1
|
Lee ASY, Lin TH, Liu YY, Wang YH, Cheng SC, Li TS, Sun CY, Chen YH. Growth inhibition and toxicity assessments of cis-3,4-diaryl-α-methylene-γ-butyrolactams in cultured human renal cancer cells and zebrafish embryos. Biochim Biophys Acta Gen Subj 2025; 1869:130761. [PMID: 39788219 DOI: 10.1016/j.bbagen.2025.130761] [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: 06/01/2024] [Revised: 12/15/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
This study aimed to compare and evaluate the growth inhibition effects of eight previously synthesized compounds, cis-3,4-diaryl-α-methylene-γ-butyrolactams (compounds 1-8), on two human renal carcinoma cell (RCC) lines: CRL-1932 (rapid growth) and HTB-44 (slow growth). MTT assays and flow cytometry were conducted, revealing that compounds 5 and 6 had the potential to induce cell death in the slow-growing RCC cells (HTB-44), while compound 8 demonstrated effectiveness in both RCC lines (HTB-44 and CRL-1932). Additionally, a non-transformed HEK293 cell line and a transgenic zebrafish with a green fluorescent kidney Tg(wt1b:egfp) were used to assess the toxicities of compounds 5, 6, and 8. The findings suggested that compound 8 was relatively non-toxic compared to the others. Western blot analysis indicated that compounds 5, 6, and 8 may interact with the P53/mTOR pathways. Based on these results, we concluded that compound 8 exhibits RCC growth inhibition properties and has lower toxicity, making it a candidate for further investigation in mammalian models.
Collapse
Affiliation(s)
- Adam Shih-Yuan Lee
- Department of Chemistry, Tamkang University, 151, Yingzhuan Road, Danshui Dist., New Taipei City 25137, Taiwan
| | - Ta-Hsien Lin
- Division of Basic Research, Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Biochemistry and Molecular Biology, National Ying Ming Chiao Tung University, Taipei, Taiwan
| | - Yen-Yu Liu
- Department of Chemistry, Tamkang University, 151, Yingzhuan Road, Danshui Dist., New Taipei City 25137, Taiwan
| | - Yun-Hsin Wang
- Department of Chemistry, Tamkang University, 151, Yingzhuan Road, Danshui Dist., New Taipei City 25137, Taiwan
| | - Shu-Chun Cheng
- Division of Nephrology, Department of Internal Medicine, Keelung Chang Gung Memorial Hospital, Keelung 204, Taiwan
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Japan
| | - Chiao-Yin Sun
- Division of Nephrology, Department of Internal Medicine, Keelung Chang Gung Memorial Hospital, Keelung 204, Taiwan; College of Medicine, Chang Gung University, Taoyuan 333, Taiwan.
| | - Yau-Hung Chen
- Department of Chemistry, Tamkang University, 151, Yingzhuan Road, Danshui Dist., New Taipei City 25137, Taiwan.
| |
Collapse
|
2
|
Tang J, Zhang Y, Zhou L, Song X, Wei Y, Qi J, Wu J, Song Z, Zhan L. Design, synthesis and biological evaluation of indoline-maleimide conjugates as potential antitumor agents for the treatment of colorectal cancer. Bioorg Med Chem 2024; 108:117786. [PMID: 38843656 DOI: 10.1016/j.bmc.2024.117786] [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: 03/26/2024] [Revised: 05/21/2024] [Accepted: 05/30/2024] [Indexed: 06/17/2024]
Abstract
An efficient protocol for direct coupling of maleimides and indolines at the C7-position was achieved under Rh(III) catalysis. Thirty four novel indoline-maleimide conjugates were prepared in good to excellent yields using this method. All compounds were evaluated for their anti-proliferative effect against colorectal cell lines. Among them, compound 3ab showed the most potent anti-proliferative activity against the CRC cells, and displayed low toxicity in the normal cell. Further investigation indicated that 3ab could effectively suppress the proliferation and migration of CRC cells, along with inducing cell cycle arrest and apoptosis. Mechanistic studies revealed that compound 3ab inhibited the proliferation of CRC cells via suppressing the AKT/GSK-3β pathway. In vivo evaluation demonstrated remarkable antitumor effect of 3ab (10 mg/kg) in the HCT116 xenograft model with no obvious toxicity, which is superior to that of 5-Fluorouracil (20 mg/kg). Therefore, conjugate 3ab could be considered as a potential CRC therapy agent for further development.
Collapse
Affiliation(s)
- Jielin Tang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yuxin Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lingling Zhou
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiangrui Song
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yusi Wei
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Ji Qi
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jianmin Wu
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Zengqiang Song
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Lingling Zhan
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
3
|
Chagaleti BK, Saravanan V, Vellapandian C, Kathiravan MK. Exploring cyclin-dependent kinase inhibitors: a comprehensive study in search of CDK-6 inhibitors using a pharmacophore modelling and dynamics approach. RSC Adv 2023; 13:33770-33785. [PMID: 38019988 PMCID: PMC10655667 DOI: 10.1039/d3ra05672d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023] Open
Abstract
Cancer prevalence and resistance issues in cancer treatment are a significant public health concern globally. Among the existing strategies in cancer therapy, targeting cyclin-dependent kinases (CDKs), especially CDK-6 is found to be one of the most promising targets, as this enzyme plays a pivotal role in cell cycle stages and cell proliferation. Cell proliferation is the characteristic feature of cancer giving rise to solid tumours. Our research focuses on creating novel compounds, specifically, pyrazolopyrimidine fused azetidinones, using a groundbreaking molecular hybridization approach to target CDK-6. Through computational investigations, ligand-based pharmacophore modelling, pharmacokinetic studies (ADMET), molecular docking, and dynamics simulations, we identified 18 promising compounds. The pharmacophore model featured one aromatic hydrophobic centre (F1: Aro/Hyd) and two H-bond acceptors (F2 and F3: Acc). Molecular docking results showed favourable binding energies (-6.5 to -8.0 kcal mol-1) and effective hydrogen bonds and hydrophobic interactions. The designed compounds demonstrated good ADMET profiles. Specifically, B6 and B18 showed low energy conformation (-7.8 kcal and -7.6 kcal), providing insights into target inhibition compared to the standard drug Palbociclib. Extensive molecular dynamics simulations confirmed the stability of these derivatives. Throughout the 100 ns simulation, the ligand-protein complexes maintained structural stability, with acceptable RMSD values. These compounds hold promise as potential leads in cancer therapy.
Collapse
Affiliation(s)
- Bharath Kumar Chagaleti
- Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology Kattankulathur-603203 India
| | - Venkatesan Saravanan
- Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology Kattankulathur-603203 India
| | - Chitra Vellapandian
- Department of Pharmacology, SRM College of Pharmacy SRMIST, Kattankulathur Chennai Tamil Nadu - 603 203 India
| | - Muthu K Kathiravan
- Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology Kattankulathur-603203 India
- Dr A. P. J. Abdul Kalam Research Lab, Department of Pharmaceutical Chemistry, SRM College of Pharmacy SRMIST, Kattankulathur Chennai Tamil Nadu - 603 203 India
| |
Collapse
|
4
|
Malebari AM, Wang S, Greene TF, O’Boyle NM, Fayne D, Khan MF, Nathwani SM, Twamley B, McCabe T, Zisterer DM, Meegan MJ. Synthesis and Antiproliferative Evaluation of 3-Chloroazetidin-2-ones with Antimitotic Activity: Heterocyclic Bridged Analogues of Combretastatin A-4. Pharmaceuticals (Basel) 2021; 14:1119. [PMID: 34832901 PMCID: PMC8624998 DOI: 10.3390/ph14111119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 12/29/2022] Open
Abstract
Antimitotic drugs that target tubulin are among the most widely used chemotherapeutic agents; however, the development of multidrug resistance has limited their clinical activity. We report the synthesis and biological properties of a series of novel 3-chloro-β-lactams and 3,3-dichloro-β-lactams (2-azetidinones) that are structurally related to the tubulin polymerisation inhibitor and vascular targeting agent, Combretastatin A-4. These compounds were evaluated as potential tubulin polymerisation inhibitors and for their antiproliferative effects in breast cancer cells. A number of the compounds showed potent activity in MCF-7 breast cancer cells, e.g., compound 10n (3-chloro-4-(3-hydroxy-4-methoxy-phenyl)-1-(3,4,5-trimethoxyphenyl)azetidin-2-one) and compound 11n (3,3-dichloro-4-(3-hydroxy-4-methoxyphenyl)-1-(3,4,5-trimethoxyphenyl)-azetidin-2-one), with IC50 values of 17 and 31 nM, respectively, and displayed comparable cellular effects to those of Combretastatin A-4. Compound 10n demonstrated minimal cytotoxicity against non-tumorigenic HEK-293T cells and inhibited the in vitro polymerisation of tubulin with significant G2/M phase cell cycle arrest. Immunofluorescence staining of MCF-7 cells confirmed that β-lactam 10n caused a mitotic catastrophe by targeting tubulin. In addition, compound 10n promoted apoptosis by regulating the expression of pro-apoptotic protein BAX and anti-apoptotic proteins Bcl-2 and Mcl-1. Molecular docking was used to explore the potential molecular interactions between novel 3-chloro-β-lactams and the amino acid residues of the colchicine binding active site cavity of β-tubulin. Collectively, these results suggest that 3-chloro-2-azetidinones, such as compound 10n, could be promising lead compounds for further clinical anti-cancer drug development.
Collapse
Affiliation(s)
- Azizah M. Malebari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Shu Wang
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (S.W.); (T.F.G.); (N.M.O.)
| | - Thomas F. Greene
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (S.W.); (T.F.G.); (N.M.O.)
| | - Niamh M. O’Boyle
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (S.W.); (T.F.G.); (N.M.O.)
| | - Darren Fayne
- Molecular Design Group, School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (D.F.); (M.F.K.)
| | - Mohemmed Faraz Khan
- Molecular Design Group, School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (D.F.); (M.F.K.)
| | - Seema M. Nathwani
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (S.M.N.); (D.M.Z.)
| | - Brendan Twamley
- School of Chemistry, Trinity College Dublin, 2 DO2R590 Dublin, Ireland; (B.T.); (T.M.)
| | - Thomas McCabe
- School of Chemistry, Trinity College Dublin, 2 DO2R590 Dublin, Ireland; (B.T.); (T.M.)
| | - Daniela M. Zisterer
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (S.M.N.); (D.M.Z.)
| | - Mary J. Meegan
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (S.W.); (T.F.G.); (N.M.O.)
| |
Collapse
|
5
|
Mandal MK, Ghosh S, Bhat HR, Naesens L, Singh UP. Synthesis and biological evaluation of substituted phenyl azetidine-2-one sulphonyl derivatives as potential antimicrobial and antiviral agents. Bioorg Chem 2020; 104:104320. [PMID: 33142428 DOI: 10.1016/j.bioorg.2020.104320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/22/2020] [Accepted: 09/24/2020] [Indexed: 10/23/2022]
Abstract
In the present study, we intend to synthesize a series of novel substituted phenyl azetidine-2-one sulphonyl derivatives. The entire set of derivatives 5 (a-t) were screened for in-vitro antibacterial, and antifungal activity, and among them eleven compounds were further screened for the antiviral activity to predict their efficacy against pathogenic viruses. Interestingly, compound 5d, 5e, 5f, 5h, 5i, and 5j showed similar or better antibacterial activity as compared to ampicillin (standard). Moreover, compounds 5h, 5i, 5j, and 5q showed good inhibitory activity against fungal strains whereas other derivatives had mild or diminished activity in comparison with standard drug clotrimazole. The antimicrobial study indicated that compounds having electron-withdrawing groups showed the highest activity. Interestingly, these tested compounds showed weak antiviral activity against Vaccinia virus, Human Coronavirus (229E), Reovirus-1, Herpes simplex virus, Sindbis virus, Coxsackievirus B4, Yellow Fever virus, and Influenza B virus in HEL cell, Vero cell, and MDCK cell cultures. The findings of the present study might open new avenues to target human disease-causing deadly microbes and viruses.
Collapse
Affiliation(s)
- Milan Kumar Mandal
- Drug Design & Discovery Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, Uttar Pradesh 211007, India
| | - Swagatika Ghosh
- Food Saftey and Drug Administration, Government of Uttar Pradesh, Lucknow, Uttar Pradesh 226018, India
| | - Hans Raj Bhat
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam 786004, India
| | - Lieve Naesens
- Rega Institute of Medical Research, KU Leuven, B-3000 Leuven, Belgium
| | - Udaya Pratap Singh
- Drug Design & Discovery Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, Uttar Pradesh 211007, India.
| |
Collapse
|
6
|
Twamley B, O’Boyle NM, Meegan MJ. Azetidin-2-ones: structures of anti-mitotic compounds based on the 1-(3,4,5-tri-meth-oxy-phen-yl)azetidin-2-one core. Acta Crystallogr E Crystallogr Commun 2020; 76:1187-1194. [PMID: 32843997 PMCID: PMC7405576 DOI: 10.1107/s2056989020008555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 06/25/2020] [Indexed: 11/15/2022]
Abstract
A series of related substituted 1-(3,4,5-tri-meth-oxy-phen-yl)azetidin-2-ones have been characterized: 3-(4-fluoro-phen-yl)-4-(4-meth-oxy-phen-yl)-1-(3,4,5-tri-meth-oxy-phen-yl)azetidin-2-one, C25H24FNO5 (1), 3-(furan-2-yl)-4-(4-meth-oxy-phen-yl)-1-(3,4,5-tri-meth-oxy-phen-yl)azetidin-2-one, C23H23NO6 (2), 4-(4-meth-oxyphen-yl)-3-(naphthalen-1-yl)-1-(3,4,5-tri-meth-oxy-phen-yl)azetidin-2-one, C29H27NO5 (3), 3-(3,4-di-meth-oxy-phen-yl)-4-(4-meth-oxy-phen-yl)-1-(3,4,5-tri-meth-oxy-phen-yl)azetidin-2-one, C27H29NO7 (4) and 4,4-bis-(4-meth-oxy-phen-yl)-3-phenyl-1-(3,4,5-tri-meth-oxy-phen-yl)azetidin-2-one, C32H31NO6 (5). All of the compounds are racemic. The lactam and 3,4,5-tri-meth-oxy-phenyl rings are approximately co-planar and the orientation of the lactam and the 4-meth-oxy-phenyl substituent is approximately orthogonal. The chiral centres, although eclipsed by geometry, have torsion angles ranging from -7.27 to 13.08° for the 3 position, and -8.69 to 13.76° for the 4 position of the β-lactam. The structures display intra-molecular C-H⋯O bonding between the 3,4,5-tri-meth-oxy-phenyl ring and the lactam ketone. Further C-H⋯O inter-actions are observed and form either an opposing meth-oxy 'buckle' to join two mol-ecules together or a cyclic dimer.
Collapse
Affiliation(s)
- Brendan Twamley
- School of Chemistry, Trinity College Dublin, Dublin 2, Ireland
| | - Niamh M. O’Boyle
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152 - 160 Pearse St, Dublin 2, Ireland
| | - Mary J. Meegan
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152 - 160 Pearse St, Dublin 2, Ireland
| |
Collapse
|
7
|
Fu DJ, Zhang YF, Chang AQ, Li J. β-Lactams as promising anticancer agents: Molecular hybrids, structure activity relationships and potential targets. Eur J Med Chem 2020; 201:112510. [PMID: 32592915 DOI: 10.1016/j.ejmech.2020.112510] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/17/2020] [Accepted: 05/20/2020] [Indexed: 01/17/2023]
Abstract
β-Lactam, commonly referred as azetidin-2-one, is a multifunctional building block for synthesizing β-amino ketones, γ-amino alcohols, and other compounds. Besides its well known antibiotic activity, this ring system exhibits a wide range of activities, attracting the attention of researchers. However, the structurally diverse β-lactam analogues as anticancer agents and their different molecular targets are poorly discussed. The purpose of this review is 3-fold: (1) to explore the molecular hybridization approach to design β-lactams hybrids as anticancer agents; (2) the structure activity relationship of the most active anticancer β-lactams and (3) to summarize their antitumor mechanisms.
Collapse
Affiliation(s)
- Dong-Jun Fu
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Yun-Feng Zhang
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - An-Qi Chang
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Jun Li
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China.
| |
Collapse
|
8
|
Hrichi H, Elkanzi NAA, Bakr RB. Novel Β-lactams and Thiazolidinone Derivatives from 1,4-dihydroquinoxaline Schiff’s Base: Synthesis, Antimicrobial Activity and Molecular Docking Studies. CHEMISTRY JOURNAL OF MOLDOVA 2020. [DOI: 10.19261/cjm.2019.647] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
9
|
da Silveira Pinto LS, Vasconcelos TRA, Gomes CRB, de Souza MVN. A Brief Review on the Development of Novel Potentially Active Azetidin-2-ones Against Cancer. CURR ORG CHEM 2020. [DOI: 10.2174/1385272824666200303115444] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Azetidin-2-ones (β-lactams) and its derivatives are an important group of heterocyclic compounds that exhibit a wide range of pharmacological properties such as antibacterial, anticancer, anti-diabetic, anti-inflammatory and anticonvulsant. Efforts have been made over the years to develop novel congeners with superior biological activities and minimal potential for undesirable side effects. The present review aimed to highlight some recent discoveries (2013-2019) on the development of novel azetidin-2-one-based compounds as potential anticancer agents.
Collapse
Affiliation(s)
- Ligia S. da Silveira Pinto
- Universidade Federal Fluminense, Instituto de Quimica, Departamento de Quimica Organica, Programa de Pos-Graduacao em Quimica. Outeiro de Sao Joao Batista, s/no, Centro, Niteroi, 24020-141, Rio de Janeiro, Brazil
| | - Thatyana R. Alves Vasconcelos
- Universidade Federal Fluminense, Instituto de Quimica, Departamento de Quimica Organica, Programa de Pos-Graduacao em Quimica. Outeiro de Sao Joao Batista, s/no, Centro, Niteroi, 24020-141, Rio de Janeiro, Brazil
| | - Claudia Regina B. Gomes
- Fundacao Oswaldo Cruz, Instituto de Tecnologia em Farmacos-Farmanguinhos. Rua Sizenando Nabuco 100, Manguinhos, 21041-250, Rio de Janeiro, RJ, Brazil
| | - Marcus Vinícius N. de Souza
- Fundacao Oswaldo Cruz, Instituto de Tecnologia em Farmacos-Farmanguinhos. Rua Sizenando Nabuco 100, Manguinhos, 21041-250, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
10
|
Ginsenoside Rh4 suppresses aerobic glycolysis and the expression of PD-L1 via targeting AKT in esophageal cancer. Biochem Pharmacol 2020; 178:114038. [PMID: 32422139 DOI: 10.1016/j.bcp.2020.114038] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/11/2020] [Indexed: 12/12/2022]
Abstract
Ginsenoside Rh4, as a bioactive component obtained from Panax notoginseng, has excellent pharmacological efficacy especially antitumor effects. However, its anticancer effects and target mechanisms in regulating human esophageal cancer are still poorly understood. Here, the results suggested that Rh4 exhibited potent anti-esophageal cancer effects in vivo and in vitro. Flow cytometric analysis and Western Blot showed that Rh4 significantly inhibited the growth by inducing G1 phase arrest. In parallel, Rh4 inhibited aerobic glycolysis in esophageal cancer cells by hindering lactate production, glucose uptake and ATP production; reducing the extracellular acidification rate (ECAR) and oxygen consumption rate (OCR); suppressing aerobic glycolysis-related protein expression. Mechanistic studies demonstrated that AKT is a possible target of Rh4, which suppresses aerobic glycolysis. Rh4 administration resulted in AKT deregulation, whereas treatment with insulin abolished the inhibitory effect of Rh4 on aerobic glycolysis. In contrast, treatment with AKT inhibitors or siRNA that silenced AKT enhanced the inhibitory effect of Rh4 on aerobic glycolysis. Moreover, molecular docking results indicated that Rh4 was able to bind to the interdomain region of AKT. Interestingly, the results revealed that Rh4 also inhibited the expression of PD-L1 via the AKT/mTOR pathway. Taken together, our findings provide important insights into the anti-esophageal cancer effects of Rh4 via suppressing aerobic glycolysis and PD-L1 expression, which indicated Rh4 could be as promising drug for clinical treatment.
Collapse
|
11
|
Cytotoxicity, anticancer, and antioxidant properties of mono and bis-naphthalimido β-lactam conjugates. Med Chem Res 2020. [DOI: 10.1007/s00044-020-02552-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
12
|
Zhang X, Jia Y. Recent Advances in β-lactam Derivatives as Potential Anticancer Agents. Curr Top Med Chem 2020; 20:1468-1480. [PMID: 32148196 DOI: 10.2174/1568026620666200309161444] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 01/22/2020] [Accepted: 01/24/2020] [Indexed: 01/26/2023]
Abstract
Cancer, accounts for around 10 million deaths annually, is the second leading cause of death globally. The continuous emergency of drug-resistant cancers and the low specificity of anticancer agents are the main challenges in the control and eradication of cancers, so it is imperative to develop novel anticancer agents. Immense efforts have been made in developing new lead compounds and novel chemotherapeutic strategies for the treatment of various forms of cancers in recent years. β-Lactam derivatives constitute versatile and attractive scaffolds for the drug discovery since these kinds of compounds possess a variety of pharmacological properties, and some of them exhibited promising potency against both drug-sensitive and drug-resistant cancer cell lines. Thus, β-lactam moiety is a useful template for the development of novel anticancer agents. This review will provide an overview of β-lactam derivatives with the potential therapeutic application for the treatment of cancers covering articles published between 2000 and 2020. The mechanisms of action, the critical aspects of design and structureactivity relationships are also discussed.
Collapse
Affiliation(s)
- Xinfen Zhang
- Department of Oncology, Zhuji Affiliated Hospital of Shaoxing University, Zhejiang Province 311800, China
| | - Yanshu Jia
- Chongqing Institute of Engineering, Chongqing 400056, China
| |
Collapse
|
13
|
Basu P, Hazra C, Baiju TV, Namboothiri INN. Synthesis of tetrahydrothiopyrano[2,3-b]indoles via [3+3] annulation of nitroallylic acetates with indoline-2-thiones. NEW J CHEM 2020. [DOI: 10.1039/c9nj04754a] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
[3+3] annulation of 1,3-binucleophilic indolin-2-thione with 1,3-bielectrophilic nitroallylic acetate via an SN2-reaction-6-endo-trig cyclization takes place with high regio- and stereoselectivity to afford tetrahydrothiopyranoindoles.
Collapse
Affiliation(s)
- Pallabita Basu
- Department of Chemistry
- Indian Institute of Technology Bombay
- Mumbai 400 076
- India
| | - Chiranjit Hazra
- Department of Chemistry
- Indian Institute of Technology Bombay
- Mumbai 400 076
- India
| | - Thekke V. Baiju
- Department of Chemistry
- Indian Institute of Technology Bombay
- Mumbai 400 076
- India
| | | |
Collapse
|
14
|
Sachdeva T, Low ML, Mai C, Cheong SL, Liew YK, Milton MD. Design, Synthesis and Characterisation of Novel Phenothiazine‐Based Triazolopyridine Derivatives: Evaluation of Anti‐Breast Cancer Activity on Human Breast Carcinoma. ChemistrySelect 2019; 4:12701-12707. [DOI: 10.1002/slct.201903203] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 10/25/2019] [Indexed: 02/05/2023]
Abstract
AbstractA series of novel phenothiazine based [1,2,4]triazolo[4, 3‐a]pyridine scaffolds were designed and synthesized in good yields by the oxidative cyclisation of phenothiazine pyridylhydrazones. Biological responses of all compounds toward a panel of human breast cancer cells (MDA‐MB‐231, MDA‐MB‐468, MCF7, SKBR3 and T47D) and human non‐tumorigenic epithelial breast cells (MCF10 A) were evaluated. Structure‐activity relationship revealed that compound with pendant phenyl ring on phenothiazine exhibited significant cytotoxic activity and apoptotic induction effects against breast cancer cell line with IC50 value 10.2 to 17.6 μM. Notably, the cytotoxic effect was 3.5 fold higher on cancer than non‐cancer cells, indicating potential control of breast cancer with lower side effects. Molecular docking studies confirmed the presence of hydrophobic contacts between appended phenyl ring, triazolopyridine and phenothiazine moieties with adjacent residues within the binding pocket of tubulin. One of the nitrogen in the triazolo ring also showed hydrogen bonding with tubulin. These tubulin interactions were also found with the taxane ring of paclitaxel. Cell cycle analysis confirmed the G2/M arrest induced by this compound on human breast cancer cells. Therefore, the potential anti‐cancer, pro‐apoptotic, and cell cycle arrest warrant further development of this molecule as a new class of anticancer agent.
Collapse
Affiliation(s)
| | - May Lee Low
- Department of Pharmaceutical Chemistry School of Pharmacy International Medical University, No. 126 Jalan Jalil Perkasa 19, Bukit Jalil 57000 Kuala Lumpur Malaysia
| | - Chun‐Wai Mai
- Department of Pharmaceutical Chemistry School of Pharmacy International Medical University, No. 126 Jalan Jalil Perkasa 19, Bukit Jalil 57000 Kuala Lumpur Malaysia
- Center for Cancer and Stem Cell Research Institute for Research, Development and Innovation International Medical University, No. 126 Jalan Jalil Perkasa 19, Bukit Jalil 57000 Kuala Lumpur Malaysia
| | - Siew Lee Cheong
- Department of Pharmaceutical Chemistry School of Pharmacy International Medical University, No. 126 Jalan Jalil Perkasa 19, Bukit Jalil 57000 Kuala Lumpur Malaysia
| | - Yun Khoon Liew
- Department of Life Sciences School of Pharmacy International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil 57000 Kuala Lumpur Malaysia
| | | |
Collapse
|
15
|
Debnath S, Kanakaraju M, Islam M, Yeeravalli R, Sen D, Das A. In silico design, synthesis and activity of potential drug-like chrysin scaffold-derived selective EGFR inhibitors as anticancer agents. Comput Biol Chem 2019; 83:107156. [PMID: 31710991 DOI: 10.1016/j.compbiolchem.2019.107156] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/26/2019] [Accepted: 10/28/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND & OBJECTIVE Epidermal growth factor receptor (EGFR) signaling pathway is one of the promising and well-established targets for anticancer therapy. The objective of the present study was to identify new EGFR inhibitors using ligand and structure-based drug designing methods, followed by a synthesis of selected inhibitors and evaluation of their activity. METHODS A series of C-7-hydroxyproton substituted chrysin derivatives were virtually drawn to generate a small compound library that was screened using 3D QSAR model created from forty-two known EGFR tyrosine kinase inhibitors. Next, the obtained hits with fitness score ≥ 1.0 were subjected to molecular docking analysis. Based on the predicted activity and XP glide score, three EGFR inhibitors were synthesized and characterized using 1H-NMR, 13C-NMR and MS. Finally, comparative in vitro investigation of the biological activity of synthesized inhibitors was performed with that of the parent molecule, chrysin. RESULTS The data depicted a 3.2-fold enhanced cytotoxicity of chrysin derivative, CHM-04 against breast cancer cells as compared with chrysin as well as its binding with EGFR protein. Furthermore, the biological activity of CHM-04 was comparable to the standard EGFR inhibitor, AG1478 in increasing apoptosis and decreasing the migratory potential of triple-negative breast cancer cells as well as significantly lowering the mammosphere forming ability of breast cancer stem cells. CONCLUSION The present study suggests CHM-04, an EGFR inhibitor possessing drug-like properties as a plausible therapeutic candidate against breast cancer.
Collapse
Affiliation(s)
- Sudhan Debnath
- Department of Chemistry, Maharaja Bir Bikram College, Agartala, 799 004, Tripura, India
| | - Manupati Kanakaraju
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, 500 007, India; Academy of Scientific & Innovative Research, 2 Rafi Marg, New Delhi, 110 001, India
| | - Minarul Islam
- Department of Chemistry, Maharaja Bir Bikram College, Agartala, 799 004, Tripura, India
| | - Ragini Yeeravalli
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, 500 007, India; Academy of Scientific & Innovative Research, 2 Rafi Marg, New Delhi, 110 001, India
| | - Debanjan Sen
- Department of Pharmacy, BCDA College of Pharmacy and Technology, Hridaypur, Kolkata, 700 127, India
| | - Amitava Das
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, 500 007, India; Academy of Scientific & Innovative Research, 2 Rafi Marg, New Delhi, 110 001, India.
| |
Collapse
|
16
|
Khanam R, Kumar R, Hejazi II, Shahabuddin S, Meena R, Jayant V, Kumar P, Bhat AR, Athar F. Piperazine clubbed with 2-azetidinone derivatives suppresses proliferation, migration and induces apoptosis in human cervical cancer HeLa cells through oxidative stress mediated intrinsic mitochondrial pathway. Apoptosis 2019; 23:113-131. [PMID: 29349707 DOI: 10.1007/s10495-018-1439-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Piperazine scaffolds or 2-azetidinone pharmacophores have been reported to show anti-cancer activities and apoptosis induction in different types of cancer cells. However, the mechanistic studies involve in induction of apoptosis addressing these two moieties for human cervical cancer cells remain uncertain. The present study emphasizes on the anti-proliferating properties and mechanism involved in induction of apoptosis for these structurally related azoles derivatives in HeLa cancer cells. 1-Phenylpiperazine clubbed with 2-azetidione derivatives (5a-5h) were synthesized, characterized using various spectroscopic techniques and evaluated for their in-vitro anti-proliferative activities and induction of apoptosis. Further, we also evaluated oxidative stress generated by these synthetic derivatives (5a-5h). Cell viability studies revealed that among all, the compound N-(3-chloro-2-(3-nitrophenyl)-4-oxoazetidin-1-yl)-2-(4-phenylpiperazin-1-yl) acetamide 5e remarkably inhibited the growth of HeLa cells in a concentration dependent manner having IC50 value of 29.44 ± 1.46 µg/ml. Morphological changes, colonies suppression and inhibition of migration clearly showed the antineoplasicity in HeLa cells treated with 5e. Simultaneously, phosphatidylserine externalization, DNA fragmentation and cell-cycle arrest showed ongoing apoptosis in the HeLa cancer cells induced by compound 5e in concentration dependent manner. Additionally, generation of intracellular ROS along with the decrease in mitochondrial membrane potential supported that compound 5e caused oxidative stress resulting in apoptosis through mitochondria mediated pathway. Elevation in the level of cytochrome c and upregulation in expression of caspase-3 clearly indicated the involvement of the intrinsic pathway of programmed cell death. In brief; compound 5e could serve as a promising lead for the development of an effective antitumor agent.
Collapse
Affiliation(s)
- Rashmin Khanam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, 110025, India
| | - Raj Kumar
- Radiation and Cancer Therapeutic Lab, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Iram Iqbal Hejazi
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, 110025, India
| | - Syed Shahabuddin
- Research Centre for Nano-Materials and Energy Technology (RCNMET), School of Science and Technology, Sunway University, 47500, Selangor, Malaysia
| | - Ramovatar Meena
- Radiation and Cancer Therapeutic Lab, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Vikrant Jayant
- Radiation and Cancer Therapeutic Lab, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Prabhat Kumar
- Radiation and Cancer Therapeutic Lab, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Abdul Roouf Bhat
- Department of Chemistry, Sri Pratap College, Cluster University, Srinagar, 190001, India.
| | - Fareeda Athar
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, 110025, India.
| |
Collapse
|
17
|
Wang S, Malebari AM, Greene TF, O'Boyle NM, Fayne D, Nathwani SM, Twamley B, McCabe T, Keely NO, Zisterer DM, Meegan MJ. 3-Vinylazetidin-2-Ones: Synthesis, Antiproliferative and Tubulin Destabilizing Activity in MCF-7 and MDA-MB-231 Breast Cancer Cells. Pharmaceuticals (Basel) 2019; 12:ph12020056. [PMID: 30979033 PMCID: PMC6630832 DOI: 10.3390/ph12020056] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/02/2019] [Accepted: 04/07/2019] [Indexed: 12/18/2022] Open
Abstract
Microtubule-targeted drugs are essential chemotherapeutic agents for various types of cancer. A series of 3-vinyl-β-lactams (2-azetidinones) were designed, synthesized and evaluated as potential tubulin polymerization inhibitors, and for their antiproliferative effects in breast cancer cells. These compounds showed potent activity in MCF-7 breast cancer cells with an IC50 value of 8 nM for compound 7s 4-[3-Hydroxy-4-methoxyphenyl]-1-(3,4,5-trimethoxyphenyl)-3-vinylazetidin-2-one) which was comparable to the activity of Combretastatin A-4. Compound 7s had minimal cytotoxicity against both non-tumorigenic HEK-293T cells and murine mammary epithelial cells. The compounds inhibited the polymerisation of tubulin in vitro with an 8.7-fold reduction in tubulin polymerization at 10 μM for compound 7s and were shown to interact at the colchicine-binding site on tubulin, resulting in significant G2/M phase cell cycle arrest. Immunofluorescence staining of MCF-7 cells confirmed that β-lactam 7s is targeting tubulin and resulted in mitotic catastrophe. A docking simulation indicated potential binding conformations for the 3-vinyl-β-lactam 7s in the colchicine domain of tubulin. These compounds are promising candidates for development as antiproiferative microtubule-disrupting agents.
Collapse
Affiliation(s)
- Shu Wang
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, 2 DO2R590 Dublin, Ireland.
| | - Azizah M Malebari
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, 2 DO2R590 Dublin, Ireland.
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Thomas F Greene
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, 2 DO2R590 Dublin, Ireland.
| | - Niamh M O'Boyle
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, 2 DO2R590 Dublin, Ireland.
| | - Darren Fayne
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, 2 DO2R590 Dublin, Ireland.
| | - Seema M Nathwani
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, 2 DO2R590 Dublin, Ireland.
| | - Brendan Twamley
- School of Chemistry, Trinity College Dublin, 2 DO2R590 Dublin, Ireland.
| | - Thomas McCabe
- School of Chemistry, Trinity College Dublin, 2 DO2R590 Dublin, Ireland.
| | - Niall O Keely
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, 2 DO2R590 Dublin, Ireland.
| | - Daniela M Zisterer
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, 2 DO2R590 Dublin, Ireland.
| | - Mary J Meegan
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, 2 DO2R590 Dublin, Ireland.
| |
Collapse
|
18
|
Manupati K, Debnath S, Goswami K, Bhoj PS, Chandak HS, Bahekar SP, Das A. Glutathione S-transferase omega 1 inhibition activates JNK-mediated apoptotic response in breast cancer stem cells. FEBS J 2019; 286:2167-2192. [PMID: 30873742 DOI: 10.1111/febs.14813] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 11/13/2018] [Accepted: 03/12/2019] [Indexed: 12/17/2022]
Abstract
Glutathione S-transferase omega 1 (GSTO1) contributes to the inactivation of a wide range of drug compounds via conjugation to glutathione during phase reactions. Chemotherapy-induced GSTO1 expression in breast cancer cells leads to chemoresistance and promotes metastasis. In search of novel GSTO1 inhibitors, we identified S2E, a thia-Michael adduct of sulfonamide chalcone with low LC50 (3.75 ± 0.73 μm) that binds to the active site of GSTO1, as revealed by molecular docking (glide score: -8.1), cellular thermal shift assay and fluorescence quenching assay (Kb ≈ 10 × 105 mol·L-1 ). Docking studies confirmed molecular interactions between GSTO1 and S2E, and identified the hydrogen bond donor Val-72 (2.14 Å) and hydrogen bond acceptor Ser-86 (2.77 Å). Best pharmacophore hypotheses could effectively map S2E and identified the 4-methyl group of the benzene sulfonamide ring as crucial to its anti-cancer activity. Lack of a thiophenyl group in another analog, 2e, reduced its efficacy as observed by cytotoxicity and pharmacophore matching. Furthermore, GSTO1 inhibition by S2E, along with tamoxifen, led to a significant increase in apoptosis and decreased migration of aggressive MDA-MB-231 cells, as well as significantly decreased migration, invasion and mammosphere formation in sorted breast cancer stem cells (CSCs, CD24- /CD44+ ). GSTO1 silencing in breast CSCs also significantly increased apoptosis and decreased migration. Mechanistically, GSTO1 inhibition activated the c-Jun N-terminal kinase stress kinase, inducing a mitochondrial apoptosis signaling pathway in breast CSCs via the pro-apoptotic proteins BAX, cytochrome c and cleaved caspase 3. Our study elucidated the role of the GSTO1 inhibitor S2E as a potential therapeutic strategy for preventing chemotherapy-induced breast CSC-mediated cancer metastasis and recurrence.
Collapse
Affiliation(s)
- Kanakaraju Manupati
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,Academy of Science and Innovative Research, New Delhi, India
| | - Sudhan Debnath
- Department of Chemistry, Maharaja Bir Bikram College, Agartala, India
| | - Kalyan Goswami
- Department of Biochemistry, Mahatma Gandhi Institute of Medical Sciences, Wardha, India
| | - Priyanka S Bhoj
- Department of Biochemistry, Mahatma Gandhi Institute of Medical Sciences, Wardha, India
| | - Hemant S Chandak
- Department of Chemistry, G. S. Science, Arts & Commerce College, Khamgaon, India
| | - Sandeep P Bahekar
- Department of Chemistry, G. S. Science, Arts & Commerce College, Khamgaon, India
| | - Amitava Das
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,Academy of Science and Innovative Research, New Delhi, India
| |
Collapse
|
19
|
Narendar Reddy T, Jayathirtha Rao V. Importance of Baylis-Hillman adducts in modern drug discovery. Tetrahedron Lett 2018. [DOI: 10.1016/j.tetlet.2018.06.023] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
20
|
Dhoke NR, Geesala R, Das A. Low Oxidative Stress-Mediated Proliferation Via JNK-FOXO3a-Catalase Signaling in Transplanted Adult Stem Cells Promotes Wound Tissue Regeneration. Antioxid Redox Signal 2018; 28:1047-1065. [PMID: 28826225 DOI: 10.1089/ars.2016.6974] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Aims: Stem cells exposed to pathological levels of reactive oxygen species (ROS) at wound sites fail to regenerate tissue. The molecular mechanism underlying differential levels of ROS-mediated regulation of stem cells remains elusive. This study elucidates the mechanistic role of catalase at 10 μM H2O2-induced proliferation of mouse bone marrow stromal (BMSC) and hematopoietic (HSPC) stem/progenitor cells. Results: BMSCs and HSPCs depicted an increased growth rate and colony formation, in the presence of 10 μM but not 100 μM concentration of H2O2, an effect that was perturbed by Vit. C. Mechanistically, JNK activation-FOXO3a nuclear translocation and binding of FOXO3a to catalase promoter at 10 μM H2O2 led to an increased expression and activity of anti-oxidant gene, catalase. This was followed by an increased proliferative phenotype via the AKT-dependent pathway that was perturbed in the presence of catalase-inhibitor, 3-aminotriazole due to an increased ROS-mediated inactivation of AKT. Preclinically, 10 μM H2O2-mediated preconditioning of BMSCs/HSPCs transplantation accelerated wound closure, enhanced catalase expression, and decreased ROS levels at the wound site. Transplantation of male donor cells into female recipient mice or GFP-labeled BMSCs or HSPCs depicted an increased engraftment and proliferation in preconditioned cell transplanted groups as compared with the wound control. Wound healing occurred via keratinocyte generation and vascularization in preconditioned BMSCs, whereas only neo-vascularization occurred in the preconditioned HSPCs transplanted groups. Innovation and Conclusion: Our study suggests a distinct role of catalase that protects BMSCs and HSPCs from low ROS and promotes proliferation. Transplantation of preconditioned stem cells enhanced wound tissue regeneration with a better antioxidant defense mechanism-as a therapeutic approach in stem cell transplantation-mediated tissue regeneration. Antioxid. Redox Signal. 28, 1047-1065.
Collapse
Affiliation(s)
- Neha R Dhoke
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Ramasatyaveni Geesala
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Amitava Das
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| |
Collapse
|
21
|
Design, Docking, and Synthesis of Quinoline-2H
-1,2,4-triazol-3(4H
)-ones as Potent Anticancer and Antitubercular Agents. ChemistrySelect 2018. [DOI: 10.1002/slct.201702279] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
22
|
Kallepu S, Kavitha M, Yeeravalli R, Manupati K, Jadav SS, Das A, Mainkar PS, Chandrasekhar S. Total Synthesis of Desmethyl Jahanyne and Its Lipo-Tetrapeptide Conjugates Derived from Parent Skeleton as BCL-2-Mediated Apoptosis-Inducing Agents. ACS OMEGA 2018; 3:63-75. [PMID: 30023766 PMCID: PMC6045489 DOI: 10.1021/acsomega.7b01634] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 12/21/2017] [Indexed: 05/25/2023]
Abstract
The total synthesis of highly potent and scarcely available marine natural product (-)-jahanyne was attempted resulting in a solution-phase synthesis of pruned versions with comparable activity. A simple and facile synthetic route was employed for the preparation of pruned congeners and would be scalable. The lipophilic tail of the natural product was synthesized from R-(+)-citronellol, utilizing easily available chemicals. All the synthesized compounds were screened for apoptotic activity against a panel of cell lines. These compounds depicted marked binding to B cell lymphoma 2 till 50 °C in cellular thermal shift analysis.
Collapse
Affiliation(s)
- Shivakrishna Kallepu
- Natural
Products Chemistry Division, Centre for Chemical Biology,
and Medicinal Chemistry
& Biotechnology, CSIR-Indian Institute
of Chemical Technology (CSIR-IICT), Hyderabad 500007, India
| | - Minnapuram Kavitha
- Natural
Products Chemistry Division, Centre for Chemical Biology,
and Medicinal Chemistry
& Biotechnology, CSIR-Indian Institute
of Chemical Technology (CSIR-IICT), Hyderabad 500007, India
- Academy
of Scientific and Innovative Research (AcSIR), New Delhi 110020, India
| | - Ragini Yeeravalli
- Natural
Products Chemistry Division, Centre for Chemical Biology,
and Medicinal Chemistry
& Biotechnology, CSIR-Indian Institute
of Chemical Technology (CSIR-IICT), Hyderabad 500007, India
- Academy
of Scientific and Innovative Research (AcSIR), New Delhi 110020, India
| | - Kanakaraju Manupati
- Natural
Products Chemistry Division, Centre for Chemical Biology,
and Medicinal Chemistry
& Biotechnology, CSIR-Indian Institute
of Chemical Technology (CSIR-IICT), Hyderabad 500007, India
- Academy
of Scientific and Innovative Research (AcSIR), New Delhi 110020, India
| | - Surender Singh Jadav
- Natural
Products Chemistry Division, Centre for Chemical Biology,
and Medicinal Chemistry
& Biotechnology, CSIR-Indian Institute
of Chemical Technology (CSIR-IICT), Hyderabad 500007, India
| | - Amitava Das
- Natural
Products Chemistry Division, Centre for Chemical Biology,
and Medicinal Chemistry
& Biotechnology, CSIR-Indian Institute
of Chemical Technology (CSIR-IICT), Hyderabad 500007, India
- Academy
of Scientific and Innovative Research (AcSIR), New Delhi 110020, India
| | - Prathama S. Mainkar
- Natural
Products Chemistry Division, Centre for Chemical Biology,
and Medicinal Chemistry
& Biotechnology, CSIR-Indian Institute
of Chemical Technology (CSIR-IICT), Hyderabad 500007, India
- Academy
of Scientific and Innovative Research (AcSIR), New Delhi 110020, India
| | - Srivari Chandrasekhar
- Natural
Products Chemistry Division, Centre for Chemical Biology,
and Medicinal Chemistry
& Biotechnology, CSIR-Indian Institute
of Chemical Technology (CSIR-IICT), Hyderabad 500007, India
- Academy
of Scientific and Innovative Research (AcSIR), New Delhi 110020, India
| |
Collapse
|
23
|
Structure-Activity Relationship Studies of β-Lactam-azide Analogues as Orally Active Antitumor Agents Targeting the Tubulin Colchicine Site. Sci Rep 2017; 7:12788. [PMID: 28986548 PMCID: PMC5630639 DOI: 10.1038/s41598-017-12912-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 09/12/2017] [Indexed: 11/29/2022] Open
Abstract
We have synthesized a series of new β-lactam-azide derivatives as orally active anti-tumor agents by targeting tubulin colchicine binding site and examined their structure activity relationship (SAR). Among them, compound 28 exhibited the most potent antiproliferative activity against MGC-803 cells with an IC50 value of 0.106 μM by induction of G2/M arrest and apoptosis and inhibition of the epithelial to mesenchymal transition. 28 acted as a novel inhibitor of tubulin polymerization by its binding to the colchicine site. SAR analysis revealed that a hydrogen atom at the C-3 position of the β-lactam was required for the potent antiproliferative activity of β-lactam-azide derivatives. Oral administration of compound 28 also effectively inhibited MGC-803 xenograft tumor growth in vivo in nude mice without causing significant loss of body weight. These results suggested that compound 28 is a promising orally active anticancer agent with potential for development of further clinical applications.
Collapse
|
24
|
Wu YC, Luo SH, Mei WJ, Cao L, Wu HQ, Wang ZY. Synthesis and biological evaluation of 4-biphenylamino-5-halo-2( 5H )-furanones as potential anticancer agents. Eur J Med Chem 2017; 139:84-94. [DOI: 10.1016/j.ejmech.2017.08.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 06/15/2017] [Accepted: 08/02/2017] [Indexed: 10/19/2022]
|
25
|
Bikshapathi R, Sai Prathima P, Yashwanth B, Rajesh P, Rao JV, Jagadeesh Kumar G, Jagadeesh N, Rao VJ. An expeditious protocol for synthesis of Baylis–Hillman derived piperazine derivatives and evaluation of their AChE inhibition. RESEARCH ON CHEMICAL INTERMEDIATES 2017. [DOI: 10.1007/s11164-017-3119-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
26
|
Reddy TN, Ravinder M, Bikshapathi R, Sujitha P, Kumar CG, Rao VJ. Design, synthesis, and biological evaluation of 4-H pyran derivatives as antimicrobial and anticancer agents. Med Chem Res 2017. [DOI: 10.1007/s00044-017-1982-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
27
|
Manupati K, Dhoke NR, Debnath T, Yeeravalli R, Guguloth K, Saeidpour S, De UC, Debnath S, Das A. Inhibiting epidermal growth factor receptor signalling potentiates mesenchymal–epithelial transition of breast cancer stem cells and their responsiveness to anticancer drugs. FEBS J 2017; 284:1830-1854. [DOI: 10.1111/febs.14084] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/27/2017] [Accepted: 04/07/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Kanakaraju Manupati
- Centre for Chemical Biology CSIR‐Indian Institute of Chemical Technology Hyderabad India
- Academy of Scientific & Innovative Research New Delhi India
| | - Neha R. Dhoke
- Centre for Chemical Biology CSIR‐Indian Institute of Chemical Technology Hyderabad India
- Academy of Scientific & Innovative Research New Delhi India
| | - Tanusree Debnath
- Department of Chemistry Maharaja Bir Bikram College Agartala Tripura India
| | - Ragini Yeeravalli
- Centre for Chemical Biology CSIR‐Indian Institute of Chemical Technology Hyderabad India
- Academy of Scientific & Innovative Research New Delhi India
| | - Kalpana Guguloth
- Centre for Chemical Biology CSIR‐Indian Institute of Chemical Technology Hyderabad India
| | - Shahrzad Saeidpour
- Centre for Chemical Biology CSIR‐Indian Institute of Chemical Technology Hyderabad India
| | - Utpal Chandra De
- Department of Chemistry Tripura University Agartala Tripura India
| | - Sudhan Debnath
- Department of Chemistry Maharaja Bir Bikram College Agartala Tripura India
| | - Amitava Das
- Centre for Chemical Biology CSIR‐Indian Institute of Chemical Technology Hyderabad India
- Academy of Scientific & Innovative Research New Delhi India
| |
Collapse
|
28
|
Geesala R, Dhoke NR, Das A. Cox-2 inhibition potentiates mouse bone marrow stem cell engraftment and differentiation-mediated wound repair. Cytotherapy 2017; 19:756-770. [PMID: 28433514 DOI: 10.1016/j.jcyt.2017.03.072] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/18/2017] [Accepted: 03/20/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND Engraftment of transplanted stem cells is often limited by cytokine and noncytokine proinflammatory mediators at the injury site. We examined the role of Cyclooxygenase-2 (Cox-2)-induced cytokine-mediated inflammation on engraftment of transplanted bone marrow stem cells (BMSCs) at the wound site. METHODS BMSCs isolated from male C57/BL6J mice were transplanted onto excisional splinting wounds in syngenic females in presence or absence of celecoxib, Cox-2 specific inhibitor (50 mg/kg, body weight [b wt]), to evaluate engraftment and wound closure. Inflammatory cell infiltration and temporal expression of inflammatory cytokines at the wound bed were determined using immunohistochemical and quantitative-real time polymerase chain reaction (qPCR) analysis, respectively. Mechanistic studies were performed on a murine macrophage cell line (J774.2) to evaluate the effect of interleukin (IL)-17A. RESULTS Celecoxib administration led to a significantly high percent of wound closure, cellular proliferation, collagen deposition, BMSCs engraftment and re-epithelialization at the wound site. Interestingly, recruitment of CD4+T cells and F4/80+ macrophages as well as BMSC transplantation induced up-regulation of Cox-2 and IL-17A gene expression levels were reverted by celecoxib administration. Exogenous supplementation of recombinant interleukin (rIL)-17 to J774.2 cells significantly increased proliferation and gene expression of cytokines -IL-1β, IL-6, IL-8, IL-18 and tumor necrosis factor (TNF)-α via nuclear translocation of nuclear factor kappa B (NFκB)p65/50 subunit. Conditioned media of rIL-17 treated J774.2 cells when supplemented to BMSCs depicted a dose-dependent increase in the number of apoptotic cells and proapoptotic protein expression that was perturbed by celecoxib or IL-17 neutralizing antibody. Finally, celecoxib led to a dose-dependent increase in BMSC differentiation into keratinocyte-like cells in vitro. CONCLUSION Celecoxib protects transplanted BMSCs from Cox-2/IL-17-induced inflammation and increases their engraftment, differentiation into keratinocytes and re-epithelialization thereby potentiating wound tissue repair.
Collapse
Affiliation(s)
- Ramasatyaveni Geesala
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India; Academy of Scientific & Innovative Research, New Delhi, India
| | - Neha R Dhoke
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India; Academy of Scientific & Innovative Research, New Delhi, India
| | - Amitava Das
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India; Academy of Scientific & Innovative Research, New Delhi, India.
| |
Collapse
|
29
|
Alegaon S, Parchure P, Araujo L, Salve P, Alagawadi K, Jalalpure S, Kumbar V. Quinoline-azetidinone hybrids: Synthesis and in vitro antiproliferation activity against Hep G2 and Hep 3B human cell lines. Bioorg Med Chem Lett 2017; 27:1566-1571. [DOI: 10.1016/j.bmcl.2017.02.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 01/31/2017] [Accepted: 02/15/2017] [Indexed: 01/24/2023]
|
30
|
Majee D, Biswas S, Mobin SM, Samanta S. Domino reaction of cyclic sulfamidate imines with Morita–Baylis–Hillman acetates promoted by DABCO: a metal-free approach to functionalized nicotinic acid derivatives. Org Biomol Chem 2017; 15:3286-3297. [DOI: 10.1039/c7ob00240h] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A series of 4,6-diarylnicotinates have been prepared in good to excellent yields via a domino reaction of cyclic sulfamidate imines with MBH acetates in 2-MeTHF promoted by DABCO under an O2 atmosphere.
Collapse
Affiliation(s)
- Debashis Majee
- Discipline of Chemistry
- Indian Institute of Technology Indore
- Simrol
- India
| | - Soumen Biswas
- Discipline of Chemistry
- Indian Institute of Technology Indore
- Simrol
- India
| | - Shaikh M. Mobin
- Discipline of Chemistry
- Indian Institute of Technology Indore
- Simrol
- India
| | - Sampak Samanta
- Discipline of Chemistry
- Indian Institute of Technology Indore
- Simrol
- India
| |
Collapse
|