1
|
Lemos ASDO, Granato JDT, Antinarelli LMR, Machado PDA, Campos LM, Bastos JPRC, Midlej VDV, Silva Neto AFD, Fabri RL, Coimbra ES. Lantana camara L. induces a multi-targeted cell death process in Leishmania amazonensis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118766. [PMID: 39222759 DOI: 10.1016/j.jep.2024.118766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/13/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
ETNOPHARMACOLOGICAL RELEVANCE Lantana camara L. is a species known for its broad spectrum of bioactivities and is commonly used in folk therapy to address inflammatory, dermatological, gastrointestinal, intestinal worms and protozoan diseases. It boasts a diverse array of secondary metabolites such as terpenes, flavonoids, and saponins. However, despite its rich chemical profile, there remains a scarcity of studies investigating its antileishmanial properties. AIM OF THE STUDY This research aims to explore the antileishmanial potential of L. camara, focusing also on its mechanism of action against Leishmania amazonensis. MATERIAL AND METHODS The ethanolic extract of L. camara leaves (LCE) was obtained through static maceration, and its phytoconstituents were identified using UFLC-QTOF-MS. The colorimetric MTT method was conducted to determine the effect of LCE on promastigotes of L. amazonensis and murine macrophages. The anti-amastigote activity was evaluated by counting intracellular parasites in macrophages after Giemsa staining. Additionally, investigations into the mechanisms underlying its action were conducted using cellular and biochemical approaches. RESULTS LCE exhibited significant activity against both promastigotes and intracellular amastigotes of L. amazonensis, with IC50 values of 12.20 μg/mL ± 0.12 and 7.09 μg/mL ± 1.24, respectively. These IC50 values indicate very promising antileishmanial activity, comparable to those found for the positive control miltefosine (5.10 μg/mL ± 1.79 and 8.96 μg/mL ± 0.50, respectively). Notably, LCE exhibited negligible cytotoxicity on macrophages (IC50 = 223.40 μg/mL ± 47.02), demonstrating selectivity towards host cells (SI = 31.50). The antileishmanial activity of LCE involved a multi-targeted cell death process, characterized by morphological and ultrastructural alterations observed through SEM and TEM analyses, as well as oxidative effects evidenced by the inhibition of trypanothione reductase, elevation of ROS and lipid levels, and mitochondrial dysfunction evaluated using DTNB, H2DCFDA, Nile red, and JC-1 assays. Additionally, extraction of ergosterol and double labeling with annexin V and PI revealed modifications to the organization and permeability of the treated parasite's plasma membrane. LCE was found to consist predominantly of terpenes, with lantadenes A, B, and C being among the eleven compounds identified through UFLC-QTOF-MS analysis. CONCLUSIONS The extract of L. camara presents a diverse array of chemical constituents, prominently featuring high terpene content, which may underlie its antileishmanial properties through a combination of apoptotic and non-apoptotic mechanisms of cell death induced by LCE. This study underscores the therapeutic potential of L. camara as a candidate for antileishmanial treatment, pending further validation.
Collapse
Affiliation(s)
- Ari Sérgio de Oliveira Lemos
- Department of Parasitology, Microbiology and Immunology, Federal University of Juiz de Fora, Juiz de Fora, MG, 36036-900, Brazil
| | - Juliana da Trindade Granato
- Department of Parasitology, Microbiology and Immunology, Federal University of Juiz de Fora, Juiz de Fora, MG, 36036-900, Brazil
| | | | - Patrícia de Almeida Machado
- Department of Parasitology, Microbiology and Immunology, Federal University of Juiz de Fora, Juiz de Fora, MG, 36036-900, Brazil
| | - Lara Melo Campos
- Bioactive Natural Products Laboratory, Department of Biochemistry, Biological Sciences Institute, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, CEP 36036-900, Brazil
| | - João Pedro Reis Costa Bastos
- Department of Parasitology, Microbiology and Immunology, Federal University of Juiz de Fora, Juiz de Fora, MG, 36036-900, Brazil
| | - Victor do Valle Midlej
- Laboratory of Cellular and Ultrastructure, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, 21040-900, Brazil
| | - Adolfo Firmino da Silva Neto
- Department of Veterinary Medicine, Faculty of Medicine, Federal University of Juiz de Fora, Juiz de Fora, MG, 36036-900, Brazil
| | - Rodrigo Luiz Fabri
- Laboratory of Bioactive Natural Products, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, CEP 36036-900, Brazil
| | - Elaine Soares Coimbra
- Department of Parasitology, Microbiology and Immunology, Federal University of Juiz de Fora, Juiz de Fora, MG, 36036-900, Brazil.
| |
Collapse
|
2
|
Goyzueta-Mamani LD, Pagliara Lage D, Barazorda-Ccahuana HL, Paco-Chipana M, Candia-Puma MA, Davila-Del-Carpio G, Galdino AS, Machado-de-Avila RA, Cordeiro Giunchetti R, D’Antonio EL, Ferraz Coelho EA, Chávez-Fumagalli MA. Exploring the Potential of Malvidin and Echiodinin as Probable Antileishmanial Agents Through In Silico Analysis and In Vitro Efficacy. Molecules 2025; 30:173. [PMID: 39795229 PMCID: PMC11722285 DOI: 10.3390/molecules30010173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Leishmaniasis, a neglected tropical disease caused by Leishmania species, presents serious public health challenges due to limited treatment options, toxicity, high costs, and drug resistance. In this study, the in vitro potential of malvidin and echioidinin is examined as antileishmanial agents against L. amazonensis, L. braziliensis, and L. infantum, comparing their effects to amphotericin B (AmpB), a standard drug. Malvidin demonstrated greater potency than echioidinin across all parasite stages and species. Against L. amazonensis, malvidin's IC50 values were 197.71 ± 17.20 µM (stationary amastigotes) and 258.07 ± 17 µM (axenic amastigotes), compared to echioidinin's 272.99 ± 29.90 μM and 335.96 ± 19.35 μM. AmpB was more potent, with IC50 values of 0.06 ± 0.01 µM and 0.10 ± 0.03 µM. Malvidin exhibited lower cytotoxicity (CC50: 2920.31 ± 80.29 µM) than AmpB (1.06 ± 0.12 µM) and a favorable selectivity index. It reduced infection rates by 35.75% in L. amazonensis-infected macrophages. The in silico analysis revealed strong binding between malvidin and Leishmania arginase, with the residues HIS139 and PRO258 playing key roles. Gene expression analysis indicated malvidin's modulation of oxidative stress and DNA repair pathways, involving genes like GLO1 and APEX1. These findings suggest malvidin's potential as a safe, natural antileishmanial compound, warranting further in vivo studies to confirm its therapeutic efficacy and pharmacokinetics in animal models.
Collapse
Affiliation(s)
- Luis Daniel Goyzueta-Mamani
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Arequipa 04000, Peru; (L.D.G.-M.); (H.L.B.-C.); (M.P.-C.); (M.A.C.-P.)
| | - Daniela Pagliara Lage
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (D.P.L.); (E.A.F.C.)
| | - Haruna Luz Barazorda-Ccahuana
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Arequipa 04000, Peru; (L.D.G.-M.); (H.L.B.-C.); (M.P.-C.); (M.A.C.-P.)
| | - Margot Paco-Chipana
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Arequipa 04000, Peru; (L.D.G.-M.); (H.L.B.-C.); (M.P.-C.); (M.A.C.-P.)
| | - Mayron Antonio Candia-Puma
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Arequipa 04000, Peru; (L.D.G.-M.); (H.L.B.-C.); (M.P.-C.); (M.A.C.-P.)
- Facultad de Ciencias Farmacéuticas, Bioquímicas y Biotecnológicas, Universidad Católica de Santa María, Arequipa 04000, Peru;
| | - Gonzalo Davila-Del-Carpio
- Facultad de Ciencias Farmacéuticas, Bioquímicas y Biotecnológicas, Universidad Católica de Santa María, Arequipa 04000, Peru;
| | - Alexsandro Sobreira Galdino
- Laboratório de Biotecnologia de Microrganismos, Universidade Federal São João Del-Rei, Divinópolis 35501-296, Brazil;
- Instituto Nacional de Ciência e Tecnologia em Biotecnologia Industrial (INCT-BI), Distrito Federal, Brasilia 70070-010, Brazil
| | | | - Rodolfo Cordeiro Giunchetti
- Laboratório de Biologia das Interações Celulares, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil;
- Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais (INCT-DT), Salvador 40110-160, Brazil
| | - Edward L. D’Antonio
- Department of Natural Sciences, University of South Carolina Beaufort, 1 University Boulevard, Bluffton, SC 29909, USA;
| | - Eduardo Antonio Ferraz Coelho
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (D.P.L.); (E.A.F.C.)
| | - Miguel Angel Chávez-Fumagalli
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Arequipa 04000, Peru; (L.D.G.-M.); (H.L.B.-C.); (M.P.-C.); (M.A.C.-P.)
| |
Collapse
|
3
|
Smith MZ, York M, Townsend KS, Martin LM, Gull T, Coghill LM, Ericsson AC, Johnson PJ. Effects of orally administered clioquinol on the fecal microbiome of horses. J Vet Intern Med 2025; 39:e17276. [PMID: 39709594 PMCID: PMC11663420 DOI: 10.1111/jvim.17276] [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: 08/13/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Whereas restoration of fecal consistency after treatment with clioquinol for chronic diarrhea and free fecal water syndrome has been attributed to its antiprotozoal properties, actions of clioquinol on the colonic bacterial microbiota have not been investigated. OBJECTIVES Characterize the dynamics of fecal microbial diversity before, during, and after PO administration of clioquinol to healthy horses. STUDY DESIGN Experimental prospective cohort study using a single horse group. METHODS Eight healthy adult horses received PO clioquinol (10 g, daily) for 7 days. Feces were obtained daily for 7 days before, during, and after conclusion of treatment, and again 3 months later. Libraries of 16S rRNA V4 region amplicons generated from fecal DNA were sequenced using the Illumina sequencing platform. Bioinformatic analysis was undertaken with QIIME2 and statistical analyses included analysis of variance (ANOVA) and permutational multivariate ANOVA (PERMANOVA). RESULTS The richness and composition of the fecal microbiome was altered after administration of clioquinol, reaching a maximum effect by the fifth day of administration. Changes included a 90% decrease in richness, and compensatory expansion of facultative anaerobes including Streptococcaceae, Enterococcaceae, and Enterobacteriaceae. Multiple horses had Salmonella cultured from feces. MAIN LIMITATIONS Limitations including lack of control group and modest sample size are obviated by robust longitudinal study design and strong effect size associated with drug exposure. CONCLUSIONS Clioquinol has broad-spectrum antibacterial effects on the fecal microbiome of horses, but spares certain bacterial families including several pathogens and pathobionts. Clioquinol should be used with caution in horses, in an environment free of contamination with fecal pathogens.
Collapse
Affiliation(s)
- Mikaila Z. Smith
- Veterinary Research Scholars Program (VRSP)University of Missouri College of Veterinary MedicineColumbia, Missouri 65211USA
| | - Mary York
- University of Missouri (MU) Bioinformatics and Analytics CoreBond Life Sciences CenterColumbia, Missouri 65201USA
- Institute for Data Science and InformaticsUniversity of MissouriColumbia, Missouri 65211USA
| | - Kile S. Townsend
- Veterinary Research Scholars Program (VRSP)University of Missouri College of Veterinary MedicineColumbia, Missouri 65211USA
- Department of Veterinary Medicine and Surgery, College of Veterinary MedicineUniversity of MissouriColumbia, Missouri 65211USA
| | - Lynn M. Martin
- Veterinary Research Scholars Program (VRSP)University of Missouri College of Veterinary MedicineColumbia, Missouri 65211USA
- Department of Veterinary Medicine and Surgery, College of Veterinary MedicineUniversity of MissouriColumbia, Missouri 65211USA
| | - Tamara Gull
- MU Veterinary Medical Diagnostic Laboratory (VMDL), College of Veterinary MedicineUniversity of MissouriColumbia, Missouri 65211USA
| | - Lyndon M. Coghill
- University of Missouri (MU) Bioinformatics and Analytics CoreBond Life Sciences CenterColumbia, Missouri 65201USA
- Department of Veterinary Pathobiology, College of Veterinary MedicineUniversity of MissouriColumbia, Missouri 65201USA
| | - Aaron C. Ericsson
- Veterinary Research Scholars Program (VRSP)University of Missouri College of Veterinary MedicineColumbia, Missouri 65211USA
- Department of Veterinary Pathobiology, College of Veterinary MedicineUniversity of MissouriColumbia, Missouri 65201USA
- University of Missouri Metagenomics CenterColumbia, Missouri 65201USA
| | - Philip J. Johnson
- Veterinary Research Scholars Program (VRSP)University of Missouri College of Veterinary MedicineColumbia, Missouri 65211USA
- Department of Veterinary Medicine and Surgery, College of Veterinary MedicineUniversity of MissouriColumbia, Missouri 65211USA
| |
Collapse
|
4
|
Granato JDT, Silva ETD, Lemos ASDO, Machado PDA, Midlej VDV, Antinarelli LMR, Silva Neto AFD, Souza MVN, Coimbra ES. 4-Quinolinylhydrazone analogues kill Leishmania (Leishmania) amazonensis by inducing apoptosis and mitochondria-dependent pathway cell death. Chem Biol Drug Des 2024; 103:e14535. [PMID: 38772877 DOI: 10.1111/cbdd.14535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/03/2024] [Accepted: 04/23/2024] [Indexed: 05/23/2024]
Abstract
Despite efforts, available alternatives for the treatment of leishmaniasis are still scarce. In this work we tested a class of 15 quinolinylhydrazone analogues and presented data that support the use of the most active compound in cutaneous leishmaniasis caused by Leishmania amazonensis. In general, the compounds showed activity at low concentrations for both parasitic forms (5.33-37.04 μM to promastigotes, and 14.31-61.98 μM to amastigotes). In addition, the best compound (MHZ15) is highly selective for the parasite. Biochemical studies indicate that the treatment of promastigotes with MHZ15 leads the loss of mitochondrial potential and increase in ROS levels as the primary effects, which triggers accumulation of lipid droplets, loss of plasma membrane integrity and apoptosis hallmarks, including DNA fragmentation and phosphatidylserine exposure. These effects were similar in the intracellular form of the parasite. However, in this parasitic form there is no change in plasma membrane integrity in the observed treatment time, which can be attributed to metabolic differences and the resilience of the amastigote. Also, ultrastructural changes such as vacuolization suggesting autophagy were observed. The in vivo effectiveness of MHZ15 in the experimental model of cutaneous leishmaniasis was carried out in mice of the BALB/c strain infected with L. amazonensis. The treatment by intralesional route showed that MHZ15 acted with great efficiency with significantly reduction in the parasite load in the injured paws and draining lymph nodes, without clinical signs of distress or compromise of animal welfare. In vivo toxicity was also evaluated and null alterations in the levels of hepatic enzymes aspartate aminotransferase, and alanine aminotransferase was observed. The data presented herein demonstrates that MHZ15 exhibits a range of favorable characteristics conducive to the development of an antileishmanial agent.
Collapse
Affiliation(s)
- Juliana da Trindade Granato
- Departamento de Parasitologia, Microbiologia e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Emerson Teixeira da Silva
- Fundação Oswaldo Cruz (Fiocruz), Instituto de Tecnologia em Fármacos Farmanguinhos, Rio de Janeiro, Brazil
| | - Ari Sérgio de Oliveira Lemos
- Departamento de Parasitologia, Microbiologia e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Patrícia de Almeida Machado
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | - Victor do Valle Midlej
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | - Luciana Maria Ribeiro Antinarelli
- Departamento de Parasitologia, Microbiologia e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Adolfo Firmino da Silva Neto
- Departamento de Medicina Veterinária, Faculdade de Medicina, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Marcus Vinícius Nora Souza
- Fundação Oswaldo Cruz (Fiocruz), Instituto de Tecnologia em Fármacos Farmanguinhos, Rio de Janeiro, Brazil
| | - Elaine Soares Coimbra
- Departamento de Parasitologia, Microbiologia e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| |
Collapse
|
5
|
Jing S, Zhang Q, Li Y, Chang H, Xiang C, Han S, Yuan G, Fan J, He H. Identification of new drug candidates against Trichomonas gallinae using high-throughput screening. Int J Parasitol Drugs Drug Resist 2023; 23:19-27. [PMID: 37562241 PMCID: PMC10424085 DOI: 10.1016/j.ijpddr.2023.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 08/01/2023] [Accepted: 08/01/2023] [Indexed: 08/12/2023]
Abstract
Trichomonas gallinae is a protozoan parasite that is the causative agent of trichomoniasis, and infects captive and wild bird species throughout the world. Although metronidazole has been the drug of choice against trichomoniasis for decades, most Trichomonas gallinae strains have developed resistance. Therefore, drugs with new modes of action or targets are urgently needed. Here, we report the development and application of a cell-based CCK-8 method for the high-throughput screening and identification of new inhibitors of Trichomonas gallinae as a beginning point for the development of new treatments for trichomoniasis. We performed the high-throughput screening of 173 anti-parasitic compounds, and found 16 compounds that were potentially effective against Trichomonas gallinae. By measuring the median inhibitory concentration (IC50) and median cytotoxic concentration (CC50), we identified 3 potentially safe and effective compounds against Trichomonas gallinae: anisomycin, fumagillin, and MG132. In conclusion, this research successfully established a high-throughput screening method for compounds and identified 3 new safe and effective compounds against Trichomonas gallinae, providing a new treatment scheme for trichomoniasis.
Collapse
Affiliation(s)
- Shengfan Jing
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, 071000, China; National Research Center for Wildlife Borne Diseases, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China
| | - Qingxun Zhang
- Beijing Milu Ecological Research Center, Beijing, 100076, China
| | - Yi Li
- National Research Center for Wildlife Borne Diseases, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China
| | - Han Chang
- National Research Center for Wildlife Borne Diseases, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China
| | - Chen Xiang
- National Research Center for Wildlife Borne Diseases, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China
| | - Shuyi Han
- National Research Center for Wildlife Borne Diseases, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China
| | - Guohui Yuan
- National Research Center for Wildlife Borne Diseases, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China
| | - Jinghui Fan
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, 071000, China.
| | - Hongxuan He
- National Research Center for Wildlife Borne Diseases, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China.
| |
Collapse
|
6
|
Sousa JPAD, Sousa JMSD, Rodrigues RRL, Nunes TADL, Machado YAA, Araujo ACD, da Silva IGM, Barros-Cordeiro KB, Báo SN, Alves MMDM, Mendonça-Junior FJB, Rodrigues KADF. Antileishmanial activity of 2-amino-thiophene derivative SB-200. Int Immunopharmacol 2023; 123:110750. [PMID: 37536181 DOI: 10.1016/j.intimp.2023.110750] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/28/2023] [Accepted: 07/30/2023] [Indexed: 08/05/2023]
Abstract
Leishmaniasis, presenting the highest number of cases worldwide is one of the most serious Neglected Tropical Diseases (NTDs). Clinical manifestations are intrinsically related to the host's immune response making immunomodulatory substances the target of numerous studies on antileishmanial activity. The currently available drugs used for treatment present various problems including high toxicity, low efficacy, and associated drug resistance. The search for therapeutic alternatives is urgent, and in this context, thiophene derivatives appear to be a promising therapeutic alternative (many have shown promising anti-leishmanial activity). The objective of this study was to investigate the antileishmanial activity of the 2-amino-thiophenic derivative SB-200. The thiophenic derivative was effective in inhibiting the growth of Leishmania braziliensis, Leishmania major, and Leishmania infantum promastigotes, obtaining respective IC50 values of 4.25 μM, 4.65 μM, and 3.96 μM. For L. infantum, it was demonstrated that the antipromastigote effect of SB-200 is associated with cell membrane integrity losses, and with morphological changes observed during scanning and transmission electron microscopy. Cytotoxicity was performed for J774.A1 macrophages and VERO cells, to obtain a CC50 of 42.52 μM and a SI of 10.74 for macrophages and a CC50 of 39.2 μM and an SI of 9.89 for VERO cells. The anti-amastigote activity of SB-200 revealed an IC50 of 2.85 μM and an SI of 14.97 against macrophages and SI of 13.8 for VERO cells. The anti-amastigote activity of SB-200 is associated with in vitro immunomodulation. For acute toxicity, SB-200 against Zophobas morio larvae permitted 100% survival. We conclude that the 2-amino-thiophenic derivative SB-200 is a promising candidate for in vivo anti-leishmania drug tests to evaluate its activity, efficacy, and safety.
Collapse
Affiliation(s)
- João Paulo Araujo de Sousa
- Infectious Disease Laboratory, Campus Ministro Reis Velloso, Federal University of Parnaíba Delta, 64202-020 Parnaíba, PI, Brasil
| | - Julyanne Maria Saraiva de Sousa
- Infectious Disease Laboratory, Campus Ministro Reis Velloso, Federal University of Parnaíba Delta, 64202-020 Parnaíba, PI, Brasil
| | - Raiza Raianne Luz Rodrigues
- Infectious Disease Laboratory, Campus Ministro Reis Velloso, Federal University of Parnaíba Delta, 64202-020 Parnaíba, PI, Brasil
| | - Thais Amanda de Lima Nunes
- Infectious Disease Laboratory, Campus Ministro Reis Velloso, Federal University of Parnaíba Delta, 64202-020 Parnaíba, PI, Brasil
| | - Yasmim Alves Aires Machado
- Infectious Disease Laboratory, Campus Ministro Reis Velloso, Federal University of Parnaíba Delta, 64202-020 Parnaíba, PI, Brasil
| | - Alexandre Carvalho de Araujo
- Infectious Disease Laboratory, Campus Ministro Reis Velloso, Federal University of Parnaíba Delta, 64202-020 Parnaíba, PI, Brasil
| | - Ingrid Gracielle Martins da Silva
- Microscopy and Microanalysis Laboratory, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Karine Brenda Barros-Cordeiro
- Microscopy and Microanalysis Laboratory, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Sônia Nair Báo
- Microscopy and Microanalysis Laboratory, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Michel Muálem de Moraes Alves
- Laboratory of Antileishmania Activity, Medicinal Plants Research Center, Federal University of Piauí, Teresina 64049-550, Brazil
| | | | | |
Collapse
|
7
|
Lima SKSD, Jesus JA, Raminelli C, Laurenti MD, Passero LFD. High Selectivity of 8-Hydroxyquinoline on Leishmania (Leishmania) and Leishmania (Viannia) Species Correlates with a Potent Therapeutic Activity In Vivo. Pharmaceuticals (Basel) 2023; 16:ph16050707. [PMID: 37242490 DOI: 10.3390/ph16050707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Leishmaniasis is a neglected disease caused by protozoa of the genus Leishmania, which causes different clinical manifestations. Drugs currently used in the treatment such as pentavalent antimonial and amphotericin B cause severe side effects in patients, and parasite resistance has been reported. Thus, it is necessary and urgent to characterize new and effective alternative drugs to replace the current chemotherapy of leishmaniasis. In this regard, it has been experimentally demonstrated that quinoline derivatives present significative pharmacological and parasitic properties. Thus, the aim of this work was to demonstrate the leishmanicidal activity of 8-hydroxyquinoline (8-HQ) in vitro and in vivo. The leishmanicidal activity (in vitro) of 8-HQ was assayed on promastigote and intracellular amastigote forms of L. (L.) amazonensis, L. (L.) infantum chagasi, L. (V.) guyanensis L. (V.) naiffi, L. (V.) lainsoni, and L. (V.) shawi. Additionally, the levels of nitric oxide and hydrogen peroxide were analyzed. The therapeutic potential of 8-HQ was analyzed in BALB/c mice infected with a strain of L. (L.) amazonensis that causes anergic cutaneous diffuse leishmaniasis. In vitro data showed that at 24 and 72 h, 8-HQ eliminated promastigote and intracellular amastigote forms of all studied species and this effect may be potentialized by nitric oxide. Furthermore, 8-HQ was more selective than miltefosine. Infected animals treated with 8-HQ by the intralesional route dramatically reduced the number of tissue parasites in the skin, and it was associated with an increase in IFN-γ and decrease in IL-4, which correlated with a reduction in inflammatory reaction in the skin. These results strongly support the idea that 8-HQ is an alternative molecule that can be employed in the treatment of leishmaniasis, given its selectivity and multispectral action in parasites from the Leishmania genus.
Collapse
Affiliation(s)
- Sarah Kymberly Santos de Lima
- Institute of Biosciences, São Paulo State University (UNESP), Praça Infante Dom Henrique, s/n, São Vicente 11330-900, Brazil
- Laboratory of Pathology of Infectious Diseases (LIM50), Department of Pathology, Medical School of São Paulo University, São Paulo 01246-903, Brazil
| | - Jéssica Adriana Jesus
- Institute of Biosciences, São Paulo State University (UNESP), Praça Infante Dom Henrique, s/n, São Vicente 11330-900, Brazil
| | - Cristiano Raminelli
- Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema 09920-000, Brazil
| | - Márcia Dalastra Laurenti
- Laboratory of Pathology of Infectious Diseases (LIM50), Department of Pathology, Medical School of São Paulo University, São Paulo 01246-903, Brazil
| | - Luiz Felipe Domingues Passero
- Institute of Biosciences, São Paulo State University (UNESP), Praça Infante Dom Henrique, s/n, São Vicente 11330-900, Brazil
- Institute for Advanced Studies of Ocean, São Paulo State University (UNESP), Rua João Francisco Bensdorp, 1178, São Vicente 11350-011, Brazil
| |
Collapse
|
8
|
Tuon FF, Dantas LR, de Souza RM, Ribeiro VST, Amato VS. Liposomal drug delivery systems for the treatment of leishmaniasis. Parasitol Res 2022; 121:3073-3082. [DOI: 10.1007/s00436-022-07659-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 09/06/2022] [Indexed: 10/14/2022]
|
9
|
Wykowski R, Fuentefria AM, de Andrade SF. Antimicrobial activity of clioquinol and nitroxoline: a scoping review. Arch Microbiol 2022; 204:535. [PMID: 35907036 PMCID: PMC9362210 DOI: 10.1007/s00203-022-03122-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/04/2022] [Indexed: 11/29/2022]
Abstract
Clioquinol and nitroxoline, two drugs with numerous pharmacological properties fallen into disuse for many decades. The first was considered dangerous due to contraindications and the second mainly because was taken as ineffective, despite its known antibacterial activity. In the last decades, the advances in pharmaceutical chemistry, molecular biology, toxicology and genetics allowed to better understand the cellular action of these compounds, some toxicological issues and/or activity scopes. Thus, a new opportunity for these drugs to be considered as potential antimicrobial agents has arisen. This review contemplates the trajectory of clioquinol and nitroxoline from their emergence to the present day, emphasizing the new studies that indicate the possibility of reintroduction for specific cases.
Collapse
Affiliation(s)
- Rachel Wykowski
- Programa de Pós-Graduação Em Microbiologia Agrícola E Do Ambiente, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Alexandre Meneghello Fuentefria
- Programa de Pós-Graduação Em Microbiologia Agrícola E Do Ambiente, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
- Departamento de Análises, Faculdade de Farmácia, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Saulo Fernandes de Andrade
- Programa de Pós-Graduação Em Microbiologia Agrícola E Do Ambiente, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil.
- Departamento de Produção de Matéria-Prima, Faculdade de Farmácia, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
10
|
Bustamante C, Díez-Mejía AF, Arbeláez N, Soares MJ, Robledo SM, Ochoa R, Varela-M. RE, Marín-Villa M. In Silico, In Vitro, and Pharmacokinetic Studies of UBMC-4, a Potential Novel Compound for Treating against Trypanosoma cruzi. Pathogens 2022; 11:pathogens11060616. [PMID: 35745470 PMCID: PMC9229894 DOI: 10.3390/pathogens11060616] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/18/2022] [Accepted: 05/21/2022] [Indexed: 12/10/2022] Open
Abstract
The lack of therapeutic alternatives for the treatment of Chagas disease, a neglected disease, drives the discovery of new drugs with trypanocidal activity. Consequently, we conducted in vitro studies using UBMC-4, a potential Trypanosoma cruzi AKT-like pleckstrin homology (PH) domain inhibitory compound found using bioinformatics tools. The half effective concentration (EC50) on intracellular amastigotes was determined at 1.85 ± 1 μM showing low cytotoxicity (LC50) > 40 μM on human cell lines tested. In order to study the lethal effect caused by the compound on epimastigotes, morphological changes were assessed by scanning and transmission electron microscopy. Progressive alterations such as flagellum inactivation, cell size reduction, nuclear structure alteration, condensation of chromatin towards the nuclear periphery, vacuole formation, and mitochondrial swelling with kinetoplast integrity loss were evidenced. In addition, apoptosis-like markers in T. cruzi were assessed by flow cytometry, demonstrating that the effect of UBMC-4 on T. cruzi AKT-like kinase reduced the tolerance to nutritional stress-triggered, apoptosis-like events, including DNA fragmentation, mitochondrial damage, and loss of plasma membrane integrity. After this, UBMC-4 was formulated for oral administration and pharmacokinetics were analyzed in a mouse model. Finally, upon oral administration of 200 mg/kg in mice, we found that a UBMC-4 plasma concentration remaining in circulation beyond 24 h after administration is well described by the two-compartment model. We conclude that UBMC-4 has an effective trypanocidal activity in vitro at low concentrations and this effect is evident in T. cruzi cell structures. In mice, UBMC-4 was well absorbed and reached plasma concentrations higher than the EC50, showing features that would aid in developing a new drug to treat Chagas disease.
Collapse
Affiliation(s)
- Christian Bustamante
- PECET-Programa de Estudio y Control de Enfermedades Tropicales, School of Medicine, Universidad de Antioquia, Medellín 050010, Colombia; (A.F.D.-M.); (N.A.); (S.M.R.)
- Correspondence: (C.B.); (M.M.-V.)
| | - Andrés Felipe Díez-Mejía
- PECET-Programa de Estudio y Control de Enfermedades Tropicales, School of Medicine, Universidad de Antioquia, Medellín 050010, Colombia; (A.F.D.-M.); (N.A.); (S.M.R.)
| | - Natalia Arbeláez
- PECET-Programa de Estudio y Control de Enfermedades Tropicales, School of Medicine, Universidad de Antioquia, Medellín 050010, Colombia; (A.F.D.-M.); (N.A.); (S.M.R.)
| | - Maurilio José Soares
- Cell Biology Laboratory, Carlos Chagas Institute/Fiocruz, Curitiba 81350-010, Paraná, Brazil;
| | - Sara M. Robledo
- PECET-Programa de Estudio y Control de Enfermedades Tropicales, School of Medicine, Universidad de Antioquia, Medellín 050010, Colombia; (A.F.D.-M.); (N.A.); (S.M.R.)
| | - Rodrigo Ochoa
- Biophysics of Tropical Diseases, Max Planck Tandem Group, Universidad de Antioquia, Medellín 050010, Colombia;
| | - Rubén E. Varela-M.
- Grupo (QUIBIO), School of Basic Sciences, Universidad Santiago de Cali, Cali 760032, Colombia;
| | - Marcel Marín-Villa
- PECET-Programa de Estudio y Control de Enfermedades Tropicales, School of Medicine, Universidad de Antioquia, Medellín 050010, Colombia; (A.F.D.-M.); (N.A.); (S.M.R.)
- Correspondence: (C.B.); (M.M.-V.)
| |
Collapse
|
11
|
Al-Mamary NI, Al-Hayali HL. Effect of Synergism of Thalidomide and Liposomal Amphotericin-B on Leishmania tropica and Leishmania donovani Promastigote. BIONATURA 2022. [DOI: 10.21931/rb/2022.07.02.59] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
This study aims to find safe and effective anti-leishmaniasis drugs; thus, the synergism between thalidomide and liposomal amphotericin-B was tested as antileishmanial on L. tropica and L. donovani promastigote in vitro. IC50, IC90 were determined at the Log phase of thalidomide and were (10), (25) µg/ml for L. tropica and (12.5), (30( µg/ml for L. donovani, Moreover IC50, IC90 were determined at Log phase of Liposomal amphotericin-B and were (5), (20) µg/ml for L. tropica and (5), (25) µg/ml for L. donovani. Additionally, synergistic effect IC50 of the two drugs were determined when Liposomal amphotericin-B fixed it, and thalidomide concentrations changed was (2.5+0.5) µg/ml on L. tropica and (2.5+1) µg/ml on L. donovan. When thalidomide was fixed, and Liposomal amphotericin-B was changed, it was (2.5+2) µg/ml for both L. tropica and L.donovani. The synergistic effect on the morphology of both promastigotes forms was observed.
Keywords. Leishmaniasis, thalidomide, liposomal amphotericin-B, synergistic.
Collapse
Affiliation(s)
- Nasma I. Al-Mamary
- Ministry of Health/ Nineveh Health Department /Makhmour Health Sector. Iraq
| | | |
Collapse
|
12
|
The antimicrobial and immunomodulatory effects of Ionophores for the treatment of human infection. J Inorg Biochem 2021; 227:111661. [PMID: 34896767 DOI: 10.1016/j.jinorgbio.2021.111661] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 10/27/2021] [Accepted: 11/03/2021] [Indexed: 12/18/2022]
Abstract
Ionophores are a diverse class of synthetic and naturally occurring ion transporter compounds which demonstrate both direct and in-direct antimicrobial properties against a broad panel of bacterial, fungal, viral and parasitic pathogens. In addition, ionophores can regulate the host-immune response during communicable and non-communicable disease states. Although the clinical use of ionophores such as Amphotericin B, Bedaquiline and Ivermectin highlight the utility of ionophores in modern medicine, for many other ionophore compounds issues surrounding toxicity, bioavailability or lack of in vivo efficacy studies have hindered clinical development. The antimicrobial and immunomodulating properties of a range of compounds with characteristics of ionophores remain largely unexplored. As such, ionophores remain a latent therapeutic avenue to address both the global burden of antimicrobial resistance, and the unmet clinical need for new antimicrobial therapies. This review will provide an overview of the broad-spectrum antimicrobial and immunomodulatory properties of ionophores, and their potential uses in clinical medicine for combatting infection.
Collapse
|
13
|
Acarbose presents in vitro and in vivo antileishmanial activity against Leishmania infantum and is a promising therapeutic candidate against visceral leishmaniasis. Med Microbiol Immunol 2021; 210:133-147. [PMID: 33870453 PMCID: PMC8053370 DOI: 10.1007/s00430-021-00707-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 04/03/2021] [Indexed: 12/01/2022]
Abstract
Treatment against visceral leishmaniasis (VL) is mainly hampered by drug toxicity, long treatment regimens and/or high costs. Thus, the identification of novel and low-cost antileishmanial agents is urgent. Acarbose (ACA) is a specific inhibitor of glucosidase-like proteins, which has been used for treating diabetes. In the present study, we show that this molecule also presents in vitro and in vivo specific antileishmanial activity against Leishmania infantum. Results showed an in vitro direct action against L. infantum promastigotes and amastigotes, and low toxicity to mammalian cells. In addition, in vivo experiments performed using free ACA or incorporated in a Pluronic® F127-based polymeric micelle system called ACA/Mic proved effective for the treatment of L. infantum-infected BALB/c mice. Treated animals presented significant reductions in the parasite load in their spleens, livers, bone marrows and draining lymph nodes when compared to the controls, as well as the development of antileishmanial Th1-type humoral and cellular responses based on high levels of IFN-γ, IL-12, TNF-α, GM-CSF, nitrite and IgG2a isotype antibodies. In addition, ACA or ACA-treated animals suffered from low organ toxicity. Treatment with ACA/Mic outperformed treatments using either Miltefosine or free ACA based on parasitological and immunological evaluations performed one and 15 days post-therapy. In conclusion, data suggest that the ACA/Mic is a potential therapeutic agent against L. infantum and merits further consideration for VL treatment.
Collapse
|
14
|
Freitas CS, Lage DP, Oliveira-da-Silva JA, Costa RR, Mendonça DVC, Martins VT, Reis TAR, Antinarelli LMR, Machado AS, Tavares GSV, Ramos FF, Brito RCF, Ludolf F, Chávez-Fumagalli MA, Roatt BM, Ramos GS, Munkert J, Ottoni FM, Campana PRV, Duarte MC, Gonçalves DU, Coimbra ES, Braga FC, Pádua RM, Coelho EAF. In vitro and in vivo antileishmanial activity of β-acetyl-digitoxin, a cardenolide of Digitalis lanata potentially useful to treat visceral leishmaniasis. ACTA ACUST UNITED AC 2021; 28:38. [PMID: 33851916 PMCID: PMC8045677 DOI: 10.1051/parasite/2021036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 04/01/2021] [Indexed: 12/11/2022]
Abstract
Current treatments of visceral leishmaniasis face limitations due to drug side effects and/or high cost, along with the emergence of parasite resistance. Novel and low-cost antileishmanial agents are therefore required. We report herein the antileishmanial activity of β-acetyl-digitoxin (b-AD), a cardenolide isolated from Digitalis lanata leaves, assayed in vitro and in vivo against Leishmania infantum. Results showed direct action of b-AD against parasites, as well as efficacy for the treatment of Leishmania-infected macrophages. In vivo experiments using b-AD-containing Pluronic® F127 polymeric micelles (b-AD/Mic) to treat L. infantum-infected mice showed that this composition reduced the parasite load in distinct organs in more significant levels. It also induced the development of anti-parasite Th1-type immunity, attested by high levels of IFN-γ, IL-12, TNF-α, GM-CSF, nitrite and specific IgG2a antibodies, in addition to low IL-4 and IL-10 contents, along with higher IFN-γ-producing CD4+ and CD8+ T-cell frequency. Furthermore, low toxicity was found in the organs of the treated animals. Comparing the therapeutic effect between the treatments, b-AD/Mic was the most effective in protecting animals against infection, when compared to the other groups including miltefosine used as a drug control. Data found 15 days after treatment were similar to those obtained one day post-therapy. In conclusion, the results obtained suggest that b-AD/Mic is a promising antileishmanial agent and deserves further studies to investigate its potential to treat visceral leishmaniasis.
Collapse
Affiliation(s)
- Camila S Freitas
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, Brazil
| | - Daniela P Lage
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, Brazil
| | - João A Oliveira-da-Silva
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, Brazil
| | - Rafaella R Costa
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, Brazil
| | - Débora V C Mendonça
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, Brazil
| | - Vívian T Martins
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, Brazil
| | - Thiago A R Reis
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, Brazil
| | - Luciana M R Antinarelli
- Departamento de Parasitologia, Microbiologia e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, 36036-900 Minas Gerais, Brazil
| | - Amanda S Machado
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, Brazil
| | - Grasiele S V Tavares
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, Brazil
| | - Fernanda F Ramos
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, Brazil
| | - Rory C F Brito
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas, Departamento de Ciências Biológicas, Insituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, 35400-000 Minas Gerais, Brazil
| | - Fernanda Ludolf
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, Brazil
| | | | - Bruno M Roatt
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas, Departamento de Ciências Biológicas, Insituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, 35400-000 Minas Gerais, Brazil
| | - Gabriela S Ramos
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901 Minas Gerais, Brazil
| | - Jennifer Munkert
- Departament Biologie, LS Pharmazeutische Biologie, Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Flaviano M Ottoni
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901 Minas Gerais, Brazil
| | - Priscilla R V Campana
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901 Minas Gerais, Brazil
| | - Mariana C Duarte
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, Brazil - Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901 Minas Gerais, Brazil
| | - Denise U Gonçalves
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, Brazil
| | - Elaine S Coimbra
- Departamento de Parasitologia, Microbiologia e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, 36036-900 Minas Gerais, Brazil
| | - Fernão C Braga
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901 Minas Gerais, Brazil
| | - Rodrigo M Pádua
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901 Minas Gerais, Brazil
| | - Eduardo A F Coelho
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, 30130-100 Minas Gerais, Brazil - Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901 Minas Gerais, Brazil
| |
Collapse
|
15
|
Olaleye OA, Kaur M, Onyenaka C, Adebusuyi T. Discovery of Clioquinol and analogues as novel inhibitors of Severe Acute Respiratory Syndrome Coronavirus 2 infection, ACE2 and ACE2 - Spike protein interaction in vitro. Heliyon 2021; 7:e06426. [PMID: 33732940 PMCID: PMC7951571 DOI: 10.1016/j.heliyon.2021.e06426] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 02/05/2021] [Accepted: 05/02/2021] [Indexed: 01/08/2023] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the etiological agent for coronavirus disease 2019 (COVID-19), has resulted in an ongoing pandemic. Presently, there are no clinically approved drugs for COVID-19. Hence, there is an urgent need to accelerate the development of effective antivirals. Herein, we discovered Clioquinol (5-chloro-7-iodo-8-quinolinol (CLQ)), a Food and Drug Administration (FDA) approved drug, and two of its analogues (7-bromo-5-chloro-8-hydroxyquinoline (CLBQ14); and 5, 7-Dichloro-8-hydroxyquinoline (CLCQ)) as potent inhibitors of SARS-CoV-2 infection-induced cytopathic effect in vitro. In addition, all three compounds showed potent anti-exopeptidase activity against recombinant human angiotensin-converting enzyme 2 (rhACE2) and inhibited the binding of rhACE2 with SARS-CoV-2 Spike (RBD) protein. CLQ displayed the highest potency in the low micromolar range, with its antiviral activity showing a strong correlation with inhibition of rhACE2 and rhACE2-RBD interaction. Altogether, our findings provide a new mode of action and molecular target for CLQ and validates this pharmacophore as a promising lead series for the clinical development of potential therapeutics for COVID-19.
Collapse
Affiliation(s)
- Omonike A. Olaleye
- Department of Pharmaceutical and Environmental Health Sciences, College of Pharmacy and Health Sciences, Texas Southern University, 3100 Cleburne St, Houston, TX 77004, USA
| | - Manvir Kaur
- Department of Pharmaceutical and Environmental Health Sciences, College of Pharmacy and Health Sciences, Texas Southern University, 3100 Cleburne St, Houston, TX 77004, USA
| | - Collins Onyenaka
- Department of Pharmaceutical and Environmental Health Sciences, College of Pharmacy and Health Sciences, Texas Southern University, 3100 Cleburne St, Houston, TX 77004, USA
| | - Tolulope Adebusuyi
- Department of Pharmaceutical and Environmental Health Sciences, College of Pharmacy and Health Sciences, Texas Southern University, 3100 Cleburne St, Houston, TX 77004, USA
| |
Collapse
|
16
|
Ivermectin presents effective and selective antileishmanial activity in vitro and in vivo against Leishmania infantum and is therapeutic against visceral leishmaniasis. Exp Parasitol 2020; 221:108059. [PMID: 33338468 DOI: 10.1016/j.exppara.2020.108059] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 12/03/2020] [Accepted: 12/13/2020] [Indexed: 12/19/2022]
Abstract
Treatment for visceral leishmaniasis (VL) is hindered mainly by the toxicity and/or high cost of therapeutic drugs. In addition, parasite resistance has been registered. Thus, there is an urgent need for the identification of novel, effective and low-cost antileishmanial agents. Since drug discovery is a long and expensive process, drug repositioning for treatment of leishmaniasis should be considered. In the present study, Ivermectin (IVE), a broad-spectrum drug used for treatment of parasitic diseases, was evaluated in vitro and in vivo against Leishmania infantum species. Results in vitro showed that IVE presented 50% Leishmania and macrophage inhibitory concentrations (IC50 and CC50, respectively) of 3.64 ± 0.48 μM and 427.50 ± 17.60 μM, respectively, with a selectivity index (SI) of 117.45; whereas Amphotericin B (AmpB), which was used as control, showed IC50 and CC50 values of 0.12 ± 0.05 μM and 1.06 ± 0.23 μM, respectively, with a corresponding SI of 8.90. Treatment with IVE effectively reduced the infection percentage and parasite burden in infected and treated macrophages and displayed a prophylactic activity by inhibiting macrophage infection with pre-treated parasites. Furthermore, preliminary studies suggested that IVE targets the parasite's mitochondria. Activity of IVE in its free format or incorporated into Pluronic® F127-based polymeric micelles (IVE/Mic) was also evaluated in vivo as a treating drug for L. infantum-infected BALB/c mice. Miltefosine was used as a control. Results showed that Miltefosine, IVE and IVE/Mic-treated animals presented significant reductions in the parasite load in their spleens, livers, bone marrows and draining lymph nodes, as well as development of an antileishmanial Th1-type immune response one and 15 days after treatment. Notably, IVE/Mic showed a better parasitological and immunological response in comparison to other alternative treatments. In conclusion, results suggest that IVE/Mic could be considered in future studies as a therapeutic alternative to treat VL.
Collapse
|
17
|
Digitoxigenin presents an effective and selective antileishmanial action against Leishmania infantum and is a potential therapeutic agent for visceral leishmaniasis. Parasitol Res 2020; 120:321-335. [PMID: 33191446 PMCID: PMC7667010 DOI: 10.1007/s00436-020-06971-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 11/05/2020] [Indexed: 12/11/2022]
Abstract
Treatment for visceral leishmaniasis (VL) is hampered mainly by drug toxicity, their high cost, and parasite resistance. Drug development is a long and pricey process, and therefore, drug repositioning may be an alternative worth pursuing. Cardenolides are used to treat cardiac diseases, especially those obtained from Digitalis species. In the present study, cardenolide digitoxigenin (DIGI) obtained from a methanolic extract of Digitalis lanata leaves was tested for its antileishmanial activity against Leishmania infantum species. Results showed that 50% Leishmania and murine macrophage inhibitory concentrations (IC50 and CC50, respectively) were of 6.9 ± 1.5 and 295.3 ± 14.5 μg/mL, respectively. With amphotericin B (AmpB) deoxycholate, used as a control drug, values of 0.13 ± 0.02 and 0.79 ± 0.12 μg/mL, respectively, were observed. Selectivity index (SI) values were of 42.8 and 6.1 for DIGI and AmpB, respectively. Preliminary studies suggested that the mechanism of action for DIGI is to cause alterations in the mitochondrial membrane potential, to increase the levels of reactive oxygen species and induce accumulation of lipid bodies in the parasites. DIGI was incorporated into Pluronic® F127-based polymeric micelles, and the formula (DIGI/Mic) was used to treat L. infantum–infected mice. Miltefosine was used as a control drug. Results showed that animals treated with either miltefosine, DIGI, or DIGI/Mic presented significant reductions in the parasite load in their spleens, livers, bone marrows, and draining lymph nodes, as well as the development of a specific Th1-type response, when compared with the controls. Results obtained 1 day after treatment were corroborated with data corresponding to 15 days after therapy. Importantly, treatment with DIGI/Mic induced better parasitological and immunological responses when compared with miltefosine- and DIGI-treated mice. In conclusion, DIGI/Mic has the potential to be used as a therapeutic agent to protect against L. infantum infection, and it is therefore worth of consideration in future studies addressing VL treatment.
Collapse
|
18
|
Multiple Drug-Induced Stress Responses Inhibit Formation of Escherichia coli Biofilms. Appl Environ Microbiol 2020; 86:AEM.01113-20. [PMID: 32826218 PMCID: PMC7580552 DOI: 10.1128/aem.01113-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/08/2020] [Indexed: 02/07/2023] Open
Abstract
The prevention of bacterial biofilm formation is one of the major current challenges in microbiology. Here, by systematically screening a large number of approved drugs for their ability to suppress biofilm formation by Escherichia coli, we identified a number of prospective antibiofilm compounds. We further demonstrated different mechanisms of action for individual compounds, from induction of replicative stress to disbalance of cation homeostasis to inhibition of bacterial attachment to the surface. Our work demonstrates the potential of drug repurposing for the prevention of bacterial biofilm formation and suggests that also for other bacteria, the activity spectrum of antibiofilm compounds is likely to be broad. In most ecosystems, bacteria exist primarily as structured surface-associated biofilms that can be highly tolerant to antibiotics and thus represent an important health issue. Here, we explored drug repurposing as a strategy to identify new antibiofilm compounds, screening over 1,000 compounds from the Prestwick Chemical Library of approved drugs for specific activities that prevent biofilm formation by Escherichia coli. Most growth-inhibiting compounds, which include known antibacterial but also antiviral and other drugs, also reduced biofilm formation. However, we also identified several drugs that were biofilm inhibitory at doses where only a weak effect or no effect on planktonic growth could be observed. The activities of the most specific antibiofilm compounds were further characterized using gene expression analysis, proteomics, and microscopy. We observed that most of these drugs acted by repressing genes responsible for the production of curli, a major component of the E. coli biofilm matrix. This repression apparently occurred through the induction of several different stress responses, including DNA and cell wall damage, and homeostasis of divalent cations, demonstrating that biofilm formation can be inhibited through a variety of molecular mechanisms. One tested drug, tyloxapol, did not affect curli expression or cell growth but instead inhibited biofilm formation by suppressing bacterial attachment to the surface. IMPORTANCE The prevention of bacterial biofilm formation is one of the major current challenges in microbiology. Here, by systematically screening a large number of approved drugs for their ability to suppress biofilm formation by Escherichia coli, we identified a number of prospective antibiofilm compounds. We further demonstrated different mechanisms of action for individual compounds, from induction of replicative stress to disbalance of cation homeostasis to inhibition of bacterial attachment to the surface. Our work demonstrates the potential of drug repurposing for the prevention of bacterial biofilm formation and suggests that also for other bacteria, the activity spectrum of antibiofilm compounds is likely to be broad.
Collapse
|
19
|
Pereira IAG, Mendonça DVC, Tavares GSV, Lage DP, Ramos FF, Oliveira-da-Silva JA, Antinarelli LMR, Machado AS, Carvalho LM, Carvalho AMRS, Salustiano IV, Reis TAR, Bandeira RS, Silva AM, Martins VT, Chávez-Fumagalli MA, Humbert MV, Roatt BM, Duarte MC, Menezes-Souza D, Coimbra ES, Leite JPV, Coelho EAF, Gonçalves DU. Parasitological and immunological evaluation of a novel chemotherapeutic agent against visceral leishmaniasis. Parasite Immunol 2020; 42:e12784. [PMID: 32772379 DOI: 10.1111/pim.12784] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/29/2020] [Accepted: 08/03/2020] [Indexed: 12/18/2022]
Abstract
AIMS Treatment for visceral leishmaniasis (VL) is hampered by the toxicity and/or high cost of drugs, as well as by emergence of parasite resistance. Therefore, there is an urgent need for new antileishmanial agents. METHODS AND RESULTS In this study, the antileishmanial activity of a diprenylated flavonoid called 5,7,3,4'-tetrahydroxy-6,8-diprenylisoflavone (CMt) was tested against Leishmania infantum and L amazonensis species. Results showed that CMt presented selectivity index (SI) of 70.0 and 165.0 against L infantum and L amazonensis promastigotes, respectively, and of 181.9 and 397.8 against respective axenic amastigotes. Amphotericin B (AmpB) showed lower SI values of 9.1 and 11.1 against L infantum and L amazonensis promastigotes, respectively, and of 12.5 and 14.3 against amastigotes, respectively. CMt was effective in the treatment of infected macrophages and caused alterations in the parasite mitochondria. L infantum-infected mice treated with miltefosine, CMt alone or incorporated in polymeric micelles (CMt/Mic) presented significant reductions in the parasite load in distinct organs, when compared to the control groups. An antileishmanial Th1-type cellular and humoral immune response were developed one and 15 days after treatment, with CMt/Mic-treated mice presenting a better protective response. CONCLUSION Our data suggest that CMt/Mic could be evaluated as a chemotherapeutic agent against VL.
Collapse
Affiliation(s)
- Isabela A G Pereira
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Débora V C Mendonça
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Grasiele S V Tavares
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daniela P Lage
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fernanda F Ramos
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - João A Oliveira-da-Silva
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luciana M R Antinarelli
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Amanda S Machado
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Lívia M Carvalho
- Departamento de Ciências Biológicas, Insituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Ana Maria R S Carvalho
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Iorrana V Salustiano
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brasil
| | - Thiago A R Reis
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Raquel S Bandeira
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Alessandra M Silva
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vívian T Martins
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Maria V Humbert
- Neisseria Research Group, Molecular Microbiology, School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, England
| | - Bruno M Roatt
- Departamento de Ciências Biológicas, Insituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Mariana C Duarte
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daniel Menezes-Souza
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Elaine S Coimbra
- Departamento de Parasitologia, Microbiologia e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - João Paulo V Leite
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brasil
| | - Eduardo A F Coelho
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Denise U Gonçalves
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
20
|
Olaleye OA, Kaur M, Onyenaka C, Adebusuyi T. Discovery of Clioquinol and Analogues as Novel Inhibitors of Severe Acute Respiratory Syndrome Coronavirus 2 Infection, ACE2 and ACE2 - Spike Protein Interaction In Vitro. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 32817951 DOI: 10.1101/2020.08.14.250480] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the etiological agent for coronavirus disease 2019 (COVID-19), has emerged as an ongoing global pandemic. Presently, there are no clinically approved vaccines nor drugs for COVID-19. Hence, there is an urgent need to accelerate the development of effective antivirals. Here in, we discovered Clioquinol (5-chloro-7-iodo-8-quinolinol (CLQ)), a FDA approved drug and two of its analogues (7-bromo-5-chloro-8-hydroxyquinoline (CLBQ14); and 5, 7-Dichloro-8-hydroxyquinoline (CLCQ)) as potent inhibitors of SARS-CoV-2 infection induced cytopathic effect in vitro . In addition, all three compounds showed potent anti-exopeptidase activity against recombinant human angiotensin converting enzyme 2 (rhACE2) and inhibited the binding of rhACE2 with SARS-CoV-2 Spike (RBD) protein. CLQ displayed the highest potency in the low micromolar range, with its antiviral activity showing strong correlation with inhibition of rhACE2 and rhACE2-RBD interaction. Altogether, our findings provide a new mode of action and molecular target for CLQ and validates this pharmacophore as a promising lead series for clinical development of potential therapeutics for COVID-19.
Collapse
|
21
|
de Chaves MA, Ferreira do Amaral T, Monteiro da Silva Rodrigues Coutinho N, Fernanda Andrzejewski Kaminski T, Teixeira ML, Flavio Souza de Oliveira L, de Andrade SF, Fuentefria AM. Synergistic association of clioquinol with antifungal drugs against biofilm forms of clinical Fusarium isolates. Mycoses 2020; 63:1069-1082. [PMID: 32662568 DOI: 10.1111/myc.13142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND The influence of biofilm on the complexity of fungal diseases has been reported in recent years, especially in non-invasive mycoses such as keratitis and onychomycosis. The difficulty in treating cases of fusariosis in the human medical clinic exemplifies this situation, because when Fusarium spp. are present in the form of biofilm, the permeation of antifungal agents is compromised. OBJECTIVES This study proposes an association of clioquinol, an inhibitor of fungal cells with antifungal drugs prescribed to combat fusariosis in humans. METHODS Susceptibility was assessed by microdilution in broth. Formation of biofilm by staining with violet crystal. Inhibition and removal of biofilm using the MTT colorimetric reagent. Time-kill combination, hypoallergenicity test, cytotoxicity test and toxicity prediction by computer analysis were also performed. RESULTS Clioquinol associated with voriconazole and ciclopirox inhibited biofilm formation. Possibly, clioquinol acts in the germination and elongation of hyphae, while voriconazole prevents cell adhesion and ciclopirox the formation of the extracellular polymeric matrix. The CLIO-VRC association reduced the biofilm formation by more than 90%, while the CLIO-CPX association prevented over 95%. None of the association was irritating, and over 90% of the leucocytes remained viable. Computational analysis does not reveal toxicity relevant to CLIO, whereas VRC and CPX showed some risks for systemic use, but suitable for topical formulations. CONCLUSIONS The combination of CLIO-VRC or CLIO-CPX proved to be a promising association strategy in the medical clinic, both in combating fungal keratitis and onychomycosis, since they prevent the initial process of establishing an infection, the formation of biofilm.
Collapse
Affiliation(s)
- Magda Antunes de Chaves
- Programa de Pós-Graduação em Microbiologia Agrícola e do Ambiente, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | | | | | - Mário Lettieri Teixeira
- Laboratório de Bioquímica e Toxicologia, Instituto Federal de Santa Catarina, Concórdia, Brazil
| | | | - Saulo Fernandes de Andrade
- Programa de Pós-Graduação em Microbiologia Agrícola e do Ambiente, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Alexandre Meneghello Fuentefria
- Programa de Pós-Graduação em Microbiologia Agrícola e do Ambiente, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
22
|
Costa CA, Lopes RM, Ferraz LS, Esteves GN, Di Iorio JF, Souza AA, de Oliveira IM, Manarin F, Judice WA, Stefani HA, Rodrigues T. Cytotoxicity of 4-substituted quinoline derivatives: Anticancer and antileishmanial potential. Bioorg Med Chem 2020; 28:115511. [DOI: 10.1016/j.bmc.2020.115511] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/08/2020] [Accepted: 04/14/2020] [Indexed: 12/11/2022]
|
23
|
Tavares GSV, Mendonça DVC, Pereira IAG, Oliveira-da-Silva JA, Ramos FF, Lage DP, Machado AS, Carvalho LM, Reis TAR, Perin L, Carvalho AMRS, Ottoni FM, Ludolf F, Freitas CS, Bandeira RS, Silva AM, Chávez-Fumagalli MA, Duarte MC, Menezes-Souza D, Alves RJ, Roatt BM, Coelho EAF. A clioquinol-containing Pluronic ® F127 polymeric micelle system is effective in the treatment of visceral leishmaniasis in a murine model. ACTA ACUST UNITED AC 2020; 27:29. [PMID: 32351209 PMCID: PMC7191975 DOI: 10.1051/parasite/2020027] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 04/14/2020] [Indexed: 12/21/2022]
Abstract
A clioquinol (ICHQ)-containing Pluronic® F127 polymeric micelle system (ICHQ/Mic) was recently shown to be effective against Leishmania amazonensis infection in a murine model. In the present study, ICHQ/Mic was tested against L. infantum infection. BALB/c mice (n = 12 per group) were infected with L. infantum stationary promastigotes through subcutaneous injection and, 45 days after challenge, received saline or were treated via the subcutaneous route with empty micelles, ICHQ or ICHQ/Mic. In addition, animals were treated with miltefosine by the oral route, as a drug control. Half of the animals were euthanized 1 and 15 days after treatment, aiming to evaluate two endpoints after therapy, when parasitological and immunological parameters were investigated. Results showed that the treatment using miltefosine, ICHQ or ICHQ/Mic induced significantly higher anti-parasite IFN-γ, IL-12, GM-CSF, nitrite and IgG2a isotype antibody levels, which were associated with low IL-4 and IL-10 production. In addition, a higher frequency of IFN-γ and TNF-α-producing CD4+ and CD8+ T-cells was found in these animals. The parasite load was evaluated in distinct organs, and results showed that the treatment using miltefosine, ICHQ or ICHQ/Mic induced significant reductions in organic parasitism in the treated and infected mice. A comparison between the treatments suggested that ICHQ/Mic was the most effective in inducing a highly polarized Th1-type response, as well as reducing the parasite load in significant levels in the treated and infected animals. Data obtained 15 days after treatment suggested maintenance of the immunological and parasitological responses. In conclusion, ICHQ/Mic could be considered in future studies for the treatment of visceral leishmaniasis.
Collapse
Affiliation(s)
- Grasiele S V Tavares
- Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Débora V C Mendonça
- Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Isabela A G Pereira
- Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Fernanda F Ramos
- Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daniela P Lage
- Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Amanda S Machado
- Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Lívia M Carvalho
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Thiago A R Reis
- Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luísa Perin
- Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Flaviano M Ottoni
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas/NUPEB, Departamento de Ciências Biológicas, Insituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Fernanda Ludolf
- Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Camila S Freitas
- Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Raquel S Bandeira
- Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Alessandra M Silva
- Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Mariana C Duarte
- Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil - Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daniel Menezes-Souza
- Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil - Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ricardo J Alves
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas/NUPEB, Departamento de Ciências Biológicas, Insituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Bruno M Roatt
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Eduardo A F Coelho
- Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil - Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
24
|
Bibi M, Choudhary MI, Yousuf S. Crystal structure and Hirshfeld surface analysis of the methanol solvate of sclareol, a labdane-type diterpenoid. Acta Crystallogr E Crystallogr Commun 2020; 76:294-297. [PMID: 32148863 PMCID: PMC7057370 DOI: 10.1107/s2056989020001474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/03/2020] [Indexed: 11/11/2022]
Abstract
The title compound, C20H36O2·CH3OH [systematic name: (3S)-4-[(S)-3-hy-droxy-3-methyl-pent-4-en-1-yl]-3,4a,8,8-tetra-methyl-deca-hydro-naphthalen-3-ol methanol monosolvate], is a methanol solvate of sclareol, a diterpene oil isolated from the medicinally important medicinal herb Salvia sclarea, commonly known as clary sage. It crystallizes in space group P1 (No. 1) with Z' = 2. The sclareol mol-ecule comprises two trans-fused cyclo-hexane rings, each having an equatorially oriented hydroxyl group, and a 3-methyl-pent-1-en-3-ol side chain. In the crystal, Os-H⋯Os, Os-H⋯Om, Om-H⋯Os and Om-H⋯Om (s = sclareol, m = methanol) hydrogen bonds connect neighboring mol-ecules into infinite [010] chains. The title compound exhibits weak anti-leishmanial activity (IC50 = 66.4 ± 1.0 µM ml-1) against standard miltefosine (IC50 = 25.8 ± 0.2 µM ml-1).
Collapse
Affiliation(s)
- Memoona Bibi
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| | - M. Iqbal Choudhary
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| | - Sammer Yousuf
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| |
Collapse
|
25
|
Cherdtrakulkiat R, Worachartcheewan A, Tantimavanich S, Lawung R, Sinthupoom N, Prachayasittikul S, Ruchirawat S, Prachayasittikul V. Discovery of novel halogenated 8‐hydroxyquinoline‐based anti‐MRSA agents: In vitro and QSAR studies. Drug Dev Res 2019; 81:127-135. [DOI: 10.1002/ddr.21611] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 09/10/2019] [Accepted: 09/21/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Rungrot Cherdtrakulkiat
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical TechnologyMahidol University Bangkok Thailand
| | - Apilak Worachartcheewan
- Department of Community Medical Technology, Faculty of Medical TechnologyMahidol University Bangkok Thailand
- Department of Clinical Chemistry, Faculty of Medical TechnologyMahidol University Bangkok Thailand
| | - Srisurang Tantimavanich
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical TechnologyMahidol University Bangkok Thailand
| | - Ratana Lawung
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical TechnologyMahidol University Bangkok Thailand
| | - Nujarin Sinthupoom
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical TechnologyMahidol University Bangkok Thailand
| | - Supaluk Prachayasittikul
- Center of Data Mining and Biomedical Informatics, Faculty of Medical TechnologyMahidol University Bangkok Thailand
| | - Somsak Ruchirawat
- Laboratory of Medicinal ChemistryChulabhorn Research Institute Bangkok Thailand
- Program in Chemical BiologyChulabhorn Graduate Institute Bangkok Thailand
- Center of Excellence on Environmental Health and Toxicology, Commission on Higher Education (CHE)Ministry of Education Thailand
| | - Virapong Prachayasittikul
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical TechnologyMahidol University Bangkok Thailand
| |
Collapse
|
26
|
Sousa JKT, Antinarelli LMR, Mendonça DVC, Lage DP, Tavares GSV, Dias DS, Ribeiro PAF, Ludolf F, Coelho VTS, Oliveira-da-Silva JA, Perin L, Oliveira BA, Alvarenga DF, Chávez-Fumagalli MA, Brandão GC, Nobre V, Pereira GR, Coimbra ES, Coelho EAF. A chloroquinoline derivate presents effective in vitro and in vivo antileishmanial activity against Leishmania species that cause tegumentary and visceral leishmaniasis. Parasitol Int 2019; 73:101966. [PMID: 31362122 DOI: 10.1016/j.parint.2019.101966] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 06/27/2019] [Accepted: 07/26/2019] [Indexed: 02/06/2023]
Abstract
The identification of new therapeutics to treat leishmaniasis is desirable, since available drugs are toxic and present high cost and/or poor availability. Therefore, the discovery of safer, more effective and selective pharmaceutical options is of utmost importance. Efforts towards the development of new candidates based on molecule analogs with known biological functions have been an interesting and cost-effective strategy. In this context, quinoline derivatives have proven to be effective biological activities against distinct diseases. In the present study, a new chloroquinoline derivate, AM1009, was in vitro tested against two Leishmania species that cause leishmaniasis. The present study analyzed the necessary inhibitory concentration to preclude 50% of the Leishmania promastigotes and axenic amastigotes (EC50 value), as well as the inhibitory concentrations to preclude 50% of the murine macrophages and human red blood cells (CC50 and RBC50 values, respectively). In addition, the treatment of infected macrophages and the inhibition of infection using pre-treated parasites were also investigated, as was the mechanism of action of the molecule in L. amazonensis. To investigate the in vivo therapeutic effect, BALB/c mice were infected with L. amazonensis and later treated with AM1009. Parasitological and immunological parameters were also evaluated. Clioquinol, a known antileishmanial quinoline derivate, and amphotericin B (AmpB), were used as molecule and drug controls, respectively. Results in both in vitro and in vivo experiments showed a better and more selective action of AM1009 to kill the in vitro parasites, as well as in treating infected mice, when compared to results obtained using clioquinol or AmpB. AM1009-treated animals presented significantly lower average lesion diameter and parasite burden in the infected tissue and organs evaluated in this study, as well as a more polarized antileishmanial Th1 immune response and low renal and hepatic toxicity. This result suggests that AM1009 should be considered a possible therapeutic target to be evaluated in future studies for treatment against leishmaniasis.
Collapse
Affiliation(s)
- Jessica K T Sousa
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, 30130-100 Belo Horizonte, Minas Gerais, Brazil
| | - Luciana M R Antinarelli
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, 30130-100 Belo Horizonte, Minas Gerais, Brazil
| | - Débora V C Mendonça
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, 30130-100 Belo Horizonte, Minas Gerais, Brazil
| | - Daniela P Lage
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, 30130-100 Belo Horizonte, Minas Gerais, Brazil
| | - Grasiele S V Tavares
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, 30130-100 Belo Horizonte, Minas Gerais, Brazil
| | - Daniel S Dias
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, 30130-100 Belo Horizonte, Minas Gerais, Brazil
| | - Patrícia A F Ribeiro
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, 30130-100 Belo Horizonte, Minas Gerais, Brazil
| | - Fernanda Ludolf
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, 30130-100 Belo Horizonte, Minas Gerais, Brazil
| | - Vinicio T S Coelho
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, 30130-100 Belo Horizonte, Minas Gerais, Brazil
| | - João A Oliveira-da-Silva
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, 30130-100 Belo Horizonte, Minas Gerais, Brazil
| | - Luísa Perin
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, 30130-100 Belo Horizonte, Minas Gerais, Brazil
| | - Bianka A Oliveira
- Pontifícia Universidade Católica de Minas Gerais, Departamento de Física e Química, Instituto de Ciências Exatas e Informática, 30535-901 Belo Horizonte, Minas Gerais, Brazil
| | - Denis F Alvarenga
- Pontifícia Universidade Católica de Minas Gerais, Departamento de Física e Química, Instituto de Ciências Exatas e Informática, 30535-901 Belo Horizonte, Minas Gerais, Brazil
| | - Miguel A Chávez-Fumagalli
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, 30130-100 Belo Horizonte, Minas Gerais, Brazil
| | - Geraldo C Brandão
- Escola de Farmácia, Universidade Federal de Ouro Preto, 35400-000 Ouro Preto, Minas Gerais, Brazil
| | - Vandack Nobre
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, 30130-100 Belo Horizonte, Minas Gerais, Brazil
| | - Guilherme R Pereira
- Pontifícia Universidade Católica de Minas Gerais, Departamento de Física e Química, Instituto de Ciências Exatas e Informática, 30535-901 Belo Horizonte, Minas Gerais, Brazil
| | - Elaine S Coimbra
- Departamento de Parasitologia, Microbiologia e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, 36036-900 Juiz de Fora, Minas Gerais, Brazil
| | - Eduardo A F Coelho
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, 30130-100 Belo Horizonte, Minas Gerais, Brazil; Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
27
|
Teixeira RI, Goulart JS, Corrêa RJ, Garden SJ, Ferreira SB, Netto-Ferreira JC, Ferreira VF, Miro P, Marin ML, Miranda MA, de Lucas NC. A photochemical and theoretical study of the triplet reactivity of furano- and pyrano-1,4-naphthoquionones towards tyrosine and tryptophan derivatives. RSC Adv 2019; 9:13386-13397. [PMID: 35519567 PMCID: PMC9063979 DOI: 10.1039/c9ra01939a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 04/22/2019] [Indexed: 01/08/2023] Open
Abstract
The photochemical reactivity of the triplet state of pyrano- and furano-1,4-naphthoquinone derivatives (1 and 2) has been examined employing nanosecond laser flash photolysis. The quinone triplets were efficiently quenched by l-tryptophan methyl ester hydrochloride, l-tyrosine methyl ester hydrochloride, N-acetyl-l-tryptophan methyl ester and N-acetyl-l-tyrosine methyl ester, substituted phenols and indole (k q ∼109 L mol-1 s-1). For all these quenchers new transients were formed in the quenching process. These were assigned to the corresponding radical pairs that resulted from a coupled electron/proton transfer from the phenols, indole, amino acids, or their esters, to the excited state of the quinone. The proton coupled electron transfer (PCET) mechanism is supported by experimental rate constants, isotopic effects and theoretical calculations. The calculations revealed differences between the hydrogen abstraction reactions of phenol and indole substrates. For the latter, the calculations indicate that electron transfer and proton transfer occur as discrete steps.
Collapse
Affiliation(s)
- Rodolfo I Teixeira
- Instituto de Química - Universidade Federal do Rio de Janeiro Cidade Universitária RJ Brazil
| | - Juliana S Goulart
- Instituto de Química - Universidade Federal do Rio de Janeiro Cidade Universitária RJ Brazil
| | - Rodrigo J Corrêa
- Instituto de Química - Universidade Federal do Rio de Janeiro Cidade Universitária RJ Brazil
| | - Simon J Garden
- Instituto de Química - Universidade Federal do Rio de Janeiro Cidade Universitária RJ Brazil
| | - Sabrina B Ferreira
- Instituto de Química - Universidade Federal do Rio de Janeiro Cidade Universitária RJ Brazil
| | | | - Vitor F Ferreira
- Universidade Federal Fluminense, Faculdade de Farmácia, Departamento de Tecnologia Farmaceûtica Niterói Santa Rosa Brazil
| | - Paula Miro
- Instituto de Tecnología Química, Universitat Politècnica de València-Consejo Superior de Investigaciones Científicas Valencia Spain
| | - M Luisa Marin
- Instituto de Tecnología Química, Universitat Politècnica de València-Consejo Superior de Investigaciones Científicas Valencia Spain
| | - Miguel A Miranda
- Instituto de Tecnología Química, Universitat Politècnica de València-Consejo Superior de Investigaciones Científicas Valencia Spain
| | - Nanci C de Lucas
- Instituto de Química - Universidade Federal do Rio de Janeiro Cidade Universitária RJ Brazil
| |
Collapse
|
28
|
Tavares GS, Mendonça DV, Lage DP, Antinarelli LM, Soyer TG, Senna AJ, Matos GF, Dias DS, Ribeiro PA, Batista JP, Poletto JM, Brandão GC, Chávez-Fumagalli MA, Pereira GR, Coimbra ES, Coelho EA. In vitro and in vivo antileishmanial activity of a fluoroquinoline derivate against Leishmania infantum and Leishmania amazonensis species. Acta Trop 2019; 191:29-37. [PMID: 30586571 DOI: 10.1016/j.actatropica.2018.12.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 10/19/2018] [Accepted: 12/21/2018] [Indexed: 01/09/2023]
Abstract
New therapeutics against leishmaniasis are desirable, since the current drugs applied against this disease complex presents problems, such as the toxicity, high cost and/or parasite resistance. In the present study, a new fluoroquinoline derivate, namely 7-chloro-N-(4-fluorophenethyl)quinolin-4-amine or GF1061, was evaluated regarding to its in vitro antileishmanial action against Leishmania infantum and L. amazonensis species, as well as by its toxicity in mammalian cells and efficacy in the treatment of infected macrophages. The mechanism of action of this molecule in L. amazonensis and the therapeutic efficacy in infected BALB/c mice were also evaluated. Results showed that GF1061 was effective against both parasite species, showing selectivity index (SI) of 38.7 and 42.7 against L. infantum and L. amazonensis promastigotes, respectively, and of 45.0 and 48.9 against the amastigotes, respectively. Amphotericin B (AmpB), used as control, showed SI values of 6.6 and 8.8 against L. infantum and L. amazonensis promastigotes, respectively, and of 2.2 and 2.7 against the amastigotes, respectively. The molecule was effective in treat infected macrophages, as well as it induced alterations in the mitochondrial membrane potential, increase in the reactive oxygen species production, and in the cell integrity of the parasites. Regarding to the in vivo experiments, BALB/c mice (n = 8 per group) were subcutaneously infected with 106L. amazonensis stationary promastigotes and, 60 days post-infection, they received saline or were treated during 10 days, once a day, with AmpB (1 mg/kg body weight) or GF1061 (5 mg/kg body weight). One day after the treatment, the infected tissue, spleen, liver, and draining lymph node (dLN) of the animals were collected, and the parasite load was evaluated. GF1061-treated mice, as compared to the saline and AmpB groups, showed significant reductions in the parasitism in the infected tissue (66% and 62%, respectively), liver (69% and 44%, respectively), spleen (71% and 38%, respectively), and dLN (72% and 48%, respectively). In conclusion, results suggested that GF1061 may be considered as a possible therapeutic target to be evaluated against leishmaniasis in other mammalian hosts.
Collapse
|
29
|
Soyer TG, Mendonça DVC, Tavares GSV, Lage DP, Dias DS, Ribeiro PAF, Perin L, Ludolf F, Coelho VTS, Ferreira ACG, Neves PHAS, Matos GF, Chávez-Fumagalli MA, Coimbra ES, Pereira GR, Coelho EAF, Antinarelli LMR. Evaluation of the in vitro and in vivo antileishmanial activity of a chloroquinolin derivative against Leishmania species capable of causing tegumentary and visceral leishmaniasis. Exp Parasitol 2019; 199:30-37. [PMID: 30817917 DOI: 10.1016/j.exppara.2019.02.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 02/09/2019] [Accepted: 02/23/2019] [Indexed: 12/23/2022]
Abstract
The treatment against leishmaniasis presents problems, since the currently used drugs are toxic and/or have high costs. In addition, parasite resistance has increased. As a consequence, in this study, a chloroquinolin derivative, namely 7-chloro-N,N-dimethylquinolin-4-amine or GF1059, was in vitro and in vivo tested against Leishmania parasites. Experiments were performed to evaluate in vitro antileishmanial activity and cytotoxicity, as well as the treatment of infected macrophages and the inhibition of infection using pre-treated parasites. This study also investigated the GF1059 mechanism of action in L. amazonensis. Results showed that the compound was highly effective against L. infantum and L. amazonensis, presenting a selectivity index of 154.6 and 86.4, respectively, against promastigotes and of 137.6 and 74.3, respectively, against amastigotes. GF1059 was also effective in the treatment of infected macrophages and inhibited the infection of these cells when parasites were pre-incubated with it. The molecule also induced changes in the parasites' mitochondrial membrane potential and cell integrity, and caused an increase in the reactive oxygen species production in L. amazonensis. Experiments performed in BALB/c mice, which had been previously infected with L. amazonensis promastigotes, and thus treated with GF1059, showed that these animals presented significant reductions in the parasite load when the infected tissue, spleen, liver, and draining lymph node were evaluated. GF1059-treated mice presented both lower parasitism and low levels of enzymatic markers, as compared to those receiving amphotericin B, which was used as control. In conclusion, data suggested that GF1059 can be considered a possible therapeutic target to be tested against leishmaniasis.
Collapse
Affiliation(s)
- Tauane G Soyer
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Débora V C Mendonça
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Grasiele S V Tavares
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daniela P Lage
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daniel S Dias
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Patrícia A F Ribeiro
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luisa Perin
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fernanda Ludolf
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vinicio T S Coelho
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Andreza C G Ferreira
- Pontifícia Universidade Católica de Minas Gerais, Departamento de Física e Química, Instituto de Ciências Exatas e Informática, Belo Horizonte, Minas Gerais, Brazil
| | - Pedro H A S Neves
- Pontifícia Universidade Católica de Minas Gerais, Departamento de Física e Química, Instituto de Ciências Exatas e Informática, Belo Horizonte, Minas Gerais, Brazil
| | - Guilherme F Matos
- Pontifícia Universidade Católica de Minas Gerais, Departamento de Física e Química, Instituto de Ciências Exatas e Informática, Belo Horizonte, Minas Gerais, Brazil
| | - Miguel A Chávez-Fumagalli
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Elaine S Coimbra
- Departamento de Parasitologia, Microbiologia e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Guilherme R Pereira
- Pontifícia Universidade Católica de Minas Gerais, Departamento de Física e Química, Instituto de Ciências Exatas e Informática, Belo Horizonte, Minas Gerais, Brazil
| | - Eduardo A F Coelho
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| | - Luciana M R Antinarelli
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
30
|
Tavares GS, Mendonça DV, Miyazaki CK, Lage DP, Soyer TG, Carvalho LM, Ottoni FM, Dias DS, Ribeiro PA, Antinarelli LM, Ludolf F, Duarte MC, Coimbra ES, Chávez-Fumagalli MA, Roatt BM, Menezes-Souza D, Barichello JM, Alves RJ, Coelho EA. A Pluronic® F127-based polymeric micelle system containing an antileishmanial molecule is immunotherapeutic and effective in the treatment against Leishmania amazonensis infection. Parasitol Int 2019; 68:63-72. [DOI: 10.1016/j.parint.2018.10.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/12/2018] [Accepted: 10/15/2018] [Indexed: 12/11/2022]
|
31
|
Senerovic L, Opsenica D, Moric I, Aleksic I, Spasić M, Vasiljevic B. Quinolines and Quinolones as Antibacterial, Antifungal, Anti-virulence, Antiviral and Anti-parasitic Agents. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1282:37-69. [PMID: 31515709 DOI: 10.1007/5584_2019_428] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Infective diseases have become health threat of a global proportion due to appearance and spread of microorganisms resistant to majority of therapeutics currently used for their treatment. Therefore, there is a constant need for development of new antimicrobial agents, as well as novel therapeutic strategies. Quinolines and quinolones, isolated from plants, animals, and microorganisms, have demonstrated numerous biological activities such as antimicrobial, insecticidal, anti-inflammatory, antiplatelet, and antitumor. For more than two centuries quinoline/quinolone moiety has been used as a scaffold for drug development and even today it represents an inexhaustible inspiration for design and development of novel semi-synthetic or synthetic agents exhibiting broad spectrum of bioactivities. The structural diversity of synthetized compounds provides high and selective activity attained through different mechanisms of action, as well as low toxicity on human cells. This review describes quinoline and quinolone derivatives with antibacterial, antifungal, anti-virulent, antiviral, and anti-parasitic activities with the focus on the last 10 years literature.
Collapse
Affiliation(s)
- Lidija Senerovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia.
| | - Dejan Opsenica
- Institute of Chemistry, Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
- Center of excellence in Environmental Chemistry and Engineering, ICTM - University of Belgrade, Belgrade, Serbia
| | - Ivana Moric
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Ivana Aleksic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Marta Spasić
- Faculty of Chemistry, University of Belgrade, Belgrade, Serbia
| | - Branka Vasiljevic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
32
|
Synthesis, leishmanicidal activity, structural descriptors and structure-activity relationship of quinoline derivatives. Future Med Chem 2018; 10:2069-2085. [PMID: 30066582 DOI: 10.4155/fmc-2018-0124] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
AIM Considering the epidemiology of leishmaniasis, the emergence of resistant parasites to the approved drugs, and severe clinical manifestations, the development of novel leishmanicidal molecules has become of considerable importance. RESULTS In this work, three commercially available and 19 synthesized quinoline derivatives were evaluated against promastigote and amastigote forms of Leishmania (Leishmania) amazonensis. In addition, structural parameters and molecular electrostatic potentials were obtained by theoretical calculations, allowing statistical (principal component analyses and hierarchical cluster analyses) and comparative (molecular electrostatic potentials vs leishmanicidal activities) studies, respectively. CONCLUSION Principal component analyses and hierarchical cluster analyses suggested volume and polar surface area as possible structural descriptors for the leishmanicidal activity. Furthermore, a comparison between molecular electrostatic potentials and leishmanicidal activities afforded a reasonable structure-activity relationship.
Collapse
|