1
|
Silva RMGD, Barbosa FC, Santos HH, Granero FO, Figueiredo CCM, Nicolau-Junior N, Hamaguchi A, Silva LP. Antioxidant and anti-glycation activities of Mandevilla velutina extract and effect on parasitemia levels in Trypanosoma cruzi experimental infection: In vivo, in vitro and in silico approaches. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118994. [PMID: 39461387 DOI: 10.1016/j.jep.2024.118994] [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: 07/08/2024] [Revised: 09/27/2024] [Accepted: 10/23/2024] [Indexed: 10/29/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Mandevilla velutina (Mart. Ex Stadelm.) Woodson, known in Brazil as "infalível" and "jalapa", is a medicinal plant native from the Cerrado region (Brazilian Savannah). The underground organ (xylopodium) of this species is prepared as ethanolic extract or infusion and it is commonly used in traditional medicine to treat snake venom. Although, locals and indigenous populations from Cerrado have used M. velutina for the treatment of infection by Trypanosoma cruzi (Chagas' disease). AIM OF THE STUDY This study aimed to evaluate the in vitro antioxidant and anti-glycation activities of the crude hydroethanolic extract of M. velutina xylopodium. Besides, it aimed to evaluate its effect on parasitemia levels in vivo T. cruzi experimental infection. In addition, this study aimed to determine possible interactions between the main compound of the extract and molecular targets associated with survival and virulence of T. cruzi in silico approaches. MATERIALS AND METHODS Determination of total polyphenols, flavonoids and steroidal aglycones content were performed. In addition, high performance liquid chromatography (HPLC) was carried out to identify main compounds of the extract. Antioxidant activity was determined by DPPH radical scavenging, ferric ion reducing power (FRAP), Thiobarbituric acid reactive species (TBARS) and Oxygen Radical Absorbance Capacity (ORAC) methods. Anti-glycation activity was demonstrated through relative mobility in electrophoresis (RME), determination of free amino groups and inhibition of AGEs formation. Determination of the action of extract in parasitemia levels was performed by T. cruzi experimental infection of mice and nitrite levels were measured in the serum of animals evaluated in this study. Molecular docking analyses of the main compound (Velutinol A) with DNA and molecular targets associated with survival and virulence of T. cruzi. RESULTS Phytoconstituents evaluation exhibited the presence polyphenols, flavonoids and steroidal aglycone, and HPLC identified the major presence of Velutinol A. Antioxidant and anti-glycation evaluations showed that the extract present significant activity in all methods evaluated. In addition, extract reduced the number of trypomastigotes and increased the survival of treated animals. The treatment using extract showed an interference in the synthesis of physiological nitric oxide as an immune response to infection. In silico assays demonstrated interaction between Velutinol A and DNA and molecular targets of T. cruzi. CONCLUSIONS The results showed that the hydroethanolic extract of M. velutina xylopodium contains bioactive compounds including polyphenols, flavonoids and steroidal aglycones (mainly Velutinol A) of which may be responsible for the antioxidant, anti-glycation and anti-parasitic activity against T. cruzi. Trypanocidal activity of M. velutina compounds may be linked to their influence on NO synthesis during infection and/or their capacity to bind and inhibit molecules associated to virulence and survival of T. cruzi.
Collapse
Affiliation(s)
- Regildo Márcio Gonçalves da Silva
- São Paulo State University (UNESP), School of Sciences, Humanities and Languages, Department of Biotechnology, Laboratory of Herbal Medicine and Natural Products, Assis, São Paulo, Brazil; São Paulo State University (UNESP), Institute of Chemistry, Araraquara, São Paulo, Brazil.
| | - Fernando Cesar Barbosa
- São Paulo State University (UNESP), School of Sciences, Humanities and Languages, Department of Biotechnology, Laboratory of Herbal Medicine and Natural Products, Assis, São Paulo, Brazil
| | - Hugo Henrique Santos
- São Paulo State University (UNESP), School of Sciences, Humanities and Languages, Department of Biotechnology, Laboratory of Herbal Medicine and Natural Products, Assis, São Paulo, Brazil
| | | | | | - Nilson Nicolau-Junior
- Federal University of Uberlândia (UFU), Institute of Biotechnology, Laboratory of Molecular Modeling, Uberlândia, Minas Gerais, Brazil
| | - Amélia Hamaguchi
- Federal University of Uberlândia (UFU), Institute of Biotechnology, Uberlândia, Minas Gerais, Brazil
| | | |
Collapse
|
2
|
Medeiros TS, Bezerra de Lima LE, Alves-Pereira EL, Alves-Silva MF, Dourado D, Fernandes-Pedrosa MDF, Figueiredo RCBQD, da Silva-Junior AA. Cationic and anionic PLGA-cholesterol hybrid nanoparticles as promising platforms to enhance the trypanocidal efficacy of benznidazole and drug delivery in Trypanosoma cruzi-infected cells. Biomed Pharmacother 2025; 183:117782. [PMID: 39755025 DOI: 10.1016/j.biopha.2024.117782] [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: 10/25/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 01/06/2025] Open
Abstract
Chagas disease is a neglected tropical disease caused by the protozoan Trypanosoma cruzi, remains a significant global health challenge. Currently, benznidazole (BNZ) is the primary treatment in many countries. However, this drug is limited by low bioavailability, significant host toxicity, and reduced efficacy in chronic disease phase. Additionally, cases of parasite resistance to treatment and low efficacy in in chronic disease phase have been reported. In this context, nanotechnology formulations for intracellular drug delivery have emerged as a promising alternative to improve the pharmacological properties of BNZ. In this study, we developed and evaluated cationic and anionic PLGA-cholesterol hybrid nanoparticles (HNPs) as innovative drug delivery systems for BNZ. These HNPs, functionalized with polyethyleneimine, were synthesized using a composition-dependent self-assembly method, yielding stable nanosystems with tuneable physicochemical properties. Furthermore, four release kinetic models were applied and Peppas-Sahlin demonstrated the best fit. In vitro assays confirmed the biocompatibility of HNPs with cardiomyoblasts at tested concentrations and revealed significantly enhanced trypanocidal activity against intracellular amastigotes compared to free BNZ. Transmission electron microscopy and fluorescence microscopy analyses highlighted effective nanoparticle internalization, with superior performance trypanocidal observed in anionic HNPs, which can be attributed to the residence of cationic in endo/lysosomal vesicles. Taken together, our results demonstrate the successful development of HNPs, underscoring their potential as a promising platform for the intracellular delivery of therapeutic agents.
Collapse
Affiliation(s)
- Thayse Silva Medeiros
- Laboratory of Pharmaceutical Technology and Biotechnology, Department of Pharmacy, Federal University of Rio Grande do Norte-UFRN, Natal, RN, Brazil; Laboratory of Cellular Biology of Pathogens, Department of Microbiology, Aggeu Magalhães Institute/FIOCRUZ-PE, Recife, PE, Brazil
| | - Lucas Eduardo Bezerra de Lima
- Laboratory of Cellular Biology of Pathogens, Department of Microbiology, Aggeu Magalhães Institute/FIOCRUZ-PE, Recife, PE, Brazil
| | - Eron Lincoln Alves-Pereira
- Laboratory of Pharmaceutical Technology and Biotechnology, Department of Pharmacy, Federal University of Rio Grande do Norte-UFRN, Natal, RN, Brazil
| | - Mariana Farias Alves-Silva
- Laboratory of Pharmaceutical Technology and Biotechnology, Department of Pharmacy, Federal University of Rio Grande do Norte-UFRN, Natal, RN, Brazil
| | - Douglas Dourado
- Laboratory of Immunopathology and Molecular Biology, Aggeu Magalhães Institute/FIOCRUZ-PE, Recife, PE, Brazil
| | - Matheus de Freitas Fernandes-Pedrosa
- Laboratory of Pharmaceutical Technology and Biotechnology, Department of Pharmacy, Federal University of Rio Grande do Norte-UFRN, Natal, RN, Brazil
| | | | - Arnóbio Antônio da Silva-Junior
- Laboratory of Pharmaceutical Technology and Biotechnology, Department of Pharmacy, Federal University of Rio Grande do Norte-UFRN, Natal, RN, Brazil.
| |
Collapse
|
3
|
Chaudhary J, Kaur G, Singh I. Synthesis strategies and anti-parasitic evaluation of novel compounds for chagas disease: Advancing drug discovery through structure-activity relationships. Eur J Med Chem 2024; 284:117203. [PMID: 39740321 DOI: 10.1016/j.ejmech.2024.117203] [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: 11/04/2024] [Revised: 12/14/2024] [Accepted: 12/22/2024] [Indexed: 01/02/2025]
Abstract
This study presents a comprehensive exploration of the synthesis of novel compounds targeting Chagas Disease (CD) caused by Trypanosoma cruzi. It is a global health threat with over 6-7 million infections worldwide. Addressing challenges in current treatments, the investigation explores diverse compound classes, including thiazoles, thiazolidinone, imidazole, pyrazole, 1,6-diphenyl-1H-pyrazolo[3,4-b] pyridine, pyrrole, naphthoquinone, neolignan, benzeneacyl hydrazones, and chalcones-based compounds. Highlighting compounds with superior trypanocidal activity compared to standard drugs. The study elucidates structure-activity relationships, emphasizing the impact of substituents, fluorine presence, and substitution patterns. Noteworthy findings include neolignan derivatives demonstrating efficacy against intracellular amastigotes and free-moving trypomastigotes, with unsaturated side chains. Benzeneacylhydrazones and chalcones, as novel classes, showed varied efficacy, with certain compounds surpassing benznidazole. A novel series of triketone compounds exhibited strong anti-parasitic activity, outperforming standard drugs. Docking study revealed that the halogen and methoxy substituted phenyl ring, thiazole, thiazolidine-4-one, quinoline, isoindoline-1,3-dione, pyrrole heterocyclic motifs can play the key role in the designing of effective inhibitors of T. cruzi. Mutually, these insights placed the foundation for the development of innovative and effective treatments for CD, addressing the urgent need for improved therapeutic options.
Collapse
Affiliation(s)
- Jitendra Chaudhary
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, India
| | - Gurdeep Kaur
- School of Chemical Engineering and Physical Sciences, Lovely Professional University, Phagwara, 144411, India
| | - Iqubal Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, India.
| |
Collapse
|
4
|
Boya P. CA, Rodriguez C, Mojica-Flores R, Urrutia JC, Cantilo-Diaz V, Barrios-Jaén M, Ng MG, Pineda L, Llanes A, Spadafora C, Mejía LC, Gutiérrez M. Antiprotozoal Natural Products from Endophytic Fungi Associated with Cacao and Coffee. Metabolites 2024; 14:575. [PMID: 39590811 PMCID: PMC11596112 DOI: 10.3390/metabo14110575] [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: 09/20/2024] [Revised: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Collectively, leishmaniasis and Chagas disease cause approximately 8 million cases and more than 40,000 deaths annually, mostly in tropical and subtropical regions. The current drugs used to treat these diseases have limitations and many undesirable side effects; hence, new drugs with better clinical profiles are needed. Fungal endophytes associated with plants are known to produce a wide array of bioactive secondary metabolites, including antiprotozoal compounds. In this study, we analyzed endophytic fungal isolates associated with Theobroma cacao and Coffea arabica crop plants, which yielded extracts with antitrypanosomatid activity. METHODS Crude extracts were subjected to bioassay-guided isolation by HPLC, followed by spectrometric and spectroscopic analyses via mass spectrometry (MS) and nuclear magnetic resonance (NMR), Results: Compounds 1-9 were isolated and displayed novel antitrypanosomal and antileishmanial activities ranging from 0.92 to 32 μM. Tandem liquid chromatography-mass spectrometry (LC-MS) analysis of the organic extracts from different strains via the feature-based Global Natural Products Social (GNPS) molecular networking platform allowed us to dereplicate a series of metabolites (10-23) in the extracts. Molecular docking simulations of the active compounds, using the 3-mercaptopyruvate sulfurtransferase protein from L. donovani (Ld3MST) and the cruzipain enzyme from T. cruzi as putative molecular targets, allowed us to suggest possible mechanisms for the action of these compounds. CONCLUSIONS The isolation of these antiprotozoal compounds confirms that crop plants like coffee and cacao harbor populations of endophytes with biomedical potential that confer added value to these crops.
Collapse
Affiliation(s)
- Cristopher A. Boya P.
- Centro de Biodiversidad y Descubrimiento de Drogas, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (C.A.B.P.); (C.R.); (R.M.-F.); (J.C.U.); (V.C.-D.); (M.B.-J.); (L.C.M.)
| | - Candelario Rodriguez
- Centro de Biodiversidad y Descubrimiento de Drogas, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (C.A.B.P.); (C.R.); (R.M.-F.); (J.C.U.); (V.C.-D.); (M.B.-J.); (L.C.M.)
- Estación Científica COIBA AIP, Ciudad del Saber, Panamá 0816-02852, Panama
| | - Randy Mojica-Flores
- Centro de Biodiversidad y Descubrimiento de Drogas, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (C.A.B.P.); (C.R.); (R.M.-F.); (J.C.U.); (V.C.-D.); (M.B.-J.); (L.C.M.)
| | - Jean Carlo Urrutia
- Centro de Biodiversidad y Descubrimiento de Drogas, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (C.A.B.P.); (C.R.); (R.M.-F.); (J.C.U.); (V.C.-D.); (M.B.-J.); (L.C.M.)
| | - Víctor Cantilo-Diaz
- Centro de Biodiversidad y Descubrimiento de Drogas, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (C.A.B.P.); (C.R.); (R.M.-F.); (J.C.U.); (V.C.-D.); (M.B.-J.); (L.C.M.)
| | - Masiel Barrios-Jaén
- Centro de Biodiversidad y Descubrimiento de Drogas, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (C.A.B.P.); (C.R.); (R.M.-F.); (J.C.U.); (V.C.-D.); (M.B.-J.); (L.C.M.)
| | - Michelle G. Ng
- Centro de Biología Molecular y Celular de Enfermedades (CBCME), Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (M.G.N.); (L.P.); (A.L.); (C.S.)
| | - Laura Pineda
- Centro de Biología Molecular y Celular de Enfermedades (CBCME), Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (M.G.N.); (L.P.); (A.L.); (C.S.)
| | - Alejandro Llanes
- Centro de Biología Molecular y Celular de Enfermedades (CBCME), Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (M.G.N.); (L.P.); (A.L.); (C.S.)
| | - Carmenza Spadafora
- Centro de Biología Molecular y Celular de Enfermedades (CBCME), Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (M.G.N.); (L.P.); (A.L.); (C.S.)
| | - Luis C. Mejía
- Centro de Biodiversidad y Descubrimiento de Drogas, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (C.A.B.P.); (C.R.); (R.M.-F.); (J.C.U.); (V.C.-D.); (M.B.-J.); (L.C.M.)
- Smithsonian Tropical Research Institute, Ancón 0843-03092, Panama
| | - Marcelino Gutiérrez
- Centro de Biodiversidad y Descubrimiento de Drogas, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panamá 0843-01103, Panama; (C.A.B.P.); (C.R.); (R.M.-F.); (J.C.U.); (V.C.-D.); (M.B.-J.); (L.C.M.)
| |
Collapse
|
5
|
Silva GM, Gomes SQ, Lopes CD, de Albuquerque S, de Paula da Silva CHT. Structural analysis and shape-based identification of novel inhibitors targeting the Trypanosoma cruzi proteasome. Int J Biol Macromol 2024; 277:134290. [PMID: 39084432 DOI: 10.1016/j.ijbiomac.2024.134290] [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: 06/13/2024] [Revised: 07/17/2024] [Accepted: 07/28/2024] [Indexed: 08/02/2024]
Abstract
There is an urgent need to develop new, safer, and more effective drugs against Chagas disease (CD) as well as related kinetoplastid diseases. Targeting and inhibiting the Trypanosoma cruzi proteasome has emerged as a promising therapeutic approach in this context. To expand the chemical space for this class of inhibitors, we performed virtual screening campaigns with emphasis on shape-based similarity and ADMET prioritization. We describe the ideation and application of robustly validated shape queries for these campaigns, which furnished 44 compounds for biological evaluation. Five hit compounds demonstrated in vitro antitrypanosomal activity by potential inhibition of T. cruzi proteasome and notable chemical diversities, particularly, LCQFTC11. Structural insights were achieved by homology modeling, sequence/structure alignment, proteasome-species comparison, docking, molecular dynamics, and MMGBSA binding affinity estimations. These methods confirmed key interactions as well as the stability of LCQFTC11 at the β4/β5 subunits' binding site of the T. cruzi proteasome, consistent with known inhibitors. Our results warrant future assay confirmation of our hit as a T. cruzi proteasome inhibitor. Importantly, we also shed light into dynamic details for a proteasome inhibition mechanism that shall be further investigated. We expect to contribute to the development of viable CD drug candidates through such a relevant approach.
Collapse
Affiliation(s)
- Guilherme Martins Silva
- Center for Drug Discovery and Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA; Computational Laboratory of Pharmaceutical Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, s/n, Ribeirão Preto, SP 14040-903, Brazil.
| | - Suzane Quintana Gomes
- Computational Laboratory of Pharmaceutical Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, s/n, Ribeirão Preto, SP 14040-903, Brazil
| | - Carla Duque Lopes
- Laboratório de Parasitologia, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, s/n, Ribeirão Preto, SP 14040-903, Brazil
| | - Sérgio de Albuquerque
- Laboratório de Parasitologia, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, s/n, Ribeirão Preto, SP 14040-903, Brazil
| | - Carlos Henrique Tomich de Paula da Silva
- Computational Laboratory of Pharmaceutical Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, s/n, Ribeirão Preto, SP 14040-903, Brazil
| |
Collapse
|
6
|
Marinho MM, da Rocha MN, Magalhães EP, Ribeiro LR, Roberto CHA, de Queiroz Almeida-Neto FW, Monteiro ML, Nunes JVS, de Menezes RRPPB, Marinho ES, de Lima Neto P, Martins AMC, Dos Santos HS. Insights of potential trypanocidal effect of the synthetic derivative (2E)-1-(4-aminophenyl)-3-(2,4-dichlorophenyl)prop-2-en-1-one: in vitro assay, MEV analysis, quantum study, molecular docking, molecular dynamics, MPO analysis, and predictive ADMET. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7797-7818. [PMID: 38722342 DOI: 10.1007/s00210-024-03138-z] [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: 06/19/2023] [Accepted: 04/30/2024] [Indexed: 10/04/2024]
Abstract
This study aims to evaluate the antitrypanosomiasis activity of a synthetic dichloro-substituted aminochalcone via in vitro assays against infected cell cultures, as well as a theoretical characterization of pharmacokinetics and pharmacodynamics against the protein targets of the evolutionary cycle of T. cruzi. The in vitro evaluation of parasite proliferation inhibition was performed via cytotoxicity analysis on mammalian host cells, effect on epimastigote and trypomastigote forms, and cell death analysis, while computer simulations characterized the electronic structure of (2E)-1-(4-aminophenyl)-3-(2,4-dichlorophenyl)prop-2-en-1-one (DCl), the mechanism of action against the proteins of the evolutionary cycle of T. cruzi: Cruzain, Trypanothione reductase, TcGAPDH, and CYP51 by molecular docking and dynamics and predictive pharmacokinetics by MPO-based ADMET. The in vitro tests showed that the DCl LC50 in order of 178.9 ± 23.9 was similar to the BZN, evidencing the effectiveness of chalcone against Trypomastigotes. Molecular docking and dynamics simulations suggest that DCl acts on the active site of the CYP51 receptor, with hydrogen interactions that showed a high degree of occupation, establishing a stable complex with the target. MPO analysis and ADMET prediction tests suggest that the compound presents an alignment between permeability and hepatic clearance, although it presents low metabolic stability. Chalcone showed stable pharmacodynamics against the CYP51 target, but can form reactive metabolites from N-conjugation and C = C epoxidation, as an indication of controlled oral dose, although the estimated LD50 rate > 500 mg/kg is a indicative of low incidence of lethality by ingestion, constituting a promising therapeutic strategy.
Collapse
Affiliation(s)
- Márcia Machado Marinho
- Department of Biological Chemistry, Regional University of Cariri, Crato, CE, Brazil
- Center for Exact Sciences and Technology, State University of Vale do Acaraú, Sobral, CE, Brazil
| | - Matheus Nunes da Rocha
- Center for Science and Technology, Postgraduate Program in Natural Sciences, State University of Ceará, Fortaleza, CE, Brazil
| | - Emanuel Paula Magalhães
- Department of Clinical and Toxicological Analysis, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Lyanna Rodrigues Ribeiro
- Department of Clinical and Toxicological Analysis, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Caio Henrique Alexandre Roberto
- Center for Science and Technology, Postgraduate Program in Natural Sciences, State University of Ceará, Fortaleza, CE, Brazil
| | | | - Marília Lopes Monteiro
- Department of Clinical and Toxicological Analysis, Federal University of Ceará, Fortaleza, CE, Brazil
| | - João Victor Serra Nunes
- Department of Clinical and Toxicological Analysis, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - Emmanuel Silva Marinho
- Center for Science and Technology, Postgraduate Program in Natural Sciences, State University of Ceará, Fortaleza, CE, Brazil
| | - Pedro de Lima Neto
- Department of Analytical Chemistry and Physical Chemistry, Federal University of Ceará, Campus do Pici, Fortaleza, CE, Brazil
| | - Alice Maria Costa Martins
- Department of Clinical and Toxicological Analysis, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Hélcio Silva Dos Santos
- Center for Exact Sciences and Technology, State University of Vale do Acaraú, Sobral, CE, Brazil.
| |
Collapse
|
7
|
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
|
8
|
Rana MM, Lu S, Menjívar TA, Fukaya K, Nakajima-Shimada J, Urabe D, Igarashi Y. Cladoic Acid, an Anti- Trypanosoma cruzi Polyketide from Cladosporium sp. TP-F2020. JOURNAL OF NATURAL PRODUCTS 2024; 87:2126-2131. [PMID: 39101838 DOI: 10.1021/acs.jnatprod.4c00567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/06/2024]
Abstract
A new polyketide, cladoic acid, was isolated from a fungus of the genus Cladosporium. The structure of the highly oxygenated trans-decalin ring with an all-E triene side chain was elucidated by extensive spectroscopic analysis. The unique chair/twist-boat conformation of the trans-decalin core and the flexibility of the B-ring were demonstrated by computer-aided conformational analysis. Cladoic acid was active against Trypanosoma cruzi and inhibited the proliferation of amastigotes and epimastigotes with IC50 values of 27 and 46 μM, respectively, but it did not show any appreciable activity against P388 murine leukemia cells, bacteria, or fungi, indicating it is a potential candidate for drug development against Chagas disease.
Collapse
Affiliation(s)
- Md Masud Rana
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, 5180 Kurokawa, Imizu-shi, Toyama 939-0398, Japan
| | - Shiyang Lu
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, 5180 Kurokawa, Imizu-shi, Toyama 939-0398, Japan
| | - Tatiana Ascencio Menjívar
- Department of Microbiology, School of Medicine, Dr. José Matías Delgado University, Campus 1, km 8 1/2 Carretera a Santa Tecla, San Salvador 01101, El Salvador
- Graduate School of Health Sciences, Gunma University, 3-39-22, Showa-machi, Maebashi, Gunma 371-8510, Japan
| | - Keisuke Fukaya
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, 5180 Kurokawa, Imizu-shi, Toyama 939-0398, Japan
| | - Junko Nakajima-Shimada
- Graduate School of Health Sciences, Gunma University, 3-39-22, Showa-machi, Maebashi, Gunma 371-8510, Japan
| | - Daisuke Urabe
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, 5180 Kurokawa, Imizu-shi, Toyama 939-0398, Japan
| | - Yasuhiro Igarashi
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, 5180 Kurokawa, Imizu-shi, Toyama 939-0398, Japan
| |
Collapse
|
9
|
Slafer B, Dessoy MA, de Oliveira RG, Mollo MC, Lee E, Matheeussen A, Maes L, Caljon G, Ferreira LLG, Krogh R, Andricopulo AD, Cruz LR, Mowbray CE, Kratz JM, Dias LC. Synthesis and Anti- Trypanosoma cruzi Activity of 3-Cyanopyridine Derivatives. ACS OMEGA 2024; 9:22360-22370. [PMID: 38799347 PMCID: PMC11112591 DOI: 10.1021/acsomega.4c01919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/14/2024] [Accepted: 04/17/2024] [Indexed: 05/29/2024]
Abstract
Chagas disease (CD) is a parasitic neglected tropical disease (NTD) caused by the protozoan Trypanosoma cruzi that affects 6 million people worldwide, often resulting in financial burden, morbidity, and mortality in endemic regions. Given a lack of highly efficient and safe treatments, new, affordable, and fit-for-purpose drugs for CD are urgently needed. In this work, we present a hit-to-lead campaign for novel cyanopyridine analogues as antichagasic agents. In a phenotypic screening against intracellular T. cruzi, hits 1 and 2 were identified and displayed promising potency combined with balanced physicochemical properties. As part of the Lead Optimization Latin America consortium, a set of 40 compounds was designed, synthesized, and tested against T. cruzi intracellular amastigotes and relevant human cell lines. The structural modifications were focused on three positions: cyanopyridine core, linker, and right-hand side. The ADME properties of selected compounds, lipophilicity, kinetic solubility, permeability, and liver microsomal stability, were evaluated. Compounds 1-9 displayed good potency (EC50T. cruzi amastigote <1 μM), and most compounds did not present significant cytotoxicity (CC50 MRC-5 = 32-64 μM). Despite the good balance between potency and selectivity, the antiparasitic activity of the series appeared to be driven by lipophilicity, making the progression of the series unfeasible due to poor ADME properties and potential promiscuity issues.
Collapse
Affiliation(s)
- Brian
W. Slafer
- Institute
of Chemistry, State University of Campinas, Campinas 13083-970, São Paulo, Brazil
| | - Marco A. Dessoy
- Institute
of Chemistry, State University of Campinas, Campinas 13083-970, São Paulo, Brazil
| | - Ramon G. de Oliveira
- Institute
of Chemistry, State University of Campinas, Campinas 13083-970, São Paulo, Brazil
| | - Maria C. Mollo
- Institute
of Chemistry, State University of Campinas, Campinas 13083-970, São Paulo, Brazil
| | - Eun Lee
- Institute
of Chemistry, State University of Campinas, Campinas 13083-970, São Paulo, Brazil
| | - An Matheeussen
- Laboratory
of Microbiology, Parasitology and Hygiene, University of Antwerp, Antwerp 2000, Belgium
| | - Louis Maes
- Laboratory
of Microbiology, Parasitology and Hygiene, University of Antwerp, Antwerp 2000, Belgium
| | - Guy Caljon
- Laboratory
of Microbiology, Parasitology and Hygiene, University of Antwerp, Antwerp 2000, Belgium
| | - Leonardo L. G. Ferreira
- Laboratory
of Medicinal and Computational Chemistry, Physics Institute of Sao Carlos, University of Sao Paulo, Sao Carlos 13566-590, São Paulo, Brazil
| | - Renata Krogh
- Laboratory
of Medicinal and Computational Chemistry, Physics Institute of Sao Carlos, University of Sao Paulo, Sao Carlos 13566-590, São Paulo, Brazil
| | - Adriano D. Andricopulo
- Laboratory
of Medicinal and Computational Chemistry, Physics Institute of Sao Carlos, University of Sao Paulo, Sao Carlos 13566-590, São Paulo, Brazil
| | - Luiza R. Cruz
- Drugs
for Neglected Diseases initiative, Geneva 1202, Switzerland
| | | | - Jadel M. Kratz
- Drugs
for Neglected Diseases initiative, Geneva 1202, Switzerland
| | - Luiz C. Dias
- Institute
of Chemistry, State University of Campinas, Campinas 13083-970, São Paulo, Brazil
| |
Collapse
|
10
|
Parvez A, Lee JS, Alam W, Tayara H, Chong KT. Integrated Computational Approaches for Drug Design Targeting Cruzipain. Int J Mol Sci 2024; 25:3747. [PMID: 38612558 PMCID: PMC11011879 DOI: 10.3390/ijms25073747] [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: 01/30/2024] [Revised: 03/15/2024] [Accepted: 03/24/2024] [Indexed: 04/14/2024] Open
Abstract
Cruzipain inhibitors are required after medications to treat Chagas disease because of the need for safer, more effective treatments. Trypanosoma cruzi is the source of cruzipain, a crucial cysteine protease that has driven interest in using computational methods to create more effective inhibitors. We employed a 3D-QSAR model, using a dataset of 36 known inhibitors, and a pharmacophore model to identify potential inhibitors for cruzipain. We also built a deep learning model using the Deep purpose library, trained on 204 active compounds, and validated it with a specific test set. During a comprehensive screening of the Drug Bank database of 8533 molecules, pharmacophore and deep learning models identified 1012 and 340 drug-like molecules, respectively. These molecules were further evaluated through molecular docking, followed by induced-fit docking. Ultimately, molecular dynamics simulation was performed for the final potent inhibitors that exhibited strong binding interactions. These results present four novel cruzipain inhibitors that can inhibit the cruzipain protein of T. cruzi.
Collapse
Affiliation(s)
- Aiman Parvez
- Department of Electronics and Information Engineering, Jeonbuk National University, Jeonju 54896, Republic of Korea; (A.P.); (W.A.)
| | - Jeong-Sang Lee
- Department of Functional Food and Biotechnology, College of Medical Sciences, Jeonju University, Jeonju 55069, Republic of Korea;
| | - Waleed Alam
- Department of Electronics and Information Engineering, Jeonbuk National University, Jeonju 54896, Republic of Korea; (A.P.); (W.A.)
| | - Hilal Tayara
- School of International Engineering and Science, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Kil To Chong
- Department of Electronics and Information Engineering, Jeonbuk National University, Jeonju 54896, Republic of Korea; (A.P.); (W.A.)
- Advances Electronics and Information Research Center, Jeonbuk National University, Jeonju 54896, Republic of Korea
| |
Collapse
|
11
|
Berhe H, Kumar Cinthakunta Sridhar M, Zerihun M, Qvit N. The Potential Use of Peptides in the Fight against Chagas Disease and Leishmaniasis. Pharmaceutics 2024; 16:227. [PMID: 38399281 PMCID: PMC10892537 DOI: 10.3390/pharmaceutics16020227] [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: 11/12/2023] [Revised: 12/28/2023] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Chagas disease and leishmaniasis are both neglected tropical diseases that affect millions of people around the world. Leishmaniasis is currently the second most widespread vector-borne parasitic disease after malaria. The World Health Organization records approximately 0.7-1 million newly diagnosed leishmaniasis cases each year, resulting in approximately 20,000-30,000 deaths. Also, 25 million people worldwide are at risk of Chagas disease and an estimated 6 million people are infected with Trypanosoma cruzi. Pentavalent antimonials, amphotericin B, miltefosine, paromomycin, and pentamidine are currently used to treat leishmaniasis. Also, nifurtimox and benznidazole are two drugs currently used to treat Chagas disease. These drugs are associated with toxicity problems such as nephrotoxicity and cardiotoxicity, in addition to resistance problems. As a result, the discovery of novel therapeutic agents has emerged as a top priority and a promising alternative. Overall, there is a need for new and effective treatments for Chagas disease and leishmaniasis, as the current drugs have significant limitations. Peptide-based drugs are attractive due to their high selectiveness, effectiveness, low toxicity, and ease of production. This paper reviews the potential use of peptides in the treatment of Chagas disease and leishmaniasis. Several studies have demonstrated that peptides are effective against Chagas disease and leishmaniasis, suggesting their use in drug therapy for these diseases. Overall, peptides have the potential to be effective therapeutic agents against Chagas disease and leishmaniasis, but more research is needed to fully investigate their potential.
Collapse
Affiliation(s)
| | | | | | - Nir Qvit
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Safed 1311502, Israel; (H.B.); (M.K.C.S.); (M.Z.)
| |
Collapse
|
12
|
Juarez-Saldivar A, Gómez-Escobedo R, Corral-Ruiz G, Chacón-Vargas KF, Horta-Montaño V, Sanchez-Torres L, Vazquez-Jimenez LK, Nogueda-Torres B, Rivera G. Repositioning FDA-Approved Drug Against Chagas Disease and Cutaneous Leishmaniosis by Structure-Based Virtual Screening. Arch Med Res 2024; 55:102958. [PMID: 38290200 DOI: 10.1016/j.arcmed.2024.102958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 12/13/2023] [Accepted: 01/11/2024] [Indexed: 02/01/2024]
Abstract
BACKGROUND Chagas disease and cutaneous leishmaniasis, two parasitic diseases caused by Trypanosoma cruzi (T. cruzi) and Leishmania mexicana (L. mexicana), respectively, have a major global impact. Current pharmacological treatments for these diseases are limited and can cause severe side effects; thus, there is a need for new antiprotozoal drugs. METHODS Using molecular docking, this work describes a structure-based virtual screening of an FDA-approved drug library against Trypanosoma cruzi and Leishmania mexicana glycolytic enzyme triosephosphate isomerase (TIM), which is highly conserved in these parasites. The selected compounds with potential dual inhibitory activity were tested in vitro to confirm their biological activity. RESULTS The study showed that five compounds: nilotinib, chlorhexidine, protriptyline, cyproheptadine, and montelukast, were more active against T. cruzi, than the reference drugs, nifurtimox and benznidazole while chlorhexidine and protriptyline were the most active against L. mexicana. CONCLUSIONS The analysis of these compounds and their structural characteristics may provide the basis for the development of new antiprotozoal agents.
Collapse
Affiliation(s)
- Alfredo Juarez-Saldivar
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa, Tamaulipas, Mexico; Unidad Academica Multidisciplinaria Reynosa-Aztlan, Universidad Autonoma de Tamaulipas, Reynosa, Mexico
| | - Rogelio Gómez-Escobedo
- Parasitology Department, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Gerardo Corral-Ruiz
- Microorganism Inmunology Laboratory, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Karla Fabiola Chacón-Vargas
- Microorganism Inmunology Laboratory, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico; Faculty of Chemical Sciences, Universidad Autónoma de Chihuahua, Chihuahua, Mexico
| | - Vanessa Horta-Montaño
- Parasitology Department, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico; Microorganism Inmunology Laboratory, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Luvia Sanchez-Torres
- Microorganism Inmunology Laboratory, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Lenci K Vazquez-Jimenez
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa, Tamaulipas, Mexico
| | - Benjamín Nogueda-Torres
- Parasitology Department, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Gildardo Rivera
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa, Tamaulipas, Mexico.
| |
Collapse
|
13
|
Gomes MC, Padilha EKA, Diniz GRA, Gomes EC, da Silva Santos-Júnior PF, Zhan P, da Siva-Júnior EF. Multi-target Compounds against Trypanosomatid Parasites and Mycobacterium tuberculosis. Curr Drug Targets 2024; 25:602-619. [PMID: 38910467 DOI: 10.2174/0113894501306843240606114854] [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: 03/09/2024] [Revised: 05/02/2024] [Accepted: 05/10/2024] [Indexed: 06/25/2024]
Abstract
Multi-target drug treatment has become popular as a substitute for traditional monotherapy. Monotherapy can lead to resistance and side effects. Multi-target drug discovery is gaining importance as data on bioactivity becomes more abundant. The design of multi-target drugs is expected to be an important development in the pharmaceutical industry in the near future. This review presents multi-target compounds against trypanosomatid parasites (Trypanosoma cruzi, T. brucei, and Leishmania sp.) and tuberculosis (Mycobacterium tuberculosis), which mainly affect populations in socioeconomically unfavorable conditions. The article analyzes the studies, including their chemical structures, viral strains, and molecular docking studies, when available. The objective of this review is to establish a foundation for designing new multi-target inhibitors for these diseases.
Collapse
Affiliation(s)
- Midiane Correia Gomes
- Research Group in Biological and Molecular Chemistry, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Campus AC. Simões, CEP 57072-970, Maceió-AL, Brazil
| | - Emanuelly Karla Araújo Padilha
- Research Group in Biological and Molecular Chemistry, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Campus AC. Simões, CEP 57072-970, Maceió-AL, Brazil
| | - Gustavo Rafael Angelo Diniz
- Research Group in Biological and Molecular Chemistry, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Campus AC. Simões, CEP 57072-970, Maceió-AL, Brazil
| | - Edilma Correia Gomes
- Research Group in Biological and Molecular Chemistry, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Campus AC. Simões, CEP 57072-970, Maceió-AL, Brazil
| | - Paulo Fernando da Silva Santos-Júnior
- Research Group in Biological and Molecular Chemistry, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Campus AC. Simões, CEP 57072-970, Maceió-AL, Brazil
| | - Peng Zhan
- Department of Medicinal - Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P.R. China
| | - Edeildo Ferreira da Siva-Júnior
- Research Group in Biological and Molecular Chemistry, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Campus AC. Simões, CEP 57072-970, Maceió-AL, Brazil
| |
Collapse
|
14
|
García-Torres I, De la Mora-De la Mora I, López-Velázquez G, Cabrera N, Flores-López LA, Becker I, Herrera-López J, Hernández R, Pérez-Montfort R, Enríquez-Flores S. Repurposing of rabeprazole as an anti- Trypanosoma cruzi drug that targets cellular triosephosphate isomerase. J Enzyme Inhib Med Chem 2023; 38:2231169. [PMID: 37401012 PMCID: PMC10351538 DOI: 10.1080/14756366.2023.2231169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/23/2023] [Accepted: 06/25/2023] [Indexed: 07/05/2023] Open
Abstract
Trypanosoma cruzi is the causative agent of American trypanosomiasis, which mainly affects populations in Latin America. Benznidazole is used to control the disease, with severe effects in patients receiving this chemotherapy. Previous studies have demonstrated the inhibition of triosephosphate isomerase from T. cruzi, but cellular enzyme inhibition has yet to be established. This study demonstrates that rabeprazole inhibits both cell viability and triosephosphate isomerase activity in T. cruzi epimastigotes. Our results show that rabeprazole has an IC50 of 0.4 µM, which is 14.5 times more effective than benznidazole. Additionally, we observed increased levels of methyl-glyoxal and advanced glycation end products after the inhibition of cellular triosephosphate isomerase by rabeprazole. Finally, we demonstrate that the inactivation mechanisms of rabeprazole on triosephosphate isomerase of T. cruzi can be achieved through the derivatization of three of its four cysteine residues. These results indicate that rabeprazole is a promising candidate against American trypanosomiasis.
Collapse
Affiliation(s)
- Itzhel García-Torres
- Laboratorio de Biomoléculas y Salud Infantil, Instituto Nacional de Pediatría, CDMX, México
| | | | | | - Nallely Cabrera
- Departamento de Bioquímica y Biología Estructural, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, CDMX, México
| | - Luis Antonio Flores-López
- CONAHCYT Instituto Nacional de Pediatría, Laboratorio de Biomoléculas y Salud Infantil, CDMX, México
| | - Ingeborg Becker
- Centro de Medicina Tropical, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, México
| | - Juliana Herrera-López
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, CDMX, México
| | - Roberto Hernández
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, CDMX, México
| | - Ruy Pérez-Montfort
- Departamento de Bioquímica y Biología Estructural, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, CDMX, México
| | - Sergio Enríquez-Flores
- Laboratorio de Biomoléculas y Salud Infantil, Instituto Nacional de Pediatría, CDMX, México
| |
Collapse
|
15
|
de Almeida GC, de Oliveira GB, da Silva Monte Z, Costa ÉCS, da Silva Falcão EP, Scotti L, Scotti MT, Oliveira Silva R, Pereira VRA, da Silva ED, Junior PAS, de Andrade Cavalcante MK, de Melo SJ. Structure-based design, optimization of lead, synthesis, and biological evaluation of compounds active against Trypanosoma cruzi. Chem Biol Drug Des 2023; 102:843-856. [PMID: 37455325 DOI: 10.1111/cbdd.14294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 04/18/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
Chagas' disease affects approximately eight million people throughout the world, especially the poorest individuals. The protozoan that causes this disease-Trypanosoma cruzi-has the enzyme cruzipain, which is the main therapeutic target. As no available medications have satisfactory effectiveness and safety, it is of fundamental importance to design and synthesize novel analogues that are more active and selective. In the present study, molecular docking and the in silico prediction of ADMET properties were used as strategies to optimize the trypanocidal activity of the pyrimidine compound ZN3F based on interactions with the target site in cruzipain. From the computational results, eight 4-amino-5-carbonitrile-pyrimidine analogues were proposed, synthesized (5a-f and 7g-h) and, tested in vitro on the trypomastigote form of the Tulahuen strain of T. cruzi. The in silico study showed that the designed analogues bond favorably to important amino acid residues of the active site in cruzipain. An in vitro evaluation of cytotoxicity was performed on L929 mammal cell lines. All derivatives inhibited the Tulahuen strain of T. cruzi and also exhibited lower toxicity to L929 cells. The 5e product, in particular, proved to be a potent, selective (IC50 = 2.79 ± 0.00 μM, selectivity index = 31.3) inhibitor of T. cruzi. The present results indicated the effectiveness of drugs based on the structure of the receptor, revealing the potential trypanocidal of pyrimidines. This study also provides information on molecular aspects for the inhibition of cruzipain.
Collapse
Affiliation(s)
- Gleybson Correia de Almeida
- Postgraduate Program in Pharmaceutical Sciences, Department of Pharmaceutical Sciences, Federal University of Pernambuco/UFPE, Recife, Brazil
| | - Gerliny Bezerra de Oliveira
- Postgraduate Program in Pharmaceutical Sciences, Department of Pharmaceutical Sciences, Federal University of Pernambuco/UFPE, Recife, Brazil
| | - Zenaide da Silva Monte
- Postgraduate Program in Pharmaceutical Sciences, Department of Pharmaceutical Sciences, Federal University of Pernambuco/UFPE, Recife, Brazil
| | - Érick Caique Santos Costa
- Postgraduate Program in Biological Sciences, Department of Biosciences, Federal University of Pernambuco/UFPE, Recife, Brazil
| | | | - Luciana Scotti
- Laboratory of Cheminformatics, Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa, Brazil
| | - Marcus Tullius Scotti
- Laboratory of Cheminformatics, Program of Natural and Synthetic Bioactive Products (PgPNSB), Health Sciences Center, Federal University of Paraíba, João Pessoa, Brazil
| | - Ricardo Oliveira Silva
- Department of Fundamental Chemistry, Center for Exact and Natural Sciences, Federal University of Pernambuco, Av. Journalist Anibal Fernandes, Recife, Brazil
| | - Valéria Rêgo Alves Pereira
- Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Federal University of Pernambuco - Campus da Av. Prof. Moraes Rego, Recife, Brazil
| | - Elis Dionisio da Silva
- Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Federal University of Pernambuco - Campus da Av. Prof. Moraes Rego, Recife, Brazil
| | - Policarpo Ademar Sales Junior
- Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Federal University of Pernambuco - Campus da Av. Prof. Moraes Rego, Recife, Brazil
| | - Marton Kaique de Andrade Cavalcante
- Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Federal University of Pernambuco - Campus da Av. Prof. Moraes Rego, Recife, Brazil
| | - Sebastião José de Melo
- Postgraduate Program in Pharmaceutical Sciences, Department of Pharmaceutical Sciences, Federal University of Pernambuco/UFPE, Recife, Brazil
- Postgraduate Program in Biological Sciences, Department of Biosciences, Federal University of Pernambuco/UFPE, Recife, Brazil
| |
Collapse
|
16
|
Laureano de Souza M, Lapierre TJWJD, Vitor de Lima Marques G, Ferraz WR, Penteado AB, Henrique Goulart Trossini G, Murta SMF, de Oliveira RB, de Oliveira Rezende C, Ferreira RS. Molecular targets for Chagas disease: validation, challenges and lead compounds for widely exploited targets. Expert Opin Ther Targets 2023; 27:911-925. [PMID: 37772733 DOI: 10.1080/14728222.2023.2264512] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/24/2023] [Indexed: 09/30/2023]
Abstract
INTRODUCTION Chagas disease (CD) imposes social and economic burdens, yet the available treatments have limited efficacy in the disease's chronic phase and cause serious adverse effects. To address this challenge, target-based approaches are a possible strategy to develop new, safe, and active treatments for both phases of the disease. AREAS COVERED This review delves into target-based approaches applied to CD drug discovery, emphasizing the studies from the last five years. We highlight the proteins cruzain (CZ), trypanothione reductase (TR), sterol 14 α-demethylase (CPY51), iron superoxide dismutase (Fe-SOD), proteasome, cytochrome b (Cytb), and cleavage and polyadenylation specificity factor 3 (CPSF3), chosen based on their biological and chemical validation as drug targets. For each, we discuss its biological relevance and validation as a target, currently related challenges, and the status of the most promising inhibitors. EXPERT OPINION Target-based approaches toward developing potential CD therapeutics have yielded promising leads in recent years. We expect a significant advance in this field in the next decade, fueled by the new options for Trypanosoma cruzi genetic manipulation that arose in the past decade, combined with recent advances in computational chemistry and chemical biology.
Collapse
Affiliation(s)
- Mariana Laureano de Souza
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | - Gabriel Vitor de Lima Marques
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Witor Ribeiro Ferraz
- Departamento de Farmacia, Faculdade de Ciencias Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - André Berndt Penteado
- Departamento de Farmacia, Faculdade de Ciencias Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | | | | | - Renata Barbosa de Oliveira
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | - Rafaela Salgado Ferreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
17
|
Cadamuro RD, Bastos IMADS, de Freitas ACO, Rosa MDS, Costa GDO, da Silva IT, Robl D, Stoco PH, Sandjo LP, Treichel H, Steindel M, Fongaro G. Bioactivity Screening and Chemical Characterization of Biocompound from Endophytic Neofusicoccum parvum and Buergenerula spartinae Isolated from Mangrove Ecosystem. Microorganisms 2023; 11:1599. [PMID: 37375101 DOI: 10.3390/microorganisms11061599] [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: 05/09/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
The discovery of biomolecules has been the subject of extensive research for several years due to their potential to combat harmful pathogens that can lead to environmental contamination and infections in both humans and animals. This study aimed to identify the chemical profile of endophytic fungi, namely Neofusicoccum parvum and Buergenerula spartinae, which were isolated from Avecinnia schaueriana and Laguncularia racemosa. We identified several HPLC-MS compounds, including Ethylidene-3,39-biplumbagin, Pestauvicolactone A, Phenylalanine, 2-Isopropylmalic acid, Fusaproliferin, Sespendole, Ansellone, Calanone derivative, Terpestacin, and others. Solid-state fermentation was conducted for 14-21 days, and methanol and dichloromethane extraction were performed to obtain a crude extract. The results of our cytotoxicity assay revealed a CC50 value > 500 μg/mL, while the virucide, Trypanosoma, leishmania, and yeast assay demonstrated no inhibition. Nevertheless, the bacteriostatic assay showed a 98% reduction in Listeria monocytogenes and Escherichia coli. Our findings suggest that these endophytic fungi species with distinct chemical profiles represent a promising niche for further exploring new biomolecules.
Collapse
Affiliation(s)
- Rafael Dorighello Cadamuro
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | | | - Ana Claudia Oliveira de Freitas
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | - Marilene da Silva Rosa
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | | | - Izabella Thaís da Silva
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
- Department of Pharmaceutical Sciences, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | - Diogo Robl
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | - Patricia Hermes Stoco
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | - Louis Pergaud Sandjo
- Department of Chemistry, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | - Helen Treichel
- Laboratory of Microbiology and Bioprocess, Federal University of Fronteira Sul, Erechim 99700970, RS, Brazil
| | - Mário Steindel
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | - Gislaine Fongaro
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| |
Collapse
|
18
|
Cox Holanda de Barros Dias M, Souza Barbalho M, Bezerra de Oliveira Filho G, Veríssimo de Oliveira Cardoso M, Lima Leite AC, da Silva Santos AC, Cristovão Silva AC, Accioly Brelaz de Castro MC, Maria Nascimento Moura D, Gomes Rebello Ferreira LF, Zaldini Hernandes M, de Freitas E Silva R, Rêgo Alves Pereira V. 1,3-Thiazole derivatives as privileged structures for anti-Trypanosoma cruzi activity: Rational design, synthesis, in silico and in vitro studies. Eur J Med Chem 2023; 257:115508. [PMID: 37267753 DOI: 10.1016/j.ejmech.2023.115508] [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: 02/15/2023] [Revised: 05/12/2023] [Accepted: 05/18/2023] [Indexed: 06/04/2023]
Abstract
Chagas disease is a deadly and centenary neglected disease that is recently surging as a potential global threat. Approximately 30% of infected individuals develop chronic Chagas cardiomyopathy and current treatment with the reference benznidazole (BZN) is ineffective for this stage. We presently report the structural planning, synthesis, characterization, molecular docking prediction, cytotoxicity, in vitro bioactivity and mechanistic studies on the anti-T. cruzi activity of a series of 16 novel 1,3-thiazoles (2-17) derived from thiosemicarbazones (1a, 1b) in a two-step and reproducible Hantzsch-based synthesis approach. The anti-T. cruzi activity was evaluated in vitro against the epimastigote, amastigote and trypomastigote forms of the parasite. In the bioactivity assays, all thiazoles were more potent than BZN against epimastigotes. We found that the compounds presented an overall increased anti-tripomastigote selectivity (Cpd 8 was 24-fold more selective) than BZN, and they mostly presented anti-amastigote activity at very low doses (from 3.65 μM, cpd 15). Mechanistic studies on cell death suggested that the series of 1,3-thiazole compounds herein reported cause parasite cell death through apoptosis, but without compromising the mitochondrial membrane potential. In silico prediction of physicochemical properties and pharmacokinetic parameters showed promising drug-like results, being all the reported compounds in compliance with Lipinski and Veber rules. In summary, our work contributes towards a more rational design of potent and selective antitripanosomal drugs, using affordable methodology to yield industrially viable drug candidates.
Collapse
Affiliation(s)
- Mabilly Cox Holanda de Barros Dias
- Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal de Pernambuco, 50740-520, Recife, Pernambuco, Brazil.
| | - Mayara Souza Barbalho
- Centro de Pesquisas Aggeu Magalhães, Fundação Oswaldo Cruz, 50670- 420, Recife, Pernambuco, Brazil
| | - Gevanio Bezerra de Oliveira Filho
- Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal de Pernambuco, 50740-520, Recife, Pernambuco, Brazil
| | | | - Ana Cristina Lima Leite
- Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal de Pernambuco, 50740-520, Recife, Pernambuco, Brazil
| | | | | | | | | | - Luiz Felipe Gomes Rebello Ferreira
- Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal de Pernambuco, 50740-520, Recife, Pernambuco, Brazil
| | - Marcelo Zaldini Hernandes
- Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal de Pernambuco, 50740-520, Recife, Pernambuco, Brazil
| | | | | |
Collapse
|
19
|
Pascual Alonso I, Almeida García F, Valdés Tresanco ME, Arrebola Sánchez Y, Ojeda Del Sol D, Sánchez Ramírez B, Florent I, Schmitt M, Avilés FX. Marine Invertebrates: A Promissory Still Unexplored Source of Inhibitors of Biomedically Relevant Metallo Aminopeptidases Belonging to the M1 and M17 Families. Mar Drugs 2023; 21:md21050279. [PMID: 37233473 DOI: 10.3390/md21050279] [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: 03/27/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/27/2023] Open
Abstract
Proteolytic enzymes, also known as peptidases, are critical in all living organisms. Peptidases control the cleavage, activation, turnover, and synthesis of proteins and regulate many biochemical and physiological processes. They are also involved in several pathophysiological processes. Among peptidases, aminopeptidases catalyze the cleavage of the N-terminal amino acids of proteins or peptide substrates. They are distributed in many phyla and play critical roles in physiology and pathophysiology. Many of them are metallopeptidases belonging to the M1 and M17 families, among others. Some, such as M1 aminopeptidases N and A, thyrotropin-releasing hormone-degrading ectoenzyme, and M17 leucyl aminopeptidase, are targets for the development of therapeutic agents for human diseases, including cancer, hypertension, central nervous system disorders, inflammation, immune system disorders, skin pathologies, and infectious diseases, such as malaria. The relevance of aminopeptidases has driven the search and identification of potent and selective inhibitors as major tools to control proteolysis with an impact in biochemistry, biotechnology, and biomedicine. The present contribution focuses on marine invertebrate biodiversity as an important and promising source of inhibitors of metalloaminopeptidases from M1 and M17 families, with foreseen biomedical applications in human diseases. The results reviewed in the present contribution support and encourage further studies with inhibitors isolated from marine invertebrates in different biomedical models associated with the activity of these families of exopeptidases.
Collapse
Affiliation(s)
- Isel Pascual Alonso
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Fabiola Almeida García
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Mario Ernesto Valdés Tresanco
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada
| | | | - Daniel Ojeda Del Sol
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | | | - Isabelle Florent
- Unité Molécules de Communication et Adaptation des Microorganismes (MCAM, UMR7245), Muséum National d'Histoire Naturelle, CNRS, CP52, 57 Rue Cuvier, 75005 Paris, France
| | - Marjorie Schmitt
- Université de Haute-Alsace, Université de Strasbourg, CNRS, LIMA UMR 7042, 68000 Mulhouse, France
| | - Francesc Xavier Avilés
- Institute for Biotechnology and Biomedicine and Department of Biochemistry, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| |
Collapse
|
20
|
Tayama Y, Mizukami S, Toume K, Komatsu K, Yanagi T, Nara T, Tieu P, Huy NT, Hamano S, Hirayama K. Anti-Trypanosoma cruzi activity of Coptis rhizome extract and its constituents. Trop Med Health 2023; 51:12. [PMID: 36859380 PMCID: PMC9976467 DOI: 10.1186/s41182-023-00502-2] [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: 12/30/2022] [Accepted: 02/09/2023] [Indexed: 03/03/2023] Open
Abstract
BACKGROUND Current therapeutic agents, including nifurtimox and benznidazole, are not sufficiently effective in the chronic phase of Trypanosoma cruzi infection and are accompanied by various side effects. In this study, 120 kinds of extracts from medicinal herbs used for Kampo formulations and 94 kinds of compounds isolated from medicinal herbs for Kampo formulations were screened for anti-T. cruzi activity in vitro and in vivo. METHODS As an experimental method, a recombinant protozoan cloned strain expressing luciferase, namely Luc2-Tulahuen, was used in the experiments. The in vitro anti-T. cruzi activity on epimastigote, trypomastigote, and amastigote forms was assessed by measuring luminescence intensity after treatment with the Kampo extracts or compounds. In addition, the cytotoxicity of compounds was tested using mouse and human feeder cell lines. The in vivo anti-T. cruzi activity was measured by a murine acute infection model using intraperitoneal injection of trypomastigotes followed by live bioluminescence imaging. RESULTS As a result, three protoberberine-type alkaloids, namely coptisine chloride, dehydrocorydaline nitrate, and palmatine chloride, showed strong anti-T. cruzi activities with low cytotoxicity. The IC50 values of these compounds differed depending on the side chain, and the most effective compound, coptisine chloride, showed a significant effect in the acute infection model. CONCLUSIONS For these reasons, coptisine chloride is a hit compound that can be a potential candidate for anti-Chagas disease drugs. In addition, it was expected that there would be room for further improvement by modifying the side chains of the basic skeleton.
Collapse
Affiliation(s)
- Yuki Tayama
- grid.174567.60000 0000 8902 2273Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523 Japan ,grid.174567.60000 0000 8902 2273Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523 Japan
| | - Shusaku Mizukami
- grid.174567.60000 0000 8902 2273Department of Immune Regulation, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan ,grid.174567.60000 0000 8902 2273School of Tropical Medicines and Global Health, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523 Japan ,grid.174567.60000 0000 8902 2273The Joint Usage/Research Center On Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, 852-8523 Japan
| | - Kazufumi Toume
- grid.267346.20000 0001 2171 836XSection of Pharmacognosy, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Katsuko Komatsu
- grid.267346.20000 0001 2171 836XSection of Pharmacognosy, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Tetsuo Yanagi
- grid.174567.60000 0000 8902 2273NEKKEN Bio-Resource Center (NBRC), Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan ,grid.174567.60000 0000 8902 2273The Joint Usage/Research Center On Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, 852-8523 Japan
| | - Takeshi Nara
- grid.411789.20000 0004 0371 1051Faculty of Pharmacy, Iryo Sosei University, Iwaki, Fukushima Japan
| | - Paul Tieu
- grid.25073.330000 0004 1936 8227Faculty of Health Sciences, McMaster University, Hamilton, ON Canada ,Online Research Club, Nagasaki, Japan
| | - Nguyen Tien Huy
- grid.174567.60000 0000 8902 2273Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523 Japan ,grid.174567.60000 0000 8902 2273School of Tropical Medicines and Global Health, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523 Japan ,Online Research Club, Nagasaki, Japan
| | - Shinjiro Hamano
- grid.174567.60000 0000 8902 2273Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523 Japan ,grid.174567.60000 0000 8902 2273Department of Parasitology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan ,grid.174567.60000 0000 8902 2273The Joint Usage/Research Center On Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, 852-8523 Japan
| | - Kenji Hirayama
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan. .,Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan. .,School of Tropical Medicines and Global Health, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan. .,The Joint Usage/Research Center On Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, 852-8523, Japan.
| |
Collapse
|
21
|
Barnadas-Carceller B, Martinez-Peinado N, Gómez LC, Ros-Lucas A, Gabaldón-Figueira JC, Diaz-Mochon JJ, Gascon J, Molina IJ, Pineda de las Infantas y Villatoro MJ, Alonso-Padilla J. Identification of compounds with activity against Trypanosoma cruzi within a collection of synthetic nucleoside analogs. Front Cell Infect Microbiol 2023; 12:1067461. [PMID: 36710960 PMCID: PMC9880260 DOI: 10.3389/fcimb.2022.1067461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/11/2022] [Indexed: 01/14/2023] Open
Abstract
Introduction Chagas disease is caused by the protozoan parasite Trypanosoma cruzi, and it is the most important neglected tropical disease in the Americas. Two drugs are available to treat the infection, but their efficacy in the chronic stage of the disease, when most cases are diagnosed, is reduced. Their tolerability is also hindered by common adverse effects, making the development of safer and efficacious alternatives a pressing need. T. cruzi is unable to synthesize purines de novo, relying on a purine salvage pathway to acquire these from its host, making it an attractive target for the development of new drugs. Methods We evaluated the anti-parasitic activity of 23 purine analogs with different substitutions in the complementary chains of their purine rings. We sequentially screened the compounds' capacity to inhibit parasite growth, their toxicity in Vero and HepG2 cells, and their specific capacity to inhibit the development of amastigotes. We then used in-silico docking to identify their likely targets. Results Eight compounds showed specific anti-parasitic activity, with IC50 values ranging from 2.42 to 8.16 μM. Adenine phosphoribosyl transferase, and hypoxanthine-guanine phosphoribosyl transferase, are their most likely targets. Discussion Our results illustrate the potential role of the purine salvage pathway as a target route for the development of alternative treatments against T. cruzi infection, highlithing the apparent importance of specific substitutions, like the presence of benzene groups in the C8 position of the purine ring, consistently associated with a high and specific anti-parasitic activity.
Collapse
Affiliation(s)
- Berta Barnadas-Carceller
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic - University of Barcelona, Barcelona, Spain
| | - Nieves Martinez-Peinado
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic - University of Barcelona, Barcelona, Spain,Secció de Parasitologia, Departament de Biologia, Sanitat i Medi Ambient, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, Barcelona, Spain
| | - Laura Córdoba Gómez
- Department of Medicinal & Organic Chemistry and Excellence Research Unit of “Chemistry Applied to Biomedicine and the Environment”, Faculty of Pharmacy, University of Granada, Granada, Spain
| | - Albert Ros-Lucas
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic - University of Barcelona, Barcelona, Spain,CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain
| | | | - Juan J. Diaz-Mochon
- Department of Medicinal & Organic Chemistry and Excellence Research Unit of “Chemistry Applied to Biomedicine and the Environment”, Faculty of Pharmacy, University of Granada, Granada, Spain,GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain,Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada-University of Granada, Granada, Spain
| | - Joaquim Gascon
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic - University of Barcelona, Barcelona, Spain,CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain
| | - Ignacio J. Molina
- Institute of Biopathology and Regenerative Medicine, Centre for Biomedical Research, University of Granada, Granada, Spain
| | - María José Pineda de las Infantas y Villatoro
- Department of Medicinal & Organic Chemistry and Excellence Research Unit of “Chemistry Applied to Biomedicine and the Environment”, Faculty of Pharmacy, University of Granada, Granada, Spain,*Correspondence: Julio Alonso-Padilla, ; María José Pineda de las Infantas y Villatoro,
| | - Julio Alonso-Padilla
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic - University of Barcelona, Barcelona, Spain,CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain,*Correspondence: Julio Alonso-Padilla, ; María José Pineda de las Infantas y Villatoro,
| |
Collapse
|
22
|
Reséndiz-Mora A, Barrera-Aveleida G, Sotelo-Rodríguez A, Galarce-Sosa I, Nevárez-Lechuga I, Santiago-Hernández JC, Nogueda-Torres B, Meza-Toledo S, Gómez-Manzo S, Wong-Baeza I, Baeza I, Wong-Baeza C. Effect of B-NIPOx in Experimental Trypanosoma cruzi Infection in Mice. Int J Mol Sci 2022; 24:ijms24010333. [PMID: 36613783 PMCID: PMC9820238 DOI: 10.3390/ijms24010333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Chagas disease is caused by Trypanosoma cruzi and represents a major public health problem, which is endemic in Latin America and emerging in the rest of the world. The two drugs that are currently available for its treatment, Benznidazole and Nifurtimox, are partially effective in the chronic phase of the disease. In this study, we designed and synthesized the benzyl ester of N-isopropyl oxamic acid (B-NIPOx), which is a non-polar molecule that crosses cell membranes. B-NIPOx is cleaved inside the parasite by carboxylesterases, releasing benzyl alcohol (a molecule with antimicrobial activity), and NIPOx, which is an inhibitor of α-hydroxy acid dehydrogenase isozyme II (HADH-II), a key enzyme in T. cruzi metabolism. We evaluated B-NIPOx cytotoxicity, its toxicity in mice, and its inhibitory activity on purified HADH-II and on T. cruzi homogenates. We then evaluated the trypanocidal activity of B-NIPOx in vitro and in vivo and its effect in the intestine of T. cruzi-infected mice. We found that B-NIPOx had higher trypanocidal activity on epimastigotes and trypomastigotes than Benznidazole and Nifurtimox, that it was more effective to reduce blood parasitemia and amastigote nests in infected mice, and that, in contrast to the reference drugs, it prevented the development of Chagasic enteropathy.
Collapse
Affiliation(s)
- Albany Reséndiz-Mora
- Laboratorio de Biomembranas, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
- Laboratorio de Enzimología, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Giovanna Barrera-Aveleida
- Laboratorio de Biomembranas, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
- Laboratorio de Enzimología, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Anahi Sotelo-Rodríguez
- Laboratorio de Biomembranas, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
- Laboratorio de Enzimología, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Iván Galarce-Sosa
- Laboratorio de Biomembranas, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
- Laboratorio de Enzimología, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Irene Nevárez-Lechuga
- Laboratorio de Biomembranas, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
- Laboratorio de Enzimología, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Juan Carlos Santiago-Hernández
- Laboratorio de Enzimología, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Benjamín Nogueda-Torres
- Laboratorio de Helmintología, Departamento de Parasitología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Sergio Meza-Toledo
- Laboratorio de Quimioterapia Experimental, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Saúl Gómez-Manzo
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico
| | - Isabel Wong-Baeza
- Laboratorio de Inmunología Molecular II, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Isabel Baeza
- Laboratorio de Biomembranas, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
- Correspondence: (I.B.); (C.W.-B.); Tel.: +52-55-5729-6000 (ext. 62326) (I.B. & C.W.-B.)
| | - Carlos Wong-Baeza
- Laboratorio de Biomembranas, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
- Correspondence: (I.B.); (C.W.-B.); Tel.: +52-55-5729-6000 (ext. 62326) (I.B. & C.W.-B.)
| |
Collapse
|
23
|
Beatriz Vermelho A, Rodrigues GC, Nocentini A, Mansoldo FRP, Supuran CT. Discovery of novel drugs for Chagas disease: is carbonic anhydrase a target for antiprotozoal drugs? Expert Opin Drug Discov 2022; 17:1147-1158. [PMID: 36039500 DOI: 10.1080/17460441.2022.2117295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Carbonic anhydrase (CA) arose significant interest as a potential new target for Chagas disease since its discovery in Trypanosoma cruzi in 2013. Benznidazole and Nifurtimox have been used for Chagas disease treatment for 60 years despite all efforts done for obtaining more efficient treatments, acting in the acute and chronic phases of illness, with fewer side effects and resistance induction. AREAS COVERED We discuss the positive and negative aspects of T. cruzi CA (TcCA) studies as a target for developing new drugs. The current research discoveries and the classes of TcCA inhibitors are reviewed. The sulfonamides and their derivatives are the main inhibitor classes, but hydroxamates and the thiols, were investigated too. These compounds inhibited the growth of the evolutive forms of the parasite. A comparative analysis was done with CAs from other Trypanosomatids and protozoans. EXPERT OPINION The search for new targets and drugs is a significant challenge worldwide, and TcCA is a potential candidate for developing new drugs. Several studied inhibitors were active against Trypanosoma cruzi, but their penetration and toxicity problems emerged. New approaches are in progress to obtain inhibitors with desired properties, allowing further steps such as tests using an adequate animal model and subsequent developments for the preclinical testing.
Collapse
Affiliation(s)
- Alane Beatriz Vermelho
- BIOINOVAR - Biotechnology Laboratories: Biocatalysis, Bioproducts, and Bioenergy, Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Giseli Capaci Rodrigues
- UNIGRANRIO - Universidade do Grande Rio Programa de Pós-Graduação em Ensino das Ciências, Rio de Janeiro, Brazil
| | - Alessio Nocentini
- Department of Neuroscience, Psychology, Drug Research, and Child's Health, Section of Pharmaceutical and Nutraceutical Sciences University of Florence, Florence, Italy
| | - Felipe R P Mansoldo
- BIOINOVAR - Biotechnology Laboratories: Biocatalysis, Bioproducts, and Bioenergy, Institute of Microbiology Paulo de Goes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudiu T Supuran
- Department of Neuroscience, Psychology, Drug Research, and Child's Health, Section of Pharmaceutical and Nutraceutical Sciences University of Florence, Florence, Italy
| |
Collapse
|
24
|
Almeida-Silva J, Menezes DS, Fernandes JMP, Almeida MC, Vasco-Dos-Santos DR, Saraiva RM, Viçosa AL, Perez SAC, Andrade SG, Suarez-Fontes AM, Vannier-Santos MA. The repositioned drugs disulfiram/diethyldithiocarbamate combined to benznidazole: Searching for Chagas disease selective therapy, preventing toxicity and drug resistance. Front Cell Infect Microbiol 2022; 12:926699. [PMID: 35967878 PMCID: PMC9372510 DOI: 10.3389/fcimb.2022.926699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/27/2022] [Indexed: 12/20/2022] Open
Abstract
Chagas disease (CD) affects at least 6 million people in 21 South American countries besides several thousand in other nations all over the world. It is estimated that at least 14,000 people die every year of CD. Since vaccines are not available, chemotherapy remains of pivotal relevance. About 30% of the treated patients cannot complete the therapy because of severe adverse reactions. Thus, the search for novel drugs is required. Here we tested the benznidazole (BZ) combination with the repositioned drug disulfiram (DSF) and its derivative diethyldithiocarbamate (DETC) upon Trypanosoma cruzi in vitro and in vivo. DETC-BZ combination was synergistic diminishing epimastigote proliferation and enhancing selective indexes up to over 10-fold. DETC was effective upon amastigotes of the BZ- partially resistant Y and the BZ-resistant Colombiana strains. The combination reduced proliferation even using low concentrations (e.g., 2.5 µM). Scanning electron microscopy revealed membrane discontinuities and cell body volume reduction. Transmission electron microscopy revealed remarkable enlargement of endoplasmic reticulum cisternae besides, dilated mitochondria with decreased electron density and disorganized kinetoplast DNA. At advanced stages, the cytoplasm vacuolation apparently impaired compartmentation. The fluorescent probe H2-DCFDA indicates the increased production of reactive oxygen species associated with enhanced lipid peroxidation in parasites incubated with DETC. The biochemical measurement indicates the downmodulation of thiol expression. DETC inhibited superoxide dismutase activity on parasites was more pronounced than in infected mice. In order to approach the DETC effects on intracellular infection, peritoneal macrophages were infected with Colombiana trypomastigotes. DETC addition diminished parasite numbers and the DETC-BZ combination was effective, despite the low concentrations used. In the murine infection, the combination significantly enhanced animal survival, decreasing parasitemia over BZ. Histopathology revealed that low doses of BZ-treated animals presented myocardial amastigote, not observed in combination-treated animals. The picrosirius collagen staining showed reduced myocardial fibrosis. Aminotransferase de aspartate, Aminotransferase de alanine, Creatine kinase, and urea plasma levels demonstrated that the combination was non-toxic. As DSF and DETC can reduce the toxicity of other drugs and resistance phenotypes, such a combination may be safe and effective.
Collapse
Affiliation(s)
- Juliana Almeida-Silva
- Innovations in Therapies, Education and Bioproducts Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Diego Silva Menezes
- Parasite Biology Laboratory, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, BA, Brazil
| | - Juan Mateus Pereira Fernandes
- Innovations in Therapies, Education and Bioproducts Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Márcio Cerqueira Almeida
- Parasite Biology Laboratory, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, BA, Brazil
| | - Deyvison Rhuan Vasco-Dos-Santos
- Innovations in Therapies, Education and Bioproducts Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Roberto Magalhães Saraiva
- Laboratory of Clinical Research on Chagas Disease, Evandro Chagas Infectious Disease Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Alessandra Lifsitch Viçosa
- Experimental Pharmacotechnics Laboratory, Department of Galenic Innovation, Institute of Drug Technology - Farmanguinhos, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Sandra Aurora Chavez Perez
- Project Management Technical Assistance, Institute of Drug Technology - Farmanguinhos, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Sônia Gumes Andrade
- Experimental Chagas Disease Laboratory, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, BA, Brazil
| | - Ana Márcia Suarez-Fontes
- Innovations in Therapies, Education and Bioproducts Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Marcos André Vannier-Santos
- Innovations in Therapies, Education and Bioproducts Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
25
|
Ros-Lucas A, Martinez-Peinado N, Bastida J, Gascón J, Alonso-Padilla J. The Use of AlphaFold for In Silico Exploration of Drug Targets in the Parasite Trypanosoma cruzi. Front Cell Infect Microbiol 2022; 12:944748. [PMID: 35909956 PMCID: PMC9329570 DOI: 10.3389/fcimb.2022.944748] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 06/23/2022] [Indexed: 11/23/2022] Open
Abstract
Chagas disease is a devastating neglected disease caused by the parasite Trypanosoma cruzi, which affects millions of people worldwide. The two anti-parasitic drugs available, nifurtimox and benznidazole, have a good efficacy against the acute stage of the infection. But this is short, usually asymptomatic and often goes undiagnosed. Access to treatment is mostly achieved during the chronic stage, when the cardiac and/or digestive life-threatening symptoms manifest. Then, the efficacy of both drugs is diminished, and their long administration regimens involve frequently associated adverse effects that compromise treatment compliance. Therefore, the discovery of safer and more effective drugs is an urgent need. Despite its advantages over lately used phenotypic screening, target-based identification of new anti-parasitic molecules has been hampered by incomplete annotation and lack of structures of the parasite protein space. Presently, the AlphaFold Protein Structure Database is home to 19,036 protein models from T. cruzi, which could hold the key to not only describe new therapeutic approaches, but also shed light on molecular mechanisms of action for known compounds. In this proof-of-concept study, we screened the AlphaFold T. cruzi set of predicted protein models to find prospective targets for a pre-selected list of compounds with known anti-trypanosomal activity using docking-based inverse virtual screening. The best receptors (targets) for the most promising ligands were analyzed in detail to address molecular interactions and potential drugs’ mode of action. The results provide insight into the mechanisms of action of the compounds and their targets, and pave the way for new strategies to finding novel compounds or optimize already existing ones.
Collapse
Affiliation(s)
- Albert Ros-Lucas
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic - University of Barcelona, Barcelona, Spain
- *Correspondence: Albert Ros-Lucas, ; Nieves Martinez-Peinado, ; Julio Alonso-Padilla,
| | - Nieves Martinez-Peinado
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic - University of Barcelona, Barcelona, Spain
- *Correspondence: Albert Ros-Lucas, ; Nieves Martinez-Peinado, ; Julio Alonso-Padilla,
| | - Jaume Bastida
- Departament de Biologia, Sanitat i Medi Ambient, Facultat de Farmàcia i Ciències de l´Alimentació, Universitat de Barcelona, Barcelona, Spain
| | - Joaquim Gascón
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic - University of Barcelona, Barcelona, Spain
- CIBERINFEC, ISCIII—CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Julio Alonso-Padilla
- Barcelona Institute for Global Health (ISGlobal), Hospital Clinic - University of Barcelona, Barcelona, Spain
- CIBERINFEC, ISCIII—CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- *Correspondence: Albert Ros-Lucas, ; Nieves Martinez-Peinado, ; Julio Alonso-Padilla,
| |
Collapse
|
26
|
Alramadhani D, Aljahdali AS, Abdulmalik O, Pierce BD, Safo MK. Metabolic Reprogramming in Sickle Cell Diseases: Pathophysiology and Drug Discovery Opportunities. Int J Mol Sci 2022; 23:7448. [PMID: 35806451 PMCID: PMC9266828 DOI: 10.3390/ijms23137448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/27/2022] [Accepted: 06/30/2022] [Indexed: 01/19/2023] Open
Abstract
Sickle cell disease (SCD) is a genetic disorder that affects millions of individuals worldwide. Chronic anemia, hemolysis, and vasculopathy are associated with SCD, and their role has been well characterized. These symptoms stem from hemoglobin (Hb) polymerization, which is the primary event in the molecular pathogenesis of SCD and contributes to erythrocyte or red blood cell (RBC) sickling, stiffness, and vaso-occlusion. The disease is caused by a mutation at the sixth position of the β-globin gene, coding for sickle Hb (HbS) instead of normal adult Hb (HbA), which under hypoxic conditions polymerizes into rigid fibers to distort the shapes of the RBCs. Only a few therapies are available, with the universal effectiveness of recently approved therapies still being monitored. In this review, we first focus on how sickle RBCs have altered metabolism and then highlight how this understanding reveals potential targets involved in the pathogenesis of the disease, which can be leveraged to create novel therapeutics for SCD.
Collapse
Affiliation(s)
- Dina Alramadhani
- Department of Medicinal Chemistry and the Institute for Structural Biology, Drug Discovery and Development, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Anfal S. Aljahdali
- Department of Pharmaceutical Chemistry, King Abdulaziz University, Alsulaymanyah, Jeddah 21589, Saudi Arabia;
| | - Osheiza Abdulmalik
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| | - B. Daniel Pierce
- Department of Biology, University of Richmond, Richmond, VA 23173, USA;
| | - Martin K. Safo
- Department of Medicinal Chemistry and the Institute for Structural Biology, Drug Discovery and Development, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA;
| |
Collapse
|
27
|
Espinosa-Bustos C, Ortiz Pérez M, Gonzalez-Gonzalez A, Zarate AM, Rivera G, Belmont-Díaz JA, Saavedra E, Cuellar MA, Vázquez K, Salas CO. New Amino Naphthoquinone Derivatives as Anti-Trypanosoma cruzi Agents Targeting Trypanothione Reductase. Pharmaceutics 2022; 14:pharmaceutics14061121. [PMID: 35745694 PMCID: PMC9228152 DOI: 10.3390/pharmaceutics14061121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/18/2022] [Accepted: 05/23/2022] [Indexed: 12/25/2022] Open
Abstract
To develop novel chemotherapeutic alternatives for the treatment of Chagas disease, in this study, a set of new amino naphthoquinone derivatives were synthesised and evaluated in vitro on the epimastigote and trypomastigote forms of Trypanosoma cruzi strains (NINOA and INC-5) and on J774 murine macrophages. The design of the new naphthoquinone derivatives considered the incorporation of nitrogenous fragments with different substitution patterns present in compounds with activity on T. cruzi, and, thus, 19 compounds were synthesised in a simple manner. Compounds 2e and 7j showed the lowest IC50 values (0.43 µM against both strains for 2e and 0.19 µM and 0.92 µM for 7j). Likewise, 7j was more potent than the reference drug, benznidazole, and was more selective on epimastigotes. To postulate a possible mechanism of action, molecular docking studies were performed on T. cruzi trypanothione reductase (TcTR), specifically at a site in the dimer interface, which is a binding site for this type of naphthoquinone. Interestingly, 7j was one of the compounds that showed the best interaction profile on the enzyme; therefore, 7j was evaluated on TR, which behaved as a non-competitive inhibitor. Finally, 7j was predicted to have a good pharmacokinetic profile for oral administration. Thus, the naphthoquinone nucleus should be considered in the search for new trypanocidal agents based on our hit 7j.
Collapse
Affiliation(s)
- Christian Espinosa-Bustos
- Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Santiago 7820436, Chile;
| | - Mariana Ortiz Pérez
- Departamento de Parasitología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Nuevo León, Francisco Villa 20, General Escobedo 66054, Mexico;
| | - Alonzo Gonzalez-Gonzalez
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Boulevard del Maestro s/n, Reynosa 88710, Mexico; (A.G.-G.); (G.R.)
| | - Ana María Zarate
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago 7820436, Chile;
| | - Gildardo Rivera
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Boulevard del Maestro s/n, Reynosa 88710, Mexico; (A.G.-G.); (G.R.)
| | - Javier A. Belmont-Díaz
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico; (J.A.B.-D.); (E.S.)
| | - Emma Saavedra
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico; (J.A.B.-D.); (E.S.)
| | - Mauricio A. Cuellar
- Centro de Investigación Farmacopea Chilena, Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Av. Gran Bretaña 1093, Valparaíso 2340000, Chile;
| | - Karina Vázquez
- Departamento de Parasitología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Nuevo León, Francisco Villa 20, General Escobedo 66054, Mexico;
- Correspondence: (K.V.); (C.O.S.)
| | - Cristian O. Salas
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago 7820436, Chile;
- Correspondence: (K.V.); (C.O.S.)
| |
Collapse
|
28
|
Brogi S, Ibba R, Rossi S, Butini S, Calderone V, Gemma S, Campiani G. Covalent Reversible Inhibitors of Cysteine Proteases Containing the Nitrile Warhead: Recent Advancement in the Field of Viral and Parasitic Diseases. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27082561. [PMID: 35458759 PMCID: PMC9029279 DOI: 10.3390/molecules27082561] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/08/2022] [Accepted: 04/13/2022] [Indexed: 12/29/2022]
Abstract
In the field of drug discovery, the nitrile group is well represented among drugs and biologically active compounds. It can form both non-covalent and covalent interactions with diverse biological targets, and it is amenable as an electrophilic warhead for covalent inhibition. The main advantage of the nitrile group as a warhead is mainly due to its milder electrophilic character relative to other more reactive groups (e.g., -CHO), reducing the possibility of unwanted reactions that would hinder the development of safe drugs, coupled to the ease of installation through different synthetic approaches. The covalent inhibition is a well-assessed design approach for serine, threonine, and cysteine protease inhibitors. The mechanism of hydrolysis of these enzymes involves the formation of a covalent acyl intermediate, and this mechanism can be exploited by introducing electrophilic warheads in order to mimic this covalent intermediate. Due to the relevant role played by the cysteine protease in the survival and replication of infective agents, spanning from viruses to protozoan parasites, we will review the most relevant and recent examples of protease inhibitors presenting a nitrile group that have been introduced to form or to facilitate the formation of a covalent bond with the catalytic cysteine active site residue.
Collapse
Affiliation(s)
- Simone Brogi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (S.B.); (V.C.)
| | - Roberta Ibba
- Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (R.I.); (S.R.); (S.B.); (G.C.)
| | - Sara Rossi
- Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (R.I.); (S.R.); (S.B.); (G.C.)
| | - Stefania Butini
- Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (R.I.); (S.R.); (S.B.); (G.C.)
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (S.B.); (V.C.)
| | - Sandra Gemma
- Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (R.I.); (S.R.); (S.B.); (G.C.)
- Correspondence:
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, DoE Department of Excellence 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (R.I.); (S.R.); (S.B.); (G.C.)
| |
Collapse
|