1
|
Liu Y, Zhang S, Tan Y. Honokiol induces apoptosis and autophagy in dexamethasone-resistant T-acute lymphoblastic leukemia CEM-C1 cells. Hematology 2024; 29:2337307. [PMID: 38573223 DOI: 10.1080/16078454.2024.2337307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 03/26/2024] [Indexed: 04/05/2024] Open
Abstract
Objective: To study whether and, if so, how honokiol overcome dexamethasone resistance in DEX-resistant CEM-C1 cells. Methods: We investigated the effect of honokiol (0-20 µM) on cell proliferation, cell cycle, cell apoptosis and autophagy in DEX-resistant CEM-C1 cells and DEX-sensitive CEM-C7 cells. We also determined the role of c-Myc protein and mRNA in the occurrence of T-ALL associated dexamethasone resistance western blot and reverse transcription-qPCR (RT-qPCR) analysis. Results: Cell Counting Kit (CCK)-8 assay shows that DEX-resistant CEM-C1 cell lines were highly resistant to dexamethasone with IC50 of 364.1 ± 29.5 µM for 48 h treatment. However, upon treatment with dexamethasone in combination with 1.5 µM of honokiol for 48 h, the IC50 of CEM-C1 cells significantly decreased to 126.2 ± 12.3 µM, and the reversal fold was 2.88. Conversely, the IC50 of CEM-C7 cells was not changed combination of dexamethasone and honokiol as compared to that of CEM-C7 cells treated with dexamethasone alone. It has been shown that honokiol induced T-ALL cell growth inhibition by apoptosis and autophagy via downregulating cell cycle-regulated proteins (Cyclin E, CDK4, and Cyclin D1) and anti-apoptotic proteins BCL-2 and upregulating pro-apoptotic proteins Bax and led to PARP cleavage. Honokiol may overcome dexamethasone resistance in DEX-resistant CEM-C1 cell lines via the suppression of c-Myc mRNA expression. Conclusion: The combination of honokiol and DEX were better than DEX alone in DEX-resistant CEM-C1 cell lines. Honokiol may regulate T-ALL-related dexamethasone resistance by affecting c-Myc.
Collapse
Affiliation(s)
- Yang Liu
- Pediatric Department, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Hubei, People's Republic of China
| | - Suqian Zhang
- Pediatric Department, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Hubei, People's Republic of China
| | - Yajuan Tan
- Pediatric Department, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Hubei, People's Republic of China
| |
Collapse
|
2
|
Kleszcz R, Dorna D, Stawny M, Paluszczak J. Honokiol Is More Potent than Magnolol in Reducing Head and Neck Cancer Cell Growth. Curr Issues Mol Biol 2024; 46:10731-10744. [PMID: 39451517 PMCID: PMC11506259 DOI: 10.3390/cimb46100637] [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: 07/29/2024] [Revised: 08/23/2024] [Accepted: 09/16/2024] [Indexed: 10/26/2024] Open
Abstract
The efficacy of treatment of head and neck squamous cell carcinoma (HNSCC) patients is still unsatisfactory, and there is an ongoing search for novel therapies. Locoregionally advanced HNSCC cases, which frequently require combined surgery and chemoradiotherapy, are especially difficult to treat. Natural compounds, like Magnolia-derived lignans-honokiol (HON) and magnolol (MAG)-can reduce cancer cell growth but retain a good safety profile and thus may show benefit as adjuvant therapeutics. The aim of this study was to evaluate the anti-cancer effects of HON and MAG in HNSCC cell lines and compare their effects between cisplatin-sensitive and cisplatin-tolerant cells. Cell viability was evaluated in FaDu and SCC-040 cells growing as monolayers and as spheroids. The effect of HON and MAG on the cell cycle, apoptosis, and gene expression was compared between wild-type FaDu cells and cisplatin persister FaDu cells. We observed that HON and MAG were more potent in reducing cell viability in cisplatin persister FaDu cells, although this effect was not directly followed by increased rates of apoptosis. Thus, HON's and MAG's capacity to affect cisplatin persister cells needs further studies. In general, we observed that HON exerted stronger cytotoxic effects than MAG in HNSCC cells, and the difference in their anti-cancer activity was especially pronounced in cells cultured in 3D.
Collapse
Affiliation(s)
- Robert Kleszcz
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, 60-806 Poznań, Poland;
| | - Dawid Dorna
- Department of Pharmaceutical Biochemistry, Doctoral School, Poznan University of Medical Sciences, 60-806 Poznań, Poland;
| | - Maciej Stawny
- Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, 60-806 Poznań, Poland;
| | - Jarosław Paluszczak
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, 60-806 Poznań, Poland;
| |
Collapse
|
3
|
Liu F, Zhang Y, Xia X, Han J, Cao L. Honokiol regulates ovarian cancer cell malignant behavior through YAP/TAZ pathway modulation. J Obstet Gynaecol Res 2024; 50:1010-1019. [PMID: 38576101 DOI: 10.1111/jog.15940] [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: 10/27/2023] [Accepted: 03/26/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Ovarian cancer (OVCA) stands as one of the most fatal gynecological malignancies. Honokiol (HNK) has been substantiated by numerous studies for its anti-tumor activity against malignancies including OVCA. Consequently, this work was designed to elucidate the impact of HNK-mediated modulation of the YAP/TAZ pathway on the biological functions of OVCA cells. METHODS OVCA cells were subjected to treatment with varying concentrations (0, 25, 50, 75, and 100 μM) of HNK, concomitant with the administration of YAP agonist (XMU). Assessment of cellular viability was executed employing the CCK-8 assay, while quantification of cellular proliferation transpired via colony formation assays. Apoptosis was ascertained using flow cytometry, and expression of apoptosis-related proteins (caspase-3, Bcl-2, Bax), EMT-related proteins (E-cadherin, N-cadherin), migration-associated proteins (MMP-2, MMP-9), and YAP/TAZ pathway-related proteins was evaluated by western blot. Transwell experiments were conducted to assess cellular migratory and invasive propensities. Xenograft tumor models were built to observe tumor growth (volume and weight), apoptosis was assessed by TUNEL staining, and Ki67 expression was evaluated through IHC. RESULTS HNK exerted inhibitory effects on the viability and proliferative capacity of OVCA cells, elicited apoptotic responses, curtailed the migratory and invasive tendencies of cells, and downregulated the YAP/TAZ pathway. Stimulation with YAP agonist (XMU-MP-1) partially attenuated the impacts of HNK on OVCA cell biology. Experiments in vivo confirmed that HNK inhibited OVCA tumor growth. CONCLUSION The outcomes of this investigation conclusively established that HNK orchestrated the modulation of the YAP/TAZ pathway, thereby exerting control over the malignant phenotypic manifestations of OVCA cells. The ascertained function of HNK in restraining cellular proliferation and tumor progression provided novel evidence of its anti-proliferative activity within OVCA cells.
Collapse
Affiliation(s)
- Fang Liu
- Department of Gynecology, The Second Affiliated Hospital of Jiaxing University, Jiaxing City, Zhejiang Province, China
| | - Yufang Zhang
- Department of Gynecology, The Second Affiliated Hospital of Jiaxing University, Jiaxing City, Zhejiang Province, China
| | - Xinyi Xia
- Department of Gynecology, The Second Affiliated Hospital of Jiaxing University, Jiaxing City, Zhejiang Province, China
| | - Jing Han
- Department of Gynecology, The Second Affiliated Hospital of Jiaxing University, Jiaxing City, Zhejiang Province, China
| | - Linyan Cao
- Department of Gynecology, The Second Affiliated Hospital of Jiaxing University, Jiaxing City, Zhejiang Province, China
| |
Collapse
|
4
|
Safe S. Specificity Proteins (Sp) and Cancer. Int J Mol Sci 2023; 24:5164. [PMID: 36982239 PMCID: PMC10048989 DOI: 10.3390/ijms24065164] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 03/10/2023] Open
Abstract
The specificity protein (Sp) transcription factors (TFs) Sp1, Sp2, Sp3 and Sp4 exhibit structural and functional similarities in cancer cells and extensive studies of Sp1 show that it is a negative prognostic factor for patients with multiple tumor types. In this review, the role of Sp1, Sp3 and Sp4 in the development of cancer and their regulation of pro-oncogenic factors and pathways is reviewed. In addition, interactions with non-coding RNAs and the development of agents that target Sp transcription factors are also discussed. Studies on normal cell transformation into cancer cell lines show that this transformation process is accompanied by increased levels of Sp1 in most cell models, and in the transformation of muscle cells into rhabdomyosarcoma, both Sp1 and Sp3, but not Sp4, are increased. The pro-oncogenic functions of Sp1, Sp3 and Sp4 in cancer cell lines were studied in knockdown studies where silencing of each individual Sp TF decreased cancer growth, invasion and induced apoptosis. Silencing of an individual Sp TF was not compensated for by the other two and it was concluded that Sp1, Sp3 and Sp4 are examples of non-oncogene addicted genes. This conclusion was strengthened by the results of Sp TF interactions with non-coding microRNAs and long non-coding RNAs where Sp1 contributed to pro-oncogenic functions of Sp/non-coding RNAs. There are now many examples of anticancer agents and pharmaceuticals that induce downregulation/degradation of Sp1, Sp3 and Sp4, yet clinical applications of drugs specifically targeting Sp TFs are not being used. The application of agents targeting Sp TFs in combination therapies should be considered for their potential to enhance treatment efficacy and decrease toxic side effects.
Collapse
Affiliation(s)
- Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
5
|
Natural carbazole alkaloid murrayafoline A displays potent anti-neuroinflammatory effect by directly targeting transcription factor Sp1 in LPS-induced microglial cells. Bioorg Chem 2022; 129:106178. [DOI: 10.1016/j.bioorg.2022.106178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 09/19/2022] [Accepted: 09/23/2022] [Indexed: 11/21/2022]
|
6
|
Myeloid cell leukemia-1 expression in cancers of the oral cavity: a scoping review. Cancer Cell Int 2022; 22:182. [PMID: 35524332 PMCID: PMC9074253 DOI: 10.1186/s12935-022-02603-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 04/20/2022] [Indexed: 12/04/2022] Open
Abstract
Background B cell lymphoma-2 (Bcl-2) family members play important roles in cell survival as well as cell death. The role of myeloid cell leukemia-1 (Mcl-1), an important member of the Bcl-2 family, is well established in hematopoietic malignancies. However, the association between Mcl-1 and oral cavity, cancers is not clearly defined. Methods A scoping review was conducted until June 30, 2021, using four major databases, PubMed, Scopus, Web of Science, and Embase. Medical subject headings keywords for Mcl-1, along with its other identifiers, and head and neck cancers (only oral cavity tumors) were used to evaluate the expression, function, molecular association, and therapeutic approach of Mcl-1 in oral cavity cancers and precancers. Findings Mcl-1 expression was associated with the progression of oral cavity cancers. The molecular mechanism and pathways of Mcl-1 in oral cavity cancers established via experimental results have been highlighted in this review. Moreover, the various synthetic and naturally derived therapeutic agents targeting Mcl-1 have been documented. Novelty/Improvement Based on our present review, Mcl-1 appears to be an effective anticancer target that can be used in the therapeutic management of oral cancers.
Collapse
|
7
|
Ashry R, Elhussiny M, Abdellatif H, Elkashty O, Abdel-Ghaffar HA, Gaballa ET, Mousa SA. Genetic Interpretation of the Impacts of Honokiol and EGCG on Apoptotic and Self-Renewal Pathways in HEp-2 Human Laryngeal CD44 high Cancer Stem Cells. Nutr Cancer 2021; 74:2152-2173. [PMID: 34590505 DOI: 10.1080/01635581.2021.1981404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Most current larynx cancer therapies are generally aimed at the global mass of tumor, targeting the non-tumorigenic cells, and unfortunately sparing the tumorigenic cancer stem cells (CSCs) that are responsible for sustained growth, metastasis, and chemo- and radioresistance. Phytochemicals and herbs have recently been introduced as therapeutic sources for eliminating CSCs. Therefore, we assessed the anti-tumor effects of two herbal ingredients, the green tea extract "Epigallocatechin-3-gallate (EGCG)" and Honokiol (HNK), on parental cells or CD44high CSCs of the human laryngeal squamous cell carcinoma cell line HEp-2. Results revealed that EGCG had a preeminent apoptotic potential on HEp-2 laryngeal CSCs. HNK conferred higher cytotoxic impacts on parental cells mostly by necrosis induction, especially with higher doses, but apoptosis induction with lower doses was also observed. The Notch signaling pathway genes were more potently suppressed by EGCG than HNK. However, HNK surpassed EGCG in downregulating the β-catenin and the Sonic Hedgehog signaling pathways genes. On a genetic basis, both agents engaged the BCL-2 family-regulated and caspase-dependent intrinsic apoptotic pathway, but EGCG and HNK triggered apoptosis via p53-independent and p53-dependent pathways, respectively. Taken together, EGCG and HNK eradicated HEp-2 human larynx cancer cells through targeting multiple self-renewal pathways and activating diverse cell death modalities.
Collapse
Affiliation(s)
- Ramy Ashry
- Institute of Toxicology, University Medical Center Mainz, Mainz, Germany.,Oral Pathology Department, Faculty of Dentistry, Mansoura University, Mansoura, Egypt
| | - Mahmoud Elhussiny
- Institute of Global Health and Human Ecology, American University in Cairo (AUC), Cairo, Egypt.,Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Hussein Abdellatif
- Department of Human and Clinical Anatomy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Sultanate of Oman.,Anatomy and Embryology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Osama Elkashty
- Oral Pathology Department, Faculty of Dentistry, Mansoura University, Mansoura, Egypt.,Craniofacial Tissue and Stem Cell Laboratory, Faculty of Dentistry, McGill University, Montreal, Canada
| | - Hassan A Abdel-Ghaffar
- Hematology Laboratory, Oncology Center, Mansoura University, Mansoura, Egypt.,Hematology section, Clinical Pathology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Essam T Gaballa
- Oral Pathology Department, Faculty of Dentistry, Mansoura University, Mansoura, Egypt.,Oral Pathology Department, Faculty of Dentistry, Delta University, Mansoura, Egypt
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, New York, USA
| |
Collapse
|
8
|
Chen DD, Wang HW, Cai XJ. Transcription factor Sp1 ameliorates sepsis-induced myocardial injury via ZFAS1/Notch signaling in H9C2 cells. Cytokine 2021; 140:155426. [PMID: 33517197 DOI: 10.1016/j.cyto.2021.155426] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/24/2020] [Accepted: 01/06/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE To investigate whether Sp1 can ameliorate sepsis-induced myocardial injury and explore the potential molecular mechanism. METHODS The embryonic cardiomyocyte cell line H9C2 and primary cultured mouse neonatal cardiomyocytes (CMNCs) were treated with LPS or phosphate-buffered saline (PBS). A mouse model of LPS-induced sepsis was established using male C57BL/6J mice and their cardiomyocytes were collected. Real-time reverse transcription-PCR (qRT-PCR) assay was used to detect the expression levels of Sp1 and ZFAS1 in cardiomyocytes. Western blotting analysis was used to assess the protein expression levels of Sp1, apoptosis-associated proteins and Notch signaling pathway related proteins. Luciferase assay was used to detect the interaction between Sp1 and ZFAS1. Cell transfection was used to generate H9C2 cells with overexpressed or knocked down of Sp1 or ZFAS1. MTT assay and flow cytometry analysis were used to test the cell proliferation and cell apoptosis ratio. RESULTS Our data revealed that the expressions of ZFAS1 and Sp1 were significantly reduced in LPS-treated H9C2 cells and primary CMNCs. The downregulation of ZFAS1 and Sp1 were also found in cardiomyocytes obtained from LPS-challenged mice. LPS induced H9C2 cell apoptosis and depressed cell proliferation was ameliorated by ZFAS1 overexpression and aggravated by ZFAS1 knockdown. Mechanistically, Luciferase assay indicated that Sp1 could bind to ZFAS1, and positively regulated ZFAS1 expression. Moreover, Notch signaling pathway participates in H9C2 cell apoptosis mediated by Sp1. CONCLUSION The present study demonstrates that Sp1 regulates LPS-induced cardiomyocyte apoptosis via ZFAS1/Notch signaling pathway, which may serve as therapeutic targets for sepsis-induced myocardial injury.
Collapse
Affiliation(s)
- Dan-Dan Chen
- Department of Critical Care Medicine, Haikou Hospital, Xiangya Medical College, Central South University, Haikou 570208, PR China
| | - Hong-Wu Wang
- Department of Critical Care Medicine, Haikou Hospital, Xiangya Medical College, Central South University, Haikou 570208, PR China
| | - Xing-Jun Cai
- Department of Respiratory and Critical Care Medicine, Hainan General Hospital, Haikou 570311, PR China.
| |
Collapse
|
9
|
Liu T, Long T, Li H. Curcumin suppresses the proliferation of oral squamous cell carcinoma through a specificity protein 1/nuclear factor-κB-dependent pathway. Exp Ther Med 2021; 21:202. [PMID: 33500696 DOI: 10.3892/etm.2021.9635] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 07/17/2020] [Indexed: 12/17/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most common cancer of the oral cavity. Curcumin (Cur), a naturally derived compound, is reported to have broad-spectrum anticancer activity and is considered as an effective nuclear factor-κB (NF-κB) inhibitor. The present study aimed to clarify the detailed molecular mechanism though which Cur regulates NF-κB pathway activity in OSCC. The viability of HSC3 and CAL33 cells following treatment with Cur was determined using a Cell Counting Kit-8 assay. The protein and mRNA expression of specificity protein 1 (Sp1), p65 and heat shock factor 1 (HSF1) was determined by western blotting and reverse transcription-quantitative PCR analysis, respectively. The NF-κB activity was measured by Dual-Luciferase reporter assay. Short hairpin RNA targeting Sp1 or control RNA was transfected into HSC3 cells using X-treme GENE HP DNA Transfection System. Colony formation assays were performed using crystal violet staining. The results demonstrated that Cur significantly inhibited the viability and colony formation ability of HSC3 and CAL33 cells. In addition, Cur decreased the expression of Sp1, p65 and HSF1 by suppressing their transcription levels. Cur decreased NF-κB activity in OSCC cells, and Sp1 downregulation enhanced the effect of Cur. The findings from the present study suggested that Cur may inhibit the proliferation of OSCC cells via a Sp1/NF-κB-dependent mechanism.
Collapse
Affiliation(s)
- Tian Liu
- Department of Stomatology, The Central Hospital of Wuhan, Wuhan, Hubei 430000, P.R. China
| | - Tian Long
- Department of Stomatology, The Central Hospital of Wuhan, Wuhan, Hubei 430000, P.R. China
| | - Haosen Li
- Department of Stomatology Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| |
Collapse
|
10
|
Bui D, Li L, Yin T, Wang X, Gao S, You M, Singh R, Hu M. Pharmacokinetic and Metabolic Profiling of Key Active Components of Dietary Supplement Magnolia officinalis Extract for Prevention against Oral Carcinoma. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:6576-6587. [PMID: 32348135 PMCID: PMC7604171 DOI: 10.1021/acs.jafc.0c01475] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Among the three key active components (KACs) of Magnolia officinalis bark extract (ME), 4-O-methylhonokiol and honokiol showed higher antiproliferation activities than magnolol in the oral squamous cancer cell lines (Cal-27, SCC-9, and SCC-4). Oral bioavailabilities of ME-KACs were poor (<0.2%) in C57BL/6 mice primarily due to their extensive first-pass phase II metabolism and poor solubilities. High plasma concentration of glucuronides upon oral administration and faster rate of glucuronidation by intestinal microsomes indicated intestine as one of the major metabolic organs for ME-KACs. Despite the increase in bioavailabilities of ME-KACs (∼8-10-fold) and decrease in AUC0-24 of glucuronides (∼10-fold) upon ME solubility enhancement, systemic exposure of ME-KACs failed to improve meaningfully. In conclusion, we propose a quality-controlled and chemically defined ME mixture, containing an optimized ratio of three KACs, delivered locally in the oral cavity as the most promising strategy for ME use as an oral cancer chemopreventive dietary supplement.
Collapse
Affiliation(s)
- Dinh Bui
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| | - Li Li
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| | - Taijun Yin
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| | - Xinli Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
- Fujian Medical University Union Hospital, Gulou District, Fuzhou City, Fujian, China
| | - Song Gao
- Department of Pharmaceutical Sciences, Texas Southern University, Houston, Texas
| | - Ming You
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Rashim Singh
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
- Corresponding Authors: [Tel: (832) 842-8320; Fax: (713) 743-1884; ] [Tel: (832) 518-9110; ]
| | - Ming Hu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
- Corresponding Authors: [Tel: (832) 842-8320; Fax: (713) 743-1884; ] [Tel: (832) 518-9110; ]
| |
Collapse
|
11
|
Ong CP, Lee WL, Tang YQ, Yap WH. Honokiol: A Review of Its Anticancer Potential and Mechanisms. Cancers (Basel) 2019; 12:E48. [PMID: 31877856 PMCID: PMC7016989 DOI: 10.3390/cancers12010048] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 12/19/2019] [Accepted: 12/19/2019] [Indexed: 12/24/2022] Open
Abstract
Cancer is characterised by uncontrolled cell division and abnormal cell growth, which is largely caused by a variety of gene mutations. There are continuous efforts being made to develop effective cancer treatments as resistance to current anticancer drugs has been on the rise. Natural products represent a promising source in the search for anticancer treatments as they possess unique chemical structures and combinations of compounds that may be effective against cancer with a minimal toxicity profile or few side effects compared to standard anticancer therapy. Extensive research on natural products has shown that bioactive natural compounds target multiple cellular processes and pathways involved in cancer progression. In this review, we discuss honokiol, a plant bioactive compound that originates mainly from the Magnolia species. Various studies have proven that honokiol exerts broad-range anticancer activity in vitro and in vivo by regulating numerous signalling pathways. These include induction of G0/G1 and G2/M cell cycle arrest (via the regulation of cyclin-dependent kinase (CDK) and cyclin proteins), epithelial-mesenchymal transition inhibition via the downregulation of mesenchymal markers and upregulation of epithelial markers. Additionally, honokiol possesses the capability to supress cell migration and invasion via the downregulation of several matrix-metalloproteinases (activation of 5' AMP-activated protein kinase (AMPK) and KISS1/KISS1R signalling), inhibiting cell migration, invasion, and metastasis, as well as inducing anti-angiogenesis activity (via the down-regulation of vascular endothelial growth factor (VEGFR) and vascular endothelial growth factor (VEGF)). Combining these studies provides significant insights for the potential of honokiol to be a promising candidate natural compound for chemoprevention and treatment.
Collapse
Affiliation(s)
| | | | - Yin Quan Tang
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor’s University Lakeside Campus, No. 1, Jalan Taylor’s, Subang Jaya 47500, Malaysia; (C.P.O.); (W.L.L.)
| | - Wei Hsum Yap
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor’s University Lakeside Campus, No. 1, Jalan Taylor’s, Subang Jaya 47500, Malaysia; (C.P.O.); (W.L.L.)
| |
Collapse
|
12
|
Choi YJ, Lee CM, Park SH, Nam MJ. Esculetin induces cell cycle arrest and apoptosis in human colon cancer LoVo cells. ENVIRONMENTAL TOXICOLOGY 2019; 34:1129-1136. [PMID: 31313495 DOI: 10.1002/tox.22815] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 06/25/2019] [Accepted: 06/26/2019] [Indexed: 06/10/2023]
Abstract
We investigated the anti-cancer effects of ESC in human colon cancer LoVo cells. Cell counting assay results showed that ESC inhibited the proliferation of LoVo cells. Cell cycle arrest results showed that cell cycle was arrested during the G0/G1 phase in the ESC-treated LoVo cells. Western blot results showed that the cell cycle inhibitory proteins p53, p27, and p21 were increased, and cyclin D1, the cell cycle progressive protein, was decreased. Sp1 is a transcription factor regulating cell proliferation, was decreased in the ESC-treated LoVo cells. Annexin V/propidium iodide staining results showed that ESC induces apoptosis in LoVo cells. Western blot results showed that Bax, cleaved caspase -3, -7, -9, and poly(ADP-ribose) polymerase, which are proapoptotic proteins, were increased and the antiapoptotic protein Bcl-2 was decreased. Taken together, ESC induced apoptosis and has an anti-cancer effect in LoVo cells.
Collapse
Affiliation(s)
- Yong J Choi
- Department of Biological Sciences, Gachon University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Chang M Lee
- Department of Biological Sciences, Gachon University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - See-Hyoung Park
- Department of Bio and Chemical Engineering, Hongik University, Sejong, Republic of Korea
| | - Myeong J Nam
- Department of Biological Sciences, Gachon University, Seongnam-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
13
|
Banik K, Ranaware AM, Deshpande V, Nalawade SP, Padmavathi G, Bordoloi D, Sailo BL, Shanmugam MK, Fan L, Arfuso F, Sethi G, Kunnumakkara AB. Honokiol for cancer therapeutics: A traditional medicine that can modulate multiple oncogenic targets. Pharmacol Res 2019; 144:192-209. [DOI: 10.1016/j.phrs.2019.04.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/18/2019] [Accepted: 04/02/2019] [Indexed: 02/07/2023]
|
14
|
Chang MT, Lee SP, Fang CY, Hsieh PL, Liao YW, Lu MY, Tsai LL, Yu CC, Liu CM. Chemosensitizing effect of honokiol in oral carcinoma stem cells via regulation of IL-6/Stat3 signaling. ENVIRONMENTAL TOXICOLOGY 2018; 33:1105-1112. [PMID: 30076764 DOI: 10.1002/tox.22587] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 05/23/2018] [Accepted: 05/31/2018] [Indexed: 06/08/2023]
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most common cancers worldwide with poor prognosis. Numerous studies have attempted to explore alternative regimens aimed at reducing cancer stem cells (CSCs) without compromising the efficacy of conventional chemoradiotherapy. The present study sought to assess the effect of a natural compound honokiol on the reduction of elevated cancer stemness, metastatic capacity, and chemoresistance of oral carcinoma stem cells (OCSCs). Our results demonstrated that honokiol attenuated the cell survival and self-renewal of OCSCs in a dose-dependent manner. Moreover, honokiol downregulated the expression of 2 selective markers of OCSCs, ALDH1, and CD44, as well as the migration and invasion abilities, indicating its potential to suppress cancer stemness. We showed that honokiol reduced the secretion of IL-6 and phosphorylation of STAT3, and the honokiol-inhibited self-renewal, invasion and colony formation were reversed by administration of IL-6. Most importantly, our data demonstrated that honokiol was able to potentiate the effect of Cisplatin, leading to a lower proportion of OCSCs and the decreased cancer stemness features. Taken together, this study demonstrated the benefits of utilizing honokiol as an adjunct therapy for OSCC treatment.
Collapse
Affiliation(s)
- Min-Te Chang
- Department of Oral and Maxillofacial Surgery, Chi Mei Medical Center, Tainan, Taiwan
| | - Shiao-Pieng Lee
- School of Dentistry, National Defense Medical Center, Taipei, Taiwan
- Department of Dentistry, Tri-Service General Hospital, Taipei, Taiwan
| | - Chih-Yuan Fang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Pei-Ling Hsieh
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Yi-Wen Liao
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
| | - Ming-Yi Lu
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Lo-Lin Tsai
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Chia Yu
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chia-Ming Liu
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
15
|
Liu F, Hu H, Zhao J, Zhang Z, Ai X, Tang L, Xie L. miR-124-3p acts as a potential marker and suppresses tumor growth in gastric cancer. Biomed Rep 2018; 9:147-155. [PMID: 30013778 PMCID: PMC6036824 DOI: 10.3892/br.2018.1113] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 01/02/2018] [Indexed: 12/19/2022] Open
Abstract
miR-124-3p has been implicated in a variety of cancers. The purpose of the present study was to investigate the expression, prognostic roles and functions of miR-124-3p in gastric cancer. Functional studies indicated that ectopic overexpression of miR-124-3p in gastric cancer cells suppressed cell viability and plate colony formation in vitro and tumor growth in vivo. In situ hybridization analysis demonstrated that decreased expression of miR-124-3p was associated with clinical stage and lymph node metastasis, as well as shorter overall survival and disease-free survival rates. Furthermore, it was observed that miR-124-3p repressed the carcinogenesis of gastric cancer by targeting Ras-related C3 botulinum toxin substrate 1 (Rac1) and specificity protein 1 (SP1). Collectively, these results indicate a potential underlying mechanism for the regulation of gastric cancer by miR-124-3p involving targeting of Rac1 and SP1. Thus, miR-124-3p may be an independent indicator of survival and treatment strategy for patients with gastric cancer.
Collapse
Affiliation(s)
- Feng Liu
- Medical School of University of South China, Hengyang, Hunan 421001, P.R. China
| | - Hongjuan Hu
- University of South China School of Nursing, Hengyang, Hunan 421001, P.R. China
| | - Jianfu Zhao
- Department of Oncology, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510630, P.R. China
| | - Zhiwei Zhang
- Medical School of University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xiaohong Ai
- The First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Liyun Tang
- The First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Liming Xie
- The First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
16
|
Deng Y, Han X, Tang S, Li C, Xiao W, Tan Z. Magnolol and Honokiol Attenuate Apoptosis of Enterotoxigenic Escherichia Coli-Induced Intestinal Epithelium by Maintaining Secretion and Absorption Homeostasis and Protecting Mucosal Integrity. Med Sci Monit 2018; 24:3348-3356. [PMID: 29782483 PMCID: PMC5990993 DOI: 10.12659/msm.910350] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 05/07/2018] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The cortex of Magnolia officinalis has long been used as an element of traditional Chinese medicine for the treatment of anxiety, chronic bronchitis, and gastrointestinal dysfunction. This study aimed to elucidate the underlying mechanism of its functional ingredients (magnolol and honokiol) in modifying the secretion and absorption homeostasis and protecting mucosal integrity in an Enterotoxigenic Escherichia coli (ETEC)-induced diarrhea mouse model. MATERIAL AND METHODS This study established a diarrhea mouse model infected by ETEC at a dosage of 0.02 ml/g live body weight (BW) in vivo. Magnolol or honokiol was followed by an intraperitoneal administration at dosages of 100, 300, and 500 mg/kg BW according to a 3×3 factorial arrangement. The useful biomarkers for evaluating the integrity of intestinal tract and histologic injury were analyzed and morphological development (including villus height, crypt depth, and ratio of villus height to crypt depth) and the expressions of inflammatory cytokines were determined by real-time PCR. RESULTS The results showed that magnolol and honokiol (500 mg/kg BW) reduced the concentrations of NO, DAO, and DLA, and iNOS activity, and the mRNA expressions of the interferon gamma (IFN-γ) and interleukin 10 (IL-10), and inhibited intestinal epithelial cell apoptosis. Magnolol and honokiol (300 mg/kg BW) elongated the villus height and crypt depth and decreased the number of goblet cells and the ratio of villus height to crypt depth. CONCLUSIONS The current results indicate that magnolol and honokiol enhance the intestinal anti-inflammatory capacities, elongate the villus height and crypt depth, and reduce goblet cell numbers to inhibit the intestinal epithelium apoptosis and effectively protect the intestinal mucosa. These results show that magnolol and honokiol protect the intestinal mucosal integrity and regulate gastrointestinal dysfunction.
Collapse
Affiliation(s)
- Yanli Deng
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, Changsha, Hunan, P.R. China
- Tea College of Guizhou University, Guiyang, Guizhou, P.R. China
| | - Xuefeng Han
- Key Laboratory for Agro-Ecological Processes in Subtropical Region, and South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, P.R. China
| | - Shaoxun Tang
- Key Laboratory for Agro-Ecological Processes in Subtropical Region, and South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, P.R. China
| | - Chengjian Li
- Department of Pharmacy, Yongzhou Vocational Technical College, Yongzhou, Hunan, P. R. China
| | - Wenjun Xiao
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, Changsha, Hunan, P.R. China
| | - Zhiliang Tan
- Key Laboratory for Agro-Ecological Processes in Subtropical Region, and South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, P.R. China
| |
Collapse
|
17
|
Safe S, Abbruzzese J, Abdelrahim M, Hedrick E. Specificity Protein Transcription Factors and Cancer: Opportunities for Drug Development. Cancer Prev Res (Phila) 2018; 11:371-382. [PMID: 29545399 DOI: 10.1158/1940-6207.capr-17-0407] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 02/14/2018] [Accepted: 02/28/2018] [Indexed: 02/06/2023]
Abstract
Specificity protein (Sp) transcription factors (TFs) such as Sp1 are critical for early development but their expression decreases with age and there is evidence that transformation of normal cells to cancer cells is associated with upregulation of Sp1, Sp3, and Sp4, which are highly expressed in cancer cells and tumors. Sp1 is a negative prognostic factor for pancreatic, colon, glioma, gastric, breast, prostate, and lung cancer patients. Functional studies also demonstrate that Sp TFs regulate genes responsible for cancer cell growth, survival, migration/invasion, inflammation and drug resistance, and Sp1, Sp3 and Sp4 are also nononcogene addiction (NOA) genes and important drug targets. The mechanisms of drug-induced downregulation of Sp TFs and pro-oncogenic Sp-regulated genes are complex and include ROS-dependent epigenetic pathways that initially decrease expression of the oncogene cMyc. Many compounds such as curcumin, aspirin, and metformin that are active in cancer prevention also exhibit chemotherapeutic activity and these compounds downregulate Sp TFs in cancer cell lines and tumors. The effects of these compounds on downregulation of Sp TFs in normal cells and the contribution of this response to their chemopreventive activity have not yet been determined. Cancer Prev Res; 11(7); 371-82. ©2018 AACR.
Collapse
Affiliation(s)
- Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas.
| | - James Abbruzzese
- Department of Medicine, Division of Oncology, Duke University School of Medicine, Durham, North Carolina
| | - Maen Abdelrahim
- GI Medical Oncology, Cockrell Center for Advanced Therapeutics, Houston Methodist Cancer Center and Institute of Academic Medicine, Houston, Texas
| | - Erik Hedrick
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| |
Collapse
|
18
|
Kang TH, Seo JH, Oh H, Yoon G, Chae JI, Shim JH. Licochalcone A Suppresses Specificity Protein 1 as a Novel Target in Human Breast Cancer Cells. J Cell Biochem 2017; 118:4652-4663. [PMID: 28498645 DOI: 10.1002/jcb.26131] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 05/11/2017] [Indexed: 12/24/2022]
Abstract
Licochalcone A (LCA), isolated from the root of Glycyrrhiza inflata, are known to have medicinal effect such as anti-oxidant, anti-bacterial, anti-viral, and anti-cancer. Though, as a pharmacological mechanism regulator, anti-cancer studies on LCA were not investigated in human breast cancer. We investigated the anti-proliferative and apoptotic effect of LCA in human breast cancer cells MCF-7 and MDA-MB-231 through MTS assay, PI staining, Annexin-V/7-AAD assay, mitochondrial membrane potential assay, multi-caspase assay, RT-PCR, Western blot analysis, and anchorage-independent cell transformation assay. Our results showed the little difference between two cells, as MCF-7 cell is both estrogen/progesterone receptor positive, there were only effect on Sp1 protein level, but not in mRNA level. Adversely, estrogen/progesterone/human epidermal growth factor receptor 2 triple negative, MDA-MB-231 showed decreased Sp1 mRNA, and protein levels. To confirm the participation of Sp1 in breast cancer cell viability, siRNA techniques were introduced. Both cells showed dysfunction of mitochondrial membrane potential and mitochondrial ROS production, which reflects it passed intracellular mitochondrial apoptosis pathway. Additionally, LCA showed the anti-proliferative and apoptotic effect in breast cancer cells through regulating Sp1 and apoptosis-related proteins in a dose- and a time-dependent manner. Consequently, LCA might be a potential anti-breast cancer drug substitute. J. Cell. Biochem. 118: 4652-4663, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Tae-Ho Kang
- Department of Dental Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 Plus, Chonbuk National University, Jeonju, 651-756, Republic of Korea
| | - Ji-Hye Seo
- Department of Dental Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 Plus, Chonbuk National University, Jeonju, 651-756, Republic of Korea
| | - Hana Oh
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 534-729, Republic of Korea
| | - Goo Yoon
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 534-729, Republic of Korea
| | - Jung-Il Chae
- Department of Dental Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 Plus, Chonbuk National University, Jeonju, 651-756, Republic of Korea
| | - Jung-Hyun Shim
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 534-729, Republic of Korea.,The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| |
Collapse
|
19
|
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: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/02/2016] [Indexed: 11/06/2022] Open
|
20
|
Honokiol inhibits sphere formation and xenograft growth of oral cancer side population cells accompanied with JAK/STAT signaling pathway suppression and apoptosis induction. BMC Cancer 2016; 16:245. [PMID: 27012679 PMCID: PMC4806492 DOI: 10.1186/s12885-016-2265-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 03/10/2016] [Indexed: 12/28/2022] Open
Abstract
Background Eliminating cancer stem cells (CSCs) has been suggested for prevention of tumor recurrence and metastasis. Honokiol, an active compound of Magnolia officinalis, had been proposed to be a potential candidate drug for cancer treatment. We explored its effects on the elimination of oral CSCs both in vitro and in vivo. Methods By using the Hoechst side population (SP) technique, CSCs-like SP cells were isolated from human oral squamous cell carcinoma (OSCC) cell lines, SAS and OECM-1. Effects of honokiol on the apoptosis and signaling pathways of SP-derived spheres were examined by Annexin V/Propidium iodide staining and Western blotting, respectively. The in vivo effectiveness was examined by xenograft mouse model and immunohistochemical tissue staining. Results The SP cells possessed higher stemness marker expression (ABCG2, Ep-CAM, Oct-4 and Nestin), clonogenicity, sphere formation capacity as well as tumorigenicity when compared to the parental cells. Treatment of these SP-derived spheres with honokiol resulted in apoptosis induction via Bax/Bcl-2 and caspase-3-dependent pathway. This apoptosis induction was associated with marked suppression of JAK2/STAT3, Akt and Erk signaling pathways in honokiol-treated SAS spheres. Consistent with its effect on JAK2/STAT3 suppression, honokiol also markedly inhibited IL-6-mediated migration of SAS cells. Accordingly, honokiol dose-dependently inhibited the growth of SAS SP xenograft and markedly reduced the immunohistochemical staining of PCNA and endothelial marker CD31 in the xenograft tumor. Conclusions Honokiol suppressed the sphere formation and xenograft growth of oral CSC-like cells in association with apoptosis induction and inhibition of survival/proliferation signaling pathways as well as angiogenesis. These results suggest its potential as an integrative medicine for combating oral cancer through targeting on CSCs. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2265-6) contains supplementary material, which is available to authorized users.
Collapse
|
21
|
Bang W, Jeon YJ, Cho JH, Lee RH, Park SM, Shin JC, Choi NJ, Choi YH, Cho JJ, Seo JM, Lee SY, Shim JH, Chae JI. β-lapachone suppresses the proliferation of human malignant melanoma cells by targeting specificity protein 1. Oncol Rep 2016; 35:1109-16. [PMID: 26718788 DOI: 10.3892/or.2015.4439] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 10/06/2015] [Indexed: 11/06/2022] Open
Abstract
β-lapachone (β-lap), a novel natural quinone derived from the bark of the Pink trumpet tree (Tabebuia avellanedae) has been demonstrated to have anticancer activity. In this study, we investigated whether β-lap exhibits anti-proliferative effects on two human malignant melanoma (HMM) cell lines, G361 and SK-MEL-28. The effects of β-lap on the HMM cell lines were investigated using 3-(4,5-dimethylthiazol-2-yl)‑5-(3-carboxymethoxyphenyl)‑2-(4-sulfophenyl-2H-tetrazolium (MTS) assay, 4',6-diamidino-2-phenylindole (DAPI) staining, Annexin V and Dead cell assay, mitochondrial membrane potential (MMP) assay and western blot analysis. We demonstrated that β-lap significantly induced apoptosis and suppressed cell viability in the HMM cells. Intriguingly, the transcription factor specificity protein 1 (Sp1) was significantly downregulated by β-lap in a dose- and time-dependent manner. Furthermore, β-lap modulated the protein expression level of the Sp1 regulatory genes including cell cycle regulatory proteins and apoptosis-associated proteins. Taken together, our findings indicated that β-lap modulates Sp1 transactivation and induces apoptotic cell death through the regulation of cell cycle- and apoptosis-associated proteins. Thus, β-lap may be used as a promising anticancer drug for cancer prevention and may improve the clinical outcome of patients with cancer.
Collapse
Affiliation(s)
- Woong Bang
- Department of Dental Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 Plus, Chonbuk National University, Jeonju 561-756, Republic of Korea
| | - Young-Joo Jeon
- Department of Dental Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 Plus, Chonbuk National University, Jeonju 561-756, Republic of Korea
| | - Jin Hyoung Cho
- Department of Dental Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 Plus, Chonbuk National University, Jeonju 561-756, Republic of Korea
| | - Ra Ham Lee
- Department of Dental Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 Plus, Chonbuk National University, Jeonju 561-756, Republic of Korea
| | - Seon-Min Park
- Pohang Center for Evaluation of Biomaterials, Pohang, Gyeongbuk 790‑834, Republic of Korea
| | - Jae-Cheon Shin
- Pohang Center for Evaluation of Biomaterials, Pohang, Gyeongbuk 790‑834, Republic of Korea
| | - Nag-Jin Choi
- Department of Animal Science, College of Agricultural and Life Science, Chonbuk National University, Jeonju 561‑756, Republic of Korea
| | - Yung Hyun Choi
- Department of Biochemistry, Dongeui University College of Oriental Medicine, Busan 614-052, Republic of Korea
| | - Jung-Jae Cho
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 534-729, Republic of Korea
| | - Jae-Min Seo
- Department of Prosthodontics, School of Dentistry and Institute of Oral Bio-Science and Research Institute of Clinical Medicine, Chonbuk National University, Jeonju 561-756, Republic of Korea
| | - Seung-Yeop Lee
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and School of Dentistry, Chonbuk National University, Jeonju 561-756, Republic of Korea
| | - Jung-Hyun Shim
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 534-729, Republic of Korea
| | - Jung-Il Chae
- Department of Dental Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 Plus, Chonbuk National University, Jeonju 561-756, Republic of Korea
| |
Collapse
|
22
|
Ju HM, Yu KW, Cho SD, Cheong SH, Kwon KH. Anti-cancer effects of traditional Korean wild vegetables in complementary and alternative medicine. Complement Ther Med 2016; 24:47-54. [DOI: 10.1016/j.ctim.2015.11.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 07/18/2015] [Accepted: 11/26/2015] [Indexed: 11/16/2022] Open
|
23
|
Cho JH, Lee RH, Jeon YJ, Shin JC, Park SM, Choi NJ, Seo KS, Yoon G, Cho SS, Kim KH, Cho JJ, Cho YS, Kim DH, Hong JT, Lee TH, Park HJ, Jung S, Seo JM, Chen H, Dong Z, Chae JI, Shim JH. Role of transcription factor Sp1 in the 4-O-methylhonokiol-mediated apoptotic effect on oral squamous cancer cells and xenograft. Int J Biochem Cell Biol 2015; 64:287-97. [PMID: 25982202 DOI: 10.1016/j.biocel.2015.05.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 04/14/2015] [Accepted: 05/07/2015] [Indexed: 11/18/2022]
Abstract
Recently, biphenolic components derived from the Magnolia family have been studied for anti-cancer, anti-stress, and anti-inflammatory pharmacological effects. However, the pharmacological mechanism of action of 4-O-methylhonokiol (MH) is not clear in oral cancer. The aim of this study was to investigate the role of MH in apoptosis and its molecular mechanism in oral squamous cell carcinoma (OSCC) cell lines, HN22 and HSC4, as well as tumor xenografts. Here, we demonstrated that MH decreased cell growth and induced apoptosis in HN22 and HSC4 cells through the regulation of specificity protein 1 (Sp1). We employed several experimental techniques such as MTS assay, DAPI staining, PI staining, Annexin-V/7-ADD staining, RT-PCR, western blot analysis, immunocytochemistry, immunohistochemistry, TUNEL assay and in vivo xenograft model analysis. MH inhibited Sp1 protein expression and reduced Sp1 protein levels via both proteasome-dependent protein degradation and inhibition of protein synthesis in HN22 and HSC4 cells; MH did not alter Sp1 mRNA levels. We found that MH directly binds Sp1 by Sepharose 4B pull-down assay and molecular modeling. In addition, treatment with MH or knocking down Sp1 expression suppressed oral cancer cell colony formation. Moreover, MH treatment effectively inhibited tumor growth and Sp1 levels in BALB/c nude mice bearing HN22 cell xenografts. These results indicated that MH inhibited cell growth, colony formation and also induced apoptosis via Sp1 suppression in OSCC cells and xenograft tumors. Thus, MH is a potent anti-cancer drug candidate for oral cancer.
Collapse
Affiliation(s)
- Jin Hyoung Cho
- Department of Oral Pharmacology, School of Dentistry and Institute of Dental Bioscience, BK21 plus, Chonbuk National University, Jeonju, Republic of Korea
| | - Ra Ham Lee
- Department of Oral Pharmacology, School of Dentistry and Institute of Dental Bioscience, BK21 plus, Chonbuk National University, Jeonju, Republic of Korea
| | - Young-Joo Jeon
- Department of Oral Pharmacology, School of Dentistry and Institute of Dental Bioscience, BK21 plus, Chonbuk National University, Jeonju, Republic of Korea
| | - Jae-Cheon Shin
- Pohang Center for Evaluation of Biomaterials, Pohang, Gyeongbuk, Republic of Korea
| | - Seon-Min Park
- Pohang Center for Evaluation of Biomaterials, Pohang, Gyeongbuk, Republic of Korea
| | - Nag-Jin Choi
- Department of Animal Science, College of Agricultural and Life Science, Chonbuk National University, Jeonju, Republic of Korea
| | - Kang Seok Seo
- Department of Animal Science and Technology, Sunchon National University, Suncheon, Republic of Korea
| | - Goo Yoon
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan-gun, Republic of Korea
| | - Seung-Sik Cho
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan-gun, Republic of Korea
| | - Ka Hwi Kim
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan-gun, Republic of Korea
| | - Jung Jae Cho
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan-gun, Republic of Korea
| | - Young Sik Cho
- Department of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Dae Hwan Kim
- College of Pharmacy, Medical Research Center, Chungbuk National University, Cheongju, Republic of Korea
| | - Jin Tae Hong
- Department of Oral Biochemistry, Dental Science Research Institute and the BK21 Project, Medical Research Center for Biomineralization Disorders, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Tae-Hoon Lee
- Department of Oral Biochemistry, Dental Science Research Institute and the BK21 Project, Medical Research Center for Biomineralization Disorders, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Hong Ju Park
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Seunggon Jung
- Department of Oral and Maxillofacial Surgery, Chonnam National University Hwasun Hospital, Gwangju, Republic of Korea
| | - Jae-Min Seo
- Department of Prosthodontics, School of Dentistry, Chonbuk National University, Jeonju, Republic of Korea
| | - Hanyong Chen
- Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Zigang Dong
- Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Jung-Il Chae
- Department of Oral Pharmacology, School of Dentistry and Institute of Dental Bioscience, BK21 plus, Chonbuk National University, Jeonju, Republic of Korea.
| | - Jung-Hyun Shim
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan-gun, Republic of Korea.
| |
Collapse
|
24
|
Cho JH, Jeon YJ, Park SM, Shin JC, Lee TH, Jung S, Park H, Ryu J, Chen H, Dong Z, Shim JH, Chae JI. Multifunctional effects of honokiol as an anti-inflammatory and anti-cancer drug in human oral squamous cancer cells and xenograft. Biomaterials 2015; 53:274-84. [DOI: 10.1016/j.biomaterials.2015.02.091] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 02/16/2015] [Accepted: 02/21/2015] [Indexed: 11/27/2022]
|
25
|
Jeon YJ, Bang W, Shin JC, Park SM, Cho JJ, Choi YH, Seo KS, Choi NJ, Shim JH, Chae JI. Downregulation of Sp1 is involved in β-lapachone-induced cell cycle arrest and apoptosis in oral squamous cell carcinoma. Int J Oncol 2015; 46:2606-12. [PMID: 25891355 DOI: 10.3892/ijo.2015.2972] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 04/08/2015] [Indexed: 11/05/2022] Open
Abstract
β-lapachone (β-lap) is a naturally occurring quinone obtained from the bark of lapacho tree (Tabebuia avellanedae) with anti-proliferative properties against various cancers. The present study investigated the cell proliferation and apoptosis effect of β-lap on two oral squamous cell carcinoma lines (OSCCs). We carried out a series of 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl-2H-tetrazolium (MTS) assays, 4',6-diamidino-2-phenylindole (DAPI) staining, cell cycle analysis, and western blot analysis to characterize β-lap and its underlying signaling pathway. We demonstrated that β-lap-treated cells significantly reduced cell proliferation but increased DNA condensation and increased sub-G1 population in OSCCs. Particularly, β-lap suppresses activation of transcription factor specificity protein 1 (Sp1) followed by apoptosis in a concentration-dependent manner in OSCCs. Furthermore, β-lap modulated protein expression levels of cell cycle regulatory proteins and apoptosis-related proteins that are known as Sp1 target genes, resulting in apoptosis. Our results collectively indicated that β-lap was able to modulate Sp1 transactivation and induce apoptosis through the regulation of cell cycle and apoptosis-related proteins. Therefore, β-lap may be used in cancer prevention and therapies to improve clinical outcome as an anticancer drug candidate.
Collapse
Affiliation(s)
- Young-Joo Jeon
- Department of Oral Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 plus, Chonbuk National University, Jeonju 561-756, Republic of Korea
| | - Woong Bang
- Department of Oral Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 plus, Chonbuk National University, Jeonju 561-756, Republic of Korea
| | - Jae-Cheon Shin
- Pohang Center for Evaluation of Biomaterials, Pohang, Gyeongbuk 790‑834, Republic of Korea
| | - Seon-Min Park
- Pohang Center for Evaluation of Biomaterials, Pohang, Gyeongbuk 790‑834, Republic of Korea
| | - Jung-Jae Cho
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 534-729, Republic of Korea
| | - Yung Hyun Choi
- Department of Biochemistry, Dongeui University College of Oriental Medicine, Busan 614-052, Republic of Korea
| | - Kang Seok Seo
- Department of Animal Science and Technology, Sunchon National University, Suncheon, Republic of Korea
| | - Nag-Jin Choi
- Department of Animal Science, College of Agricultural and Life Science, Chonbuk National University, Jeonju 651-756, Republic of Korea
| | - Jung-Hyun Shim
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 534-729, Republic of Korea
| | - Jung-Il Chae
- Department of Oral Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 plus, Chonbuk National University, Jeonju 561-756, Republic of Korea
| |
Collapse
|
26
|
Cheng S, Castillo V, Eliaz I, Sliva D. Honokiol suppresses metastasis of renal cell carcinoma by targeting KISS1/KISS1R signaling. Int J Oncol 2015; 46:2293-8. [PMID: 25846316 PMCID: PMC4441299 DOI: 10.3892/ijo.2015.2950] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 02/10/2015] [Indexed: 01/01/2023] Open
Abstract
Renal cell carcinoma (RCC) is a common urological cancer worldwide and is known to have a high risk of metastasis, which is considered responsible for more than 90% of cancer associated deaths. Honokiol is a small-molecule biphenol isolated from Magnolia spp. bark and has been shown to be a potential anticancer agent involved in multiple facets of signal transduction. In this study, we demonstrated that honokiol inhibited the invasion and colony formation of highly metastatic RCC cell line 786-0 in a dose-dependent manner. DNA-microarray data showed the significant upregulation of metastasis-suppressor gene KISS1 and its receptor, KISS1R. The upregulation was confirmed by qRT-PCR analysis. Overexpression of KISS1 and KISS1R was detected by western blotting at the translation level as well. Of note, the decreased invasive and colonized capacities were reversed by KISS1 knockdown. Taken together, the results first indicate that activation of KISS1/KISS1R signaling by honokiol suppresses multistep process of metastasis, including invasion and colony formation, in RCC cells 786-0. Honokiol may be considered as a natural agent against RCC metastasis.
Collapse
Affiliation(s)
- Shujie Cheng
- Cancer Research Laboratory, Methodist Research Institute, Indiana University Health, Indianapolis, IN, USA
| | - Victor Castillo
- Cancer Research Laboratory, Methodist Research Institute, Indiana University Health, Indianapolis, IN, USA
| | - Isaac Eliaz
- Amitabha Medical Clinic and Healing Center, Santa Rosa, CA, USA
| | - Daniel Sliva
- Cancer Research Laboratory, Methodist Research Institute, Indiana University Health, Indianapolis, IN, USA
| |
Collapse
|
27
|
Jeon YJ, Bang W, Choi YH, Shim JH, Chae JI. Beta-Lapachone Suppresses Non-small Cell Lung Cancer Proliferation through the Regulation of Specificity Protein 1. Biol Pharm Bull 2015; 38:1302-8. [PMID: 26328485 DOI: 10.1248/bpb.b15-00159] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Lung cancer is the leading cause of cancer-related death worldwide, and non-small cell lung cancer (NSCLC) is the most common pathological type with a reported frequency of about 85% of all cases. Despite recent advances in therapeutic agents and targeted therapies, the prognosis for NSCLC remains poor, and therefore it is important to identify the biological targets of this complex disease since a blockade of such targets would affect multiple downstream signaling cascades. β-Lapachone (β-Lap) is an antiproliferative agent that selectively induces apoptosis-related cell death in a variety of human cancer cells. However, the mechanisms of its action require further investigation. In this study, we show that treatment with β-lap triggers apoptosis and cell-cycle arrest in two NSCLC cell lines: H1299 and NCI-H358. The transcription factor specificity protein 1 (Sp1) was markedly inhibited by β-lap in a dose- and time-dependent manner. Furthermore, β-lap modulated the protein expression levels of the Sp1 regulatory genes, including cell-cycle regulatory proteins and antiapoptotic proteins, resulting in apoptosis. Taken together, our results indicate that β-lap may be a potential antiproliferative agent candidate by inducing apoptotic cell death in NSCLC tissue through downregulation of Sp1.
Collapse
Affiliation(s)
- Young-Joo Jeon
- Department of Dental Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 plus, Chonbuk National University
| | | | | | | | | |
Collapse
|
28
|
Chae JI, Jeon YJ, Shim JH. Anti-proliferative properties of kahweol in oral squamous cancer through the regulation specificity protein 1. Phytother Res 2014; 28:1879-86. [PMID: 25196544 DOI: 10.1002/ptr.5217] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 07/24/2014] [Accepted: 07/28/2014] [Indexed: 12/20/2022]
Abstract
Kahweol, the coffee-specific deterpene, has been shown to have potential anti-cancer effects against several cancers. However, the molecular mechanisms underlying the anti-cancer activity of kahweol have not yet established. In this study, we investigated whether kahweol could show anti-cancer effects on oral squamous cell lines (OSCCs), HN22 and HSC4. We conducted an 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxy-phenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay, 4'-6-diamidino2-phenylindole (DAPI) staining, propidium iodide staining, immunocytochemistry, and Western blot analysis for the characterization of kahweol and the underlying signaling pathway. We determined that kahweol-treated cells showed significantly decreased cell viability and increased nuclear condensation and an increased sub-G1 population in OSCCs. Interestingly, suppression of the transcription factor specificity protein 1 (Sp1) was followed by induced apoptosis by kahweol in a dose-dependent manner. In addition, kahweol modulated the protein expression level of the Sp1 regulatory genes including cell cycle regulatory proteins and anti-apoptotic proteins, resulting in apoptosis. Taken together, results from these findings suggest that kahweol may be a potential anti-cancer drug candidate to induce apoptotic cell death through downregulation of Sp1 in OSCCs.
Collapse
Affiliation(s)
- Jung-Il Chae
- Department of Oral Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 plus, Chonbuk National University, Jeonju, 651-756, Korea
| | | | | |
Collapse
|
29
|
CHO JINHYOUNG, SHIN JAECHEON, CHO JUNGJAE, CHOI YUNGHYUN, SHIM JUNGHYUN, CHAE JUNGIL. Esculetin (6,7-dihydroxycoumarin): A potential cancer chemopreventive agent through suppression of Sp1 in oral squamous cancer cells. Int J Oncol 2014; 46:265-71. [DOI: 10.3892/ijo.2014.2700] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 09/10/2014] [Indexed: 11/05/2022] Open
|
30
|
Cho JJ, Chae JI, Yoon G, Kim KH, Cho JH, Cho SS, Cho YS, Shim JH. Licochalcone A, a natural chalconoid isolated from Glycyrrhiza inflata root, induces apoptosis via Sp1 and Sp1 regulatory proteins in oral squamous cell carcinoma. Int J Oncol 2014; 45:667-74. [PMID: 24858379 DOI: 10.3892/ijo.2014.2461] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 05/07/2014] [Indexed: 11/06/2022] Open
Abstract
Licochalcone A (LCA), a chalconoid derived from root of Glycyrrhiza inflata, has been known to possess a wide range of biological functions such as antitumor, anti-angiogenesis, antiparasitic, anti-oxidant, antibacterial and anti-inflammatory effects. However, the anticancer effects of LCA on oral squamous cell carcinoma (OSCC) have not been reported. Our data showed that LCA inhibited OSCC cell (HN22 and HSC4) growth in a concentration- and time-dependent manner. Mechanistically, it was mediated via downregulation of specificity protein 1 (Sp1) expression and subsequent regulation of Sp1 downstream proteins such as p27, p21, cyclin D1, Mcl-1 and survivin. Here, we found that LCA caused apoptotic cell death in HSC4 and HN22 cells, as characterized by sub-G1 population, nuclear condensation, Annexin V staining, and multi-caspase activity and apoptotic regulatory proteins such as Bax, Bid, Bcl(-xl), caspase-3 and PARP. Consequently, this study strongly suggests that LCA induces apoptotic cell death of OSCC cells via downregulation of Sp1 expression, prompting its potential use for the treatment of human OSCC.
Collapse
Affiliation(s)
- Jung Jae Cho
- Natural Medicine Research Institute, Department of Pharmacy, College of Pharmacy, Mokpo National University, Jeonnam 534-729, Republic of Korea
| | - Jung-Il Chae
- Department of Oral Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 plus, Chonbuk National University, Jeonju 651-756, Republic of Korea
| | - Goo Yoon
- Natural Medicine Research Institute, Department of Pharmacy, College of Pharmacy, Mokpo National University, Jeonnam 534-729, Republic of Korea
| | - Ka Hwi Kim
- Natural Medicine Research Institute, Department of Pharmacy, College of Pharmacy, Mokpo National University, Jeonnam 534-729, Republic of Korea
| | - Jin Hyoung Cho
- Department of Oral Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 plus, Chonbuk National University, Jeonju 651-756, Republic of Korea
| | - Seung-Sik Cho
- Natural Medicine Research Institute, Department of Pharmacy, College of Pharmacy, Mokpo National University, Jeonnam 534-729, Republic of Korea
| | - Young Sik Cho
- College of Pharmacy, Keimyung University, Dalseo-gu, Daegu 704-701, Republic of Korea
| | - Jung-Hyun Shim
- Natural Medicine Research Institute, Department of Pharmacy, College of Pharmacy, Mokpo National University, Jeonnam 534-729, Republic of Korea
| |
Collapse
|
31
|
Bernaskova M, Kretschmer N, Schuehly W, Huefner A, Weis R, Bauer R. Synthesis of tetrahydrohonokiol derivates and their evaluation for cytotoxic activity against CCRF-CEM leukemia, U251 glioblastoma and HCT-116 colon cancer cells. Molecules 2014; 19:1223-37. [PMID: 24448063 PMCID: PMC6270748 DOI: 10.3390/molecules19011223] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 01/09/2014] [Accepted: 01/13/2014] [Indexed: 01/27/2023] Open
Abstract
Biphenyl neolignans such as honokiol and magnolol, which are the major active constituents of the Asian medicinal plant Magnolia officinalis, are known to exert a multitude of pharmacological and biological activities. Among these, cytotoxic and tumor growth inhibitory activity against various tumour cell lines are well-documented. To further elucidate the cytotoxic effects of honokiol derivatives, derivatizations were performed using tetrahydrohonokiol as a scaffold. The derivatizations comprised the introduction of functional groups, e.g., nitro and amino groups, as well as alkylation. This way, 18 derivatives, of which 13 were previously undescribed compounds, were evaluated against CCRF-CEM leukemia cells, U251 glioblastoma and HCT-116 colon cancer cells. The results revealed no significant cytotoxic effects in any of the three tested cell lines at a test concentration of 10 µM.
Collapse
Affiliation(s)
- Marketa Bernaskova
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, Universitätsplatz 1, University of Graz, 8010 Graz, Austria.
| | - Nadine Kretschmer
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, Universitätsplatz 1, University of Graz, 8010 Graz, Austria.
| | - Wolfgang Schuehly
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, Universitätsplatz 1, University of Graz, 8010 Graz, Austria.
| | - Antje Huefner
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, Universitätsplatz 1, University of Graz, 8010 Graz, Austria.
| | - Robert Weis
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, Universitätsplatz 1, University of Graz, 8010 Graz, Austria.
| | - Rudolf Bauer
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, Universitätsplatz 1, University of Graz, 8010 Graz, Austria.
| |
Collapse
|
32
|
Averett C, Arora S, Zubair H, Singh S, Bhardwaj A, Singh AP. Molecular Targets of Honokiol: A Promising Phytochemical for Effective Cancer Management. Enzymes 2014; 36:175-93. [PMID: 27102704 DOI: 10.1016/b978-0-12-802215-3.00009-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Honokiol is a bioactive, biphenolic phytochemical, present in the aerial parts of plants classified under the genus Magnolia. It has been an important constituent of Asian traditional medicine and is used against many ailments. Honokiol possesses potent antioxidative, anti-inflammatory, antiangiogenic, and anticancer activities by targeting a variety of signaling molecules. Consequently, there has been immense interest in exploring its utility as a novel chemopreventive and therapeutic agent against several malignancies. In this chapter, we review the structure-function relationship of honokiol and its derivative compounds, the impact of honokiol on various phenotypes associated with cancer progression and metastasis, and its prominent molecular targets and pharmacokinetics. Clearly, the available data generate significant interest in this novel phytochemical and emerging information continues to provide strong support for its potential applicability in cancer management.
Collapse
Affiliation(s)
- Courey Averett
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Sumit Arora
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Haseeb Zubair
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Seema Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Arun Bhardwaj
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Ajay P Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA.
| |
Collapse
|