1
|
Es-Sounni B, Harboul K, Mouhib A, Alanazi AS, Hefnawy M, Bakhouch M, Benali T, Hammani K, Mazoir N, El Yazidi M, Benharref A, Fahim M. Ruthenium(II) Complex-Based Tetradentate Schiff Bases: Synthesis, Spectroscopic, Antioxidant, and Antibacterial Investigations. Int J Mol Sci 2024; 25:7879. [PMID: 39063120 PMCID: PMC11277530 DOI: 10.3390/ijms25147879] [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: 06/01/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
In this work, we describe the synthesis of novel Ruthenium (II) complex-based salen Schiff bases. The obtained Ruthenium (II) complexes are characterized using usual spectroscopic and spectrometric techniques, viz., IR, UV-Vis, NMR (1H and 13C), powder X-ray diffraction, and HRMS. Further techniques, such as DTA-TGA and elemental analysis, are used to well establish the structure of the obtained complexes. Octahedral geometries are tentatively proposed for the new Ru(II) complexes. The measured molar conductance for the Ruthenium (II) complexes shows their electrolytic nature (4.24-4.44 S/m). The new Ru(II) complexes are evaluated for their antioxidant and antibacterial activities. The DPPH radical scavenging, FRAP, and total antioxidant capacity (TAC) assays show that the obtained complexes are more potent than the used positive control. They also exhibit promising antibacterial responses against pathogen bacteria: [RuH2L3Cl2] exhibits an important inhibition against Bacillus subtilis DSM 6633, with an inhibition zone of 21 ± 1.41 mm with an MIC value of 0.39 mg/mL, and Proteus mirabilis INH, with 16.50 ± 0.70 mm and an MIC value of 0.78 mg/mL, while [RuH2L2Cl2] exerts interesting antibacterial effects versus Bacillus subtilis DSM 6633 (21 ± 1.41 mm) and Proteus mirabilis INH (25.5 ± 0.70 mm) with equal MIC values of 0.97 mg/mL.
Collapse
Affiliation(s)
- Bouchra Es-Sounni
- Laboratory of Innovative Materials and Biotechnology of Naturel Resources, Faculty of Sciences, Moulay Ismail University, Meknes 50000, Morocco; (B.E.-S.)
| | - Kaoutar Harboul
- Laboratory of Natural Resources and Environment, Polydisciplinary Faculty of Taza, Sidi Mohamed Ben Abdellah University of Fez, Taza 30050, Morocco (T.B.); (K.H.)
| | - Ayoub Mouhib
- Bioorganic Chemistry Team, Department of Chemistry, Faculty of Sciences, Chouaïb Doukkali University, El Jadida 24000, Morocco
| | - Ashwag S. Alanazi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh 1167, Saudi Arabia;
| | - Mohamed Hefnawy
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Mohamed Bakhouch
- Bioorganic Chemistry Team, Department of Chemistry, Faculty of Sciences, Chouaïb Doukkali University, El Jadida 24000, Morocco
| | - Taoufiq Benali
- Laboratory of Natural Resources and Environment, Polydisciplinary Faculty of Taza, Sidi Mohamed Ben Abdellah University of Fez, Taza 30050, Morocco (T.B.); (K.H.)
- Environment and Health Team, Polydisciplinary Faculty of Safi, Cadi Ayyad University, Safi 46000, Morocco
| | - Khalil Hammani
- Laboratory of Natural Resources and Environment, Polydisciplinary Faculty of Taza, Sidi Mohamed Ben Abdellah University of Fez, Taza 30050, Morocco (T.B.); (K.H.)
| | - Noureddine Mazoir
- Bioorganic Chemistry Team, Department of Chemistry, Faculty of Sciences, Chouaïb Doukkali University, El Jadida 24000, Morocco
| | - Mohamed El Yazidi
- Engineering Laboratory of Organometallic and Molecular Materials and Environment, Faculty of Sciences Dhar El Mahraz, University Sidi Mohamed Ben Abdellah, Fez 30000, Morocco;
| | - Ahmed Benharref
- Laboratory of Natural Substances Chemistry, Faculty of Sciences Semlalia, Cadi Ayyad University, Marrakech 40000, Morocco;
| | - Mohammed Fahim
- Laboratory of Innovative Materials and Biotechnology of Naturel Resources, Faculty of Sciences, Moulay Ismail University, Meknes 50000, Morocco; (B.E.-S.)
| |
Collapse
|
2
|
Salvan da Rosa J, Bramorski Mohr ET, Lubschinski TL, Vieira GN, Rossa TA, Mandolesi Sá M, Dalmarco EM. Interference in Macrophage Balance (M1/M2): The Mechanism of Action Responsible for the Anti-Inflammatory Effect of a Fluorophenyl-Substituted Imidazole. Mediators Inflamm 2024; 2024:9528976. [PMID: 38405621 PMCID: PMC10894048 DOI: 10.1155/2024/9528976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/19/2024] [Accepted: 02/03/2024] [Indexed: 02/27/2024] Open
Abstract
Traditionally, the treatment of inflammatory conditions has focused on the inhibition of inflammatory mediator production; however, many conditions are refractory to this classical approach. Recently, an alternative has been presented by researchers to solve this problem: The immunomodulation of cells closely related to inflammation. Hence, macrophages, a critical key in both innate and acquired immunity, have been presented as an alternative target for the development of new medicines. In this work, we tested the fluorophenyl-imidazole for its anti-inflammatory activity and possible immunomodulatory effect on RAW 264.7 macrophages. We also evaluated the anti-inflammatory effect of the compound, and the macrophage repolarization to M2 was confirmed by the ability of the compound to reduce the M1 markers TNF-α, IL-6, MCP-1, IL-12p70, IFN-γ, and TLR4, the high levels of p65 phosphorylated, iNOS and COX-2 mRNA expression, and the fact that the compound was not able to induce the production of M1 markers when used in macrophages without lipopolysaccharide (LPS) stimulation. Moreover, fluorophenyl-imidazole had the ability to increase the M2 markers IL-4, IL-13, CD206, apoptosis and phagocytosis levels, arginase-1, and FIZZ-1 mRNA expression before LPS stimulation. Similarly, it was also able to induce the production of these same M2 markers in macrophages without being induced with LPS. These results reinforce the affirmation that the fluorophenyl-imidazole has an important anti-inflammatory effect and demonstrates that this effect is due to immunomodulatory activity, having the ability to trigger a repolarization of macrophages from M1 to M2a. These facts suggest that this molecule could be used as an alternative scaffold for the development of a new medicine to treat inflammatory conditions, where the anti-inflammatory and proregenerative properties of M2a macrophages are desired.
Collapse
Affiliation(s)
- Julia Salvan da Rosa
- Department of Clinical Analysis, Center for Health Sciences, Campus Universitário—Trindade, Universidade Federal de Santa Catarina, Florianópolis 88040-970, SC, Brazil
| | - Eduarda Talita Bramorski Mohr
- Department of Clinical Analysis, Center for Health Sciences, Campus Universitário—Trindade, Universidade Federal de Santa Catarina, Florianópolis 88040-970, SC, Brazil
| | - Tainá Larissa Lubschinski
- Department of Clinical Analysis, Center for Health Sciences, Campus Universitário—Trindade, Universidade Federal de Santa Catarina, Florianópolis 88040-970, SC, Brazil
| | - Guilherme Nicácio Vieira
- Department of Clinical Analysis, Center for Health Sciences, Campus Universitário—Trindade, Universidade Federal de Santa Catarina, Florianópolis 88040-970, SC, Brazil
| | - Thais Andreia Rossa
- Department of Chemistry, Center for Physical and Mathematical Sciences, Campus Universitário—Trindade, Universidade Federal de Santa Catarina, Florianópolis 88040-970, SC, Brazil
| | - Marcus Mandolesi Sá
- Department of Chemistry, Center for Physical and Mathematical Sciences, Campus Universitário—Trindade, Universidade Federal de Santa Catarina, Florianópolis 88040-970, SC, Brazil
| | - Eduardo Monguilhott Dalmarco
- Department of Clinical Analysis, Center for Health Sciences, Campus Universitário—Trindade, Universidade Federal de Santa Catarina, Florianópolis 88040-970, SC, Brazil
| |
Collapse
|
3
|
Giacomazzo GE, Conti L, Fagorzi C, Pagliai M, Andreini C, Guerri A, Perito B, Mengoni A, Valtancoli B, Giorgi C. Ruthenium(II) Polypyridyl Complexes and Metronidazole Derivatives: A Powerful Combination in the Design of Photoresponsive Antibacterial Agents Effective under Hypoxic Conditions. Inorg Chem 2023; 62:7716-7727. [PMID: 37163381 DOI: 10.1021/acs.inorgchem.3c00214] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Ruthenium(II) polypyridyl complexes (RPCs) are gaining momentum in photoactivated chemotherapy (PACT), thanks to the possibility of overcoming the classical reliance on molecular oxygen of photodynamic therapy while preserving the selective drug activation by using light. However, notwithstanding the intriguing perspectives, the translation of such an approach in the development of new antimicrobials has been only barely considered. Herein, MTZH-1 and MTZH-2, two novel analogues of metronidazole (MTZ), a mainstay drug in the treatment of anaerobic bacterial infections, were designed and inserted in the strained ruthenium complexes [Ru(tpy)(dmp)(MTZ-1)]PF6 (Ru2) and [Ru(tpy)(dmp)(MTZ-2)]PF6 (Ru3) (tpy = terpyridine, dmp = 2,9-dimethyl-1,10-phenanthroline) (Chart 1). Analogously to the parental compound [Ru(tpy)(dmp)(5NIM)]PF6 (Ru1) (5-nitroimidazolate), the Ru(II)-imidazolate coordination of MTZ derivatives resulted in promising Ru(II) photocages, capable to easily unleash the bioactive ligands upon light irradiation and increase the antibacterial activity against Bacillus subtilis, which was chosen as a model of Gram-positive bacteria. The photoreleased 5-nitroimidazole-based ligands led to remarkable phototoxicities under hypoxic conditions (<1% O2), with the lead compound Ru3 that exhibited the highest potency across the series, being comparable to the one of the clinical drug MTZ. Besides, the chemical architectures of MTZ derivatives made their interaction with NimAunfavorable, being NimA a model of reductases responsible for bacterial resistance against 5-nitroimidazole-based antibiotics, thus hinting at their possible use to combat antimicrobial resistance. This work may therefore provide fundamental knowledge in the design of novel photoresponsive tools to be used in the fight against infectious diseases. For the first time, the effectiveness of the "photorelease antimicrobial therapy" under therapeutically relevant hypoxic conditions was demonstrated.
Collapse
Affiliation(s)
- Gina Elena Giacomazzo
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, Sesto Fiorentino, 50019 Firenze, Italy
| | - Luca Conti
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, Sesto Fiorentino, 50019 Firenze, Italy
| | - Camilla Fagorzi
- Department of Biology, University of Florence, Via Madonna del Piano 6, Sesto Fiorentino, 50019 Firenze, Italy
| | - Marco Pagliai
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, Sesto Fiorentino, 50019 Firenze, Italy
| | - Claudia Andreini
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, Sesto Fiorentino, 50019 Firenze, Italy
- Magnetic Resonance Center (CERM), University of Florence, Via Luigi Sacconi 6, Sesto Fiorentino, 50019 Firenze, Italy
| | - Annalisa Guerri
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, Sesto Fiorentino, 50019 Firenze, Italy
| | - Brunella Perito
- Department of Biology, University of Florence, Via Madonna del Piano 6, Sesto Fiorentino, 50019 Firenze, Italy
| | - Alessio Mengoni
- Department of Biology, University of Florence, Via Madonna del Piano 6, Sesto Fiorentino, 50019 Firenze, Italy
| | - Barbara Valtancoli
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, Sesto Fiorentino, 50019 Firenze, Italy
| | - Claudia Giorgi
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, Sesto Fiorentino, 50019 Firenze, Italy
| |
Collapse
|
4
|
Li SR, Tan YM, Zhang L, Zhou CH. Comprehensive Insights into Medicinal Research on Imidazole-Based Supramolecular Complexes. Pharmaceutics 2023; 15:1348. [PMID: 37242590 PMCID: PMC10222694 DOI: 10.3390/pharmaceutics15051348] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
The electron-rich five-membered aromatic aza-heterocyclic imidazole, which contains two nitrogen atoms, is an important functional fragment widely present in a large number of biomolecules and medicinal drugs; its unique structure is beneficial to easily bind with various inorganic or organic ions and molecules through noncovalent interactions to form a variety of supramolecular complexes with broad medicinal potential, which is being paid an increasing amount of attention regarding more and more contributions to imidazole-based supramolecular complexes for possible medicinal application. This work gives systematical and comprehensive insights into medicinal research on imidazole-based supramolecular complexes, including anticancer, antibacterial, antifungal, antiparasitic, antidiabetic, antihypertensive, and anti-inflammatory aspects as well as ion receptors, imaging agents, and pathologic probes. The new trend of the foreseeable research in the near future toward imidazole-based supramolecular medicinal chemistry is also prospected. It is hoped that this work provides beneficial help for the rational design of imidazole-based drug molecules and supramolecular medicinal agents and more effective diagnostic agents and pathological probes.
Collapse
Affiliation(s)
- Shu-Rui Li
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Yi-Min Tan
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Ling Zhang
- School of Chemical Technology, Shijiazhuang University, Shijiazhuang 050035, China
| | - Cheng-He Zhou
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| |
Collapse
|
5
|
Ceramella J, Troiano R, Iacopetta D, Mariconda A, Pellegrino M, Catalano A, Saturnino C, Aquaro S, Sinicropi MS, Longo P. Synthesis of Novel N-Heterocyclic Carbene-Ruthenium (II) Complexes, “Precious” Tools with Antibacterial, Anticancer and Antioxidant Properties. Antibiotics (Basel) 2023; 12:antibiotics12040693. [PMID: 37107055 PMCID: PMC10135378 DOI: 10.3390/antibiotics12040693] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Ruthenium N-heterocyclic carbene (Ru-NHC) complexes show interesting physico-chemical properties as catalysts and potential in medicinal chemistry, exhibiting multiple biological activities, among them anticancer, antimicrobial, antioxidant, and anti-inflammatory. Herein, we designed and synthesized a new series of Ru-NHC complexes and evaluated their biological activities as anticancer, antibacterial, and antioxidant agents. Among the newly synthesized complexes, RANHC-V and RANHC-VI are the most active against triple-negative human breast cancer cell lines MDA-MB-231. These compounds were selective in vitro inhibitors of the human topoisomerase I activity and triggered cell death by apoptosis. Furthermore, the Ru-NHC complexes’ antimicrobial activity was studied against Gram-positive and -negative bacteria, revealing that all the complexes possessed the best antibacterial activity against the Gram-positive Staphylococcus aureus, at a concentration of 25 µg/mL. Finally, the antioxidant effect was assessed by DPPH and ABTS radicals scavenging assays, resulting in a higher ability for inhibiting the ABTS•+, with respect to the well-known antioxidant Trolox. Thus, this work provides encouraging insights for further development of novel Ru-NHC complexes as potent chemotherapeutic agents endowed with multiple biological properties.
Collapse
Affiliation(s)
- Jessica Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende, Italy
| | - Rubina Troiano
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| | - Domenico Iacopetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende, Italy
| | - Annaluisa Mariconda
- Department of Science, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Michele Pellegrino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende, Italy
| | - Alessia Catalano
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70126 Bari, Italy
| | - Carmela Saturnino
- Department of Science, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Stefano Aquaro
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende, Italy
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende, Italy
| | - Pasquale Longo
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| |
Collapse
|
6
|
Catalano A, Mariconda A, Sinicropi MS, Ceramella J, Iacopetta D, Saturnino C, Longo P. Biological Activities of Ruthenium NHC Complexes: An Update. Antibiotics (Basel) 2023; 12:365. [PMID: 36830276 PMCID: PMC9952499 DOI: 10.3390/antibiotics12020365] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Ruthenium N-heterocyclic carbene (NHC) complexes have unique physico-chemical properties as catalysts and a huge potential in medicinal chemistry and pharmacology, exhibiting a variety of notable biological activities. In this review, the most recent studies on ruthenium NHC complexes are summarized, focusing specifically on antimicrobial and antiproliferative activities. Ruthenium NHC complexes are generally active against Gram-positive bacteria, such as Bacillus subtilis, Staphylococcus aureus, Micrococcus luteus, Listeria monocytogenes and are seldom active against Gram-negative bacteria, including Salmonella typhimurium, Pseudomonas aeruginosa and Escherichia coli and fungal strains of Candida albicans. The antiproliferative activity was tested against cancer cell lines of human colon, breast, cervix, epidermis, liver and rat glioblastoma cell lines. Ruthenium NHC complexes generally demonstrated cytotoxicity higher than standard anticancer drugs. Further studies are needed to explore the mechanism of action of these interesting compounds.
Collapse
Affiliation(s)
- Alessia Catalano
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70126 Bari, Italy
| | | | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Jessica Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Domenico Iacopetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Carmela Saturnino
- Department of Science, University of Basilicata, 85100 Potenza, Italy
| | - Pasquale Longo
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| |
Collapse
|
7
|
Reactivity of a nitrosyl ruthenium complex and its potential impact on the fate of DNA - An in vitro investigation. J Inorg Biochem 2023; 238:112052. [PMID: 36334365 DOI: 10.1016/j.jinorgbio.2022.112052] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/21/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
The role of metal complexes on facing DNA has been a topic of major interest. However, metallonitrosyl compounds have been poorly investigated regarding their reactivities and interaction with DNA. A nitrosyl compound, cis-[Ru(bpy)2(SO3)(NO)](PF6)(A), showed a variety of promising biological activities catching our attention. Here, we carried out a series of studies involving the interaction and damage of DNA mediated by the metal complex A and its final product after NO release, cis-[Ru(bpy)2(SO3)(H2O](B). The fate of DNA with these metal complexes was investigated upon light or chemical stimuli using electrophoresis, electronic absorption spectroscopy, circular dichroism, size-exclusion resin, mass spectrometry, electron spin resonance (ESR) and viscometry. Since many biological disorders involve the production of oxidizing species, it is important to evaluate the reactivity of these compounds under such conditions as well. Indeed, the metal complex B exhibited important reactivity with H2O2 enabling DNA degradation, with detection of an unusual oxygenated intermediate. ESR spectroscopy detected mainly the DMPO-OOH adduct, which only emerges if H2O2 and O2 are present together. This result indicated HOO• as a key radical likely involved in DNA damage as supported by agarose gel electrophoresis. Notably, the nitrosyl ruthenium complex did not show evidence of direct DNA damage. However, its aqua product should be carefully considered as potentially harmful to DNA deserving further in vivo studies to better address any genotoxicity.
Collapse
|
8
|
Synthesis, characterization and in vitro cytotoxicity of ruthenium(II) metronidazole complexes: Cell cycle arrest at G1/S transition and apoptosis induction in MCF-7 cells. J Inorg Biochem 2022; 237:112022. [PMID: 36244314 DOI: 10.1016/j.jinorgbio.2022.112022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/25/2022] [Accepted: 10/02/2022] [Indexed: 11/05/2022]
Abstract
Ruthenium compounds are known to be potential drug candidates since they offer the potential for reduced toxicity. Furthermore, the various oxidation states, different mechanisms of action and ligand substitution kinetics give them advantages over platinum-based complexes, making them suitable for use in biological applications. So, herein, novel ruthenium(II) complexes with metronidazole as ligand were obtained [RuCl(MTNZ)(dppb)(4,4'-Mebipy)]PF6 (1), [RuCl(MTNZ)(dppb)(4,4'-Methoxybipy)]PF6 (2), [RuCl(MTNZ)(dppb)(bipy)]PF6 (3) and [RuCl(MTNZ)(dppb)(phen)]PF6 (4) where, MTNZ = metronidazole, dppb = 1,4-bis(diphenylphosphino)butane, 4,4'-Mebipy = 4,4'-dimethyl-2,2'-bipyridine, 4,4'-Methoxybipy = 4,4'-dimethoxy-2,2'-bipyridine, bipy = 2,2'-bipyridine and phen = 1,10-phenanthroline. The complexes were characterized by elemental analysis, molar conductivity, infrared and UV-Vis spectroscopy, cyclic voltammetry, 31P{1H}, 1H, 13C{1H} and Dept 135 NMR and mass spectrometry. The interaction of complexes 1-4 with DNA was evaluated, and their cytotoxicity profiles were determined on four different tumor cell lines derived from human cancers (SK-MEL-147, melanoma; HepG2, hepatocarcinoma; MCF-7, estrogen-positive breast cancer; A549, non-small cell lung cancer). We demonstrated that complexes (1) and (3) are promising antitumor agents once inhibited the proliferative behavior of MCF-7 cells and induced apoptosis.
Collapse
|
9
|
Synthesis, characterization, antioxidant potential, and cytotoxicity screening of new Cu(II) complexes with 4-(arylchalcogenyl)-1H-pyrazoles ligands. J Inorg Biochem 2022; 237:112013. [PMID: 36183642 DOI: 10.1016/j.jinorgbio.2022.112013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 09/16/2022] [Accepted: 09/18/2022] [Indexed: 01/18/2023]
Abstract
Two new Cu(II) complexes based on 4-(arylchalcogenyl)-1H-pyrazoles monodentate bis(ligand) containing selenium or sulfur groups (2a and 2b) have been synthesized and characterized by IR spectroscopy, high-resolution mass spectrometry (HRMS), and by X-ray crystallography. In the effort to propose new applications for the biomedical area, we evaluated the antioxidant activity and cytotoxicity of the newly synthesized complexes. The antioxidant activity of the Cu(II) complexes (2a - 2b) were assessed through their ability to inhibit the formation of reactive species (RS) induced by sodium azide and to scavenge the synthetic radicals 2,2-diphenyl-1-picrylhydrazyl (DPPH) and 2,2-azinobis-3-ethylbenzothiazoline-6-sulfonic acid (ABTS+). Both copper complexes containing selenium (2a) and sulfur (2b) presented in vitro antioxidant activity. The (1a - 1b and 2a - 2b) compounds did not show cytotoxicity in V79 cells at low concentrations. Furthermore, the antiproliferative activity of free ligands (1a - 1b) and their complexes (2a - 2b) were tested against two human tumor cell lines: MCF-7 (breast adenocarcinoma) and HepG2 (hepatocarcinoma). Also, 2a was tested against U2OS (osteosarcoma). Our results demonstrated that 1a and 1b show little or no growth inhibition activities on human cell lines.The 2a compound exhibited good cytotoxic activity toward human tumor cell lines. However, 2a showed no selectivity, with a selectivity index of 1.12-1.40. Complex 2b was selective for the MCF-7 human tumor cell lines with IC50 of 59 ± 2 μM. This study demonstrates that the Cu(II) complexes 2a and 2b represent promising antitumoral compounds, and further studies are necessary to understand the molecular mechanisms of these effects.
Collapse
|
10
|
Mohamed A, Chilingerian JN, Bali P, Obonyo M, Debnath A. A Bioluminescence-Based Drug Screen Identifies Activities of Fexinidazole and Its Metabolites against Helicobacter pylori. Antibiotics (Basel) 2022; 11:1605. [PMID: 36421252 PMCID: PMC9686901 DOI: 10.3390/antibiotics11111605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2023] Open
Abstract
Helicobacter pylori is responsible for a wide range of gastric diseases, including gastric cancer and gastritis. With half of the world’s population infected by H. pylori and the current standard of care associated with suboptimal outcomes, a search for more effective drugs is critical. To facilitate drug screening for H. pylori, we developed a microtiter plate-based compound screening method that is faster and can screen multiple compounds. We identified activities of fexinidazole and its sulfoxide and sulfone metabolites against H. pylori. Both fexinidazole and its metabolites exhibited equipotency against SS1, 60190, and G27 strains, which were about 3–6-fold more potent than the currently used metronidazole. We also determined the minimal inhibitory concentration (MIC) of metronidazole, fexinidazole, and its metabolites against these strains by a traditional agar plate-based method. While MIC values of fexinidazole and metronidazole were similar against all the strains, both sulfoxide and sulfone showed lower MIC values than metronidazole against SS1 and 60190. Given the recent FDA approval of fexinidazole, our data on the in vitro antibacterial activities of fexinidazole and its metabolites support further evaluation of this drug with the goal of producing an alternative nitro-based antimicrobial with good safety profiles for the treatment of H. pylori infection.
Collapse
|
11
|
Miri AH, Kamankesh M, Llopis-Lorente A, Liu C, Wacker MG, Haririan I, Asadzadeh Aghdaei H, Hamblin MR, Yadegar A, Rad-Malekshahi M, Zali MR. The Potential Use of Antibiotics Against Helicobacter pylori Infection: Biopharmaceutical Implications. Front Pharmacol 2022; 13:917184. [PMID: 35833028 PMCID: PMC9271669 DOI: 10.3389/fphar.2022.917184] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Helicobacter pylori (H. pylori) is a notorious, recalcitrant and silent germ, which can cause a variety of debilitating stomach diseases, including gastric and duodenal ulcers and gastric cancer. This microbe predominantly colonizes the mucosal layer of the human stomach and survives in the inhospitable gastric microenvironment, by adapting to this hostile milieu. In this review, we first discuss H. pylori colonization and invasion. Thereafter, we provide a survey of current curative options based on polypharmacy, looking at pharmacokinetics, pharmacodynamics and pharmaceutical microbiology concepts, in the battle against H. pylori infection.
Collapse
Affiliation(s)
- Amir Hossein Miri
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mojtaba Kamankesh
- Polymer Chemistry Department, School of Science, University of Tehran, Tehran, Iran
| | - Antoni Llopis-Lorente
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Chenguang Liu
- College of Marine Life Science, Ocean University of China, Qingdao, China
| | - Matthias G. Wacker
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | - Ismaeil Haririan
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mazda Rad-Malekshahi
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Benhassine A, Chouiter MI, Kara Ali M, Kacem-Chaouche N, Merazig H, Bencharif M, Belfaitah A. New Cd(II) complex derived from (1-methylimidazol-2-yl) methanol: Synthesis, crystal structure, spectroscopic study, DFT and TD-DFT calculations, antimicrobial activity and free-radical scavenging capacity. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
13
|
Juszczak M, Kluska M, Kosińska A, Rudolf B, Woźniak K. Antioxidant Activity of Ruthenium Cyclopentadienyl Complexes Bearing Succinimidato and Phthalimidato Ligands. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27092803. [PMID: 35566156 PMCID: PMC9101797 DOI: 10.3390/molecules27092803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/22/2022] [Accepted: 04/24/2022] [Indexed: 11/22/2022]
Abstract
In these studies, we investigated the antioxidant activity of three ruthenium cyclopentadienyl complexes bearing different imidato ligands: (η5-cyclopentadienyl)Ru(CO)2-N-methoxysuccinimidato (1), (η5-cyclopentadienyl)Ru(CO)2-N-ethoxysuccinimidato (2), and (η5-cyclopentadienyl)Ru(CO)2-N-phthalimidato (3). We studied the effects of ruthenium complexes 1–3 at a low concentration of 50 µM on the viability and the cell cycle of peripheral blood mononuclear cells (PBMCs) and HL-60 leukemic cells exposed to oxidative stress induced by hydrogen peroxide (H2O2). Moreover, we examined the influence of these complexes on DNA oxidative damage, the level of reactive oxygen species (ROS), and superoxide dismutase (SOD) activity. We have observed that ruthenium complexes 1–3 increase the viability of both normal and cancer cells decreased by H2O2 and also alter the HL-60 cell cycle arrested by H2O2 in the sub-G1 phase. In addition, we have shown that ruthenium complexes reduce the levels of ROS and oxidative DNA damage in both cell types. They also restore SOD activity reduced by H2O2. Our results indicate that ruthenium complexes 1–3 bearing succinimidato and phthalimidato ligands have antioxidant activity without cytotoxic effect at low concentrations. For this reason, the ruthenium complexes studied by us should be considered interesting molecules with clinical potential that require further detailed research.
Collapse
Affiliation(s)
- Michał Juszczak
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; (M.J.); (M.K.)
| | - Magdalena Kluska
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; (M.J.); (M.K.)
| | - Aneta Kosińska
- Department of Organic Chemistry, Faculty of Chemistry, University of Lodz, 91-403 Lodz, Poland; (A.K.); (B.R.)
| | - Bogna Rudolf
- Department of Organic Chemistry, Faculty of Chemistry, University of Lodz, 91-403 Lodz, Poland; (A.K.); (B.R.)
| | - Katarzyna Woźniak
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; (M.J.); (M.K.)
- Correspondence:
| |
Collapse
|
14
|
The nitric oxide pathway is involved in the anti-inflammatory effect of the rutheniumcomplex [Ru(bpy)2(2-MIM)(NO)](PF6)3. Eur J Pharmacol 2022; 921:174869. [PMID: 35247379 DOI: 10.1016/j.ejphar.2022.174869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 02/28/2022] [Indexed: 12/15/2022]
Abstract
Metal coordination complexes are chemotherapeutic and anti-inflammatory agents. The ruthenium complex FOR811A ([Ru(bpy)2(2-MIM)Cl](PF6)3) FOR811A was evaluated in mice models of acute inflammation and behavioral tests. Animals received FOR811A (3, 10 or 30 mg/Kg; i.p.), indomethacin (20 mg/Kg; i.p.), L-NAME (20 mg/Kg; i.v.) aminoguanidine (50 mg/Kg; i.p.) or dexamethasone (0.5 mg/Kg; s.c.) 30 min before inflammatory stimulation. Paw edema was induced by carrageenan (400 μg/paw), TNF-α or L-arginine (15 nmol/paw) (5 ng/paw) and evaluated by hydroplestimometry 4 h later. Peritonitis was induced by carrageenan (500 μg; i.p.) and evaluated 4 h later for hypernociception and quantification of total/differential leukocytes, total protein reduced glutathione (GSH) and myeloperoxidase (MPO). FOR811A inhibited the paw edema induced by carrageenan at 3 (64%; p < 0.0001), 10 (73%; p < 0.0001) and 30 mg/kg (66%; p < 0.0001), and at 10 mg/kg that induced with L-arginine by 75% or TNF-α by 55% (p = 0.0012). Paw tissues histological analysis showed reduction in mast cells (46%; p = 0.0027), leukocyte infiltrate (66%; p < 0.0001), edema and hemorrhagic areas. Immunohistochemical evaluation revealed inhibition of iNOS (62%; p < 0.0001) and TNF-α (35%; p < 0.0001). In the peritonitis model FOR811A increased (2.8X; p < 0.0001) hypernociceptive threshold, reduced total leukocytes (29%; p < 0.0001), neutrophils (47%; p = 0.0003) and total proteins (36%; p = 0.0082). FOR811A also inhibited MPO (47%; p = 0.0296) and increased GSH (1.8X; p < 0.0001). In the behavioral tests, FOR811A reduced (30.6%) the number of crossings in the open field, and increased (16%) the number of falls in the Rota rod. Concluding, FOR811A presents anti-inflammatory and antioxidant effects, via nitric oxide pathway.
Collapse
|
15
|
Nikitin E, Shpakovsky D, Tyurin VY, Kazak A, Gracheva YA, Vasilichin V, Pavlyukov M, Mironova E, Gontcharenko V, Lyssenko K, Antonets A, Dubova L, Shevtsov P, Shevtsova E, Shamraeva M, Shtil A, Milaeva E. Novel organotin complexes with phenol and imidazole moieties for optimized antitumor properties. J Organomet Chem 2022. [DOI: 10.1016/j.jorganchem.2021.122212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
16
|
da Silva Filho PM, Andrade AL, Lopes JBAC, Pinheiro ADA, de Vasconcelos MA, Fonseca SGDC, Lopes LGDF, Sousa EHS, Teixeira EH, Longhinotti E. The biofilm inhibition activity of a NO donor nanosilica with enhanced antibiotics action. Int J Pharm 2021; 610:121220. [PMID: 34687814 DOI: 10.1016/j.ijpharm.2021.121220] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/30/2021] [Accepted: 10/16/2021] [Indexed: 02/07/2023]
Abstract
Nitric oxide (NO) has emerged as a promising antibacterial agent, where NO donor compounds have been explored. Here, we investigated the role of a silica nanoparticle containing nitroprusside (MPSi-NP) as a NO donor agent against methicillin-sensitive (ATCC 25,923 and ATCC 12228) and methicillin-resistant (ATCC 700,698 and ATCC 35984) Staphylococcus strains. Biofilm inhibition was studied along with antibiotic activity in combination with standard antibiotics (ampicillin and tetracycline). MPSi-NP exhibited thermal release of 63% of NO within 24 h, while free nitroprusside released only 18% during a dialysis assay, indicating an assisted release of NO mediated by the nanoparticles. This nanomaterial showed only a moderate activity in blocking biofilm production, but exhibited a significant decrease in the number of viable bacterial cells (over 600-fold for Staphylococcus aureus ATCC 700,698 and Staphylococcus epidermidis ATCC 35984). Remarkably, even using MPSi-NP at concentrations below any antibacterial action, its combination with ampicillin promoted a significant decrease in MIC for resistant strains of S. aureus ATCC 700,698 (2-fold) and S. epidermidis ATCC 35,984 (4-fold). A carbopol-based gel formulation with MPSi-NP (0.5% w/w) was prepared and showed a zone of inhibition of 7.7 ± 0.6 mm for S. epidermidis ATCC 35984. Topical use of MPSi-NP in combination with antibiotics might be a manageable strategy to prevent and eventually treat complicated resistant bacterial infections.
Collapse
Affiliation(s)
- Pedro Martins da Silva Filho
- Departamento de Química Analítica e Físico-Química, Universidade Federal do Ceará, Fortaleza, CE 60440-900, Brazil; Laboratório de Bioinorgânica, Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, PO Box 12200, Campus do Pici s/n, Fortaleza, CE 60440-900, Brazil
| | - Alexandre Lopes Andrade
- Laboratório Integrado de Biomoléculas, Departamento de Patologia e Medicina Legal, Universidade Federal do Ceará, CEP, Fortaleza, CE 60430-270, Brazil
| | - Jessica Barros Arrais Cruz Lopes
- Laboratório Integrado de Biomoléculas, Departamento de Patologia e Medicina Legal, Universidade Federal do Ceará, CEP, Fortaleza, CE 60430-270, Brazil
| | - Aryane de Azevedo Pinheiro
- Laboratório Integrado de Biomoléculas, Departamento de Patologia e Medicina Legal, Universidade Federal do Ceará, CEP, Fortaleza, CE 60430-270, Brazil
| | - Mayron Alves de Vasconcelos
- Laboratório Integrado de Biomoléculas, Departamento de Patologia e Medicina Legal, Universidade Federal do Ceará, CEP, Fortaleza, CE 60430-270, Brazil; Departamento de Ciências Biológicas, Faculdade de Ciências Exatas e Naturais, Universidade do Estado do Rio Grande do Norte, Mossoró, RN 59610-090, Brazil; Universidade do Estado de Minas Gerais, Unidade de Divinópolis, Divinópolis, MG 35501-170, Brazil
| | | | - Luiz Gonzaga de França Lopes
- Laboratório de Bioinorgânica, Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, PO Box 12200, Campus do Pici s/n, Fortaleza, CE 60440-900, Brazil
| | - Eduardo Henrique Silva Sousa
- Laboratório de Bioinorgânica, Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, PO Box 12200, Campus do Pici s/n, Fortaleza, CE 60440-900, Brazil.
| | - Edson Holanda Teixeira
- Laboratório Integrado de Biomoléculas, Departamento de Patologia e Medicina Legal, Universidade Federal do Ceará, CEP, Fortaleza, CE 60430-270, Brazil.
| | - Elisane Longhinotti
- Departamento de Química Analítica e Físico-Química, Universidade Federal do Ceará, Fortaleza, CE 60440-900, Brazil.
| |
Collapse
|
17
|
de Oliveira Neto J, Marinho MM, Silveira JADM, Rocha DG, Lima NCB, Gouveia Júnior FS, Lopes LGDF, de Sousa EHS, Martins AMC, Marinho AD, Jorge RJB, Monteiro HSA. Synthesis and potential vasorelaxant effect of a novel ruthenium-based nitro complex. J Inorg Biochem 2021; 228:111666. [PMID: 34923187 DOI: 10.1016/j.jinorgbio.2021.111666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 11/10/2021] [Accepted: 11/10/2021] [Indexed: 12/11/2022]
Abstract
This study aimed to investigate the synthesis and potential vasodilator effect of a novel ruthenium complex, cis-[Ru(bpy)2(2-MIM)(NO2)]PF6 (bpy = 2,2'-bipyridine and 2-MIM = 2-methylimidazole) (FOR711A), containing an imidazole derivative via an in silico molecular docking model using β1 H-NOX (Heme-nitric oxide/oxygen binding) domain proteins of reduced and oxidized soluble guanylate cyclase (sGC). In addition, pharmacokinetic properties in the human organism were predicted through computational simulations and the potential for acute irritation of FOR711A was also investigated in vitro using the hen's egg chorioallantoic membrane (HET-CAM). FOR711A interacted with sites of the β1 H-NOX domain of reduced and oxidized sGC, demonstrating shorter bond distances to several residues and negative values of total energy. The predictive study revealed molar refractivity (RM): 127.65; Log Po/w = 1.29; topological polar surface area (TPSA): 86.26 Å2; molar mass (MM) = 541.55 g/mol; low solubility, high unsaturation index, high gastrointestinal absorption; toxicity class 4; failure to cross the blood-brain barrier and to react with cytochrome P450 (CYP) enzymes CYP1A2, CYP2C19, CYP2C9, CYP2D6 and CYP3A4. After the HET-CAM assay, the FOR711A complex was classified as non-irritant (N.I.) and its vasodilator effect was confirmed through greater evidence of blood vessels after the administration and ending of the observation period of 5 min. These results suggest that FOR711A presented a potential stimulator/activator effect of sGC via NO/sGC/cGMP. However, results indicate it needs a vehicle for oral administration.
Collapse
Affiliation(s)
- Joselito de Oliveira Neto
- Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Coronel Nunes de Melo St., 1127, 60.430-275 Fortaleza, CE, Brazil; Drug Research and Development Center (NPDM), Federal University of Ceará, Coronel Nunes de Melo St., 1000, 60.430-275 Fortaleza, CE, Brazil
| | - Márcia Machado Marinho
- State University of Ceará, Iguatu Faculty of Education, Science and Letters, Iguatu, CE, Brazil
| | - João Alison de Moraes Silveira
- Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Coronel Nunes de Melo St., 1127, 60.430-275 Fortaleza, CE, Brazil; Drug Research and Development Center (NPDM), Federal University of Ceará, Coronel Nunes de Melo St., 1000, 60.430-275 Fortaleza, CE, Brazil.
| | - Danilo Galvão Rocha
- Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Coronel Nunes de Melo St., 1127, 60.430-275 Fortaleza, CE, Brazil; Drug Research and Development Center (NPDM), Federal University of Ceará, Coronel Nunes de Melo St., 1000, 60.430-275 Fortaleza, CE, Brazil
| | - Natália Cavalcante Barbosa Lima
- Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Coronel Nunes de Melo St., 1127, 60.430-275 Fortaleza, CE, Brazil; Drug Research and Development Center (NPDM), Federal University of Ceará, Coronel Nunes de Melo St., 1000, 60.430-275 Fortaleza, CE, Brazil
| | | | | | | | - Alice Maria Costa Martins
- Department of Clinical and Toxicological Analysis, School of Pharmacy, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Aline Diogo Marinho
- Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Coronel Nunes de Melo St., 1127, 60.430-275 Fortaleza, CE, Brazil; Drug Research and Development Center (NPDM), Federal University of Ceará, Coronel Nunes de Melo St., 1000, 60.430-275 Fortaleza, CE, Brazil
| | - Roberta Jeane Bezerra Jorge
- Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Coronel Nunes de Melo St., 1127, 60.430-275 Fortaleza, CE, Brazil; Drug Research and Development Center (NPDM), Federal University of Ceará, Coronel Nunes de Melo St., 1000, 60.430-275 Fortaleza, CE, Brazil
| | - Helena Serra Azul Monteiro
- Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Coronel Nunes de Melo St., 1127, 60.430-275 Fortaleza, CE, Brazil; Drug Research and Development Center (NPDM), Federal University of Ceará, Coronel Nunes de Melo St., 1000, 60.430-275 Fortaleza, CE, Brazil
| |
Collapse
|
18
|
Giacomazzo GE, Conti L, Guerri A, Pagliai M, Fagorzi C, Sfragano PS, Palchetti I, Pietraperzia G, Mengoni A, Valtancoli B, Giorgi C. Nitroimidazole-Based Ruthenium(II) Complexes: Playing with Structural Parameters to Design Photostable and Light-Responsive Antibacterial Agents. Inorg Chem 2021; 61:6689-6694. [PMID: 34793162 DOI: 10.1021/acs.inorgchem.1c03032] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
5-Nitroimidazole (5NIMH), chosen as a molecular model of nitroimidazole derivatives, which represent a broad-spectrum class of antimicrobials, was incorporated into the ruthenium complexes [Ru(tpy)(phen)(5NIM)]PF6 (1) and [Ru(tpy)(dmp)(5NIM)]PF6 (2) (tpy = terpyridine, phen = phenanthroline, dmp = 2,9-dimethyl-1,10-phenanthroline). Besides the uncommon metal coordination of 5-nitroimidazole in its imidazolate form (5NIM), the different architectures of the spectator ligands (phen and dmp) were exploited to tune the "mode of action" of the resulting complexes, passing from a photostable compound where the redox properties of 5NIMH are preserved (1) to one suitable for the nitroimidazole phototriggered release (2) and whose antibacterial activity against B. subtilis, chosen as cellular model, is effectively improved upon light exposure. This study may provide a fundamental knowledge on the use of Ru(II)-polypyridyl complexes to incorporate and/or photorelease biologically relevant nitroimidazole derivatives in the design of a novel class of antimicrobials.
Collapse
Affiliation(s)
- Gina Elena Giacomazzo
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Florence, Italy
| | - Luca Conti
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Florence, Italy
| | - Annalisa Guerri
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Florence, Italy
| | - Marco Pagliai
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Florence, Italy
| | - Camilla Fagorzi
- Department of Biology, University of Florence, Via Madonna del Piano 6, 50019 Sesto Fiorentino, Florence, Italy
| | - Patrick Severin Sfragano
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Florence, Italy
| | - Ilaria Palchetti
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Florence, Italy
| | - Giangaetano Pietraperzia
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Florence, Italy.,European Laboratory for Non-Linear Spectroscopy (LENS), Via Nello Carrara 1, 50019 Sesto Fiorentino, Florence, Italy
| | - Alessio Mengoni
- Department of Biology, University of Florence, Via Madonna del Piano 6, 50019 Sesto Fiorentino, Florence, Italy
| | - Barbara Valtancoli
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Florence, Italy
| | - Claudia Giorgi
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Florence, Italy
| |
Collapse
|
19
|
da Silva GM, da Silva MC, Nascimento DVG, Lima Silva EM, Gouvêa FFF, de França Lopes LG, Araújo AV, Ferraz Pereira KN, de Queiroz TM. Nitric Oxide as a Central Molecule in Hypertension: Focus on the Vasorelaxant Activity of New Nitric Oxide Donors. BIOLOGY 2021; 10:1041. [PMID: 34681140 PMCID: PMC8533285 DOI: 10.3390/biology10101041] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/08/2021] [Accepted: 10/09/2021] [Indexed: 12/15/2022]
Abstract
Cardiovascular diseases include all types of disorders related to the heart or blood vessels. High blood pressure is an important risk factor for cardiac complications and pathological disorders. An increase in circulating angiotensin-II is a potent stimulus for the expression of reactive oxygen species and pro-inflammatory cytokines that activate oxidative stress, perpetuating a deleterious effect in hypertension. Studies demonstrate the capacity of NO to prevent platelet or leukocyte activation and adhesion and inhibition of proliferation, as well as to modulate inflammatory or anti-inflammatory reactions and migration of vascular smooth muscle cells. However, in conditions of low availability of NO, such as during hypertension, these processes are impaired. Currently, there is great interest in the development of compounds capable of releasing NO in a modulated and stable way. Accordingly, compounds containing metal ions coupled to NO are being investigated and are widely recognized as having great relevance in the treatment of different diseases. Therefore, the exogenous administration of NO is an attractive and pharmacological alternative in the study and treatment of hypertension. The present review summarizes the role of nitric oxide in hypertension, focusing on the role of new NO donors, particularly the metal-based drugs and their protagonist activity in vascular function.
Collapse
Affiliation(s)
- Gabriela Maria da Silva
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Mirelly Cunha da Silva
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Déborah Victória Gomes Nascimento
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Ellen Mayara Lima Silva
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Fabíola Furtado Fialho Gouvêa
- School of Technical Health, Health Sciences Center, Federal University of Paraíba, João Pessoa 58.051-900, PB, Brazil;
| | - Luiz Gonzaga de França Lopes
- Laboratory of Bioinorganic Chemistry, Department of Organic and Inorganic Chemistry, Federal University of Ceará, Fortaleza 60.020-181, CE, Brazil;
| | - Alice Valença Araújo
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Kelli Nogueira Ferraz Pereira
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Thyago Moreira de Queiroz
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| |
Collapse
|