1
|
Bernardini G, Trezza A, Petricci E, Romagnoli G, Zambardino D, Manetti F, Braconi D, Geminiani M, Santucci A. A Comprehensive In Vitro and In Silico Approach for Targeting 4-Hydroxyphenyl Pyruvate Dioxygenase: Towards New Therapeutics for Alkaptonuria. Int J Mol Sci 2025; 26:3181. [PMID: 40243989 PMCID: PMC11988800 DOI: 10.3390/ijms26073181] [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: 01/30/2025] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
Alkaptonuria (AKU) is an ultra-rare genetic disorder caused by mutations in the homogentisate 1,2-dioxygenase (HGD) gene, leading to the accumulation of homogentisic acid (HGA). Current treatment options are limited, with Nitisinone (Orfadin or NTBC) being the only approved drug. However, its long-term use raises concerns due to significant adverse effects, highlighting the urgent need for safer alternatives. AKU manifests with progressive and often painful symptoms, severely impacting patients' quality of life. Identifying new therapeutic approaches to inhibit 4-hydroxyphenyl pyruvate dioxygenase (4-HPPD) is critical to improving outcomes for AKU patients. In this study, we present a novel integrated in vitro and in silico strategy to assess the residence time of 4-HPPD inhibitors. In particular, we evaluated several features of a set of triketone compounds including their inhibitory efficacy, residence time, and ochronotic pigment accumulation. By means of our integrated approach, we investigated the pharmacokinetic and pharmacodynamics properties of novel 4-HPPD inhibitors and provided a promising foundation for the development of safer and more effective treatments for AKU.
Collapse
Affiliation(s)
- Giulia Bernardini
- ONE-HEALTH Lab, Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (G.B.); (A.T.); (E.P.); (G.R.); (D.Z.); (D.B.); (A.S.)
| | - Alfonso Trezza
- ONE-HEALTH Lab, Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (G.B.); (A.T.); (E.P.); (G.R.); (D.Z.); (D.B.); (A.S.)
| | - Elena Petricci
- ONE-HEALTH Lab, Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (G.B.); (A.T.); (E.P.); (G.R.); (D.Z.); (D.B.); (A.S.)
| | - Giulia Romagnoli
- ONE-HEALTH Lab, Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (G.B.); (A.T.); (E.P.); (G.R.); (D.Z.); (D.B.); (A.S.)
| | - Demetra Zambardino
- ONE-HEALTH Lab, Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (G.B.); (A.T.); (E.P.); (G.R.); (D.Z.); (D.B.); (A.S.)
| | - Fabrizio Manetti
- ONE-HEALTH Lab, Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (G.B.); (A.T.); (E.P.); (G.R.); (D.Z.); (D.B.); (A.S.)
| | - Daniela Braconi
- ONE-HEALTH Lab, Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (G.B.); (A.T.); (E.P.); (G.R.); (D.Z.); (D.B.); (A.S.)
| | - Michela Geminiani
- ONE-HEALTH Lab, Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (G.B.); (A.T.); (E.P.); (G.R.); (D.Z.); (D.B.); (A.S.)
| | - Annalisa Santucci
- ONE-HEALTH Lab, Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (G.B.); (A.T.); (E.P.); (G.R.); (D.Z.); (D.B.); (A.S.)
- MetabERN, Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy
| |
Collapse
|
2
|
Frusciante L, Geminiani M, Shabab B, Olmastroni T, Roncucci N, Mastroeni P, Salvini L, Lamponi S, Trezza A, Santucci A. Enhancing Industrial Hemp ( Cannabis sativa) Leaf By-Products: Bioactive Compounds, Anti-Inflammatory Properties, and Potential Health Applications. Int J Mol Sci 2025; 26:548. [PMID: 39859264 PMCID: PMC11765263 DOI: 10.3390/ijms26020548] [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: 11/26/2024] [Revised: 12/21/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
The sustainable utilization of biomass-derived bioactives addresses the growing demand for natural health products and supports sustainable development goals by reducing reliance on synthetic chemicals in healthcare. Cannabis sativa biomass, in particular, has emerged as a valuable resource within this context. This study focuses on the hydroethanolic extract of C. sativa leaves (CSE), which exhibited significant levels of phenolic compounds contributing to robust antioxidant activity. Evaluation using potassium ferricyanide, ABTS, and DPPH methods revealed potent radical scavenging activity comparable to the Trolox standard. UPLC-MS/MS profiling identified cannabinoids as the predominant secondary metabolites in CSE, with flavonoids also present in substantial quantities. This study investigated the anti-inflammatory potential of CSE on RAW 264.7 macrophages and IL-1β-stimulated C-20/A4 immortalized human chondrocytes, demonstrating protective effects without cytotoxic or mutagenic effects. Mechanistically, CSE reduced inflammation by inhibiting the MAPK and NF-κB signaling pathways. In silico approaches showed the ability of CSE's main metabolites to bind and influence MAPK and NF-κB activity, confirming in vitro evidence. Incorporating C. sativa leaf extract into a hyaluronic acid-based formulation showed biotechnological promise for treating joint inflammation. Future research should aim to elucidate the molecular mechanisms underlying these effects and explore the potential of CSE-derived compounds in mitigating osteoarthritis progression. This approach highlights the significance of utilizing annually increasing biomass waste for sustainable bioactivity and environmental impact reduction.
Collapse
Affiliation(s)
- Luisa Frusciante
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (L.F.); (B.S.); (T.O.); (P.M.); (S.L.); (A.T.); (A.S.)
| | - Michela Geminiani
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (L.F.); (B.S.); (T.O.); (P.M.); (S.L.); (A.T.); (A.S.)
- SienabioACTIVE, University of Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Behnaz Shabab
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (L.F.); (B.S.); (T.O.); (P.M.); (S.L.); (A.T.); (A.S.)
| | - Tommaso Olmastroni
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (L.F.); (B.S.); (T.O.); (P.M.); (S.L.); (A.T.); (A.S.)
| | - Neri Roncucci
- Tenuta di Mensanello, Località Mensanello, 34, 53034 Colle di Val d’Elsa, Italy;
| | - Pierfrancesco Mastroeni
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (L.F.); (B.S.); (T.O.); (P.M.); (S.L.); (A.T.); (A.S.)
| | - Laura Salvini
- Fondazione Toscana Life Sciences, Strada del Petriccio e Belriguardo, 53100 Siena, Italy;
| | - Stefania Lamponi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (L.F.); (B.S.); (T.O.); (P.M.); (S.L.); (A.T.); (A.S.)
- SienabioACTIVE, University of Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Alfonso Trezza
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (L.F.); (B.S.); (T.O.); (P.M.); (S.L.); (A.T.); (A.S.)
| | - Annalisa Santucci
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 53100 Siena, Italy; (L.F.); (B.S.); (T.O.); (P.M.); (S.L.); (A.T.); (A.S.)
- SienabioACTIVE, University of Siena, Via Aldo Moro, 53100 Siena, Italy
- ARTES 4.0, Viale Rinaldo Piaggio, 34, 56025 Pontedera, Italy
| |
Collapse
|
3
|
Ramunno A, Vitale RM, Amodeo P, Crescenzi C, Panti A, Fiorenzani P, De Luca M, Spizzirri UG, Restuccia D, Aiello F, Fusi F. Bioguided Identification of Polymethoxyflavones as Novel Vascular Ca V1.2 Channel Blockers from Citrus Peel. Molecules 2024; 29:5693. [PMID: 39683852 DOI: 10.3390/molecules29235693] [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: 10/14/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
The huge amount of citrus peel produced worldwide represents an economic burden for society. However, this agricultural by-product is a rich source of natural molecules, potentially endowed with interesting pharmacological activities. In this regard, we decided to investigate if the polymethoxyflavones contained in citrus peel waste could be exploited as novel vasorelaxant agents. A hydroalcoholic blond orange (Citrus sinensis) peel extract, obtained by ultrasonication, was partitioned in dichloromethane. Column chromatography allowed for the isolation of four polymethoxyflavones, namely, scutellarein tetramethyl ether, nobiletin, tangeretin, and sinensetin, identified by nuclear magnetic resonance (NMR) spectroscopy and UPLC-HRMS/MS and confirmed by multivariate curve resolution of NMR fractional spectra. The four molecules showed interesting in vitro vasorelaxant activity, at least, in part, due to the blockade of smooth muscle CaV1.2 channels. Molecular modeling and docking analysis elucidated the binding mode of the polymethoxyflavones at the homology model of the rat CaV1.2c subunit and provided the structural basis to rationalise the highest activity of scutellarein tetramethyl ether in the set and the dramatic effect of the additional methoxy group occurring in nobiletin and sinensetin. In conclusion, citrus peel can be considered a freely available, valuable source of vasoactive compounds worthy of pharmaceutical and/or nutraceutical exploitation.
Collapse
Affiliation(s)
- Anna Ramunno
- Department of Pharmacy, University of Salerno, 84084 Fisciano, SA, Italy
| | - Rosa Maria Vitale
- Institute of Biomolecular Chemistry-National Research Council (ICB-CNR), 80078 Pozzuoli, NA, Italy
| | - Pietro Amodeo
- Institute of Biomolecular Chemistry-National Research Council (ICB-CNR), 80078 Pozzuoli, NA, Italy
| | - Carlo Crescenzi
- Department of Pharmacy, University of Salerno, 84084 Fisciano, SA, Italy
| | - Alice Panti
- Department of Life Sciences, University of Siena, 53100 Siena, TS, Italy
| | - Paolo Fiorenzani
- Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, 53100 Siena, TS, Italy
| | - Michele De Luca
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy
| | - Umile Gianfranco Spizzirri
- Ionian Department of Law, Economics and Environment, University of Bari Aldo Moro, 74123 Taranto, TA, Italy
| | - Donatella Restuccia
- Department of Management, University of Roma La Sapienza, 00161 Rome, RM, Italy
| | - Francesca Aiello
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy
| | - Fabio Fusi
- Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, 53100 Siena, TS, Italy
| |
Collapse
|
4
|
Dias P, Salam R, Pourová J, Vopršalová M, Konečný L, Jirkovský E, Duintjer Tebbens J, Mladěnka P. The quercetin metabolite 4-methylcatechol causes vasodilation via voltage-gated potassium (K V) channels. Food Funct 2024; 15:11047-11059. [PMID: 39422021 DOI: 10.1039/d3fo04672a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Dietary polyphenols have been associated with many beneficial cardiovascular effects. However, these effects are rather attributed to small phenolic metabolites formed by the gut microbiota, which reach sufficient concentrations in systemic circulation. 4-Methylcatechol (4-MC) is one such metabolite. As it is shown to possess considerable vasorelaxant effects, this study aimed to unravel its mechanism of action. To this end, experimental in vitro and in silico approaches were employed. In the first step, isometric tension recordings were performed on rat aortic rings. 4-MC potentiated the effect of cyclic nucleotides, but the effect was not mediated by either soluble guanylyl cyclase (sGC), modification of cyclic adenosine monophosphate levels, or protein kinase G. Hence, downstream targets such as calcium or potassium channels were considered. Inhibition of voltage-gated K+ channels (KV) markedly decreased the effect of 4-MC, and vasodilation was partly decreased by inhibition of the KV7 isoform. Contrarily, other types of K+ channels or L-type Ca2+ channels were not involved. In silico reverse docking confirmed that 4-MC binds to KV7.4 through hydrogen bonding and hydrophobic interactions. In particular, it interacts with two crucial residues for KV7.4 activation: Trp242 and Phe246. In summary, our findings suggested that 4-MC exerts vasorelaxation by opening KV channels with the involvement of KV7.4.
Collapse
Affiliation(s)
- Patrícia Dias
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Hradec Kralove, 500 05, Czech Republic.
- Division of Outcomes & Translational Sciences, Pelotonia Research Center, The Ohio State University, 2255 Kenny Rd, Columbus, OH, USA
| | - Rudy Salam
- Department of Biophysics and Physical Chemistry, Faculty of Pharmacy, Charles University, Hradec Kralove, 500 05, Czech Republic
- Department of Pharmacy, Faculty of Medicine, Brawijaya University, Malang, Indonesia
| | - Jana Pourová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Hradec Kralove, 500 05, Czech Republic.
| | - Marie Vopršalová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Hradec Kralove, 500 05, Czech Republic.
| | - Lukáš Konečný
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Hradec Kralove, 500 05, Czech Republic.
| | - Eduard Jirkovský
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Hradec Kralove, 500 05, Czech Republic.
| | - Jurjen Duintjer Tebbens
- Department of Biophysics and Physical Chemistry, Faculty of Pharmacy, Charles University, Hradec Kralove, 500 05, Czech Republic
| | - Přemysl Mladěnka
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Hradec Kralove, 500 05, Czech Republic.
| |
Collapse
|
5
|
Falbo F, Carullo G, Panti A, Spiga O, Gianibbi B, Ahmed A, Campiani G, Ramunno A, Aiello F, Fusi F. Exploring the chemical space around chrysin to develop novel vascular Ca V1.2 channel blockers, promising vasorelaxant agents. Arch Pharm (Weinheim) 2024; 357:e2400536. [PMID: 39239992 DOI: 10.1002/ardp.202400536] [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: 07/03/2024] [Revised: 08/18/2024] [Accepted: 08/19/2024] [Indexed: 09/07/2024]
Abstract
The flavonoid chrysin is an effective vascular CaV1.2 channel blocker. The aim of this study was to explore the chemical space around chrysin to identify the structural features that can be modified to develop novel and more effective blockers. Four derivatives (Chrysin 1-4) were synthesised and a functional, electrophysiology and molecular docking approach was pursued to assess their binding mode to CaV1.2 channels and their activity in vascular preparations. Methylation of the 5- and 7-OH of the chrysin backbone caused a marked reduction of the Ca2+ antagonistic potency and efficacy. However, C-8 derivatives showed biophysical features similar to those of the parent compound and, like nicardipine, bound with high affinity to and stabilised the CaV1.2 channel in its inactivated state. The vasorelaxant effects of the four derivatives appeared vessel-specific, addressing the molecules' derivatization towards different targets. In conclusion, the scaffold of chrysin may be considered a valuable starting point for the development of innovative vascular CaV1.2 channel blockers.
Collapse
Affiliation(s)
- Federica Falbo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Gabriele Carullo
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Alice Panti
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Ottavia Spiga
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Beatrice Gianibbi
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Amer Ahmed
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Giuseppe Campiani
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Anna Ramunno
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | - Francesca Aiello
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Fabio Fusi
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, Siena, Italy
| |
Collapse
|
6
|
Ibba R, Sestito S, Ambrosio FA, Marchese E, Costa G, Fiorentino FP, Fusi F, Marchesi I, Polini B, Chiellini G, Alcaro S, Piras S, Carta A. Discovery of pyridoquinoxaline-based new P-gp inhibitors as coadjutant against Multi Drug Resistance in cancer. Eur J Med Chem 2024; 276:116647. [PMID: 38981337 DOI: 10.1016/j.ejmech.2024.116647] [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: 04/05/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/11/2024]
Abstract
Multi-drug resistance (MDR) is a serious challenge in contemporary clinical practice and is mostly responsible for the failure of cancer medication therapies. Several experimental evidence links MDR to the overexpression of the drug efflux transporter P-gp, therefore, the discovery of novel P-glycoprotein inhibitors is required to treat or prevent MDR and to improve the absorption of chemotherapy drugs via the gastrointestinal system. In this work, we explored a series of novel pyridoquinoxaline-based derivatives designed from parental compounds, previously proved active in enhancing anticancer drugs in MDR nasopharyngeal carcinoma (KB). Among them, derivative 10d showed the most potent and selective inhibition of fluorescent dye efflux, if compared to reference compounds (MK-571, Novobiocin, Verapamil), and the highest MDR reversal activity when co-administered with the chemotherapeutic agents Vincristine and Etoposide, at non-cytotoxic concentrations. Molecular modelling predicted the two compound 10d binding mode in a ratio of 2:1 with the target protein. No cytotoxicity was observed in healthy microglia cells and off-target investigations showed the absence of CaV1.2 channel blockade. In summary, our findings indicated that 10d could potentially be a novel therapeutic coadjutant by inhibiting P-gp transport function in vitro, thereby reversing cancer multidrug resistance.
Collapse
Affiliation(s)
- Roberta Ibba
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100, Sassari, Italy.
| | - Simona Sestito
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, 07100, Sassari, Italy.
| | | | - Emanuela Marchese
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Campus "S. Venuta", 88100, Catanzaro, Italy.
| | - Giosuè Costa
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Campus "S. Venuta", 88100, Catanzaro, Italy; Net4Science Academic Spin-Off, University "Magna Græcia" of Catanzaro, Campus "S. Venuta", 88100, Catanzaro, Italy.
| | | | - Fabio Fusi
- Department of Biotechnologies, Chemistry and Pharmacy, University of Siena, 53100, Siena, Italy.
| | | | - Beatrice Polini
- Department of Pathology, University of Pisa, 56100, Pisa, Italy.
| | - Grazia Chiellini
- Department of Pathology, University of Pisa, 56100, Pisa, Italy.
| | - Stefano Alcaro
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Campus "S. Venuta", 88100, Catanzaro, Italy; Net4Science Academic Spin-Off, University "Magna Græcia" of Catanzaro, Campus "S. Venuta", 88100, Catanzaro, Italy.
| | - Sandra Piras
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100, Sassari, Italy.
| | - Antonio Carta
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100, Sassari, Italy.
| |
Collapse
|
7
|
Gautheron G, Péraldi-Roux S, Vaillé J, Belhadj S, Patyra A, Bayle M, Youl E, Omhmmed S, Guyot M, Cros G, Guichou JF, Uzan B, Movassat J, Quignard JF, Neasta J, Oiry C. The flavonoid resokaempferol improves insulin secretion from healthy and dysfunctional pancreatic β-cells. Br J Pharmacol 2024. [PMID: 39327688 DOI: 10.1111/bph.17304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 06/20/2024] [Accepted: 07/09/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND AND PURPOSE The pharmacology of flavonoids on β-cell function is largely undefined especially in the context of defective secretion of insulin. We sought to identify flavonoids that increased the insulin-secreting function of β-cells and to explore the underlying mechanisms. EXPERIMENTAL APPROACH INS-1 β-cells in culture and islets of Langerhans isolated from control and diabetic male rats were used for insulin secretion experiments. Pharmacological and electrophysiological approaches were used for mechanistic studies. KEY RESULTS Among a set of flavonoids, exposure of INS-1 β-cells to resokaempferol (ResoK) enhanced glucose-stimulated insulin secretion and therefore we further characterised its activity and its pharmacological mechanism. ResoK glucose-dependently enhanced insulin secretion in INS-1 β-cells and pancreatic islets isolated from rats. Mechanistically, whole cell patch clamp recordings in INS-1 cells showed that ResoK rapidly and dose-dependently enhanced the L-type Ca2+ current whereas it was inactive towards T-type Ca2+ current. Accordingly, pharmacological inhibition of L-type Ca2+ current but not T-type Ca2+ current blocked the effects of ResoK on glucose-stimulated insulin secretion. ResoK was still active on dysfunctional β-cells as it ameliorated glucose-stimulated insulin secretion in glucotoxicity-induced dysfunctional INS-1 cells and in pancreatic islets isolated from diabetic rats. CONCLUSION AND IMPLICATIONS ResoK is a glucose-dependent activator of insulin secretion. Our results indicated that the effects of ResoK on insulin secretion involved its capacity to stimulate L-type Ca2+ currents in cultured β-cells. As ResoK was also effective on dysfunctional β-cells, our work provides a new approach to stimulating insulin secretion, using compounds based on the structure of ResoK.
Collapse
Affiliation(s)
| | | | - Justine Vaillé
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Sahla Belhadj
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Andrzej Patyra
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
- Department of Pharmaceutical Biology, Medical University of Warsaw, Warsaw, Poland
| | - Morgane Bayle
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Estelle Youl
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | | | - Mélanie Guyot
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Gérard Cros
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | | | - Benjamin Uzan
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Jamileh Movassat
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, France
| | - Jean-François Quignard
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, Pessac, France
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, Pessac, France
| | - Jérémie Neasta
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Catherine Oiry
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| |
Collapse
|
8
|
Mastroeni P, Trezza A, Geminiani M, Frusciante L, Visibelli A, Santucci A. HGA Triggers SAA Aggregation and Accelerates Fibril Formation in the C20/A4 Alkaptonuria Cell Model. Cells 2024; 13:1501. [PMID: 39273071 PMCID: PMC11394027 DOI: 10.3390/cells13171501] [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: 08/06/2024] [Revised: 08/31/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
Alkaptonuria (AKU) is a rare autosomal recessive metabolic disorder caused by mutations in the homogentisate 1,2-dioxygenase (HGD) gene, leading to the accumulation of homogentisic acid (HGA), causing severe inflammatory conditions. Recently, the presence of serum amyloid A (SAA) has been reported in AKU tissues, classifying AKU as novel secondary amyloidosis; AA amyloidosis is characterized by the extracellular tissue deposition of fibrils composed of fragments of SAA. AA amyloidosis may complicate several chronic inflammatory conditions, like rheumatoid arthritis, ankylosing spondylitis, inflammatory bowel disease, chronic infections, neoplasms, etc. Treatments of AA amyloidosis relieve inflammatory disorders by reducing SAA concentrations; however, no definitive therapy is currently available. SAA regulation is a crucial step to improve AA secondary amyloidosis treatments. Here, applying a comprehensive in vitro and in silico approach, we provided evidence that HGA is a disruptor modulator of SAA, able to enhance its polymerization, fibril formation, and aggregation upon SAA/SAP colocalization. In silico studies deeply dissected the SAA misfolding molecular pathway and SAA/HGA binding, suggesting novel molecular insights about it. Our results could represent an important starting point for identifying novel therapeutic strategies in AKU and AA secondary amyloidosis-related diseases.
Collapse
Affiliation(s)
- Pierfrancesco Mastroeni
- ONE-HEALTH Lab, Department of Biotechnology, Chemistry and Pharmacy, University of Siena Via Aldo Moro, 53100 Siena, Italy; (P.M.); (A.T.); (L.F.); (A.V.); (A.S.)
| | - Alfonso Trezza
- ONE-HEALTH Lab, Department of Biotechnology, Chemistry and Pharmacy, University of Siena Via Aldo Moro, 53100 Siena, Italy; (P.M.); (A.T.); (L.F.); (A.V.); (A.S.)
| | - Michela Geminiani
- ONE-HEALTH Lab, Department of Biotechnology, Chemistry and Pharmacy, University of Siena Via Aldo Moro, 53100 Siena, Italy; (P.M.); (A.T.); (L.F.); (A.V.); (A.S.)
| | - Luisa Frusciante
- ONE-HEALTH Lab, Department of Biotechnology, Chemistry and Pharmacy, University of Siena Via Aldo Moro, 53100 Siena, Italy; (P.M.); (A.T.); (L.F.); (A.V.); (A.S.)
| | - Anna Visibelli
- ONE-HEALTH Lab, Department of Biotechnology, Chemistry and Pharmacy, University of Siena Via Aldo Moro, 53100 Siena, Italy; (P.M.); (A.T.); (L.F.); (A.V.); (A.S.)
| | - Annalisa Santucci
- ONE-HEALTH Lab, Department of Biotechnology, Chemistry and Pharmacy, University of Siena Via Aldo Moro, 53100 Siena, Italy; (P.M.); (A.T.); (L.F.); (A.V.); (A.S.)
- MetabERN, Department of Biotechnology, Chemistry and Pharmacy, University of Siena Via Aldo Moro, 53100 Siena, Italy
| |
Collapse
|
9
|
Son NT, Gianibbi B, Panti A, Spiga O, Bastos JK, Fusi F. Vasorelaxant Activity of (2S)-Sakuranetin and Other Flavonoids Isolated from the Green Propolis of the Caatinga Mimosa tenuiflora. PLANTA MEDICA 2024; 90:454-468. [PMID: 38599606 DOI: 10.1055/a-2294-7042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Some in vitro and in vivo evidence is consistent with the cardiovascular beneficial activity of propolis. As the single actors responsible for this effect have never been identified, an in-depth investigation of flavonoids isolated from the green propolis of the Caatinga Mimosa tenuiflora was performed and their mechanism of action was described. A comprehensive electrophysiology, functional, and molecular docking approach was applied. Most flavanones and flavones were effective CaV1.2 channel blockers with a potency order of (2S)-sakuranetin > eriodictyol-7,3'-methyl ether > quercetin 3-methyl ether > 5,4'-dihydroxy-6,7-dimethoxyflavanone > santin > axillarin > penduletin > kumatakenin, ermanin and viscosine being weak or modest stimulators. Except for eriodictyol 5-O-methyl ether, all the flavonoids were also effective spasmolytic agents of vascular rings, kumatakenin and viscosine also showing an endothelium-dependent activity. (2S)-Sakuranetin also stimulated KCa1.1 channels both in single myocytes and vascular rings. In silico analysis provided interesting insights into the mode of action of (2S)-sakuranetin within both CaV1.2 and KCa1.1 channels. The green propolis of the Caatinga Mimosa tenuiflora is a valuable source of multi-target vasoactive flavonoids: this evidence reinforces its nutraceutical value in the cardiovascular disease prevention arena.
Collapse
Affiliation(s)
- Ninh The Son
- Institute of Chemistry, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
- Department of Chemistry, Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Caugiay, Hanoi, Vietnam
| | - Beatrice Gianibbi
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Siena, Italy
| | - Alice Panti
- Dipartimento di Scienze della Vita, Università di Siena, Siena, Italy
| | - Ottavia Spiga
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Siena, Italy
| | - Jairo Kenupp Bastos
- School of Pharmaceutical Sciences of Ribeirão Preto, Ribeirão Preto, University of São Paulo, Ribeirão Preto-SP, Brazil
| | - Fabio Fusi
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Siena, Italy
| |
Collapse
|
10
|
Son NT, Gianibbi B, Panti A, Spiga O, Bastos JK, Fusi F. 3,3'-O-dimethylquercetin: A bi-functional vasodilator isolated from green propolis of the Caatinga Mimosa tenuiflora. Eur J Pharmacol 2024; 967:176400. [PMID: 38331336 DOI: 10.1016/j.ejphar.2024.176400] [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: 09/19/2023] [Revised: 01/29/2024] [Accepted: 02/06/2024] [Indexed: 02/10/2024]
Abstract
In the search for novel, bi-functional compounds acting as CaV1.2 channel blockers and K+ channel stimulators, which represent an effective therapy for hypertension, 3,3'-O-dimethylquercetin was isolated for the first time from Brazilian Caatinga green propolis. Its effects were investigated through electrophysiological, functional, and computational approaches. In rat tail artery myocytes, 3,3'-O-dimethylquercetin blocked Ba2+ currents through CaV1.2 channels (IBa1.2) in a concentration-dependent manner, with the inhibition being reversed upon washout. The compound also shifted the voltage dependence of the steady-state inactivation curve to more negative potentials without affecting the slope of the inactivation and activation curves. Furthermore, the flavonoid stimulated KCa1.1 channel currents (IKCa1.1). In silico simulations provided additional evidence for the binding of 3,3'-O-dimethylquercetin to KCa1.1 and CaV1.2 channels and elucidated its mechanism of action. In depolarized rat tail artery rings, the flavonoid induced a concentration-dependent relaxation. Moreover, in rat aorta rings its antispasmodic effect was inversely related to the transmembrane K+ gradient. In conclusion, 3,3'-O-dimethylquercetin demonstrates effective in vitro vasodilatory properties, encouraging the exploration of its scaffold to develop novel derivatives for potential use in the treatment of hypertension.
Collapse
Affiliation(s)
- Ninh The Son
- School of Pharmaceutical Sciences of Ribeirão Preto, Ribeirão Preto, University of São Paulo, Avenida Professor Doutor Zeferino Vaz, S/N, 14040-903, Ribeirão Preto-SP, Brazil; Institute of Chemistry, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet, Caugiay, Hanoi, 10000, Vietnam; Department of Chemistry, Graduate University of Science and Technology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Caugiay, Hanoi, 10000, Vietnam
| | - Beatrice Gianibbi
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro 2, 53100, Siena, Italy
| | - Alice Panti
- Dipartimento di Scienze della Vita, Università di Siena, Via A. Moro 2, 53100, Siena, Italy
| | - Ottavia Spiga
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro 2, 53100, Siena, Italy
| | - Jairo Kenupp Bastos
- School of Pharmaceutical Sciences of Ribeirão Preto, Ribeirão Preto, University of São Paulo, Avenida Professor Doutor Zeferino Vaz, S/N, 14040-903, Ribeirão Preto-SP, Brazil.
| | - Fabio Fusi
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro 2, 53100, Siena, Italy.
| |
Collapse
|
11
|
Ahmed A, Trezza A, Gentile M, Paccagnini E, Panti A, Lupetti P, Spiga O, Bova S, Fusi F. Dynamin-independent Ca V1.2 and K Ca1.1 channels regulation and vascular tone modulation by the mitochondrial fission inhibitors dynasore and dyngo-4a. Eur J Pharmacol 2023; 951:175786. [PMID: 37179045 DOI: 10.1016/j.ejphar.2023.175786] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 05/03/2023] [Accepted: 05/10/2023] [Indexed: 05/15/2023]
Abstract
A role for mitochondrial fission in vascular contraction has been proposed based on the vasorelaxant activity of the dynamin (and mitochondrial fission) inhibitors mdivi-1 and dynasore. However, mdivi-1 is capable to inhibit Ba2+ currents through CaV1.2 channels (IBa1.2), stimulate KCa1.1 channel currents (IKCa1.1), and modulate pathways key to the maintenance of vessel active tone in a dynamin-independent manner. Using a multidisciplinary approach, the present study demonstrates that dynasore, like mdivi-1, is a bi-functional vasodilator, blocking IBa1.2 and stimulating IKCa1.1 in rat tail artery myocytes, as well as promoting relaxation of rat aorta rings pre-contracted by either high K+ or phenylephrine. Conversely, its analogue dyngo-4a, though inhibiting mitochondrial fission triggered by phenylephrine and stimulating IKCa1.1, did not affect IBa1.2 but potentiated both high K+- and phenylephrine-induced contractions. Docking and molecular dynamics simulations identified the molecular basis supporting the different activity of dynasore and dyngo-4a at CaV1.2 and KCa1.1 channels. Mito-tempol only partially counteracted the effects of dynasore and dyngo-4a on phenylephrine-induced tone. In conclusion, the present data, along with previous observations (Ahmed et al., 2022) rise caution for the use of dynasore, mdivi-1, and dyngo-4a as tools to investigate the role of mitochondrial fission in vascular contraction: to this end, a selective dynamin inhibitor and/or a different experimental approach are needed.
Collapse
Affiliation(s)
- Amer Ahmed
- Dipartimento di Scienze della Vita, Università di Siena, via A. Moro 2, 53100, Siena, Italy
| | - Alfonso Trezza
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, via A. Moro 2, 53100, Siena, Italy
| | - Mariangela Gentile
- Dipartimento di Scienze della Vita, Università di Siena, via A. Moro 2, 53100, Siena, Italy
| | - Eugenio Paccagnini
- Dipartimento di Scienze della Vita, Università di Siena, via A. Moro 2, 53100, Siena, Italy
| | - Alice Panti
- Dipartimento di Scienze della Vita, Università di Siena, via A. Moro 2, 53100, Siena, Italy
| | - Pietro Lupetti
- Dipartimento di Scienze della Vita, Università di Siena, via A. Moro 2, 53100, Siena, Italy
| | - Ottavia Spiga
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, via A. Moro 2, 53100, Siena, Italy
| | - Sergio Bova
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Fabio Fusi
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, via A. Moro 2, 53100, Siena, Italy.
| |
Collapse
|
12
|
Chen M, Wu Q. Roles and mechanisms of natural drugs on sinus node dysfunction. Biomed Pharmacother 2023; 164:114777. [PMID: 37229801 DOI: 10.1016/j.biopha.2023.114777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/18/2023] [Accepted: 04/23/2023] [Indexed: 05/27/2023] Open
Abstract
Sinus node dysfunction is a common arrhythmia disorder with a high incidence and significant social and economic burden. Currently, there are no effective drugs for treating chronic sinus node dysfunction. The disease is associated with ion channel disturbances caused by aging, fibrosis, inflammation, oxidative stress, and autonomic dysfunction. Natural active substances and Chinese herbal medicines have been widely used and extensively studied in the medical community for the treatment of arrhythmias. Multiple studies have demonstrated that various active ingredients and Chinese herbal medicines, such as astragaloside IV, quercetin, and ginsenosides, exhibit antioxidant effects, reduce fibrosis, and maintain ion channel stability, providing promising drugs for treating sinus node dysfunction. This article summarizes the research progress on natural active ingredients and Chinese herbal formulas that regulate sick sinoatrial node function, providing valuable references for the treatment of sinus node dysfunction.
Collapse
Affiliation(s)
- Meilian Chen
- Quanzhou Hospital of Traditional Chinese Medicine, Fujian 362000, China
| | - Qiaomin Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
13
|
Carullo G, Falbo F, Ahmed A, Trezza A, Gianibbi B, Nicolotti O, Campiani G, Aiello F, Saponara S, Fusi F. Artificial intelligence-driven identification of morin analogues acting as Ca V1.2 channel blockers: Synthesis and biological evaluation. Bioorg Chem 2023; 131:106326. [PMID: 36563413 DOI: 10.1016/j.bioorg.2022.106326] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/02/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Morin is a vasorelaxant flavonoid, whose activity is ascribable to CaV1.2 channel blockade that, however, is weak as compared to that of clinically used therapeutic agents. A conventional strategy to circumvent this drawback is to synthesize new derivatives differently decorated and, in this context, morin-derivatives able to interact with CaV1.2 channels were found by employing the potential of PLATO in target fishing and reverse screening. Three different derivatives (5a-c) were selected as promising tools, synthesized, and investigated in in vitro functional studies using rat aorta rings and rat tail artery myocytes. 5a-c were found more effective vasorelaxant agents than the naturally occurring parent compound and antagonized both electro- and pharmaco-mechanical coupling in an endothelium-independent manner. 5a, the series' most potent, reduced also Ca2+ mobilization from intracellular store sites. Furthermore, 5a≈5c > 5b inhibited Ba2+ current through CaV1.2 channels. However, compound 5a caused also a concentration-dependent inhibition of KCa1.1 channel currents.
Collapse
Affiliation(s)
- Gabriele Carullo
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Federica Falbo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Ed. Polifunzionale, 87036, Rende (CS), Italy
| | - Amer Ahmed
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Alfonso Trezza
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Beatrice Gianibbi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Orazio Nicolotti
- Department of Pharmacy- Drug Sciences, University of Bari "Aldo Moro", Via Orabona 4, 70125 Bari, Italy
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Francesca Aiello
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Ed. Polifunzionale, 87036, Rende (CS), Italy.
| | - Simona Saponara
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Fabio Fusi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| |
Collapse
|
14
|
Targeting neuronal calcium channels and GSK3β for Alzheimer's disease with naturally-inspired Diels-Alder adducts. Bioorg Chem 2022; 129:106152. [PMID: 36155094 DOI: 10.1016/j.bioorg.2022.106152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/08/2022] [Accepted: 09/10/2022] [Indexed: 11/22/2022]
Abstract
The complexity of neurodegenerative diseases, among which Alzheimer's disease plays a pivotal role, poses one of the tough therapeutic challenges of present time. In this perspective, a multitarget approach appears as a promising strategy to simultaneously interfere with different defective pathways. In this paper, a structural simplification plan was performed on our previously reported multipotent polycyclic compounds, in order to obtain a simpler pharmacophoric central core with improved pharmacokinetic properties, while maintaining the modulating activity on neuronal calcium channels and glycogen synthase kinase 3-beta (GSK-3β), as validated targets to combat Alzheimer's disease. The molecular pruning approach applied here led to tetrahydroisoindole-dione (1), tetrahydromethanoisoindole-dione (2) and tetrahydroepoxyisoindole-dione (3) structures, easily affordable by Diels-Alder cycloaddition. Preliminary data indicated structure 3 as the most appropriate, thus a SAR study was performed by introducing different substituents, selected on the basis of the commercial availability of the furan derivatives required for the synthetic procedure. The results indicated compound 10 as a promising, structurally atypical, safe and BBB-penetrating Cav modulator, inhibiting both L- and N-calcium channels, likely responsible for the Ca2+ overload observed in Alzheimer's disease. In a multitarget perspective, compound 11 appeared as an effective prototype, endowed with improved Cav inhibitory activity, with respect to the reference drug nifedipine, and encouraging modulating activity on GSK-3β.
Collapse
|
15
|
Carullo G, Saponara S, Ahmed A, Gorelli B, Mazzotta S, Trezza A, Gianibbi B, Campiani G, Fusi F, Aiello F. Novel Labdane Diterpenes-Based Synthetic Derivatives: Identification of a Bifunctional Vasodilator That Inhibits Ca V1.2 and Stimulates K Ca1.1 Channels. Mar Drugs 2022; 20:md20080515. [PMID: 36005518 PMCID: PMC9410420 DOI: 10.3390/md20080515] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 07/30/2022] [Accepted: 08/11/2022] [Indexed: 11/26/2022] Open
Abstract
Sesquiterpenes such as leucodin and the labdane-type diterpene manool are natural compounds endowed with remarkably in vitro vasorelaxant and in vivo hypotensive activities. Given their structural similarity with the sesquiterpene lactone (+)-sclareolide, this molecule was selected as a scaffold to develop novel vasoactive agents. Functional, electrophysiology, and molecular dynamics studies were performed. The opening of the five-member lactone ring in the (+)-sclareolide provided a series of labdane-based small molecules, promoting a significant in vitro vasorelaxant effect. Electrophysiology data identified 7 as a CaV1.2 channel blocker and a KCa1.1 channel stimulator. These activities were also confirmed in the intact vascular tissue. The significant antagonism caused by the CaV1.2 channel agonist Bay K 8644 suggested that 7 might interact with the dihydropyridine binding site. Docking and molecular dynamic simulations provided the molecular basis of the CaV1.2 channel blockade and KCa1.1 channel stimulation produced by 7. Finally, 7 reduced coronary perfusion pressure and heart rate, while prolonging conduction and refractoriness of the atrioventricular node, likely because of its Ca2+ antagonism. Taken together, these data indicate that the labdane scaffold represents a valuable starting point for the development of new vasorelaxant agents endowed with negative chronotropic properties and targeting key pathways involved in the pathophysiology of hypertension and ischemic cardiomyopathy.
Collapse
Affiliation(s)
- Gabriele Carullo
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Simona Saponara
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Amer Ahmed
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Beatrice Gorelli
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Sarah Mazzotta
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Alfonso Trezza
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Beatrice Gianibbi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Fabio Fusi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
- Correspondence: (F.F.); (F.A.)
| | - Francesca Aiello
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Edif. Polifunzionale, 87036 Arcavacata di Rende, Italy
- Correspondence: (F.F.); (F.A.)
| |
Collapse
|
16
|
Ahmed A, Trezza A, Gentile M, Paccagnini E, Lupetti P, Spiga O, Bova S, Fusi F. The drp-1-mediated mitochondrial fission inhibitor mdivi-1 impacts the function of ion channels and pathways underpinning vascular smooth muscle tone. Biochem Pharmacol 2022; 203:115205. [DOI: 10.1016/j.bcp.2022.115205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/07/2022] [Accepted: 08/02/2022] [Indexed: 11/24/2022]
|
17
|
Cuong NM, Son NT, Nhan NT, Fukuyama Y, Ahmed A, Saponara S, Trezza A, Gianibbi B, Vigni G, Spiga O, Fusi F. Vietnamese Dalbergia tonkinensis: A Promising Source of Mono- and Bifunctional Vasodilators. Molecules 2022; 27:molecules27144505. [PMID: 35889386 PMCID: PMC9324545 DOI: 10.3390/molecules27144505] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 02/04/2023] Open
Abstract
Hypertension is a risk factor for cardiovascular diseases, which are the main cause of morbidity and mortality in the world. In the search for new molecules capable of targeting KCa1.1 and CaV1.2 channels, the expression of which is altered in hypertension, the in vitro vascular effects of a series of flavonoids extracted from the heartwoods, roots, and leaves of Dalbergia tonkinensis Prain, widely used in traditional medicine, were assessed. Rat aorta rings, tail artery myocytes, and docking and molecular dynamics simulations were used to analyse their effect on these channels. Formononetin, orobol, pinocembrin, and biochanin A showed a marked myorelaxant activity, particularly in rings stimulated by moderate rather than high KCl concentrations. Ba2+ currents through CaV1.2 channels (IBa1.2) were blocked in a concentration-dependent manner by sativanone, 3′-O-methylviolanone, pinocembrin, and biochanin A, while it was stimulated by ambocin. Sativanone, dalsissooside, and eriodictyol inhibited, while tectorigenin 7-O-[β-D-apiofuranosyl-(1→6)-β-D-glucopyranoside], ambocin, butin, and biochanin A increased IKCa1.1. In silico analyses showed that biochanin A, sativanone, and pinocembrin bound with high affinity in target-sensing regions of both channels, providing insight into their potential mechanism of action. In conclusion, Dalbergia tonkinensis is a valuable source of mono- and bifunctional, vasoactive scaffolds for the development of novel antihypertensive drugs.
Collapse
Affiliation(s)
- Nguyen Manh Cuong
- Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet, Caugiay, Hanoi 122100, Vietnam
- Correspondence: (N.M.C.); (F.F.)
| | - Ninh The Son
- Institute of Chemistry, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet, Caugiay, Hanoi 122100, Vietnam;
| | - Ngu Truong Nhan
- Faculty of Science and Technology, Tay Nguyen University, 567 Le Duan, Ea Tam, Buon Ma Thuot 630000, Vietnam;
| | - Yoshiyasu Fukuyama
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima 770-8514, Japan;
| | - Amer Ahmed
- Dipartimento di Scienze della Vita, Università di Siena, Via A. Moro 2, 53100 Siena, Italy; (A.A.); (S.S.)
| | - Simona Saponara
- Dipartimento di Scienze della Vita, Università di Siena, Via A. Moro 2, 53100 Siena, Italy; (A.A.); (S.S.)
| | - Alfonso Trezza
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro 2, 53100 Siena, Italy; (A.T.); (B.G.); (G.V.); (O.S.)
| | - Beatrice Gianibbi
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro 2, 53100 Siena, Italy; (A.T.); (B.G.); (G.V.); (O.S.)
| | - Ginevra Vigni
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro 2, 53100 Siena, Italy; (A.T.); (B.G.); (G.V.); (O.S.)
| | - Ottavia Spiga
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro 2, 53100 Siena, Italy; (A.T.); (B.G.); (G.V.); (O.S.)
| | - Fabio Fusi
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro 2, 53100 Siena, Italy; (A.T.); (B.G.); (G.V.); (O.S.)
- Correspondence: (N.M.C.); (F.F.)
| |
Collapse
|