1
|
Bushi A, Ma Y, Adu-Amankwaah J, Wang R, Cui F, Xiao R, Zhao J, Yuan J, Tan R. G protein-coupled estrogen receptor biased signaling in health and disease. Pharmacol Ther 2025; 269:108822. [PMID: 39978643 DOI: 10.1016/j.pharmthera.2025.108822] [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: 08/30/2024] [Revised: 02/03/2025] [Accepted: 02/12/2025] [Indexed: 02/22/2025]
Abstract
G protein-coupled estrogen receptor (GPER) is now recognized for its pivotal role in cellular signaling, influencing diverse physiological processes and disease states. Unlike classical estrogen receptors, GPER exhibits biased signaling, wherein ligand binding triggers selective pathways over others, significantly impacting cellular responses. This review explores the nuanced mechanisms of biased signaling mediated by GPER, underscoring its relevance in cardiovascular health, neurological function, immune modulation, and oncogenic processes. Despite its critical implications, biased signaling through GPER remains underexplored compared to traditional signaling paradigms. We explore recent progress in understanding GPER signaling specificity and its potential therapeutic implications across various diseases. Future research directions aim to uncover the molecular basis of biased signaling, develop selective ligands, and translate these insights into personalized therapeutic approaches. Exploiting the therapeutic potential of GPER biased signaling represents a promising frontier in precision medicine, offering innovative strategies to address unmet medical needs.
Collapse
Affiliation(s)
- Aisha Bushi
- School international education, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yixuan Ma
- First Clinical Medical School, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Joseph Adu-Amankwaah
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Rong Wang
- The second clinical college, China Medical University, Shenyang, Liaoning 110122, China
| | - Fen Cui
- Research Institution of Behavioral Medicine Education, Jining Medical University, Jining 272067, China
| | - Rui Xiao
- Second Clinical Medical School, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jinming Zhao
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang 110122, China; Department of Pathology, The First Hospital of China Medical University, Shenyang 110001, China.
| | - Jinxiang Yuan
- Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, Shandong, China.
| | - Rubin Tan
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
2
|
Chun D, Lee SB, Chun S, Choi SH, Hong J, Lee H, Lee J, Hong S. Synthesis of N-heterocyclic compounds using N, N-dimethylacetamides as an electrophilic carbon source. Org Biomol Chem 2024; 22:8617-8624. [PMID: 39360784 DOI: 10.1039/d4ob01417k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
In this study, N-heterocyclic compounds were synthesized using nitrogen-containing nucleophilic substrates and electrophilic carbon sources derived from N,N-dimethylacetamide (DMAc). Depending on the nucleophilic groups, N-heterocyclic compounds such as 4-quinazolinones, pyrrole-quinoxalines, and dihydro-benzothiadiazine dioxides were produced. Carbon, adjacent to the nitrogen in DMAc, was activated in the presence of FeCl3·6H2O and di-t-butyl peroxide (DTBP). This procedure was considered an economical synthetic method because it utilized iron catalysts and DMAc as an electrophilic carbon source and a solvent.
Collapse
Affiliation(s)
- Dayoung Chun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
| | - Seok Beom Lee
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Simin Chun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
| | - Seung Hyun Choi
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
| | - Junhwa Hong
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
| | - Honghui Lee
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
| | - Jinwoo Lee
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
| | - Suckchang Hong
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
3
|
Żabińska M, Wiśniewska K, Węgrzyn G, Pierzynowska K. Exploring the physiological role of the G protein-coupled estrogen receptor (GPER) and its associations with human diseases. Psychoneuroendocrinology 2024; 166:107070. [PMID: 38733757 DOI: 10.1016/j.psyneuen.2024.107070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/15/2024] [Accepted: 05/05/2024] [Indexed: 05/13/2024]
Abstract
Estrogen is a group of hormones that collaborate with the nervous system to impact the overall well-being of all genders. It influences many processes, including those occurring in the central nervous system, affecting learning and memory, and playing roles in neurodegenerative diseases and mental disorders. The hormone's action is mediated by specific receptors. Significant roles of classical estrogen receptors, ERα and ERβ, in various diseases were known since many years, but after identifying a structurally and locationally distinct receptor, the G protein-coupled estrogen receptor (GPER), its role in human physiology and pathophysiology was investigated. This review compiles GPER-related information, highlighting its impact on homeostasis and diseases, while putting special attention on functions and dysfunctions of this receptor in neurobiology and biobehavioral processes. Understanding the receptor modulation possibilities is essential for therapy, as disruptions in receptors can lead to diseases or disorders, irrespective of correct estrogen levels. We conclude that studies on the GPER receptor have the potential to develop therapies that regulate estrogen and positively impact human health.
Collapse
Affiliation(s)
- Magdalena Żabińska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, Gdansk 80-308, Poland
| | - Karolina Wiśniewska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, Gdansk 80-308, Poland
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, Gdansk 80-308, Poland
| | - Karolina Pierzynowska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, Gdansk 80-308, Poland.
| |
Collapse
|
4
|
Abbas MA, Al-Kabariti AY, Sutton C. Comprehensive understanding of the role of GPER in estrogen receptor-alpha negative breast cancer. J Steroid Biochem Mol Biol 2024; 241:106523. [PMID: 38636681 DOI: 10.1016/j.jsbmb.2024.106523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 11/01/2023] [Accepted: 04/14/2024] [Indexed: 04/20/2024]
Abstract
G protein-coupled estrogen receptor (GPER) plays a prominent role in facilitating the rapid, non-genomic signaling of estrogens in breast cancer cells. Herein, a comprehensive overview of the role of GPER in ER-ɑ-negative breast cancer is provided. Activation of GPER affected proliferation, metastasis and epithelial mesenchymal transition in ER-ɑ negative breast cancer cells. Clinical studies have demonstrated that GPER positivity was strongly correlated with larger tumor size and advanced clinical stage, suggesting that GPER/ERK signaling may play a role in promoting tumor progression. Strong evidence existed that environmental contaminants like bisphenol A have a carcinogenic potential mediated by GPER activation. The complexity of the cross talk between GPER and other receptors including ER-β, ER-α36, Estrogen-related receptor α (ERRα) and androgen receptor has been discussed. The potential utility of small molecules and phytoestrogens targeting GPER, adds valuable insights into its therapeutic potential. This review holds promises in advancing our understanding of GPER role in ER-ɑ-negative breast cancer. Overall, the consequences of GPER activation are still an area of active research and the implication are not entirely clear.
Collapse
Affiliation(s)
- Manal A Abbas
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan; Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Aya Y Al-Kabariti
- Department of Biopharmaceutics and Clinical Pharmacy, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan; Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Amman 19328, Jordan.
| | - Chris Sutton
- School of Chemistry and Biosciences, University of Bradford, Bradford BD7 1DP, UK
| |
Collapse
|
5
|
Giannessi L, Lupo MG, Rossi I, Martina MG, Vilella A, Bodria M, Giuliani D, Zimetti F, Zanotti I, Potì F, Bernini F, Ferri N, Radi M. Identification of 4-amino-2-Pyridones as new potent PCSK9 inhibitors: From phenotypic hit discovery to in vivo tolerability. Eur J Med Chem 2024; 265:116063. [PMID: 38160616 DOI: 10.1016/j.ejmech.2023.116063] [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: 06/09/2023] [Revised: 12/15/2023] [Accepted: 12/15/2023] [Indexed: 01/03/2024]
Abstract
Among the strategies to overcome the underperformance of statins in cardiovascular diseases (CVDs), the development of drugs targeting the Proprotein Convertase Subtilisin-like Kexin type 9 (PCSK9) is considered one of the most promising. However, only anti-PCSK9 biological drugs have been approved to date, and orally available small-molecules for the treatment of hypercholesterolemic conditions are still missing on the market. In the present work, we describe the application of a phenotypic approach to the identification and optimization of 4-amino-2-pyridone derivatives as a new chemotype with anti-PCSK9 activity. Starting from an in-house collection of compounds, functional assays on HepG2 cells followed by a chemistry-driven hit optimization campaign, led to the potent anti-PCSK9 candidate 5c. This compound, at 5 μM, totally blocked PCSK9 secretion from HepG2 cells, significantly increased LDL receptor (LDLR) expression, and acted cooperatively with simvastatin by reducing its induction of PCSK9 expression. Finally, compound 5c also proved to be well tolerated in C57BL/6J mice at the tested concentration (40 mg/kg) with no sign of toxicity or behavior modifications.
Collapse
Affiliation(s)
- Lisa Giannessi
- Dipartimento di Scienze Degli Alimenti e Del Farmaco (DipALIFAR), Università Degli Studi di Parma, Viale Delle Scienze, 27/A, 43124, Parma, Italy
| | | | - Ilaria Rossi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 351131, Padova, Italy
| | - Maria Grazia Martina
- Dipartimento di Scienze Degli Alimenti e Del Farmaco (DipALIFAR), Università Degli Studi di Parma, Viale Delle Scienze, 27/A, 43124, Parma, Italy
| | - Antonietta Vilella
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125, Modena, Italy
| | - Martina Bodria
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125, Modena, Italy
| | - Daniela Giuliani
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125, Modena, Italy
| | - Francesca Zimetti
- Dipartimento di Scienze Degli Alimenti e Del Farmaco (DipALIFAR), Università Degli Studi di Parma, Viale Delle Scienze, 27/A, 43124, Parma, Italy
| | - Ilaria Zanotti
- Dipartimento di Scienze Degli Alimenti e Del Farmaco (DipALIFAR), Università Degli Studi di Parma, Viale Delle Scienze, 27/A, 43124, Parma, Italy
| | - Francesco Potì
- Department of Medicine and Surgery, Unit of Neuroscience, University of Parma, 43125, Parma, Italy
| | - Franco Bernini
- Dipartimento di Scienze Degli Alimenti e Del Farmaco (DipALIFAR), Università Degli Studi di Parma, Viale Delle Scienze, 27/A, 43124, Parma, Italy
| | - Nicola Ferri
- Department of Medicine, University of Padova, 35128, Padova, Italy; Veneto Institute of Molecular Medicine, Padua, 35129, Italy.
| | - Marco Radi
- Dipartimento di Scienze Degli Alimenti e Del Farmaco (DipALIFAR), Università Degli Studi di Parma, Viale Delle Scienze, 27/A, 43124, Parma, Italy.
| |
Collapse
|
6
|
Barghi Lish A, Foroumadi A, Kolvari E, Safari F. Synthesis and Biological Evaluation of 12-Aryl-11-hydroxy-5,6-dihydropyrrolo[2″,1″:3',4']pyrazino[1',2':1,5]pyrrolo[2,3- d]pyridazine-8(9 H)-one Derivatives as Potential Cytotoxic Agents. ACS OMEGA 2023; 8:42212-42224. [PMID: 38024677 PMCID: PMC10653054 DOI: 10.1021/acsomega.3c04167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023]
Abstract
In the present paper, a facile and efficient synthetic procedure has been applied to obtain dihydrodipyrrolo[1,2-a:2',1'-c]pyrazine-2,3-dicarboxylates (5a-s), which have subsequently gone through the cyclization in the presence of hydrazine hydrate to afford 12-aryl-11-hydroxy-5,6-dihydropyrrolo[2″,1″:3',4']pyrazino[1',2':1,5]pyrrolo[2,3-d]pyridazine-8(9H)-ones (7a-q). The molecular structures of these novel compounds were extensively examined through the analysis of spectroscopic data in combination with X-ray crystallography techniques. Following that, the in vitro cytotoxic activities of all derivatives against three human cancer cell lines (Panc-1, PC3, and MDA-MB-231) were comprehensively evaluated alongside the assessment on normal human dermal fibroblast (HDF) cells using the MTT assay. Among the compounds, the 3-nitrophenyl derivative (7m) from the second series showed the best antiproliferative activity against all tested cell lines, particularly against Panc-1 cell line, (IC50 = 12.54 μM), being nearly twice as potent as the standard drug etoposide. The induction of apoptosis and sub-G1 cell cycle arrest in Panc-1 cancer cells by compound 7m was confirmed through further assessment. Moreover, the inhibition of kinases and the induction of cellular apoptosis by compound 7m in Panc-1 cancer cells were validated using the Western blotting assay.
Collapse
Affiliation(s)
- Azam Barghi Lish
- Department
of Chemistry, Semnan University, Semnan 35351-19111, Iran
| | - Alireza Foroumadi
- Department
of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran1417614411, Iran
- Drug
Design and Development Research Center, The Institute of Pharmaceutical
Sciences (TIPS), Tehran University of Medical
Sciences, Tehran 1417614411, Iran
| | - Eskandar Kolvari
- Department
of Chemistry, Semnan University, Semnan 35351-19111, Iran
| | - Fatemeh Safari
- Department
of Biology, Faculty of Science, University
of Guilan, Rasht 4193833697, Iran
| |
Collapse
|
7
|
Hatami M, Kouchak M, Kheirollah A, Khorsandi L, Rashidi M. Quercetin-loaded solid lipid nanoparticles exhibit antitumor activity and suppress the proliferation of triple-negative MDA-MB 231 breast cancer cells: implications for invasive breast cancer treatment. Mol Biol Rep 2023; 50:9417-9430. [PMID: 37831347 DOI: 10.1007/s11033-023-08848-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 09/26/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUND Quercetin (QC) is a naturally occurring flavonoid found in abundance in fruits and vegetables. Its anti-cancer and anti-inflammatory properties have been previously demonstrated, but its low bioavailability hampers its clinical use. Triple-negative breast cancer is a subtype of breast cancer with a poor response to chemotherapy. This study investigates the anti-cancer effects of quercetin-solid lipid nanoparticles (QC-SLN) on the triple-negative breast cancer cell line MDA-MB231. MATERIALS AND METHODS MCF-7 and MDA-MB231 cells were treated with 18.9 µM of QC and QC-SLN for 48 h. Cell viability, apoptosis, colony formation assay, and the anti-angiogenic effects of the treatment were evaluated. RESULTS QC-SLN displayed optimal properties (particle size of 154 nm, zeta potential of -27.7 mV, encapsulation efficiency of 99.6%, and drug loading of 1.81%) and exhibited sustained release of QC over 72 h. Compared to the QC group, the QC-SLN group showed a significant decrease in cell viability, colony formation, angiogenesis, and a substantial increase in apoptosis through the modulation of Bax and Bcl-2 at both gene and protein levels. The augmentation in the proportion of cleaved-to-pro caspases 3 and 9, as well as poly (ADP-ribose) polymerase (PARP), under the influence of QC-SLN, was conspicuously observed in both cancer cell lines. CONCLUSIONS This study showcases quercetin-solid lipid nanoparticles (QC-SLN) as a promising therapy for triple-negative breast cancer. The optimized QC-SLN formulation improved physicochemical properties and sustained quercetin release, resulting in reduced cell viability, colony formation, angiogenesis, and increased apoptosis in the MDA-MB231 cell line. These effects were driven by modulating Bax and Bcl-2 expression, activating caspases 3 and 9, and poly (ADP-ribose) polymerase (PARP). Further in vivo studies are needed to confirm QC-SLN's efficacy and safety.
Collapse
Affiliation(s)
- Mahdi Hatami
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institution, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Kouchak
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Alireza Kheirollah
- Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Layasadat Khorsandi
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institution, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mojtaba Rashidi
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institution, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
8
|
Damai M, Guzzardi N, Lewis V, Rao ZX, Sykes D, Patel B. Crafting mono- and novel bis-methylated pyrroloquinoxaline derivatives from a shared precursor and its application in the total synthesis of marinoquinoline A. RSC Adv 2023; 13:29561-29567. [PMID: 37822662 PMCID: PMC10562898 DOI: 10.1039/d3ra05952a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/03/2023] [Indexed: 10/13/2023] Open
Abstract
The synthesis of mono- and novel bis-methylated pyrrolo[1,2-a]quinoxalines through the addition of unstable methyl radicals to aryl isocyanides is described contingent upon the reaction conditions employed. The strategy has been effectively employed in the total synthesis of the natural product marinoquinoline A.
Collapse
Affiliation(s)
- Margarita Damai
- School of Human Sciences, London Metropolitan University 166-220 Holloway Road London N7 8DB UK
| | - Norman Guzzardi
- School of Human Sciences, London Metropolitan University 166-220 Holloway Road London N7 8DB UK
| | - Viliyana Lewis
- School of Human Sciences, London Metropolitan University 166-220 Holloway Road London N7 8DB UK
| | - Zenobia X Rao
- School of Human Sciences, London Metropolitan University 166-220 Holloway Road London N7 8DB UK
| | - Daniel Sykes
- School of Human Sciences, London Metropolitan University 166-220 Holloway Road London N7 8DB UK
| | - Bhaven Patel
- School of Human Sciences, London Metropolitan University 166-220 Holloway Road London N7 8DB UK
| |
Collapse
|
9
|
Carullo G, Mazzotta S, Ceramella J, Iacopetta D, Ramunno A, Rosano C, Brizzi A, Campiani G, Aiello F, Sinicropi MS. Development of 1-(2-aminophenyl)pyrrole-based amides acting as human topoisomerase I inhibitors. Arch Pharm (Weinheim) 2023; 356:e2300270. [PMID: 37452410 DOI: 10.1002/ardp.202300270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023]
Abstract
Topoisomerases are ubiquitous enzymes in the human body, particularly involved in cancer development and progression. Topoisomerase I (topoI) performs DNA relaxation reactions by "controlled rotation" rather than by "strand passage." The inhibition of topoI has become a useful strategy to control cancer cell proliferation. Nowadays, different compounds have undergone clinical trials, but the search for new molecular entities is necessary and benefits from medicinal chemistry efforts. Pyrrole-based compounds emerged as promising antiproliferative agents, with particular interest in breast cancer therapy and topoI inhibition. Starting from these observations and based on the scaffold-hopping approach, we developed a small library of 1-(2-aminophenyl)pyrrole-based amides (7a-f) as new anticancer agents. Tested on a panel of cancer cell lines, 7a-f displayed the most interesting profile in MDA-MB-231 cells, where the most active compounds, 7d-f, were able to induce death by apoptosis. Direct enzymatic assays and docking simulations on the topoI active site (PDB: 1A35) revealed the inhibitory activity and potential binding site for the newly developed 1-(2-aminophenyl)pyrrole-based amides.
Collapse
Affiliation(s)
- Gabriele Carullo
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Siena, Italy
| | - Sarah Mazzotta
- Dipartimento di Chimica, Università degli Studi di Milano, Milano, Italy
| | - Jessica Ceramella
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione, Università della Calabria, Rende, Italy
| | - Domenico Iacopetta
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione, Università della Calabria, Rende, Italy
| | - Anna Ramunno
- Dipartimento di Farmacia, Università degli Studi di Salerno, Fisciano, Italy
| | - Camillo Rosano
- Unità di Proteomica e Spettrometria di Massa, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Antonella Brizzi
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Siena, Italy
| | - Giuseppe Campiani
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Siena, Italy
| | - Francesca Aiello
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione, Università della Calabria, Rende, Italy
| | - Maria S Sinicropi
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione, Università della Calabria, Rende, Italy
| |
Collapse
|
10
|
Carullo G, Di Bonaventura G, Rossi S, Lupetti V, Tudino V, Brogi S, Butini S, Campiani G, Gemma S, Pompilio A. Development of Quinazolinone Derivatives as Modulators of Virulence Factors of Pseudomonas aeruginosa Cystic Fibrosis Strains. Molecules 2023; 28:6535. [PMID: 37764311 PMCID: PMC10536951 DOI: 10.3390/molecules28186535] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Pseudomonas aeruginosa (PA), one of the ESKAPE pathogens, is an opportunistic Gram-negative bacterium responsible for nosocomial infections in humans but also for infections in patients affected by AIDS, cancer, or cystic fibrosis (CF). Treatment of PA infections in CF patients is a global healthcare problem due to the ability of PA to gain antibiotic tolerance through biofilm formation. Anti-virulence compounds represent a promising approach as adjuvant therapy, which could reduce or eliminate the pathogenicity of PA without impacting its growth. Pyocyanin is one of the virulence factors whose production is modulated by the Pseudomonas quinolone signal (PQS) through its receptor PqsR. Different PqsR modulators have been synthesized over the years, highlighting this new powerful therapeutic strategy. Based on the promising structure of quinazolin-4(3H)-one, we developed compounds 7a-d, 8a,b, 9, 10, and 11a-f able to reduce biofilm formation and the production of virulence factors (pyocyanin and pyoverdine) at 50 µM in two PA strains responsible for CF acute and chronic infections. The developed compounds did not reduce the cell viability of IB3-1 bronchial CF cells, and computational studies confirmed the potential ability of novel compounds to act as potential Pqs system modulators.
Collapse
Affiliation(s)
- Gabriele Carullo
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (G.C.); (S.R.); (V.T.); (S.B.); (G.C.)
| | - Giovanni Di Bonaventura
- Department of Medical, Oral and Biotechnological Sciences, School of Medicine and Health Sciences “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (G.D.B.); (V.L.); (A.P.)
| | - Sara Rossi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (G.C.); (S.R.); (V.T.); (S.B.); (G.C.)
| | - Veronica Lupetti
- Department of Medical, Oral and Biotechnological Sciences, School of Medicine and Health Sciences “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (G.D.B.); (V.L.); (A.P.)
| | - Valeria Tudino
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (G.C.); (S.R.); (V.T.); (S.B.); (G.C.)
| | - Simone Brogi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy;
| | - Stefania Butini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (G.C.); (S.R.); (V.T.); (S.B.); (G.C.)
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (G.C.); (S.R.); (V.T.); (S.B.); (G.C.)
| | - Sandra Gemma
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (G.C.); (S.R.); (V.T.); (S.B.); (G.C.)
| | - Arianna Pompilio
- Department of Medical, Oral and Biotechnological Sciences, School of Medicine and Health Sciences “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (G.D.B.); (V.L.); (A.P.)
| |
Collapse
|
11
|
Arterburn JB, Prossnitz ER. G Protein-Coupled Estrogen Receptor GPER: Molecular Pharmacology and Therapeutic Applications. Annu Rev Pharmacol Toxicol 2023; 63:295-320. [PMID: 36662583 PMCID: PMC10153636 DOI: 10.1146/annurev-pharmtox-031122-121944] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The actions of estrogens and related estrogenic molecules are complex and multifaceted in both sexes. A wide array of natural, synthetic, and therapeutic molecules target pathways that produce and respond to estrogens. Multiple receptors promulgate these responses, including the classical estrogen receptors of the nuclear hormone receptor family (estrogen receptors α and β), which function largely as ligand-activated transcription factors, and the 7-transmembrane G protein-coupled estrogen receptor, GPER, which activates a diverse array of signaling pathways. The pharmacology and functional roles of GPER in physiology and disease reveal important roles in responses to both natural and synthetic estrogenic compounds in numerous physiological systems. These functions have implications in the treatment of myriad disease states, including cancer, cardiovascular diseases, and metabolic disorders. This review focuses on the complex pharmacology of GPER and summarizes major physiological functions of GPER and the therapeutic implications and ongoing applications of GPER-targeted compounds.
Collapse
Affiliation(s)
- Jeffrey B Arterburn
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, New Mexico, USA
- University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA;
| | - Eric R Prossnitz
- University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA;
- Center of Biomedical Research Excellence in Autophagy, Inflammation and Metabolism, and Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| |
Collapse
|
12
|
Wang C, Zhang Y, Zhang T, Shi L, Geng Z, Xing D. Proteolysis-targeting chimaeras (PROTACs) as pharmacological tools and therapeutic agents: advances and future challenges. J Enzyme Inhib Med Chem 2022; 37:1667-1693. [PMID: 35702041 PMCID: PMC9225776 DOI: 10.1080/14756366.2022.2076675] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Proteolysis-targeting chimaeras (PROTACs) have been developed to be an emerging technology for targeted protein degradation and attracted the favour of academic institutions, large pharmaceutical enterprises, and biotechnology companies. The mechanism is based on the inhibition of protein function by hijacking a ubiquitin E3 ligase for protein degradation. The heterobifunctional PROTACs contain a ligand for recruiting an E3 ligase, a linker, and another ligand to bind with the protein targeted for degradation. To date, PROTACs targeting ∼70 proteins, many of which are clinically validated drug targets, have been successfully developed with several in clinical trials for diseases therapy. In this review, the recent advances in PROTACs against clinically validated drug targets are summarised and the chemical structure, cellular and in vivo activity, pharmacokinetics, and pharmacodynamics of these PROTACs are highlighted. In addition, the potential advantages, challenges, and prospects of PROTACs technology in disease treatment are discussed.
Collapse
Affiliation(s)
- Chao Wang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, China
| | - Yujing Zhang
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao University, Qingdao, China.,School of Pharmacy, Qingdao University, Qingdao, China
| | - Tingting Zhang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, China
| | - Lingyu Shi
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, China
| | - Zhongmin Geng
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, China
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, China.,School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
13
|
Chen Y, He C, Chen J, Zhang G, Yu Y. Palladium(II)-catalyzed tandem reaction for the assembly of 1,3-disubstituted pyrrolo[1,2-α]pyrazines. SYNTHETIC COMMUN 2022. [DOI: 10.1080/00397911.2022.2112605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Affiliation(s)
- Yitong Chen
- Zhejiang Province Key Laboratory of Anti-cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P.R. of China
| | - Chang He
- Zhejiang Province Key Laboratory of Anti-cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P.R. of China
| | - Jun Chen
- Department of Pharmacy, Yongkang First People’s Hospital Affiliated to Hangzhou Medical College, Yongkang, P.R. of China
| | - Guolin Zhang
- Zhejiang Province Key Laboratory of Anti-cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P.R. of China
| | - Yongping Yu
- Zhejiang Province Key Laboratory of Anti-cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P.R. of China
| |
Collapse
|
14
|
Samanta SK, Sarkar R, Sengupta U, Das S, Ganguly D, Hasija A, Chopra D, Bera MK. A direct entry to polycyclic quinoxaline derivatives via I 2-DMSO mediated oxidative decarboxylation of α-amino acids and the subsequent Pictet-Spengler cyclization reaction. Org Biomol Chem 2022; 20:4650-4658. [PMID: 35612282 DOI: 10.1039/d2ob00503d] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
A facile and highly efficient iodine-promoted strategy has been delineated for the synthesis of indolo and pyrrolo[1,2-a]quinoxaline derivatives via an oxidative Pictet-Spengler type amino cyclo-annulation reaction using ∝-amino acids as aldehyde surrogates. The concomitant benzylic oxidation and the compatibility of different starting materials under standard conditions made the current method versatile. The salient features of the protocol such as readily available starting materials, inexpensive promoters, environmental benignity, broad substrate scope, scalability, and good to excellent yield make the method more attractive to practitioners of organic synthesis.
Collapse
Affiliation(s)
- Surya Kanta Samanta
- Department of Chemistry, Indian Institute of Engineering Science and Technology (IIEST), Shibpur, Howrah 711 103, WB, India.
| | - Rumpa Sarkar
- Department of Chemistry, Indian Institute of Engineering Science and Technology (IIEST), Shibpur, Howrah 711 103, WB, India.
| | - Utsav Sengupta
- Department of Chemistry, Indian Institute of Engineering Science and Technology (IIEST), Shibpur, Howrah 711 103, WB, India.
| | - Sayan Das
- Centre for Health Science and Technology, JIS Institute of Advanced Studies and Research, JIS University, Kolkata, India
| | - Debabani Ganguly
- Centre for Health Science and Technology, JIS Institute of Advanced Studies and Research, JIS University, Kolkata, India
| | - Avantika Hasija
- Crystallography and Crystal Chemistry Laboratory, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal 462066, India
| | - Deepak Chopra
- Crystallography and Crystal Chemistry Laboratory, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal 462066, India
| | - Mrinal K Bera
- Department of Chemistry, Indian Institute of Engineering Science and Technology (IIEST), Shibpur, Howrah 711 103, WB, India.
| |
Collapse
|
15
|
Feng LS, Gao C, Liu FW, Wang XP, Zhang ZL. Recent updates on the anticancer activity of quinoxaline hybrids (Jan. 2017-Jan. 2022). Curr Top Med Chem 2022; 22:1426-1441. [DOI: 10.2174/1568026622666220428093955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/18/2022] [Accepted: 02/18/2022] [Indexed: 11/22/2022]
Abstract
Abstract:
Cancer as one of the leading causes of death among non-communicable diseases has already posed a heavy burden on the world health system. Chemotherapy is one of the most effective approaches for cancer treatment, but multidrug resistance, lack of efficacy, and toxic side effects hamper efficacious cancer chemotherapy, creating an urgent need to develop novel, more effective and less toxic anticancer therapeutics. Quinoxalines as fascinating structures constitute an important class of heterocycles in drug discovery. Quinoxaline hybrids could exert anticancer activity through diverse mechanisms and possess profound in vitro and in vivo efficacy against various cancers including multidrug-resistant forms. Thus, quinoxaline hybrids represent useful templates for the control and eradication of cancer. The purpose of the present review article is to provide an emphasis on the recent developments (Jan. 2017-Jan. 2022) in quinoxaline hybrids with insights into their in vitro and in vivo anticancer potential as well as structure-activity relationships (SARs) to facilitate further rational design of more effective candidates.
Collapse
|
16
|
Priyanka HP, Thiyagaraj A, Krithika G, Nair RS, Hopper W, ThyagaRajan S. 17β-Estradiol Concentration and Direct β 2-Adrenoceptor Inhibition Determine Estrogen-Mediated Reversal of Adrenergic Immunosuppression. Ann Neurosci 2022; 29:32-52. [PMID: 35875427 PMCID: PMC9305908 DOI: 10.1177/09727531211070541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 11/29/2021] [Indexed: 11/17/2022] Open
Abstract
Background: Sympathetic innervation of lymphoid organs, and the presence of 17β-estradiol (estrogen or E2) and adrenergic receptors (ARs) on lymphocytes, suggests that sympathetic stimulation and hormonal activation may influence immune functions. Purpose: Modeling and simulating these pathways may help to understand the dynamics of neuroendocrine-immune modulation at the cellular and molecular levels. Methods: Dose- and receptor-dependent effects of E2 and AR subtype-specific agonists were established in vitro on lymphocytes from young male Sprague-Dawley rats and were modeled in silico using the MATLAB Simbiology toolbox. Kinetic principles were assigned to define receptor–ligand dynamics, and concentration/time plots were obtained using Ode15s solvers at different time intervals for key regulatory molecules. Comparisons were drawn between in silico and in vitro data for validating the constructed model with sensitivity analysis of key regulatory molecules to assess their individual impacts on the dynamics of the system. Finally, docking studies were conducted with key ligands E2 and norepinephrine (NE) to understand the mechanistic principles underlying their interactions. Results: Adrenergic activation triggered proapoptotic signals, while E2 enhanced survival signals, showing opposing effects as observed in vitro. Treatment of lymphocytes with E2 shows a 10-fold increase in survival signals in a dose-dependent manner. Cyclic adenosine monophosphate (cAMP) activation is crucial for the activation of survival signals through extracellular signal-regulated kinase (p-ERK) and cAMP responsive element binding (p-CREB) protein. Docking studies showed the direct inhibition of ERK by NE and β2-AR by E2 explaining how estrogen signaling overrides NE-mediated immunosuppression in vitro. Conclusion: The cross-talk between E2 and adrenergic signaling pathways determines lymphocyte functions in a receptor subtype and coactivation-dependent manner in health and disease.
Collapse
Affiliation(s)
- Hannah P. Priyanka
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
- Inspire Lab, Institute of Advanced Research in Health Sciences, Tamil Nadu Government Multi Super Specialty Hospital, Chennai, Tamil Nadu, India
| | - A. Thiyagaraj
- Department of Bioinformatics, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
- Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - G. Krithika
- Centre of Advanced Study in Crystallography and Biophysics, University of Madras Guindy, Campus, Chennai, Tamil Nadu, India
| | - R. S. Nair
- Inspire Lab, Institute of Advanced Research in Health Sciences, Tamil Nadu Government Multi Super Specialty Hospital, Chennai, Tamil Nadu, India
| | - W. Hopper
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
- Department of Bioinformatics, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - S. ThyagaRajan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| |
Collapse
|
17
|
Wang C, Zhang Y, Wang J, Xing D. VHL-based PROTACs as potential therapeutic agents: Recent progress and perspectives. Eur J Med Chem 2022; 227:113906. [PMID: 34656901 DOI: 10.1016/j.ejmech.2021.113906] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 02/06/2023]
Abstract
Proteolysis targeting chimeras (PROTACs), which hijack proteins of interest (POIs) and recruit E3 ligases for target degradation via the ubiquitin-proteasome pathway, are a novel drug discovery paradigm that has been widely used as biological tools and medicinal molecules with the potential of clinical application value. To date, a wide variety of small molecule PROTACs have been developed. Importantly, VHL-based PROTACs have emerged to be a promising approach for proteins, including those non-druggable ones, such as transcriptional factors and scaffold proteins. VHL-based PRTOACs have been developed for the treatment of diseases that are difficult to be dealt with by conventional methods, such as radiotherapy, chemotherapy, and small molecule inhibitors. In this review, the recent advances of VHL-based PRTOACs were summarized, and the chances and challenges associated with this area were also highlighted.
Collapse
Affiliation(s)
- Chao Wang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China.
| | - Yujing Zhang
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao University, Qingdao, 266071, Shandong, China; School of Pharmacy, Qingdao University, Qingdao, 266021, Shandong, China.
| | - Jie Wang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China.
| | - Dongming Xing
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
18
|
Green Synthesis of New Pyrrolo [1,2-a] quinoxalines as Antiproliferative Agents in GPER-expressing Breast Cancer Cells. J CHEM-NY 2021. [DOI: 10.1155/2021/5596816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
4,5-Dihydropyrrolo [1,2-a]quinoxalines are interesting druggable scaffolds, with multifaceted biological properties, including anticancer properties targeting the G protein-coupled estrogen receptor 1 (GPER). In this work, the synthesis and preliminary antiproliferative activity of a small set of new 4,5-dihydropyrrolo[1,2-a]quinoxalines (18-20) and pyrrolo[1,2-a]quinoxalines (21, 22) has been reported, inspired by known antiproliferative agents (G-1, G-15, and G-36). The synthesis of the pyrroloquinoxalinic core was employed following the Pictet–Spengler reaction, using the surfactant p-dodecylbenzene sulphonic acid (p-DBSA), as catalyst. It demonstrated efficiency in the catalysis of the 4-phenylpyrrole [1,2-a] quinoxaline type compound formation in mild solvents such as water, ethanol, and hydroalcoholic solutions. In addition, the reactions proceeded in a short time (between 15 and 120 minutes) at room temperature and with high yields. The in vitro MTT assays showed that the presence of isopropyl groups furnished promising antiproliferative compounds. Although, the acetyl group provided also antiproliferative effects, breaking down its responsibility in the GPER transactivation. Nevertheless, it is possible to conclude that the 4,5-dihydropyrrolo[1,2-a]quinoxalines remain a feasible scaffold to develop anticancer agents against GPER-expressing cells.
Collapse
|
19
|
Selvendran S, Das S, Waidha K, Venkatesan S, Balamurali MM, Basu B, Rajendran S. Pyrrole‐Fused Benzoxazinones/Quinoxalinones: Molecular Dynamic Simulation, Antiproliferative and Antibacterial Activities. ChemistrySelect 2021. [DOI: 10.1002/slct.202103015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Suresh Selvendran
- Chemistry Division School of Advanced Sciences Vellore Institute of Technology Chennai Campus Chennai 600127 Tamilnadu India
| | - Souvik Das
- Department of Neuroendocrinology and Experimental Hematology Chittaranjan National Cancer Institute Kolkata 700 026 West Bengal India
| | - Kamran Waidha
- DRDO-Defence Institute of High Altitude Research (DIHAR), Leh Ladakh, UT 194101 India
| | - Swathi Venkatesan
- Chemistry Division School of Advanced Sciences Vellore Institute of Technology Chennai Campus Chennai 600127 Tamilnadu India
| | - M. M. Balamurali
- Chemistry Division School of Advanced Sciences Vellore Institute of Technology Chennai Campus Chennai 600127 Tamilnadu India
| | - Biswarup Basu
- Department of Neuroendocrinology and Experimental Hematology Chittaranjan National Cancer Institute Kolkata 700 026 West Bengal India
| | - Saravanakumar Rajendran
- Chemistry Division School of Advanced Sciences Vellore Institute of Technology Chennai Campus Chennai 600127 Tamilnadu India
| |
Collapse
|
20
|
Lee JH, Yoon SH, Nam S, Kim I. One-pot three-component coupling access to 1,2-dihydropyrrolo[1,2-a]pyrazine-1-phosphonates: multi-functionalization of a pyrazine unit. Org Biomol Chem 2021; 19:6066-6084. [PMID: 34137765 DOI: 10.1039/d1ob00885d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A new pyrrolo[1,2-a]pyrazine chemical space with a poly-substituted pyrazine unit was readily accessed by Sc(OTf)3-catalyzed one-pot three-component coupling of a pyrrole derivative, amine, and trialkylphosphite under environment-friendly conditions. The formation of multiple bonds (two C-N and one C-P) via a domino process consisting of the chemoselective Kabachnik-Fields reaction and intramolecular cyclodehydration allowed for the construction of highly functionalized pyrazines.
Collapse
Affiliation(s)
- Jeong Hwa Lee
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea.
| | - Seok Hyun Yoon
- Graduate Program of Industrial Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea
| | - Seonghyeon Nam
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea.
| | - Ikyon Kim
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea.
| |
Collapse
|
21
|
Ahn J, Lee SB, Song I, Chun S, Oh DC, Hong S. Synthesis of 4-Aryl Pyrrolo[1,2-α]quinoxalines via Iron-Catalyzed Oxidative Coupling from an Unactivated Methyl Arene. J Org Chem 2021; 86:7390-7402. [PMID: 34028267 DOI: 10.1021/acs.joc.1c00371] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Herein, we describe the direct synthesis of pyrrolo[1,2-α]quinoxaline via oxidative coupling between methyl arene and 1-(2-aminophenyl) pyrroles. Oxidation of the benzylic carbon of the methyl arene was achieved by di-t-butyl peroxide in the presence of an iron catalyst, followed by conversion to an activated aldehyde in situ. Oxygen played a crucial role in the oxidation process to accelerate benzaldehyde formation. Subsequent Pictet-Spengler-type annulation completed the quinoxaline structure. The protocol tolerated various kinds of functional groups and provided 22 4-aryl pyrrolo[1,2-α]quinoxalines when various methyl arene derivatives were used. The developed method proceeded in air, and all catalysts, reagents, and solvents were easily accessible.
Collapse
Affiliation(s)
- Jiwon Ahn
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Seok Beom Lee
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Injae Song
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Simin Chun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Dong-Chan Oh
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Suckchang Hong
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
22
|
Togiti UK, Shukla AK, Bhattacharya A. Pyrrolo[1,2-a]quinoxalines from chalcones: An alternate route. Tetrahedron Lett 2021. [DOI: 10.1016/j.tetlet.2021.153008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
23
|
D’Arrigo G, Gianquinto E, Rossetti G, Cruciani G, Lorenzetti S, Spyrakis F. Binding of Androgen- and Estrogen-Like Flavonoids to Their Cognate (Non)Nuclear Receptors: A Comparison by Computational Prediction. Molecules 2021; 26:1613. [PMID: 33799482 PMCID: PMC8001607 DOI: 10.3390/molecules26061613] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/02/2021] [Accepted: 03/10/2021] [Indexed: 12/24/2022] Open
Abstract
Flavonoids are plant bioactives that are recognized as hormone-like polyphenols because of their similarity to the endogenous sex steroids 17β-estradiol and testosterone, and to their estrogen- and androgen-like activity. Most efforts to verify flavonoid binding to nuclear receptors (NRs) and explain their action have been focused on ERα, while less attention has been paid to other nuclear and non-nuclear membrane androgen and estrogen receptors. Here, we investigate six flavonoids (apigenin, genistein, luteolin, naringenin, quercetin, and resveratrol) that are widely present in fruits and vegetables, and often used as replacement therapy in menopause. We performed comparative computational docking simulations to predict their capability of binding nuclear receptors ERα, ERβ, ERRβ, ERRγ, androgen receptor (AR), and its variant ART877A and membrane receptors for androgens, i.e., ZIP9, GPRC6A, OXER1, TRPM8, and estrogens, i.e., G Protein-Coupled Estrogen Receptor (GPER). In agreement with data reported in literature, our results suggest that these flavonoids show a relevant degree of complementarity with both estrogen and androgen NR binding sites, likely triggering genomic-mediated effects. It is noteworthy that reliable protein-ligand complexes and estimated interaction energies were also obtained for some suggested estrogen and androgen membrane receptors, indicating that flavonoids could also exert non-genomic actions. Further investigations are needed to clarify flavonoid multiple genomic and non-genomic effects. Caution in their administration could be necessary, until the safe assumption of these natural molecules that are largely present in food is assured.
Collapse
Affiliation(s)
- Giulia D’Arrigo
- Department of Drug Science and Technology, University of Turin, Via Giuria 9, 10125 Turin, Italy; (G.D.); (E.G.)
| | - Eleonora Gianquinto
- Department of Drug Science and Technology, University of Turin, Via Giuria 9, 10125 Turin, Italy; (G.D.); (E.G.)
| | - Giulia Rossetti
- Institute for Neuroscience and Medicine (INM-9) and Institute for Advanced Simulations (IAS-5) “Computational Biomedicine”, Forschungszentrum Jülich, 52425 Jülich, Germany
- Jülich Supercomputing Center (JSC), Forschungszentrum Jülich, 52425 Jülich, Germany
- Department of Neurology, RWTH, Aachen University, 52074 Aachen, Germany;
| | - Gabriele Cruciani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy;
| | - Stefano Lorenzetti
- Istituto Superiore di Sanità (ISS), Department of Food Safety, Nutrition and Veterinary Public Health, Viale Regina Elena 299, 00161 Rome, Italy
| | - Francesca Spyrakis
- Department of Drug Science and Technology, University of Turin, Via Giuria 9, 10125 Turin, Italy; (G.D.); (E.G.)
| |
Collapse
|
24
|
Ulhaq ZS, Soraya GV, Milliana A, Tse WKF. Association between GPER gene polymorphisms and GPER expression levels with cancer predisposition and progression. Heliyon 2021; 7:e06428. [PMID: 33748487 PMCID: PMC7970143 DOI: 10.1016/j.heliyon.2021.e06428] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/27/2021] [Accepted: 03/02/2021] [Indexed: 12/26/2022] Open
Abstract
Estrogen is a female sex steroid hormone that plays a significant role in physiological functions. Evidence suggests that estrogen-signaling pathways are closely linked to cancer development and progression. The novel G protein-coupled estrogen receptor (GPER or GPR30) has been shown to influence cancer predisposition and progression, although results of related studies remain equivocal. Thus, this meta-analysis aimed to estimate the relationship between GPER gene polymorphisms and GPER expression levels, with cancer predisposition and progression. The pooled results showed that two GPER polymorphisms, rs3808350 and rs3808351, were significantly associated with cancer predisposition, especially in the Asian population, but no significant association was detected for rs11544331. In parallel, we also found that cancer aggressiveness and progression correlated with rs3808351 and GPER expression in cancerous tissues. Altogether, our findings suggest that GPER plays a pivotal role in cancer pathogenesis and progression. We suggest that rs3808350 and rs3808351 may be used as a prospective biomarker for cancer screening; while rs3808351 and GPER expression can be used to examine the prognosis of patients with cancer. Further biological studies are warranted to confirm our findings.
Collapse
Affiliation(s)
- Zulvikar Syambani Ulhaq
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Maulana Malik Ibrahim State Islamic University of Malang, Batu, East Java, 65151, Indonesia
| | - Gita Vita Soraya
- Department of Biochemistry, Faculty of Medicine, Hasanuddin University, Makassar, South Sulawesi, 90245, Indonesia
| | - Alvi Milliana
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Maulana Malik Ibrahim State Islamic University of Malang, Batu, East Java, 65151, Indonesia
| | - William Ka Fai Tse
- Center for Promotion of International Education and Research, Faculty of Agriculture, Kyushu University, Fukuoka, 8190395, Japan
| |
Collapse
|
25
|
Mazzotta S, Governa P, Borgonetti V, Marcolongo P, Nanni C, Gamberucci A, Manetti F, Pessina F, Carullo G, Brizzi A, Aiello F. Pinocembrin and its linolenoyl ester derivative induce wound healing activity in HaCaT cell line potentially involving a GPR120/FFA4 mediated pathway. Bioorg Chem 2021; 108:104657. [PMID: 33556697 DOI: 10.1016/j.bioorg.2021.104657] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 12/17/2022]
Abstract
Wound healing represents an urgent need from the clinical point of view. Several diseases result in wound conditions which are difficult to treat, such as in the case of diabetic foot ulcer. Starting from there, the medicinal research has focused on various targets over the years, including GPCRs as new wound healing drug targets. In line with this, GPR120, known to be an attractive target in type 2 diabetes drug discovery, was studied to finalize the development of new wound healing agents. Pinocembrin (HW0) was evaluated as a suitable compound for interacting with GPR120, and was hybridized with fatty acids, which are known endogenous GPR120 ligands, to enhance the wound healing potential and GPR120 interactions. HW0 and its 7-linolenoyl derivative (HW3) were found to be innovative wound healing agents. Immunofluorescence and functional assays suggested that their activity was mediated by GPR120, and docking simulations showed that the compounds could share the same pocket occupied by the known GPR120 agonist, TUG-891.
Collapse
Affiliation(s)
- Sarah Mazzotta
- Department of Pharmaceutical Sciences, Via Luigi Mangiagalli 25, 20133 Milano, Italy
| | - Paolo Governa
- Department of Biotechnology, Chemistry and Pharmacy - DoE 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Vittoria Borgonetti
- Department of Neuroscience, Psychology, Pharmacology and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy
| | - Paola Marcolongo
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Claudio Nanni
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Alessandra Gamberucci
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Fabrizio Manetti
- Department of Biotechnology, Chemistry and Pharmacy - DoE 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy.
| | - Federica Pessina
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy.
| | - Gabriele Carullo
- Department of Biotechnology, Chemistry and Pharmacy - DoE 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy.
| | - Antonella Brizzi
- Department of Biotechnology, Chemistry and Pharmacy - DoE 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Francesca Aiello
- Department of Pharmacy, Health and Nutritional Sciences - DoE 2018-2022, University of Calabria, Ed. Polifunzionale, 87036 Arcavacata di Rende (CS), Italy
| |
Collapse
|
26
|
Khan SU, Ahemad N, Chuah LH, Naidu R, Htar TT. G protein-coupled estrogen receptor-1: homology modeling approaches and application in screening new GPER-1 modulators. J Biomol Struct Dyn 2020; 40:3325-3335. [PMID: 33164654 DOI: 10.1080/07391102.2020.1844059] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
G protein-coupled receptors (GPCRs) belong to the largest family of protein targets comprising over 800 members in which at least 500 members are the therapeutic targets. Among the GPCRs, G protein-coupled estrogen receptor-1 (GPER-1) has shown to have the ability in estrogen signaling. As GPER-1 plays a critical role in several physiological responses, GPER-1 has been considered as a potential therapeutic target to treat estrogen-based cancers and other non-communicable diseases. However, the progress in the understanding of GPER-1 structure and function is relatively slow due to the availability of a only a few selective GPER-1 modulators. As with many GPCRs, the X-ray crystal structure of GPER-1 is yet to be resolved and thus has led the researchers to search for new GPER-1 modulators using homology models of GPER-1. In this review, we aim to summarize various approaches used in the generation of GPER-1 homology model and their applications that have resulted in new GPER-1 ligands.
Collapse
Affiliation(s)
- Shafi Ullah Khan
- School of Pharmacy, Monash University Malaysia, Subang Jaya, Selangor, Malaysia
| | - Nafees Ahemad
- School of Pharmacy, Monash University Malaysia, Subang Jaya, Selangor, Malaysia.,Tropical Medicine and Biology Multidisciplinary Platform, Monash University Malaysia, Subang Jaya, Selangor, Malaysia
| | - Lay-Hong Chuah
- School of Pharmacy, Monash University Malaysia, Subang Jaya, Selangor, Malaysia.,Advanced Engineering Platform, Monash University Malaysia, Subang Jaya, Selangor, Malaysia
| | - Rakesh Naidu
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Selangor, Malaysia
| | - Thet Thet Htar
- School of Pharmacy, Monash University Malaysia, Subang Jaya, Selangor, Malaysia
| |
Collapse
|
27
|
Chun S, Ahn J, Putta RR, Lee SB, Oh DC, Hong S. Direct Synthesis of Pyrrolo[1,2-α]quinoxalines via Iron-Catalyzed Transfer Hydrogenation between 1-(2-Nitrophenyl)pyrroles and Alcohols. J Org Chem 2020; 85:15314-15324. [DOI: 10.1021/acs.joc.0c02145] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Simin Chun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Jiwon Ahn
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Ramachandra Reddy Putta
- BK 21 Plus Project, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Seok Beom Lee
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Dong-Chan Oh
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Suckchang Hong
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
28
|
Grande F, Occhiuzzi MA, Lappano R, Cirillo F, Guzzi R, Garofalo A, Jacquot Y, Maggiolini M, Rizzuti B. Computational Approaches for the Discovery of GPER Targeting Compounds. Front Endocrinol (Lausanne) 2020; 11:517. [PMID: 32849301 PMCID: PMC7417359 DOI: 10.3389/fendo.2020.00517] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/26/2020] [Indexed: 12/12/2022] Open
Abstract
Estrogens exert a panel of biological activities mainly through the estrogen receptors α and β, which belong to the nuclear receptor superfamily. Diverse studies have shown that the G protein-coupled estrogen receptor 1 (GPER, previously known as GPR30) also mediates the multifaceted effects of estrogens in numerous pathophysiological events, including neurodegenerative, immune, metabolic, and cardiovascular disorders and the progression of different types of cancer. In particular, GPER is implicated in hormone-sensitive tumors, albeit diverse issues remain to be deeply investigated. As such, this receptor may represent an appealing target for therapeutics in different diseases. The yet unavailable complete GPER crystallographic structure, and its relatively low sequence similarity with the other members of the G protein-coupled receptor (GPCR) family, hamper the possibility to discover compounds able to modulate GPER activity. Consequently, a reliable molecular model of this receptor is required for the design of suitable ligands. To date, convergent approaches involving structure-based drug design and virtual ligand screening have led to the identification of several GPER selective ligands, thus providing important information regarding its mode of action and function. In this survey, we summarize results obtained through computer-aided techniques devoted to the assessment of GPER ligands toward their usefulness in innovative treatments of different diseases.
Collapse
Affiliation(s)
- Fedora Grande
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Maria A. Occhiuzzi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Francesca Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
- Department of Physics, University of Calabria, Rende, Italy
| | - Rita Guzzi
- Department of Physics, University of Calabria, Rende, Italy
- CNR-NANOTEC, Licryl-UOS Cosenza and CEMIF.Cal, Department of Physics, University of Calabria, Rende, Italy
| | - Antonio Garofalo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Yves Jacquot
- Cibles Thérapeutiques et Conception de Médicaments (CiTCoM), CNRS UMR 8038, INSERM U1268, Faculté de Pharmacie de Paris, Université de Paris, Paris, France
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Bruno Rizzuti
- CNR-NANOTEC, Licryl-UOS Cosenza and CEMIF.Cal, Department of Physics, University of Calabria, Rende, Italy
| |
Collapse
|
29
|
Zacarías-Lara OJ, Méndez-Luna D, Martínez-Ruíz G, García-Sanchéz JR, Fragoso-Vázquez MJ, Bello M, Becerra-Martínez E, García-Vázquez JB, Correa-Basurto J. Synthesis and In Vitro Evaluation of Tetrahydroquinoline Derivatives as Antiproliferative Compounds of Breast Cancer via Targeting the GPER. Anticancer Agents Med Chem 2020; 19:760-771. [PMID: 30451119 DOI: 10.2174/1871520618666181119094144] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/03/2018] [Accepted: 11/04/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Some reports have demonstrated the role of the G Protein-coupled Estrogen Receptor (GPER) in growth and proliferation of breast cancer cells. OBJECTIVE In an effort to develop new therapeutic strategies against breast cancer, we employed an in silico study to explore the binding modes of tetrahydroquinoline 2 and 4 to be compared with the reported ligands G1 and G1PABA. METHODS This study aimed to design and filter ligands by in silico studies determining their Lipinski's rule, toxicity and binding properties with GPER to achieve experimental assays as anti-proliferative compounds of breast cancer cell lines. RESULTS In silico studies suggest as promissory two tetrahydroquinoline 2 and 4 which contain a carboxyl group instead of the acetyl group (as is needed for G1 synthesis), which add low (2) and high hindrance (4) chemical moieties to explore the polar, hydrophobic and hindrance effects. Docking and molecular dynamics simulations of the target compounds were performed with GPER to explore their binding mode and free energy values. In addition, the target small molecules were synthesized and assayed in vitro using breast cancer cells (MCF-7 and MDA-MB-231). Experimental assays showed that compound 2 decreased cell proliferation, showing IC50 values of 50µM and 25µM after 72h of treatment of MCF-7 and MDA-MB-231 cell lines, respectively. Importantly, compound 2 showed a similar inhibitory effect on proliferation as G1 compound in MDA-MB-231 cells, suggesting that both ligands reach the GPER-binding site in a similar way, as was demonstrated through in silico studies. CONCLUSION A concentration-dependent inhibition of cell proliferation occurred with compound 2 in the two cell lines regardless of GPER.
Collapse
Affiliation(s)
- Oscar J Zacarías-Lara
- Laboratorio de Diseno y Desarrollo de Nuevos Farmacos e Innovacion Biotecnologica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Diaz Miron, 11340 Mexico, CDMX, Mexico
| | - David Méndez-Luna
- Laboratorio de Diseno y Desarrollo de Nuevos Farmacos e Innovacion Biotecnologica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Diaz Miron, 11340 Mexico, CDMX, Mexico
| | - Gustavo Martínez-Ruíz
- Unidad de Investigacion en Enfermedades Oncologicas, Hospital Infantil de Mexico, Federico Gomez, Mexico
| | - José R García-Sanchéz
- Laboratorio de Oncologia Molecular y Estres Oxidativo, Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Diaz Miron, 11340 Mexico, CDMX, Mexico
| | - Manuel J Fragoso-Vázquez
- Laboratorio de Diseno y Desarrollo de Nuevos Farmacos e Innovacion Biotecnologica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Diaz Miron, 11340 Mexico, CDMX, Mexico.,Departamento de Química Orgánica, Escuela Nacional de Ciencias, Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala, México, CDMX., 11340 México
| | - Martiniano Bello
- Laboratorio de Diseno y Desarrollo de Nuevos Farmacos e Innovacion Biotecnologica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Diaz Miron, 11340 Mexico, CDMX, Mexico
| | - Elvia Becerra-Martínez
- Laboratorio de RMN, Centro de Nanociencias y Micro y Nanotecnologias, Instituto Politecnico Nacional, Calle Luis Enrique Erro s/n, Unidad Profesional Adolfo Lopez Mateos, Gustavo A, Madero, 07738 Mexico, Ciudad de Mexico, Mexico
| | - Juan B García-Vázquez
- Laboratorio de Diseno y Desarrollo de Nuevos Farmacos e Innovacion Biotecnologica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Diaz Miron, 11340 Mexico, CDMX, Mexico
| | - José Correa-Basurto
- Laboratorio de Diseno y Desarrollo de Nuevos Farmacos e Innovacion Biotecnologica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Escuela Superior de Medicina, Instituto Politecnico Nacional, Plan de San Luis y Diaz Miron, 11340 Mexico, CDMX, Mexico
| |
Collapse
|
30
|
Seo Y, Lee JH, Park SH, Namkung W, Kim I. Expansion of chemical space based on a pyrrolo[1,2-a]pyrazine core: Synthesis and its anticancer activity in prostate cancer and breast cancer cells. Eur J Med Chem 2019; 188:111988. [PMID: 31901746 DOI: 10.1016/j.ejmech.2019.111988] [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: 10/24/2019] [Revised: 12/10/2019] [Accepted: 12/17/2019] [Indexed: 12/24/2022]
Abstract
In connection with our continued research to generate new aza-fused heteroaromatic chemical scaffolds, we developed a highly atom-economical three-component route to novel 3,4-dihydropyrrolo[1,2-a]pyrazine ring skeleton multi-functionalized on the pyrazine unit. This [4+1+1] annulation approach led us to gain access to a new N-fused bicyclic chemical space having two distinctive functional groups (heteroaryl and aroyl) in a trans manner. Investigation of anticancer activity of the synthesized compounds and their derivatives revealed that (3R*,4S*)-3-(4-bromophenyl)-4-(4-fluorobenzoyl)-2-(2-oxo-2-phenylethyl)-3,4-dihydropyrrolo[1,2-a]pyrazin-2-ium bromide (3h) has potent anticancer activity. 3h significantly inhibited cell viability in prostate cancer cells (PC-3) and breast cancer cells (MCF-7) with IC50 value of 1.18 ± 0.05 μM and 1.95 ± 0.04 μM, respectively. In addition, 3h strongly reduced cell migration in a dose dependent manner, and induced apoptosis via caspase-3 activation and cleavage of PARP in PC-3 and MCF-7 cells. Our results in this study imply that 3h can be a potential anticancer agent against prostate cancer and breast cancer.
Collapse
Affiliation(s)
- Yohan Seo
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85, Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea
| | - Jeong Hwa Lee
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85, Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea
| | - So-Hyeon Park
- Graduate Program of Industrial Pharmaceutical Science, Yonsei University, Incheon, 21983, Republic of Korea
| | - Wan Namkung
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85, Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea; Interdisciplinary Program of Integrated OMICS for Biomedical Science Graduate School, Yonsei University, Seoul, 03722, Republic of Korea.
| | - Ikyon Kim
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85, Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Republic of Korea.
| |
Collapse
|
31
|
Effects of cholecalciferol supplementation on serum angiogenic biomarkers in breast cancer patients treated with tamoxifen: A controlled randomized clinical trial. Nutrition 2019; 72:110656. [PMID: 31901710 DOI: 10.1016/j.nut.2019.110656] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 11/04/2019] [Accepted: 11/08/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The aim of this study was to investigate the effects of cholecalciferol supplementation on serum levels of angiogenic parameters in patients with breast cancer (BC) who were treated with tamoxifen. METHODS This was a pilot-based, randomized, triple-blind, placebo-controlled clinical trial with 52 patients with BC randomly assigned to either an intervention group receiving weekly 50 000 IU cholecalciferol or a placebo group for 8 wk. At baseline and at end of study, serum levels of angiogenic growth factors such as vascular endothelial growth factor (VEGF)-A, angiopoietin (Ang)-2, hypoxia-inducible factor (Hif)-1, and high-sensitivity C-reactive protein were measured by enzyme-linked immunosorbent assay. Every 4 wk, a completed 3-d, 24-h dietary record and daily sunlight exposure checklist were collected and anthropometric variables were measured. RESULTS The ultimate number of participants in each arm was 22 for analyses. For premenopausal women, cholecalciferol supplementation resulted in a significant decrease in serum levels of Ang-2 and VEGF-A after 8 wk of treatment (P < 0.05). In the absence of vascular invasion, supplementation led to a significant decrease in Ang-2 levels compared with the placebo group (P < 0.05). Supplementation caused significant increases in Hif-1 in patients diagnosed with the infiltration of tumors into vascular or lymphatic vessels (P < 0.05). CONCLUSION Cholecalciferol supplementation achieved sufficient efficacy among patients with BC taking tamoxifen and could be effective in the reduction of angiogenic biomarkers particularly dependent on the infiltration status of the tumor to vessels. Further studies with larger subgroups should be investigated.
Collapse
|
32
|
A highly divergent Pictet-Spengler approach for pyrrolo[1,2-a]quinoxalines from aryl amine using 1,2-dinitrobenzene as an oxidant. Tetrahedron Lett 2019. [DOI: 10.1016/j.tetlet.2019.151250] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
33
|
Guo WY, Zeng SMZ, Deora GS, Li QS, Ruan BF. Estrogen Receptor α (ERα)-targeting Compounds and Derivatives: Recent Advances in Structural Modification and Bioactivity. Curr Top Med Chem 2019; 19:1318-1337. [PMID: 31215379 DOI: 10.2174/1568026619666190619142504] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 04/29/2019] [Accepted: 05/05/2019] [Indexed: 12/17/2022]
Abstract
Breast cancer is the most common cancer suffered by female, and the second highest cause of cancer-related death among women worldwide. At present, hormone therapy is still the main treatment route and can be divided into three main categories: selective estrogen receptor modulators (SERMs), selective estrogen receptor downregulators (SERDs), and aromatase inhibitors (AIs). However, breast cancer is difficult to cure even after several rounds of anti-estrogen therapy and most drugs have serious side-effects. Here, we review the literature published over the past five years regarding the isolation and synthesis of analogs and their derivatives.
Collapse
Affiliation(s)
- Wei-Yun Guo
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Shang-Ming-Zhu Zeng
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Girdhar Singh Deora
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Qing-Shan Li
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Ban-Feng Ruan
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| |
Collapse
|
34
|
|
35
|
Cappello AR, Aiello F, Polerà N, Armentano B, Casaburi I, Di Gioia ML, Loizzo MR, Dolce V, Pezzi V, Tundis R. In vitro anti-proliferative and anti-bacterial properties of new C7 benzoate derivatives of pinocembrin. Nat Prod Res 2019; 35:1783-1791. [PMID: 31311327 DOI: 10.1080/14786419.2019.1641805] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
In the present work, the in vitro anti-proliferative and anti-bacterial activities of three semi-synthetic benzoate pinocembrin derivatives, isolated from the aerial parts of Glycyrrhiza glabra L., were investigated. As occurs in most natural compounds, the bioavailability of pinocembrin is very poor, therefore it should be improved by chemical strategies aimed to prolong its shelf life and, consequently, its activity. On this basis, three benzoate derivatives of pinocembrin (a1-a3) were synthesised and assayed in order to ascertain their biological value. Among them, compound a1 showed the highest anti-proliferative activity on a wide panel of cancer cell lines, as well as low toxic effects on non-malignant breast cells. The calculated IC50 values in HeLa and SKBR3 cells were 8.5 and 12.7 µM, respectively. Briefly, a1 treatment increased ROS levels, induced mitochondrial membrane damage leading to necrotic death of HeLa cells. Moreover, a1 displayed a promising anti-bacterial activity.
Collapse
Affiliation(s)
- Anna R Cappello
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Francesca Aiello
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Nicoletta Polerà
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Biagio Armentano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Ivan Casaburi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Maria Luisa Di Gioia
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Monica R Loizzo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Vincenza Dolce
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Vincenzo Pezzi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Rosa Tundis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| |
Collapse
|
36
|
Kim J, Park M, Choi J, Singh DK, Kwon HJ, Kim SH, Kim I. Design, synthesis, and biological evaluation of novel pyrrolo[1,2-a]pyrazine derivatives. Bioorg Med Chem Lett 2019; 29:1350-1356. [DOI: 10.1016/j.bmcl.2019.03.044] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 02/27/2019] [Accepted: 03/27/2019] [Indexed: 12/31/2022]
|
37
|
Dagar A, Bae GH, Lee JH, Kim I. Tandem [4+1+1] Annulation Approach to 4-Acyl-3,4-dihydropyrrolo[1,2-a]pyrazines: Diastereoselective Construction of Dihydropyrazine Units from Pyrroles. J Org Chem 2019; 84:6916-6927. [DOI: 10.1021/acs.joc.9b00724] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Anuradha Dagar
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Gi Hun Bae
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Jeong Hwa Lee
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Ikyon Kim
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| |
Collapse
|
38
|
Niazvand F, Orazizadeh M, Khorsandi L, Abbaspour M, Mansouri E, Khodadadi A. Effects of Quercetin-Loaded Nanoparticles on MCF-7 Human Breast Cancer Cells. ACTA ACUST UNITED AC 2019; 55:medicina55040114. [PMID: 31013662 PMCID: PMC6524048 DOI: 10.3390/medicina55040114] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 02/15/2019] [Accepted: 04/16/2019] [Indexed: 12/27/2022]
Abstract
Background and objectives: Previous studies have shown anti-tumor activity of quercetin (QT). However, the low bioavailability of QT has restricted its use. This study aimed to assess the toxic effect of QT encapsulated in solid lipid nanoparticles (QT-SLNs) on the growth of MCF-7 human breast cancer cells. Materials and Methods: MCF-7 and MCF-10A (non-tumorigenic cell line) cell lines treated with 25 µmol/mL of QT or QT-SLNs for 48 h. Cell viability, colony formation, oxidative stress, and apoptosis were evaluated to determine the toxic effects of the QT-SLNs. Results: The QT-SLNs with appropriate characteristics (particle size of 85.5 nm, a zeta potential of −22.5 and encapsulation efficiency of 97.6%) were prepared. The QT-SLNs showed sustained QT release until 48 h. Cytotoxicity assessments indicated that QT-SLNs inhibited MCF-7 cells growth with a low IC50 (50% inhibitory concentration) value, compared to the free QT. QT-SLNs induced a significant decrease in the viability and proliferation of MCF-7 cells, compared to the free QT. QT-SLN significantly increased reactive oxygen species (ROS) level and MDA contents and significantly decreased antioxidant enzyme activity in the MCF-7 cells. Following QT-SLNs treatment, the expression of the Bcl-2 protein significantly decreased, whereas Bx expression showed a significant increase in comparison with free QT-treated cells. Furthermore, The QT-SLNs significantly increased apoptotic and necrotic indexes in MCF-7 cells. Viability, proliferation, oxidative stress and apoptosis of MCF-10A cells were not affected by QT or QT-SLNs. Conclusions: According to the results of this study, SLN significantly enhanced the toxic effect of QT against human breast cancer cells.
Collapse
Affiliation(s)
- Firoozeh Niazvand
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Mahmoud Orazizadeh
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Layasadat Khorsandi
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Mohammadreza Abbaspour
- Targeted Drug Delivery Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Esrafil Mansouri
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Ali Khodadadi
- Cancer Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
39
|
Perri M, Aiello F, Cione E, Carullo G, Amendola L, Mazzotta S, Caroleo MC. Investigation of TNBC in vitro Antiproliferative Effects of Versatile Pirrolo[1,2-a]quinoxaline Compounds. Front Mol Biosci 2019; 6:12. [PMID: 30915341 PMCID: PMC6423061 DOI: 10.3389/fmolb.2019.00012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 02/19/2019] [Indexed: 11/19/2022] Open
Abstract
The triple-negative breast cancer (TNBC) is characterized by a more aggressive nature and poorer prognosis, nowadays none pharmaceutical approach is still available. For this reason, the research of new active compounds and attractive targets represents an interesting field. In this context MDA- MB-231 cell line was selected to evaluate the antiproliferative effects of new [1,2-a]-pyrroloquinoxaline derivatives. The MTT assay revealed that the amine forms of synthesized molecules were more active compared to iminic ones at 72 h of incubation. The antiproliferative effect of the most promising compounds highlighted the formation of autophagic vacuoles.
Collapse
Affiliation(s)
- Mariarita Perri
- Department of Pharmacy Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Francesca Aiello
- Department of Pharmacy Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Erika Cione
- Department of Pharmacy Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Gabriele Carullo
- Department of Pharmacy Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Luisa Amendola
- Department of Pharmacy Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Sarah Mazzotta
- Química Orgánica Y Farmacéutica, Universidad de Sevilla, Seville, Spain
| | - Maria Cristina Caroleo
- Department of Pharmacy Health and Nutritional Sciences, University of Calabria, Rende, Italy
| |
Collapse
|
40
|
Sibiya MA, Raphoko L, Mangokoana D, Makola R, Nxumalo W, Matsebatlela TM. Induction of Cell Death in Human A549 Cells Using 3-(Quinoxaline-3-yl) Prop-2-ynyl Methanosulphonate and 3-(Quinoxaline-3-yl) Prop-2-yn-1-ol. Molecules 2019; 24:E407. [PMID: 30678061 PMCID: PMC6384999 DOI: 10.3390/molecules24030407] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/12/2019] [Accepted: 01/14/2019] [Indexed: 12/21/2022] Open
Abstract
Despite major advancements in the development of various chemotherapeutic agents, treatment for lung cancer remains costly, ineffective, toxic to normal non-cancerous cells, and still hampered by a high level of remissions. A novel cohort of quinoxaline derivatives designed to possess a wide spectrum of biological activities was synthesized with promising targeted and selective anticancer drug activity. Hence, this study was aimed at determining in vitro anticancer activity effects of a newly synthesized class of 3-(quinoxaline-3-yl) prop-2-ynyl quinoxaline derivatives on A549 lung cancer cells. An assessment of the quinoxaline derivatives ferric reducing power, free radical scavenging activity, cytotoxic activity, and ability to induce reactive oxygen species (ROS) production was performed using the Ferric Reducing Antioxidant Power (FRAP), 2,2-diphenyl-1-picryl-hydrazyl (DPPH), 3-[4,5-dimethylthiazole-2-yl]-2,5-diphenyltetrazolium bromide (MTT) and 2',7'-dichlorodihydrofluorescein diacetate (H₂DCFDA) assays, respectively. The ability of the quinoxaline derivatives to induce apoptosis in A549 cells was assessed using the Acridine Orange/Ethidium Bromide (AO/EB) and Annexin V-FITC/Dead Cell Assay. Of the four quinoxaline derivatives tested, 3-(quinoxaline-3-yl) prop-2-ynyl methanosulphate (LA-39B) and 3-(quinoxaline-3-yl) prop-2-yn-1-ol (LA-55) displayed a dose-dependent reducing power, free-radical scavenging activity, inhibition of cell viability, and stimulation of ROS production which was accompanied by induction of apoptosis in A549 lung cancer cells. None of the quinoxaline derivatives induced cell death or ROS production in non-cancerous Raw 267.4 macrophage cells. Cytotoxicity was observed in A549 lung cancer, HeLa cervical cancer, and MCF-7 breast cancer cells albeit inhibition was more pronounced in A549 cells. The results of the study suggest that 3-(quinoxaline-3-yl) prop-2-ynyl methanosulphate and 3-(quinoxaline-3-yl) prop-2-yn-1-ol induce apoptotic cell death in A549 lung cancer cells.
Collapse
Affiliation(s)
- Mixo Aunny Sibiya
- Department of Biochemistry, Microbiology and Biotechnology, School of Molecular and Life Sciences, University of Limpopo, Sovenga 0727, South Africa.
| | - Lerato Raphoko
- Chemistry Department, School of Physical and Mineral Sciences, University of Limpopo, Sovenga 0727, South Africa.
| | - Dikgale Mangokoana
- Department of Biochemistry, Microbiology and Biotechnology, School of Molecular and Life Sciences, University of Limpopo, Sovenga 0727, South Africa.
| | - Raymond Makola
- Department of Biochemistry, Microbiology and Biotechnology, School of Molecular and Life Sciences, University of Limpopo, Sovenga 0727, South Africa.
| | - Winston Nxumalo
- Chemistry Department, School of Physical and Mineral Sciences, University of Limpopo, Sovenga 0727, South Africa.
| | - Thabe Moses Matsebatlela
- Department of Biochemistry, Microbiology and Biotechnology, School of Molecular and Life Sciences, University of Limpopo, Sovenga 0727, South Africa.
| |
Collapse
|
41
|
G-Protein Coupled Estrogen Receptor in Breast Cancer. Int J Mol Sci 2019; 20:ijms20020306. [PMID: 30646517 PMCID: PMC6359026 DOI: 10.3390/ijms20020306] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/10/2019] [Accepted: 01/12/2019] [Indexed: 12/16/2022] Open
Abstract
The G-protein coupled estrogen receptor (GPER), an alternate estrogen receptor (ER) with a structure distinct from the two canonical ERs, being ERα, and ERβ, is expressed in 50% to 60% of breast cancer tissues and has been presumed to be associated with the development of tamoxifen resistance in ERα positive breast cancer. On the other hand, triple-negative breast cancer (TNBC) constitutes 15% to 20% of breast cancers and frequently displays a more aggressive behavior. GPER is prevalent and involved in TNBC and can be a therapeutic target. However, contradictory results exist regarding the function of GPER in breast cancer, proliferative or pro-apoptotic. A better understanding of the GPER, its role in breast cancer, and the interactions with the ER and epidermal growth factor receptor will be beneficial for the disease management and prevention in the future.
Collapse
|
42
|
Krishna T, Reddy TN, Laxminarayana E, Kalita D. Copper-Catalyzed One-Pot Synthesis of Pyrrolo[1,2-a
]quinoxaline Derivatives from 1-(2-Aminophenyl)-pyrroles and Aldehydes. ChemistrySelect 2019. [DOI: 10.1002/slct.201803538] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Thalishetti Krishna
- Technology Development Centre; Custom Pharmaceutical Services, Dr. Reddy's Laboratories Ltd; Hyderabad 500049 India
- Department of Chemistry; Jawaharlal Nehru Technological University Hyderabad, Kukatpally, Hyderabad -; 500 085 Telangana India
| | - Thatikonda Narendar Reddy
- Crop Protection Chemicals Division; CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Telangana; Hyderabad-500007 India
| | - Eppakayala Laxminarayana
- Department of Chemistry, Sreenidhi Institute of Science and Technology (Autonomous), Ghatkesar; Hyderabad- 501301, Telangana India
| | - Dipak Kalita
- Technology Development Centre; Custom Pharmaceutical Services, Dr. Reddy's Laboratories Ltd; Hyderabad 500049 India
| |
Collapse
|
43
|
Khan SU, Ahemad N, Chuah LH, Naidu R, Htar TT. Sequential ligand- and structure-based virtual screening approach for the identification of potential G protein-coupled estrogen receptor-1 (GPER-1) modulators. RSC Adv 2019; 9:2525-2538. [PMID: 35520492 PMCID: PMC9059856 DOI: 10.1039/c8ra09318k] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 01/04/2019] [Indexed: 11/26/2022] Open
Abstract
G protein-coupled estrogen receptor-1 (GPER-1) is a seven transmembrane receptor, responsible for mediating rapid estrogen signaling in many physiological responses in reproductive, nervous, endocrine, immune and cardiovascular systems. Due to unavailability of the crystal structure of GPER-1, we have performed sequential ligand-based virtual screening (LBVS) and structure-based screening (SBVS) to identify potential GPER-1 modulators. LBVS and SBVS approaches were validated retrospectively using the Receiver Operating Curve (ROC) plot and the early Enrichment Factor (EF). LBVS was performed based on a GPER-1 agonist, G1, as a query model for screening of the eMolecules library using the Rapid Overlay of Chemical Structure (ROCS) and the electrostatic potential screening (EON) approaches. Top-scored hits from LBVS were further screened by SBVS. SBVS was based on generating homology models of GPER-1 and subsequent molecular docking studies. Using Chemguass4 score, we filtered the final hits with the higher score in comparison to G1 (Chemguass4 score = −11.575). The top-ranked hits were clustered based on similarity in their scaffolds. Prospective validation was performed by evaluating the antiproliferative activity of synthesized compounds (SK0 and SK0P) which were representative of top hits obtained from our virtual screening approach. This paper presents the application of sequential ligand- and structure-based virtual screening approach for the identification of G protein-coupled estrogen receptor-1 (GPER-1/GPR30) modulators.![]()
Collapse
Affiliation(s)
- Shafi Ullah Khan
- School of Pharmacy
- Monash University Malaysia
- 47500 Subang Jaya
- Malaysia
| | - Nafees Ahemad
- School of Pharmacy
- Monash University Malaysia
- 47500 Subang Jaya
- Malaysia
- Tropical Medicine and Biology Multidisciplinary Platform
| | - Lay-Hong Chuah
- School of Pharmacy
- Monash University Malaysia
- 47500 Subang Jaya
- Malaysia
- Advanced Engineering Platform
| | - Rakesh Naidu
- Jeffrey Cheah School of Medicine and Health Sciences
- Monash University Malaysia
- 47500 Subang Jaya
- Malaysia
| | - Thet Thet Htar
- School of Pharmacy
- Monash University Malaysia
- 47500 Subang Jaya
- Malaysia
| |
Collapse
|