1
|
Lee WJ, Jo JH, Jang SI, Jung EJ, Hwang JM, Bae JW, Ha JJ, Kim DH, Kwon WS. The natural flavonoid compound deguelin suppresses sperm (Sus Scrofa) functions through abnormal activation of the PI3K/AKT pathway. Reprod Toxicol 2023; 120:108426. [PMID: 37353039 DOI: 10.1016/j.reprotox.2023.108426] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/05/2023] [Accepted: 06/20/2023] [Indexed: 06/25/2023]
Abstract
Deguelin is a natural flavonoid extracted from plants belonging to the Lonchocarpus, Derris, or Tephrosia genera. It inhibits AKT activity in tumors and has the potential to be used as a treatment for malignant tumors. However, the risks associated with the use of deguelin on male fertility have not yet been explained in detail. Therefore, this study was conducted to investigate the effects of deguelin on sperm functions during capacitation. First, boar spermatozoa were exposed to different concentrations of deguelin (0.1, 1, 10, 50, and 100 μM). Next, sperm functional assessments, such as sperm motility, capacitation status, intracellular ATP level, and cell viability, were performed. The expression levels of PI3K/AKT-related proteins and the phosphorylation of their tyrosine residues were also evaluated by western blotting. No significant difference was observed in cell viability; however, deguelin considerably decreased sperm motility and motion kinematics in a dose-dependent manner. Although no significant difference was observed in the capacitation status, acrosome reaction decreased at high concentrations of deguelin (50 and 100 μM). Furthermore, intracellular ATP levels were significantly decreased in all deguelin treatment groups compared with those in the control group. Results of western blotting revealed that deguelin substantially diminished tyrosine phosphorylation. Interestingly, in contrast to previous studies showing that deguelin inhibits AKT activity, our results showed that it increased the expression of PI3K/AKT pathway-related proteins. Collectively, these findings indicate that deguelin exerts negative effects on sperm functions due to abnormal PI3K/AKT signaling activation. We believe that this is the first study to provide evidence that deguelin can regulate sperm functions independent of PI3K/AKT pathway inhibition. Furthermore, its detrimental effects on male fertility should be considered while developing or using deguelin as a therapeutic agent.
Collapse
Affiliation(s)
- Woo-Jin Lee
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju, Gyeongsangbuk-do 37224, the Republic of Korea
| | - Jae-Hwan Jo
- Department of Animal Biotechnology, Kyungpook National University, Sangju, Gyeongsangbuk-do 37224, the Republic of Korea
| | - Seung-Ik Jang
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju, Gyeongsangbuk-do 37224, the Republic of Korea
| | - Eun-Ju Jung
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju, Gyeongsangbuk-do 37224, the Republic of Korea
| | - Ju-Mi Hwang
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju, Gyeongsangbuk-do 37224, the Republic of Korea
| | - Jeong-Won Bae
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju, Gyeongsangbuk-do 37224, the Republic of Korea
| | - Jae Jung Ha
- Gyeongbuk Livestock Research Institute, Yeongju, Gyeongsangbuk-do 36052, the Republic of Korea
| | - Dae-Hyun Kim
- Gyeongbuk Livestock Research Institute, Yeongju, Gyeongsangbuk-do 36052, the Republic of Korea.
| | - Woo-Sung Kwon
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju, Gyeongsangbuk-do 37224, the Republic of Korea; Department of Animal Biotechnology, Kyungpook National University, Sangju, Gyeongsangbuk-do 37224, the Republic of Korea; Research Center for Horse Industry, Kyungpook National University, Sangju, Gyeongsangbuk-do 37224, the Republic of Korea.
| |
Collapse
|
2
|
Liu H, Xu L, Zhou L, Han W, Li Z, Liu C. DBP induced autophagy and necrotic apoptosis in HepG2 cells via the mitochondrial damage pathway. Food Chem Toxicol 2023; 176:113782. [PMID: 37059380 DOI: 10.1016/j.fct.2023.113782] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 04/05/2023] [Accepted: 04/11/2023] [Indexed: 04/16/2023]
Abstract
Phthalate esters (PAEs) are widely present in human tissues and pose significant health risks. In this study, HepG2 cells were treated with 0.0625, 0.125, 0.25, 0.5 and 1 mM Dibutyl phthalate (DBP) for 48 h to investigate mitochondrial toxicity. The results showed that DBP caused mitochondrial damage, autophagy, apoptosis and necroptosis; Transcriptomics analysis identified that MAPK and PI3K were significant factors in the cytotoxic changes induced by DBP; N-Acetyl-L-cysteine (NAC), SIRT1 activator, ERK inhibitor, p38 inhibitor and ERK siRNA treatments counteracted the changes of SIRT1/PGC-1α and Nrf2 pathway-related proteins, autophagy and necroptotic apoptosis proteins induced by DBP. While PI3K and Nrf2 inhibitors exacerbated the changes in SIRT1/PGC-1α, Nrf2-associated proteins and autophagy and necroptosis proteins induced by DBP. In addition, the autophagy inhibitor 3-MA alleviated the increase in DBP-induced necroptosis proteins. These results suggested that DBP-induced oxidative stress activated the MAPK pathway, inhibited the PI3K pathway, which in turn inhibited the SIRT1/PGC-1α pathway and Nrf2 pathway, thereby causing cell autophagy and necroptosis.
Collapse
Affiliation(s)
- Huan Liu
- College of Food Science, South China Agricultural University, Guangzhou, 510642, China; Guangdong Provincial Key Laboratory of Food Quality and Safety, Guangzhou, 510642, China.
| | - Linjing Xu
- College of Food Science, South China Agricultural University, Guangzhou, 510642, China; Guangdong Provincial Key Laboratory of Food Quality and Safety, Guangzhou, 510642, China.
| | - Lizi Zhou
- College of Food Science, South China Agricultural University, Guangzhou, 510642, China; Guangdong Provincial Key Laboratory of Food Quality and Safety, Guangzhou, 510642, China.
| | - Wenna Han
- College of Food Science, South China Agricultural University, Guangzhou, 510642, China; Guangdong Provincial Key Laboratory of Food Quality and Safety, Guangzhou, 510642, China.
| | - Zhongyi Li
- College of Food Science, South China Agricultural University, Guangzhou, 510642, China; Guangdong Provincial Key Laboratory of Food Quality and Safety, Guangzhou, 510642, China.
| | - Chunhong Liu
- College of Food Science, South China Agricultural University, Guangzhou, 510642, China; Guangdong Provincial Key Laboratory of Food Quality and Safety, Guangzhou, 510642, China.
| |
Collapse
|
3
|
Tuli HS, Mittal S, Loka M, Aggarwal V, Aggarwal D, Masurkar A, Kaur G, Varol M, Sak K, Kumar M, Sethi G, Bishayee A. Deguelin targets multiple oncogenic signaling pathways to combat human malignancies. Pharmacol Res 2021; 166:105487. [PMID: 33581287 DOI: 10.1016/j.phrs.2021.105487] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/23/2021] [Accepted: 02/07/2021] [Indexed: 02/07/2023]
Abstract
Cancer is an anomalous growth and differentiation of cells known to be governed by oncogenic factors. Plant-based natural metabolites have been well recognized to possess chemopreventive properties. Deguelin, a natural rotenoid, is among the class of bioactive phytoconstituents from a diverse range of plants with potential antineoplastic effects in different cancer subtypes. However, the precise mechanisms of how deguelin inhibits tumor progression remains elusive. Deguelin has shown promising results in targeting the hallmarks of tumor progression via inducing tumor apoptosis, cell cycle arrest, and inhibition of angiogenesis and metastasis. Based on initial scientific excerpts, deguelin has been reported to inhibit tumor growth via different signaling pathways, including mitogen-activated protein kinase, phosphoinositide 3-kinase, serine/threonine protein kinase B (also known as Akt), mammalian target of rapamycin, nuclear factor-κB, matrix metalloproteinase (MMP)-2, MMP-9 and caspase-3, caspase-8, and caspase-9. This review summarizes the mechanistic insights of antineoplastic action of deguelin to gain a clear understanding of its therapeutic effects in cancer. The anticancer potential of deguelin with respect to its efficacy in targeting tumorigenesis via nanotechnological approaches is also investigated. The initial scientific findings have presented deguelin as a promising antitumorigenic agent which can be used for monotherapy as well as synergistically to augment efficacy of chemotherapeutic treatment regimes.
Collapse
Affiliation(s)
- Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala 133 207, Haryana, India.
| | - Sonam Mittal
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Mariam Loka
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
| | - Vaishali Aggarwal
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA15260, USA
| | - Diwakar Aggarwal
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala 133 207, Haryana, India
| | - Akshara Masurkar
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, Narsee Monjee Institute of Management Studies University, Mumbai 400 056, Maharashtra, India
| | - Ginpreet Kaur
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, Narsee Monjee Institute of Management Studies University, Mumbai 400 056, Maharashtra, India
| | - Mehmet Varol
- Department of Molecular Biology and Genetics, Faculty of Science, Mugla Sitki Kocman University, Mugla TR48000, Turkey
| | | | - Manoj Kumar
- Department of Chemistry, Maharishi Markandeshwar (Deemed to be University), Sadopur 134007, Haryana, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA.
| |
Collapse
|
4
|
An Electrophilic Deguelin Analogue Inhibits STAT3 Signaling in H- Ras-Transformed Human Mammary Epithelial Cells: The Cysteine 259 Residue as a Potential Target. Biomedicines 2020; 8:biomedicines8100407. [PMID: 33053804 PMCID: PMC7600869 DOI: 10.3390/biomedicines8100407] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/04/2020] [Accepted: 10/09/2020] [Indexed: 11/17/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a point of convergence for numerous oncogenic signals that are often constitutively activated in many cancerous or transformed cells and some stromal cells in the tumor microenvironment. Persistent STAT3 activation in malignant cells stimulates proliferation, survival, angiogenesis, invasion, and tumor-promoting inflammation. STAT3 undergoes activation through phosphorylation on tyrosine 705, which facilitates its dimerization. Dimeric STAT3 translocates to the nucleus, where it regulates the transcription of genes involved in cell proliferation, survival, etc. In the present study, a synthetic deguelin analogue SH48, discovered by virtual screening, inhibited the phosphorylation, nuclear translocation, and transcriptional activity of STAT3 in H-ras transformed human mammary epithelial MCF-10A cells (MCF10A-ras). We speculated that SH48 bearing an α,β-unsaturated carbonyl group could interact with a thiol residue of STAT3, thereby inactivating this transcription factor. Non-electrophilic analogues of SH48 failed to inhibit STAT3 activation, lending support to the above supposition. By utilizing a biotinylated SH48, we were able to demonstrate the complex formation between SH48 and STAT3. SH48 treatment to MCF10A-ras cells induced autophagy, which was verified by staining with a fluorescent acidotropic probe, LysoTracker Red, as well as upregulating the expression of LC3II and p62. In conclusion, the electrophilic analogue of deguelin interacts with STAT3 and inhibits its activation in MCF10A-ras cells, which may account for its induction of autophagic death.
Collapse
|
5
|
Liao W, Liu X, Yang Q, Liu H, Liang B, Jiang J, Huang J, Ning C, Zang N, Zhou B, Liao Y, Chen J, Tian L, Ho W, Abdullah AS, Kong L, Liang H, Chen H, Ye L. Deguelin inhibits HCV replication through suppressing cellular autophagy via down regulation of Beclin1 expression in human hepatoma cells. Antiviral Res 2020; 174:104704. [PMID: 31917237 DOI: 10.1016/j.antiviral.2020.104704] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 12/15/2019] [Accepted: 01/02/2020] [Indexed: 02/06/2023]
Abstract
AIMS Deguelin, a natural compound derived from Mundulea sericea (Leguminosae) and some other plants exhibits an activity to inhibit autophagy, a cellular machinery required for hepatitis C virus (HCV) replication. This study aimed to illuminate the impact of deguelin on HCV replication and mechanism(s) involved. METHODS HCV JFH-1-Huh7 infectious system was used for the investigation. Real time RT-PCR, Western blot, fluorescent microscopy assay were used to measure the expression levels of viral or cellular factors. Overexpression and silencing expression techniques were used to determine the role of key cellular factors. RESULTS Deguelin treatment of Huh7 cells significantly inhibited HCV JFH-1 replication in a dose- and time-dependent manner. Deguelin treatment suppressed autophagy in Huh7 cells, evidenced by the decrease of LC3B-II levels, the conversion of LC3B-I to LC3B-II, and the formation of GFP-LC3 puncta as well as the increase of p62 level in deguelin-treated cells compared with control cells. HCV infection could induce autophagy which was also suppressed by deguelin treatment. Mechanism research reveals that deguelin inhibited expression of Beclin1, which is a key cellular factor for the initiation of the autophagosome formation in autophagy. Overexpression or silencing expression of Beclin1 in deguelin-treated Huh7 cells could weaken or enhance the inhibitory effect on autophagy by deguelin, respectively, and thus partially recover or further inhibit HCV replication correspondingly. CONCLUSIONS Deguelin may serve as a novel anti-HCV compound via its inhibitory effect on autophagy, which warrants further investigation as a potential therapeutic agent for HCV infection.
Collapse
Affiliation(s)
- Weibo Liao
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Geriatrics Digestion Department of Internal Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xin Liu
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Quanlue Yang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Huifang Liu
- Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Bingyu Liang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Junjun Jiang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jiegang Huang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Chuanyi Ning
- Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Ning Zang
- Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Bo Zhou
- Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yanyan Liao
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jingzhao Chen
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Geriatrics Digestion Department of Internal Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Li Tian
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Geriatrics Digestion Department of Internal Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Wenzhe Ho
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA, 19140, USA
| | - Abu S Abdullah
- Boston University School of Medicine, Boston Medical Center, Boston, MA, 02118, USA
| | - Lingbao Kong
- Institute of Pathogenic Microorganism, College of Bioscience and Bioengineering, Jiangxi Agricultural University, Nanchang, 330045, Jiangxi, China
| | - Hao Liang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| | - Hui Chen
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Geriatrics Digestion Department of Internal Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| | - Li Ye
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
6
|
范 俣, 刘 瑞, 丁 晓, 上官 信, 吴 新. [Deguelin inhibits proliferation and regulates the expression of MCM3-CDC45 in MCF-7 and H1299 cells in vitro]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:1545-1550. [PMID: 29180339 PMCID: PMC6779631 DOI: 10.3969/j.issn.1673-4254.2017.11.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To observe the effects of deguelin on the proliferation of breast cancer MCF-7 cells and lung cancer H1299 cells in vitro and the expression of minichromosome maintenance protein 3 (MCM3) and CDC45 in the cells. METHODS MTT assay was used to evaluate the proliferation of MCF-7 and H1299 cells exposed to different concentrations of deguelin for 48, 72 or 96 h. The growth of the cells was observed microscopically and the changes of MCM3 and CDC45 expressions in MCF-7 and H1299 cells following deguelin treatment were detected with fluorescence quantitative PCR. RESULTS The proliferation of MCF-7 cells was significantly inhibited by exposure to 0.25, 0.5, 1, 5, 10, 30, and 50 µmol/L deguelin for 48, 72, and 96 h in a concentration- and time-dependent manner. In MCF-7 cells, the IC50 of deguelin at 48, 72, and 96 h was 9, 3, and 2 µmol/L, respectively. Deguelin treatments of H1299 cells at 0.5, 1, 5, 10, 30, 50, and 100 µmol/L also resulted in a concentration- and time-dependent inhibition of the cell growth with an IC50 at 96 h of 2 µmol/L. Optical microscopy of the cells revealed a decreased number of viable cells with obvious cell shrinkage following deguelin treatments. The expression of MCM3 and CDC45 were significantly reduced in the cells after deguelin treatments. CONCLUSION Deguelin can inhibit the proliferation of MCF-7 and H1299 cells in vitro and down-regulate the expression of MCM3 and CDC45 in the cells.
Collapse
Affiliation(s)
- 俣琳 范
- 南方医科大学研究生院,广东 广州 510515Graduate School of Southern Medical University, Guangzhou 510515, China
- 广州军区广州总医院药学部,广东 广州 510010Department of Pharmacy, General Hospital of Guangzhou Military Command, Guangzhou 510010, China
| | - 瑞瑾 刘
- 广州军区广州总医院药学部,广东 广州 510010Department of Pharmacy, General Hospital of Guangzhou Military Command, Guangzhou 510010, China
| | - 晓艳 丁
- 广州军区广州总医院药学部,广东 广州 510010Department of Pharmacy, General Hospital of Guangzhou Military Command, Guangzhou 510010, China
| | - 信一 上官
- 广州军区广州总医院药学部,广东 广州 510010Department of Pharmacy, General Hospital of Guangzhou Military Command, Guangzhou 510010, China
| | - 新荣 吴
- 南方医科大学研究生院,广东 广州 510515Graduate School of Southern Medical University, Guangzhou 510515, China
- 广州军区广州总医院药学部,广东 广州 510010Department of Pharmacy, General Hospital of Guangzhou Military Command, Guangzhou 510010, China
| |
Collapse
|
7
|
Screening of a small, well-curated natural product-based library identifies two rotenoids with potent nematocidal activity against Haemonchus contortus. Vet Parasitol 2017; 244:172-175. [DOI: 10.1016/j.vetpar.2017.07.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/05/2017] [Accepted: 07/06/2017] [Indexed: 12/20/2022]
|
8
|
Preston S, Korhonen PK, Mouchiroud L, Cornaglia M, McGee SL, Young ND, Davis RA, Crawford S, Nowell C, Ansell BRE, Fisher GM, Andrews KT, Chang BCH, Gijs MAM, Sternberg PW, Auwerx J, Baell J, Hofmann A, Jabbar A, Gasser RB. Deguelin exerts potent nematocidal activity
via
the mitochondrial respiratory chain. FASEB J 2017; 31:4515-4532. [DOI: 10.1096/fj.201700288r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 06/12/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Sarah Preston
- Faculty of Veterinary and Agricultural SciencesUniversity of MelbourneParkvilleVictoriaAustralia
- Faculty of Science and TechnologyFederation UniversityBallaratVictoriaAustralia
| | - Pasi K. Korhonen
- Faculty of Veterinary and Agricultural SciencesUniversity of MelbourneParkvilleVictoriaAustralia
| | - Laurent Mouchiroud
- Laboratory of Integrative and Systems PhysiologyÉcole Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Matteo Cornaglia
- Laboratory of MicrosystemsÉcole Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Sean L. McGee
- Metabolic Research UnitMetabolic Reprogramming LaboratorySchool of Medicine, Faculty of Health, Deakin UniversityWaurn PondsVictoriaAustralia
| | - Neil D. Young
- Faculty of Veterinary and Agricultural SciencesUniversity of MelbourneParkvilleVictoriaAustralia
| | - Rohan A. Davis
- Griffith Institute for Drug DiscoveryGriffith UniversityNathanQueenslandAustralia
| | - Simon Crawford
- School of Biosciences, University of MelbourneParkvilleVictoriaAustralia
| | - Cameron Nowell
- Drug Discovery BiologyMonash University Institute of Pharmaceutical SciencesMonash UniversityParkvilleVictoriaAustralia
| | - Brendan R. E. Ansell
- Faculty of Veterinary and Agricultural SciencesUniversity of MelbourneParkvilleVictoriaAustralia
| | - Gillian M. Fisher
- Griffith Institute for Drug DiscoveryGriffith UniversityNathanQueenslandAustralia
| | - Katherine T. Andrews
- Griffith Institute for Drug DiscoveryGriffith UniversityNathanQueenslandAustralia
| | - Bill C. H. Chang
- Faculty of Veterinary and Agricultural SciencesUniversity of MelbourneParkvilleVictoriaAustralia
- Yourgene BioscienceTaipeiTaiwan
| | - Martin A. M. Gijs
- Laboratory of MicrosystemsÉcole Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Paul W. Sternberg
- Division of Biology and Biological EngineeringCalifornia Institute of TechnologyPasadenaCaliforniaUSA
| | - Johan Auwerx
- Laboratory of Integrative and Systems PhysiologyÉcole Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Jonathan Baell
- Medicinal ChemistryMonash University Institute of Pharmaceutical SciencesMonash UniversityParkvilleVictoriaAustralia
| | - Andreas Hofmann
- Faculty of Veterinary and Agricultural SciencesUniversity of MelbourneParkvilleVictoriaAustralia
- Griffith Institute for Drug DiscoveryGriffith UniversityNathanQueenslandAustralia
| | - Abdul Jabbar
- Faculty of Veterinary and Agricultural SciencesUniversity of MelbourneParkvilleVictoriaAustralia
| | - Robin B. Gasser
- Faculty of Veterinary and Agricultural SciencesUniversity of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
9
|
Deguelin and Its Role in Chronic Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 929:363-375. [DOI: 10.1007/978-3-319-41342-6_16] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/21/2023]
|
10
|
Liu YP, Lee JJ, Lai TC, Lee CH, Hsiao YW, Chen PS, Liu WT, Hong CY, Lin SK, Ping Kuo MY, Lu PJ, Hsiao M. Suppressive function of low-dose deguelin on the invasion of oral cancer cells by downregulating tumor necrosis factor alpha-induced nuclear factor-kappa B signaling. Head Neck 2015; 38 Suppl 1:E524-34. [PMID: 25784049 DOI: 10.1002/hed.24034] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Deguelin has both antiproliferation and antimetastasis activities. However, high-dose deguelin elicits many undesired side effects. The purpose of this study was to investigate whether the low-dose deguelin can prevent the metastasis of oral cancer. METHODS The dose effects of deguelin on metastasis of oral cancer cells were analyzed by in vitro invasion assay and an orthotropic xenograft mouse model. The involvement of tumor necrosis factor alpha (TNF-α)-induced nuclear factor-kappa B (NF-κB) signaling was examined by Western blot and reporter assay. RESULTS Low-dose deguelin, which has minimal cytotoxicity, significantly inhibited the invasion and migration of oral cancer cells. These inhibitory effects of low-dose deguelin were mediated by suppressing TNF-α-induced activation of IκB kinase leading to the inhibition of IκB phosphorylation, NF-κB transcriptional activity, and matrix metalloproteinase-2 (MMP2) expression. The low-dose deguelin treatment significantly inhibited tumor growth and invasion without systemic toxicity. CONCLUSION The low-dose deguelin suppressed the invasion and migration of oral cancer by downregulating TNF-α-induced NF-κB signaling. © 2015 Wiley Periodicals, Inc. Head Neck 38: E524-E534, 2016.
Collapse
Affiliation(s)
- Yu-Peng Liu
- Department of Genome Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Jih-Jong Lee
- Department of Veterinary Medicine, College of Bio-Resources and Agriculture, National Taiwan University, Taipei, Taiwan
| | | | - Chien-Hsin Lee
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Ya-Wen Hsiao
- Department of Cardiology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Po-Shen Chen
- Graduate School of Biomedical Sciences, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Wei-Ting Liu
- Institute of Pharmacology, National Cheng-Kung University, Tainan, Taiwan
| | - Chi-Yuan Hong
- Institute of Clinical Dentistry, School of Dentistry, National Taiwan University Hospital, Taipei, Taiwan
| | - Se-Kwan Lin
- Institute of Clinical Dentistry, School of Dentistry, National Taiwan University Hospital, Taipei, Taiwan
| | - Mark-Yen Ping Kuo
- Institute of Clinical Dentistry, School of Dentistry, National Taiwan University Hospital, Taipei, Taiwan
| | - Pei-Jung Lu
- Institute of Clinical Medicine, National Cheng-Kung University, Tainan, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|