1
|
Di Sotto A, Valipour M, Azari A, Di Giacomo S, Irannejad H. Benzoindolizidine Alkaloids Tylophorine and Lycorine and Their Analogues with Antiviral, Anti-Inflammatory, and Anticancer Properties: Promises and Challenges. Biomedicines 2023; 11:2619. [PMID: 37892993 PMCID: PMC10603990 DOI: 10.3390/biomedicines11102619] [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: 08/09/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
Ongoing viral research, essential for public health due to evolving viruses, gains significance owing to emerging viral infections such as the SARS-CoV-2 pandemic. Marine and plant alkaloids show promise as novel potential pharmacological strategies. In this narrative review, we elucidated the potential of tylophorine and lycorine, two naturally occurring plant-derived alkaloids with a shared benzoindolizidine scaffold, as antiviral agents to be potentially harnessed against respiratory viral infections. Possible structure-activity relationships have also been highlighted. The substances and their derivatives were found to be endowed with powerful and broad-spectrum antiviral properties; moreover, they were able to counteract inflammation, which often underpins the complications of viral diseases. At last, their anticancer properties hold promise not only for advancing cancer research but also for mitigating the oncogenic effects of viruses. This evidence suggests that tylophorine and lycorine could effectively counteract the pathogenesis of respiratory viral disease and its harmful effects. Although common issues about the pharmacologic development of natural substances remain to be addressed, the collected evidence highlights a possible interest in tylophorine and lycorine as antiviral and/or adjuvant strategies and encourages future more in-depth pre-clinical and clinical investigations to overcome their drawbacks and harness their power for therapeutic purposes.
Collapse
Affiliation(s)
- Antonella Di Sotto
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy;
| | - Mehdi Valipour
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Aala Azari
- Environment and Health, Department of Public Health and Primary Care, KU Leuven, Herestraat 49, 3000 Leuven, Belgium;
| | - Silvia Di Giacomo
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy;
- Department of Food Safety, Nutrition and Veterinary Public Health, National Institute of Health, 00161 Rome, Italy
| | - Hamid Irannejad
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari 48471-93698, Iran;
| |
Collapse
|
2
|
Chen SR, Li ZQ, Xu J, Ding MY, Shan YM, Cheng YC, Zhang GX, Sun YW, Wang YQ, Wang Y. Celastrol attenuates hepatitis C virus translation and inflammatory response in mice by suppressing heat shock protein 90β. Acta Pharmacol Sin 2023; 44:1637-1648. [PMID: 36882503 PMCID: PMC10374583 DOI: 10.1038/s41401-023-01067-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/18/2023] [Indexed: 03/09/2023] Open
Abstract
Hepatitis C virus (HCV) infection is one of the major factors to trigger a sustained hepatic inflammatory response and hence hepatocellular carcinoma (HCC), but direct-acting-antiviral (DAAs) was not efficient to suppress HCC development. Heat shock protein 90 kDa (HSP90) is highly abundant in different types of cancers, and especially controls protein translation, endoplasmic reticulum stress, and viral replication. In this study we investigated the correlation between the expression levels of HSP90 isoforms and inflammatory response marker NLRP3 in different types of HCC patients as well as the effect of a natural product celastrol in suppression of HCV translation and associated inflammatory response in vivo. We identified that the expression level of HSP90β isoform was correlated with that of NLRP3 in the liver tissues of HCV positive HCC patients (R2 = 0.3867, P < 0.0101), but not in hepatitis B virus-associated HCC or cirrhosis patients. We demonstrated that celastrol (3, 10, 30 μM) dose-dependently suppressed the ATPase activity of both HSP90α and HSP90β, while its anti-HCV activity was dependent on the Ala47 residue in the ATPase pocket of HSP90β. Celastrol (200 nM) halted HCV internal ribosomal entry site (IRES)-mediated translation at the initial step by disrupting the association between HSP90β and 4EBP1. The inhibitory activity of celastrol on HCV RNA-dependent RNA polymerase (RdRp)-triggered inflammatory response also depended on the Ala47 residue of HSP90β. Intravenous injection of adenovirus expressing HCV NS5B (pAde-NS5B) in mice induced severe hepatic inflammatory response characterized by significantly increased infiltration of immune cells and hepatic expression level of Nlrp3, which was dose-dependently ameliorated by pretreatment with celastrol (0.2, 0.5 mg/kg, i.p.). This study reveals a fundamental role of HSP90β in governing HCV IRES-mediated translation as well as hepatic inflammation, and celastrol as a novel inhibitor of HCV translation and associated inflammation by specifically targeting HSP90β, which could be developed as a lead for the treatment of HSP90β positive HCV-associated HCC.
Collapse
Affiliation(s)
- Shao-Ru Chen
- Institute of Chinese Medical Sciences and State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao SAR, China
| | - Zheng-Qing Li
- Institute of Chinese Medical Sciences and State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao SAR, China
| | - Jun Xu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Mo-Yu Ding
- Institute of Chinese Medical Sciences and State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao SAR, China
| | - Ya-Ming Shan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Yung-Chi Cheng
- Department of Pharmacology, Yale University, New Haven, CT, 06510, USA
| | - Gao-Xiao Zhang
- Institute of New Drug Research and Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine, Jinan University College of Pharmacy, Guangzhou, 510632, China
| | - Ye-Wei Sun
- Institute of New Drug Research and Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine, Jinan University College of Pharmacy, Guangzhou, 510632, China
| | - Yu-Qiang Wang
- Institute of New Drug Research and Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine, Jinan University College of Pharmacy, Guangzhou, 510632, China
- Guangzhou Magpie Pharmaceuticals Co., Ltd., Guangzhou International Business Incubator, Guangzhou, 510663, China
| | - Ying Wang
- Institute of Chinese Medical Sciences and State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao SAR, China.
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao SAR, China.
- Minister of Education Science Center for Precision Oncology, University of Macau, Macao SAR, China.
- Minister of Education Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
3
|
Reimche I, Yu H, Ariantari NP, Liu Z, Merkens K, Rotfuß S, Peter K, Jungwirth U, Bauer N, Kiefer F, Neudörfl JM, Schmalz HG, Proksch P, Teusch N. Phenanthroindolizidine Alkaloids Isolated from Tylophora ovata as Potent Inhibitors of Inflammation, Spheroid Growth, and Invasion of Triple-Negative Breast Cancer. Int J Mol Sci 2022; 23:ijms231810319. [PMID: 36142230 PMCID: PMC9499467 DOI: 10.3390/ijms231810319] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
Triple-negative breast cancer (TNBC), representing the most aggressive form of breast cancer with currently no targeted therapy available, is characterized by an inflammatory and hypoxic tumor microenvironment. To date, a broad spectrum of anti-tumor activities has been reported for phenanthroindolizidine alkaloids (PAs), however, their mode of action in TNBC remains elusive. Thus, we investigated six naturally occurring PAs extracted from the plant Tylophora ovata: O-methyltylophorinidine (1) and its five derivatives tylophorinidine (2), tylophoridicine E (3), 2-demethoxytylophorine (4), tylophoridicine D (5), and anhydrodehydrotylophorinidine (6). In comparison to natural (1) and for more-in depth studies, we also utilized a sample of synthetic O-methyltylophorinidine (1s). Our results indicate a remarkably effective blockade of nuclear factor kappa B (NFκB) within 2 h for compounds (1) and (1s) (IC50 = 17.1 ± 2.0 nM and 3.3 ± 0.2 nM) that is different from its effect on cell viability within 24 h (IC50 = 13.6 ± 0.4 nM and 4.2 ± 1 nM). Furthermore, NFκB inhibition data for the additional five analogues indicate a structure–activity relationship (SAR). Mechanistically, NFκB is significantly blocked through the stabilization of its inhibitor protein kappa B alpha (IκBα) under normoxic as well as hypoxic conditions. To better mimic the TNBC microenvironment in vitro, we established a 3D co-culture by combining the human TNBC cell line MDA-MB-231 with primary murine cancer-associated fibroblasts (CAF) and type I collagen. Compound (1) demonstrates superiority against the therapeutic gold standard paclitaxel by diminishing spheroid growth by 40% at 100 nM. The anti-proliferative effect of (1s) is distinct from paclitaxel in that it arrests the cell cycle at the G0/G1 state, thereby mediating a time-dependent delay in cell cycle progression. Furthermore, (1s) inhibited invasion of TNBC monoculture spheroids into a matrigel®-based environment at 10 nM. In conclusion, PAs serve as promising agents with presumably multiple target sites to combat inflammatory and hypoxia-driven cancer, such as TNBC, with a different mode of action than the currently applied chemotherapeutic drugs.
Collapse
Affiliation(s)
- Irene Reimche
- Department of Biomedical Sciences, Institute of Health Research and Education, University of Osnabrück, 49090 Osnabrück, Germany
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Haiqian Yu
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Ni Putu Ariantari
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich Heine University, 40225 Düsseldorf, Germany
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, Udayana University, Bali 80361, Indonesia
| | - Zhen Liu
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Kay Merkens
- Department of Chemistry, University of Cologne, 50923 Cologne, Germany
| | - Stella Rotfuß
- Department of Biomedical Sciences, Institute of Health Research and Education, University of Osnabrück, 49090 Osnabrück, Germany
| | - Karin Peter
- Department of Biomedical Sciences, Institute of Health Research and Education, University of Osnabrück, 49090 Osnabrück, Germany
| | - Ute Jungwirth
- Department of Life Sciences, Centre for Therapeutic Innovation, University of Bath, Bath BA2 7AY, UK
| | - Nadine Bauer
- European Institute of Molecular Imaging, University of Münster, 48149 Münster, Germany
| | - Friedemann Kiefer
- European Institute of Molecular Imaging, University of Münster, 48149 Münster, Germany
- Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | | | | | - Peter Proksch
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Nicole Teusch
- Department of Biomedical Sciences, Institute of Health Research and Education, University of Osnabrück, 49090 Osnabrück, Germany
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich Heine University, 40225 Düsseldorf, Germany
- Correspondence: ; Tel.: +49-211-81-14163
| |
Collapse
|
4
|
Mang Z, Zhang S, Bai J, Li M, Li H. Design, Synthesis and in Vitro Evaluation of Tylophorine Derivatives as Possible Antitumor Agents. Chem Biodivers 2020; 17:e2000066. [PMID: 32567792 DOI: 10.1002/cbdv.202000066] [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: 01/29/2020] [Accepted: 06/19/2020] [Indexed: 11/07/2022]
Abstract
Structural simplification and modification of natural products are always very important resources to antitumor drugs. By introducing various aminomethyl groups and amide groups into the phenanthrene ring of tylophorine, a novel series of tylophorine derivatives have been designed and synthesized, and their antiproliferative activities against MCF-7, A549 and HepG-2 cells have been evaluated, too. The results indicated that most of the prepared compounds exhibited good antitumor activities. Especially, one compound with an {ethyl[2-(morpholin-4-yl)ethyl]amino}methyl group at the side chain exhibited the most significant cytotoxic effects.
Collapse
Affiliation(s)
- Zhiguo Mang
- State Key Laboratory of Bioengineering Reactor, Shanghai Key Laboratory of New Drug Design, and School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, P. R. China
| | - Shuai Zhang
- State Key Laboratory of Bioengineering Reactor, Shanghai Key Laboratory of New Drug Design, and School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, P. R. China
| | - Jing Bai
- State Key Laboratory of Bioengineering Reactor, Shanghai Key Laboratory of New Drug Design, and School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, P. R. China.,School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Dongqing South Road, Guizhou, 550025, P. R. China
| | - Meijuan Li
- State Key Laboratory of Bioengineering Reactor, Shanghai Key Laboratory of New Drug Design, and School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, P. R. China
| | - Hao Li
- State Key Laboratory of Bioengineering Reactor, Shanghai Key Laboratory of New Drug Design, and School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, P. R. China
| |
Collapse
|
5
|
Renner J, Thakur A, Rutz PM, Cowley JM, Evangelista JL, Kumar P, Prater MB, Stolley RM, Louie J. Total Synthesis of Indolizidine Alkaloids via Nickel-Catalyzed (4 + 2) Cyclization. Org Lett 2020; 22:924-928. [DOI: 10.1021/acs.orglett.9b04479] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Jonas Renner
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-8450, United States
| | - Ashish Thakur
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-8450, United States
| | - Philipp M. Rutz
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-8450, United States
| | - Jacob M. Cowley
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-8450, United States
| | - Judah L. Evangelista
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-8450, United States
| | - Puneet Kumar
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-8450, United States
| | - Matthew B. Prater
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-8450, United States
| | - Ryan M. Stolley
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-8450, United States
| | - Janis Louie
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-8450, United States
| |
Collapse
|
6
|
Bach DH, Lee SK. The Potential Impacts of Tylophora Alkaloids and their Derivatives in Modulating Inflammation, Viral Infections, and Cancer. Curr Med Chem 2019; 26:4709-4725. [PMID: 30047325 DOI: 10.2174/0929867325666180726123339] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/16/2018] [Accepted: 05/24/2018] [Indexed: 12/12/2022]
Abstract
Cancer chemotherapies or antitumor agents mainly remain the backbone of current treatment based on killing the rapidly dividing cancer cell such as tylophora alkaloids and their analogues which have also demonstrated anticancer potential through diverse biological pathways including regulation of the immune system. The introduction of durable clinically effective monoclonal antibodies, however, unmasked a new era of cancer immunotherapies. Therefore, the understanding of cancer pathogenesis will provide new possible treatment options, including cancer immunotherapy and targeted agents. Combining cytotoxic agents and immunotherapies may offer several unique advantages that are complementary to and potentially synergistic with biologic modalities. Herein, we highlight the dynamic mechanism of action of immune modulation in cancer and the immunological aspects of the orally active antitumor agents tylophora alkaloids and their analogues. We also suggest that future cancer treatments will rely on the development of combining tumor-targeted agents and biologic immunotherapies.
Collapse
Affiliation(s)
- Duc-Hiep Bach
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul, Korea
| | - Sang Kook Lee
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
7
|
Joa H, Blažević T, Grojer C, Zeller I, Heiss EH, Atanasov AG, Feldler I, Gruzdaitis P, Czaloun C, Proksch P, Messner B, Bernhard D, Dirsch VM. Tylophorine reduces protein biosynthesis and rapidly decreases cyclin D1, inhibiting vascular smooth muscle cell proliferation in vitro and in organ culture. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 60:152938. [PMID: 31078367 DOI: 10.1016/j.phymed.2019.152938] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/19/2019] [Accepted: 04/22/2019] [Indexed: 05/13/2023]
Abstract
BACKGROUND Tylophorine (TYL) is an alkaloid with antiproliferative action in cancer cells. Vascular smooth muscle cell (VSMC) proliferation and neointima formation contribute to restenosis after percutaneous coronary interventions. HYPOTHESIS/PURPOSE Our goal was to examine the potential of TYL to inhibit VSMC proliferation and migration, and to dissect underlying signaling pathways. STUDY DESIGN AND METHODS TYL was administered to platelet-derived growth factor (PDGF-BB)-stimulated, serum-stimulated, quiescent and unsynchronized VSMC of rat and human origin. BrdU incorporation and resazurin conversion were used to assess cell proliferation. Cell cycle progression was analyzed by flow cytometry of propidium iodide-stained nuclei. Expression profiles of proteins and mRNAs were determined using western blot analysis and RT-qPCR. The Click-iT OPP Alexa Fluor 488 assay was used to monitor protein biosynthesis. RESULTS TYL inhibited PDGF-BB-induced proliferation of rat aortic VSMCs by arresting cells in G1 phase of the cell cycle with an IC50 of 0.13 µmol/l. The lack of retinoblastoma protein phosphorylation and cyclin D1 downregulation corroborated a G1 arrest. Inhibition of proliferation and cyclin D1 downregulation were species- and stimulus-independent. TYL also decreased levels of p21 and p27 proteins, although at later time points than observed for cyclin D1. Co-treatment of VSMC with TYL and MG132 or cycloheximide (CHX) excluded proteasome activation by TYL as the mechanism of action. Comparable time-dependent downregulation of cyclin D1, p21 and p27 in TYL- or CHX-treated cells, together with decreased protein synthesis observed in the Click-iT assay, suggests that TYL is a protein synthesis inhibitor. Besides proliferation, TYL also suppressed migration of PDGF-activated VSMC. In a human saphenous vein organ culture model for graft disease, TYL potently inhibited intimal hyperplasia. CONCLUSION This unique activity profile renders TYL an interesting lead for the treatment of vasculo-proliferative disorders, such as restenosis.
Collapse
Affiliation(s)
- Helge Joa
- Department of Pharmacognosy, University of Vienna, Althanstraße 14, Vienna 1090, Austria; vasopharm GmbH, Friedrich-Bergius-Ring 15, 97076 Würzburg, Germany
| | - Tina Blažević
- Department of Pharmacognosy, University of Vienna, Althanstraße 14, Vienna 1090, Austria.
| | - Christoph Grojer
- Department of Pharmacognosy, University of Vienna, Althanstraße 14, Vienna 1090, Austria; Pfizer Corp. Austria GmbH, Floridsdorfer Hauptstraße 1, 1210 Wien, Austria
| | - Iris Zeller
- Cardiac Surgery Research Laboratory, Department of Surgery, Medical University of Vienna, Währinger Gürtel 18-20, Vienna 1090, Austria; Division of Clinical Microbiology, Department of Laboratory Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Elke H Heiss
- Department of Pharmacognosy, University of Vienna, Althanstraße 14, Vienna 1090, Austria
| | - Atanas G Atanasov
- Department of Pharmacognosy, University of Vienna, Althanstraße 14, Vienna 1090, Austria; Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, 05-552, Jastrzebiec, Poland
| | - Ines Feldler
- Department of Pharmacognosy, University of Vienna, Althanstraße 14, Vienna 1090, Austria
| | - Päivi Gruzdaitis
- Department of Pharmacognosy, University of Vienna, Althanstraße 14, Vienna 1090, Austria; Pharmaceutical Information Centre Ltd., Korkeavuorenkatu 35, 00130 Helsinki, Finland
| | - Christa Czaloun
- Department of Pharmacognosy, University of Vienna, Althanstraße 14, Vienna 1090, Austria; Auge Gottes Apotheke, Nussdorfer Straße 79, 1090 Wien, Austria
| | - Peter Proksch
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich-Heine University, Universitätsstraße 1, Düsseldorf 40225, Germany
| | - Barbara Messner
- Cardiac Surgery Research Laboratory, Department of Surgery, Medical University of Vienna, Währinger Gürtel 18-20, Vienna 1090, Austria
| | - David Bernhard
- Cardiac Surgery Research Laboratory, Department of Surgery, Medical University of Vienna, Währinger Gürtel 18-20, Vienna 1090, Austria; Center for Medical Research, Johannes Kepler University Linz, Krankenhausstr. 7a, 4020 Linz, Austria
| | - Verena M Dirsch
- Department of Pharmacognosy, University of Vienna, Althanstraße 14, Vienna 1090, Austria
| |
Collapse
|
8
|
Tetracenomycin X Exerts Antitumour Activity in Lung Cancer Cells through the Downregulation of Cyclin D1. Mar Drugs 2019; 17:md17010063. [PMID: 30669360 PMCID: PMC6357012 DOI: 10.3390/md17010063] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 01/10/2019] [Accepted: 01/15/2019] [Indexed: 12/19/2022] Open
Abstract
Tetracenomycin X (Tcm X) has been reported to have antitumour activity in various cancers, but there have not been any studies on its activity with respect to lung cancer to date. Therefore, this study aims to investigate the anti-lung cancer activity of Tcm X. In this study, we found that tetracenomycin X showed antitumour activity in vivo and selectively inhibited the proliferation of lung cancer cells without influencing lung fibroblasts. In addition, apoptosis and autophagy did not contribute to the antitumour activity. Tetracenomycin X exerts antitumour activity through cell cycle arrest induced by the downregulation of cyclin D1. To explore the specific mechanism, we found that tetracenomycin X directly induced cyclin D1 proteasomal degradation and indirectly downregulated cyclin D1 via the activation of p38 and c-JUN proteins. All these findings were explored for the first time, which indicated that tetracenomycin X may be a powerful antimitotic class of anticancer drug candidates for the treatment of lung cancer in the future.
Collapse
|
9
|
Endo R, Takashima N, Nekooki-Machida Y, Komi Y, Hui KKW, Takao M, Akatsu H, Murayama S, Sawa A, Tanaka M. TAR DNA-Binding Protein 43 and Disrupted in Schizophrenia 1 Coaggregation Disrupts Dendritic Local Translation and Mental Function in Frontotemporal Lobar Degeneration. Biol Psychiatry 2018; 84:509-521. [PMID: 29752072 PMCID: PMC6123275 DOI: 10.1016/j.biopsych.2018.03.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 02/06/2018] [Accepted: 03/07/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND Neurodegenerative diseases involving protein aggregation often accompany psychiatric symptoms. Frontotemporal lobar degeneration (FTLD) associated with TAR DNA-binding protein 43 (TDP-43) aggregation is characterized by progressive neuronal atrophy in frontal and temporal lobes of cerebral cortex. Furthermore, patients with FTLD display mental dysfunction in multiple behavioral dimensions. Nevertheless, their molecular origin for psychiatric symptoms remains unclear. METHODS In FTLD neurons and mouse models with TDP-43 aggregates, we examined coaggregation between TDP-43 and disrupted in schizophrenia 1 (DISC1), a key player in the pathology of mental conditions and its effects on local translation in dendrites and psychiatric behaviors. The protein coaggregation and the expression level of synaptic proteins were also investigated with postmortem brains from patients with FTLD (n = 6). RESULTS We found cytosolic TDP-43/DISC1 coaggregates in brains of both FTLD mouse model and patients with FTLD. At the mechanistic levels, the TDP-43/DISC1 coaggregates disrupted the activity-dependent dendritic local translation through impairment of translation initiation and, in turn, reduced synaptic protein expression. Behavioral deficits detected in FTLD model mice were ameliorated by exogenous DISC1 expression. CONCLUSIONS Our findings reveal a novel role of the aggregate-prone TDP-43/DISC1 protein complex in regulating local translation, which affects aberrant behaviors relevant to multiple psychiatric dimensions.
Collapse
Affiliation(s)
- Ryo Endo
- Laboratory for Protein Conformation Diseases, RIKEN Brain Science Institute, Japan
| | - Noriko Takashima
- Laboratory for Protein Conformation Diseases, RIKEN Brain Science Institute, Japan
| | - Yoko Nekooki-Machida
- Laboratory for Protein Conformation Diseases, RIKEN Brain Science Institute, Japan
| | - Yusuke Komi
- Laboratory for Protein Conformation Diseases, RIKEN Brain Science Institute, Japan
| | - Kelvin Kai-Wan Hui
- Laboratory for Protein Conformation Diseases, RIKEN Brain Science Institute, Japan
| | - Masaki Takao
- Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Japan,Department of Neurology, Saitama Medical University, Japan
| | - Hiroyasu Akatsu
- Choju Medical Institute, Fukushimura Hospital, Japan,Department of Medicine for Aging in Place and Community-Based Medical Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Shigeo Murayama
- Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Japan
| | - Akira Sawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD.
| | - Motomasa Tanaka
- Laboratory for Protein Conformation Diseases, RIKEN Brain Science Institute, Wako, Japan.
| |
Collapse
|
10
|
Wang Y, Lee S, Ha Y, Lam W, Chen SR, Dutschman GE, Gullen EA, Grill SP, Cheng Y, Fürstner A, Francis S, Baker DC, Yang X, Lee KH, Cheng YC. Tylophorine Analogs Allosterically Regulates Heat Shock Cognate Protein 70 And Inhibits Hepatitis C Virus Replication. Sci Rep 2017; 7:10037. [PMID: 28855547 PMCID: PMC5577180 DOI: 10.1038/s41598-017-08815-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 07/19/2017] [Indexed: 12/12/2022] Open
Abstract
Tylophorine analogs have been shown to exhibit diverse activities against cancer, inflammation, arthritis, and lupus in vivo. In this study, we demonstrated that two tylophorine analogs, DCB-3503 and rac-cryptopleurine, exhibit potent inhibitory activity against hepatitis C virus (HCV) replication in genotype 1b Con 1 isolate. The inhibition of HCV replication is at least partially mediated through cellular heat shock cognate protein 70 (Hsc70). Hsc70 associates with the HCV replication complex by primarily binding to the poly U/UC motifs in HCV RNA. The interaction of DCB-3503 and rac-cryptopleurine with Hsc70 promotes the ATP hydrolysis activity of Hsc70 in the presence of the 3' poly U/UC motif of HCV RNA. Regulating the ATPase activity of Hsc70 may be one of the mechanisms by which tylophorine analogs inhibit HCV replication. This study demonstrates the novel anti-HCV activity of tylophorine analogs. Our results also highlight the importance of Hsc70 in HCV replication.
Collapse
Affiliation(s)
- Ying Wang
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA. .,Institute of Chinese Medical Sciences and State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau, SAR, China.
| | - Sangwon Lee
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Ya Ha
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Wing Lam
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Shao-Ru Chen
- Institute of Chinese Medical Sciences and State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau, SAR, China
| | - Ginger E Dutschman
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Elizabeth A Gullen
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Susan P Grill
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Yao Cheng
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Alois Fürstner
- Max-Planck-Institut für Kohlenforschung, 45470, Mülheim/Ruhr, Germany
| | - Samson Francis
- Department of Chemistry, The University of Tennessee, Knoxville, TN, 37996, USA
| | - David C Baker
- Department of Chemistry, The University of Tennessee, Knoxville, TN, 37996, USA
| | - Xiaoming Yang
- Natural Products Research Laboratories, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Kuo-Hsiung Lee
- Natural Products Research Laboratories, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA.,Chinese Medicine Research and Development Center, China Medical University and Hospital, Taichung, Taiwan
| | - Yung-Chi Cheng
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
11
|
Tylophorine Analog DCB-3503 Inhibited Cyclin D1 Translation through Allosteric Regulation of Heat Shock Cognate Protein 70. Sci Rep 2016; 6:32832. [PMID: 27596272 PMCID: PMC5011780 DOI: 10.1038/srep32832] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 08/15/2016] [Indexed: 02/06/2023] Open
Abstract
Tylophorine analog DCB-3503 is a potential anticancer and immunosuppressive agent that suppresses the translation of cellular regulatory proteins, including cyclin D1, at the elongation step. However, the molecular mechanism underlying this phenomenon remains unknown. This study demonstrates that DCB-3503 preferentially binds to heat shock cognate protein 70 (HSC70), which is a determinant for cyclin D1 translation by binding to the 3′-untranslated region (3′ UTR) of its mRNA. DCB-3503 allosterically regulates the ATPase and chaperone activities of HSC70 by promoting ATP hydrolysis in the presence of specific RNA binding motifs (AUUUA) of cyclin D1 mRNA. The suppression of cyclin D1 translation by DCB-3503 is not solely caused by perturbation of the homeostasis of microRNAs, although the microRNA processing complex is dissociated with DCB-3503 treatment. This study highlights a novel regulatory mechanism of protein translation with AUUUA motifs in the 3′ UTR of mRNA by HSC70, and its activity can be allosterically modulated by DCB-3503. DCB-3503 may be used to treat malignancies, such as hepatocellular carcinoma or breast cancer with elevated expression of cyclin D1.
Collapse
|
12
|
Wen T, Wang Z, Meng X, Wu M, Li Y, Wu X, Zhao L, Wang P, Yin Z, Li-Ling J, Wang Q. Synthesis of novel tylophorine derivatives and evaluation of their anti-inflammatory activity. ACS Med Chem Lett 2014; 5:1027-31. [PMID: 25221661 DOI: 10.1021/ml500255j] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 07/23/2014] [Indexed: 12/23/2022] Open
Abstract
We have previously demonstrated that DCB-3503, a tylophorine analogue, has an anti-inflammatory property in murine models for autoimmune diseases. However, its mechanism remains unknown. Here, we have synthesized 34 derivatives of DCB-3503 and investigated their effects on T cells differentiation and TNF-α production. Six derivatives (4, 9, 13, 19, 31, and 32) could significantly promote the expression of Foxp3. Among these, the IC50 of 31 and 32 was about 500 μM. Eight analogues (1, 2, 4, 9, 12, 18, 19, and 21) showed anti-TNF-α effect in Raw 264.7 cells and murine splenocytes, of which 18 and 19 were most significant. Moreover, 31 and 18 showed a better activity and cell survival ratio when compared with DCB-3503 at various concentrations. In summary, we have demonstrated the anti-inflammatory characteristics of 34 novel tylophorine derivatives and discussed their structure-activity relationship in order to explore their therapeutic potentials for inflammatory diseases.
Collapse
Affiliation(s)
- Ti Wen
- Department
of Medical Oncology, First Hospital of China Medical University, Shengyang 110001, China
| | - Ziwen Wang
- State
Key Laboratory of Elemento-Organic Chemistry, Research Institute of
Elemento-Organic Chemistry, Nankai University, Tianjin 300071, China
- Tianjin
Key Laboratory of Structure and Performance for Functional Molecules,
Key Laboratory of Inorganic−Organic Hybrid Functional Material
Chemistry, Ministry of Education, College of Chemistry, Tianjin Normal University, Tianjin 300387, China
| | - Xianyi Meng
- College
of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Meng Wu
- State
Key Laboratory of Elemento-Organic Chemistry, Research Institute of
Elemento-Organic Chemistry, Nankai University, Tianjin 300071, China
| | - Yangguang Li
- College
of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Xiaoli Wu
- College
of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Liqing Zhao
- College
of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Puyue Wang
- College
of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Zhinan Yin
- College
of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Jesse Li-Ling
- Joint
Key Laboratory for Bio-Resource Research and Utilization of Sichuan
and Chongqing, Institute of Medical Genetics, School of Life Science, Sichuan University, Chengdu 610041, China
| | - Qingmin Wang
- State
Key Laboratory of Elemento-Organic Chemistry, Research Institute of
Elemento-Organic Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
13
|
Miguélez J, Boto A, Marín R, Díaz M. Simplification of antitumoral phenanthroindolizidine alkaloids: Short synthesis of cytotoxic indolizidinone and pyrrolidine analogs. Eur J Med Chem 2013; 66:540-54. [DOI: 10.1016/j.ejmech.2013.06.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 06/03/2013] [Accepted: 06/06/2013] [Indexed: 12/22/2022]
|
14
|
Lai CY, Pan SL, Yang XM, Chang LH, Chang YL, Yang PC, Lee KH, Teng CM. Depletion of 4E-BP1 and regulation of autophagy lead to YXM110-induced anticancer effects. Carcinogenesis 2013; 34:2050-60. [PMID: 23633518 DOI: 10.1093/carcin/bgt146] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Natural products have always been a profuse database for developing new chemotherapeutics. YXM110 is a newly synthesized phenanthroquinolizidines that exhibits excellent anticancer activity in numerous cancer cells. In this study, we examined the anticancer mechanisms of YXM110 both in vitro and in vivo. Protein level of 4E-binding protein 1, which is crucial in cap-independent translation, was decreased significantly after YXM110 treatment via c-Jun N-terminal kinases-mediated proteasomal degradation. Moreover, the effects of YXM110 were associated with several characteristics of autophagy, including accumulation of autophagic vacuoles, elevation of Atg12-Atg5 and light chain 3 (LC3)-II, and levels of GFP-LC3 puncta. The results suggested that depletion of Mcl-1 contributes to YXM110-triggered autophagy, whereas downregulation of lysosomal-related genes could cause autophagy impairment. Furthermore, YXM110-induced cell death was prevented by autophagy inhibitor 3-methyladenine and Atg5 silencing, indicating that YXM110-mediated autophagy impairment leads to cancer cell death. These data reveal key mechanisms that support the further development of YXM110 as a promising anticancer agent.
Collapse
Affiliation(s)
- Chin-Yu Lai
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
15
|
The molecular mechanism and potential role of heat shock-induced p53 protein accumulation. Mol Cell Biochem 2013; 378:161-9. [PMID: 23456460 DOI: 10.1007/s11010-013-1607-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Accepted: 02/23/2013] [Indexed: 02/08/2023]
Abstract
Workers who are exposed to extreme heat or work in hot environments may be at risk of heat stress. Exposure to extreme heat can result in occupational illnesses and injuries. On the other hand, local and regional heat therapy has been used for the treatment of some cancers, such as liver cancer, lung cancer, and kidney cancer. Although heat stress has been shown to induce the accumulation of p53 protein, a key regulator of cell cycle, apoptosis, DNA repair, and autophagy, how it regulates p53 protein accumulation and what the p53 targets are remain unclear. Here, we show that, among various genotoxic stresses, including ionizing radiation (IR) and ultraviolet (UV) radiation, heat stress contributes significantly to increase p53 protein levels in normal liver cells and liver cancer cells. Heat stress did not increase p53 mRNA expression as well as p53 promoter activity. However, heat stress enhanced the half-life of p53 protein. Moreover, heat stress increased the expression of puma and light chain 3 (LC-3), which are associated with the apoptotic and autophagic function of p53, respectively, whereas it did not change the expression of the cell cycle regulators p21, 14-3-3δ, and GADD45α, suggesting that heat-triggered alteration of p53 selectively modulates the downstream targets of p53. Our study provides a novel mechanism by which heat shock stimulates p53 protein accumulation, which is different from common DNA damages, such as IR and UV, and also provides new molecular basis for heat injuries or heat therapy.
Collapse
|
16
|
Wang Y, Wong HC, Gullen EA, Lam W, Yang X, Shi Q, Lee KH, Cheng YC. Cryptopleurine analogs with modification of e ring exhibit different mechanism to rac-cryptopleurine and tylophorine. PLoS One 2012; 7:e51138. [PMID: 23251437 PMCID: PMC3519526 DOI: 10.1371/journal.pone.0051138] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 10/29/2012] [Indexed: 01/01/2023] Open
Abstract
Tylophorine analogs exhibit a broad range of pharmacological activities, including anti-cancer, anti-inflammatory, anti-autoimmune, and anti-virus effects. Structure-activity relationship study of different structure tylophorine analogs can provide further understanding of their biological activity. Modifications on the E ring of the quinolizidine moiety of cryptopleurine analogs changed the potency and the selective inhibitory effect on NF-κB, AP-1, and CRE signaling pathways. Functional cryptopleurine analogs showed potent inhibition of NF-κB signaling pathway in both HepG2 and HEK-293 cell lines. The E ring structure analogs also differed in suppression of protein translation, and expression of cyclin D1. Our results showed that DCB-3503 or Rac-cryptopleurine could be a scaffold for modification to yield compounds with different mechanisms of action.
Collapse
Affiliation(s)
- Ying Wang
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Hui-Chyn Wong
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Elizabeth A. Gullen
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Wing Lam
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Xiaoming Yang
- Natural Products Research Laboratories, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Qian Shi
- Natural Products Research Laboratories, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Kuo-Hsiung Lee
- Natural Products Research Laboratories, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Chinese Medicine Research and Development Center, China Medical University and Hospital, Taichung, Taiwan
- * E-mail: (YCC); (KHL)
| | - Yung-Chi Cheng
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, United States of America
- * E-mail: (YCC); (KHL)
| |
Collapse
|
17
|
Niphakis MJ, Gay BC, Hong KH, Bleeker NP, Georg GI. Synthesis and evaluation of the anti-proliferative and NF-κB activities of a library of simplified tylophorine analogs. Bioorg Med Chem 2012; 20:5893-900. [DOI: 10.1016/j.bmc.2012.07.044] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2012] [Revised: 07/14/2012] [Accepted: 07/24/2012] [Indexed: 01/20/2023]
|
18
|
Wen T, Li Y, Wu M, Chen X, Han L, Bao X, Wang Z, Wang K, Hu Y, Zhou X, Wu Z, Wang P, Hong Z, Zhao L, Wang Q, Yin Z. A novel tylophorine analog NK-007 ameliorates colitis through inhibition of innate immune response. Int Immunopharmacol 2012; 14:487-94. [PMID: 22929538 DOI: 10.1016/j.intimp.2012.08.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 07/09/2012] [Accepted: 08/13/2012] [Indexed: 12/21/2022]
Abstract
In this study, we synthesized (±)-tylophorine malate (NK-007), an analog of tylophorine (DCB3503), and analyzed its anti-inflammatory effect in vivo using a dextran sulfate sodium (DSS)-induced colitis model and an acetic acid-induced colitis model. As indicated by disease activity index (DAI) and degree of macroscopic colonic damage, NK-007 can significantly suppress colitis. To delineate the underlying mechanism, we have explored the influence of NK-007 on the production of TNF-α by murine primary bone marrow-derived dendritic cells (BMDCs) as well as monocyte/macrophage cell line Raw 264.7 triggered by lipopolysaccharide (LPS). For both types of innate immune cells, NK-007 showed a potent TNF-α inhibitory effect, and has in addition reduced the expression of IL-12 in BMDCs. Moreover, Raw cells treated with NK-007 also showed decreased phosphorylation of NF-κB, which may explain the protective immune-regulatory effect of NK-007 for experimental colitis.
Collapse
Affiliation(s)
- Ti Wen
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Yang X, Shi Q, Yang SC, Chen CY, Yu SL, Bastow KF, Morris-Natschke SL, Wu PC, Lai CY, Wu TS, Pan SL, Teng CM, Lin JC, Yang PC, Lee KH. Antitumor agents 288: design, synthesis, SAR, and biological studies of novel heteroatom-incorporated antofine and cryptopleurine analogues as potent and selective antitumor agents. J Med Chem 2011; 54:5097-107. [PMID: 21668000 DOI: 10.1021/jm200330s] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Novel heteroatom-incorporated antofine and cryptopleurine analogues were designed, synthesized, and tested against a panel of five cancer cell lines. Two new S-13-oxo analogues (11 and 16) exhibited potent cell growth inhibition in vitro (GI(50): 9 nM and 20 nM). Interestingly, both compounds displayed improved selectivity among different cancer cell lines, in contrast to the natural products antofine and cryptopleurine. Mechanism of action (MOA) studies suggested that R-antofine promotes dysregulation of DNA replication during early S phase, while no similar effects were observed for 11 and 15 on corresponding replication initiation complexes. Compound 11 also showed greatly reduced cytotoxicity against normal cells and moderate antitumor activity against HT-29 human colorectal adenocarcinoma xenograft in mice without overt toxicity.
Collapse
Affiliation(s)
- Xiaoming Yang
- Natural Products Research Laboratories, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599-7568, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
|