1
|
Sa M, da Silva M, Ball B, Geddes-McAlister J. Revealing the dynamics of fungal disease with proteomics. Mol Omics 2025; 21:173-184. [PMID: 40066820 DOI: 10.1039/d4mo00222a] [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: 05/16/2025]
Abstract
The occurrence and distribution of new and re-emerging fungal pathogens, along with rates of antifungal resistance, are rising across the globe, and correspondingly, so are our awareness and call for action to address this public health concern. To effectively detect, monitor, and treat fungal infections, biological insights into the mechanisms that regulate pathogenesis, influence survival, and promote resistance are urgently needed. Mass spectrometry-based proteomics is a high-resolution technique that enables the identification and quantification of proteins across diverse biological systems to better understand the biology driving phenotypes. In this review, we highlight dynamic and innovative applications of proteomics to characterize three critical fungal pathogens (i.e., Candida spp., Cryptococcus spp., and Aspergillus spp.) causing disease in humans. We present strategies to investigate the host-pathogen interface, virulence factor production, and protein-level drivers of antifungal resistance. Through these studies, new opportunities for biomarker development, drug target discovery, and immune system remodeling are discussed, supporting the use of proteomics to combat a plethora of fungal diseases threatening global health.
Collapse
Affiliation(s)
- Mariana Sa
- Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, N1G 2W1, Canada.
| | - Mayara da Silva
- Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, N1G 2W1, Canada.
| | - Brianna Ball
- Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, N1G 2W1, Canada.
| | | |
Collapse
|
2
|
Lucas Dos Santos AT, Audilene de Freitas M, Queiroz da Silva ML, Silva FDS, Guilhermino Dos Santos A, Silva Menêses AV, Farias NS, Pereira Carneiro JN, Alencar Fonseca VJ, Silva Dos Santos H, Rogenio da Silva Mendes F, Silva J, Marinho MM, Marinho ES, Melo Coutinho HD, Bezerra Morais-Braga MF. In silico activity and effect of synthetic chalcones on Candida albicans and Candida tropicalis biofilms. Biochimie 2025; 234:29-39. [PMID: 40113030 DOI: 10.1016/j.biochi.2025.03.004] [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: 09/28/2024] [Revised: 03/12/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Abstract
Biofilm formation is considered one of the most important virulence factors for Candida species, which presents an extracellular matrix of polymeric substances that limits the passage of antifungals, leading to fungal resistance. Therefore, the present study investigated the biofilm eradication effect of synthetic chalcones against Candida albicans and Candida tropicalis. Molecular docking studies were conducted to verify the mechanism of action of chalcones on Candida species proteins. The biofilm eradication effect was determined using crystal violet methodology to quantify biomass and Thiazolyl blue tetrazolium bromide (MTT) to verify the influence on metabolic activity. A molecular docking study was also carried out with Candida proteins using the Protein Data Bank repository (https://www.rcsb.org/) and Autodocktools™ software. The results showed that (1E,4E)-1,5-diphenylpenta-1,4-dien-3-one (DB-Acetone), (1E,3E,6E,8E)-1,9-diphenylnona-1,3,6,8-tetraen-5-one (DB-CNM), and (1E,4E)-1,5-bis(4-methoxyphenyl)penta-1,4-dien-3-one (DB-Anisal) were able to eradicate the biomass of C. albicans CA INCQS 40006 (ATCC 10231), while fluconazole only reduced the biomass at the three tested concentrations (IC50, IC50 × 10, and IC50 × 20) against C. tropicalis CT INCQS 40042 (ATCC 13803). Both chalcones and fluconazole successfully reduced metabolic activity across all tested strains. The molecular docking study concluded that DB-Acetone, DB-Anisal, and DB-CNM exhibited significant affinity energy values toward the binding sites of C. albicans and C. tropicalis. It is concluded that the synthetic chalcones showed promising results in inhibiting Candida spp. biofilm, demonstrating efficacy in reducing biomass as well as metabolic activity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Jacilene Silva
- Regional University of Cariri - URCA, Pimenta, Crato, Ceará, Brazil
| | | | | | | | | |
Collapse
|
3
|
Strieder Philippsen G, Augusto Vicente Seixas F. Computational approach based on freely accessible tools for antimicrobial drug design. Bioorg Med Chem Lett 2025; 115:130010. [PMID: 39486485 DOI: 10.1016/j.bmcl.2024.130010] [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: 08/05/2024] [Revised: 10/15/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024]
Abstract
Antimicrobial drug development is crucial for public health, especially with the emergence of pandemics and drug resistance that prompts the search for new therapeutic resources. In this context, in silico assays consist of a valuable approach in the rational drug design because they enable a faster and more cost-effective identification of drug candidates compared to in vitro screening. However, once a potential drug is identified, in vitro and in vivo assays are essential to verify the expected activity of the compound and advance it through the subsequent stages of drug development. This work aims to outline an in silico protocol that utilizes only freely available computational tools for identifying new potential antimicrobial agents, which is also suitable in the broad spectrum of drug design. Additionally, this paper reviews relevant computational methods in this context and provides a summary of information concerning the protein-ligand interaction.
Collapse
|
4
|
Rana D, Beladiya J, Sheth D, Kumar H, Jindal AB, Shah G, Sharma A, Dash SK, Shrivastava SK, Benival D. Investigating a novel therapeutic composition for dry eye syndrome management: In vitro and in vivo studies. Int J Pharm 2024; 666:124783. [PMID: 39353497 DOI: 10.1016/j.ijpharm.2024.124783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/27/2024] [Accepted: 09/28/2024] [Indexed: 10/04/2024]
Abstract
Dry eye syndrome (DES) presents a significant challenge in ophthalmic care, necessitating innovative approaches for effective management. This research article introduces a multifaceted strategy to address DES through the development of ocular inserts utilizing advanced technologies such as hot-melt extrusion (HME) and the CaliCut post-extrusion system. The formulation includes key ingredients targeting different layers of the tear film and associated inflammation, including hydroxypropyl cellulose (HPC), polyethylene glycol (PEG), castor oil, and dexamethasone. The study incorporates a Design of Experiments (DoE) approach, integrating HME and the precise stretching and cutting technique of CaliCut for manufacturing consistency and dimensional control of the inserts. The developed insert(s) have been systematically characterized for their physicochemical properties, release profile, and in vivo efficacy. In silico molecular docking studies have also been conducted to assess the binding affinities of formulation components with ocular mucin, elucidating their binding affinities. Preliminary results demonstrate promising potential for the developed insert in managing DES, offering preservative-free treatment, sustained drug delivery, and improved patient compliance. This study highlights the integration of advanced technologies and formulation strategies in ocular drug delivery for effective DES management.
Collapse
Affiliation(s)
- Dhwani Rana
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), India
| | - Jayesh Beladiya
- Department of Pharmacology, L.M. College of Pharmacy, Ahmedabad, India
| | - Devang Sheth
- Department of Pharmacology, L.M. College of Pharmacy, Ahmedabad, India
| | - Hansal Kumar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology-Banaras Hindu University, Varanasi 221005, India
| | - Anil B Jindal
- Department of Pharmacy, Birla Institute of Technology and Science Pilani (Bits Pilani), Pilani Campus, Rajasthan 333031, India
| | - Gunjan Shah
- Gunjan Eye Hospital, Ahmedabad 380063, India
| | - Amit Sharma
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), India
| | - Sanat Kumar Dash
- Department of Pharmacy, Birla Institute of Technology and Science Pilani (Bits Pilani), Pilani Campus, Rajasthan 333031, India
| | - Sushant Kumar Shrivastava
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology-Banaras Hindu University, Varanasi 221005, India
| | - Derajram Benival
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), India.
| |
Collapse
|
5
|
Souza LBFC, de Oliveira Bento A, Lourenço EMG, Ferreira MRA, Oliveira WN, Soares LAL, G. Barbosa E, Rocha HAO, Chaves GM. Mechanism of action and synergistic effect of Eugenia uniflora extract in Candida spp. PLoS One 2024; 19:e0303878. [PMID: 39137202 PMCID: PMC11321568 DOI: 10.1371/journal.pone.0303878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 05/01/2024] [Indexed: 08/15/2024] Open
Abstract
The limited arsenal of antifungal drugs have prompted the search for novel molecules with biological activity. This study aimed to characterize the antifungal mechanism of action of Eugenia uniflora extract and its synergistic activity with commercially available antifungal drugs on the following Candida species: C. albicans, C. tropicalis, C. glabrata, C. parapsilosis and C. dubliniensis. In silico analysis was performed to predict antifungal activity of the major compounds present in the extract. Minimal inhibitory concentrations (MICs) were determined in the presence of exogenous ergosterol and sorbitol. Yeast cells were grown in the presence of stressors. The loss of membrane integrity was assessed using propidium iodide staining (fluorescence emission). Synergism between the extract and antifungal compounds (in addition to time kill-curves) was determined. Molecular docking revealed possible interactions between myricitrin and acid gallic and enzymes involved in ergosterol and cell wall biosynthesis. Candida cells grown in the presence of the extract with addition of exogenous ergosterol and sorbitol showed 2 to 8-fold increased MICs. Strains treated with the extract revealed greater loss of membrane integrity when compared to their Fluconazole counterparts, but this effect was less pronounced than the membrane damage caused by Amphotericin B. The extract also made the strains more susceptible to Congo red and Calcofluor white. A synergistic action of the extract with Fluconazole and Micafungin was observed. The E. uniflora extract may be a viable option for the treatment of Candida infections.
Collapse
Affiliation(s)
- Luanda B. F. C. Souza
- Medical and Molecular Mycology Laboratory, Department of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Aurélio de Oliveira Bento
- Medical and Molecular Mycology Laboratory, Department of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Estela M. G. Lourenço
- Department of Pharmacy, Federal University of Rio Grande do Norte, Natal, Brazil
- Laboratory of Synthesis and Transformation of Organic Molecules, LP4, Institute of Chemistry, Federal University of Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Magda R. A. Ferreira
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, Brazil
| | - Wogenes N. Oliveira
- Department of Pharmacy, Federal University of Rio Grande do Norte, Natal, Brazil
| | | | - Euzébio G. Barbosa
- Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, Brazil
| | - Hugo A. O. Rocha
- Department of Biochemistry, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Guilherme Maranhão Chaves
- Medical and Molecular Mycology Laboratory, Department of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, Natal, Brazil
| |
Collapse
|
6
|
Kim M, Nowakowska A, Kim J, Kim YB. Anti-Influenza A Potential of Tagetes erecta Linn. Extract Based on Bioinformatics Analysis and In Vitro Assays. Int J Mol Sci 2024; 25:7065. [PMID: 39000173 PMCID: PMC11241564 DOI: 10.3390/ijms25137065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/14/2024] [Accepted: 06/19/2024] [Indexed: 07/16/2024] Open
Abstract
Tagetes erecta Linn. (TE) is traditionally used to treat cardiovascular, renal, and gastrointestinal diseases. In this study, we investigated the active compounds and targets of TE extract that may exert antiviral effects against influenza A. Active compounds and targets of TE extract were identified using the Traditional Chinese Medicine Systems Pharmacology database (TCSMP). The influenza A-related gene set was screened using GeneCards and the Kyoto Encyclopedia of Genes and Genomes (KEGG). A protein-protein interaction (PPI) network was built to establish the hub targets. Pathway and target studies were conducted using Gene Expression Omnibus (GEO). The interactions between active compounds and potential targets were assessed by molecular docking. An in vitro study was performed using antiviral and plaque reduction assays. From the compound and target search, we identified 6 active compounds and 95 potential targets. We retrieved 887 influenza-associated target genes and determined 14 intersecting core targets between TE and influenza. After constructing a compound-target network, we discovered lutein and beta-carotene to be the key compounds. Next, PPI network analysis identified the top three hub genes associated with influenza (IL-6, HIF1A, and IL-1β). Similarly, GEO analysis revealed IL-6, TGFB1, and CXCL8 to be the top three target genes. In our docking study, we identified that lutein and IL-6 had the strongest bindings. Our in vitro experimental results revealed that the TE extract exhibited therapeutic rather than prophylactic effects on influenza disease. We identified lutein as a main active compound in TE extract, and IL-6 as an important target associated with influenza, by using data mining and bioinformatics. Our in vitro findings indicated that TE extract exerted protective properties against the influenza A virus. We speculated that lutein, as a key active component in TE extract, is largely responsible for its antiviral effects. Therefore, we suggest TE extract as an alternative in the treatment of influenza.
Collapse
Affiliation(s)
| | | | | | - Young Bong Kim
- Department of Biomedical Science and Engineering, Konkuk University, Seoul 05029, Republic of Korea; (M.K.); (A.N.); (J.K.)
| |
Collapse
|
7
|
Araújo GR, da Costa PCQG, Nogueira PL, Alves DDN, Ferreira AR, da Silva PR, de Andrade JC, de Sousa NF, Loureiro PBA, Sobral MV, Sousa DP, Scotti MT, de Castro RD, Scotti L. In Silico and In Vitro Evaluation of the Antifungal Activity of a New Chromone Derivative against Candida spp. BIOTECH 2024; 13:16. [PMID: 38921048 PMCID: PMC11201913 DOI: 10.3390/biotech13020016] [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: 04/10/2024] [Revised: 05/10/2024] [Accepted: 05/20/2024] [Indexed: 06/27/2024] Open
Abstract
Candida species are frequently implicated in the development of both superficial and invasive fungal infections, which can impact vital organs. In the quest for novel strategies to combat fungal infections, there has been growing interest in exploring synthetic and semi-synthetic products, particularly chromone derivatives, renowned for their antimicrobial properties. In the analysis of the antifungal activity of the compound (E)-benzylidene-chroman-4-one against Candida, in silico and laboratory tests were performed to predict possible mechanisms of action pathways, and in vitro tests were performed to determine antifungal activity (MIC and MFC), to verify potential modes of action on the fungal cell membrane and wall, and to assess cytotoxicity in human keratinocytes. The tested compound exhibited predicted affinity for all fungal targets, with the highest predicted affinity observed for thymidylate synthase (-102.589 kJ/mol). MIC and CFM values ranged from 264.52 μM (62.5 μg/mL) to 4232.44 μM (1000 μg/mL). The antifungal effect likely occurs due to the action of the compound on the plasma membrane. Therefore, (E)-benzylidene-chroman-4-one showed fungicidal-like activity against Candida spp., possibly targeting the plasma membrane.
Collapse
Affiliation(s)
- Gleycyelly Rodrigues Araújo
- Department of Clinical and Social Dentistry, Federal University of Paraíba, Campus I, João Pessoa 58051-900, PB, Brazil;
| | - Palloma Christine Queiroga Gomes da Costa
- Postgraduate Program in Dentistry, Department of Clinic and Social Dentistry, Center of Health Sciences, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (P.C.Q.G.d.C.); (P.L.N.); (D.d.N.A.); (P.R.d.S.)
| | - Paula Lima Nogueira
- Postgraduate Program in Dentistry, Department of Clinic and Social Dentistry, Center of Health Sciences, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (P.C.Q.G.d.C.); (P.L.N.); (D.d.N.A.); (P.R.d.S.)
| | - Danielle da Nóbrega Alves
- Postgraduate Program in Dentistry, Department of Clinic and Social Dentistry, Center of Health Sciences, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (P.C.Q.G.d.C.); (P.L.N.); (D.d.N.A.); (P.R.d.S.)
| | - Alana Rodrigues Ferreira
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (A.R.F.); (J.C.d.A.); (N.F.d.S.); (P.B.A.L.); (M.V.S.); (D.P.S.); (M.T.S.); (R.D.d.C.)
- Health Sciences Center, Federal University of Paraíba, Campus I, João Pessoa 58051-900, PB, Brazil
| | - Pablo R. da Silva
- Postgraduate Program in Dentistry, Department of Clinic and Social Dentistry, Center of Health Sciences, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (P.C.Q.G.d.C.); (P.L.N.); (D.d.N.A.); (P.R.d.S.)
| | - Jéssica Cabral de Andrade
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (A.R.F.); (J.C.d.A.); (N.F.d.S.); (P.B.A.L.); (M.V.S.); (D.P.S.); (M.T.S.); (R.D.d.C.)
- Health Sciences Center, Federal University of Paraíba, Campus I, João Pessoa 58051-900, PB, Brazil
| | - Natália F. de Sousa
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (A.R.F.); (J.C.d.A.); (N.F.d.S.); (P.B.A.L.); (M.V.S.); (D.P.S.); (M.T.S.); (R.D.d.C.)
- Health Sciences Center, Federal University of Paraíba, Campus I, João Pessoa 58051-900, PB, Brazil
| | - Paulo Bruno Araujo Loureiro
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (A.R.F.); (J.C.d.A.); (N.F.d.S.); (P.B.A.L.); (M.V.S.); (D.P.S.); (M.T.S.); (R.D.d.C.)
- Health Sciences Center, Federal University of Paraíba, Campus I, João Pessoa 58051-900, PB, Brazil
| | - Marianna Vieira Sobral
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (A.R.F.); (J.C.d.A.); (N.F.d.S.); (P.B.A.L.); (M.V.S.); (D.P.S.); (M.T.S.); (R.D.d.C.)
- Health Sciences Center, Federal University of Paraíba, Campus I, João Pessoa 58051-900, PB, Brazil
| | - Damião P. Sousa
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (A.R.F.); (J.C.d.A.); (N.F.d.S.); (P.B.A.L.); (M.V.S.); (D.P.S.); (M.T.S.); (R.D.d.C.)
- Health Sciences Center, Federal University of Paraíba, Campus I, João Pessoa 58051-900, PB, Brazil
| | - Marcus Tullius Scotti
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (A.R.F.); (J.C.d.A.); (N.F.d.S.); (P.B.A.L.); (M.V.S.); (D.P.S.); (M.T.S.); (R.D.d.C.)
- Health Sciences Center, Federal University of Paraíba, Campus I, João Pessoa 58051-900, PB, Brazil
| | - Ricardo Dias de Castro
- Postgraduate Program in Dentistry, Department of Clinic and Social Dentistry, Center of Health Sciences, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (P.C.Q.G.d.C.); (P.L.N.); (D.d.N.A.); (P.R.d.S.)
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (A.R.F.); (J.C.d.A.); (N.F.d.S.); (P.B.A.L.); (M.V.S.); (D.P.S.); (M.T.S.); (R.D.d.C.)
- Health Sciences Center, Federal University of Paraíba, Campus I, João Pessoa 58051-900, PB, Brazil
| | - Luciana Scotti
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (A.R.F.); (J.C.d.A.); (N.F.d.S.); (P.B.A.L.); (M.V.S.); (D.P.S.); (M.T.S.); (R.D.d.C.)
- Health Sciences Center, Federal University of Paraíba, Campus I, João Pessoa 58051-900, PB, Brazil
- Institute of Drugs and Medicines Research, Federal University of Paraíba, Via Ipê Amarelo, S/N, João Pessoa 58051-900, PB, Brazil
| |
Collapse
|
8
|
Manzano JAH, Brogi S, Calderone V, Macabeo APG, Austriaco N. Globospiramine Exhibits Inhibitory and Fungicidal Effects against Candida albicans via Apoptotic Mechanisms. Biomolecules 2024; 14:610. [PMID: 38927014 PMCID: PMC11201426 DOI: 10.3390/biom14060610] [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: 04/15/2024] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Candidiasis is considered an emerging public health concern because of the occurrence of drug-resistant Candida strains and the lack of an available structurally diverse antifungal drug armamentarium. The indole alkaloid globospiramine from the anticandidal Philippine medicinal plant Voacanga globosa exhibits a variety of biological activities; however, its antifungal properties remain to be explored. In this study, we report the in vitro anticandidal activities of globospiramine against two clinically relevant Candida species (C. albicans and C. tropicalis) and the exploration of its possible target proteins using in silico methods. Thus, the colony-forming unit (CFU) viability assay revealed time- and concentration-dependent anticandidal effects of the alkaloid along with a decrease in the number of viable CFUs by almost 50% at 60 min after treatment. The results of the MIC and MFC assays indicated inhibitory and fungicidal effects of globospiramine against C. albicans (MIC = 8 µg/mL; MFC = 8 µg/mL) and potential fungistatic effects against C. tropicalis at lower concentrations (MIC = 4 µg/mL; MFC > 64 µg/mL). The FAM-FLICA poly-caspase assay showed metacaspase activation in C. albicans cells at concentrations of 16 and 8 µg/mL, which agreed well with the MIC and MFC values. Molecular docking and molecular dynamics simulation experiments suggested globospiramine to bind strongly with 1,3-β-glucan synthase and Als3 adhesin-enzymes indirectly involved in apoptosis-driven candidal inhibition.
Collapse
Affiliation(s)
- Joe Anthony H. Manzano
- The Graduate School, University of Santo Tomas, España Blvd., Manila 1015, Philippines;
- UST Laboratories for Vaccine Science, Molecular Biology and Biotechnology, Research Center for the Natural and Applied Sciences, University of Santo Tomas, España Blvd., Manila 1015, Philippines;
- Laboratory for Organic Reactivity, Discovery, and Synthesis (LORDS), Research Center for the Natural and Applied Sciences, University of Santo Tomas, España Blvd., Manila 1015, Philippines
| | - Simone Brogi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy;
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy;
| | - Allan Patrick G. Macabeo
- Laboratory for Organic Reactivity, Discovery, and Synthesis (LORDS), Research Center for the Natural and Applied Sciences, University of Santo Tomas, España Blvd., Manila 1015, Philippines
- Department of Chemistry, College of Science, University of Santo Tomas, España Blvd., Manila 1015, Philippines
| | - Nicanor Austriaco
- UST Laboratories for Vaccine Science, Molecular Biology and Biotechnology, Research Center for the Natural and Applied Sciences, University of Santo Tomas, España Blvd., Manila 1015, Philippines;
- Department of Biological Sciences, College of Science, University of Santo Tomas, España Blvd., Manila 1015, Philippines
| |
Collapse
|
9
|
Ouf SA, Gomha SM, Farag B, Zaki ME, Ewies MM, Sharawy IA, Khalil FO, Mahmoud HK. Synthesis of novel Bis-1,2,4-Triazolo[3,4-b][1,3,4]Thiadiazines from natural camphoric acid as potential anti-candidal agents. RESULTS IN CHEMISTRY 2024; 7:101406. [DOI: 10.1016/j.rechem.2024.101406] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025] Open
|
10
|
Benjamin I, Benson CU, Adalikwu SA, Nduoma FA, Akor FO, Odey MO, Ezeani EC, Anyambula IA, Odume MA, Louis H. Investigating the potential of thiazolyl carbohydrazides derivatives as anti-Candida albicans agents: An intuition from molecular modelling, pharmacokinetic evaluation, and molecular docking analysis. CHEMICAL PHYSICS IMPACT 2023; 7:100275. [DOI: 10.1016/j.chphi.2023.100275] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
|
11
|
Almeida-Paes R, Frases S. New drugs and other strategies for the treatment of fungal infections. Future Microbiol 2023; 18:997-1000. [PMID: 37721205 DOI: 10.2217/fmb-2023-0169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2023] Open
Affiliation(s)
- Rodrigo Almeida-Paes
- Laboratório de Micologia, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, 21040-360, Brazil
- Rede Micologia - FAPERJ, Rio de Janeiro, 21941-902, Brazil
| | - Susana Frases
- Rede Micologia - FAPERJ, Rio de Janeiro, 21941-902, Brazil
- Laboratório de Biofísica de Fungos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil. Cidade Universitária, Ilha do Fundão, Rio de Janeiro, Brasil
| |
Collapse
|
12
|
Todaro B, Ottalagana E, Luin S, Santi M. Targeting Peptides: The New Generation of Targeted Drug Delivery Systems. Pharmaceutics 2023; 15:1648. [PMID: 37376097 DOI: 10.3390/pharmaceutics15061648] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/22/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Peptides can act as targeting molecules, analogously to oligonucleotide aptamers and antibodies. They are particularly efficient in terms of production and stability in physiological environments; in recent years, they have been increasingly studied as targeting agents for several diseases, from tumors to central nervous system disorders, also thanks to the ability of some of them to cross the blood-brain barrier. In this review, we will describe the techniques employed for their experimental and in silico design, as well as their possible applications. We will also discuss advancements in their formulation and chemical modifications that make them even more stable and effective. Finally, we will discuss how their use could effectively help to overcome various physiological problems and improve existing treatments.
Collapse
Affiliation(s)
- Biagio Todaro
- NEST Laboratory, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Elisa Ottalagana
- NEST Laboratory, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
- Fondazione Pisana per la Scienza, Via Ferruccio Giovannini 13, San Giuliano Terme, 56017 Pisa, Italy
| | - Stefano Luin
- NEST Laboratory, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Melissa Santi
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| |
Collapse
|
13
|
Karine Marcomini E, Negri M. Fungal quorum-sensing molecules and antiseptics: a promising strategy for biofilm modulation? Drug Discov Today 2023:103624. [PMID: 37224996 DOI: 10.1016/j.drudis.2023.103624] [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: 01/30/2023] [Revised: 04/27/2023] [Accepted: 05/17/2023] [Indexed: 05/26/2023]
Abstract
New strategies to control fungal biofilms are essential, especially those that interfere in the biofilm organization process and cellular communication, known as quorum sensing. The effect of antiseptics and quorum-sensing molecules (QSMs) have been considered with regard to this; however, little has been elucidated, particularly because studies are often restricted to the action of antiseptics and QSMs against a few fungal genera. In this review, we discuss progress reported in the literature thus far and analyze, through in silico methods, 13 fungal QSMs with regard to their physicochemical, pharmacological, and toxicity properties, including their mutagenicity, tumorigenicity, hepatotoxicity, and nephrotoxicity. From these in silico analyses, we highlight 4-hydroxyphenylacetic acid and tryptophol as having satisfactory properties and, thus, propose that these should be investigated further as antifungal agents. We also recommend future in vitro approaches to determine the association of QSMs with commonly used antiseptics as potential antibiofilm agents.
Collapse
|
14
|
Dalwadi SM, Hunt A, Bonnen MD, Ghebre YT. Computational approaches for drug repurposing in oncology: untapped opportunity for high value innovation. Front Oncol 2023; 13:1198284. [PMID: 37274281 PMCID: PMC10233043 DOI: 10.3389/fonc.2023.1198284] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 05/05/2023] [Indexed: 06/06/2023] Open
Abstract
Historically, the effort by academia and industry to develop new chemical entities into lifesaving drugs has limited success in meeting the demands of today's healthcare. Repurposing drugs that are originally approved by the United States Food and Drug Administration or by regulatory authorities around the globe is an attractive strategy to rapidly develop much-needed therapeutics for oncologic indications that extend from treating cancer to managing treatment-related complications. This review discusses computational approaches to harness existing drugs for new therapeutic use in oncology.
Collapse
Affiliation(s)
| | | | - Mark D. Bonnen
- Department of Radiation Oncology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Yohannes T. Ghebre
- Department of Radiation Oncology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
15
|
Ibrahim MS, Farag B, Y. Al-Humaidi J, Zaki MEA, Fathalla M, Gomha SM. Mechanochemical Synthesis and Molecular Docking Studies of New Azines Bearing Indole as Anticancer Agents. Molecules 2023; 28:3869. [PMID: 37175279 PMCID: PMC10180502 DOI: 10.3390/molecules28093869] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/05/2023] [Accepted: 04/07/2023] [Indexed: 05/15/2023] Open
Abstract
The development of new approaches for the synthesis of new bioactive heterocyclic derivatives is of the utmost importance for pharmaceutical industry. In this regard, the present study reports the green synthesis of new benzaldazine and ketazine derivatives via the condensation of various carbonyl compounds (aldehydes and ketones with the 3-(1-hydrazineylideneethyl)-1H-indole using the grinding method with one drop of acetic acid). Various spectroscopic techniques were used to identify the structures of the synthesized derivatives. Furthermore, the anticancer activities of the reported azine derivatives were evaluated against colon, hepatocellular, and breast carcinoma cell lines using the MTT technique with doxorubicin as a reference medication. The findings suggested that the synthesized derivatives exhibited potential anti-tumor activities toward different cell lines. For example, 3c, 3d, 3h, 9, and 13 exhibited interesting activity with an IC50 value of 4.27-8.15 µM towards the HCT-116 cell line as compared to doxorubicin (IC50 = 5.23 ± 0.29 µM). In addition, 3c, 3d, 3h, 9, 11, and 13 showed excellent cytotoxic activities (IC50 = 4.09-9.05 µM) towards the HePG-2 cell line compared to doxorubicin (IC50 = 4.50 ± 0.20 µM), and 3d, 3h, 9, and 13 demonstrated high potency (IC50 = 6.19-8.39 µM) towards the breast cell line (MCF-7) as compared to the reference drug (IC50 = 4.17 ± 0.20 µM). The molecular interactions between derivatives 3a-h, 7, 9, 11, 13, and the CDK-5 enzyme (PDB ID: 3IG7) were studied further using molecular docking indicating a high level of support for the experimental results. Furthermore, the drug-likeness analysis of the reported derivatives indicated that derivative 9 (binding affinity = -8.34 kcal/mol) would have a better pharmacokinetics, drug-likeness, and oral bioavailability as compared to doxorubicin (-7.04 kcal/mol). These results along with the structure-activity relationship (SAR) of the reported derivatives will pave the way for the design of additional azines bearing indole with potential anticancer activities.
Collapse
Affiliation(s)
- Mohamed S. Ibrahim
- Department of Chemistry, Faculty of Science, Islamic University of Madinah, Madinah 42351, Saudi Arabia; (M.S.I.); (M.F.)
| | - Basant Farag
- Department of Chemistry, Faculty of Science, Zagazig University, Zagazig 44519, Egypt;
| | - Jehan Y. Al-Humaidi
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia;
| | - Magdi E. A. Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia;
| | - Maher Fathalla
- Department of Chemistry, Faculty of Science, Islamic University of Madinah, Madinah 42351, Saudi Arabia; (M.S.I.); (M.F.)
| | - Sobhi M. Gomha
- Department of Chemistry, Faculty of Science, Islamic University of Madinah, Madinah 42351, Saudi Arabia; (M.S.I.); (M.F.)
- Department of Chemistry, Faculty of Science, Cairo University, Cairo 12613, Egypt
| |
Collapse
|
16
|
Sulistyowaty MI, Putra GS, Budiati T, Indrianingsih AW, Anwari F, Kesuma D, Matsunami K, Yamauchi T. Synthesis, In Silico Study, Antibacterial and Antifungal Activities of N-phenylbenzamides. Int J Mol Sci 2023; 24:ijms24032745. [PMID: 36769066 PMCID: PMC9917131 DOI: 10.3390/ijms24032745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Antibiotic and antifungal resistance problems have been prevalent in recent decades. One of the efforts to solve the problems is to develop new medicines with more potent antibacterial and antifungal activity. N-phenylbenzamides have the potential to be developed as antibacterial and antifungal medicine. This study aimed to synthesize N-phenylbenzamides and evaluate their in silico and in vitro antibacterial and antifungal activities. The in silico studies conducted absorption, distribution, metabolism, excretion and toxicity (ADMET) predictions along with molecular docking studies. ADMET predictions used pkCSM software online, while the docking studies used MVD software (Molegro ® Virtual Docker version 5.5) on Aminoglycosid-2 ″-phosphotransferase-IIa (APH2 ″-IIa) enzyme with protein data bank (PDB) ID code 3HAV as antibacterial and aspartic proteinases enzyme (Saps) with PDB ID code 2QZX as an antifungal. In vitro, antibacterial and antifungal tests were carried out using the zone of inhibition (ZOI) method. The five N-phenylbenzamides (3a-e) were successfully synthesized with a high yield. Based on in silico and in vitro studies, compounds 3a-e have antibacterial and antifungal activities, where they can inhibit the growth of Gram-positive bacteria (Staphylococcus aureus), Gram-negative (Escherichia coli), and Candida albicans. Therefore, compounds 3a-e can be developed as a topical antibacterial and antifungal agent.
Collapse
Affiliation(s)
- Melanny Ika Sulistyowaty
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Surabaya 60115, Indonesia
- Correspondence: (M.I.S.); (D.K.)
| | - Galih Satrio Putra
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Negeri Malang, Malang 65151, Indonesia
| | - Tutuk Budiati
- Faculty of Pharmacy, Widya Mandala Catholic University, Surabaya 60265, Indonesia
| | - Anastasia Wheni Indrianingsih
- Research Center for Food Technology and Processing, National Research and Innovation Agency (BRIN), Yogyakarta 55861, Indonesia
| | - Farida Anwari
- Medical Laboratory Science, University of Anwar Medika, Sidoarjo 61262, Indonesia
| | - Dini Kesuma
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Surabaya, Surabaya 60293, Indonesia
- Correspondence: (M.I.S.); (D.K.)
| | - Katsuyoshi Matsunami
- Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima 734-8553, Japan
| | - Takayasu Yamauchi
- Faculty of Pharmaceutical Science, Hoshi University, 2-4-41, Ebara, Shinagawa, Tokyo 142-8501, Japan
| |
Collapse
|
17
|
Kashyap D, Jakhmola S, Tiwari D, Kumar R, Moorthy NSHN, Elangovan M, Brás NF, Jha HC. Plant derived active compounds as potential anti SARS-CoV-2 agents: an in-silico study. J Biomol Struct Dyn 2022; 40:10629-10650. [PMID: 34225565 DOI: 10.1080/07391102.2021.1947384] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Plants are a valued potential source of drugs for a variety of diseases and are often considered less toxic to humans. We investigated antiviral compounds that may potentially target SARS-CoV-2 antigenic spike (S) and host proteins; angiotensin-converting enzyme2 (ACE2), and transmembrane serine protease2 (TMPRSS2). We scrutinized 36 phytochemicals from 15 Indian medicinal plants known to be effective against RNA viruses via molecular docking. Besides, the TMPRSS2 structure was modeled and validated using the SWISS-MODEL. Docking was performed using Autodock Vina and 4.2 followed by visualization of the docking poses on Pymol version 2.4.0 and Discovery Studio Visualizer. Molecular docking showed that 12 out of 36 active compounds interacted efficiently with S, ACE2, and TMPRSS2 proteins. The ADMET profile generated using the swissADME and pkCSM server revealed that these compounds were possessed druggable properties. The Amber 12 simulation package was used to carry out energy minimizations and molecular dynamics (MD) simulations. The total simulation time for both S protein: WFA and S protein: WND complexes was 300 ns (100 ns per replica). A total of 120 structures were extracted from the last 60 ns of each MD simulation for further analysis. MM-PBSA and MM-GBSA were employed to assess the binding energy of each ligand and the receptor-binding domain of the viral S-protein. The methods suggested that WND and WFA showed thermodynamically favorable binding energies, and the S protein had a higher affinity with WND. Interestingly, Leu455 hotspot residue in the S protein, also predicted to participate in binding with ACE2, was engaged by WND and WFA. HighlightsPlants' natural active compounds may aid in the development of COVID-19 therapeutics.MD simulation study revealed stable binding of withanolide D and withaferin A with spike proteinWithanolide D and withaferin A could be effective against SARS-CoV-2 spike protein.Discovery of druggable agents that have less or lack of binding affinity with ACE2 to avoid the organs associated with comorbidities.According to ADMET selected phytochemicals may be used as druggable compounds.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Dharmendra Kashyap
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Shweta Jakhmola
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Deeksha Tiwari
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Rajesh Kumar
- Department of Physics, Indian Institute of Technology Indore, Indore, India
| | | | | | - Natércia F Brás
- LAQV, REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - Hem Chandra Jha
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| |
Collapse
|
18
|
A machine learning based model accurately predicts cellular response to electric fields in multiple cell types. Sci Rep 2022; 12:9912. [PMID: 35705588 PMCID: PMC9200721 DOI: 10.1038/s41598-022-13925-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/30/2022] [Indexed: 11/24/2022] Open
Abstract
Many cell types migrate in response to naturally generated electric fields. Furthermore, it has been suggested that the external application of an electric field may be used to intervene in and optimize natural processes such as wound healing. Precise cell guidance suitable for such optimization may rely on predictive models of cell migration, which do not generalize. Here, we present a machine learning model that can forecast directedness of cell migration given a timeseries of previous directedness and electric field values. This model is trained using time series galvanotaxis data of mammalian cranial neural crest cells obtained through time-lapse microscopy of cells cultured at 37 °C in a galvanotaxis chamber at ambient pressure. Next, we show that our modeling approach can be used for a variety of cell types and experimental conditions with very limited training data using transfer learning methods. We adapt the model to predict cell behavior for keratocytes (room temperature, ~ 18–20 °C) and keratinocytes (37 °C) under similar experimental conditions with a small dataset (~ 2–5 cells). Finally, this model can be used to perform in silico studies by simulating cell migration lines under time-varying and unseen electric fields. We demonstrate this by simulating feedback control on cell migration using a proportional–integral–derivative (PID) controller. This data-driven approach provides predictive models of cell migration that may be suitable for designing electric field based cellular control mechanisms for applications in precision medicine such as wound healing.
Collapse
|
19
|
Guerah NEH, Zerrouki K, Benslama O, Daran JC, Bouacida S, Bouchene R. New polymorph for Cd(II) chloro‑bridged coordination polymer based on 3-aminopyrazin-2-carboxylic acid: Synthesis, structural characterization, Hirshfeld surface analysis, thermal properties and molecular docking study on the antifungal activity. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132681] [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]
|
20
|
Gao A, Kouznetsova VL, Tsigelny IF. Machine-Learning-Based Virtual Screening to Repurpose Drugs for Treatment of Candida albicans Infection. Mycoses 2022; 65:794-805. [PMID: 35639510 DOI: 10.1111/myc.13475] [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: 04/17/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Approximately 30% of Candida genus isolates are resistant to all currently available antifungal drugs and it is highly important to develop new treatments. Additionally, many current drugs are toxic and cause unwanted side effects. 1,3-beta-glucan synthase is an essential enzyme that builds the cell walls of Candida. OBJECTIVES Targeting CaFKS1, a subunit of the synthase, could be used to fight Candida. METHODS In the present study, a machine-learning model based on chemical descriptors was trained to recognize drugs that inhibit CaFKS1. The model attained 96.72% accuracy for classifying between active and inactive drug compounds. Descriptors for FDA-approved and other drugs were calculated and the model was used to predict the potential activity of these drugs against CaFKS1. RESULTS Several drugs, including goserelin and icatibant, were detected as active with high confidence. Many of the drugs, interestingly, were gonadotrophin-releasing hormone (GnRH) antagonists or agonists. A literature search found that five of the predicted drugs inhibit Candida experimentally. CONCLUSIONS This study yields promising drugs to be repurposed to combat Candida albicans infection. Future steps include testing the drugs on fungal cells in vitro.
Collapse
Affiliation(s)
- Andrew Gao
- REHS Program, San Diego Supercomputer Center, University of California at San Diego, La Jolla, Calif, USA.,MAP Program, University of California at San Diego, La Jolla, Calif, USA
| | - Valentina L Kouznetsova
- San Diego Supercomputer Center, University of California at San Diego, La Jolla, Calif, USA.,BiAna, La Jolla, Calif, USA
| | - Igor F Tsigelny
- San Diego Supercomputer Center, University of California at San Diego, La Jolla, Calif, USA.,BiAna, La Jolla, Calif, USA.,Department of Neurosciences, University of California at San Diego, La Jolla, Calif, USA
| |
Collapse
|
21
|
Rocha da Silva C, Sá LGDAV, Dos Santos EV, Ferreira TL, Coutinho TDNP, Moreira LEA, de Sousa Campos R, de Andrade CR, Barbosa da Silva WM, de Sá Carneiro I, Silva J, Dos Santos HS, Marinho ES, Cavalcanti BC, de Moraes MO, Júnior HVN, Andrade Neto JB. Evaluation of the antifungal effect of chlorogenic acid against strains of Candida spp. resistant to fluconazole: apoptosis induction and in silico analysis of the possible mechanisms of action. J Med Microbiol 2022; 71. [PMID: 35575783 DOI: 10.1099/jmm.0.001526] [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] [Indexed: 01/19/2023] Open
Abstract
Introduction. Candida spp. are commensal fungal pathogens of humans, but when there is an imbalance in the microbiota, or weak host immunity, these yeasts can become pathogenic, generating high medical costs.Gap Statement. With the increase in resistance to conventional antifungals, the development of new therapeutic strategies is necessary. This study evaluated the in vitro antifungal activity of chlorogenic acid against fluconazole-resistant strains of Candida spp. Mechanism of action through flow cytometry and in silico analyses, as well as molecular docking assays with ALS3 and SAP5, important proteins in the pathogenesis of Candida albicans associated with the adhesion process and biofilm formation.Results. The chlorogenic acid showed in vitro antifungal activity against the strains tested, causing reduced cell viability, increased potential for mitochondrial depolarization and production of reactive oxygen species, DNA fragmentation and phosphatidylserine externalization, indicating an apoptotic process. Concerning the analysis through docking, the complexes formed between chlorogenic acid and the targets Thymidylate Kinase, CYP51, 1Yeast Cytochrome BC1 Complex e Exo-B-(1,3)-glucanase demonstrated more favourable binding energy. In addition, chlorogenic acid presented significant interactions with the ALS3 active site residues of C. albicans, important in the adhesion process and resistance to fluconazole. Regarding molecular docking with SAP5, no significant interactions were found between chlorogenic acid and the active site of the enzyme.Conclusion. We concluded that chlorogenic acid has potential use as an adjuvant in antifungal therapies, due to its anti-Candida activity and ability to interact with important drug targets.
Collapse
Affiliation(s)
- Cecília Rocha da Silva
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Lívia Gurgel do Amaral Valente Sá
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil.,Christus University Center (UNICHRISTUS), Fortaleza, CE, Brazil
| | | | | | | | - Lara Elloyse Almeida Moreira
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Rosana de Sousa Campos
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Christus University Center (UNICHRISTUS), Fortaleza, CE, Brazil
| | | | | | - Igor de Sá Carneiro
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil
| | - Jacilene Silva
- Department of Chemistry, Group of Theoretical Chemistry and Electrochemistry (GQTE), State University of Ceará, Limoeiro do Norte, Ceará, Brazil
| | - Hélcio Silva Dos Santos
- Science and Technology Centre, Course of Chemistry, State University Vale do Acaraú, Sobral, CE, Brazil
| | - Emmanuel Silva Marinho
- Department of Chemistry, Group of Theoretical Chemistry and Electrochemistry (GQTE), State University of Ceará, Limoeiro do Norte, Ceará, Brazil
| | - Bruno Coelho Cavalcanti
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil.,Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Manoel Odorico de Moraes
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil.,Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Hélio Vitoriano Nobre Júnior
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - João Batista Andrade Neto
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil.,Christus University Center (UNICHRISTUS), Fortaleza, CE, Brazil
| |
Collapse
|
22
|
Handayani S, Nurdiana N, Winarsih S, Endharti AT. Holothurin Compound from Sea Cucumber (Holothuria sp.) as Antifungal Alternative against Candida Infections. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.8086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: The previous studies have identified chemical compounds in sea cucumbers that have antifungal properties. However, further information on the underlying antifungal needed to be updated.
AIM: This study aimed to discover efficient antifungal treatments against candidiasis disease.
MATERIALS AND METHODS: This study analyzed the antifungal activity from Holothurin against Candida albicans in silico using molecular docking and minimum inhibitory concentration (MIC).
RESULTS: The results revealed that holothurin has a binding affinity of −7.9 kcal/mol and MIC value of 1.5 mg/ml.
CONCLUSION: Holothurin may inhibit the infection of C. albicans. Furthermore, additional research is required to validate the activity of this compound.
Collapse
|
23
|
Silva SL, de Oliveira Pereira F, Cordeiro LV, Diniz Neto H, Dos Santos Maia M, da Silva Souza HD, de Athayde-Filho PF, Scotti MT, Scotti L, de Oliveira Lima E. Antifungal activity of 2-Chloro-N-phenylacetamide, docking and molecular dynamics studies against clinical isolates of Candida tropicalis and Candida parapsilosis. J Appl Microbiol 2022; 132:3601-3617. [PMID: 35179275 DOI: 10.1111/jam.15498] [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: 12/28/2021] [Revised: 02/06/2022] [Accepted: 02/14/2022] [Indexed: 11/28/2022]
Abstract
AIMS This study evaluated the antifungal, antibiofilm, and molecular docking of 2-Chloro-N-phenylacetamide against clinical isolates of Candida tropicalis and Candida parapsilosis. METHODS AND RESULTS MIC of the test drugs was determined by microdilution. A1Cl obtained MIC values ranging from 16 and 256 μg/mL. Fluconazole MIC ranging from 16 and 512 μg/mL. MIC of A1Cl showed fungicide activity, emphasizing the solid antifungal potential of this drug. An association study was performed with A1Cl and fluconazole (checkerboard), revealing indifference by decreasing. Thus, we conducted this study using A1Cl isolated. In the micromorphological assay, the test drugs reduced the production of virulence structures compared to the control (concentration-dependent effect). A1Cl inhibited in vitro biofilm formation at all concentrations tested (1/4MIC to 8xMIC) (p<0.05) and reduced mature biofilm biomass (p<0.05) against C. tropicalis and C. parapsilosis. In the ex vivo biofilm susceptibility testing (human nails fragments), A1Cl inhibited biofilm formation and reduced mature biofilm biomass (p<0.05) more than 50% at MIC. Fluconazole had a similar effect at 4xMIC. In silico studies suggest that the mechanism of antifungal activity of A1Cl involves the inhibition of the enzyme dihydrofolate reductase rather than geranylgeranyltransferase-I. CONCLUSIONS The results suggest that A1Cl is a promising antifungal agent. Furthermore, this activity is related to attenuation of expression of virulence factors and antibiofilm effects against C. tropicalis and C. parapsilosis. SIGNIFICANCE AND IMPACT OF THE STUDY Our study provides the first evidence that A1Cl, a novel synthetic drug, has fungicidal effects against C. tropicalis and C. parapsilosis. Furthermore, in vitro and ex vivo biofilms assays have demonstrated the potential antibiofilm of A1Cl. The mechanism of action involves inhibiting the enzyme dihydrofolate reductase, which was supported by in silico analyses. Therefore, this potential can be explored as a therapeutic alternative for onychomycosis and, at the same time, contribute to decreasing the resistance of clinical isolates of C. tropicalis and C. parapsilosis.
Collapse
Affiliation(s)
- Shellygton Lima Silva
- Postgraduate Program in Natural and Bioactive Synthetic Products, Department of Pharmaceutical Sciences, Federal University of Paraiba, João Pessoa, Brazil
| | - Fillipe de Oliveira Pereira
- Biochemistry Laboratory, Academic Unit of Health, Education and Health Center, Federal University of Campina Grande, Cuité, Brazil
| | - Laisa Vilar Cordeiro
- Postgraduate Program in Natural and Bioactive Synthetic Products, Department of Pharmaceutical Sciences, Federal University of Paraiba, João Pessoa, Brazil
| | - Hermes Diniz Neto
- Postgraduate Program in Natural and Bioactive Synthetic Products, Department of Pharmaceutical Sciences, Federal University of Paraiba, João Pessoa, Brazil
| | - Mayara Dos Santos Maia
- Postgraduate Program in Natural and Bioactive Synthetic Products, Department of Pharmaceutical Sciences, Federal University of Paraiba, João Pessoa, Brazil
| | - Helivado Diogenes da Silva Souza
- Bioenergy and Organic Synthesis Research Laboratory, Department of Chemistry, University of Paraiba, João Pessoa, Paraíba, Brazil
| | - Petrônio F de Athayde-Filho
- Bioenergy and Organic Synthesis Research Laboratory, Department of Chemistry, University of Paraiba, João Pessoa, Paraíba, Brazil
| | - Marcus Tullius Scotti
- Postgraduate Program in Natural and Bioactive Synthetic Products, Department of Pharmaceutical Sciences, Federal University of Paraiba, João Pessoa, Brazil
| | - Luciana Scotti
- Postgraduate Program in Natural and Bioactive Synthetic Products, Department of Pharmaceutical Sciences, Federal University of Paraiba, João Pessoa, Brazil
| | - Edeltrudes de Oliveira Lima
- Postgraduate Program in Natural and Bioactive Synthetic Products, Department of Pharmaceutical Sciences, Federal University of Paraiba, João Pessoa, Brazil
| |
Collapse
|
24
|
Kasabe UI, Kale KB, Rode NR, Shelar AV, Patil RH, Mhaske PC, Chaskar MG. Synthesis and antifungal screening of tetramethyl hexahydro-1 H-xanthene-1,8(2 H)-dione derivatives as potential inhibitors of morphogenesis and biofilm formation in Candida albicans. NEW J CHEM 2022; 46:2128-2139. [DOI: 10.1039/d1nj04168a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
A series of hexahydro-1H-xanthene-1,8(2H)-dione derivatives were synthesized. All the derivatives were screened for their anti-virulence properties againstCandida albicans.In silicostudies were performed to corroborate the experimentally observed facts.
Collapse
Affiliation(s)
- Umesh I. Kasabe
- Department of Chemistry, Baburaoji Gholap College (Affiliated to Savitribai Phule Pune University, Pune), Sangvi, Pune-411027, India
| | - Kishor B. Kale
- Department of Chemistry, Nowrosjee Wadia College (Affiliated to Savitribai Phule Pune University, Pune), Pune-411001, India
| | - Nitin R. Rode
- Department of Chemistry, Nowrosjee Wadia College (Affiliated to Savitribai Phule Pune University, Pune), Pune-411001, India
| | - Amruta V. Shelar
- Department of Technology, Savitribai Phule Pune University, Pune-411007, India
| | - Rajendra H. Patil
- Department of Biotechnology, Savitribai Phule Pune University, Pune-411007, India
| | - Pravin C. Mhaske
- Department of Chemistry, Sir Parashurambhau College (Affiliated to Savitribai Phule Pune University, Pune), Pune-411030, India
| | - Manohar G. Chaskar
- Department of Chemistry, Baburaoji Gholap College (Affiliated to Savitribai Phule Pune University, Pune), Sangvi, Pune-411027, India
- Dean, Science and Technology, Savitribai Phule Pune University, Pune-411007, India
| |
Collapse
|
25
|
Hajj RE, Tawk L, Itani S, Hamie M, Ezzeddine J, El Sabban M, El Hajj H. Toxoplasmosis: Current and Emerging Parasite Druggable Targets. Microorganisms 2021; 9:microorganisms9122531. [PMID: 34946133 PMCID: PMC8707595 DOI: 10.3390/microorganisms9122531] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/10/2021] [Accepted: 11/10/2021] [Indexed: 12/19/2022] Open
Abstract
Toxoplasmosis is a prevalent disease affecting a wide range of hosts including approximately one-third of the human population. It is caused by the sporozoan parasite Toxoplasma gondii (T. gondii), which instigates a range of symptoms, manifesting as acute and chronic forms and varying from ocular to deleterious congenital or neuro-toxoplasmosis. Toxoplasmosis may cause serious health problems in fetuses, newborns, and immunocompromised patients. Recently, associations between toxoplasmosis and various neuropathies and different types of cancer were documented. In the veterinary sector, toxoplasmosis results in recurring abortions, leading to significant economic losses. Treatment of toxoplasmosis remains intricate and encompasses general antiparasitic and antibacterial drugs. The efficacy of these drugs is hindered by intolerance, side effects, and emergence of parasite resistance. Furthermore, all currently used drugs in the clinic target acute toxoplasmosis, with no or little effect on the chronic form. In this review, we will provide a comprehensive overview on the currently used and emergent drugs and their respective parasitic targets to combat toxoplasmosis. We will also abridge the repurposing of certain drugs, their targets, and highlight future druggable targets to enhance the therapeutic efficacy against toxoplasmosis, hence lessening its burden and potentially alleviating the complications of its associated diseases.
Collapse
Affiliation(s)
- Rana El Hajj
- Department of Biological Sciences, Beirut Arab University, P.O. Box 11-5020, Riad El Solh, Beirut 1107 2809, Lebanon;
| | - Lina Tawk
- Department of Medical Laboratory Sciences, Faculty of Health Sciences, University of Balamand, Beirut 1100 2807, Lebanon; (L.T.); (J.E.)
| | - Shaymaa Itani
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Riad El-Solh, Beirut 1107 2020, Lebanon; (S.I.); (M.H.)
| | - Maguy Hamie
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Riad El-Solh, Beirut 1107 2020, Lebanon; (S.I.); (M.H.)
| | - Jana Ezzeddine
- Department of Medical Laboratory Sciences, Faculty of Health Sciences, University of Balamand, Beirut 1100 2807, Lebanon; (L.T.); (J.E.)
| | - Marwan El Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Riad El-Solh, Beirut 1107 2020, Lebanon;
| | - Hiba El Hajj
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236, Riad El-Solh, Beirut 1107 2020, Lebanon; (S.I.); (M.H.)
- Correspondence: ; Tel.: +961–1-350000 (ext. 4897)
| |
Collapse
|
26
|
Doustimotlagh AH, Taheri S, Mansourian M, Eftekhari M. Extraction and Identification of Two Flavonoids in Phlomoides hyoscyamoides as an Endemic Plant of Iran: The Role of Quercetin in the Activation of the Glutathione Peroxidase, the Improvement of the Hydroxyproline and Protein Oxidation in Bile Duct-Ligated Rats. Curr Comput Aided Drug Des 2021; 16:629-640. [PMID: 31481005 DOI: 10.2174/1573409915666190903163335] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/22/2019] [Accepted: 08/02/2019] [Indexed: 01/04/2023]
Abstract
BACKGROUND Cholestatic liver disease, a serious chronic condition that develops progressive hepatic degeneration through free radicals. OBJECTIVE The present study was designed to extract and identify two flavonoids in Phlomoides hyoscyamoides plant, native to Iran and evaluate the role of quercetin identified on the liver injury among bile ductligated rats. METHODS This study was conducted on 25 male Wistar rats within three groups of sham control, mere bile duct-ligated, and bile duct-ligated with quercetin. The bile duct-ligated animals received quercetin at a dose of 50 mg/kg/day for 10 days, followed by biochemical tests, oxidative stress markers, activity of antioxidant enzymes and hematoxylin and eosin staining. Molecular docking was used to explore the interactive behavior of quercetin with glutathione peroxidase. RESULTS According to analyses of the obtained extract, two main active ingredients of P. hyoscyamoides were rutin and quercetin. Bile duct-ligated group showed a significant liver necrosis, a clear increase in plasma and tissue oxidative stress parameters, and a decrease in glutathione peroxidase activity as compared to sham control group. Quercetin injection in bile duct-ligated rats resulted in significant decrease in hydroxyproline, protein carbonyl and histopathologic indexes and significant increase in glutathione peroxidase activity (P-value≤0.05). Based on the molecular docking, the quercetin was able to regulate the glutathione peroxidase activity. CONCLUSION The quercetin acts as an enzyme inducer by renewing the glutathione peroxidase activity and inhibiting the oxidation of proteins and hence decreases the oxidative stress. These results could be a sign of confirming the positive role of quercetin in attenuating the liver damage and degeneration.
Collapse
Affiliation(s)
- Amir H Doustimotlagh
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran,Department of Clinical Biochemistry, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Salman Taheri
- Chemistry & Chemical Engineering Research Center of Iran, P.O. Box 14335-186, Tehran, Iran
| | - Mahboubeh Mansourian
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran,Department of Pharmacology, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Mahdieh Eftekhari
- Department of Pharmacognosy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Amaral JL, Souza PFN, Oliveira JTA, Freire VN, Sousa DOB. Computational approach, scanning electron and fluorescence microscopies revealed insights into the action mechanisms of anticandidal peptide Mo-CBP 3-PepIII. Life Sci 2021; 281:119775. [PMID: 34186044 DOI: 10.1016/j.lfs.2021.119775] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 06/09/2021] [Accepted: 06/20/2021] [Indexed: 11/17/2022]
Abstract
AIMS The Candida genus is composed of opportunistic pathogens that threaten public health. Given the increase in resistance to current drugs, it is necessary to develop new drugs to treat infections by these pathogens. Antimicrobial peptides are promising alternative molecules with low cost, broad action spectrum and low resistance induction. This study aimed to clarify the action mechanisms of synthetic peptides against Candida albicans. MAIN METHODS The mode of action of the anticandidal peptides Mo-CBP3-PepIII were analyzed through molecular dynamics and quantum biochemistry methods against Exo-β-1,3-glucanase (EXG), vital to cell wall metabolism. Furthermore, scanning electron (SEM) and fluorescence (FM) microscopies were employed to corroborate the in silico data. KEY FINDINGS Mo-CBP3-PepIII strongly interacted with EXG (-122.2 kcal mol-1) at the active site, higher than the commercial inhibitor pepstatin. Also, molecular dynamics revealed the insertion of Mo-CBP3-PepIII into the yeast membrane. SEM analyses revealed that Mo-CBP3-PepIII induced cracks and scars of the cell wall and FM analyses confirmed the pore formation on the Candida membrane. SIGNIFICANCE Mo-CBP3-PepIII has strong potential as a new drug with a broad spectrum of action, given its different mode of action compared to conventional drugs.
Collapse
Affiliation(s)
- Jackson L Amaral
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Ceará CEP 60.440-554, Brazil; Department of Physics, Federal University of Ceará, Fortaleza, Ceará CEP 60.440-554, Brazil.
| | - Pedro F N Souza
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Ceará CEP 60.440-554, Brazil
| | - Jose T A Oliveira
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Ceará CEP 60.440-554, Brazil
| | - Valder N Freire
- Department of Physics, Federal University of Ceará, Fortaleza, Ceará CEP 60.440-554, Brazil
| | - Daniele O B Sousa
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Ceará CEP 60.440-554, Brazil.
| |
Collapse
|
28
|
Molecular Human Targets of Bioactive Alkaloid-Type Compounds from Tabernaemontana cymose Jacq. Molecules 2021; 26:molecules26123765. [PMID: 34205626 PMCID: PMC8234993 DOI: 10.3390/molecules26123765] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/10/2021] [Accepted: 06/18/2021] [Indexed: 12/29/2022] Open
Abstract
Alkaloids are a group of secondary metabolites that have been widely studied for the discovery of new drugs due to their properties on the central nervous system and their anti-inflammatory, antioxidant and anti-cancer activities. Molecular docking was performed for 10 indole alkaloids identified in the ethanol extract of Tabernaemontana cymosa Jacq. with 951 human targets involved in different diseases. The results were analyzed through the KEGG and STRING databases, finding the most relevant physiological associations for alkaloids. The molecule 5-oxocoronaridine proved to be the most active molecule against human proteins (binding energy affinity average = −9.2 kcal/mol) and the analysis of the interactions between the affected proteins pointed to the PI3K/ Akt/mTOR signaling pathway as the main target. The above indicates that indole alkaloids from T. cymosa constitute a promising source for the search and development of new treatments against different types of cancer.
Collapse
|
29
|
Uppar V, Chandrashekharappa S, Shivamallu C, P S, Kollur SP, Ortega-Castro J, Frau J, Flores-Holguín N, Basarikatti AI, Chougala M, Mohan M M, Banuprakash G, Jayadev, Venugopala KN, Nandeshwarappa BP, Veerapur R, Al-Kheraif AA, Elgorban AM, Syed A, Mudnakudu-Nagaraju KK, Padmashali B, Glossman-Mitnik D. Investigation of Antifungal Properties of Synthetic Dimethyl-4-Bromo-1-(Substituted Benzoyl) Pyrrolo[1,2-a] Quinoline-2,3-Dicarboxylates Analogues: Molecular Docking Studies and Conceptual DFT-Based Chemical Reactivity Descriptors and Pharmacokinetics Evaluation. Molecules 2021; 26:2722. [PMID: 34066433 PMCID: PMC8124935 DOI: 10.3390/molecules26092722] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 02/07/2023] Open
Abstract
Candida albicans, an opportunistic fungal pathogen, frequently colonizes immune-compromised patients and causes mild to severe systemic reactions. Only few antifungal drugs are currently in use for therapeutic treatment. However, evolution of a drug-resistant C. albicans fungal pathogen is of major concern in the treatment of patients, hence the clinical need for novel drug design and development. In this study, in vitro screening of novel putative pyrrolo[1,2-a]quinoline derivatives as the lead drug targets and in silico prediction of the binding potential of these lead molecules against C. albicans pathogenic proteins, such as secreted aspartic protease 3 (SAP3; 2H6T), surface protein β-glucanase (3N9K) and sterol 14-alpha demethylase (5TZ1), were carried out by molecular docking analyses. Further, biological activity-based QSAR and theoretical pharmacokinetic analysis were analyzed. Here, in vitro screening of novel analogue derivatives as drug targets against C. albicans showed inhibitory potential in the concentration of 0.4 µg for BQ-06, 07 and 08, 0.8 µg for BQ-01, 03, and 05, 1.6 µg for BQ-04 and 12.5 µg for BQ-02 in comparison to the standard antifungal drug fluconazole in the concentration of 30 µg. Further, in silico analysis of BQ-01, 03, 05 and 07 analogues docked on chimeric 2H6T, 3N9K and 5TZ1 revealed that these analogues show potential binding affinity, which is different from the therapeutic antifungal drug fluconazole. In addition, these molecules possess good drug-like properties based on the determination of conceptual Density Functional Theory (DFT)-based descriptors, QSAR and pharmacokinetics. Thus, the study offers significant insight into employing pyrrolo[1,2-a]quinoline analogues as novel antifungal agents against C. albicans that warrants further investigation.
Collapse
Affiliation(s)
- Vijayakumar Uppar
- Department of Chemistry, School of Basic Science, Rani Channamma University, Belagavi 591156, Karnataka, India; (V.U.); (A.I.B.)
| | - Sandeep Chandrashekharappa
- Institute for Stem Cell Science and Regenerative Medicine, NCBS, TIFR, GKVK-Campus Bellary road, Bengaluru 560065, Karnataka, India;
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) Raebareli, Lucknow (UP) 226002, India;
| | - Chandan Shivamallu
- Department of Biotechnology & Bioinformatics, Faculty of Life Sciences, JSS Academy of Higher Education and Research, Mysore 570015, Karnataka, India; (C.S.); (M.M.M.)
| | - Sushma P
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) Raebareli, Lucknow (UP) 226002, India;
| | - Shiva Prasad Kollur
- Department of Sciences, Amrita School of Arts and Sciences, Amrita Vishwa Vidyapeetham, Mysuru 570026, Karnataka, India;
| | - Joaquín Ortega-Castro
- Departament de Química, Universitat de les Illes Balears, E-07122 Palma de Malllorca, Spain; (J.O.-C.); (J.F.)
| | - Juan Frau
- Departament de Química, Universitat de les Illes Balears, E-07122 Palma de Malllorca, Spain; (J.O.-C.); (J.F.)
| | - Norma Flores-Holguín
- Laboratorio Virtual NANOCOSMOS, Departamento de Medio Ambiente y Energía, Centro de Investigación en Materiales Avanzados, Chihuahua, Chih 31136, Mexico; (N.F.-H.); (D.G.-M.)
| | - Atiyaparveen I. Basarikatti
- Department of Chemistry, School of Basic Science, Rani Channamma University, Belagavi 591156, Karnataka, India; (V.U.); (A.I.B.)
| | - Mallikarjun Chougala
- Department of Biotechnology, JSS College of Arts, Commerce and Science (Autonomous), Mysore 570025, Karnataka, India;
| | - Mrudula Mohan M
- Department of Biotechnology & Bioinformatics, Faculty of Life Sciences, JSS Academy of Higher Education and Research, Mysore 570015, Karnataka, India; (C.S.); (M.M.M.)
| | - Govindappa Banuprakash
- Department of Chemistry, SJB Institute of Technology, Bengaluru 560060, Kengeri, India; (G.B.); (J.)
| | - Jayadev
- Department of Chemistry, SJB Institute of Technology, Bengaluru 560060, Kengeri, India; (G.B.); (J.)
| | - Katharigatta N. Venugopala
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
- Department of Biotechnology and Food Technology, Faculty of Applied Sciences, Durban University of Technology, Durban 4001, South Africa
| | - Belakatte P. Nandeshwarappa
- Department of Studies in Chemistry, Shivagangothri, Davangere University, Davangere 577007, Karnataka, India;
| | - Ravindra Veerapur
- Department of Metallurgy and Materials Engineering, Malawi Institute of Technology, Malawi University of Science and Technology, P.O. Box-5916 Limbe, Malawi;
| | - Abdulaziz A. Al-Kheraif
- Dental Biomaterials Research Chair, Dental Health Department, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia;
| | - Abdallah M. Elgorban
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (A.M.E.); (A.S.)
| | - Asad Syed
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (A.M.E.); (A.S.)
| | - Kiran K. Mudnakudu-Nagaraju
- Department of Biotechnology & Bioinformatics, Faculty of Life Sciences, JSS Academy of Higher Education and Research, Mysore 570015, Karnataka, India; (C.S.); (M.M.M.)
| | - Basavaraj Padmashali
- Department of Chemistry, School of Basic Science, Rani Channamma University, Belagavi 591156, Karnataka, India; (V.U.); (A.I.B.)
| | - Daniel Glossman-Mitnik
- Laboratorio Virtual NANOCOSMOS, Departamento de Medio Ambiente y Energía, Centro de Investigación en Materiales Avanzados, Chihuahua, Chih 31136, Mexico; (N.F.-H.); (D.G.-M.)
| |
Collapse
|
30
|
da Nóbrega Alves D, Monteiro AFM, Andrade PN, Lazarini JG, Abílio GMF, Guerra FQS, Scotti MT, Scotti L, Rosalen PL, de Castro RD. Docking Prediction, Antifungal Activity, Anti-Biofilm Effects on Candida spp., and Toxicity against Human Cells of Cinnamaldehyde. Molecules 2020; 25:molecules25245969. [PMID: 33339401 PMCID: PMC7767272 DOI: 10.3390/molecules25245969] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/17/2022] Open
Abstract
Objective: This study evaluated the antifungal activity of cinnamaldehyde on Candida spp. In vitro and in situ assays were carried out to test cinnamaldehyde for its anti-Candida effects, antibiofilm activity, effects on fungal micromorphology, antioxidant activity, and toxicity on keratinocytes and human erythrocytes. Statistical analysis was performed considering α = 5%. Results: The minimum inhibitory concentration (MIC) and minimum fungicidal concentration (MFC) of cinnamaldehyde ranged from 18.91 μM to 37.83 μM. MIC values did not change in the presence of 0.8 M sorbitol, whereas an 8-fold increase was observed in the presence of ergosterol, suggesting that cinnamaldehyde may act on the cell membrane, which was subsequently confirmed by docking analysis. The action of cinnamaldehyde likely includes binding to enzymes involved in the formation of the cytoplasmic membrane in yeast cells. Cinnamaldehyde-treated microcultures showed impaired cellular development, with an expression of rare pseudo-hyphae and absence of chlamydoconidia. Cinnamaldehyde reduced biofilm adherence by 64.52% to 33.75% (p < 0.0001) at low concentrations (378.3–151.3 µM). Cinnamaldehyde did not show antioxidant properties. Conclusions: Cinnamaldehyde showed fungicidal activity through a mechanism of action likely related to ergosterol complexation; it was non-cytotoxic to keratinocytes and human erythrocytes and showed no antioxidant activity.
Collapse
Affiliation(s)
- Danielle da Nóbrega Alves
- Graduate Program in Natural and Synthetic Bioactive Products (PgPNSB), Department of Clinic and Social Dentistry, Center for Health Sciences, Federal University of Paraiba, João Pessoa-PB 58051-900, Brazil;
| | - Alex France Messias Monteiro
- Graduate Program in Natural and Synthetic Bioactive Products (PgPNSB), Department of Pharmaceutical Sciences, Center for Health Sciences, Federal University of Paraíba, João Pessoa-PB 58051-900, Brazil;
| | - Patrícia Néris Andrade
- Experimental Pharmacology and Cell Culture Laboratory, Center for Health Sciences, Federal University of Paraiba, João Pessoa-PB 58051-900, Brazil;
| | - Josy Goldoni Lazarini
- Department of Bioscience, Piracicaba Dental School, University of Campinas, Campinas-SP 13414-903, Brazil; (J.G.L.); (P.L.R.)
| | - Gisely Maria Freire Abílio
- Department of Physiology and Pathology, Center for Health Sciences, Federal University of Paraíba, João Pessoa-PB 58051-900, Brazil;
| | - Felipe Queiroga Sarmento Guerra
- Department of Pharmaceutical Sciences, Center for Health Sciences, Federal University of Paraíba, João Pessoa-PB 58051-900, Brazil;
| | - Marcus Tullius Scotti
- Graduate Program in Natural and Synthetic Bioactive Products (PgPNSB), Department of Chemistry, Center for Health Sciences, Federal University of Paraíba, João Pessoa-PB 58051-900, Brazil;
| | - Luciana Scotti
- Graduate Program in Natural and Synthetic Bioactive Products (PgPNSB), Cheminformatics Laboratory, Center for Health Sciences, Federal University of Paraíba, João Pessoa-PB 58051-900, Brazil;
| | - Pedro Luiz Rosalen
- Department of Bioscience, Piracicaba Dental School, University of Campinas, Campinas-SP 13414-903, Brazil; (J.G.L.); (P.L.R.)
- Biological Sciences Graduate Program (PPGCB), Institute of Biomedical Sciences (ICB), Federal University of Alfenas (UNIFAL-MG), Alfenas 37130-000, Brazil
| | - Ricardo Dias de Castro
- Department of Clinic and Social Dentistry, Center for Health Sciences, Federal University of Paraiba, João Pessoa-PB 58051-900, Brazil
- Correspondence: ; Tel.: +55-83-3216-7742
| |
Collapse
|
31
|
Dabade SJ, Mandloi D, Bajaj A. Molecular Docking and QSAR Studies of Coumarin Derivatives as NMT Inhibitors: Simple Structural Features as Potential Modulators of Antifungal Activity. LETT DRUG DES DISCOV 2020. [DOI: 10.2174/1570180817999200617105711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Background:
Treatments of fungal diseases, including Candidiasis, remain not up to
scratch in spite of the mounting catalog of synthetic antifungal agents. These have served as the
impetus for investigating new antifungal agents based on natural products. Consequently, genetic
algorithm-multiple linear regression (GA-MLR) based QSAR (Quantitative Structure-Activity Relationship)
studies of coumarin analogues along with molecular docking were carried out.
Methods:
Coumarin analogues with their MIC values were used to generate the training and test
sets of compounds for QSAR models development; the analogues were also docked into the binding
pocket of NMT (MyristoylCoA: protein N-myristoyltransferase).
Results and Discussion:
The statistical parameters for internal and external validation of QSAR
analysis (R2 = 0.830, Q2 = 0.758, R2Pred = 0.610 and R2m overall = 0.683 ), Y Randomization, Ridge
trace, VIF, tolerance and model criteria of Golbraikh and Tropsha data illustrate the robustness of
the best proposed QSAR model. Most of the analogues bind to the electrostatic, hydrophobic
clamp and display hydrogen bonding with amino acid residues of NMT. Interestingly, the most
active coumarin analogue (MolDock score of -189.257) was docked deeply within the binding
pocket of NMT, thereby displaying hydrogen bonding with Tyr107, Leu451, Leu450, Gln226,
Cys393 and Leu394 amino acid residues.
Conclusion:
The combinations of descriptors from various descriptor subsets in QSAR analysis
have highlighted the role of atomic properties such as polarizability and atomic van der Waals volume
to explain the inhibitory activity. The models and related information may pave the way for
important insight into the designing of putative NMT inhibitors for Candida albicans.
Collapse
Affiliation(s)
- Sapna Jain Dabade
- Department of Applied Science, SAGE University, Indore & Research Scholar at School of Chemical Sciences, Devi Ahilya Vishwavidyalaya, Indore, India
| | - Dheeraj Mandloi
- Institute of Engineering and Technology, Devi Ahilya Vishwavidyalaya, Indore, India
| | - Amritlal Bajaj
- School of Chemical Sciences, Devi Ahilya Vishwavidyalaya, Indore, India
| |
Collapse
|
32
|
Gurgel do Amaral Valente Sá L, da Silva CR, Neto JBDA, do Nascimento FBSA, Barroso FDD, da Silva LJ, Cabral VPDF, Barbosa AD, Silva J, Marinho ES, de Moraes MO, Rios MEF, Cavalcanti BC, Lima ISP, Júnior HVN. Antifungal activity of etomidate against growing biofilms of fluconazole-resistant Candida spp. strains, binding to mannoproteins and molecular docking with the ALS3 protein. J Med Microbiol 2020; 69:1221-1227. [PMID: 32894212 DOI: 10.1099/jmm.0.001241] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
This study evaluated the effect of etomidate against biofilms of Candida spp. and analysed through molecular docking the interaction of this drug with ALS3, an important protein for fungal adhesion. Three fluconazole-resistant fungi were used: Candida albicans, Candida parapsilosis and Candida tropicalis. Growing biofilms were exposed to etomidate at 31.25-500 µg ml-1. Then, an ALS3 adhesive protein from C. albicans was analysed through a molecular mapping technique, composed of a sequence of algorithms to perform molecular mapping simulation based on classic force field theory. Etomidate showed antifungal activity against growing biofilms of resistant C. albicans, C. parapsilosis and C. tropicalis at all concentrations used in the study. The etomidate coupling analysis revealed three interactions with the residues of interest compared to hepta-threonine, which remained at the ALS3 site. In addition, etomidate decreased the expression of mannoproteins on the surface of C. albicans. These results revealed that etomidate inhibited the growth of biofilms.
Collapse
Affiliation(s)
- Lívia Gurgel do Amaral Valente Sá
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, 60430-275, CE, Brazil.,Department of Clinical and Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, 60430-170, CE, Brazil
| | - Cecília Rocha da Silva
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, 60430-275, CE, Brazil.,Department of Clinical and Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, 60430-170, CE, Brazil
| | - João Batista de Andrade Neto
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, 60430-275, CE, Brazil.,Department of Clinical and Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, 60430-170, CE, Brazil
| | - Francisca Bruna Stefany Aires do Nascimento
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, 60430-275, CE, Brazil.,Department of Clinical and Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, 60430-170, CE, Brazil
| | - Fátima Daiana Dias Barroso
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, 60430-275, CE, Brazil.,Department of Clinical and Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, 60430-170, CE, Brazil
| | - Lisandra Juvêncio da Silva
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, 60430-275, CE, Brazil.,Department of Clinical and Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, 60430-170, CE, Brazil
| | - Vitória Pessoa de Farias Cabral
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, 60430-275, CE, Brazil.,Department of Clinical and Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, 60430-170, CE, Brazil
| | - Amanda Dias Barbosa
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, 60430-275, CE, Brazil.,Department of Clinical and Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, 60430-170, CE, Brazil
| | - Jacilene Silva
- Department of Chemistry, Group of Theoretical Chemistry and Electrochemistry (GQTE), State University of Ceará, Limoeiro do Norte, 62930-000, CE, Brazil
| | - Emmanuel Silva Marinho
- Department of Chemistry, Group of Theoretical Chemistry and Electrochemistry (GQTE), State University of Ceará, Limoeiro do Norte, 62930-000, CE, Brazil
| | - Manoel Odorico de Moraes
- Department of Physiology and Pharmacology, Neuropharmacology Laboratory, Federal University of Ceará, Fortaleza, 60430-275, CE, Brazil
| | | | - Bruno Coêlho Cavalcanti
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, 60430-275, CE, Brazil
| | - Iri Sandro Pampolha Lima
- Departament of Pharmacology, School of Medicine, Federal University of Ceará, Barbalha, 63048-080, CE, Brazil
| | | |
Collapse
|
33
|
LeBlanc EV, Polvi EJ, Veri AO, Privé GG, Cowen LE. Structure-guided approaches to targeting stress responses in human fungal pathogens. J Biol Chem 2020; 295:14458-14472. [PMID: 32796038 DOI: 10.1074/jbc.rev120.013731] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 08/11/2020] [Indexed: 11/06/2022] Open
Abstract
Fungi inhabit extraordinarily diverse ecological niches, including the human body. Invasive fungal infections have a devastating impact on human health worldwide, killing ∼1.5 million individuals annually. The majority of these deaths are attributable to species of Candida, Cryptococcus, and Aspergillus Treating fungal infections is challenging, in part due to the emergence of resistance to our limited arsenal of antifungal agents, necessitating the development of novel therapeutic options. Whereas conventional antifungal strategies target proteins or cellular components essential for fungal growth, an attractive alternative strategy involves targeting proteins that regulate fungal virulence or antifungal drug resistance, such as regulators of fungal stress responses. Stress response networks enable fungi to adapt, grow, and cause disease in humans and include regulators that are highly conserved across eukaryotes as well as those that are fungal-specific. This review highlights recent developments in elucidating crystal structures of fungal stress response regulators and emphasizes how this knowledge can guide the design of fungal-selective inhibitors. We focus on the progress that has been made with highly conserved regulators, including the molecular chaperone Hsp90, the protein phosphatase calcineurin, and the small GTPase Ras1, as well as with divergent stress response regulators, including the cell wall kinase Yck2 and trehalose synthases. Exploring structures of these important fungal stress regulators will accelerate the design of selective antifungals that can be deployed to combat life-threatening fungal diseases.
Collapse
Affiliation(s)
- Emmanuelle V LeBlanc
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Elizabeth J Polvi
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Amanda O Veri
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Gilbert G Privé
- Departments of Medical Biophysics and Biochemistry, University of Toronto, Toronto, Ontario, Canada.,Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
34
|
Liu ZQ. Bridging free radical chemistry with drug discovery: A promising way for finding novel drugs efficiently. Eur J Med Chem 2019; 189:112020. [PMID: 32006794 DOI: 10.1016/j.ejmech.2019.112020] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 12/06/2019] [Accepted: 12/27/2019] [Indexed: 02/07/2023]
Abstract
Many diseases have been regarded to correlate with the in vivo oxidative damages, which are caused by overproduced free radicals from metabolic process or reactive oxygen species (ROS). This background motivates chemists to explore free radical reactions and to design a number of antioxidants, but whether free radical chemistry can be applied to accelerate the efficacy of the drug discovery is still underrepresented. Herein, in light of recent findings as well as kinetics on free radical reaction, the discipline of free radical chemistry is introduced to be a novel tool for finding potential drugs from antioxidant libraries accumulated during the study on free radical chemistry. These antioxidants provide with such abundant types of structural skeleton that might be employed to inhibit oxidations in different biological microenvironments. Although the in vitro characterization on the antioxidative property exerts a potential role of an antioxidant as a prodrug, the in vivo investigation on the property for quenching free radicals will make a final decision for the antioxidant whether it is worthy to be further explored pharmacologically. Therefore, it is reasonable to expect that bridging free radical chemistry with the pharmacological research will provide with a succinct way for finding novel drugs efficiently.
Collapse
Affiliation(s)
- Zai-Qun Liu
- Department of Organic Chemistry, College of Chemistry, Jilin University, No.2519 Jiefang Road, Changchun, 130021, People's Republic of China.
| |
Collapse
|
35
|
Selection of potential anti-adhesion drugs by in silico approaches targeted to ALS3 from Candida albicans. Biotechnol Lett 2019; 41:1391-1401. [DOI: 10.1007/s10529-019-02747-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 10/18/2019] [Indexed: 12/25/2022]
|