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
|
Amaral M, Romanelli MM, Asiki H, Bicker J, Lage DP, Freitas CS, Taniwaki NN, Lago JHG, Coelho EAF, Falcão A, Fortuna A, Anderson EA, Tempone AG. Synthesis of a dehydrodieugenol B derivative as a lead compound for visceral leishmaniasis-mechanism of action and in vivo pharmacokinetic studies. Antimicrob Agents Chemother 2024; 68:e0083124. [PMID: 39382276 PMCID: PMC11539218 DOI: 10.1128/aac.00831-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 09/16/2024] [Indexed: 10/10/2024] Open
Abstract
Leishmaniasis is a parasitic neglected tropical disease, affecting 12 million people. Available treatments present several limitations, with an increasing number of resistance cases. In the search for new chemotherapies, the natural product dehydrodieugenol B was used as a scaffold for the synthesis of a series of derivatives, resulting in the discovery of the promising analog [4-(4-(5-allyl-3-methoxy-2-((4-methoxybenzyl)oxy)phenoxy)-3-methoxybenzyl)morpholine, 1]. In this work, we investigated the effect of compound 1 on cell signaling in Leishmania (L.) infantum, culminating in cell death, as well as its immunomodulatory effect in the host cell. Additionally, we performed a pharmacokinetic profile study in an animal model. After treatment, compound 1 induced the alkalinization of acidocalcisomes and concomitant Ca2+ release in the parasite. These events may induce depolarization of the mitochondrial potential, with successive collapse of the bioenergetic system, leading to a reduction of ATP and reactive oxygen species (ROS) levels. The analysis of total proteins and protein profile by matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF/MS) demonstrated that compound 1 also altered the parasite proteins after treatment. Transmission electron microscopy studies revealed ultrastructural damage to mitochondria; together, these data suggest that compound 1 may promote autophagic cell death. Additionally, compound 1 also induced an immunomodulatory effect in host cells, with a reduction of Th1 and Th2 cytokine response, characterizing an anti-inflammatory compound. The obtained pharmacokinetic profile in rats enhances the potential of the compound, with a mean plasma half-life (T1/2) of 21 h. These data reinforce the potential of compound 1 as a new lead for future efficacy studies.
Collapse
Affiliation(s)
- Maiara Amaral
- Laboratory of Physiopathology, Instituto Butantan, São Paulo, Brazil
- Instituto de Medicina Tropical, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Maiara M. Romanelli
- Instituto de Medicina Tropical, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Hannah Asiki
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Oxford, United Kingdom
| | - Joana Bicker
- Laboratory of Pharmacology and Pharmaceutical Care, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute of Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| | - Daniela P. Lage
- Laboratório de Pesquisa do 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
| | - Camila S. Freitas
- Laboratório de Pesquisa do 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
| | - Noemi N. Taniwaki
- Laboratory of Electron Microscopy, Instituto Adolfo Lutz, São Paulo, Brazil
| | - Joao Henrique G. Lago
- Center for Natural and Human Sciences, Federal University of ABC, Santo Andre, Brazil
| | - Eduardo A. F. Coelho
- Laboratório de Pesquisa do 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
| | - Amílcar Falcão
- Laboratory of Pharmacology and Pharmaceutical Care, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute of Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| | - Ana Fortuna
- Laboratory of Pharmacology and Pharmaceutical Care, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute of Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| | - Edward A. Anderson
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Oxford, United Kingdom
| | - Andre G. Tempone
- Instituto de Medicina Tropical, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
3
|
da Silva Bortoleti BT, Camargo PG, Gonçalves MD, Tomiotto-Pellissier F, Silva TF, Concato VM, Detoni MB, Bidóia DL, da Silva Lima CH, Rodrigues CR, Bispo MDLF, de Macedo FC, Conchon-Costa I, Miranda-Sapla MM, Wowk PF, Pavanelli WR. Effect of a thiohydantoin salt derived from l-Arginine on Leishmania amazonensis and infected cells: Insights from biological effects to molecular docking interactions. Chem Biol Interact 2024; 403:111216. [PMID: 39218371 DOI: 10.1016/j.cbi.2024.111216] [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: 02/27/2024] [Revised: 07/12/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Leishmaniasis is a neglected tropical disease caused by parasites of the genus Leishmania and is responsible for more than 1 million new cases and 70,000 deaths annually worldwide. Treatment has high costs, toxicity, complex and long administration time, several adverse effects, and drug-resistant strains, therefore new therapies are urgently needed. Synthetic compounds have been highlighted in the medicinal chemistry field as a strong option for drug development against different diseases. Organic salts (OS) have multiple biological activities, including activity against protozoa such as Leishmania spp. This study aimed to investigate the in vitro leishmanicidal activity and death mechanisms of a thiohydantoin salt derived from l-arginine (ThS) against Leishmania amazonensis. We observed that ThS treatment inhibited promastigote proliferation, increased ROS production, phosphatidylserine exposure and plasma membrane permeabilization, loss of mitochondrial membrane potential, lipid body accumulation, autophagic vacuole formation, cell cycle alteration, and morphological and ultrastructural changes, showing parasites death. Additionally, ThS presents low cytotoxicity in murine macrophages (J774A.1), human monocytes (THP-1), and sheep erythrocytes. ThS in vitro cell treatment reduced the percentage of infected macrophages and the number of amastigotes per macrophage by increasing ROS production and reducing TNF-α levels. These results highlight the potential of ThS among thiohydantoins, mainly related to the arginine portion, as a leishmanicidal drug for future drug strategies for leishmaniasis treatment. Notably, in silico investigation of key targets from L. amazonensis, revealed that a ThS compound from the l-arginine amino acid strongly interacts with arginase (ARG) and TNF-α converting enzyme (TACE), suggesting its potential as a Leishmania inhibitor.
Collapse
Affiliation(s)
- Bruna Taciane da Silva Bortoleti
- Biosciences and Biotechnology Postgraduate Program, Carlos Chagas Institute, (ICC/Fiocruz/PR), Curitiba, Paraná, Brazil; State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil
| | - Priscila Goes Camargo
- Federal University of Rio de Janeiro, Faculty of Pharmacy, Rio de Janeiro, Rio de Janeiro, Brazil; State University of Londrina (UEL/PR), Chemistry Department, Londrina, Paraná, Brazil
| | - Manoela Daiele Gonçalves
- State University of Londrina (UEL/PR), Laboratory of Biotransformation and Phytochemistry, Londrina, Paraná, Brazil
| | - Fernanda Tomiotto-Pellissier
- Biosciences and Biotechnology Postgraduate Program, Carlos Chagas Institute, (ICC/Fiocruz/PR), Curitiba, Paraná, Brazil; State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil
| | - Taylon Felipe Silva
- State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil
| | - Virginia Marcia Concato
- State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil
| | - Mariana Barbosa Detoni
- State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil
| | - Danielle Larazin Bidóia
- State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil
| | | | - Carlos Rangel Rodrigues
- Federal University of Rio de Janeiro, Faculty of Pharmacy, Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Ivete Conchon-Costa
- State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil
| | | | - Pryscilla Fanini Wowk
- Carlos Chagas Institute (ICC/Fiocruz/PR), Molecular Immunology and Cellular Group, Curitiba, Paraná, Brazil.
| | - Wander Rogério Pavanelli
- State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil.
| |
Collapse
|
4
|
Oliveira SSC, Marinho FA, Sangenito LS, Seabra SH, Menna-Barreto RF, d’Avila CM, Santos ALS, Branquinha MH. Susceptibility of Leishmania amazonensis Axenic Amastigotes to the Calpain Inhibitor MDL28170. Trop Med Infect Dis 2024; 9:259. [PMID: 39591265 PMCID: PMC11598141 DOI: 10.3390/tropicalmed9110259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Leishmaniasis encompasses a group of neglected diseases caused by flagellated protozoa belonging to the Leishmania genus, associated with high morbidity and mortality. The search for compounds with anti-Leishmania activity that exhibit lower toxicity and can overcome the emergence of resistant strains remains a significant goal. In this context, the calpain inhibitor MDL28170 has previously demonstrated deleterious effects against promastigote forms of Leishmania amazonensis, which led us to investigate its role on axenic amastigote forms. The calpain inhibitor MDL28170 was able to decrease the viability of amastigotes in a typically dose-dependent manner. The treatment with the IC50 dose (13.5 μM) for 72 h led to significant amastigote lysis and increased cell-to-cell aggregation. Ultrastructural analysis revealed several cellular alterations, including disruption of the trans-Golgi network and the formation of autophagosomes when treated with MDL28170 at ½ × IC50 dose. Additionally, mitochondrial swelling and the formation of concentric membranous structures inside the mitochondrion were observed after incubation with the IC50 dose. These results reinforce the potential application of the calpain inhibitor MDL28170 against L. amazonensis, highlighting its effectiveness and possible mechanism of action against the parasite.
Collapse
Affiliation(s)
- Simone S. C. Oliveira
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (S.S.C.O.); (F.A.M.); (L.S.S.)
| | - Fernanda A. Marinho
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (S.S.C.O.); (F.A.M.); (L.S.S.)
| | - Leandro S. Sangenito
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (S.S.C.O.); (F.A.M.); (L.S.S.)
- Instituto Federal de Educação, Ciência e Tecnologia do Rio de Janeiro, Campus Nilópolis, Rio de Janeiro 26530-060, Brazil
| | - Sergio H. Seabra
- Laboratório de Biologia Celular e Tecidual, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Rio de Janeiro 28013-602, Brazil;
| | - Rubem F. Menna-Barreto
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro 21040-360, Brazil;
| | - Claudia M. d’Avila
- Laboratório de Doenças Parasitárias, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro 21040-360, Brazil;
| | - André L. S. Santos
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (S.S.C.O.); (F.A.M.); (L.S.S.)
- Programa de Pós-Graduação em Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-909, Brazil
| | - Marta H. Branquinha
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (S.S.C.O.); (F.A.M.); (L.S.S.)
| |
Collapse
|
5
|
Pozo-Martínez J, Arán VJ, Zúñiga-Bustos M, Parra-Magna S, Rocha-Valderrama E, Liempi A, Castillo C, Olea-Azar C, Moncada-Basualto M. In Vitro Evaluation of New 5-Nitroindazolin-3-one Derivatives as Promising Agents against Trypanosoma cruzi. Int J Mol Sci 2024; 25:11107. [PMID: 39456891 PMCID: PMC11508334 DOI: 10.3390/ijms252011107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Chagas disease is a prevalent health problem in Latin America which has received insufficient attention worldwide. Current treatments for this disease, benznidazole and nifurtimox, have limited efficacy and may cause side effects. A recent study proposed investigating a wide range of nitroindazole and indazolone derivatives as feasible treatments. Therefore, it is proposed that adding a nitro group at the 5-position of the indazole and indazolone structure could enhance trypanocidal activity by inducing oxidative stress through activation of the nitro group by NTRs (nitroreductases). The study results indicate that the nitro group advances free radical production, as confirmed by several analyses. Compound 5a (5-nitro-2-picolyl-indazolin-3-one) shows the most favorable trypanocidal activity (1.1 ± 0.3 µM in epimastigotes and 5.4 ± 1.0 µM in trypomastigotes), with a selectivity index superior to nifurtimox. Analysis of the mechanism of action indicated that the nitro group at the 5-position of the indazole ring induces the generation of reactive oxygen species (ROS), which causes apoptosis in the parasites. Computational docking studies reveal how the compounds interact with critical residues of the NTR and FMNH2 (flavin mononucleotide reduced) in the binding site, which is also present in active ligands. The lipophilicity of the studied series was shown to influence their activity, and the nitro group was found to play a crucial role in generating free radicals. Further investigations are needed of derivatives with comparable lipophilic characteristics and the location of the nitro group in different positions of the base structure.
Collapse
Affiliation(s)
- Josué Pozo-Martínez
- Department of Molecular Pharmacology and Clinical, Faculty of Medicine, University of Chile, Santiago 8380453, Chile;
- Laboratorio de Química-Médica, Facultad de Ciencia y Tecnología, Universidad del Azuay, Av. 24 de Mayo 777, Cuenca 010204, Ecuador
| | - Vicente J. Arán
- Instituto de Química Médica (CSIC), Juan de la Cierva 3, 28006 Madrid, Spain;
| | - Matías Zúñiga-Bustos
- Instituto Universitario de Investigación y Desarrollo Tecnológico, Universidad Tecnológica Metropolitana, Santiago 8940577, Chile; (M.Z.-B.)
| | - Sebastián Parra-Magna
- Instituto Universitario de Investigación y Desarrollo Tecnológico, Universidad Tecnológica Metropolitana, Santiago 8940577, Chile; (M.Z.-B.)
- Free Radical and Antioxidants Laboratory, Inorganic and Analytical Department, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago 8380492, Chile
| | - Esteban Rocha-Valderrama
- Instituto Universitario de Investigación y Desarrollo Tecnológico, Universidad Tecnológica Metropolitana, Santiago 8940577, Chile; (M.Z.-B.)
- Free Radical and Antioxidants Laboratory, Inorganic and Analytical Department, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago 8380492, Chile
| | - Ana Liempi
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile (C.C.)
| | - Christian Castillo
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile (C.C.)
| | - Claudio Olea-Azar
- Department of Molecular Pharmacology and Clinical, Faculty of Medicine, University of Chile, Santiago 8380453, Chile;
| | - Mauricio Moncada-Basualto
- Instituto Universitario de Investigación y Desarrollo Tecnológico, Universidad Tecnológica Metropolitana, Santiago 8940577, Chile; (M.Z.-B.)
| |
Collapse
|
6
|
Barbosa DCS, Holanda VN, Lima EMA, Cavalcante MKA, Brelaz-de-Castro MCA, Chaves EJF, Rocha GB, Silva CJO, Oliveira RN, Figueiredo RCBQ. 1,2,4-Oxadiazole Derivatives: Physicochemical Properties, Antileishmanial Potential, Docking and Molecular Dynamic Simulations of Leishmania infantum Target Proteins. Molecules 2024; 29:4654. [PMID: 39407583 PMCID: PMC11478322 DOI: 10.3390/molecules29194654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 09/16/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Visceral leishmaniasis (VL), caused by protozoa of the genus Leishmania, remains a significant public health concern due to its potentially lethal nature if untreated. Current chemotherapy options are limited by severe toxicity and drug resistance. Derivatives of 1,2,4-oxadiazole have emerged as promising drug candidates due to their broad biological activity. This study investigated the effects of novel 1,2,4-oxadiazole derivatives (Ox1-Ox7) on Leishmania infantum, the etiological agent of VL. In silico predictions using SwissADME suggest that these compounds have high oral absorption and good bioavailability. Among them, Ox1 showed the most promise, with higher selectivity against promastigotes and lower cytotoxicity towards L929 fibroblasts and J774.G8 macrophages. Ox1 exhibited selectivity indices of 18.7 and 61.7 against L. infantum promastigotes and amastigotes, respectively, compared to peritoneal macrophages. Ultrastructural analyses revealed severe morphological damage in both parasite forms, leading to cell death. Additionally, Ox1 decreased the mitochondrial membrane potential in promastigotes, as shown by flow cytometry. Molecular docking and dynamic simulations indicated a strong affinity of Ox1 for the L. infantum CYP51 enzyme. Overall, Ox1 is a promising and effective compound against L. infantum.
Collapse
Affiliation(s)
- Deyzi C. S. Barbosa
- Department of Microbiology, Aggeu Magalhães Institute (IAM-FIOCRUZ), Recife 50740-465, PE, Brazil;
| | - Vanderlan N. Holanda
- Department of Biomedicine, University Center of Vitória de Santo Antão (UNIVISA), Vitória de Santo Antão 55610-050, PE, Brazil
| | - Elton M. A. Lima
- Center for Exact and Natural Sciences, Federal University of Pernambuco (UFPE), Recife 50740-560, PE, Brazil
| | - Marton K. A. Cavalcante
- Parasitology Laboratory, Academic Center of Vitória, Federal University of Pernambuco (UFPE), Recife 50670-420, PE, Brazil
- Department of Immunology, Aggeu Magalhães Institute (IAM-FIOCRUZ), Recife 50740-465, PE, Brazil
| | - Maria Carolina A. Brelaz-de-Castro
- Parasitology Laboratory, Academic Center of Vitória, Federal University of Pernambuco (UFPE), Recife 50670-420, PE, Brazil
- Department of Immunology, Aggeu Magalhães Institute (IAM-FIOCRUZ), Recife 50740-465, PE, Brazil
| | - Elton J. F. Chaves
- Department of Chemistry, Federal University of Paraíba (UFPB), João Pessoa 58051-900, PB, Brazil
| | - Gerd B. Rocha
- Department of Chemistry, Federal University of Paraíba (UFPB), João Pessoa 58051-900, PB, Brazil
| | - Carla J. O. Silva
- Department of Fundamental Chemistry, Federal University of Pernambuco (UFPE), Recife 50740-540, PE, Brazil;
| | - Ronaldo N. Oliveira
- Department of Chemistry, Federal Rural University of Pernambuco (UFRPE), Recife 52171-900, PE, Brazil;
| | | |
Collapse
|
7
|
Mendes IC, Dos Reis Bertoldo W, Miranda-Junior AS, Assis AVD, Repolês BM, Ferreira WRR, Chame DF, Souza DDL, Pavani RS, Macedo AM, Franco GR, Serra E, Perdomo V, Menck CFM, da Silva Leandro G, Fragoso SP, Barbosa Elias MCQ, Machado CR. DNA lesions that block transcription induce the death of Trypanosoma cruzi via ATR activation, which is dependent on the presence of R-loops. DNA Repair (Amst) 2024; 141:103726. [PMID: 39096697 DOI: 10.1016/j.dnarep.2024.103726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 06/25/2024] [Accepted: 07/07/2024] [Indexed: 08/05/2024]
Abstract
Trypanosoma cruzi is the etiological agent of Chagas disease and a peculiar eukaryote with unique biological characteristics. DNA damage can block RNA polymerase, activating transcription-coupled nucleotide excision repair (TC-NER), a DNA repair pathway specialized in lesions that compromise transcription. If transcriptional stress is unresolved, arrested RNA polymerase can activate programmed cell death. Nonetheless, how this parasite modulates these processes is unknown. Here, we demonstrate that T. cruzi cell death after UV irradiation, a genotoxic agent that generates lesions resolved by TC-NER, depends on active transcription and is signaled mainly by an apoptotic-like pathway. Pre-treated parasites with α-amanitin, a selective RNA polymerase II inhibitor, become resistant to such cell death. Similarly, the gamma pre-irradiated cells are more resistant to UV when the transcription processes are absent. The Cockayne Syndrome B protein (CSB) recognizes blocked RNA polymerase and can initiate TC-NER. Curiously, CSB overexpression increases parasites' cell death shortly after UV exposure. On the other hand, at the same time after irradiation, the single-knockout CSB cells show resistance to the same treatment. UV-induced fast death is signalized by the exposition of phosphatidylserine to the outer layer of the membrane, indicating a cell death mainly by an apoptotic-like pathway. Furthermore, such death is suppressed in WT parasites pre-treated with inhibitors of ataxia telangiectasia and Rad3-related (ATR), a key DDR kinase. Signaling for UV radiation death may be related to R-loops since the overexpression of genes associated with the resolution of these structures suppress it. Together, results suggest that transcription blockage triggered by UV radiation activates an ATR-dependent apoptosis-like mechanism in T. cruzi, with the participation of CSB protein in this process.
Collapse
Affiliation(s)
- Isabela Cecilia Mendes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Willian Dos Reis Bertoldo
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Adalberto Sales Miranda-Junior
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Antônio Vinícius de Assis
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Bruno Marçal Repolês
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Wesley Roger Rodrigues Ferreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Daniela Ferreira Chame
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Daniela De Laet Souza
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Raphael Souza Pavani
- Laboratório Especial de Ciclo Celular, Instituto Butantan, São Paulo, MG, São Paulo, SP 05503-900, Brazil
| | - Andrea Mara Macedo
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Glória Regina Franco
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Esteban Serra
- Instituto de Biología Molecular y Celular de Rosario, CONICET, 2000 Rosario, Santa Fe, Argentina; Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, 2000 Rosario, Santa Fe, Argentina
| | - Virginia Perdomo
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, 2000 Rosario, Santa Fe, Argentina
| | - Carlos Frederico Martins Menck
- Departamento de Microbiologia, Instituto de Ciências Biomédicas (ICB), Universidade de São Paulo (USP), São Paulo, SP 05508-900, Brazil
| | - Giovana da Silva Leandro
- Departamento de Microbiologia, Instituto de Ciências Biomédicas (ICB), Universidade de São Paulo (USP), São Paulo, SP 05508-900, Brazil
| | | | | | - Carlos Renato Machado
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil.
| |
Collapse
|
8
|
Blanco CM, de Souza HADS, Martins PDC, Almeida-Silva J, Suarez-Fontes AM, Chaves YO, Vannier-Santos MA, Pratt-Riccio LR, Daniel-Ribeiro CT, Lopes SCP, Totino PRR. Cell Death of P. vivax Blood Stages Occurs in Absence of Classical Apoptotic Events and Induces Eryptosis of Parasitized Host Cells. Pathogens 2024; 13:673. [PMID: 39204273 PMCID: PMC11357032 DOI: 10.3390/pathogens13080673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/28/2024] [Accepted: 08/06/2024] [Indexed: 09/03/2024] Open
Abstract
Elucidation of pathways regulating parasite cell death is believed to contribute to identification of novel therapeutic targets for protozoan diseases, and in this context, apoptosis-like cell death has been reported in different groups of protozoa, in which metacaspases seem to play a role. In the genus Plasmodium, apoptotic markers have been detected in P. falciparum and P. berghei, and no study focusing on P. vivax cell death has been reported so far. In the present study, we investigated the susceptibility of P. vivax to undergo apoptotic cell death after incubating mature trophozoites with the classical apoptosis inducer staurosporine. As assessed by flow cytometry assays, staurosporine inhibited parasite intraerythrocytic development, which was accompanied by a decrease in cell viability, evidenced by reduced plasmodial mitochondrial activity. However, typical signs of apoptosis, such as DNA fragmentation, chromatin condensation, and nuclear segregation, were not detected in the parasites induced to cell death, and no significant alteration in metacaspase gene expression (PvMCA1) was observed under cell death stimulus. Interestingly, dying parasites positively modulated cell death (eryptosis) of host erythrocytes, which was marked by externalization of phosphatidylserine and cell shrinkage. Our study shows for the time that P. vivax blood stages may not be susceptible to apoptosis-like processes, while they could trigger eryptosis of parasitized cells by undergoing cell death. Further studies are required to elucidate the cellular machinery involved in cell death of P. vivax parasites as well as in the modulation of host cell death.
Collapse
Affiliation(s)
- Carolina Moreira Blanco
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz, Fiocruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal), Secretaria de Vigilância em Saúde e Ambiente (SVSA), Ministério da Saúde, Rio de Janeiro 21040-360, Brazil; (C.M.B.); (H.A.d.S.d.S.); (P.d.C.M.); (L.R.P.-R.); (C.T.D.-R.)
| | - Hugo Amorim dos Santos de Souza
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz, Fiocruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal), Secretaria de Vigilância em Saúde e Ambiente (SVSA), Ministério da Saúde, Rio de Janeiro 21040-360, Brazil; (C.M.B.); (H.A.d.S.d.S.); (P.d.C.M.); (L.R.P.-R.); (C.T.D.-R.)
| | - Priscilla da Costa Martins
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz, Fiocruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal), Secretaria de Vigilância em Saúde e Ambiente (SVSA), Ministério da Saúde, Rio de Janeiro 21040-360, Brazil; (C.M.B.); (H.A.d.S.d.S.); (P.d.C.M.); (L.R.P.-R.); (C.T.D.-R.)
| | - Juliana Almeida-Silva
- Laboratório de Inovações em Terapia, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, Brazil; (J.A.-S.); (M.A.V.-S.)
| | - Ana Marcia Suarez-Fontes
- Laboratório de Inovações em Terapia, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, Brazil; (J.A.-S.); (M.A.V.-S.)
| | - Yury Oliveira Chaves
- Instituto Leônidas e Maria Deane, Fiocruz Amazônia, Manaus 69057-070, Brazil; (Y.O.C.); (S.C.P.L.)
| | - Marcos André Vannier-Santos
- Laboratório de Inovações em Terapia, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, Brazil; (J.A.-S.); (M.A.V.-S.)
| | - Lilian Rose Pratt-Riccio
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz, Fiocruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal), Secretaria de Vigilância em Saúde e Ambiente (SVSA), Ministério da Saúde, Rio de Janeiro 21040-360, Brazil; (C.M.B.); (H.A.d.S.d.S.); (P.d.C.M.); (L.R.P.-R.); (C.T.D.-R.)
| | - Cláudio Tadeu Daniel-Ribeiro
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz, Fiocruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal), Secretaria de Vigilância em Saúde e Ambiente (SVSA), Ministério da Saúde, Rio de Janeiro 21040-360, Brazil; (C.M.B.); (H.A.d.S.d.S.); (P.d.C.M.); (L.R.P.-R.); (C.T.D.-R.)
| | - Stefanie Costa Pinto Lopes
- Instituto Leônidas e Maria Deane, Fiocruz Amazônia, Manaus 69057-070, Brazil; (Y.O.C.); (S.C.P.L.)
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado (FMT-HVD), Manaus 69040-000, Brazil
| | - Paulo Renato Rivas Totino
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz, Fiocruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal), Secretaria de Vigilância em Saúde e Ambiente (SVSA), Ministério da Saúde, Rio de Janeiro 21040-360, Brazil; (C.M.B.); (H.A.d.S.d.S.); (P.d.C.M.); (L.R.P.-R.); (C.T.D.-R.)
| |
Collapse
|
9
|
Rivas F, Del Mármol C, Scalese G, Pérez Díaz L, Machado I, Blacque O, Salazar F, Coitiño EL, Benítez D, Medeiros A, Comini M, Gambino D. Multifunctional Organometallic Compounds Active against Infective Trypanosomes: Ru(II) Ferrocenyl Derivatives with Two Different Bioactive Ligands. Inorg Chem 2024; 63:11667-11687. [PMID: 38860314 DOI: 10.1021/acs.inorgchem.4c01125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
Human African trypanosomiasis (HAT, sleeping sickness) and American trypanosomiasis (Chagas disease) are endemic zoonotic diseases caused by genomically related trypanosomatid protozoan parasites (Trypanosoma brucei and Trypanosoma cruzi, respectively). Just a few old drugs are available for their treatment, with most of them sharing poor safety, efficacy, and pharmacokinetic profiles. Only fexinidazole has been recently incorporated into the arsenal for the treatment of HAT. In this work, new multifunctional Ru(II) ferrocenyl compounds were rationally designed as potential agents against these pathogens by including in a single molecule 1,1'-bis(diphenylphosphino)ferrocene (dppf) and two bioactive bidentate ligands: pyridine-2-thiolato-1-oxide ligand (mpo) and polypyridyl ligands (NN). Three [Ru(mpo)(dppf)(NN)](PF6) compounds and their derivatives with chloride as a counterion were synthesized and fully characterized in solid state and solution. They showed in vitro activity on bloodstream T. brucei (EC50 = 31-160 nM) and on T. cruzi trypomastigotes (EC50 = 190-410 nM). Compounds showed the lowest EC50 values on T. brucei when compared to the whole set of metal-based compounds previously developed by us. In addition, several of the Ru compounds showed good selectivity toward the parasites, particularly against the highly proliferative bloodstream form of T. brucei. Interaction with DNA and generation of reactive oxygen species (ROS) were ruled out as potential targets and modes of action of the Ru compounds. Biochemical assays and in silico analysis led to the insight that they are able to inhibit the NADH-dependent fumarate reductase from T. cruzi. One representative hit induced a mild oxidation of low molecular weight thiols in T. brucei. The compounds were stable for at least 72 h in two different media and more lipophilic than both bioactive ligands, mpo and NN. An initial assessment of the therapeutic efficacy of one of the most potent and selective candidates, [Ru(mpo)(dppf)(bipy)]Cl, was performed using a murine infection model of acute African trypanosomiasis. This hit compound lacks acute toxicity when applied to animals in the dose/regimen described, but was unable to control parasite proliferation in vivo, probably because of its rapid clearance or low biodistribution in the extracellular fluids. Future studies should investigate the pharmacokinetics of this compound in vivo and involve further research to gain deeper insight into the mechanism of action of the compounds.
Collapse
Affiliation(s)
- Feriannys Rivas
- Área Química Inorgánica, Facultad de Química, Universidad de la República, 11800 Montevideo, Uruguay
| | - Carolina Del Mármol
- Área Química Inorgánica, Facultad de Química, Universidad de la República, 11800 Montevideo, Uruguay
| | - Gonzalo Scalese
- Área Química Inorgánica, Facultad de Química, Universidad de la República, 11800 Montevideo, Uruguay
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
| | - Leticia Pérez Díaz
- Sección Genómica Funcional, Facultad de Ciencias, Universidad de la República, 11400 Montevideo, Uruguay
| | - Ignacio Machado
- Área Química Analítica, Facultad de Química, Universidad de la República, 11800 Montevideo, Uruguay
| | - Olivier Blacque
- Department of Chemistry, University of Zurich, CH 8057 Zurich, Switzerland
| | - Fabiana Salazar
- Laboratorio de Química Teórica y Computacional (LQTC), Instituto de Química Biológica, Facultad de Ciencias, and Centro de Investigaciones Biomédicas (CeInBio), Universidad de la República, 11400 Montevideo, Uruguay
| | - E Laura Coitiño
- Laboratorio de Química Teórica y Computacional (LQTC), Instituto de Química Biológica, Facultad de Ciencias, and Centro de Investigaciones Biomédicas (CeInBio), Universidad de la República, 11400 Montevideo, Uruguay
| | - Diego Benítez
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
| | - Andrea Medeiros
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, 11800 Montevideo, Uruguay
| | - Marcelo Comini
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
| | - Dinorah Gambino
- Área Química Inorgánica, Facultad de Química, Universidad de la República, 11800 Montevideo, Uruguay
| |
Collapse
|
10
|
Bethencourt-Estrella CJ, López-Arencibia A, Lorenzo-Morales J, Piñero JE. Global Health Priority Box: Discovering Flucofuron as a Promising Antikinetoplastid Compound. Pharmaceuticals (Basel) 2024; 17:554. [PMID: 38794125 PMCID: PMC11123942 DOI: 10.3390/ph17050554] [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: 02/09/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
Leishmaniasis, produced by Leishmania spp., and Chagas disease, produced by Trypanosoma cruzi, affect millions of people around the world. The treatments for these pathologies are not entirely effective and produce some side effects. For these reasons, it is necessary to develop new therapies that are more active and less toxic for patients. Some initiatives, such as the one carried out by the Medicines for Malaria Venture, allow for the screening of a large number of compounds of different origins to find alternatives to the lack of trypanocide treatments. In this work, 240 compounds were tested from the Global Health Priority Box (80 compounds with confirmed activity against drug-resistant malaria, 80 compounds for screening against neglected and zoonotic diseases and diseases at risk of drug resistance, and 80 compounds with activity against various vector species) against Trypanosoma cruzi and Leishmania amazonensis. Flucofuron, a compound with activity against vectors and with previous activity reported against Staphylococcus spp. and Schistosoma spp., demonstrates activity against L. amazonensis and T. cruzi and produces programmed cell death in the parasites. Flucofuron seems to be a good candidate for continuing study and proving its use as a trypanocidal agent.
Collapse
Affiliation(s)
- Carlos J. Bethencourt-Estrella
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203 La Laguna, Spain;
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38203 La Laguna, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Atteneri López-Arencibia
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203 La Laguna, Spain;
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38203 La Laguna, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jacob Lorenzo-Morales
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203 La Laguna, Spain;
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38203 La Laguna, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José E. Piñero
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203 La Laguna, Spain;
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38203 La Laguna, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
11
|
Bethencourt-Estrella CJ, Delgado-Hernández S, López-Arencibia A, San Nicolás-Hernández D, Salazar-Villatoro L, Omaña-Molina M, Tejedor D, García-Tellado F, Lorenzo-Morales J, Piñero JE. Acrylonitrile derivatives: In vitro activity and mechanism of cell death induction against Trypanosoma cruzi and Leishmania amazonensis. Int J Parasitol Drugs Drug Resist 2024; 24:100531. [PMID: 38484645 PMCID: PMC10950693 DOI: 10.1016/j.ijpddr.2024.100531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/16/2024] [Accepted: 02/27/2024] [Indexed: 03/24/2024]
Abstract
Leishmaniasis and Chagas disease are parasitic infections that affect millions of people worldwide, producing thousands of deaths per year. The current treatments against these pathologies are not totally effective and produce some side effects in the patients. Acrylonitrile derivatives are a group of compounds that have shown activity against these two diseases. In this work, four novels synthetic acrylonitriles were evaluated against the intracellular form and extracellular forms of L. amazonensis and T. cruzi. The compounds 2 and 3 demonstrate to have good selectivity indexes against both parasites, specifically the compound 3 against the amastigote form (SI = 6 against L. amazonensis and SI = 7.4 against T. cruzi). In addition, the parasites treated with these two compounds demonstrate to produce a programmed cell death, since they were positive for the events studied related to this type of death, including chromatin condensation, accumulation of reactive oxygen species and alteration of the mitochondrial membrane potential. In conclusion, this work confirms that acrylonitriles is a source of possible new compounds against kinetoplastids, however, more studies are needed to corroborate this activity.
Collapse
Affiliation(s)
- Carlos J Bethencourt-Estrella
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203 La Laguna, Islas Canarias, Tenerife, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, Islas Canarias, Tenerife, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Samuel Delgado-Hernández
- Instituto de Productos Naturales y Agrobiología, Consejo Superior de Investigaciones Científicas, Avda. Fco. Sánchez 3, 38206 La Laguna, Islas Canarias, Tenerife, Spain
| | - Atteneri López-Arencibia
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203 La Laguna, Islas Canarias, Tenerife, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, Islas Canarias, Tenerife, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Desirée San Nicolás-Hernández
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203 La Laguna, Islas Canarias, Tenerife, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, Islas Canarias, Tenerife, Spain
| | - Lizbeth Salazar-Villatoro
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico 07360, Mexico
| | - Maritza Omaña-Molina
- Facultad de Estudios Superiores Iztacala, Medicina, Universidad Nacional Autónoma de México (UNAM), Tlalnepantla 54090, Mexico
| | - David Tejedor
- Instituto de Productos Naturales y Agrobiología, Consejo Superior de Investigaciones Científicas, Avda. Fco. Sánchez 3, 38206 La Laguna, Islas Canarias, Tenerife, Spain.
| | - Fernando García-Tellado
- Instituto de Productos Naturales y Agrobiología, Consejo Superior de Investigaciones Científicas, Avda. Fco. Sánchez 3, 38206 La Laguna, Islas Canarias, Tenerife, Spain
| | - Jacob Lorenzo-Morales
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203 La Laguna, Islas Canarias, Tenerife, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, Islas Canarias, Tenerife, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - José E Piñero
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203 La Laguna, Islas Canarias, Tenerife, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, Islas Canarias, Tenerife, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| |
Collapse
|
12
|
Selener MG, Borgo J, Sarratea MB, Delfino MA, Laurella LC, Cerny N, Gomez J, Coll M, Malchiodi EL, Bivona AE, Barrera P, Redko FC, Catalán CAN, Alberti AS, Sülsen VP. Trypanocidal and Anti-Inflammatory Effects of Three ent-Kaurane Diterpenoids from Gymnocoronis spilanthoides var. subcordata (Asteraceae). Pharmaceutics 2024; 16:415. [PMID: 38543309 PMCID: PMC10975076 DOI: 10.3390/pharmaceutics16030415] [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: 01/03/2024] [Revised: 02/24/2024] [Accepted: 02/27/2024] [Indexed: 01/03/2025] Open
Abstract
Chagas disease, caused by the protozoan Trypanosoma cruzi, affects 6-7 million people worldwide. The dichloromethane extract obtained from the aerial parts of Gymnocoronis spilanthoides var subcordata showed trypanocidal activity in vitro. The fractionation of the dewaxed organic extract via column chromatography led to the isolation of three diterpenoids: ent-9α,11α-dihydroxy-15-oxo-kaur-16-en-19-oic acid or adenostemmoic acid B, (16R)-ent-11α-hydroxy-15-oxokauran-19-oic acid and ent-11α-hydroxy-15-oxo-kaur-16-en-19-oic acid. These compounds showed IC50 values of 10.6, 15.9 and 4.8 µM against T. cruzi epimastigotes, respectively. When tested against amastigotes, the diterpenoids afforded IC50 values of 6.1, 19.5 and 60.6 µM, respectively. The cytotoxicity of the compounds was tested on mammalian cells using an MTT assay, resulting in CC50s of 321.8, 23.3 and 14.8 µM, respectively. The effect of adenostemmoic acid B on T. cruzi was examined at the ultrastructural level using transmission microscopy. Treatment with 20 μM for 48 h stimulated the formation of abnormal cytosolic membranous structures in the parasite. This compound also showed an anti-inflammatory effect in murine macrophages stimulated with LPS and other TLR agonists. Treatment of macrophages with adenostemmoic acid B was able to reduce TNF secretion and nitric oxide production, while increasing IL-10 production. The combination of adenostemmoic acid B with benznidazole resulted in greater inhibition of NF-kB and a decrease in nitrite concentration. The administration of adenostemmoic acid B to mice infected with trypomastigotes of T. cruzi at the dose of 1 mg/kg/day for five days produced a significant decrease in parasitemia levels and weight loss. Treatment with the association with benznidazole increased the survival time of the animals. In view of these results, adenostemmoic acid B could be considered a promising candidate for further studies in the search for new treatments for Chagas disease.
Collapse
Affiliation(s)
- Mariana G. Selener
- Cátedra de Farmacognosia, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Piso 2, Buenos Aires C1113AAD, Argentina; (M.G.S.); (J.B.); (L.C.L.); (F.C.R.)
- Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), CONICET-Universidad de Buenos Aires, Junín 956, Piso 2, Buenos Aires C1113AAD, Argentina
| | - Jimena Borgo
- Cátedra de Farmacognosia, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Piso 2, Buenos Aires C1113AAD, Argentina; (M.G.S.); (J.B.); (L.C.L.); (F.C.R.)
- Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), CONICET-Universidad de Buenos Aires, Junín 956, Piso 2, Buenos Aires C1113AAD, Argentina
| | - Maria Belen Sarratea
- Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET-Universidad de Buenos Aires, Junín 956, Piso 4, Buenos Aires C1113AAD, Argentina; (M.B.S.); (E.L.M.); (A.E.B.)
| | - Maria Alicia Delfino
- Cátedra de Inmunología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Piso 4, Buenos Aires C1113AAD, Argentina;
| | - Laura C. Laurella
- Cátedra de Farmacognosia, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Piso 2, Buenos Aires C1113AAD, Argentina; (M.G.S.); (J.B.); (L.C.L.); (F.C.R.)
- Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), CONICET-Universidad de Buenos Aires, Junín 956, Piso 2, Buenos Aires C1113AAD, Argentina
| | - Natacha Cerny
- Departamento de Microbiología, Parasitología e Inmunología-IMPAM (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, Buenos Aires C1121ABG, Argentina;
| | - Jessica Gomez
- Facultad de Ciencias Médicas, Instituto de Histología y Embriología “Dr. Mario H. Burgos” (IHEM), Universidad Nacional de Cuyo-CONICET, CC 56, Mendoza 5500, Argentina; (J.G.); (M.C.); (P.B.)
| | - Mauro Coll
- Facultad de Ciencias Médicas, Instituto de Histología y Embriología “Dr. Mario H. Burgos” (IHEM), Universidad Nacional de Cuyo-CONICET, CC 56, Mendoza 5500, Argentina; (J.G.); (M.C.); (P.B.)
| | - Emilio L. Malchiodi
- Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET-Universidad de Buenos Aires, Junín 956, Piso 4, Buenos Aires C1113AAD, Argentina; (M.B.S.); (E.L.M.); (A.E.B.)
- Cátedra de Inmunología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Piso 4, Buenos Aires C1113AAD, Argentina;
| | - Augusto E. Bivona
- Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET-Universidad de Buenos Aires, Junín 956, Piso 4, Buenos Aires C1113AAD, Argentina; (M.B.S.); (E.L.M.); (A.E.B.)
- Cátedra de Inmunología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Piso 4, Buenos Aires C1113AAD, Argentina;
| | - Patricia Barrera
- Facultad de Ciencias Médicas, Instituto de Histología y Embriología “Dr. Mario H. Burgos” (IHEM), Universidad Nacional de Cuyo-CONICET, CC 56, Mendoza 5500, Argentina; (J.G.); (M.C.); (P.B.)
| | - Flavia C. Redko
- Cátedra de Farmacognosia, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Piso 2, Buenos Aires C1113AAD, Argentina; (M.G.S.); (J.B.); (L.C.L.); (F.C.R.)
- Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), CONICET-Universidad de Buenos Aires, Junín 956, Piso 2, Buenos Aires C1113AAD, Argentina
| | - César A. N. Catalán
- Instituto de Química Orgánica, Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Ayacucho 471, San Miguel de Tucumán T4000INI, Argentina;
| | - Andrés Sánchez Alberti
- Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET-Universidad de Buenos Aires, Junín 956, Piso 4, Buenos Aires C1113AAD, Argentina; (M.B.S.); (E.L.M.); (A.E.B.)
- Departamento de Microbiología, Parasitología e Inmunología-IMPAM (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, Buenos Aires C1121ABG, Argentina;
| | - Valeria P. Sülsen
- Cátedra de Farmacognosia, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Piso 2, Buenos Aires C1113AAD, Argentina; (M.G.S.); (J.B.); (L.C.L.); (F.C.R.)
- Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), CONICET-Universidad de Buenos Aires, Junín 956, Piso 2, Buenos Aires C1113AAD, Argentina
| |
Collapse
|
13
|
James MR, Aufiero MA, Vesely EM, Dhingra S, Liu KW, Hohl TM, Cramer RA. Aspergillus fumigatus cytochrome c impacts conidial survival during sterilizing immunity. mSphere 2023; 8:e0030523. [PMID: 37823656 PMCID: PMC10871163 DOI: 10.1128/msphere.00305-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/29/2023] [Indexed: 10/13/2023] Open
Abstract
IMPORTANCE Aspergillus fumigatus can cause a life-threatening infection known as invasive pulmonary aspergillosis (IPA), which is marked by fungus-attributable mortality rates of 20%-30%. Individuals at risk for IPA harbor genetic mutations or incur pharmacologic defects that impair myeloid cell numbers and/or function, exemplified by bone marrow transplant recipients, patients that receive corticosteroid therapy, or patients with chronic granulomatous disease (CGD). However, treatments for Aspergillus infections remain limited, and resistance to the few existing drug classes is emerging. Recently, the World Health Organization classified A. fumigatus as a critical priority fungal pathogen. Our cell death research identifies an important aspect of fungal biology that impacts susceptibility to leukocyte killing. Furthering our understanding of mechanisms that mediate the outcome of fungal-leukocyte interactions will increase our understanding of both the underlying fungal biology governing cell death and innate immune evasion strategies utilized during mammalian infection pathogenesis. Consequently, our studies are a critical step toward leveraging these mechanisms for novel therapeutic advances.
Collapse
Affiliation(s)
- Matthew R. James
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth, Hanover, New Hampshire, USA
| | - Mariano A. Aufiero
- Louis V Gerstner Jr. Graduate School of Biomedical Sciences, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Elisa M. Vesely
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth, Hanover, New Hampshire, USA
| | - Sourabh Dhingra
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth, Hanover, New Hampshire, USA
| | - Ko-Wei Liu
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth, Hanover, New Hampshire, USA
| | - Tobias M. Hohl
- Louis V Gerstner Jr. Graduate School of Biomedical Sciences, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Infectious Disease Service, Department of Medicine, Memorial Hospital, New York, New York, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Robert A. Cramer
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth, Hanover, New Hampshire, USA
| |
Collapse
|
14
|
Gong Z, Mao W, Jin F, Zhang S, Zhao J, Ren P, Yu Z, Bai Y, Wang C, Cao J, Liu B. Prostaglandin D 2 regulates Escherichia coli-induced inflammatory responses through TLR2, TLR4, and NLRP3 in macrophages. Prostaglandins Other Lipid Mediat 2023; 169:106772. [PMID: 37669705 DOI: 10.1016/j.prostaglandins.2023.106772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 08/27/2023] [Accepted: 08/29/2023] [Indexed: 09/07/2023]
Abstract
Prostaglandin D2 (PGD2) synthesis is closely associated with the innate immune response mediated by pattern recognition receptors (PPRs). We determined PGD2 synthesis whether mediated by Toll-like receptor 2 (TLR2), TLR4 and Nod-like receptor pyrin domain-containing protein 3 (NLRP3) in Escherichia coli (E. coli)-, lipopolysaccharide (LPS)- and Braun lipoprotein (BLP)-stimulated macrophages. Our data demonstrate that TLR2, TLR4, and NLRP3 could regulate the synthesis of PGD2 through cyclo-oxygenase-2 (COX-2) and hematopoietic PGD synthase (H-PGDS) in E. coli-, LPS- or BLP-stimulated macrophages, suggesting that TLR2, TLR4, and NLRP3 are critical in regulating PGD2 secretion by controlling PGD2 synthetase expression in E. coli-, LPS- or BLP-stimulated macrophages. The H-PGDS (a PGD2 specific synthase) inhibitor pre-treatment could down-regulate the secretion of TNF-α, RANTES and IL-10 in LPS- and E. coli-stimulated macrophage. Meanwhile, H-PGDS inhibitor could down-regulate the secretion of TNF-α, while up-regulated RANTES and IL-10 secretion in BLP-stimulated macrophages, suggesting that PGD2 could regulate the secretion of cytokines and chemokines in E. coli-, LPS- or BLP-stimulated macrophages. Furthermore, exogenous PGD2 regulates the secretion of cytokines and chemokines through activation of MAPK and NF-κB signaling pathways after E. coli-, LPS- or BLP stimulation in macrophages. Taken together, PGD2 is found able to regulate E. coli-induced inflammatory responses through TLR2, TLR4, and NLRP3 in macrophages.
Collapse
Affiliation(s)
- Zhiguo Gong
- Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot, China
| | - Wei Mao
- Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot, China
| | - Feng Jin
- Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot, China
| | - Shuangyi Zhang
- Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot, China
| | - Jiamin Zhao
- Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot, China
| | - Peipei Ren
- Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot, China
| | - Zhuoya Yu
- Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot, China
| | - Yunjie Bai
- Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot, China
| | - Chao Wang
- Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot, China
| | - Jinshan Cao
- Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot, China.
| | - Bo Liu
- Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011 Hohhot, China.
| |
Collapse
|
15
|
Utage B, Patole M, Nagvenkar P, Gacche R. Prosopis juliflora (Sw.) DC.induces apoptotic-like programmed cell death in Leishmania donovani via over production of oxidative stress, mitochondrial dysfunction and ATP depletion. J Tradit Complement Med 2023; 13:611-622. [PMID: 38020554 PMCID: PMC10658441 DOI: 10.1016/j.jtcme.2023.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 05/03/2023] [Accepted: 06/29/2023] [Indexed: 12/01/2023] Open
Abstract
Background Leishmaniasis is endemic in more than 60 countries with a large number of mortality cases. The current chemotherapy approaches employed for managing the leishmaniasis is associated with severe side effects. Therefore there is a need to develop effective, safe, and cost affordable antileishmanial drug candidates. Purpose of the study This study was designed to evaluate the in vitro antileishmanial activity of a Prosopis juliflora leaves extract (PJLME) towards the Leishmania donovani parasites. Material and methods PJLME was evaluated for its cytotoxicity against the L. donovani parasites and the mouse macrophage cells. Further, various in vitro experiments like ROS assay, mitochondrial membrane potential assay, annexin v assay, cell cycle assay, and caspase 3/7 assay were performed to understand the mechanism of cell death. Phytochemical profiling of P. juliflorawas performed by utilizing HPTLC and GC-MS analysis. Results PJLME demonstrated antileishmanial activity at a remarkably lower concentration of IC50 6.5 μg/mL. Of note, interestingly PJLME IC50 concentration has not demonstrated cytotoxicity against the mouse macrophage cell line. Performed experiments confirmed ROS inducing potential of PJLME which adversely affected the mitochondrial membrane potential and caused loss of mitochondrial membrane potential and thereby ATP levels. PJLME also arrested the cell cycle and induced apoptotic-like cell death in PJLME treated L. donovani promastigotes. Conclusion The results clearly established the significance of Prosopis juliflora as an effective and safe natural resource for managing visceral leishmaniasis. The findings can be used as a baseline reference for developing novel leads/formulations for effective management of visceral leishmaniasis.
Collapse
Affiliation(s)
| | - Milind Patole
- National Centre for Cell Science, NCCS Complex, Pune, 411007, MS, India
| | - Punam Nagvenkar
- National Centre for Cell Science, NCCS Complex, Pune, 411007, MS, India
| | - Rajesh Gacche
- Department of Biotechnology, Savitribai Phule Pune University, Pune, 411007, MS, India
| |
Collapse
|
16
|
Pereira PML, Fernandes BT, dos Santos VR, Cabral WRC, Lovo-Martins MI, Alonso L, Lancheros CAC, de Paula JC, Camargo PG, Suzukawa HT, Alonso A, Macedo F, Nakamura CV, Tavares ER, de Lima Ferreira Bispo M, Yamauchi LM, Pinge-Filho P, Yamada-Ogatta SF. Antiprotozoal Activity of Benzoylthiourea Derivatives against Trypanosoma cruzi: Insights into Mechanism of Action. Pathogens 2023; 12:1012. [PMID: 37623972 PMCID: PMC10457850 DOI: 10.3390/pathogens12081012] [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: 06/30/2023] [Revised: 07/30/2023] [Accepted: 08/01/2023] [Indexed: 08/26/2023] Open
Abstract
For decades, only two nitroheterocyclic drugs have been used as therapeutic agents for Chagas disease. However, these drugs present limited effectiveness during the chronic phase, possess unfavorable pharmacokinetic properties, and induce severe adverse effects, resulting in low treatment adherence. A previous study reported that N-(cyclohexylcarbamothioyl) benzamide (BTU-1), N-(tert-butylcarbamothioyl) benzamide (BTU-2), and (4-bromo-N-(3-nitrophenyl) carbamothioyl benzamide (BTU-3) present selective antiprotozoal activity against all developmental forms of Trypanosoma cruzi Y strain. In this study, we investigated the mechanism of action of these compounds through microscopy and biochemical analyses. Transmission electron microscopy analysis showed nuclear disorganization, changes in the plasma membrane with the appearance of blebs and extracellular arrangements, intense vacuolization, mitochondrial swelling, and formation of myelin-like structures. Biochemical results showed changes in the mitochondrial membrane potential, reactive oxygen species content, lipid peroxidation, and plasma membrane fluidity. In addition, the formation of autophagic vacuoles was observed. These findings indicate that BTU-1, BTU-2, and BTU-3 induced profound morphological, ultrastructural, and biochemical alterations in epimastigote forms, triggering an autophagic-dependent cell death pathway.
Collapse
Affiliation(s)
- Patrícia Morais Lopes Pereira
- Graduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (P.M.L.P.); (B.T.F.); (W.R.C.C.); (H.T.S.); (P.P.-F.)
- Laboratory of Molecular Biology of Microorganisms, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (V.R.d.S.); (E.R.T.)
| | - Bruna Terci Fernandes
- Graduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (P.M.L.P.); (B.T.F.); (W.R.C.C.); (H.T.S.); (P.P.-F.)
- Laboratory of Molecular Biology of Microorganisms, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (V.R.d.S.); (E.R.T.)
| | - Vitória Ribeiro dos Santos
- Laboratory of Molecular Biology of Microorganisms, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (V.R.d.S.); (E.R.T.)
| | - Weslei Roberto Correia Cabral
- Graduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (P.M.L.P.); (B.T.F.); (W.R.C.C.); (H.T.S.); (P.P.-F.)
- Laboratory of Molecular Biology of Microorganisms, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (V.R.d.S.); (E.R.T.)
| | - Maria Isabel Lovo-Martins
- Laboratory of Experimental Immunopathology, Department of Immunology, Parasitology and General Pathology, State University of Londrina, Londrina 86057-970, Brazil;
| | - Lais Alonso
- Institute of Physics, Federal University of Goiás, Goiania 74690-900, Brazil; (L.A.); (A.A.)
| | | | | | - Priscila Goes Camargo
- Laboratory of Medicinal Molecules Synthesis, Department of Chemistry, State University of Londrina, Londrina 86057-970, Brazil; (P.G.C.); (M.d.L.F.B.)
| | - Helena Tiemi Suzukawa
- Graduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (P.M.L.P.); (B.T.F.); (W.R.C.C.); (H.T.S.); (P.P.-F.)
- Laboratory of Molecular Biology of Microorganisms, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (V.R.d.S.); (E.R.T.)
| | - Antônio Alonso
- Institute of Physics, Federal University of Goiás, Goiania 74690-900, Brazil; (L.A.); (A.A.)
| | - Fernando Macedo
- Laboratory of Medicinal Molecules Synthesis, Department of Chemistry, State University of Londrina, Londrina 86057-970, Brazil; (P.G.C.); (M.d.L.F.B.)
| | - Celso Vataru Nakamura
- Laboratory of Technological Innovation in the Development of Drugs and Cosmetics, Department of Basic Health Sciences, State University of Maringá, Maringa 87020-900, Brazil;
| | - Eliandro Reis Tavares
- Laboratory of Molecular Biology of Microorganisms, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (V.R.d.S.); (E.R.T.)
| | - Marcelle de Lima Ferreira Bispo
- Laboratory of Medicinal Molecules Synthesis, Department of Chemistry, State University of Londrina, Londrina 86057-970, Brazil; (P.G.C.); (M.d.L.F.B.)
| | - Lucy Megumi Yamauchi
- Graduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (P.M.L.P.); (B.T.F.); (W.R.C.C.); (H.T.S.); (P.P.-F.)
- Laboratory of Molecular Biology of Microorganisms, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (V.R.d.S.); (E.R.T.)
| | - Phileno Pinge-Filho
- Graduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (P.M.L.P.); (B.T.F.); (W.R.C.C.); (H.T.S.); (P.P.-F.)
- Laboratory of Experimental Immunopathology, Department of Immunology, Parasitology and General Pathology, State University of Londrina, Londrina 86057-970, Brazil;
| | - Sueli Fumie Yamada-Ogatta
- Graduate Program in Microbiology, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (P.M.L.P.); (B.T.F.); (W.R.C.C.); (H.T.S.); (P.P.-F.)
- Laboratory of Molecular Biology of Microorganisms, Department of Microbiology, State University of Londrina, Londrina 86057-970, Brazil; (V.R.d.S.); (E.R.T.)
| |
Collapse
|
17
|
James MR, Aufiero MA, Vesely EM, Dhingra S, Liu KW, Hohl TM, Cramer RA. Aspergillus fumigatus cytochrome c impacts conidial survival during sterilizing immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.07.544103. [PMID: 37333187 PMCID: PMC10274773 DOI: 10.1101/2023.06.07.544103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Invasive pulmonary aspergillosis (IPA) is a life-threatening infection caused by species in the ubiquitous fungal genus Aspergillus . While leukocyte-generated reactive oxygen species (ROS) are critical for the clearance of fungal conidia from the lung and resistance to IPA, the processes that govern ROS-dependent fungal cell death remain poorly defined. Using a flow cytometric approach that monitors two independent cell death markers, an endogenous histone H2A:mRFP nuclear integrity reporter and Sytox Blue cell impermeable (live/dead) stain, we observed that loss of A. fumigatus cytochrome c ( cycA ) results in reduced susceptibility to cell death from hydrogen peroxide (H 2 O 2 ) treatment. Consistent with these observations in vitro , loss of cycA confers resistance to both NADPH-oxidase -dependent and -independent killing by host leukocytes. Fungal ROS resistance is partly mediated in part by Bir1, a homolog to survivin in humans, as Bir1 overexpression results in decreased ROS-induced conidial cell death and reduced killing by innate immune cells in vivo . We further report that overexpression of the Bir1 N-terminal BIR domain in A. fumigatus conidia results in altered expression of metabolic genes that functionally converge on mitochondrial function and cytochrome c ( cycA ) activity. Together, these studies demonstrate that cycA in A. fumigatus contributes to cell death responses that are induced by exogenous H 2 O 2 and by host leukocytes. Importance Aspergillus fumigatus can cause a life-threatening infection known as invasive pulmonary aspergillosis (IPA), which is marked by fungus-attributable mortality rates of 20%-30%. Individuals at risk of IPA harbor genetic mutations or incur pharmacologic defects that impair myeloid cell numbers and/or function, exemplified by bone marrow transplant recipients, patients that receive corticosteroid therapy, or patients with Chronic Granulomatous Disease (CGD). However, treatments for Aspergillus infections remains limited, and resistance to the few existing drug classes is emerging. Recently, the World Health Organization (WHO) classified A. fumigatus as a critical priority fungal pathogen. Our research identifies an important aspect of fungal biology that impacts susceptibility to leukocyte killing. Furthering our understanding of mechanisms that mediate the outcome of fungal-leukocyte interactions will increase our understanding of both the underlying fungal biology governing cell death and innate immune evasion strategies utilized during mammalian infection pathogenesis. Consequently, our studies are a critical step toward leveraging these mechanisms for novel therapeutic advances.
Collapse
|
18
|
Bethencourt-Estrella CJ, Delgado-Hernández S, López-Arencibia A, San Nicolás-Hernández D, Tejedor D, García-Tellado F, Lorenzo-Morales J, Piñero JE. In vitro activity and mechanism of cell death induction of cyanomethyl vinyl ethers derivatives against Trypanosoma cruzi. Int J Parasitol Drugs Drug Resist 2023; 22:72-80. [PMID: 37311268 PMCID: PMC10276036 DOI: 10.1016/j.ijpddr.2023.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 06/15/2023]
Abstract
Chagas disease causes a problematic pathology that can lead to megacolon and heart disease, and can even cause the death of the patient. Current therapies for this disease are the same as they were 50 years ago, are not fully effective and have strong side effects. The lack of a safe and effective therapy makes it necessary to search for new, less toxic and totally effective compounds against this parasite. In this work, the antichagasic activity of 46 novel cyanomethyl vinyl ether derivatives was studied. In addition, to elucidate the type of cell death that these compounds produce in parasites, several events related to programmed cell death were studied. The results highlight four more selective compounds, E63, E64, E74 and E83, which also appear to trigger programmed cell death, and are therefore postulated as good candidates to use in future therapeutics for Chagas disease.
Collapse
Affiliation(s)
- Carlos J Bethencourt-Estrella
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203, La Laguna, Tenerife, Islas Canarias, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, Tenerife, Islas Canarias, Spain.
| | - Samuel Delgado-Hernández
- Instituto de Productos Naturales y Agrobiología, Consejo Superior de Investigaciones Científicas, Avda. Fco. Sánchez 3, 38206, La Laguna, Tenerife, Islas Canarias, Spain; Departamento de Química, Unidad Departamental de Química Analítica, Universidad de La Laguna (ULL), Tenerife, 38206, Spain.
| | - Atteneri López-Arencibia
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203, La Laguna, Tenerife, Islas Canarias, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, Tenerife, Islas Canarias, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28220, Madrid, Spain.
| | - Desirée San Nicolás-Hernández
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203, La Laguna, Tenerife, Islas Canarias, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, Tenerife, Islas Canarias, Spain.
| | - David Tejedor
- Instituto de Productos Naturales y Agrobiología, Consejo Superior de Investigaciones Científicas, Avda. Fco. Sánchez 3, 38206, La Laguna, Tenerife, Islas Canarias, Spain.
| | - Fernando García-Tellado
- Instituto de Productos Naturales y Agrobiología, Consejo Superior de Investigaciones Científicas, Avda. Fco. Sánchez 3, 38206, La Laguna, Tenerife, Islas Canarias, Spain.
| | - Jacob Lorenzo-Morales
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203, La Laguna, Tenerife, Islas Canarias, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, Tenerife, Islas Canarias, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28220, Madrid, Spain.
| | - José E Piñero
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203, La Laguna, Tenerife, Islas Canarias, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, Tenerife, Islas Canarias, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28220, Madrid, Spain.
| |
Collapse
|
19
|
San Nicolás-Hernández D, Hernández-Álvarez E, Bethencourt-Estrella CJ, López-Arencibia A, Sifaoui I, Bazzocchi IL, Lorenzo-Morales J, Jiménez IA, Piñero JE. Multi-target withaferin-A analogues as promising anti-kinetoplastid agents through the programmed cell death. Biomed Pharmacother 2023; 164:114879. [PMID: 37210899 DOI: 10.1016/j.biopha.2023.114879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/09/2023] [Accepted: 05/12/2023] [Indexed: 05/23/2023] Open
Abstract
Leishmaniasis and Chagas disease, two of the most prevalent neglected tropical diseases, are a world health problem. The harsh reality of these infective diseases is the absence of effective and safe therapies. In this framework, natural products play an important role in overcoming the current need to development new antiparasitic agents. The present study reports the synthesis, antikinetoplastid screening, mechanism study of fourteen withaferin A derivatives (2-15). Nine of them (2-6, 8-10 and 12) showed a potent dose-dependent inhibitory effect on the proliferation of Leishmania amazonensis and L. donovani promastigotes and Trypanosoma cruzi epimastigotes with IC50 values ranging from 0.19 to 24.01 µM. Outstandingly, the fully acetylated derivative 10 (4,27-diacetylwithaferin A) was the most potent compound showing IC50 values of 0.36, 2.82 and 0.19 µM against L. amazonensis, L. donovani and T. cruzi, respectively. Furthermore, analogue 10 exhibited approximately 18 and 36-fold greater antikinetoplastid activity, on L. amazonensis and T. cruzi, than the reference drugs. The activity was accompanied by significantly lower cytotoxicity on the murine macrophage cell line. Moreover, compounds 2, 3, 5-7, 9 and 10 showed more potent activity than the reference drug against the intracellular amastigotes forms of L. amazonensis and T.cruzi, with a good selectivity index on a mammalian cell line. In addition, withaferin A analogues 3, 5-7, 9 and 10 induce programmed cell death through a process of apoptosis-like and autophagy. These results strengthen the anti-parasitic potential of withaferin A-related steroids against neglected tropical diseases caused by Leishmania spp. and T. cruzi parasites.
Collapse
Affiliation(s)
- Desirée San Nicolás-Hernández
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avenida Astrofísico Francisco Sánchez, S/N, 38203 La Laguna, Tenerife, Canary Islands, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38200 La Laguna, Tenerife, Spain
| | - Eduardo Hernández-Álvarez
- Instituto Universitario de Bio-Orgánica Antonio González, Departamento de Química Orgánica, Universidad de La Laguna, Avenida Astrofísico Francisco Sánchez 2, 38206 La Laguna, Tenerife, Canary Islands, Spain
| | - Carlos J Bethencourt-Estrella
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avenida Astrofísico Francisco Sánchez, S/N, 38203 La Laguna, Tenerife, Canary Islands, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38200 La Laguna, Tenerife, Spain
| | - Atteneri López-Arencibia
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avenida Astrofísico Francisco Sánchez, S/N, 38203 La Laguna, Tenerife, Canary Islands, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38200 La Laguna, Tenerife, Spain.
| | - Ines Sifaoui
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avenida Astrofísico Francisco Sánchez, S/N, 38203 La Laguna, Tenerife, Canary Islands, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38200 La Laguna, Tenerife, Spain
| | - Isabel L Bazzocchi
- Instituto Universitario de Bio-Orgánica Antonio González, Departamento de Química Orgánica, Universidad de La Laguna, Avenida Astrofísico Francisco Sánchez 2, 38206 La Laguna, Tenerife, Canary Islands, Spain
| | - Jacob Lorenzo-Morales
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avenida Astrofísico Francisco Sánchez, S/N, 38203 La Laguna, Tenerife, Canary Islands, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38200 La Laguna, Tenerife, Spain
| | - Ignacio A Jiménez
- Instituto Universitario de Bio-Orgánica Antonio González, Departamento de Química Orgánica, Universidad de La Laguna, Avenida Astrofísico Francisco Sánchez 2, 38206 La Laguna, Tenerife, Canary Islands, Spain.
| | - José E Piñero
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avenida Astrofísico Francisco Sánchez, S/N, 38203 La Laguna, Tenerife, Canary Islands, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38200 La Laguna, Tenerife, Spain
| |
Collapse
|
20
|
Ilu A, Chia MA, Cataldi TR, Labate CA, Ebiloma GU, Yusuf PO, Shuaibu MN, Balogun EO. Type-I like metalloproteinase in the venom of the West African saw-scaled carpet viper (Echis ocellatus) has anti-trypanosomal activity against African trypanosomes. Toxicon 2023; 229:107138. [PMID: 37127124 DOI: 10.1016/j.toxicon.2023.107138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/26/2023] [Accepted: 04/26/2023] [Indexed: 05/03/2023]
Abstract
African trypanosomiasis is an infectious disease caused by hemoparasites of the genus Trypanosoma and remains a major health problem in Africa - killing around 4000 people and animals worth an estimated $5 billion, annually. The absence of a vaccine and satisfactory drug against African trypanosomiasis (AT) necessitates the continued search for new chemotherapy options. Owing to the rich biochemical diversity in snake venom, it has recently become a source of therapeutic peptides that are being explored for the development of novel drug candidates for diverse ailments such as cancers and infectious diseases. To explore this, Echis ocellatus venom (EOV) was investigated for the presence of an anti-Trypanosoma factor, with the subsequent aim to isolate and identify it. Crude EOV was collected and tested in vitro on the bloodstream form (BSF) i.e. long and slender morphological form of Trypanosoma brucei and T. congolense. This initial testing was followed by a sequential anti-trypanosomal assay guided purification of EOV using ethanol precipitation, distillation, and ion exchange (IEX) chromatography to obtain the active trypanocidal component. The purified anti-Trypanosoma factor, estimated to be a 52-kDa protein on SDS-PAGE, was subjected to in-gel trypsin digestion and 2D RP HPLC-MS/MS to identify the protein. The anti-Trypanosoma factor was revealed to be a zinc-dependent metalloproteinase that contains the HEXXHXXGXXH adamalysin motif. This protein may provide a conceptual framework for the possible design of a safe and effective anti-trypanosomal peptide for the treatment of AT.
Collapse
Affiliation(s)
- Ameh Ilu
- Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria; Africa Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, Zaria, Nigeria
| | - Mathias A Chia
- Department of Botany, Ahmadu Bello University, Zaria, Nigeria
| | - Thais R Cataldi
- Department of Genetics, Laboratório Multiusuários Centralizado de Genômica Funcional Aplicada à Agropecuária e Agroenergia, University of Sao Paulo, Piracicaba, Brazil
| | - Carlos A Labate
- Department of Genetics, Laboratório Multiusuários Centralizado de Genômica Funcional Aplicada à Agropecuária e Agroenergia, University of Sao Paulo, Piracicaba, Brazil
| | - Godwin U Ebiloma
- School of Health & Life Sciences, Teesside University, Middlesbrough, United Kingdom
| | - Peter O Yusuf
- Department of Veterinary Pharmacology and Toxicology, Ahmadu Bello University, Zaria, Nigeria
| | - Mohammed N Shuaibu
- Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria; Africa Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, Zaria, Nigeria; Centre for Biotechnology Research and Training, Ahmadu Bello University, Zaria, Nigeria
| | - Emmanuel O Balogun
- Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria; Africa Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, Zaria, Nigeria; Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan; Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| |
Collapse
|
21
|
Nicolás-Hernández DS, Rodríguez-Expósito RL, López-Arencibia A, Bethencourt-Estrella CJ, Sifaoui I, Salazar-Villatoro L, Omaña-Molina M, Fernández JJ, Díaz-Marrero AR, Piñero JE, Lorenzo-Morales J. Meroterpenoids from Gongolaria abies-marina against Kinetoplastids: In Vitro Activity and Programmed Cell Death Study. Pharmaceuticals (Basel) 2023; 16:ph16040476. [PMID: 37111233 PMCID: PMC10146491 DOI: 10.3390/ph16040476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/16/2023] [Accepted: 03/18/2023] [Indexed: 04/29/2023] Open
Abstract
Leishmaniasis and Chagas disease affect millions of people worldwide. The available treatments against these parasitic diseases are limited and display multiple undesired effects. The brown alga belonging to the genus Gongolaria has been previously reported as a source of compounds with different biological activities. In a recent study from our group, Gongolaria abies-marine was proven to present antiamebic activity. Hence, this brown alga could be a promising source of interesting molecules for the development of new antiprotozoal drugs. In this study, four meroterpenoids were isolated and purified from a dichloromethane/ethyl acetate crude extract through a bioguided fractionation process targeting kinetoplastids. Moreover, the in vitro activity and toxicity were evaluated, and the induction of programmed cell death was checked in the most active and less toxic compounds, namely gongolarone B (2), 6Z-1'-methoxyamentadione (3) and 1'-methoxyamentadione (4). These meroterpenoids triggered mitochondrial malfunction, oxidative stress, chromatin condensation and alterations of the tubulin network. Furthermore, a transmission electron microscopy (TEM) image analysis showed that meroterpenoids (2-4) induced the formation of autophagy vacuoles and ER and Golgi complex disorganization. The obtained results demonstrated that the mechanisms of action at the cellular level of these compounds were able to induce autophagy as well as an apoptosis-like process in the treated parasites.
Collapse
Affiliation(s)
- Desirée San Nicolás-Hernández
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38206 La Laguna, Tenerife, Spain
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38206 La Laguna, Tenerife, Spain
- Red de Investigación Colaborativa en Enfermedades Tropicales (RICET), Instituto de Salud Carlos III, 28006 Madrid, Spain
| | - Rubén L Rodríguez-Expósito
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38206 La Laguna, Tenerife, Spain
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38206 La Laguna, Tenerife, Spain
- Red de Investigación Colaborativa en Enfermedades Tropicales (RICET), Instituto de Salud Carlos III, 28006 Madrid, Spain
| | - Atteneri López-Arencibia
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38206 La Laguna, Tenerife, Spain
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38206 La Laguna, Tenerife, Spain
- Red de Investigación Colaborativa en Enfermedades Tropicales (RICET), Instituto de Salud Carlos III, 28006 Madrid, Spain
| | - Carlos J Bethencourt-Estrella
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38206 La Laguna, Tenerife, Spain
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38206 La Laguna, Tenerife, Spain
- Red de Investigación Colaborativa en Enfermedades Tropicales (RICET), Instituto de Salud Carlos III, 28006 Madrid, Spain
| | - Ines Sifaoui
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38206 La Laguna, Tenerife, Spain
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38206 La Laguna, Tenerife, Spain
- Red de Investigación Colaborativa en Enfermedades Tropicales (RICET), Instituto de Salud Carlos III, 28006 Madrid, Spain
| | - Lizbeth Salazar-Villatoro
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico 07360, Mexico
| | - Maritza Omaña-Molina
- Facultad de Estudios Superiores Iztacala, Medicina, Universidad Nacional Autónoma de México (UNAM), Tlalnepantla 54090, Mexico
| | - José J Fernández
- Instituto Universitario de Bio-Orgánica Antonio González, Departamento de Química Orgánica, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez 3, 38206 La Laguna, Tenerife, Spain
| | - Ana R Díaz-Marrero
- Instituto Universitario de Bio-Orgánica Antonio González, Departamento de Química Orgánica, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez 3, 38206 La Laguna, Tenerife, Spain
- Instituto de Productos Naturales y Agrobiología, Consejo Superior de Investigaciones Científicas, Avda. Astrofísico Fco. Sánchez 3, 38206 La Laguna, Tenerife, Spain
| | - José E Piñero
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38206 La Laguna, Tenerife, Spain
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38206 La Laguna, Tenerife, Spain
- Red de Investigación Colaborativa en Enfermedades Tropicales (RICET), Instituto de Salud Carlos III, 28006 Madrid, Spain
- Consorcio Centro de Investigación Biomédica En Red (CIBER) de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28006 Madrid, Spain
| | - Jacob Lorenzo-Morales
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38206 La Laguna, Tenerife, Spain
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38206 La Laguna, Tenerife, Spain
- Red de Investigación Colaborativa en Enfermedades Tropicales (RICET), Instituto de Salud Carlos III, 28006 Madrid, Spain
- Consorcio Centro de Investigación Biomédica En Red (CIBER) de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28006 Madrid, Spain
| |
Collapse
|
22
|
Romano PS, Akematsu T, Besteiro S, Bindschedler A, Carruthers VB, Chahine Z, Coppens I, Descoteaux A, Alberto Duque TL, He CY, Heussler V, Le Roch KG, Li FJ, de Menezes JPB, Menna-Barreto RFS, Mottram JC, Schmuckli-Maurer J, Turk B, Tavares Veras PS, Salassa BN, Vanrell MC. Autophagy in protists and their hosts: When, how and why? AUTOPHAGY REPORTS 2023; 2:2149211. [PMID: 37064813 PMCID: PMC10104450 DOI: 10.1080/27694127.2022.2149211] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 11/15/2022] [Indexed: 03/12/2023]
Abstract
Pathogenic protists are a group of organisms responsible for causing a variety of human diseases including malaria, sleeping sickness, Chagas disease, leishmaniasis, and toxoplasmosis, among others. These diseases, which affect more than one billion people globally, mainly the poorest populations, are characterized by severe chronic stages and the lack of effective antiparasitic treatment. Parasitic protists display complex life-cycles and go through different cellular transformations in order to adapt to the different hosts they live in. Autophagy, a highly conserved cellular degradation process, has emerged as a key mechanism required for these differentiation processes, as well as other functions that are crucial to parasite fitness. In contrast to yeasts and mammals, protist autophagy is characterized by a modest number of conserved autophagy-related proteins (ATGs) that, even though, can drive the autophagosome formation and degradation. In addition, during their intracellular cycle, the interaction of these pathogens with the host autophagy system plays a crucial role resulting in a beneficial or harmful effect that is important for the outcome of the infection. In this review, we summarize the current state of knowledge on autophagy and other related mechanisms in pathogenic protists and their hosts. We sought to emphasize when, how, and why this process takes place, and the effects it may have on the parasitic cycle. A better understanding of the significance of autophagy for the protist life-cycle will potentially be helpful to design novel anti-parasitic strategies.
Collapse
Affiliation(s)
- Patricia Silvia Romano
- Laboratorio de Biología de Trypanosoma cruzi y de la célula hospedadora. Instituto de Histología y Embriología de Mendoza. Universidad Nacional de Cuyo. (IHEM-CONICET-UNCUYO). Facultad de Ciencias Médicas. Universidad Nacional de Cuyo. Av. Libertador 80 (5500), Mendoza, Argentina
| | - Takahiko Akematsu
- Department of Biosciences, College of Humanities and Sciences, Nihon University, Tokyo, Japan
| | | | | | - Vern B Carruthers
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Zeinab Chahine
- Department of Molecular, Cell and Systems Biology, University of California Riverside, CA, USA
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology. Department of Molecular Microbiology and Immunology. Johns Hopkins Malaria Research Institute. Johns Hopkins University Bloomberg School of Public Health. Baltimore 21205, MD, USA
| | - Albert Descoteaux
- Centre Armand-Frappier Santé Biotechnologie, Institut national de la recherche scientifique, Laval, QC
| | - Thabata Lopes Alberto Duque
- Autophagy Inflammation and Metabolism Center, University of New Mexico Health Sciences Center, Albuquerque, NM, USA; Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Cynthia Y He
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Volker Heussler
- Institute of Cell Biology.University of Bern. Baltzerstr. 4 3012 Bern
| | - Karine G Le Roch
- Department of Molecular, Cell and Systems Biology, University of California Riverside, CA, USA
| | - Feng-Jun Li
- Department of Biological Sciences, National University of Singapore, Singapore
| | | | | | - Jeremy C Mottram
- York Biomedical Research Institute, Department of Biology, University of York, York, UK
| | | | - Boris Turk
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, SI-1000 Ljubljana, Slovenia
| | - Patricia Sampaio Tavares Veras
- Laboratory of Host-Parasite Interaction and Epidemiology, Gonçalo Moniz Institute, Fiocruz-Bahia
- National Institute of Science and Technology of Tropical Diseases - National Council for Scientific Research and Development (CNPq)
| | - Betiana Nebai Salassa
- Laboratorio de Biología de Trypanosoma cruzi y de la célula hospedadora. Instituto de Histología y Embriología de Mendoza. Universidad Nacional de Cuyo. (IHEM-CONICET-UNCUYO). Facultad de Ciencias Médicas. Universidad Nacional de Cuyo. Av. Libertador 80 (5500), Mendoza, Argentina
| | - María Cristina Vanrell
- Laboratorio de Biología de Trypanosoma cruzi y de la célula hospedadora. Instituto de Histología y Embriología de Mendoza. Universidad Nacional de Cuyo. (IHEM-CONICET-UNCUYO). Facultad de Ciencias Médicas. Universidad Nacional de Cuyo. Av. Libertador 80 (5500), Mendoza, Argentina
| |
Collapse
|
23
|
Santos TAC, Silva KP, Souza GB, Alves PB, Menna-Barreto RFS, Scher R, Fernandes RPM. Chalcone Derivative Induces Flagellar Disruption and Autophagic Phenotype in Phytomonas serpens In Vitro. Pathogens 2023; 12:pathogens12030423. [PMID: 36986345 PMCID: PMC10051746 DOI: 10.3390/pathogens12030423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/03/2023] [Accepted: 03/05/2023] [Indexed: 03/11/2023] Open
Abstract
Phytomonas serpens is a trypanosomatid phytoparasite, found in a great variety of species, including tomato plants. It is a significant problem for agriculture, causing high economic loss. In order to reduce the vegetal infections, different strategies have been used. The biological activity of molecules obtained from natural sources has been widely investigated to treat trypanosomatids infections. Among these compounds, chalcones have been shown to have anti-parasitic and anti-inflammatory effects, being described as having a remarkable activity on trypanosomatids, especially in Leishmania species. Here, we evaluated the antiprotozoal activity of the chalcone derivative (NaF) on P. serpens promastigotes, while also assessing its mechanism of action. The results showed that treatment with the derivative NaF for 24 h promotes an important reduction in the parasite proliferation (IC50/24 h = 23.6 ± 4.6 µM). At IC50/24 h concentration, the compound induced an increase in reactive oxygen species (ROS) production and a shortening of the unique flagellum of the parasites. Electron microscopy evaluation reinforced the flagellar phenotype in treated promastigotes, and a dilated flagellar pocket was frequently observed. The treatment also promoted a prominent autophagic phenotype. An increased number of autophagosomes were detected, presenting different levels of cargo degradation, endoplasmic reticulum profiles surrounding different cellular structures, and the presence of concentric membranar structures inside the mitochondrion. Chalcone derivatives may present an opportunity to develop a treatment for the P. serpens infection, as they are easy to synthesize and are low in cost. In order to develop a new product, further studies are still necessary.
Collapse
Affiliation(s)
- Tamiris A. C. Santos
- Laboratório de Enzimologia, Departamento de Fisiologia, Universidade Federal de Sergipe, São Cristóvão 49100-000, SE, Brazil
- Programa de Pós-Graduação em Agricultura e Biodiversidade, Universidade Federal de Sergipe, São Cristóvão 49100-000, SE, Brazil
| | - Kleiton P. Silva
- Laboratório de Enzimologia, Departamento de Fisiologia, Universidade Federal de Sergipe, São Cristóvão 49100-000, SE, Brazil
- Programa de Pós-Graduação em Agricultura e Biodiversidade, Universidade Federal de Sergipe, São Cristóvão 49100-000, SE, Brazil
| | - Gabriella B. Souza
- Laboratório de Química, Universidade Federal de Sergipe, São Cristóvão 49100-000, SE, Brazil
| | - Péricles B. Alves
- Laboratório de Química, Universidade Federal de Sergipe, São Cristóvão 49100-000, SE, Brazil
| | - Rubem F. S. Menna-Barreto
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, RJ, Brazil
- Correspondence: (R.F.S.M.-B.); (R.P.M.F.)
| | - Ricardo Scher
- Programa de Pós-Graduação em Agricultura e Biodiversidade, Universidade Federal de Sergipe, São Cristóvão 49100-000, SE, Brazil
- Laboratório de Biologia Celular e Imunologia do Câncer e Leishmania, Universidade Federal de Sergipe, São Cristóvão 49100-000, SE, Brazil
| | - Roberta P. M. Fernandes
- Laboratório de Enzimologia, Departamento de Fisiologia, Universidade Federal de Sergipe, São Cristóvão 49100-000, SE, Brazil
- Programa de Pós-Graduação em Agricultura e Biodiversidade, Universidade Federal de Sergipe, São Cristóvão 49100-000, SE, Brazil
- Correspondence: (R.F.S.M.-B.); (R.P.M.F.)
| |
Collapse
|
24
|
Kang MS, Kim S, Kim DS, Yu HS, Lee JE. The amoebicidal effect of Torreya nucifera extract on Acanthamoeba lugdunensis. PLoS One 2023; 18:e0281141. [PMID: 36745609 PMCID: PMC9901751 DOI: 10.1371/journal.pone.0281141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 01/15/2023] [Indexed: 02/07/2023] Open
Abstract
As the number of contact lens users increases, contact lens induced corneal infection is becoming more common. Acanthamoeba keratitis (AK) is a type of those which is caused by Acanthamoeba species, and may cause severe ocular inflammation and visual loss. We evaluated whether Torreya nucifera (T. nucifera) extract has an anti-amoebic effect and studied its mechanism of action on Acanthamoeba lugdunensis (A. lugdunensis). Cell viability was tested using the alamarBlue™ method, and the cell death mechanism was confirmed using the Tali® Apoptosis Kit. The SYTOX® Green assay was performed to check the plasma membrane permeability. The JC-1 dye was used to measure the mitochondrial membrane potential. A CellTiter-Glo® Luminescent Assay was used to measure the adenosine-triphosphate (ATP) level. Morphological changes in the mitochondria were examined by transmission electron microscopy (TEM). Cystic changes and a decrease in cell viability after treatment with T. nucifera were observed. Both apoptotic and necrotic cells were found in the Tali® Apoptosis assay. There was no significant difference in plasma membrane permeability between the control and T. nucifera treated groups. The collapse of the mitochondrial membrane potential and reduced ATP level in A. lugdunensis was confirmed in the groups treated with T. nucifera. Structural damage to the mitochondria was observed on TEM in the groups treated with T. nucifera. T. nucifera showed an anti-amoebic effect on A. lugdunensis, by inducing the loss of mitochondrial membrane potential. Thus, it could be a future therapeutic agent for AK.
Collapse
Affiliation(s)
- Min Seung Kang
- Department of Ophthalmology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, South Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Sangyoon Kim
- Department of Ophthalmology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, South Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Da Som Kim
- Research Center of Manufacturing Process and Analytical Development, Aprogen, Osong, South Korea
| | - Hak Sun Yu
- Department of Parasitology and Tropical Medicine, Pusan National University School of Medicine, Yangsan, South Korea
| | - Ji Eun Lee
- Department of Ophthalmology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, South Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
- Department of Ophthalmology, Pusan National University School of Medicine, Yangsan, South Korea
| |
Collapse
|
25
|
San Nicolás-Hernández D, Bethencourt-Estrella CJ, López-Arencibia A, Hernández-Álvarez E, Sifaoui I, Bazzocchi IL, Lorenzo-Morales J, Jiménez IA, Piñero JE. Withaferin A-silyl ether analogs as potential anti-kinetoplastid agents targeting the programmed cell death. Biomed Pharmacother 2023; 157:114012. [PMID: 36399830 DOI: 10.1016/j.biopha.2022.114012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/17/2022] Open
Abstract
Current therapies of leishmaniasis and Chagas disease, two of the most widespread neglected tropical diseases, have limited efficacy and toxic side effects. In this regard, natural products play an important role in overcoming the current need for new antiparasitic agents. The present study reports the leishmanicidal and trypanocidal activities of twenty-four known silyl-ether derivatives of withaferin A. Eleven compounds from this series (4, 7, 8, 10, 12, 15, 17, 18, 20, 22 and 25) showed a potent dose-dependent inhibitory effect on the proliferation of Leishmania amazonensis promastigotes and Trypanosoma cruzi epimastigotes respectively, even higher than the references drugs, miltefosine and benznidazole. Among them, the most promising compound, derivative 10, exhibited approximately 34-fold higher leishmanicidal activity and 49-fold higher trypanocidal activity compared to the reference drugs, as well as lower cytotoxicity. Moreover, compounds 4, 7, 10, 12 and 15 were more active than the reference drugs against the amastigote forms of L. amazonensis, presenting a high selectivity index. Assays performed to study the ATP levels, mitochondrial membrane potential, plasma membrane permeability, chromatin condensation, reactive oxygen species and autophagy indicated that these withaferin A-silyl analogs appear to induce events characteristic of apoptosis-like and also autophagy leading to programmed cell death. These findings support the therapeutic potential of withaferin A-related steroids as anti-Leishmania and Trypanosoma agents.
Collapse
Affiliation(s)
- Desirée San Nicolás-Hernández
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofısico Fco. Sanchez, S/N, 38203 La Laguna, Tenerife, Canary Islands, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38200 La Laguna, Tenerife, Spain; Red de Investigación Cooperativa en Enfermedades Tropicales (RICET), Inst. de Salud Carlos III, Madrid, Spain.
| | - Carlos J Bethencourt-Estrella
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofısico Fco. Sanchez, S/N, 38203 La Laguna, Tenerife, Canary Islands, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38200 La Laguna, Tenerife, Spain; Red de Investigación Cooperativa en Enfermedades Tropicales (RICET), Inst. de Salud Carlos III, Madrid, Spain.
| | - Atteneri López-Arencibia
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofısico Fco. Sanchez, S/N, 38203 La Laguna, Tenerife, Canary Islands, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38200 La Laguna, Tenerife, Spain; Red de Investigación Cooperativa en Enfermedades Tropicales (RICET), Inst. de Salud Carlos III, Madrid, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Inst. de Salud Carlos III, Madrid, Spain.
| | - Eduardo Hernández-Álvarez
- Instituto Universitario de Bio-Orgánica Antonio González, Departamento de Química Orgánica, Universidad de La Laguna, Avenida Astrofísico Francisco Sánchez 2, 38206 La Laguna, Tenerife, Canary Islands, Spain.
| | - Ines Sifaoui
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofısico Fco. Sanchez, S/N, 38203 La Laguna, Tenerife, Canary Islands, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38200 La Laguna, Tenerife, Spain; Red de Investigación Cooperativa en Enfermedades Tropicales (RICET), Inst. de Salud Carlos III, Madrid, Spain.
| | - Isabel L Bazzocchi
- Instituto Universitario de Bio-Orgánica Antonio González, Departamento de Química Orgánica, Universidad de La Laguna, Avenida Astrofísico Francisco Sánchez 2, 38206 La Laguna, Tenerife, Canary Islands, Spain.
| | - Jacob Lorenzo-Morales
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofısico Fco. Sanchez, S/N, 38203 La Laguna, Tenerife, Canary Islands, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38200 La Laguna, Tenerife, Spain; Red de Investigación Cooperativa en Enfermedades Tropicales (RICET), Inst. de Salud Carlos III, Madrid, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Inst. de Salud Carlos III, Madrid, Spain.
| | - Ignacio A Jiménez
- Instituto Universitario de Bio-Orgánica Antonio González, Departamento de Química Orgánica, Universidad de La Laguna, Avenida Astrofísico Francisco Sánchez 2, 38206 La Laguna, Tenerife, Canary Islands, Spain.
| | - José E Piñero
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofısico Fco. Sanchez, S/N, 38203 La Laguna, Tenerife, Canary Islands, Spain; Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38200 La Laguna, Tenerife, Spain; Red de Investigación Cooperativa en Enfermedades Tropicales (RICET), Inst. de Salud Carlos III, Madrid, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Inst. de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
26
|
Xie Y, Liang H, Jiang N, Liu D, Zhang N, Li Q, Zhang K, Sang X, Feng Y, Chen R, Zhang Y, Chen Q. Graphene quantum dots induce cascadic apoptosis via interaction with proteins associated with anti-oxidation after endocytosis by Trypanosoma brucei. Front Immunol 2022; 13:1022050. [PMID: 36561761 PMCID: PMC9763322 DOI: 10.3389/fimmu.2022.1022050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022] Open
Abstract
Trypanosoma brucei, the pathogen causing African sleeping sickness (trypanosomiasis) in humans, causes debilitating diseases in many regions of the world, but mainly in African countries with tropical and subtropical climates. Enormous efforts have been devoted to controlling trypanosomiasis, including expanding vector control programs, searching for novel anti-trypanosomial agents, and developing vaccines, but with limited success. In this study, we systematically investigated the effect of graphene quantum dots (GQDs) on trypanosomal parasites and their underlying mechanisms. Ultrasmall-sized GQDs can be efficiently endocytosed by T. brucei and with no toxicity to mammalian-derived cells, triggering a cascade of apoptotic reactions, including mitochondrial disorder, intracellular reactive oxygen species (ROS) elevation, Ca2+ accumulation, DNA fragmentation, adenosine triphosphate (ATP) synthesis impairment, and cell cycle arrest. All of these were caused by the direct interaction between GQDs and the proteins associated with cell apoptosis and anti-oxidation responses, such as trypanothione reductase (TryR), a key protein in anti-oxidation. GQDs specifically inhibited the enzymatic activity of TryR, leading to a reduction in the antioxidant capacity and, ultimately, parasite apoptotic death. These data, for the first time, provide a basis for the exploration of GQDs in the development of anti-trypanosomials.
Collapse
Affiliation(s)
- Yiwei Xie
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Hongrui Liang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Ning Jiang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Dingyuan Liu
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Naiwen Zhang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Qilong Li
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Kai Zhang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Xiaoyu Sang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Ying Feng
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Ran Chen
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Yiwei Zhang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China
| | - Qijun Chen
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China,Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, Shenyang, China,Research Unit for Pathogenic Mechanism of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang Agricultural University, Shenyang, China,*Correspondence: Qijun Chen,
| |
Collapse
|
27
|
Rivas F, Del Mármol C, Scalese G, Pérez-Díaz L, Machado I, Blacque O, Medeiros A, Comini M, Gambino D. New multifunctional Ru(II) organometallic compounds show activity against Trypanosoma brucei and Leishmania infantum. J Inorg Biochem 2022; 237:112016. [PMID: 36244312 DOI: 10.1016/j.jinorgbio.2022.112016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/24/2022] [Accepted: 09/26/2022] [Indexed: 11/06/2022]
Abstract
Human African trypanosomiasis (sleeping sickness) and leishmaniasis are prevalent zoonotic diseases caused by genomically related trypanosomatid protozoan parasites (Trypanosoma brucei and Leishmania spp). Additionally, both are co-endemic in certain regions of the world. Only a small number of old drugs exist for their treatment, with most of them sharing poor safety, efficacy, and pharmacokinetic profiles. In this work, new multifunctional Ru(II) ferrocenyl compounds were rationally designed as potential agents against these trypanosomatid parasites by including in a single molecule 1,1'-bis(diphenylphosphino)ferrocene (dppf) and two bioactive bidentate ligands: 8-hydroxyquinoline derivatives (8HQs) and polypyridyl ligands (NN). Three [Ru(8HQs)(dppf)(NN)](PF6) compounds were synthesized and fully characterized. They showed in vitro activity on bloodstream Trypanosoma brucei (IC50 140-310 nM) and on Leishmania infantum promastigotes (IC50 3.0-4.8 μM). The compounds showed good selectivity towards T. brucei in respect to J774 murine macrophages as mammalian cell model (SI 15-38). Changing hexafluorophosphate counterion by chloride led to a three-fold increase in activity on both parasites and to a two to three-fold increase in selectivity towards the pathogens. The compounds affect in vitro at least the targets of the individual bioactive moieties included in the new chemical entities: DNA and generation of ROS. The compounds are stable in solution and are more lipophilic than the free bioactive ligands. No clear correlation between lipophilicity, interaction with DNA or generation of ROS and activity was detected, which agrees with their overall similar anti-trypanosoma potency and selectivity. These compounds are promising candidates for further drug development.
Collapse
Affiliation(s)
- Feriannys Rivas
- Área Química Inorgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay; Programa de Posgrado en Química, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Carolina Del Mármol
- Área Química Inorgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Gonzalo Scalese
- Área Química Inorgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Leticia Pérez-Díaz
- Laboratorio de Interacciones Moleculares, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Ignacio Machado
- Área Química Analítica, Universidad de la República, Montevideo, Uruguay
| | - Olivier Blacque
- Department of Chemistry, University of Zurich, Zurich, Switzerland
| | - Andrea Medeiros
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, Montevideo, Uruguay; Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Marcelo Comini
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Dinorah Gambino
- Área Química Inorgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
28
|
Sandes JM, de Figueiredo RCBQ. The endoplasmic reticulum of trypanosomatids: An unrevealed road for chemotherapy. Front Cell Infect Microbiol 2022; 12:1057774. [PMID: 36439218 PMCID: PMC9684732 DOI: 10.3389/fcimb.2022.1057774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 10/24/2022] [Indexed: 01/04/2024] Open
Abstract
The endoplasmic reticulum (ER) of higher eukaryotic cells forms an intricate membranous network that serves as the main processing facility for folding and assembling of secreted and membrane proteins. The ER is a highly dynamic organelle that interacts with other intracellular structures, as well as endosymbiotic pathogenic and non-pathogenic microorganisms. A strict ER quality control (ERQC) must work to ensure that proteins entering the ER are folded and processed correctly. Unfolded or misfolded proteins are usually identified, selected, and addressed to Endoplasmic Reticulum-Associated Degradation (ERAD) complex. Conversely, when there is a large demand for secreted proteins or ER imbalance, the accumulation of unfolded or misfolded proteins activates the Unfold Protein Response (UPR) to restore the ER homeostasis or, in the case of persistent ER stress, induces the cell death. Pathogenic trypanosomatids, such as Trypanosoma cruzi, Trypanosoma brucei and Leishmania spp are the etiological agents of important neglected diseases. These protozoans have a complex life cycle alternating between vertebrate and invertebrate hosts. The ER of trypanosomatids, like those found in higher eukaryotes, is also specialized for secretion, and depends on the ERAD and non-canonical UPR to deal with the ER stress. Here, we reviewed the basic aspects of ER biology, organization, and quality control in trypanosomatids. We also focused on the unusual way by which T. cruzi, T. brucei, and Leishmania spp. respond to ER stress, emphasizing how these parasites' ER-unrevealed roads might be an attractive target for chemotherapy.
Collapse
Affiliation(s)
- Jana Messias Sandes
- Laboratório de Biologia Celular e Molecular de Patógenos, Departamento de Microbiologia, Instituto Aggeu Magalhães, Recife, Brazil
- Laboratório de Microscopia Eletrônica, Instituto Keizo Assami, Universidade Federal de Pernambuco, Recife, Brazil
| | | |
Collapse
|
29
|
Knieß R, Leeder W, Reißig P, Geyer FK, Göringer HU. Core-Shell DNA-Cholesterol Nanoparticles Exert Lysosomolytic Activity in African Trypanosomes. Chembiochem 2022; 23:e202200410. [PMID: 36040754 PMCID: PMC9826209 DOI: 10.1002/cbic.202200410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/18/2022] [Indexed: 01/11/2023]
Abstract
Trypanosoma brucei is the causal infectious agent of African trypanosomiasis in humans and Nagana in livestock. Both diseases are currently treated with a small number of chemotherapeutics, which are hampered by a variety of limitations reaching from efficacy and toxicity complications to drug-resistance problems. Here, we explore the forward design of a new class of synthetic trypanocides based on nanostructured, core-shell DNA-lipid particles. In aqueous solution, the particles self-assemble into micelle-type structures consisting of a solvent-exposed, hydrophilic DNA shell and a hydrophobic lipid core. DNA-lipid nanoparticles have membrane-adhesive qualities and can permeabilize lipid membranes. We report the synthesis of DNA-cholesterol nanoparticles, which specifically subvert the membrane integrity of the T. brucei lysosome, killing the parasite with nanomolar potencies. Furthermore, we provide an example of the programmability of the nanoparticles. By functionalizing the DNA shell with a spliced leader (SL)-RNA-specific DNAzyme, we target a second trypanosome-specific pathway (dual-target approach). The DNAzyme provides a backup to counteract the recovery of compromised parasites, which reduces the risk of developing drug resistance.
Collapse
Affiliation(s)
- Robert Knieß
- Molecular GeneticsTechnical University DarmstadtSchnittspahnstr. 1064287DarmstadtGermany
| | - Wolf‐Matthias Leeder
- Molecular GeneticsTechnical University DarmstadtSchnittspahnstr. 1064287DarmstadtGermany
| | - Paul Reißig
- Molecular GeneticsTechnical University DarmstadtSchnittspahnstr. 1064287DarmstadtGermany
| | - Felix Klaus Geyer
- Molecular GeneticsTechnical University DarmstadtSchnittspahnstr. 1064287DarmstadtGermany
| | - H. Ulrich Göringer
- Molecular GeneticsTechnical University DarmstadtSchnittspahnstr. 1064287DarmstadtGermany
| |
Collapse
|
30
|
Scariot DB, Staneviciute A, Zhu J, Li X, Scott EA, Engman DM. Leishmaniasis and Chagas disease: Is there hope in nanotechnology to fight neglected tropical diseases? Front Cell Infect Microbiol 2022; 12:1000972. [PMID: 36189341 PMCID: PMC9523166 DOI: 10.3389/fcimb.2022.1000972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 08/30/2022] [Indexed: 11/22/2022] Open
Abstract
Nanotechnology is revolutionizing many sectors of science, from food preservation to healthcare to energy applications. Since 1995, when the first nanomedicines started being commercialized, drug developers have relied on nanotechnology to improve the pharmacokinetic properties of bioactive molecules. The development of advanced nanomaterials has greatly enhanced drug discovery through improved pharmacotherapeutic effects and reduction of toxicity and side effects. Therefore, highly toxic treatments such as cancer chemotherapy, have benefited from nanotechnology. Considering the toxicity of the few therapeutic options to treat neglected tropical diseases, such as leishmaniasis and Chagas disease, nanotechnology has also been explored as a potential innovation to treat these diseases. However, despite the significant research progress over the years, the benefits of nanotechnology for both diseases are still limited to preliminary animal studies, raising the question about the clinical utility of nanomedicines in this field. From this perspective, this review aims to discuss recent nanotechnological developments, the advantages of nanoformulations over current leishmanicidal and trypanocidal drugs, limitations of nano-based drugs, and research gaps that still must be filled to make these novel drug delivery systems a reality for leishmaniasis and Chagas disease treatment.
Collapse
Affiliation(s)
- Debora B. Scariot
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, and Simpson Querrey Institute, Northwestern University, Evanston and Chicago, IL, United States
| | - Austeja Staneviciute
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, and Simpson Querrey Institute, Northwestern University, Evanston and Chicago, IL, United States
| | - Jennifer Zhu
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, and Simpson Querrey Institute, Northwestern University, Evanston and Chicago, IL, United States
| | - Xiaomo Li
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Pathology, Northwestern University, Chicago, IL, United States
| | - Evan A. Scott
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, and Simpson Querrey Institute, Northwestern University, Evanston and Chicago, IL, United States
| | - David M. Engman
- Department of Pathology, Northwestern University, Chicago, IL, United States
| |
Collapse
|
31
|
Monteiro ML, Lima DB, Freire KA, Nicolaski Pedron C, Magalhães EP, Silva BP, García-Jareño AB, Silva Oliveira C, Nunes JVS, Marinho MM, Menezes RRPPBD, Orzaéz M, Oliveira Junior VX, Martins AMC. Rational design of a trypanocidal peptide derived from Dinoponera quadriceps venom. Eur J Med Chem 2022; 241:114624. [DOI: 10.1016/j.ejmech.2022.114624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/06/2022] [Accepted: 07/21/2022] [Indexed: 11/27/2022]
|
32
|
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
|
33
|
Bethencourt-Estrella CJ, Delgado-Hernández S, López-Arencibia A, San Nicolás-Hernández D, Tejedor D, García-Tellado F, Lorenzo-Morales J, Piñero JE. In vitro activity and cell death mechanism induced by acrylonitrile derivatives against Leishmania amazonensis. Bioorg Chem 2022; 124:105872. [DOI: 10.1016/j.bioorg.2022.105872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/06/2022] [Accepted: 05/10/2022] [Indexed: 11/24/2022]
|
34
|
Costa Catta-Preta CM, Cézar de Azevedo-Martins A, de Souza W, Motta MCM. Effect of the endoplasmic reticulum stressor tunicamycin in Angomonas deanei heat-shock protein expression and on the association with the endosymbiotic bacterium. Exp Cell Res 2022; 417:113162. [PMID: 35460679 DOI: 10.1016/j.yexcr.2022.113162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/04/2022] [Accepted: 04/16/2022] [Indexed: 01/01/2023]
Abstract
The endoplasmic reticulum (ER) presents unique properties to establishing bacterium symbiosis in eukaryotic cells since it synthesizes and glycosylates essential molecules like proteins and lipids. Tunicamycin (TM) is an antibiotic that inhibits the first step in the N-linked glycosylation in eukaryotes and has been used as an ER stress inducer to activate the Unfolded Protein Response (UPR). Mutualistic symbiosis in trypanosomatids is characterized by structural adaptations and intense metabolic exchanges, thus we investigated the effects of TM in the association between Angomonas deanei and its symbiotic bacterium, through ultrastructural and proteomic approaches. Cells treated with the inhibitor showed a decrease in proliferation, enlargement of the ER and Golgi cisternae and an increased distance between the symbiont and the ER. TM proved to be an important tool to better understand ER stress in trypanosomatids, since changes in protein composition were observed in the host protozoan, especially the expression of the Hsp90 chaperone. Furthermore, data obtained indicates the importance of the ER for the adaptation and maintenance of symbiotic associations between prokaryotes and eukaryotes, considering that this organelle has recognized importance in the biogenesis and division of cell structures.
Collapse
Affiliation(s)
- Carolina Moura Costa Catta-Preta
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21491-590, Rio de Janeiro, RJ, Brazil
| | - Allan Cézar de Azevedo-Martins
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21491-590, Rio de Janeiro, RJ, Brazil
| | - Wanderley de Souza
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21491-590, Rio de Janeiro, RJ, Brazil; Centro Nacional de Biologia Estrutural e Bioimagem, RJ, Brazil
| | - Maria Cristina M Motta
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21491-590, Rio de Janeiro, RJ, Brazil; Centro Nacional de Biologia Estrutural e Bioimagem, RJ, Brazil.
| |
Collapse
|
35
|
Pedra-Rezende Y, Macedo IS, Midlej V, Mariante RM, Menna-Barreto RFS. Different Drugs, Same End: Ultrastructural Hallmarks of Autophagy in Pathogenic Protozoa. Front Microbiol 2022; 13:856686. [PMID: 35422792 PMCID: PMC9002357 DOI: 10.3389/fmicb.2022.856686] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/02/2022] [Indexed: 01/18/2023] Open
Abstract
Protozoan parasites interact with a wide variety of organisms ranging from bacteria to humans, representing one of the most common causes of parasitic diseases and an important public health problem affecting hundreds of millions of people worldwide. The current treatment for these parasitic diseases remains unsatisfactory and, in some cases, very limited. Treatment limitations together with the increased resistance of the pathogens represent a challenge for the improvement of the patient’s quality of life. The continuous search for alternative preclinical drugs is mandatory, but the mechanisms of action of several of these compounds have not been described. Electron microscopy is a powerful tool for the identification of drug targets in almost all cellular models. Interestingly, ultrastructural analysis showed that several classes of antiparasitic compounds induced similar autophagic phenotypes in trypanosomatids, trichomonadids, and apicomplexan parasites as well as in Giardia intestinalis and Entamoeba spp. with the presence of an increased number of autophagosomes as well as remarkable endoplasmic reticulum profiles surrounding different organelles. Autophagy is a physiological process of eukaryotes that maintains homeostasis by the self-digestion of nonfunctional organelles and/or macromolecules, limiting redundant and damaged cellular components. Here, we focus on protozoan autophagy to subvert drug effects, discussing its importance for successful chemotherapy.
Collapse
Affiliation(s)
- Yasmin Pedra-Rezende
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Isabela S Macedo
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Victor Midlej
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Rafael M Mariante
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | | |
Collapse
|
36
|
Mendes A, Armada A, Cabral LIL, Amado PSM, Campino L, Cristiano MLS, Cortes S. 1,2,4-Trioxolane and 1,2,4,5-Tetraoxane Endoperoxides against Old-World Leishmania Parasites: In Vitro Activity and Mode of Action. Pharmaceuticals (Basel) 2022; 15:ph15040446. [PMID: 35455443 PMCID: PMC9024893 DOI: 10.3390/ph15040446] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/01/2022] [Accepted: 04/02/2022] [Indexed: 12/15/2022] Open
Abstract
Leishmaniasis remains one of the ten Neglected Tropical Diseases with significant morbidity and mortality in humans. Current treatment of visceral leishmaniasis is difficult due to a lack of effective, non-toxic, and non-extensive medications. This study aimed to evaluate the selectivity of 12 synthetic endoperoxides (1,2,4-trioxolanes; 1,2,4,5-tetraoxanes) and uncover their biochemical effects on Leishmania parasites responsible for visceral leishmaniasis. The compounds were screened for in vitro activity against L. infantum and L. donovani and for cytotoxicity in two monocytic cell lines (J774A.1 and THP-1) using the methyl thiazol tetrazolium assay. Reactive oxygen species formation, apoptosis, and mitochondrial impairment were measured by flow cytometry. The compounds exhibited fair to moderate anti-proliferative activity against promastigotes of the 2 Leishmania species, with IC50 values ranging from 13.0 ± 1.7 µM to 793.0 ± 37.2 µM. Tetraoxanes LC132 and LC138 demonstrated good leishmanicidal activity on L. infantum amastigotes (IC50 13.2 ± 5.2 and 23.9 ± 2.7 µM) with low cytotoxicity in mammalian cells (SIs 22.1 and 118.6), indicating selectivity towards the parasite. Furthermore, LC138 was able to induce late apoptosis and dose-dependent oxidative stress without affecting mithocondria. Compounds LC132 and LC138 can be further explored as potential antileishmanial chemotypes.
Collapse
Affiliation(s)
- Andreia Mendes
- Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), Rua da Junqueira, 100, 1349-008 Lisboa, Portugal; (A.M.); (A.A.); (L.C.)
| | - Ana Armada
- Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), Rua da Junqueira, 100, 1349-008 Lisboa, Portugal; (A.M.); (A.A.); (L.C.)
- Global Health Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), Rua da Junqueira, 100, 1349-008 Lisboa, Portugal
| | - Lília I. L. Cabral
- Centro de Ciências do Mar (CCMAR), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; (L.I.L.C.); (P.S.M.A.)
- Departamento de Química e Farmácia, Faculdade de Ciências e Tecnologia, Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Patrícia S. M. Amado
- Centro de Ciências do Mar (CCMAR), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; (L.I.L.C.); (P.S.M.A.)
- Departamento de Química e Farmácia, Faculdade de Ciências e Tecnologia, Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Lenea Campino
- Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), Rua da Junqueira, 100, 1349-008 Lisboa, Portugal; (A.M.); (A.A.); (L.C.)
| | - Maria L. S. Cristiano
- Centro de Ciências do Mar (CCMAR), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; (L.I.L.C.); (P.S.M.A.)
- Departamento de Química e Farmácia, Faculdade de Ciências e Tecnologia, Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
- Correspondence: (M.L.S.C.); (S.C.)
| | - Sofia Cortes
- Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), Rua da Junqueira, 100, 1349-008 Lisboa, Portugal; (A.M.); (A.A.); (L.C.)
- Global Health Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), Rua da Junqueira, 100, 1349-008 Lisboa, Portugal
- Correspondence: (M.L.S.C.); (S.C.)
| |
Collapse
|
37
|
Chloride substitution on 2-hydroxy-3,4,6-trimethoxyphenylchalcones improves in vitro selectivity on Trypanosoma cruzi strain Y. Chem Biol Interact 2022; 361:109920. [DOI: 10.1016/j.cbi.2022.109920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 03/14/2022] [Accepted: 03/30/2022] [Indexed: 01/12/2023]
|
38
|
das Chagas Almeida A, Meinel RS, Leal YL, Silva TP, Glanzmann N, Mendonça DVC, Perin L, Cunha-Júnior EF, Coelho EAF, Melo RCN, da Silva AD, Coimbra ES. Functionalized 1,2,3-triazolium salts as potential agents against visceral leishmaniasis. Parasitol Res 2022; 121:1389-1406. [PMID: 35169883 DOI: 10.1007/s00436-022-07431-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 01/07/2022] [Indexed: 10/19/2022]
Abstract
Visceral leishmaniasis (VL) is the most severe clinical form of leishmaniasis, being fatal if untreated. In search of a more effective treatment for VL, one of the main strategies is the development and screening of new antileishmanial compounds. Here, we reported the synthesis of seven new acetyl functionalized 1,2,3-triazolium salts, together with four 1,2,3-triazole precursors, and investigated their effect against different strains of L. infantum from dogs and humans. The 1,2,3-triazolium salts exhibited better activity than the 1,2,3-triazole derivatives with IC50 range from 0.12 to 8.66 μM and, among them, compound 5 showed significant activity against promastigotes (IC50 from 4.55 to 5.28 μM) and intracellular amastigotes (IC50 from 5.36 to 7.92 μM), with the best selective index (SI ~ 6-9) and reduced toxicity. Our findings, using biochemical and ultrastructural approaches, demonstrated that compound 5 targets the mitochondrion of L. infantum promastigotes, leading to the formation of reactive oxygen species (ROS), increase of the mitochondrial membrane potential, and mitochondrial alteration. Moreover, quantitative transmission electron microscopy (TEM) revealed that compound 5 induces the reduction of promastigote size and cytoplasmic vacuolization. Interestingly, the effect of compound 5 was not associated with apoptosis or necrosis of the parasites but, instead, seems to be mediated through a pathway involving autophagy, with a clear detection of autophagic vacuoles in the cytoplasm by using both a fluorescent marker and TEM. As for the in vivo studies, compound 5 showed activity in a mouse model of VL at 20 mg/kg, reducing the parasite load in both spleen and liver (59.80% and 26.88%, respectively). Finally, this compound did not induce hepatoxicity or nephrotoxicity and was able to normalize the altered biochemical parameters in the infected mice. Thus, our findings support the use of 1,2,3-triazolium salts as potential agents against visceral leishmaniasis.
Collapse
Affiliation(s)
- Ayla das Chagas Almeida
- Núcleo de Pesquisas Em Parasitologia, Departamento de Parasitologia, Microbiologia e Imunologia, I.C.B., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Raíssa Soares Meinel
- SINTBIOMOL, Departamento de Química, I.C.E., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Yasmim Lopes Leal
- SINTBIOMOL, Departamento de Química, I.C.E., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Thiago P Silva
- Laboratório de Biologia Celular, Departamento de Biologia, Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Nícolas Glanzmann
- SINTBIOMOL, Departamento de Química, I.C.E., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Débora Vasconcelos Costa 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
| | - Luísa 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
| | - Edézio Ferreira Cunha-Júnior
- Laboratório de Imunoparasitologia, Unidade Integrada de Pesquisa Em Produtos Bioativos e Biociências, Universidade Federal Do Rio de Janeiro, Campus UFRJ-Macaé, Macaé, 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
| | - Rossana C N Melo
- Laboratório de Biologia Celular, Departamento de Biologia, Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Adilson David da Silva
- SINTBIOMOL, Departamento de Química, I.C.E., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Elaine Soares Coimbra
- Núcleo de Pesquisas Em Parasitologia, Departamento de Parasitologia, Microbiologia e Imunologia, I.C.B., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil.
| |
Collapse
|
39
|
Carloto ACM, Bortoleti BTDS, Rodrigues ACJ, Silva TF, Tomiotto-Pellissier F, Bidóia DL, Gonçalves MD, Assolini JP, Dekker RFH, Barbosa-Dekker AM, Costa IN, Conchon-Costa I, Miranda-Sapla MM, Pavanelli WR. Botryosphaeran, [(1 → 3)(1 → 6)-β-D-glucan], induces apoptosis-like death in promastigotes of Leishmania amazonensis, and exerts a leishmanicidal effect on infected macrophages by activating NF-kB and producing pro-inflammatory molecules. Chem Biol Interact 2022; 351:109713. [PMID: 34699765 DOI: 10.1016/j.cbi.2021.109713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 09/30/2021] [Accepted: 10/20/2021] [Indexed: 12/11/2022]
Abstract
Leishmaniasis is an infectious-parasitic disease caused by the protozoan Leishmania spp. The available treatments are based upon expensive drugs bearing adverse side-effects. The search for new therapeutic alternatives that present a more effective action without causing adverse effects to the patient is therefore important. The objective of this study was to evaluate the in vitro effect of botryosphaeran, a (1 → 3)(1 → 6)-β-D-glucan, on the promastigote and intracellular amastigote forms of Leishmania amazonensis. The direct activity of botryosphaeran on promastigote forms was evaluated in vitro and inhibited proliferation, the IC50 7 μg/mL in 48 h was calculated. After 48 h treatment, botryosphaeran induced nitric oxide production (NO), caused mitochondrial membrane hyperpolarization, increased reactive oxygen species (ROS), and accumulation of lipid vesicles in promastigotes, resulting in apoptosis, necrosis and autophagy, and was accompanied by morphological and ultrastructural changes. The range of concentrations used did not alter the viability of peritoneal macrophages from BALB/c mice and erythrocytes of sheep. Botryosphaeran was able to reduce the number of infected macrophages and the number of amastigotes per macrophage at 12.5 μg/mL (50.75% ± 6.48), 25 μg/mL (55.66% ± 3.93) and 50 μg/mL (72.9% ± 6.98), and IC50 9.3 μg/mL (±0.66) for intracellular amastigotes forms. The leishmanicidal effect was due to activation of NF-κB and promoted an increase in pro-inflammatory cytokines (TNF-α and IL-6), iNOS and microbial-derived ROS and NO, in addition to decreasing the levels of SOD. Based upon the data obtained, we infer that botryosphaeran exerted an active leishmanicidal and immunomodulatory effect, acting on promastigotes through autophagic, apoptotic and necrosis processes, and in the intracellular amastigote form, through the action of ROS and NO.
Collapse
Affiliation(s)
- Amanda Cristina Machado Carloto
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, 86057-970, Londrina, Paraná, Brazil.
| | - Bruna Taciane da Silva Bortoleti
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, 86057-970, Londrina, Paraná, Brazil; Gonçalo Moniz Institute (FIOCRUZ/Bahia), 40296-710, Salvador, Bahia, Brazil
| | - Ana Carolina Jacob Rodrigues
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, 86057-970, Londrina, Paraná, Brazil; Carlos Chagas Institute (ICC/FIOCRUZ/Paraná), 81310-020, Curitiba, Paraná, Brazil
| | - Taylon Felipe Silva
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, 86057-970, Londrina, Paraná, Brazil
| | - Fernanda Tomiotto-Pellissier
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, 86057-970, Londrina, Paraná, Brazil; Department of Medical Pathology, Health Sciences Sector, Federal University of Paraná, 80060-240, Curitiba, Paraná, Brazil
| | - Danielle Lazarin Bidóia
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, 86057-970, Londrina, Paraná, Brazil
| | - Manoela Daiele Gonçalves
- Biotransformation and Phytochemistry Laboratory, Chemistry Department, Exact Sciences Center, State University of Londrina, 86057-970, Londrina, Paraná, Brazil
| | - João Paulo Assolini
- Alto Vale University of Rio Do Peixe, 89500-000, Caçador, Santa Catarina, Brazil
| | - Robert F H Dekker
- Postgradute Program in Environmental Engineering, Paraná Technological University, Londrina Campus, 86036-370, Londrina, Paraná, Brazil; Beta-Glucan Pharmaceuticals EIRELI, Lote 24A, Zirconia Block, Paraná Technological University, Londrina Campus, Avenue João Miguel Caram 731, 86036-700, Londrina, Paraná, Brazil
| | - Aneli M Barbosa-Dekker
- Postgradute Program in Environmental Engineering, Paraná Technological University, Londrina Campus, 86036-370, Londrina, Paraná, Brazil; Beta-Glucan Pharmaceuticals EIRELI, Lote 24A, Zirconia Block, Paraná Technological University, Londrina Campus, Avenue João Miguel Caram 731, 86036-700, Londrina, Paraná, Brazil
| | - Idessania Nazareth Costa
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, 86057-970, Londrina, Paraná, Brazil
| | - Ivete Conchon-Costa
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, 86057-970, Londrina, Paraná, Brazil
| | - Milena Menegazzo Miranda-Sapla
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, 86057-970, Londrina, Paraná, Brazil
| | - Wander Rogério Pavanelli
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, 86057-970, Londrina, Paraná, Brazil.
| |
Collapse
|
40
|
Antiprotozoal Effect of Snake Venoms and Their Fractions: A Systematic Review. Pathogens 2021; 10:pathogens10121632. [PMID: 34959587 PMCID: PMC8707848 DOI: 10.3390/pathogens10121632] [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: 11/12/2021] [Revised: 12/08/2021] [Accepted: 12/13/2021] [Indexed: 11/17/2022] Open
Abstract
Background: Protozoal infection is a lingering public health issue of great concern, despite efforts to produce drugs and vaccines against it. Recent breakthrough research has discovered alternative antiprotozoal agents encompassing the use of snake venoms and their components to cure these infections. This study collated the existing literature to examine the antiprotozoal effect of snake venoms and their fractions. Methods: We conducted a systematic review following the PRISMA guidelines. The PubMed and Embase databases were searched from their inception until 13 October 2021. Articles were screened at the title, abstract and full-text phases. Some additional studies were obtained through the manual search process. Results: We identified 331 studies via the electronic database and manual searches, of which 55 reporting the antiprotozoal effect of snake venoms and their components were included in the review. Around 38% of studies examined the effect of whole crude venoms, and a similar percentage evaluated the effect of a proportion of enzymatic phospholipase A2 (PLA2). In particular, this review reports around 36 PLA2 activities and 29 snake crude venom activities. We also report the notable phenomenon of synergism with PLA2 isoforms of Bothrops asper. Importantly, limited attention has been given so far to the antiprotozoal efficacies of metalloproteinase, serine protease and three-finger toxins, although these venom components have been identified as significant components of the dominant venom families. Conclusion: This study highlights the impact of snake venoms and their fractions on controlling protozoal infections and suggests the need to examine further the effectiveness of other venom components, such as metalloproteinase, serine protease and three-finger toxins. Future research questions in this field must be redirected toward synergism in snake venom components, based on pharmacological usage and in the context of toxicology. Ascertaining the effects of snake venoms and their components on other protozoal species that have not yet been studied is imperative.
Collapse
|
41
|
Silveira GO, Coelho HS, Amaral MS, Verjovski-Almeida S. Long non-coding RNAs as possible therapeutic targets in protozoa, and in Schistosoma and other helminths. Parasitol Res 2021; 121:1091-1115. [PMID: 34859292 DOI: 10.1007/s00436-021-07384-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 11/14/2021] [Indexed: 12/26/2022]
Abstract
Long non-coding RNAs (lncRNAs) emerged in the past 20 years due to massive amounts of scientific data regarding transcriptomic analyses. They have been implicated in a plethora of cellular processes in higher eukaryotes. However, little is known about lncRNA possible involvement in parasitic diseases, with most studies only detecting their presence in parasites of human medical importance. Here, we review the progress on lncRNA studies and their functions in protozoans and helminths. In addition, we show an example of knockdown of one lncRNA in Schistosoma mansoni, SmLINC156349, which led to in vitro parasite adhesion, motility, and pairing impairment, with a 20% decrease in parasite viability and 33% reduction in female oviposition. Other observed phenotypes were a decrease in the proliferation rate of both male and female worms and their gonads, and reduced female lipid and vitelline droplets that are markers for well-developed vitellaria. Impairment of female worms' vitellaria in SmLINC156349-silenced worms led to egg development deficiency. All those results demonstrate the great potential of the tools and methods to characterize lncRNAs as potential new therapeutic targets. Further, we discuss the challenges and limitations of current methods for studying lncRNAs in parasites and possible solutions to overcome them, and we highlight the future directions of this exciting field.
Collapse
Affiliation(s)
- Gilbert O Silveira
- Laboratório de Parasitologia, Instituto Butantan, São Paulo, SP, 05503-900, Brazil.,Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Helena S Coelho
- Laboratório de Parasitologia, Instituto Butantan, São Paulo, SP, 05503-900, Brazil
| | - Murilo S Amaral
- Laboratório de Parasitologia, Instituto Butantan, São Paulo, SP, 05503-900, Brazil.
| | - Sergio Verjovski-Almeida
- Laboratório de Parasitologia, Instituto Butantan, São Paulo, SP, 05503-900, Brazil. .,Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, 05508-900, Brazil.
| |
Collapse
|
42
|
López-Arencibia A, Sifaoui I, Reyes-Batlle M, Bethencourt-Estrella CJ, San Nicolás-Hernández D, Lorenzo-Morales J, Piñero JE. Discovery of New Chemical Tools against Leishmania amazonensis via the MMV Pathogen Box. Pharmaceuticals (Basel) 2021; 14:1219. [PMID: 34959620 PMCID: PMC8708704 DOI: 10.3390/ph14121219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/12/2021] [Accepted: 11/22/2021] [Indexed: 01/04/2023] Open
Abstract
The protozoan parasite Leishmania causes a spectrum of diseases and there are over 1 million infections each year. Current treatments are toxic, expensive, and difficult to administer, and resistance to them is emerging. In this study, we screened the antileishmanial activity of the Pathogen Box compounds from the Medicine for Malaria Venture against Leishmania amazonensis, and compared their structures and cytotoxicity. The compounds MMV676388 (3), MMV690103 (5), MMV022029 (7), MMV022478 (9) and MMV021013 (10) exerted a significant dose-dependent inhibition effect on the proliferation of L. amazonensis promastigotes and intracellular amastigotes. Moreover, studies on the mechanism of cell death showed that compounds 3 and 5 induced an apoptotic process while the compounds 7, 9 and 10 seem to induce an autophagic mechanism. The present findings underline the potential of these five molecules as novel therapeutic leishmanicidal agents.
Collapse
Affiliation(s)
- Atteneri López-Arencibia
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Campus de Anchieta, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203 La Laguna, Spain; (I.S.); (M.R.-B.); (C.J.B.-E.); (D.S.N.-H.); (J.E.P.)
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38203 La Laguna, Spain
- Red de Investigación Colaborativa en Enfermedades Tropicales (RICET), 28006 Madrid, Spain
- Consorcio Centro de Investigacion Biomedica en Red M.P. (CIBER) de Enfermedades Infecciosas, Instituto de Salud Carlos III, 28006 Madrid, Spain
| | - Ines Sifaoui
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Campus de Anchieta, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203 La Laguna, Spain; (I.S.); (M.R.-B.); (C.J.B.-E.); (D.S.N.-H.); (J.E.P.)
- Red de Investigación Colaborativa en Enfermedades Tropicales (RICET), 28006 Madrid, Spain
- Consorcio Centro de Investigacion Biomedica en Red M.P. (CIBER) de Enfermedades Infecciosas, Instituto de Salud Carlos III, 28006 Madrid, Spain
| | - María Reyes-Batlle
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Campus de Anchieta, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203 La Laguna, Spain; (I.S.); (M.R.-B.); (C.J.B.-E.); (D.S.N.-H.); (J.E.P.)
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38203 La Laguna, Spain
- Red de Investigación Colaborativa en Enfermedades Tropicales (RICET), 28006 Madrid, Spain
- Consorcio Centro de Investigacion Biomedica en Red M.P. (CIBER) de Enfermedades Infecciosas, Instituto de Salud Carlos III, 28006 Madrid, Spain
| | - Carlos J. Bethencourt-Estrella
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Campus de Anchieta, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203 La Laguna, Spain; (I.S.); (M.R.-B.); (C.J.B.-E.); (D.S.N.-H.); (J.E.P.)
- Red de Investigación Colaborativa en Enfermedades Tropicales (RICET), 28006 Madrid, Spain
- Consorcio Centro de Investigacion Biomedica en Red M.P. (CIBER) de Enfermedades Infecciosas, Instituto de Salud Carlos III, 28006 Madrid, Spain
| | - Desirée San Nicolás-Hernández
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Campus de Anchieta, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203 La Laguna, Spain; (I.S.); (M.R.-B.); (C.J.B.-E.); (D.S.N.-H.); (J.E.P.)
- Red de Investigación Colaborativa en Enfermedades Tropicales (RICET), 28006 Madrid, Spain
- Consorcio Centro de Investigacion Biomedica en Red M.P. (CIBER) de Enfermedades Infecciosas, Instituto de Salud Carlos III, 28006 Madrid, Spain
| | - Jacob Lorenzo-Morales
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Campus de Anchieta, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203 La Laguna, Spain; (I.S.); (M.R.-B.); (C.J.B.-E.); (D.S.N.-H.); (J.E.P.)
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38203 La Laguna, Spain
- Red de Investigación Colaborativa en Enfermedades Tropicales (RICET), 28006 Madrid, Spain
- Consorcio Centro de Investigacion Biomedica en Red M.P. (CIBER) de Enfermedades Infecciosas, Instituto de Salud Carlos III, 28006 Madrid, Spain
| | - José E. Piñero
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Campus de Anchieta, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203 La Laguna, Spain; (I.S.); (M.R.-B.); (C.J.B.-E.); (D.S.N.-H.); (J.E.P.)
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, 38203 La Laguna, Spain
- Red de Investigación Colaborativa en Enfermedades Tropicales (RICET), 28006 Madrid, Spain
- Consorcio Centro de Investigacion Biomedica en Red M.P. (CIBER) de Enfermedades Infecciosas, Instituto de Salud Carlos III, 28006 Madrid, Spain
| |
Collapse
|
43
|
Bethencourt-Estrella CJ, Nocchi N, López-Arencibia A, San Nicolás-Hernández D, Souto ML, Suárez-Gómez B, Díaz-Marrero AR, Fernández JJ, Lorenzo-Morales J, Piñero JE. Antikinetoplastid Activity of Sesquiterpenes Isolated from the Zoanthid Palythoa aff. clavata. Pharmaceuticals (Basel) 2021; 14:ph14111095. [PMID: 34832876 PMCID: PMC8625207 DOI: 10.3390/ph14111095] [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: 09/24/2021] [Revised: 10/23/2021] [Accepted: 10/25/2021] [Indexed: 02/07/2023] Open
Abstract
Leishmaniasis and Chagas disease are neglected tropical diseases that cause problems in developing countries. The causative agents, Leishmania spp. and Trypanosoma cruzi, produce a clinical picture that can be fatal for the patient, such as Chagas heart disease, visceral leishmaniasis and megacolon, among others. Current treatments for these diseases are not very effective and highly toxic, since they require very prolonged treatments. The development of innovative, effective and safe drugs to fight infections caused by these parasites remains a challenge. For this reason, in recent years, there has been an increase in the search for new therapies. In this study, the antikinetoplastid activity of 13 sesquiterpene lactones obtained from Palythoa aff. clavata was screened against L. amazonensis, L. donovani and T. cruzi. The results revealed that the sesquiterpene lactones anhydroartemorin (2), cis,trans-costunolide-14-acetate (3) and 4-hydroxyarbusculin A (11) were the most selective against the kinetoplastid species studied. These molecules seem to induce the mechanisms involved in an apoptotic-like death or programmed cell death (PCD) in the kinetoplastids, and since they do not cause necrosis, the inflammatory events associated with this type of cell death will not be triggered.
Collapse
Affiliation(s)
- Carlos J. Bethencourt-Estrella
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez s/n, 38203 La Laguna, Spain; (C.J.B.-E.); (A.L.-A.); (D.S.N.-H.)
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez s/n, 38203 La Laguna, Spain
- Red de Investigación Cooperativa en Enfermedades Tropicales (RICET), 28029 Madrid, Spain
| | - Nathalia Nocchi
- Instituto Universitario de Bio-Orgánica Antonio González (IUBO AG), Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez 2, 38206 La Laguna, Spain; (N.N.); (M.L.S.); (B.S.-G.)
- Departamento de Química Orgánica, Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez s/n, 38203 La Laguna, Spain
| | - Atteneri López-Arencibia
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez s/n, 38203 La Laguna, Spain; (C.J.B.-E.); (A.L.-A.); (D.S.N.-H.)
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez s/n, 38203 La Laguna, Spain
- Red de Investigación Cooperativa en Enfermedades Tropicales (RICET), 28029 Madrid, Spain
| | - Desirée San Nicolás-Hernández
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez s/n, 38203 La Laguna, Spain; (C.J.B.-E.); (A.L.-A.); (D.S.N.-H.)
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez s/n, 38203 La Laguna, Spain
- Red de Investigación Cooperativa en Enfermedades Tropicales (RICET), 28029 Madrid, Spain
| | - María L. Souto
- Instituto Universitario de Bio-Orgánica Antonio González (IUBO AG), Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez 2, 38206 La Laguna, Spain; (N.N.); (M.L.S.); (B.S.-G.)
- Departamento de Química Orgánica, Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez s/n, 38203 La Laguna, Spain
| | - Blanca Suárez-Gómez
- Instituto Universitario de Bio-Orgánica Antonio González (IUBO AG), Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez 2, 38206 La Laguna, Spain; (N.N.); (M.L.S.); (B.S.-G.)
| | - Ana R. Díaz-Marrero
- Instituto Universitario de Bio-Orgánica Antonio González (IUBO AG), Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez 2, 38206 La Laguna, Spain; (N.N.); (M.L.S.); (B.S.-G.)
- Correspondence: (A.R.D.-M.); (J.J.F.); (J.L.-M.); (J.E.P.)
| | - José J. Fernández
- Instituto Universitario de Bio-Orgánica Antonio González (IUBO AG), Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez 2, 38206 La Laguna, Spain; (N.N.); (M.L.S.); (B.S.-G.)
- Departamento de Química Orgánica, Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez s/n, 38203 La Laguna, Spain
- Correspondence: (A.R.D.-M.); (J.J.F.); (J.L.-M.); (J.E.P.)
| | - Jacob Lorenzo-Morales
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez s/n, 38203 La Laguna, Spain; (C.J.B.-E.); (A.L.-A.); (D.S.N.-H.)
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez s/n, 38203 La Laguna, Spain
- Red de Investigación Cooperativa en Enfermedades Tropicales (RICET), 28029 Madrid, Spain
- Consorcio Centro de Investigacion Biomedica en Red M.P. (CIBER) de Enfermedades Infecciosas, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (A.R.D.-M.); (J.J.F.); (J.L.-M.); (J.E.P.)
| | - José E. Piñero
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez s/n, 38203 La Laguna, Spain; (C.J.B.-E.); (A.L.-A.); (D.S.N.-H.)
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez s/n, 38203 La Laguna, Spain
- Red de Investigación Cooperativa en Enfermedades Tropicales (RICET), 28029 Madrid, Spain
- Consorcio Centro de Investigacion Biomedica en Red M.P. (CIBER) de Enfermedades Infecciosas, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (A.R.D.-M.); (J.J.F.); (J.L.-M.); (J.E.P.)
| |
Collapse
|
44
|
Gonçalves MD, Bortoleti BTDS, Tomiotto-Pellissier F, Concato VM, de Matos RLN, Silva TF, Rodrigues ACJ, Carloto ACM, Costa IN, Lazarin-Bidóia D, Miranda-Sapla MM, Pavanelli WR, Arakawa NS, Conchon-Costa I. Grandiflorenic acid isolated from Sphagneticola trilobata against Trypanosoma cruzi: Toxicity, mechanisms of action and immunomodulation. Toxicol In Vitro 2021; 78:105267. [PMID: 34688839 DOI: 10.1016/j.tiv.2021.105267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 11/30/2022]
Abstract
Grandiflorenic acid (GFA) is one of the main kaurane diterpenes found in different parts of Sphagneticola trilobata. It has several biological activities, especially antiprotozoal action. In turn, Chagas disease is a complex systemic disease caused by the protozoan Trypanosoma cruzi, and the drugs available to treat it involve significant side effects and impose an urgent need to search for therapeutic alternatives. In this context, our goal was to determine the effect of GFA on trypomastigote and intracellular amastigote forms. Our results showed that GFA treatment led to significantly less viability of trypomastigote forms, with morphological and ultrastructural changes in the parasites treated with IC50 of GFA (24.60 nM), and larger levels of reactive oxygen species (ROS), mitochondrial depolarization, lipid droplets accumulation, presence of autophagic vacuoles, phosphatidylserine exposure, and plasma membrane damage. In addition, the GFA treatment was able to reduce the percentage of infected cells and the number of amastigotes per macrophage (J774A.1) without showing cytotoxicity in mammalian cell lines (J774A.1, LLCMK2, THP-1, AMJ2-C11), in addition to increasing TNF-α and reducing IL-6 levels in infected macrophages. In conclusion, the GFA treatment exerted influence on trypomastigote forms through an apoptosis-like mechanism and by eliminating intracellular parasites via TNF-α/ROS pathway, without generating cellular cytotoxicity.
Collapse
Affiliation(s)
| | - Bruna Taciane da Silva Bortoleti
- Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, PR, Brazil; Biosciences and Biotechnology Postgraduate Program, Carlos Chagas Institute, ICC/Fiocruz/PR, Curitiba, PR, Brazil
| | - Fernanda Tomiotto-Pellissier
- Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, PR, Brazil; Biosciences and Biotechnology Postgraduate Program, Carlos Chagas Institute, ICC/Fiocruz/PR, Curitiba, PR, Brazil
| | - Virginia Marcia Concato
- Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, PR, Brazil
| | | | - Taylon Felipe Silva
- Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, PR, Brazil
| | - Ana Carolina Jacob Rodrigues
- Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, PR, Brazil; Biosciences and Biotechnology Postgraduate Program, Carlos Chagas Institute, ICC/Fiocruz/PR, Curitiba, PR, Brazil
| | | | - Idessania Nazareth Costa
- Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, PR, Brazil
| | - Danielle Lazarin-Bidóia
- Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, PR, Brazil
| | | | - Wander Rogério Pavanelli
- Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, PR, Brazil
| | - Nilton Syogo Arakawa
- Department of Chemical, Center of Exact Sciences, State University of Londrina, PR, Brazil
| | - Ivete Conchon-Costa
- Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, PR, Brazil
| |
Collapse
|
45
|
Bortoleti BTDS, Gonçalves MD, Tomiotto-Pellissier F, Camargo PG, Assolini JP, Concato VM, Detoni MB, Bidóia DL, Bispo MDLF, Lima CHDS, de Macedo FC, Conchon-Costa I, Miranda-Sapla MM, Wowk PF, Pavanelli WR. Investigation of the antileishmanial activity and mechanisms of action of acetyl-thiohydantoins. Chem Biol Interact 2021; 351:109690. [PMID: 34637778 DOI: 10.1016/j.cbi.2021.109690] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 09/28/2021] [Accepted: 10/07/2021] [Indexed: 12/20/2022]
Abstract
The currently available treatment options for leishmaniasis are associated with high costs, severe side effects, and high toxicity. In previous studies, thiohydantoins demonstrated some pharmacological activities and were shown to be potential hit compounds with antileishmanial properties. The present study further explored the antileishmanial effect of acetyl-thiohydantoins against Leishmania amazonensis and determined the main processes involved in parasite death. We observed that compared to thiohydantoin nuclei, acetyl-thiohydantoin treatment inhibited the proliferation of promastigotes. This treatment caused alterations in cell cycle progression and parasite size and caused morphological and ultrastructural changes. We then investigated the mechanisms involved in the death of the protozoan; there was an increase in ROS production, phosphatidylserine exposure, and plasma membrane permeabilization and a loss of mitochondrial membrane potential, resulting in an accumulation of lipid bodies and the formation of autophagic vacuoles on these parasites and confirming an apoptosis-like process. In intracellular amastigotes, selected acetyl-thiohydantoins reduced the percentage of infected macrophages and the number of amastigotes/macrophages by increasing ROS production and reducing TNF-α levels. Moreover, thiohydantoins did not induce cytotoxicity in murine macrophages (J774A.1), human monocytes (THP-1), or sheep erythrocytes. In silico and in vitro analyses showed that acetyl-thiohydantoins exerted in vitro antileishmanial effects on L. amazonensis promastigotes in apoptosis-like and amastigote forms by inducing ROS production and reducing TNF-α levels, indicating that they are good candidates for drug discovery studies in leishmaniasis treatment. Additionally, we carried out molecular docking analyses of acetyl-thiohydantoins on two important targets of Leishmania amazonensis: arginase and TNF-alpha converting enzyme. The results suggested that the acetyl groups in the N1-position of the thiohydantoin ring and the ring itself could be pharmacophoric groups due to their affinity for binding amino acid residues at the active site of both enzymes via hydrogen bond interactions. These results demonstrate that thiohydantoins are promising hit compounds that could be used as antileishmanial agents.
Collapse
Affiliation(s)
- Bruna Taciane da Silva Bortoleti
- Biosciences and Biotechnology Postgraduate Program, Carlos Chagas Institute, (ICC/Fiocruz/PR), Curitiba, Paraná, Brazil; State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil
| | - Manoela Daiele Gonçalves
- State University of Londrina (UEL/PR), Laboratory of Biotransformation and Phytochemistry, Londrina, Paraná, Brazil
| | - Fernanda Tomiotto-Pellissier
- Biosciences and Biotechnology Postgraduate Program, Carlos Chagas Institute, (ICC/Fiocruz/PR), Curitiba, Paraná, Brazil; State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil
| | - Priscila Goes Camargo
- State University of Londrina (UEL/PR), Laboratory of Research on Bioactive Molecules, Londrina, Paraná, Brazil
| | - João Paulo Assolini
- State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil
| | - Virginia Marcia Concato
- State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil
| | - Mariana Barbosa Detoni
- State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil
| | - Danielle Larazin Bidóia
- State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil
| | | | | | - Fernando Cesar de Macedo
- State University of Londrina (UEL/PR), Laboratory of Research on Bioactive Molecules, Londrina, Paraná, Brazil
| | - Ivete Conchon-Costa
- State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil
| | | | | | - Wander Rogério Pavanelli
- State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil.
| |
Collapse
|
46
|
Dos Santos NS, de Pinho FA, Hlavac NRC, Nunes TL, Almeida NR, Solcà MS, Varjão BM, Portela RW, Rugani JN, Rêgo FD, Barrouin-Melo SM, Soares RP. Feline Leishmaniasis Caused by Leishmania infantum: Parasite Sequencing, Seropositivity, and Clinical Characterization in an Endemic Area From Brazil. Front Vet Sci 2021; 8:734916. [PMID: 34513979 PMCID: PMC8424124 DOI: 10.3389/fvets.2021.734916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/02/2021] [Indexed: 11/13/2022] Open
Abstract
Zoonotic leishmaniasis caused by Leishmania infantum is a disease of One Health concern since human and animal cases and environmental damage are interconnected. L. infantum has a complex epidemiological cycle with multiple hosts, including mammals-humans, domestic, and wild animals-and arthropod vectors. Knowledge on mammal infections in endemic areas is crucial for developing control strategies. This work aimed to detect and characterize L. infantum infection in domestic cats from areas where human and canine leishmaniasis cases occur. No cases of feline leishmaniasis (FeL) had been previously reported in those areas. Five municipalities from Bahia state were chosen, comprising 2,480.8 km2 with 1,103,866 inhabitants. Ninety domiciliated and/or sheltered cats underwent clinical examination and serology by a rapid reference test recommended by the Brazilian government. Cytology, PCR, and parasite DNA sequencing were performed in bone marrow samples. Rapid tests detected antibodies in 5.6% (5/90) of the cats. Leishmania infantum infection was confirmed in 7.8% (7/90) of the cats by PCR, sequencing, and parasite isolation. Three out of the five municipalities (60%) had infected cats, and PCR positivity varied from 6.9 to 29%. One cat was categorized as harboring active L. infantum infection with amastigote forms in bone marrow smears. No clinical signs were detected at the first clinical exam, but 1 month later the cat developed severe FeL. The cat isolate was grown in culture, typed and its DNA sequence was homologous to the L. infantum reference strain (PP75). In conclusion, cats are potential hosts and may acquire L. infantum in endemic areas where canine and human cases occur. For cats, the need for surveillance, differential diagnosis and clinical care is highly recommended since a fast clinical progression of FeL developed in a subclinical animal. An accurate standardized immunodiagnostic assay for FeL is warranted.
Collapse
Affiliation(s)
- Nara Santos Dos Santos
- Laboratory of Veterinary Infectious Diseases, Teaching Hospital of Veterinary Medicine, Federal University of Bahia (UFBA), Salvador, Brazil
| | - Flaviane Alves de Pinho
- Laboratory of Veterinary Infectious Diseases, Teaching Hospital of Veterinary Medicine, Federal University of Bahia (UFBA), Salvador, Brazil.,Department of Veterinary Anatomy, Pathology and Clinics of the School of Veterinary Medicine and Zootechny, Federal University of Bahia, Salvador, Brazil
| | - Nicole Regina Capacchi Hlavac
- Department of Veterinary Anatomy, Pathology and Clinics of the School of Veterinary Medicine and Zootechny, Federal University of Bahia, Salvador, Brazil
| | - Talyta Lins Nunes
- Department of Veterinary Anatomy, Pathology and Clinics of the School of Veterinary Medicine and Zootechny, Federal University of Bahia, Salvador, Brazil
| | - Nádia Rossi Almeida
- Department of Veterinary Preventive Medicine and Animal Production of the School of Veterinary Medicine and Zootechny, Federal University of Bahia, Salvador, Brazil
| | - Manuela Silva Solcà
- Department of Veterinary Preventive Medicine and Animal Production of the School of Veterinary Medicine and Zootechny, Federal University of Bahia, Salvador, Brazil
| | - Bruno Milen Varjão
- Laboratory of Veterinary Infectious Diseases, Teaching Hospital of Veterinary Medicine, Federal University of Bahia (UFBA), Salvador, Brazil
| | - Ricardo Wagner Portela
- Laboratory of Immunology and Molecular Biology, Institute of Health Sciences, Federal University of Bahia, Salvador, BA, Brazil
| | | | - Felipe Dutra Rêgo
- Instituto Rene Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Stella Maria Barrouin-Melo
- Laboratory of Veterinary Infectious Diseases, Teaching Hospital of Veterinary Medicine, Federal University of Bahia (UFBA), Salvador, Brazil.,Department of Veterinary Anatomy, Pathology and Clinics of the School of Veterinary Medicine and Zootechny, Federal University of Bahia, Salvador, Brazil
| | | |
Collapse
|
47
|
Amentoflavone isolated from Selaginella sellowii Hieron induces mitochondrial dysfunction in Leishmania amazonensis promastigotes. Parasitol Int 2021; 86:102458. [PMID: 34509671 DOI: 10.1016/j.parint.2021.102458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/26/2021] [Accepted: 09/06/2021] [Indexed: 11/21/2022]
Abstract
Leishmaniasis chemotherapy is a bottleneck in disease treatment. Although available, chemotherapy is limited, toxic, painful, and does not lead to parasite clearance, with parasite resistance also being reported. Therefore, new therapeutic options are being investigated, such as plant-derived anti-parasitic compounds. Amentoflavone is the most common biflavonoid in the Selaginella genus, and its antileishmanial activity has already been described on Leishmania amazonensis intracellular amastigotes but its direct action on the parasite is controversial. In this work we demonstrate that amentoflavone is active on L. amazonensis promastigotes (IC50 = 28.5 ± 2.0 μM) and amastigotes. Transmission electron microscopy of amentoflavone-treated promastigotes showed myelin-like figures, autophagosomes as well as enlarged mitochondria. Treated parasites also presented multiple lipid droplets and altered basal body organization. Similarly, intracellular amastigotes presented swollen mitochondria, membrane fragments in the lumen of the flagellar pocket as well as autophagic vacuoles. Flow cytometric analysis after TMRE staining showed that amentoflavone strongly decreased mitochondrial membrane potential. In silico analysis shows that amentoflavone physic-chemical, drug-likeness and bioavailability characteristics suggest it might be suitable for oral administration. We concluded that amentoflavone presents a direct effect on L. amazonensis parasites, causing mitochondrial dysfunction and parasite killing. Therefore, all results point for the potential of amentoflavone as a promising candidate for conducting advanced studies for the development of drugs against leishmaniasis.
Collapse
|
48
|
Scalese G, Machado I, Salinas G, Pérez-Díaz L, Gambino D. Heteroleptic Oxidovanadium(V) Complexes with Activity against Infective and Non-Infective Stages of Trypanosoma cruzi. Molecules 2021; 26:5375. [PMID: 34500808 PMCID: PMC8433833 DOI: 10.3390/molecules26175375] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 12/18/2022] Open
Abstract
Five heteroleptic compounds, [VVO(IN-2H)(L-H)], where L are 8-hydroxyquinoline derivatives and IN is a Schiff base ligand, were synthesized and characterized in both the solid and solution state. The compounds were evaluated on epimastigotes and trypomastigotes of Trypanosoma cruzi as well as on VERO cells, as a mammalian cell model. Compounds showed activity against trypomastigotes with IC50 values of 0.29-3.02 μM. IN ligand and the new [VVO2(IN-H)] complex showed negligible activity. The most active compound [VVO(IN-2H)(L2-H)], with L2 = 5-chloro-7-iodo-8-hydroxyquinoline, showed good selectivity towards the parasite and was selected to carry out further biological studies. Stability studies suggested a partial decomposition in solution. [VVO(IN-2H)(L2-H)] affects the infection potential of cell-derived trypomastigotes. Low total vanadium uptake by parasites and preferential accumulation in the soluble proteins fraction were determined. A trypanocide effect was observed when incubating epimastigotes with 10 × IC50 values of [VVO(IN-2H)(L2-H)] and the generation of ROS after treatments was suggested. Fluorescence competition measurements with DNA:ethidium bromide adduct showed a moderate DNA interaction of the complexes. In vivo toxicity study on C. elegans model showed no toxicity up to a 100 μM concentration of [VVO(IN-2H)(L2-H)]. This compound could be considered a prospective anti-T. cruzi agent that deserves further research.
Collapse
Affiliation(s)
- Gonzalo Scalese
- Área Química Inorgánica, Facultad de Química, Universidad de la República, Montevideo 11800, Uruguay;
- Programa de Posgrados de la Facultad de Química, Universidad de la República, Montevideo 11800, Uruguay
| | - Ignacio Machado
- Área Química Analítica, Facultad de Química, Universidad de la República, Montevideo 11800, Uruguay;
| | - Gustavo Salinas
- Worm Biology Lab, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay;
- Departamento de Biociencias, Facultad de Química, Montevideo 11800, Uruguay
| | - Leticia Pérez-Díaz
- Laboratorio de Interacciones Moleculares, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay;
| | - Dinorah Gambino
- Área Química Inorgánica, Facultad de Química, Universidad de la República, Montevideo 11800, Uruguay;
| |
Collapse
|
49
|
Kourbeli V, Chontzopoulou E, Moschovou K, Pavlos D, Mavromoustakos T, Papanastasiou IP. An Overview on Target-Based Drug Design against Kinetoplastid Protozoan Infections: Human African Trypanosomiasis, Chagas Disease and Leishmaniases. Molecules 2021; 26:molecules26154629. [PMID: 34361781 PMCID: PMC8348971 DOI: 10.3390/molecules26154629] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 02/06/2023] Open
Abstract
The protozoan diseases Human African Trypanosomiasis (HAT), Chagas disease (CD), and leishmaniases span worldwide and therefore their impact is a universal concern. The present regimen against kinetoplastid protozoan infections is poor and insufficient. Target-based design expands the horizon of drug design and development and offers novel chemical entities and potential drug candidates to the therapeutic arsenal against the aforementioned neglected diseases. In this review, we report the most promising targets of the main kinetoplastid parasites, as well as their corresponding inhibitors. This overview is part of the Special Issue, entitled "Advances of Medicinal Chemistry against Kinetoplastid Protozoa (Trypanosoma brucei, Trypanosoma cruzi and Leishmania spp.) Infections: Drug Design, Synthesis and Pharmacology".
Collapse
Affiliation(s)
- Violeta Kourbeli
- Department of Pharmacy, Division of Pharmaceutical Chemistry, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupoli-Zografou, 157 84 Athens, Greece;
| | - Eleni Chontzopoulou
- Department of Organic Chemistry, Faculty of Chemistry, National and Kapodistrian University of Athens, Panepistimioupoli-Zografou, 157 71 Athens, Greece; (E.C.); (K.M.); (D.P.); (T.M.)
| | - Kalliopi Moschovou
- Department of Organic Chemistry, Faculty of Chemistry, National and Kapodistrian University of Athens, Panepistimioupoli-Zografou, 157 71 Athens, Greece; (E.C.); (K.M.); (D.P.); (T.M.)
| | - Dimitrios Pavlos
- Department of Organic Chemistry, Faculty of Chemistry, National and Kapodistrian University of Athens, Panepistimioupoli-Zografou, 157 71 Athens, Greece; (E.C.); (K.M.); (D.P.); (T.M.)
| | - Thomas Mavromoustakos
- Department of Organic Chemistry, Faculty of Chemistry, National and Kapodistrian University of Athens, Panepistimioupoli-Zografou, 157 71 Athens, Greece; (E.C.); (K.M.); (D.P.); (T.M.)
| | - Ioannis P. Papanastasiou
- Department of Pharmacy, Division of Pharmaceutical Chemistry, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupoli-Zografou, 157 84 Athens, Greece;
- Correspondence:
| |
Collapse
|
50
|
Glanzmann N, Antinarelli LMR, da Costa Nunes IK, Pereira HMG, Coelho EAF, Coimbra ES, da Silva AD. Synthesis and biological activity of novel 4-aminoquinoline/1,2,3-triazole hybrids against Leishmania amazonensis. Biomed Pharmacother 2021; 141:111857. [PMID: 34323702 DOI: 10.1016/j.biopha.2021.111857] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/18/2021] [Accepted: 06/24/2021] [Indexed: 01/07/2023] Open
Abstract
Quinoline and 1,2,3-triazoles are well-known nitrogen-based heterocycles presenting diverse pharmacological properties, although their antileishmanial activity is still poorly exploited. As an effort to contribute with studies involving these interesting chemical groups, in the present study, a series of compounds derived from 4-aminoquinoline and 1,2,3-triazole were synthetized and biological studies using L. amazonensis species were performed. The results pointed that the derivative 4, a hybrid of 4-aminoquinoline/1,2,3-triazole exhibited the best antileishmanial action, with inhibitory concentration (IC50) values of ~1 µM against intramacrophage amastigotes of L. amazonensis , and being 16-fold more active to parasites than to the host cell. The mechanism of action of derivative 4 suggest a multi-target action on Leishmania parasites, since the treatment of L. amazonensis promastigotes caused mitochondrial membrane depolarization, accumulation of ROS products, plasma membrane permeabilization, increase in neutral lipids, exposure of phosphatidylserine to the cell surface, changes in the cell cycle and DNA fragmentation. The results suggest that the antileishmanial effect of this compound is primarily altering critical biochemical processes for the correct functioning of organelles and macromolecules of parasites, with consequent cell death by processes related to apoptosis-like and necrosis. No up-regulation of reactive oxygen and nitrogen intermediates was promoted by derivative 4 on L. amazonensis -infected macrophages, suggesting a mechanism of action independent from the activation of the host cell. In conclusion, data suggest that derivative 4 presents selective antileishmanial effect, which is associated with multi-target action, and can be considered for future studies for the treatment against disease.
Collapse
Affiliation(s)
- Nícolas Glanzmann
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais 36.036-900, Brazil
| | - Luciana Maria Ribeiro Antinarelli
- Departamento de Parasitologia, Microbiologia e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais 36.036-900, Brazil; Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 30.130-100, Brazil
| | - Isabelle Karine da Costa Nunes
- Laboratório de Apoio ao Desenvolvimento Tecnológico, Polo de Química, Universidade Federal do Rio de Janeiro, Cidade Universitária Ilha do Fundão, Rio de Janeiro 21.941-598, Brazil
| | - Henrique Marcelo Gualberto Pereira
- Laboratório de Apoio ao Desenvolvimento Tecnológico, Polo de Química, Universidade Federal do Rio de Janeiro, Cidade Universitária Ilha do Fundão, Rio de Janeiro 21.941-598, Brazil
| | - 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, Minas Gerais 30.130-100, Brazil; Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Elaine Soares Coimbra
- Departamento de Parasitologia, Microbiologia e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais 36.036-900, Brazil
| | - Adilson David da Silva
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais 36.036-900, Brazil.
| |
Collapse
|