oncol 2014; 45:2085-91. [PMID: 25175557 DOI: 10.3892/ijo.2014.2624] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 07/22/2014] [Indexed: 11/06/2022] Open
Abstract
Nuclear factor-κB (NF-κB) activation has been implicated not only in carcinogenesis but also in cancer cell invasion and metastatic process; therefore, targeting the NF-κB pathway is an attractive strategy for controlling meta-stasis. Amongst 56 chemically defined compounds derived from natural products, we have identified a new phytochemical compound Hirsutine, which strongly suppresses NF-κB activity in murine 4T1 breast cancer cells. In accordance with the NF-κB inhibition, Hirsutine reduced the metastatic potential of 4T1 cells, as seen in the inhibition of the migration and invasion capacity of 4T1 cells. Hirsutine further inhibited the constitutive expression of MMP-2 and MMP-9 in 4T1 cells, and reduced the in vivo lung metastatic potential of 4T1 cells in the experimental model. Given that the migration of human breast cancer cells was also inhibited, our present study implies that Hirsutine is an attractive phytochemical compound for reducing metastasis potential of cancer cells by regulating tumor-promoting NF-κB activity.
Collapse
Affiliation(s)
- Chenghua Lou
- Division of Pathogenic Biochemistry, Department of Bioscience, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Kei Takahashi
- Laboratory of Cancer Biology and Molecular Immunology, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo 113-0033, Japan
| | - Tatsuro Irimura
- Laboratory of Cancer Biology and Molecular Immunology, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo 113-0033, Japan
| | - Ikuo Saiki
- Division of Pathogenic Biochemistry, Department of Bioscience, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Yoshihiro Hayakawa
- Division of Pathogenic Biochemistry, Department of Bioscience, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan
| |
Collapse
|