1
|
Jiang Y, Tang Y, Li Y, Liu L, Yue K, Li X, Qiu P, Yin R, Jiang T. Psammaplin A analogues with modified disulfide bond targeting histone deacetylases: Synthesis and biological evaluation. Eur J Med Chem 2024; 275:116541. [PMID: 38851056 DOI: 10.1016/j.ejmech.2024.116541] [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: 03/21/2024] [Revised: 05/02/2024] [Accepted: 05/26/2024] [Indexed: 06/10/2024]
Abstract
Psammaplin A (PsA), a symmetrical bromotyrosine-derived disulfide marine metabolite, has been reported could inhibit HDAC1/2/3 through its thiol monomer. Inspired by the disuflide bond structure of this marine natural product, we designed and synthesized a series of PsA analogues, in which the disulfide bond of PsA was replaced with diselenide bond or cyclic disulfide/diselenide/selenenylsulfide motifs. We also studied the HDAC inhibition, cell growth inhibition, and apoptosis induction of these PsA analogues. The results showed that, all the synthetic diselenide analogues and cyclic selenenyl sulfide compounds exhibited better antiproferative activity than their counterpart of disulfide analogues. Among the prepared analogues, diselenide analogue P-503 and P-116 significantly increased the ability of inhibiting HDAC6 and induced apoptosis and G2/M cell cycle arrest. However, cyclic selenenylsulfides analogues P-111 lost its HDAC inhibitory ability and exhibited no effect on cell cycle and apoptosis, indicating that the anti-proliferative mechanism of cyclic selenenylsulfides analogues has changed.
Collapse
Affiliation(s)
- Yukun Jiang
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Ya Tang
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Yuxuan Li
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Lu Liu
- Marine Biomedical Research Institute of Qingdao, Qingdao, 266237, China
| | - Kairui Yue
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Xiaoyang Li
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Peiju Qiu
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Marine Biomedical Research Institute of Qingdao, Qingdao, 266237, China.
| | - Ruijuan Yin
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Marine Biomedical Research Institute of Qingdao, Qingdao, 266237, China.
| | - Tao Jiang
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| |
Collapse
|
2
|
Wahi A, Jain P, Sinhari A, Jadhav HR. Progress in discovery and development of natural inhibitors of histone deacetylases (HDACs) as anti-cancer agents. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:675-702. [PMID: 37615708 DOI: 10.1007/s00210-023-02674-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/12/2023] [Indexed: 08/25/2023]
Abstract
The study of epigenetic translational modifications had drawn great interest for the last few decades. These processes play a vital role in many diseases and cancer is one of them. Histone acetyltransferase (HAT) and histone deacetylases (HDACs) are key enzymes involved in the acetylation and deacetylation of histones and ultimately in post-translational modifications. Cancer frequently exhibits epigenetic changes, particularly disruption in the expression and activity of HDACs. It includes the capacity to regulate proliferative signalling, circumvent growth inhibitors, escape cell death, enable replicative immortality, promote angiogenesis, stimulate invasion and metastasis, prevent immunological destruction, and genomic instability. The majority of tumours develop and spread as a result of HDAC dysregulation. As a result, HDAC inhibitors (HDACis) were developed, and they today stand as a very promising therapeutic approach. One of the most well-known and efficient therapies for practically all cancer types is chemotherapy. However, the efficiency and safety of treatment are constrained by higher toxicity. The same has been observed with the synthetic HDACi. Natural products, owing to many advantages over synthetic compounds for cancer treatment have always been a choice for therapy. Hence, naturally available molecules are of particular interest for HDAC inhibition and HDAC has drawn the attention of the research fraternity due to their potential to offer a diverse array of chemical structures and bioactive compounds. This diversity opens up new avenues for exploring less toxic HDAC inhibitors to reduce side effects associated with conventional synthetic inhibitors. The review presents comprehensive details on natural product HDACi, their mechanism of action and their biological effects. Moreover, this review provides a brief discussion on the structure activity relationship of selected natural HDAC inhibitors and their analogues which can guide future research to discover selective, more potent HDACi with minimal toxicity.
Collapse
Affiliation(s)
- Abhishek Wahi
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, DPSRU, New Delhi, 110017, India
| | - Priti Jain
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, DPSRU, New Delhi, 110017, India.
| | - Apurba Sinhari
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Vidya Vihar, Pilani, Rajasthan, 333031, India
| | - Hemant R Jadhav
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Vidya Vihar, Pilani, Rajasthan, 333031, India
| |
Collapse
|
3
|
Abdelrahman KS, Hassan HA, Abdel-Aziz SA, Marzouk AA, Shams R, Osawa K, Abdel-Aziz M, Konno H. Development and Assessment of 1,5-Diarylpyrazole/Oxime Hybrids Targeting EGFR and JNK-2 as Antiproliferative Agents: A Comprehensive Study through Synthesis, Molecular Docking, and Evaluation. Molecules 2023; 28:6521. [PMID: 37764297 PMCID: PMC10537604 DOI: 10.3390/molecules28186521] [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: 08/25/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
New 1,5-diarylpyrazole oxime hybrid derivatives (scaffolds A and B) were designed, synthesized, and then their purity was verified using a variety of spectroscopic methods. A panel of five cancer cell lines known to express EGFR and JNK-2, including human colorectal adenocarcinoma cell line DLD-1, human cervical cancer cell line Hela, human leukemia cell line K562, human pancreatic cell line SUIT-2, and human hepatocellular carcinoma cell line HepG2, were used to biologically evaluate for their in vitro cytotoxicity for all the synthesized compounds 7a-j, 8a-j, 9a-c, and 10a-c. The oxime containing compounds 8a-j and 10a-c were more active as antiproliferative agents than their non-oxime congeners 7a-j and 9a-c. Compounds 8d, 8g, 8i, and 10c inhibited EGFR with IC50 values ranging from 8 to 21 µM when compared with sorafenib. Compound 8i inhibited JNK-2 as effectively as sorafenib, with an IC50 of 1.0 µM. Furthermore, compound 8g showed cell cycle arrest at the G2/M phase in the cell cycle analysis of the Hela cell line, whereas compound 8i showed combined S phase and G2 phase arrest. According to docking studies, oxime hybrid compounds 8d, 8g, 8i, and 10c exhibited binding free energies ranging from -12.98 to 32.30 kcal/mol at the EGFR binding site whereas compounds 8d and 8i had binding free energies ranging from -9.16 to -12.00 kcal/mol at the JNK-2 binding site.
Collapse
Affiliation(s)
- Kamal S. Abdelrahman
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt; (S.A.A.-A.); (A.A.M.)
| | - Heba A. Hassan
- Department of Medicinal Chemistry Faculty of Pharmacy, Minia University, Minia 61519, Egypt; (H.A.H.); (M.A.-A.)
| | - Salah A. Abdel-Aziz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt; (S.A.A.-A.); (A.A.M.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Deraya University, Minia 61768, Egypt
| | - Adel A. Marzouk
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt; (S.A.A.-A.); (A.A.M.)
- National Center for Natural Products Research, School of Pharmacy, University of Missippi, Oxford, MS 38677, USA
| | - Raef Shams
- Emergent Bioengineering Materials Research Team, RIKEN Centre for Emergent Matter Science, RIKEN, Wako 351-0198, Saitama, Japan;
| | - Keima Osawa
- Graduate School of Science and Engineering, Yamagata University, Yonezawa 992-8510, Yamagata, Japan;
| | - Mohamed Abdel-Aziz
- Department of Medicinal Chemistry Faculty of Pharmacy, Minia University, Minia 61519, Egypt; (H.A.H.); (M.A.-A.)
| | - Hiroyuki Konno
- Graduate School of Science and Engineering, Yamagata University, Yonezawa 992-8510, Yamagata, Japan;
| |
Collapse
|
4
|
Maxfield K, Payne M, Chamberland S. Total Synthesis and Biological Evaluation of Clavatadines C-E. ACS OMEGA 2022; 7:22915-22929. [PMID: 35811874 PMCID: PMC9260760 DOI: 10.1021/acsomega.2c02913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/09/2022] [Indexed: 06/15/2023]
Abstract
We described herein the application of a convergent and protecting-group avoidant approach that led to the first total synthesis of the marine natural products clavatadine D (4) and E (5), and the second total synthesis of clavatadine C (3). In each case, a key amide-coupling afforded an immediate precursor of each natural product in a rapid manner from structurally similar western and eastern portions that derived from an ester of l-tyrosine and butane-1,4-diamine, respectively. A deprotection step free of detectable byproducts cleanly provided the remaining known members of the clavatadine family of natural products. Each total synthesis required five steps (longest linear sequence) with overall yields of 30-37%, 26-39%, and 28-50% for clavatadine C (3), D (4), and E (5), respectively. A screen of their potential anticancer activity against the NCI-60 cell line panel revealed cytotoxicity levels up to 38% across a broad spectrum of tumor types. Although clavatadine C (3) was relatively benign, clavatadine D (4) exhibited 20-38% growth inhibition against a wide array of cancer cell types including leukemia, non-small-cell lung, colon, ovarian, and breast. Clavatadine E (5) was active against two types of human brain tumors.
Collapse
|
5
|
Tian C, Huang S, Xu Z, Liu W, Li D, Liu M, Zhu C, Wu L, Jiang X, Ding H, Zhao Q. Design, synthesis, and biological evaluation of β-carboline 1,3,4-oxadiazole based hybrids as HDAC inhibitors with potential antitumor effects. Bioorg Med Chem Lett 2022; 64:128663. [PMID: 35272009 DOI: 10.1016/j.bmcl.2022.128663] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/28/2022] [Accepted: 03/03/2022] [Indexed: 12/11/2022]
Abstract
A series of novel β-carboline 1,3,4-oxadiazole based hybrids were designed, synthesized, and tested for cytotoxicity and HDAC inhibition. Among the target compounds, compound ZDLT-1 displayed high inhibitory activity for class I HDACs 1, 2, and 3, and potent anti-proliferative activity against HCT116 cells with an IC50 value of 0.173 ± 0.018 μM, it also exhibited better inhibitory activity with an IC50 value of 6 nM for HDAC6 than SAHA (IC50 = 15 nM). Furthermore, the pharmacological experiment of Hoechst staining, colony formation, cell apoptosis assay, and wound healing scratch assay indicated that compound ZDLT-1 was a potent cytotoxic agent against HCT116 cells with cell apoptosis induction. Further, in silico prediction of physicochemical properties, drug-likeness, and ADME parameters suggested that compound ZDLT-1 is a promising anticancer agent. Taken together, the high potency cytotoxicity and class I HDACs inhibitory activity of compound ZDLT-1, which with the β-carboline 1,3,4-oxadiazole based hybrids as potent anticancer agents could be nominated for further modification and optimization.
Collapse
Affiliation(s)
- Caizhi Tian
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, PR China; School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Shuoqi Huang
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, PR China; School of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Zihua Xu
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, PR China
| | - Wenwu Liu
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, PR China; School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Deping Li
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, PR China; School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Mingyue Liu
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, PR China; School of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Chengze Zhu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Limeng Wu
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, PR China; School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xiaowen Jiang
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, PR China; School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Huaiwei Ding
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Qingchun Zhao
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, PR China; School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
6
|
Qiu X, Zhu L, Wang H, Tan Y, Yang Z, Yang L, Wan L. From natural products to HDAC inhibitors: An overview of drug discovery and design strategy. Bioorg Med Chem 2021; 52:116510. [PMID: 34826681 DOI: 10.1016/j.bmc.2021.116510] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 10/09/2021] [Accepted: 10/15/2021] [Indexed: 02/08/2023]
Abstract
Histone deacetylases (HDACs) play a key role in the homeostasis of protein acetylation in histones and have recently emerged as a therapeutic target for numerous diseases. The inhibition of HDACs may block angiogenesis, arrest cell growth, and lead to differentiation and apoptosis in tumour cells. Thus, HDAC inhibitors (HDACi) have received increasing attention and many of which are developed from natural sources. In the past few decades, naturally occurring HDACi have been identified to have potent anticancer activities, some of which have demonstrated promising therapeutic effects on haematological malignancies. In this review, we summarized the discovery and modification of HDAC inhibitors from natural sources, novel drug design that uses natural products as parent nuclei, and dual target design strategies that combine HDAC with non-HDAC targets.
Collapse
Affiliation(s)
- Xiang Qiu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lv Zhu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huan Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yan Tan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhuang Yang
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Linyu Yang
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Li Wan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
7
|
Schepetkin IA, Plotnikov MB, Khlebnikov AI, Plotnikova TM, Quinn MT. Oximes: Novel Therapeutics with Anticancer and Anti-Inflammatory Potential. Biomolecules 2021; 11:biom11060777. [PMID: 34067242 PMCID: PMC8224626 DOI: 10.3390/biom11060777] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023] Open
Abstract
Oximes have been studied for decades because of their significant roles as acetylcholinesterase reactivators. Over the last twenty years, a large number of oximes have been reported with useful pharmaceutical properties, including compounds with antibacterial, anticancer, anti-arthritis, and anti-stroke activities. Many oximes are kinase inhibitors and have been shown to inhibit over 40 different kinases, including AMP-activated protein kinase (AMPK), phosphatidylinositol 3-kinase (PI3K), cyclin-dependent kinase (CDK), serine/threonine kinases glycogen synthase kinase 3 α/β (GSK-3α/β), Aurora A, B-Raf, Chk1, death-associated protein-kinase-related 2 (DRAK2), phosphorylase kinase (PhK), serum and glucocorticoid-regulated kinase (SGK), Janus tyrosine kinase (JAK), and multiple receptor and non-receptor tyrosine kinases. Some oximes are inhibitors of lipoxygenase 5, human neutrophil elastase, and proteinase 3. The oxime group contains two H-bond acceptors (nitrogen and oxygen atoms) and one H-bond donor (OH group), versus only one H-bond acceptor present in carbonyl groups. This feature, together with the high polarity of oxime groups, may lead to a significantly different mode of interaction with receptor binding sites compared to corresponding carbonyl compounds, despite small changes in the total size and shape of the compound. In addition, oximes can generate nitric oxide. This review is focused on oximes as kinase inhibitors with anticancer and anti-inflammatory activities. Oximes with non-kinase targets or mechanisms of anti-inflammatory activity are also discussed.
Collapse
Affiliation(s)
- Igor A. Schepetkin
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA;
| | - Mark B. Plotnikov
- Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, 634028 Tomsk, Russia;
| | - Andrei I. Khlebnikov
- Kizhner Research Center, National Research Tomsk Polytechnic University, 634050 Tomsk, Russia;
- Scientific Research Institute of Biological Medicine, Altai State University, 656049 Barnaul, Russia
| | - Tatiana M. Plotnikova
- Department of Pharmacology, Siberian State Medical University, 634050 Tomsk, Russia;
| | - Mark T. Quinn
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA;
- Correspondence: ; Tel.: +1-406-994-4707; Fax: +1-406-994-4303
| |
Collapse
|
8
|
Ju Han H, Sub Byun W, Ho Lee G, Kyung Kim W, Jang K, Yang S, Yang J, Woo Ha M, Hong S, Lee J, Shin J, Bong Oh K, Kook Lee S, Park HG. Synthesis and biological activity of selenopsammaplin A and its analogues as antitumor agents with DOT1L inhibitory activity. Bioorg Med Chem 2021; 35:116072. [PMID: 33636429 DOI: 10.1016/j.bmc.2021.116072] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 12/17/2022]
Abstract
Disruptor of telomeric silencing-1 like (DOT1L) is a histone H3 methyltransferase which specifically catalyzes the methylation of histone H3 lysine-79 residue. Recent findings demonstrate that DOT1L is abnormally overexpressed and the upregulated DOT1L evokes the proliferation and metastasis in human breast cancer cells. Therefore, the DOT1L inhibitor is considered a promising strategy to treat breast cancers. Non-nucleoside DOT1L inhibitors, selenopsammaplin A and its analogues, were firstly reported in the present study. Selenopsammaplin A was newly designed and synthesized with 25% overall yield in 8 steps from 3-bromo-4-hydroxybenzaldahyde, and thirteen analogues of selenopsammaplin A were prepared for structure-activity relationship studies of their cytotoxicity against cancer cells and inhibitory activity toward DOT1L for antitumor potential. All synthetic selenopsammaplin A analogues exhibited the higher cytotoxicity compared to psammaplin A with up to 6 - 60 times depending on cancer cells, and most analogues showed significant inhibitory activities against DOT1L. Among the prepared analogues, the phenyl analogue (10) possessed the most potent activity with both cytotoxicity and inhibition of DOT1L. Compound 10 also exhibited the antitumor and antimetastatic activity in an orthotopic mouse metastasis model implanted with MDA-MB-231 human breast cancer cells. These biological findings suggest that analogue 10 is a promising candidate for development as a cancer chemotherapeutic agent in breast cancers.
Collapse
Affiliation(s)
- Hae Ju Han
- Research Institute of Pharmaceutical Science and College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea.
| | - Woong Sub Byun
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea.
| | - Gyu Ho Lee
- Research Institute of Pharmaceutical Science and College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea
| | - Won Kyung Kim
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea
| | - Kyungkuk Jang
- Research Institute of Pharmaceutical Science and College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea
| | - Sehun Yang
- Research Institute of Pharmaceutical Science and College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea
| | - Jewon Yang
- Research Institute of Pharmaceutical Science and College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea
| | - Min Woo Ha
- College of Pharmacy, Jeju National University, 102 Jejudaehak-ro, Jeju 63243, Republic of Korea; Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, 102 Jejudaehak-ro, Jeju, 63243, Republic of Korea
| | - Suckchang Hong
- Research Institute of Pharmaceutical Science and College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea
| | - Jeeyeon Lee
- Research Institute of Pharmaceutical Science and College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea
| | - Jongheon Shin
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea
| | - Ki Bong Oh
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul 151-921, Republic of Korea
| | - Sang Kook Lee
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea.
| | - Hyeung-Geun Park
- Research Institute of Pharmaceutical Science and College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea.
| |
Collapse
|
9
|
Design, synthesis and biological evaluation of oxime lacking Psammaplin inspired chemical libraries as anti-cancer agents. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2020.129173] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
10
|
Bao Y, Xu Q, Wang L, Wei Y, Hu B, Wang J, Liu D, Zhao L, Jing Y. Studying Histone Deacetylase Inhibition and Apoptosis Induction of Psammaplin A Monomers with Modified Thiol Group. ACS Med Chem Lett 2021; 12:39-47. [PMID: 33488962 DOI: 10.1021/acsmedchemlett.0c00369] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 11/25/2020] [Indexed: 12/30/2022] Open
Abstract
Psammaplin A (PsA) is a bromotyrosine disulfide dimer with histone deacetylase (HDAC) inhibition and acts through reduced monomer PsA-SH. We studied the connection of HDAC inhibition, cell growth inhibition, and apoptosis induction of PsA-SH by modifying the -SH group with deletion (6a) or replacement with hydroxamic acid (10b) or benzamide (12g). PsA-SH inhibits HDAC1/2/3 and 6a loses the HDAC inhibition ability. 10b inhibits HDAC1/2/3/6 while 12g shows selective inhibition of HDAC3. PsA-SH and 10b, but neither 6a nor 12g, induce apoptosis in human leukemia HL-60 cells associated with increased acetylation of Histone H3. PsA-SH and 10b inhibit growth of several solid tumor cell lines in vitro and Lewis lung cancer cell growth in vivo. PsA-SH is a simple scaffold for developing selective HDAC inhibitors and induces apoptosis through inhibiting HDAC1/2.
Collapse
Affiliation(s)
- Yu Bao
- Department of Pharmacology, Liaoning Key Lab of Targeting Drugs for Hematological Malignancies, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, R. P. China
| | - Qihao Xu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Lin Wang
- Department of Pharmacology, Liaoning Key Lab of Targeting Drugs for Hematological Malignancies, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, R. P. China
| | - Yunfei Wei
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Baichun Hu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Jian Wang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Dan Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Linxiang Zhao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yongkui Jing
- Department of Pharmacology, Liaoning Key Lab of Targeting Drugs for Hematological Malignancies, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, R. P. China
| |
Collapse
|
11
|
Byun WS, Lee GH, Park HG, Lee SK. Inhibition of DOT1L by Half-Selenopsammaplin A Analogs Suppresses Tumor Growth and EMT-Mediated Metastasis in Triple-Negative Breast Cancer. Pharmaceuticals (Basel) 2020; 14:ph14010018. [PMID: 33379275 PMCID: PMC7824081 DOI: 10.3390/ph14010018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/18/2020] [Accepted: 12/24/2020] [Indexed: 12/26/2022] Open
Abstract
Due to a lack of hormone receptors, current treatment strategies for triple-negative breast cancer (TNBC) are limited with frequent disease recurrence and metastasis. Recent findings have suggested that aberrant methylation of histone H3 lysine 79 residue (H3K79me) by the histone methyltransferase disruptor of telomeric silencing 1-like (DOT1L) is a potential therapeutic target for TNBC clinical management. Therefore, we developed DOT1L inhibitors as potential antitumor agents against TNBC cells. We reveal that a synthetic half-selenopsammaplin A analog 9l (subsequently known as 9l) exhibited inhibitory activity against DOT1L-mediated H3K79 methylation, and showed antitumor activity in TNBC cells. The analog 9l also significantly inhibited TNBC invasion and migration via the modulation of epithelial-mesenchymal transition (EMT) markers, including N-cadherin and vimentin downregulation and E-cadherin upregulation. In an MDA-MB-231/Luc-implanted orthotopic mouse metastasis model, treatment with 9l effectively inhibited tumor growth and lung metastasis via DOT1L regulatory activity and EMT processes. Taken together, these findings highlight the potential of 9l as a novel therapeutic candidate for treating metastatic TNBC via DOT1L modulation.
Collapse
Affiliation(s)
- Woong Sub Byun
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Korea;
| | - Gyu Ho Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea;
| | - Hyeung-geun Park
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea;
- Correspondence: (H.-g.P.); (S.K.L.)
| | - Sang Kook Lee
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Korea;
- Correspondence: (H.-g.P.); (S.K.L.)
| |
Collapse
|
12
|
Rieux C, Goffinont S, Coste F, Tber Z, Cros J, Roy V, Guérin M, Gaudon V, Bourg S, Biela A, Aucagne V, Agrofoglio L, Garnier N, Castaing B. Thiopurine Derivative-Induced Fpg/Nei DNA Glycosylase Inhibition: Structural, Dynamic and Functional Insights. Int J Mol Sci 2020; 21:ijms21062058. [PMID: 32192183 PMCID: PMC7139703 DOI: 10.3390/ijms21062058] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/12/2020] [Accepted: 03/14/2020] [Indexed: 02/06/2023] Open
Abstract
DNA glycosylases are emerging as relevant pharmacological targets in inflammation, cancer and neurodegenerative diseases. Consequently, the search for inhibitors of these enzymes has become a very active research field. As a continuation of previous work that showed that 2-thioxanthine (2TX) is an irreversible inhibitor of zinc finger (ZnF)-containing Fpg/Nei DNA glycosylases, we designed and synthesized a mini-library of 2TX-derivatives (TXn) and evaluated their ability to inhibit Fpg/Nei enzymes. Among forty compounds, four TXn were better inhibitors than 2TX for Fpg. Unexpectedly, but very interestingly, two dithiolated derivatives more selectively and efficiently inhibit the zincless finger (ZnLF)-containing enzymes (human and mimivirus Neil1 DNA glycosylases hNeil1 and MvNei1, respectively). By combining chemistry, biochemistry, mass spectrometry, blind and flexible docking and X-ray structure analysis, we localized new TXn binding sites on Fpg/Nei enzymes. This endeavor allowed us to decipher at the atomic level the mode of action for the best TXn inhibitors on the ZnF-containing enzymes. We discovered an original inhibition mechanism for the ZnLF-containing Fpg/Nei DNA glycosylases by disulfide cyclic trimeric forms of dithiopurines. This work paves the way for the design and synthesis of a new structural class of inhibitors for selective pharmacological targeting of hNeil1 in cancer and neurodegenerative diseases.
Collapse
Affiliation(s)
- Charlotte Rieux
- Centre de Biophysique Moléculaire, UPR4301 CNRS, rue Charles Sadron, CEDEX 2, F-45071 Orléans, France; (C.R.); (S.G.); (F.C.); (J.C.); (M.G.); (V.G.); (A.B.); (V.A.)
| | - Stéphane Goffinont
- Centre de Biophysique Moléculaire, UPR4301 CNRS, rue Charles Sadron, CEDEX 2, F-45071 Orléans, France; (C.R.); (S.G.); (F.C.); (J.C.); (M.G.); (V.G.); (A.B.); (V.A.)
| | - Franck Coste
- Centre de Biophysique Moléculaire, UPR4301 CNRS, rue Charles Sadron, CEDEX 2, F-45071 Orléans, France; (C.R.); (S.G.); (F.C.); (J.C.); (M.G.); (V.G.); (A.B.); (V.A.)
| | - Zahira Tber
- Institut de Chimie Organique et Analytique, UMR7311 CNRS-Orleans University, Université d’Orléans, Pôle de Chimie, rue de Chartres, F-45100 Orléans, France; (Z.T.); (S.B.); (L.A.)
| | - Julien Cros
- Centre de Biophysique Moléculaire, UPR4301 CNRS, rue Charles Sadron, CEDEX 2, F-45071 Orléans, France; (C.R.); (S.G.); (F.C.); (J.C.); (M.G.); (V.G.); (A.B.); (V.A.)
| | - Vincent Roy
- Institut de Chimie Organique et Analytique, UMR7311 CNRS-Orleans University, Université d’Orléans, Pôle de Chimie, rue de Chartres, F-45100 Orléans, France; (Z.T.); (S.B.); (L.A.)
- Université d’Orléans, UFR Sciences et Techniques, rue de Chartres, 45100 Orléans, France
- Correspondence: (V.R.); (N.G.); (B.C.)
| | - Martine Guérin
- Centre de Biophysique Moléculaire, UPR4301 CNRS, rue Charles Sadron, CEDEX 2, F-45071 Orléans, France; (C.R.); (S.G.); (F.C.); (J.C.); (M.G.); (V.G.); (A.B.); (V.A.)
- Université d’Orléans, UFR Sciences et Techniques, rue de Chartres, 45100 Orléans, France
| | - Virginie Gaudon
- Centre de Biophysique Moléculaire, UPR4301 CNRS, rue Charles Sadron, CEDEX 2, F-45071 Orléans, France; (C.R.); (S.G.); (F.C.); (J.C.); (M.G.); (V.G.); (A.B.); (V.A.)
| | - Stéphane Bourg
- Institut de Chimie Organique et Analytique, UMR7311 CNRS-Orleans University, Université d’Orléans, Pôle de Chimie, rue de Chartres, F-45100 Orléans, France; (Z.T.); (S.B.); (L.A.)
| | - Artur Biela
- Centre de Biophysique Moléculaire, UPR4301 CNRS, rue Charles Sadron, CEDEX 2, F-45071 Orléans, France; (C.R.); (S.G.); (F.C.); (J.C.); (M.G.); (V.G.); (A.B.); (V.A.)
| | - Vincent Aucagne
- Centre de Biophysique Moléculaire, UPR4301 CNRS, rue Charles Sadron, CEDEX 2, F-45071 Orléans, France; (C.R.); (S.G.); (F.C.); (J.C.); (M.G.); (V.G.); (A.B.); (V.A.)
| | - Luigi Agrofoglio
- Institut de Chimie Organique et Analytique, UMR7311 CNRS-Orleans University, Université d’Orléans, Pôle de Chimie, rue de Chartres, F-45100 Orléans, France; (Z.T.); (S.B.); (L.A.)
- Université d’Orléans, UFR Sciences et Techniques, rue de Chartres, 45100 Orléans, France
| | - Norbert Garnier
- Centre de Biophysique Moléculaire, UPR4301 CNRS, rue Charles Sadron, CEDEX 2, F-45071 Orléans, France; (C.R.); (S.G.); (F.C.); (J.C.); (M.G.); (V.G.); (A.B.); (V.A.)
- Université d’Orléans, UFR Sciences et Techniques, rue de Chartres, 45100 Orléans, France
- Correspondence: (V.R.); (N.G.); (B.C.)
| | - Bertrand Castaing
- Centre de Biophysique Moléculaire, UPR4301 CNRS, rue Charles Sadron, CEDEX 2, F-45071 Orléans, France; (C.R.); (S.G.); (F.C.); (J.C.); (M.G.); (V.G.); (A.B.); (V.A.)
- Correspondence: (V.R.); (N.G.); (B.C.)
| |
Collapse
|
13
|
Byun WS, Kim WK, Han HJ, Chung HJ, Jang K, Kim HS, Kim S, Kim D, Bae ES, Park S, Lee J, Park HG, Lee SK. Targeting Histone Methyltransferase DOT1L by a Novel Psammaplin A Analog Inhibits Growth and Metastasis of Triple-Negative Breast Cancer. Mol Ther Oncolytics 2019; 15:140-152. [PMID: 31720371 PMCID: PMC6838941 DOI: 10.1016/j.omto.2019.09.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 09/19/2019] [Indexed: 01/24/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most intractable cancer in women with a high risk of metastasis. While hyper-methylation of histone H3 catalyzed by disruptor of telomeric silencing 1-like (DOT1L), a specific methyltransferase for histone H3 at lysine residue 79 (H3K79), is reported as a potential target for TNBCs, early developed nucleoside-type DOT1L inhibitors are not sufficient for effective inhibition of growth and metastasis of TNBC cells. We found that TNBC cells had a high expression level of DOT1L and a low expression level of E-cadherin compared to normal breast epithelial cells and non-TNBC cells. Here, a novel psammaplin A analog (PsA-3091) exhibited a potent inhibitory effect of DOT1L-mediated H3K79 methylation. Consistently, PsA-3091 also significantly inhibited the proliferation, migration, and invasion of TNBC cells along with the augmented expression of E-cadherin and the suppression of N-cadherin, ZEB1, and vimentin expression. In an orthotopic mouse model, PsA-3091 effectively inhibited lung metastasis and tumor growth by the regulation of DOT1L activity and EMT biomarkers. Together, we report here a new template of DOT1L inhibitor and suggest that targeting DOT1L-mediated H3K79 methylation by a novel PsA analog may be a promising strategy for the treatment of metastatic breast cancer patients.
Collapse
Affiliation(s)
- Woong Sub Byun
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Won Kyung Kim
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Hae Ju Han
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Hwa-Jin Chung
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyungkuk Jang
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Han Sun Kim
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Sunghwa Kim
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Donghwa Kim
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Eun Seo Bae
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Sunghyouk Park
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Jeeyeon Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyeung-geun Park
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Sang Kook Lee
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
14
|
Liu XJ, Liu HY, Wang HX, Shi YP, Tang R, Zhang S, Chen BQ. Synthesis and antitumor evaluation of novel fused heterocyclic 1,2,4-triazolo[3,4-b]-1,3,4-thiadiazole derivatives. Med Chem Res 2019. [DOI: 10.1007/s00044-019-02409-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
15
|
Kshatriya R, Kambale D, Mali S, Jejurkar VP, Lokhande P, Chaudhari HK, Saha S. Brønsted Acid Catalyzed Domino Synthesis of Functionalized 4H‐Chromens and Their ADMET, Molecular Docking and Antibacterial Studies. ChemistrySelect 2019. [DOI: 10.1002/slct.201901775] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Rajpratap Kshatriya
- Department of Dyestuff TechnologyInstitute of Chemical Technology, Matunga Mumbai Maharashtra- 400019
| | - Dayanand Kambale
- Department of ChemistrySavitribai Phule Pune University, Ganeshkhind Pune Maharashtra- 411007
| | - Suraj Mali
- Department of Pharmaceutical Science and TechnologyInstitute of Chemical Technology, Matunga Mumbai Maharashtra- 400019
| | - Valmik P. Jejurkar
- Department of Dyestuff TechnologyInstitute of Chemical Technology, Matunga Mumbai Maharashtra- 400019
| | - Pradeep Lokhande
- Department of ChemistrySavitribai Phule Pune University, Ganeshkhind Pune Maharashtra- 411007
| | - Hemchandra K. Chaudhari
- Department of Pharmaceutical Science and TechnologyInstitute of Chemical Technology, Matunga Mumbai Maharashtra- 400019
| | - Satyajit Saha
- Department of Dyestuff TechnologyInstitute of Chemical Technology, Matunga Mumbai Maharashtra- 400019
| |
Collapse
|
16
|
Marine-Derived Natural Lead Compound Disulfide-Linked Dimer Psammaplin A: Biological Activity and Structural Modification. Mar Drugs 2019; 17:md17070384. [PMID: 31252563 PMCID: PMC6669562 DOI: 10.3390/md17070384] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 06/20/2019] [Accepted: 06/25/2019] [Indexed: 12/16/2022] Open
Abstract
Marine natural products are considered to be valuable resources that are furnished with diverse chemical structures and various bioactivities. To date, there are seven compounds derived from marine natural products which have been approved as therapeutic drugs by the U.S. Food and Drug Administration. Numerous bromotyrosine derivatives have been isolated as a type of marine natural products. Among them, psammaplin A, including the oxime groups and carbon-sulfur bonds, was the first identified symmetrical bromotyrosine-derived disulfide dimer. It has been found to have a broad bioactive spectrum, especially in terms of antimicrobial and antiproliferative activities. The highest potential indole-derived psammaplin A derivative, UVI5008, is used as an epigenetic modulator with multiple enzyme inhibitory activities. Inspired by these reasons, psammaplin A has gradually become a research focus for pharmacologists and chemists. To the best of our knowledge, there is no systematic review about the biological activity and structural modification of psammaplin A. In this review, the pharmacological effects, total synthesis, and synthesized derivatives of psammaplin A are summarized.
Collapse
|
17
|
Saima, Soni I, Lavekar AG, Shukla M, Equbal D, Sinha AK, Chopra S. Biocatalytic synthesis of diaryl disulphides and their bio-evaluation as potent inhibitors of drug-resistant Staphylococcus aureus. Drug Dev Res 2018; 80:171-178. [PMID: 30565263 DOI: 10.1002/ddr.21507] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/06/2018] [Accepted: 11/28/2018] [Indexed: 11/11/2022]
Abstract
Staphylococcus aureus is a WHO Priority II pathogen for its capability to cause acute to chronic infections and to resist antibiotics, thus severely impacting healthcare systems worldwide. In this context, it is urgently desired to discover novel molecules to thwart the continuing emergence of antimicrobial resistance. Disulphide containing small molecules has gained prominence as antibacterials. As their conventional synthesis requires tedious synthetic procedure and sometimes toxic reagents, a green and environmentally benign protocol for their synthesis has been developed through which a series of molecules were obtained and evaluated for antibacterial activity against ESKAPE pathogen panel. The hit compound was tested for cytotoxicity against Vero cells to determine its selectivity index and time-kill kinetics was determined. The activity of hit was determined against a panel of S. aureus multi-drug resistant clinical isolates. Also, its ability to synergize with FDA approved drugs was tested as was its ability to reduce biofilm. We identified bis(2-bromophenyl) disulphide (2t) as possessing equipotent antimicrobial activity against S. aureus including MRSA and VRSA strains. Further, 2t exhibited a selectivity index of 25 with concentration-dependent bactericidal activity, synergized with all drugs tested and significantly reduced preformed biofilm. Taken together, 2t exhibits all properties to be positioned as novel scaffold for anti-staphylococcal therapy.
Collapse
Affiliation(s)
- Saima
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Isha Soni
- Division of Microbiology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Aditya G Lavekar
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Manjulika Shukla
- Division of Microbiology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Danish Equbal
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Arun K Sinha
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Sidharth Chopra
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India.,Division of Microbiology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| |
Collapse
|
18
|
Goettge MN, Cioni JP, Ju KS, Pallitsch K, Metcalf WW. PcxL and HpxL are flavin-dependent, oxime-forming N-oxidases in phosphonocystoximic acid biosynthesis in Streptomyces. J Biol Chem 2018; 293:6859-6868. [PMID: 29540479 PMCID: PMC5936822 DOI: 10.1074/jbc.ra118.001721] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/12/2018] [Indexed: 12/13/2022] Open
Abstract
Several oxime-containing small molecules have useful properties, including antimicrobial, insecticidal, anticancer, and immunosuppressive activities. Phosphonocystoximate and its hydroxylated congener, hydroxyphosphonocystoximate, are recently discovered oxime-containing natural products produced by Streptomyces sp. NRRL S-481 and Streptomyces regensis NRRL WC-3744, respectively. The biosynthetic pathways for these two compounds are proposed to diverge at an early step in which 2-aminoethylphosphonate (2AEPn) is converted to (S)-1-hydroxy-2-aminoethylphosphonate ((S)-1H2AEPn) in S. regensis but not in Streptomyces sp. NRRL S-481). Subsequent installation of the oxime moiety into either 2AEPn or (S)-1H2AEPn is predicted to be catalyzed by PcxL or HpxL from Streptomyces sp. NRRL S-481 and S. regensis NRRL WC-3744, respectively, whose sequence and predicted structural characteristics suggest they are unusual N-oxidases. Here, we show that recombinant PcxL and HpxL catalyze the FAD- and NADPH-dependent oxidation of 2AEPn and 1H2AEPn, producing a mixture of the respective aldoximes and nitrosylated phosphonic acid products. Measurements of catalytic efficiency indicated that PcxL has almost an equal preference for 2AEPn and (R)-1H2AEPn. 2AEPn was turned over at a 10-fold higher rate than (R)-1H2AEPn under saturating conditions, resulting in a similar but slightly lower kcat/Km We observed that (S)-1H2AEPn is a relatively poor substrate for PcxL but is clearly the preferred substrate for HpxL, consistent with the proposed biosynthetic pathway in S. regensis. HpxL also used both 2AEPn and (R)-1H2AEPn, with the latter inhibiting HpxL at high concentrations. Bioinformatic analysis indicated that PcxL and HpxL are members of a new class of oxime-forming N-oxidases that are broadly dispersed among bacteria.
Collapse
Affiliation(s)
- Michelle N Goettge
- From the Department of Microbiology and the Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 and
| | - Joel P Cioni
- From the Department of Microbiology and the Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 and
| | - Kou-San Ju
- From the Department of Microbiology and the Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 and
| | - Katharina Pallitsch
- the Institute of Organic Chemistry, University of Vienna, 1090 Vienna, Austria
| | - William W Metcalf
- From the Department of Microbiology and the Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 and
| |
Collapse
|
19
|
Design, synthesis and spectroscopic and crystallographic characterisation of novel functionalized pyrazole derivatives: biological evaluation for their cytotoxic, angiogenic and antioxidant activities. RESEARCH ON CHEMICAL INTERMEDIATES 2018. [DOI: 10.1007/s11164-018-3445-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
20
|
Cincinelli R, Musso L, Artali R, Guglielmi M, Bianchino E, Cardile F, Colelli F, Pisano C, Dallavalle S. Camptothecin-psammaplin A hybrids as topoisomerase I and HDAC dual-action inhibitors. Eur J Med Chem 2018; 143:2005-2014. [DOI: 10.1016/j.ejmech.2017.11.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 11/06/2017] [Accepted: 11/07/2017] [Indexed: 12/15/2022]
|
21
|
Gomes NGM, Dasari R, Chandra S, Kiss R, Kornienko A. Marine Invertebrate Metabolites with Anticancer Activities: Solutions to the "Supply Problem". Mar Drugs 2016; 14:E98. [PMID: 27213412 PMCID: PMC4882572 DOI: 10.3390/md14050098] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 04/29/2016] [Accepted: 05/05/2016] [Indexed: 02/07/2023] Open
Abstract
Marine invertebrates provide a rich source of metabolites with anticancer activities and several marine-derived agents have been approved for the treatment of cancer. However, the limited supply of promising anticancer metabolites from their natural sources is a major hurdle to their preclinical and clinical development. Thus, the lack of a sustainable large-scale supply has been an important challenge facing chemists and biologists involved in marine-based drug discovery. In the current review we describe the main strategies aimed to overcome the supply problem. These include: marine invertebrate aquaculture, invertebrate and symbiont cell culture, culture-independent strategies, total chemical synthesis, semi-synthesis, and a number of hybrid strategies. We provide examples illustrating the application of these strategies for the supply of marine invertebrate-derived anticancer agents. Finally, we encourage the scientific community to develop scalable methods to obtain selected metabolites, which in the authors' opinion should be pursued due to their most promising anticancer activities.
Collapse
Affiliation(s)
- Nelson G M Gomes
- REQUIMTE/LAQV, Laboratory of Pharmacognosy, Department of Chemistry, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira No. 228, 4050-313 Porto, Portugal.
| | - Ramesh Dasari
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX 78666, USA.
| | - Sunena Chandra
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX 78666, USA.
| | - Robert Kiss
- Laboratoire de Cancérologie et de Toxicologie Expérimentale, Faculté de Pharmacie, Université Libre de Bruxelles, Campus de la Plaine, CP205/1, Boulevard du Triomphe, 1050 Brussels, Belgium.
| | - Alexander Kornienko
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX 78666, USA.
| |
Collapse
|
22
|
Wen J, Bao Y, Niu Q, Liu J, Yang J, Wang W, Jiang T, Fan Y, Li K, Wang J, Zhao L, Liu D. Synthesis, biological evaluation and molecular modeling studies of psammaplin A and its analogs as potent histone deacetylases inhibitors and cytotoxic agents. Bioorg Med Chem Lett 2015; 26:4372-6. [PMID: 27460171 DOI: 10.1016/j.bmcl.2015.12.094] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 09/05/2015] [Accepted: 12/28/2015] [Indexed: 02/02/2023]
Abstract
In this study, a concise synthetic method of psammaplin A was achieved from 3-bromo-4-hydroxybenzaldahyde and hydantoin through a four-step synthesis via Knoevenagel condensation, hydrolysis, oximation and amidation in 37% overall yield. A collection of novel psammaplin A analogs focused on the variations of substituents at the benzene ring and modifications at the oxime moiety were synthesized. Among all the synthesized compounds, 5d and 5e showed better HDAC inhibition than psammaplin A and comparable cytotoxicity against four cancer cell lines (PC-3, MCF-7, A549 and HL-60). Molecular docking and dynamics simulation revealed that (i) hydrogen atom of the oxime group interacts with Asp99 of HDAC1 through a water bridged hydrogen bond and (ii) a hydroxyl group is optimal attached on the para-position of benzene, interacting with Glu203 at the entrance to the active site tunnel.
Collapse
Affiliation(s)
- Jiachen Wen
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yu Bao
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qun Niu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiang Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jinyu Yang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Wanqiao Wang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Tao Jiang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yinbo Fan
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Kun Li
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jian Wang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Linxiang Zhao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Dan Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
23
|
Lee JY, Lee MY, Ha MW, Won TH, Cho HJ, Shin J, Park HG, Kim DD. Determination and validation of psammaplin A and its derivatives in rat plasma by liquid chromatography-tandem mass spectrometry and its application in pharmacokinetic study. J Chromatogr B Analyt Technol Biomed Life Sci 2015; 1000:155-62. [PMID: 26232561 DOI: 10.1016/j.jchromb.2015.07.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Revised: 07/01/2015] [Accepted: 07/05/2015] [Indexed: 10/23/2022]
Abstract
A liquid chromatography-tandem mass (LC-MS/MS) method was developed for the determination of psammaplin A (PsA) and its newly synthesized derivatives (PsA 107, PsA 109, and PsA 123) in rat plasma using bupropion as an internal standard (IS). The plasma samples were deproteinized with acetonitrile. Chromatographic separation was performed on hydro-RP column (75×2.0mm, 80Å, 4μm) with isocratic elution using 5mM ammonium formate buffer/acetonitrile (30:70, v/v) at a flow rate of 0.4mL/min and the total run time was 5min. Mass spectrometric detection was performed with positive electrospray ionization (ESI) in multiple reaction monitoring (MRM) mode. The ion transitions monitored were m/z 663.2→331.0, 687.2→343.1, 587.3→293.1, 563.3→281.0, and 240.0→184.0 for PsA, PsA 107, PsA 109, PsA 123, and IS, respectively. All analytes showed good linearity over the concentration range of 5.00-5000ng/mL (r(2)≥0.994). The lower limit of quantification was 5ng/mL for PsA and its three PsA derivatives. Within- and between-run precisions (relative standard deviation, RSD) were less than 9.66% and accuracy (relative error, RE) ranged from -9.34% to 7.25%. Established method was successfully applied to the investigation of pharmacokinetic properties of PsA and its derivatives in rats after intravenous administration at a dose of 2mg/kg.
Collapse
Affiliation(s)
- Jae-Young Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Mee Yeon Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Min Woo Ha
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Tae Hyung Won
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Hyun-Jong Cho
- College of Pharmacy, Kangwon National University, Chuncheon 200-701, Republic of Korea
| | - Jongheon Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Hyeung-geun Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Dae-Duk Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea.
| |
Collapse
|