1
|
Rubio-Hernández M, Alcolea V, Barbosa da Silva E, Giardini MA, M Fernandes TH, Martínez-Sáez N, O'Donoghue AJ, Siqueira-Neto JL, Pérez-Silanes S. Synthesis and Biological Evaluation of New Chalcogen Semicarbazone ( S, Se) and Their Azole Derivatives against Chagas Disease. J Med Chem 2024. [PMID: 39485736 DOI: 10.1021/acs.jmedchem.4c01535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Chagas disease is caused by the eukaryote parasite Trypanosoma cruzi. Current treatment exhibits limited efficacy and selenium-based compounds emerged as promising candidates for new therapies which is surpassing its bioisoster, sulfur. We designed new thiosemicarbazones, thiazoles, selenosemicarbazones and selenazoles, using isosteric substitution. We synthesized 57 new chalcogen compounds which were evaluated against T. cruzi, C2C12 cells and cruzain, the main target of this parasite. Additionally, human cathepsin L, was tested for selectivity. Three compounds were selected, based on their activity against the intracellular amastigotes (EC50 < 1 μM, SI > 10) and cruzain (IC50 < 100 nM, SI > 5.55) which compared favorably with the approved drug, Benznidazole, and the well-established cruzain inhibitor K777. Seleno-compounds demonstrated enhanced activity and selenazoles showed a decrease in selenium-associated toxicity. Compound 4-methyl-2-(2-(1-(3-nitrophenyl)ethylidene)hydrazineyl)-1,3-selenazole (Se2h) emerged as a promising candidate, and its binding to cruzain was investigated. Pharmacokinetic assessment was conducted, showing a favorable profile for subsequent in vivo assays.
Collapse
Affiliation(s)
- Mercedes Rubio-Hernández
- ISTUN Institute of Tropical Health, Department of Pharmaceutical Sciences, Universidad de Navarra, 31008 Pamplona, Spain
| | - Verónica Alcolea
- ISTUN Institute of Tropical Health, Department of Pharmaceutical Sciences, Universidad de Navarra, 31008 Pamplona, Spain
| | - Elany Barbosa da Silva
- Skaggs School of Pharmacy and Pharmaceutical Sciences and Center for Discovery and Innovation in Parasitic Diseases, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Miriam A Giardini
- Skaggs School of Pharmacy and Pharmaceutical Sciences and Center for Discovery and Innovation in Parasitic Diseases, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Thaís H M Fernandes
- Skaggs School of Pharmacy and Pharmaceutical Sciences and Center for Discovery and Innovation in Parasitic Diseases, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Nuria Martínez-Sáez
- Department of Pharmaceutical Sciences, Universidad de Navarra, 31008 Pamplona, Spain
| | - Anthony J O'Donoghue
- Skaggs School of Pharmacy and Pharmaceutical Sciences and Center for Discovery and Innovation in Parasitic Diseases, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Jair L Siqueira-Neto
- Skaggs School of Pharmacy and Pharmaceutical Sciences and Center for Discovery and Innovation in Parasitic Diseases, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Silvia Pérez-Silanes
- ISTUN Institute of Tropical Health, Department of Pharmaceutical Sciences, Universidad de Navarra, 31008 Pamplona, Spain
| |
Collapse
|
2
|
da Silva AD, Fracasso M, Bottari NB, Palma TV, Engelmann AM, Castro MFV, Assmann CE, Mostardeiro V, Reichert KP, Nauderer J, da Veiga ML, da Rocha MIUM, Milleti LC, das Neves GB, Gundel S, Ourique AF, Monteiro SG, Morsch VM, Chitolina MR, Da Silva AS. Effects of Free and Nanoencapsulated Benznidazole in Acute Trypanosoma cruzi Infection: Role of Cholinergic Pathway and Redox Status. Pharmaceuticals (Basel) 2024; 17:1397. [PMID: 39459036 PMCID: PMC11510717 DOI: 10.3390/ph17101397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/04/2024] [Accepted: 10/05/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: The Trypanosoma cruzi infection promotes an intense inflammatory process that affects several tissues. The cholinergic system may exert a regulatory immune response and control the inflammatory process. This study aimed to evaluate the comparative effect of free and nanoencapsulated benznidazole in acute T. cruzi infection to assess hematological, biochemical, and oxidative status triggered by the cholinergic system. Methods: For this, fifty female Swiss mice were distributed in eight groups, i.e., uninfected and infected animals under four treatment protocols: untreated (control-CT); vehicle treatment (Eudragit L 100-EL-100); benznidazole treatment (BNZ); and nanoencapsulated benznidazole treatment (NBNZ). After eight treatment days, the animals were euthanized for sample collection. Results: The peak of parasitemia was at day 7 p.i., and the BNZ and NBNZ controlled and reduced the parasite rate but showed no efficacy in terms of total elimination of parasites analyzed by RT-PCR in both infected groups. The infection promotes significant anemia, leukopenia, and thrombocytopenia, which the BNZ improves. There was an increase in AChE activity during infection, leading to a pro-inflammatory response and an increase in M1 and M2 mACh receptors in the BNZ group, showing that the treatment interacted with the cholinergic pathway. In addition, a pro-oxidative response was characterized in the infection and mainly in the infected BNZ and NBNZ groups. The histopathological analysis showed significative splenomegaly and inflammatory infiltrate in the heart, liver, and spleen. Conclusions: The administration of the BNZ or NBNZ reverses hematological, hepatic, and renal alterations through cholinergic signaling and stimulates a pro-inflammatory response during acute T. cruzi infection.
Collapse
Affiliation(s)
- Aniélen D. da Silva
- Department of Biochemistry and Molecular Biology, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (A.D.d.S.); (M.F.); (T.V.P.); (A.M.E.); (M.F.V.C.); (C.E.A.); (V.M.); (K.P.R.); (J.N.); (V.M.M.); (M.R.C.)
| | - Mateus Fracasso
- Department of Biochemistry and Molecular Biology, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (A.D.d.S.); (M.F.); (T.V.P.); (A.M.E.); (M.F.V.C.); (C.E.A.); (V.M.); (K.P.R.); (J.N.); (V.M.M.); (M.R.C.)
| | - Nathieli B. Bottari
- Department of Microbiology and Parasitology, Universidade Federal de Pelotas, Pelotas 96015-560, Brazil;
| | - Taís V. Palma
- Department of Biochemistry and Molecular Biology, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (A.D.d.S.); (M.F.); (T.V.P.); (A.M.E.); (M.F.V.C.); (C.E.A.); (V.M.); (K.P.R.); (J.N.); (V.M.M.); (M.R.C.)
| | - Ana M. Engelmann
- Department of Biochemistry and Molecular Biology, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (A.D.d.S.); (M.F.); (T.V.P.); (A.M.E.); (M.F.V.C.); (C.E.A.); (V.M.); (K.P.R.); (J.N.); (V.M.M.); (M.R.C.)
| | - Milagros F. V. Castro
- Department of Biochemistry and Molecular Biology, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (A.D.d.S.); (M.F.); (T.V.P.); (A.M.E.); (M.F.V.C.); (C.E.A.); (V.M.); (K.P.R.); (J.N.); (V.M.M.); (M.R.C.)
| | - Charles E. Assmann
- Department of Biochemistry and Molecular Biology, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (A.D.d.S.); (M.F.); (T.V.P.); (A.M.E.); (M.F.V.C.); (C.E.A.); (V.M.); (K.P.R.); (J.N.); (V.M.M.); (M.R.C.)
| | - Vitor Mostardeiro
- Department of Biochemistry and Molecular Biology, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (A.D.d.S.); (M.F.); (T.V.P.); (A.M.E.); (M.F.V.C.); (C.E.A.); (V.M.); (K.P.R.); (J.N.); (V.M.M.); (M.R.C.)
| | - Karine P. Reichert
- Department of Biochemistry and Molecular Biology, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (A.D.d.S.); (M.F.); (T.V.P.); (A.M.E.); (M.F.V.C.); (C.E.A.); (V.M.); (K.P.R.); (J.N.); (V.M.M.); (M.R.C.)
| | - Jelson Nauderer
- Department of Biochemistry and Molecular Biology, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (A.D.d.S.); (M.F.); (T.V.P.); (A.M.E.); (M.F.V.C.); (C.E.A.); (V.M.); (K.P.R.); (J.N.); (V.M.M.); (M.R.C.)
| | - Marcelo L. da Veiga
- Department of Pathology, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (M.L.d.V.); (M.I.U.M.d.R.)
| | - Maria Izabel U. M. da Rocha
- Department of Pathology, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (M.L.d.V.); (M.I.U.M.d.R.)
| | - Luiz Claudio Milleti
- Department of Animal Production, Universidade do Estado de Santa Catarina, Lages 88520-000, SC, Brazil; (L.C.M.); (G.B.d.N.)
| | - Gabriella B. das Neves
- Department of Animal Production, Universidade do Estado de Santa Catarina, Lages 88520-000, SC, Brazil; (L.C.M.); (G.B.d.N.)
| | - Samanta Gundel
- Center Science Heath, Universidade Franciscana, Santa Maria 97010-491, RS, Brazil; (S.G.); (A.F.O.)
| | - Aline F. Ourique
- Center Science Heath, Universidade Franciscana, Santa Maria 97010-491, RS, Brazil; (S.G.); (A.F.O.)
| | - Silvia G. Monteiro
- Department of Microbiology and Parasitology, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil;
| | - Vera M. Morsch
- Department of Biochemistry and Molecular Biology, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (A.D.d.S.); (M.F.); (T.V.P.); (A.M.E.); (M.F.V.C.); (C.E.A.); (V.M.); (K.P.R.); (J.N.); (V.M.M.); (M.R.C.)
| | - Maria Rosa Chitolina
- Department of Biochemistry and Molecular Biology, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (A.D.d.S.); (M.F.); (T.V.P.); (A.M.E.); (M.F.V.C.); (C.E.A.); (V.M.); (K.P.R.); (J.N.); (V.M.M.); (M.R.C.)
| | - Aleksandro S. Da Silva
- Department of Animal Science, Universidade do Estado de Santa Catarina, Chapecó 89815-630, SC, Brazil
| |
Collapse
|
3
|
Saliu JA. Machine Learning-Based Approach to Identify Inhibitors of Sterol-14-Alpha Demethylase: A Study on Chagas Disease. Bioinform Biol Insights 2024; 18:11779322241262635. [PMID: 39081668 PMCID: PMC11287730 DOI: 10.1177/11779322241262635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 05/23/2024] [Indexed: 08/02/2024] Open
Abstract
Objectives Chagas Disease, caused by the parasite Trypanosoma cruzi, remains a significant public health concern, particularly in Latin America. The current standard treatment for Chagas Disease, benznidazole, is associated with various side effects, necessitating the search for alternative therapeutic options. In this study, we aimed to identify potential therapeutics for Chagas Disease through a comprehensive computational analysis. Methods A library of compounds derived from Cananga odorata was screened using a combination of pharmacophore modeling, structure-based screening, and quantitative structure-activity relationship (QSAR) analysis. The pharmacophore model facilitated the efficient screening of the compound library, while the structure-based screening identified hit compounds with promising inhibitory potential against the target enzyme, sterol-14-alpha demethylase. Results The QSAR model predicted the bioactivity of the hit compounds, revealing one compound to exhibit superior activity compared to benznidazole. Evaluation of the physicochemical, pharmacokinetic, toxicity, and medicinal chemistry properties of the hit compounds indicated their drug-like characteristics, oral bioavailability, ease of synthesis, and reduced toxicity profiles. Conclusion Overall, our findings present a promising avenue for the discovery of novel therapeutics for Chagas Disease. The identified hit compounds possess favorable drug-like properties and demonstrate potent inhibitory effects against the target enzyme. Further in vitro and in vivo studies are warranted to validate their efficacy and safety profiles.
Collapse
Affiliation(s)
- Jamiyu A Saliu
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Nigeria
| |
Collapse
|
4
|
Torchelsen FKVDS, Mazzeti AL, Mosqueira VCF. Drugs in preclinical and early clinical development for the treatment of Chagas´s disease: the current status. Expert Opin Investig Drugs 2024; 33:575-590. [PMID: 38686546 DOI: 10.1080/13543784.2024.2349289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2024]
Abstract
INTRODUCTION Chagas disease is spreading faster than expected in different countries, and little progress has been reported in the discovery of new drugs to combat Trypanosoma cruzi infection in humans. Recent clinical trials have ended with small hope. The pathophysiology of this neglected disease and the genetic diversity of parasites are exceptionally complex. The only two drugs available to treat patients are far from being safe, and their efficacy in the chronic phase is still unsatisfactory. AREAS COVERED This review offers a comprehensive examination and critical review of data reported in the last 10 years, and it is focused on findings of clinical trials and data acquired in vivo in preclinical studies. EXPERT OPINION The in vivo investigations classically in mice and dog models are also challenging and time-consuming to attest cure for infection. Poorly standardized protocols, availability of diagnosis methods and disease progression markers, the use of different T. cruzi strains with variable benznidazole sensitivities, and animals in different acute and chronic phases of infection contribute to it. More synchronized efforts between research groups in this field are required to put in evidence new promising substances, drug combinations, repurposing strategies, and new pharmaceutical formulations to impact the therapy.
Collapse
Affiliation(s)
- Fernanda Karoline Vieira da Silva Torchelsen
- School of Pharmacy, Federal University of Ouro Preto, Ouro Preto, Brazil
- Post-Graduation Program in Pharmaceutical Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Ana Lia Mazzeti
- Department of Biomedical Sciences and Health, Academic Unit of Passos, University of Minas Gerais State, Passos, Brazil
| | | |
Collapse
|
5
|
Murta SMF, Lemos Santana PA, Jacques Dit Lapierre TJW, Penteado AB, El Hajje M, Navarro Vinha TC, Liarte DB, de Souza ML, Goulart Trossini GH, de Oliveira Rezende Júnior C, de Oliveira RB, Ferreira RS. New drug discovery strategies for the treatment of benznidazole-resistance in Trypanosoma cruzi, the causative agent of Chagas disease. Expert Opin Drug Discov 2024; 19:741-753. [PMID: 38715393 DOI: 10.1080/17460441.2024.2349155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/25/2024] [Indexed: 05/22/2024]
Abstract
INTRODUCTION Benznidazole, the drug of choice for treating Chagas Disease (CD), has significant limitations, such as poor cure efficacy, mainly in the chronic phase of CD, association with side effects, and parasite resistance. Understanding parasite resistance to benznidazole is crucial for developing new drugs to treat CD. AREAS COVERED Here, the authors review the current understanding of the molecular basis of benznidazole resistance. Furthermore, they discuss the state-of-the-art methods and critical outcomes employed to evaluate the efficacy of potential drugs against T. cruzi, aiming to select better compounds likely to succeed in the clinic. Finally, the authors describe the different strategies employed to overcome resistance to benznidazole and find effective new treatments for CD. EXPERT OPINION Resistance to benznidazole is a complex phenomenon that occurs naturally among T. cruzi strains. The combination of compounds that inhibit different metabolic pathways of the parasite is an important strategy for developing a new chemotherapeutic protocol.
Collapse
Affiliation(s)
- Silvane Maria Fonseca Murta
- Grupo de Genômica Funcional de Parasitos - Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Pedro Augusto Lemos Santana
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | - André Berndt Penteado
- Departamento de Farmacia, Faculdade de Ciencias Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Marissa El Hajje
- Departamento de Farmacia, Faculdade de Ciencias Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | | | | | - Mariana Laureano de Souza
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, 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
|
6
|
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
|
7
|
Jayawardhana S, Ward AI, Francisco AF, Lewis MD, Taylor MC, Kelly JM, Olmo F. Benznidazole treatment leads to DNA damage in Trypanosoma cruzi and the persistence of rare widely dispersed non-replicative amastigotes in mice. PLoS Pathog 2023; 19:e1011627. [PMID: 37956215 PMCID: PMC10681306 DOI: 10.1371/journal.ppat.1011627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/27/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Benznidazole is the front-line drug used to treat infections with Trypanosoma cruzi, the causative agent of Chagas disease. However, for reasons that are unknown, treatment failures are common. When we examined parasites that survived benznidazole treatment in mice using highly sensitive in vivo and ex vivo bioluminescence imaging, we found that recrudescence is not due to persistence of parasites in a specific organ or tissue that preferentially protects them from drug activity. Surviving parasites are widely distributed and located in host cells where the vast majority contained only one or two amastigotes. Therefore, infection relapse does not arise from a small number of intact large nests. Rather, persisters are either survivors of intracellular populations where co-located parasites have been killed, or amastigotes in single/low-level infected cells exist in a state where they are less susceptible to benznidazole. To better assess the nature of parasite persisters, we exposed infected mammalian cell monolayers to a benznidazole regimen that reduces the intracellular amastigote population to <1% of the pre-treatment level. Of host cells that remained infected, as with the situation in vivo, the vast majority contained only one or two surviving intracellular amastigotes. Analysis, based on non-incorporation of the thymidine analogue EdU, revealed these surviving parasites to be in a transient non-replicative state. Furthermore, treatment with benznidazole led to widespread parasite DNA damage. When the small number of parasites which survive in mice after non-curative treatment were assessed using EdU labelling, this revealed that these persisters were also initially non-replicative. A possible explanation could be that triggering of the T. cruzi DNA damage response pathway by the activity of benznidazole metabolites results in exit from the cell cycle as parasites attempt DNA repair, and that metabolic changes associated with non-proliferation act to reduce drug susceptibility. Alternatively, a small percentage of the parasite population may pre-exist in this non-replicative state prior to treatment.
Collapse
Affiliation(s)
- Shiromani Jayawardhana
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Alexander I. Ward
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Amanda F. Francisco
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Michael D. Lewis
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Martin C. Taylor
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - John M. Kelly
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Francisco Olmo
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
8
|
González-González A, Vázquez C, Encalada R, Saavedra E, Vázquez-Jiménez LK, Ortiz-Pérez E, Bolognesi ML, Rivera G. Phenothiazine-based virtual screening, molecular docking, and molecular dynamics of new trypanothione reductase inhibitors of Trypanosoma cruzi. Mol Inform 2023; 42:e2300069. [PMID: 37490403 DOI: 10.1002/minf.202300069] [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: 03/21/2023] [Revised: 06/26/2023] [Accepted: 07/25/2023] [Indexed: 07/27/2023]
Abstract
Phenothiazine derivatives can unselectively inhibit the trypanothione-dependent antioxidant system enzyme trypanothione reductase (TR). A virtual screening of 2163 phenothiazine derivatives from the ZINC15 and PubChem databases docked on the active site of T. cruzi TR showed that 285 compounds have higher affinity than the natural ligand trypanothione disulfide. 244 compounds showed higher affinity toward the parasite's enzyme than to its human homolog glutathione reductase. Protein-ligand interaction profiling predicted that the main interactions for the top scored compounds were with residues important for trypanothione disulfide binding: Phe396, Pro398, Leu399, His461, Glu466, and Glu467, particularly His461, which participates in catalysis. Two compounds with the desired profiles, ZINC1033681 (Zn_C687) and ZINC10213096 (Zn_C216), decreased parasite growth by 20 % and 50 %, respectively. They behaved as mixed-type inhibitors of recombinant TR, with Ki values of 59 and 47 μM, respectively. This study provides a further understanding of the potential of phenothiazine derivatives as TR inhibitors.
Collapse
Affiliation(s)
- Alonzo González-González
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, 88710, Reynosa, México
| | - Citlali Vázquez
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, 14080, Mexico City, Mexico
| | - Rusely Encalada
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, 14080, Mexico City, Mexico
| | - Emma Saavedra
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, 14080, Mexico City, Mexico
| | - Lenci K Vázquez-Jiménez
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, 88710, Reynosa, México
| | - Eyra Ortiz-Pérez
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, 88710, Reynosa, México
| | - María Laura Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, I-40126, Bologna, Italy
| | - Gildardo Rivera
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, 88710, Reynosa, México
| |
Collapse
|
9
|
Martinez MZ, Olmo F, Taylor MC, Caudron F, Wilkinson SR. Dissecting the interstrand crosslink DNA repair system of Trypanosoma cruzi. DNA Repair (Amst) 2023; 125:103485. [PMID: 36989950 DOI: 10.1016/j.dnarep.2023.103485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/08/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023]
Abstract
DNA interstrand crosslinks (ICLs) are toxic lesions that can block essential biological processes. Here we show Trypanosoma cruzi, the causative agent of Chagas disease, is susceptible to ICL-inducing compounds including mechlorethamine and novel nitroreductase-activated prodrugs that have potential in treating this infection. To resolve such lesions, cells co-opt enzymes from "classical" DNA repair pathways that alongside dedicated factors operate in replication-dependent and -independent mechanisms. To assess ICL repair in T. cruzi, orthologues of SNM1, MRE11 and CSB were identified and their function assessed. The T. cruzi enzymes could complement the mechlorethamine susceptibility phenotype displayed by corresponding yeast and/or T. brucei null confirming their role as ICL repair factors while GFP-tagged TcSNM1, TcMRE11 and TcCSB were shown to localise to the nuclei of insect and/or intracellular form parasites. Gene disruption demonstrated that while each activity was non-essential for T. cruzi viability, nulls displayed a growth defect in at least one life cycle stage with TcMRE11-deficient trypomastigotes also compromised in mammalian cell infectivity. Phenotyping revealed all nulls were more susceptible to mechlorethamine than controls, a trait complemented by re-expression of the deleted gene. To assess interplay, the gene disruption approach was extended to generate T. cruzi deficient in TcSNM1/TcMRE11 or in TcSNM1/TcCSB. Analysis demonstrated these activities functioned across two ICL repair pathways with TcSNM1 and TcMRE11 postulated to operate in a replication-dependent system while TcCSB helps resolve transcription-blocking lesions. By unravelling how T. cruzi repairs ICL damage, specific inhibitors targeting repair components could be developed and used to increase the potency of trypanocidal ICL-inducing compounds.
Collapse
Affiliation(s)
- Monica Zavala Martinez
- School of Biological & Chemical Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Francisco Olmo
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Martin C Taylor
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Fabrice Caudron
- School of Biological & Chemical Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Shane R Wilkinson
- School of Biological & Chemical Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK.
| |
Collapse
|
10
|
González-González A, Sánchez-Sánchez O, Krauth-Siegel RL, Bolognesi ML, Gớmez-Escobedo R, Nogueda-Torres B, Vázquez-Jiménez LK, Saavedra E, Encalada R, Espinoza-Hicks JC, Paz-González AD, Rivera G. In Vitro and In Silico Analysis of New n-Butyl and Isobutyl Quinoxaline-7-carboxylate 1,4-di- N-oxide Derivatives against Trypanosoma cruzi as Trypanothione Reductase Inhibitors. Int J Mol Sci 2022; 23:13315. [PMID: 36362102 PMCID: PMC9655728 DOI: 10.3390/ijms232113315] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/24/2022] [Accepted: 10/27/2022] [Indexed: 09/29/2023] Open
Abstract
American trypanosomiasis is a worldwide health problem that requires attention due to ineffective treatment options. We evaluated n-butyl and isobutyl quinoxaline-7-carboxylate 1,4-di-N-oxide derivatives against trypomastigotes of the Trypanosoma cruzi strains NINOA and INC-5. An in silico analysis of the interactions of 1,4-di-N-oxide on the active site of trypanothione reductase (TR) and an enzyme inhibition study was carried out. The n-butyl series compound identified as T-150 had the best trypanocidal activity against T. cruzi trypomastigotes, with a 13% TR inhibition at 44 μM. The derivative T-147 behaved as a mixed inhibitor with Ki and Ki' inhibition constants of 11.4 and 60.8 µM, respectively. This finding is comparable to the TR inhibitor mepacrine (Ki = 19 µM).
Collapse
Affiliation(s)
- Alonzo González-González
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico
| | - Oscar Sánchez-Sánchez
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico
| | - R. Luise Krauth-Siegel
- Center of Biochemistry, Heidelberg University, Im Neuenheimer Feld 328, 69120 Heidelberg, Germany
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, I-40126 Bologna, Italy
| | - Rogelio Gớmez-Escobedo
- Departamento de Parasitología, Escuela Nacional de Ciencias Biológicas Instituto Politécnico Nacional, Ciudad de Mexico 07738, Mexico
| | - Benjamín Nogueda-Torres
- Departamento de Parasitología, Escuela Nacional de Ciencias Biológicas Instituto Politécnico Nacional, Ciudad de Mexico 07738, Mexico
| | - Lenci K. Vázquez-Jiménez
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico
| | - Emma Saavedra
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de Mexico 14080, Mexico
| | - Rusely Encalada
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de Mexico 14080, Mexico
| | | | - Alma D. Paz-González
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico
| | - Gildardo Rivera
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico
| |
Collapse
|
11
|
Santi AMM, Ribeiro JM, Reis-Cunha JL, Burle-Caldas GDA, Santos IFM, Silva PA, Resende DDM, Bartholomeu DC, Teixeira SMR, Murta SMF. Disruption of multiple copies of the Prostaglandin F2alpha synthase gene affects oxidative stress response and infectivity in Trypanosoma cruzi. PLoS Negl Trop Dis 2022; 16:e0010845. [PMID: 36260546 PMCID: PMC9581433 DOI: 10.1371/journal.pntd.0010845] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/26/2022] [Indexed: 11/08/2022] Open
Abstract
Chagas disease, caused by the protozoan Trypanosoma cruzi, is a serious chronic parasitic disease, currently treated with Nifurtimox (NFX) and Benznidazole (BZ). In addition to high toxicity, these drugs have low healing efficacy, especially in the chronic phase of the disease. The existence of drug-resistant T. cruzi strains and the occurrence of cross-resistance between BZ and NFX have also been described. In this context, it is urgent to study the metabolism of these drugs in T. cruzi, to better understand the mechanisms of resistance. Prostaglandin F2α synthase (PGFS) is an enzyme that has been correlated with parasite resistance to BZ, but the mechanism by which resistance occurs is still unclear. Our results show that the genome of the CL Brener clone of T. cruzi, contains five PGFS sequences and three potential pseudogenes. Using CRISPR/Cas9 we generated knockout cell lines in which all PGFS sequences were disrupted, as shown by PCR and western blotting analyses. The PGFS deletion did not alter the growth of the parasites or their susceptibility to BZ and NFX when compared to wild-type (WT) parasites. Interestingly, NTR-1 transcripts were shown to be upregulated in ΔPGFS mutants. Furthermore, the ΔPGFS parasites were 1.6 to 1.7-fold less tolerant to oxidative stress generated by menadione, presented lower levels of lipid bodies than the control parasites during the stationary phase, and were less infective than control parasites.
Collapse
Affiliation(s)
- Ana Maria Murta Santi
- Grupo Genômica Funcional de Parasitos, Instituto René Rachou, Fiocruz Minas, Belo Horizonte, Minas Gerais, Brazil
| | - Juliana Martins Ribeiro
- Grupo Genômica Funcional de Parasitos, Instituto René Rachou, Fiocruz Minas, Belo Horizonte, Minas Gerais, Brazil
| | - João Luís Reis-Cunha
- Departamento de Parasitologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Departamento de Medicina Veterinária Preventiva, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | | | - Paula Alves Silva
- Grupo Genômica Funcional de Parasitos, Instituto René Rachou, Fiocruz Minas, Belo Horizonte, Minas Gerais, Brazil
| | - Daniela de Melo Resende
- Grupo Genômica Funcional de Parasitos, Instituto René Rachou, Fiocruz Minas, Belo Horizonte, Minas Gerais, Brazil
| | | | | | - Silvane Maria Fonseca Murta
- Grupo Genômica Funcional de Parasitos, Instituto René Rachou, Fiocruz Minas, Belo Horizonte, Minas Gerais, Brazil
- * E-mail:
| |
Collapse
|
12
|
Travi BL. Current status of antihistamine drugs repurposing for infectious diseases. MEDICINE IN DRUG DISCOVERY 2022. [DOI: 10.1016/j.medidd.2022.100140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
13
|
Functionalized Nitroimidazole Scaffold Construction and Their Pharmaceutical Applications: A 1950–2021 Comprehensive Overview. Pharmaceuticals (Basel) 2022; 15:ph15050561. [PMID: 35631389 PMCID: PMC9144801 DOI: 10.3390/ph15050561] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/28/2022] [Accepted: 04/01/2022] [Indexed: 11/16/2022] Open
Abstract
Nitroimidazole represents one of the most essential and unique scaffolds in drug discovery since its discovery in the 1950s. It was K. Maeda in Japan who reported in 1953 the first nitroimidazole as a natural product from Nocardia mesenterica with antibacterial activity, which was later identified as Azomycin 1 (2-nitroimidazole) and remained in focus until now. This natural antibiotic was the starting point for synthesizing numerous analogs and regio-isomers, leading to several life-saving drugs and clinical candidates against a number of diseases, including infections (bacterial, viral, parasitic) and cancers, as well as imaging agents in medicine/diagnosis. In the present decade, the nitroimidazole scaffold has again been given two life-saving drugs (Delamanid and Pretomanid) used to treat MDR (multi-drug resistant) tuberculosis. Keeping in view the highly successful track-record of the nitroimidazole scaffold in providing breakthrough therapeutic drugs, this comprehensive review focuses explicitly on presenting the activity profile and synthetic chemistry of functionalized nitroimidazole (2-, 4- and 5-nitroimidazoles as well as the fused nitroimidazoles) based drugs and leads published from 1950 to 2021. The present review also presents the miscellaneous examples in each class. In addition, the mutagenic profile of nitroimidazole-based drugs and leads and derivatives is also discussed.
Collapse
|
14
|
Antimicrobials and Resistance Part II: Antifungals, Antivirals, and Antiparasitics. J Am Acad Dermatol 2022; 86:1207-1226. [DOI: 10.1016/j.jaad.2021.11.065] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/27/2021] [Accepted: 11/29/2021] [Indexed: 11/18/2022]
|
15
|
Farani PSG, Ferreira BIS, Gibaldi D, Lannes-Vieira J, Moreira OC. Modulation of miR-145-5p and miR-146b-5p levels is linked to reduced parasite load in H9C2 Trypanosoma cruzi infected cardiomyoblasts. Sci Rep 2022; 12:1436. [PMID: 35082354 PMCID: PMC8791985 DOI: 10.1038/s41598-022-05493-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 01/06/2022] [Indexed: 11/15/2022] Open
Abstract
In the heart tissue of acutely Trypanosoma cruzi-infected mice miR-145-5p and miR-146b-5p are, respectively, downregulated and upregulated. Here, we used the H9C2 rat cardiomyoblast cell line infected with the Colombian T. cruzi strain to investigate the parasite-host cell interplay, focusing on the regulation of miR-145-5p and miR-146b-5p expression. Next, we explored the effects of interventions with the trypanosomicidal drug Benznidazole (Bz) alone or combined with Pentoxifylline (PTX), a methylxanthine derivative shown to modulate immunological and cardiac abnormalities in a model of chronic chagasic cardiomyopathy, on parasite load and expression of miR-145-5p and miR-146b-5p. The infection of H9C2 cells with trypomastigote forms allowed parasite cycle with intracellular forms multiplication and trypomastigote release. After 48 and 144 h of infection, upregulation of miR-145-5p (24 h: 2.38 ± 0.26; 48 h: 3.15 ± 0.9-fold change) and miR-146b-5b (24 h: 2.60 ± 0.46; 48 h: 2.97 ± 0.23-fold change) was detected. The peak of both miRNA levels paralleled with release of trypomastigote forms. Addition of 3 µM and 10 µM of Bz 48 h after infection reduced parasite load but did not interfere with miR-145-5p and miR-146b-5p levels. Addition of PTX did not interfere with Bz-induced parasite control efficacy. Conversely, combined Bz + PTX treatment decreased the levels of both microRNAs, resembling the expression levels detected in non-infected H9C2 cells. Moreover, the use of miR-145-5p and miR-146b-5p mimic/inhibitor systems before infection of H9C2 cells decreased parasite load, 72 h postinfection. When H9C2 cells were treated with miR-145-5p and miR-146b-5p mimic/inhibitor 48 h after infection, all the used systems, except the miR-146b-5p inhibitor, reduced parasite load. Altogether, our data indicate that these microRNAs putatively control signaling pathways crucial for parasite–host cell interaction. Thus, miR-145-5p and miR-146b-5p deserve to be further investigated as biomarkers of parasite control and tools to identify therapeutic adjuvants to etiological treatment in Chagas disease.
Collapse
Affiliation(s)
- Priscila Silva Grijó Farani
- Real Time PCR Platform RPT09A, Laboratory of Molecular Biology and Endemic Diseases, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,Laboratory of Biology of the Interactions, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Beatriz Iandra Silva Ferreira
- Real Time PCR Platform RPT09A, Laboratory of Molecular Biology and Endemic Diseases, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Daniel Gibaldi
- Laboratory of Biology of the Interactions, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Joseli Lannes-Vieira
- Laboratory of Biology of the Interactions, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Otacilio Cruz Moreira
- Real Time PCR Platform RPT09A, Laboratory of Molecular Biology and Endemic Diseases, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.
| |
Collapse
|
16
|
Beilstein S, El Phil R, Sahraoui SS, Scapozza L, Kaiser M, Mäser P. Laboratory Selection of Trypanosomatid Pathogens for Drug Resistance. Pharmaceuticals (Basel) 2022; 15:ph15020135. [PMID: 35215248 PMCID: PMC8879015 DOI: 10.3390/ph15020135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 11/16/2022] Open
Abstract
The selection of parasites for drug resistance in the laboratory is an approach frequently used to investigate the mode of drug action, estimate the risk of emergence of drug resistance, or develop molecular markers for drug resistance. Here, we focused on the How rather than the Why of laboratory selection, discussing different experimental set-ups based on research examples with Trypanosoma brucei, Trypanosoma cruzi, and Leishmania spp. The trypanosomatids are particularly well-suited to illustrate different strategies of selecting for drug resistance, since it was with African trypanosomes that Paul Ehrlich performed such an experiment for the first time, more than a century ago. While breakthroughs in reverse genetics and genome editing have greatly facilitated the identification and validation of candidate resistance mutations in the trypanosomatids, the forward selection of drug-resistant mutants still relies on standard in vivo models and in vitro culture systems. Critical questions are: is selection for drug resistance performed in vivo or in vitro? With the mammalian or with the insect stages of the parasites? Under steady pressure or by sudden shock? Is a mutagen used? While there is no bona fide best approach, we think that a methodical consideration of these questions provides a helpful framework for selection of parasites for drug resistance in the laboratory.
Collapse
Affiliation(s)
- Sabina Beilstein
- Department Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, 4051 Basel, Switzerland; (S.B.); (M.K.)
- Swiss TPH, University of Basel, Petersplatz 1, 4002 Basel, Switzerland
| | - Radhia El Phil
- School of Pharmaceutical Sciences, University of Geneva, 1205 Geneva, Switzerland; (R.E.P.); (S.S.S.); (L.S.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| | - Suzanne Sherihan Sahraoui
- School of Pharmaceutical Sciences, University of Geneva, 1205 Geneva, Switzerland; (R.E.P.); (S.S.S.); (L.S.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| | - Leonardo Scapozza
- School of Pharmaceutical Sciences, University of Geneva, 1205 Geneva, Switzerland; (R.E.P.); (S.S.S.); (L.S.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| | - Marcel Kaiser
- Department Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, 4051 Basel, Switzerland; (S.B.); (M.K.)
- Swiss TPH, University of Basel, Petersplatz 1, 4002 Basel, Switzerland
| | - Pascal Mäser
- Department Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, 4051 Basel, Switzerland; (S.B.); (M.K.)
- Swiss TPH, University of Basel, Petersplatz 1, 4002 Basel, Switzerland
- Correspondence: ; Tel.: +41-61-284-8338
| |
Collapse
|
17
|
Martinez SJ, Nardella GN, Rodríguez ME, Rivero CV, Agüero F, Romano PS. Biological features of TcM: A new Trypanosoma cruzi isolate from Argentina classified into TcV lineage. CURRENT RESEARCH IN MICROBIAL SCIENCES 2022; 3:100152. [PMID: 35909611 PMCID: PMC9325899 DOI: 10.1016/j.crmicr.2022.100152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
TcM is a new T. cruzi isolate that belongs to DTU TcV. TcV is a T. cruzi linage prevalent in human infections of Argentina. TcM is less virulent that TcY strain. TcM displays slow-growing rate and muscle tissue tropism. TcM is more susceptible to benznidazole than TcY.
Trypanosoma cruzi, the etiologic agent of Chagas disease (CD) presents a wide genetic and phenotypic diversity that is classified into seven lineages or discrete typing units (DTU: TcI to TcVI and Tcbat). Although isolates and strains that belong to a particular group can share some attributes, such as geographic distribution, others like growth rate, cell tropism, and response to treatment can be highly variable. In addition, studies that test new trypanocidal drugs are frequently conducted on T. cruzi strains maintained for a long time in axenic culture, resulting in changes in parasite virulence and other important features. This work aimed to isolate and characterize a new T. cruzi strain from a chronic Chagas disease patient. The behavior of this isolate was studied by using standard in vitro assays and in vivo mice infection tests and compared with the T. cruzi Y strain (TcY), broadly used in research laboratories worldwide. Data showed that TcM behaves as a slow-growing strain in vitro that develops chronic infections in mice and displays high tropism to muscular tissues, in accordance with its clinical performance. In contrast, the Y strain behaved as an acute strain that can infect different types of cells and tissues. Interestingly, TcM, which belongs to DTU TcV, is more susceptible to benznidazole than TcY, a TcII strain considered moderately resistant to this drug. These differential properties contribute to the characterization of a TcV strain, one of the main lineages in the southern countries of South America, and open the possibility to introduce changes that improve the management of Chagas patients in the future
Collapse
Affiliation(s)
- Santiago José Martinez
- Laboratorio de Biología de Trypanosoma cruzi y la célula hospedadora. Instituto de Histología y Embriología, Universidad Nacional de Cuyo (IHEM-CONICET-UNCUYO), Mendoza, Argentina
| | | | - Matías Exequiel Rodríguez
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (IIB-CONICET-UNSAM), Buenos Aires, Argentina
| | - Cynthia Vanesa Rivero
- Laboratorio de Biología de Trypanosoma cruzi y la célula hospedadora. Instituto de Histología y Embriología, Universidad Nacional de Cuyo (IHEM-CONICET-UNCUYO), Mendoza, Argentina
| | - Fernán Agüero
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (IIB-CONICET-UNSAM), Buenos Aires, Argentina
| | - Patricia Silvia Romano
- Laboratorio de Biología de Trypanosoma cruzi y la célula hospedadora. Instituto de Histología y Embriología, Universidad Nacional de Cuyo (IHEM-CONICET-UNCUYO), Mendoza, Argentina
- Facultad de Ciencias Médicas. Universidad Nacional de Cuyo (FCM-UNCUYO), Mendoza, Argentina
- Corresponding author at: Instituto de Histología y Embriología, Universidad Nacional de Cuyo (IHEM-CONICET-UNCUYO), Casilla de Correo 56, Centro Universitario, Parque General San Martín, (5500) Mendoza, Argentina
| |
Collapse
|
18
|
Dumoulin PC, Burleigh BA. Metabolic flexibility in Trypanosoma cruzi amastigotes: implications for persistence and drug sensitivity. Curr Opin Microbiol 2021; 63:244-249. [PMID: 34455305 DOI: 10.1016/j.mib.2021.07.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/19/2021] [Accepted: 07/24/2021] [Indexed: 12/18/2022]
Abstract
Throughout their life cycle, parasitic organisms experience a variety of environmental conditions. To ensure persistence and transmission, some protozoan parasites are capable of adjusting their replication or converting to distinct life cycle stages. Trypanosoma cruzi is a 'generalist' parasite that is competent to infect various insect (triatomine) vectors and mammalian hosts. Within the mammalian host, T. cruzi replicates intracellularly as amastigotes and can persist for the lifetime of the host. The persistence of the parasites in tissues can lead to the development of Chagas disease. Recent work has identified growth plasticity and metabolic flexibility as aspects of amastigote biology that are important determinants of persistence in varied growth conditions and under drug pressure. A better understanding of the link between amastigote and host/tissue metabolism will aid in the development of new drugs or therapies that can limit disease pathology.
Collapse
Affiliation(s)
- Peter C Dumoulin
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, United States.
| | - Barbara A Burleigh
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, United States
| |
Collapse
|
19
|
Maldonado E, Morales-Pison S, Urbina F, Solari A. Molecular and Functional Characteristics of DNA Polymerase Beta-Like Enzymes From Trypanosomatids. Front Cell Infect Microbiol 2021; 11:670564. [PMID: 34422676 PMCID: PMC8375306 DOI: 10.3389/fcimb.2021.670564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 06/08/2021] [Indexed: 11/16/2022] Open
Abstract
Trypanosomatids are a group of primitive unicellular eukaryotes that can cause diseases in plants, insects, animals, and humans. Kinetoplast genome integrity is key to trypanosomatid cell survival and viability. Kinetoplast DNA (kDNA) is usually under attack by reactive oxygen and nitric species (ROS and RNS), damaging the DNA, and the cells must remove and repair those oxidatively generated lesions in order to survive and proliferate. Base excision repair (BER) is a well-conserved pathway for DNA repair after base damage, single-base loss, and single-strand breaks, which can arise from ROS, RSN, environmental genotoxic agents, and UV irradiation. A powerful BER system has been described in the T. cruzi kinetoplast and it is mainly carried out by DNA polymerase β (pol β) and DNA polymerase β-PAK (pol β-PAK), which are kinetoplast-located in T. cruzi as well as in other trypanosomatids. Both pol β and pol β-PAK belong to the X-family of DNA polymerases (pol X family), perform BER in trypanosomatids, and display intrinsic 5-deoxyribose phosphate (dRP) lyase and DNA polymerase activities. However, only Pol β-PAK is able to carry out trans-lesion synthesis (TLS) across 8oxoG lesions. T. cruzi cells overexpressing pol β are more resistant to ROS and are also more efficient to repair 8oxoG compared to control cells. Pol β seems to play a role in kDNA replication, since it associates with kinetoplast antipodal sites in those development stages in trypanosomatids which are competent for cell replication. ROS treatment of cells induces the overexpression of pol β, indicating that plays a role in kDNA repair. In this review, we will summarize the main features of trypanosomatid minicircle kDNA replication and the biochemical characteristics of pol β-like enzymes and their involvement in BER and kDNA replication. We also summarize key structural features of trypanosomatid pol β compared to their mammalian (human) counterpart.
Collapse
Affiliation(s)
- Edio Maldonado
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Sebastian Morales-Pison
- Laboratorio de Genética Molecular Humana, Programa de Genética Humana, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Fabiola Urbina
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Aldo Solari
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
20
|
Dattani A, Drammeh I, Mahmood A, Rahman M, Szular J, Wilkinson SR. Unraveling the antitrypanosomal mechanism of benznidazole and related 2-nitroimidazoles: From prodrug activation to DNA damage. Mol Microbiol 2021; 116:674-689. [PMID: 34061384 DOI: 10.1111/mmi.14763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/17/2021] [Accepted: 05/29/2021] [Indexed: 12/27/2022]
Abstract
Nitroheterocycles represent an important class of compound used to treat trypanosomiasis. They often function as prodrugs and can undergo type I nitroreductase (NTR1)-mediated activation before promoting their antiparasitic activities although the nature of these downstream effects has yet to be determined. Here, we show that in an NTR1-dependent process, benznidazole promotes DNA damage in the nuclear genome of Trypanosoma brucei, providing the first direct link between activation of this prodrug and a downstream trypanocidal mechanism. Phenotypic and protein expression studies revealed that components of the trypanosome's homologous recombination (HR) repair pathway (TbMRE11, γH2A, TbRAD51) cooperate to resolve the benznidazole-induced damage, indicating that the prodrug-induced lesions are most likely double stand DNA breaks, while the sequence/recruitment kinetics of these factors parallels that in other eukaryotes HR systems. When extended to other NTR1-activated 2-nitroimidazoles, some were shown to promote DNA damage. Intriguingly, the lesions induced by these required TbMRE11 and TbCSB activities to fix leading us to postulate that TbCSB may operate in systems other than the transcription-coupled nucleotide excision repair pathway. Understanding how existing trypanosomal drugs work will aid future drug design and help unlock novel reactions/pathways that could be exploited as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Ambika Dattani
- School of Biological & Chemical Sciences, Queen Mary University of London, London, UK
| | - Isatou Drammeh
- School of Biological & Chemical Sciences, Queen Mary University of London, London, UK
| | - Aishah Mahmood
- School of Biological & Chemical Sciences, Queen Mary University of London, London, UK
| | - Mahbubur Rahman
- School of Biological & Chemical Sciences, Queen Mary University of London, London, UK
| | - Joanna Szular
- School of Biological & Chemical Sciences, Queen Mary University of London, London, UK
| | - Shane R Wilkinson
- School of Biological & Chemical Sciences, Queen Mary University of London, London, UK
| |
Collapse
|
21
|
Pandey RP, Nascimento MS, Moore CE, Raj VS, Kalil J, Cunha-Neto E. New Approaches for the Treatment of Chagas Disease. Curr Drug Targets 2021; 22:835-841. [PMID: 33238855 DOI: 10.2174/1389450121999201124122643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 11/22/2022]
Abstract
Chagas disease, caused by the protozoan Trypanosoma cruzi is a neglected tropical disease with high prevalence (5.7 million in Latin America, WHO 2015), significant burden, and significant morbimortality mostly due to severe heart disorders during the chronic phase of infection. Chagas disease is endemic in Latin America, and medical care for the disease is the major expense for Brazil's Universal Healthcare System (Sistema Único de Saúde (SUS). The efficacy of the available drugs benznidazole and nifurtimox are low for the chronic phase of Chagas disease, the phase in which most patients are diagnosed, and there are frequent side effects, and drug resistance occurs. The rapid deployment of new drug regimens that are effective for the chronic phase treatment is low-cost and less toxic than the currently available therapy, which is a global priority. Repurposing drugs already in clinical use with other combinations would be the fastest and safest strategy for treating Chagas disease patients. We hypothesize that the combined treatment using repurposing drugs with benznidazole will be more efficacious than benznidazole alone. This needs to be tested further both in vitro and in animal models to understand the efficacy of the treatment before performing human clinical trials. We further hypothesize that producing nanoparticle formulation of the drugs can reduce their toxicity and improve therapeutic use.
Collapse
Affiliation(s)
- Ramendra Pati Pandey
- Centre for Drug Design Discovery and Development (C4D), SRM University, DelhiNCR, Rajiv Gandhi Education City, Sonepat - 131 029, Haryana, India
| | - Marilda Savoia Nascimento
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Catrin E Moore
- Nuffield Department of Medicine, University of Oxford, Big Data Institute, Li Ka Shing Centre for Health Information and Discovery Old Road Campus, Headington, Oxford, OX3 7LF, United Kingdom
| | - V Samuel Raj
- Centre for Drug Design Discovery and Development (C4D), SRM University, DelhiNCR, Rajiv Gandhi Education City, Sonepat - 131 029, Haryana, India
| | - Jorge Kalil
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Edecio Cunha-Neto
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
22
|
Florentino PTV, Mendes D, Vitorino FNL, Martins DJ, Cunha JPC, Mortara RA, Menck CFM. DNA damage and oxidative stress in human cells infected by Trypanosoma cruzi. PLoS Pathog 2021; 17:e1009502. [PMID: 33826673 PMCID: PMC8087042 DOI: 10.1371/journal.ppat.1009502] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 04/30/2021] [Accepted: 03/25/2021] [Indexed: 12/11/2022] Open
Abstract
Trypanosoma cruzi is the etiologic agent of Chagas' disease. Infected cells with T. cruzi activate several responses that promote unbalance of reactive oxygen species (ROS) that may cause DNA damage that activate cellular responses including DNA repair processes. In this work, HeLa cells and AC16 human cardiomyocyte cell line were infected with T. cruzi to investigate host cell responses at genome level during parasites intracellular life cycle. In fact, alkaline sensitive sites and oxidized DNA bases were detected in the host cell genetic material particularly in early stages of infection. These DNA lesions were accompanied by phosphorylation of the histone H2Ax, inducing γH2Ax, a marker of genotoxic stress. Moreover, Poly [ADP-ribose] polymerase-1 (PARP1) and 8-oxoguanine glycosylase (OGG1) are recruited to host cell nuclei, indicating activation of the DNA repair process. In infected cells, chromatin-associated proteins are carbonylated, as a possible consequence of oxidative stress and the nuclear factor erythroid 2-related factor 2 (NRF2) is induced early after infection, suggesting that the host cell antioxidant defenses are activated. However, at late stages of infection, NRF2 is downregulated. Interestingly, host cells treated with glutathione precursor, N-acetyl cysteine, NRF2 activator (Sulforaphane), and also Benznidonazol (BNZ) reduce parasite burst significantly, and DNA damage. These data indicate that the balance of oxidative stress and DNA damage induction in host cells may play a role during the process of infection itself, and interference in these processes may hamper T. cruzi infection, revealing potential target pathways for the therapy support.
Collapse
Affiliation(s)
- Pilar T. V. Florentino
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Davi Mendes
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Davi J. Martins
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Julia P. C. Cunha
- Special Laboratory of Cell Cycle, Butantan Institute, São Paulo, Brazil
| | - Renato A. Mortara
- Department of Microbiology, Imunology & Parasitology, Escola Paulista de Medicina Federal University of São Paulo, São Paulo, Brazil
| | - Carlos F. M. Menck
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
23
|
Mazzeti AL, Capelari-Oliveira P, Bahia MT, Mosqueira VCF. Review on Experimental Treatment Strategies Against Trypanosoma cruzi. J Exp Pharmacol 2021; 13:409-432. [PMID: 33833592 PMCID: PMC8020333 DOI: 10.2147/jep.s267378] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/03/2021] [Indexed: 12/11/2022] Open
Abstract
Chagas disease is a neglected tropical disease caused by the protozoan Trypanosoma cruzi. Currently, only nitroheterocyclic nifurtimox (NFX) and benznidazole (BNZ) are available for the treatment of Chagas disease, with limitations such as variable efficacy, long treatment regimens and toxicity. Different strategies have been used to discover new active molecules for the treatment of Chagas disease. Target-based and phenotypic screening led to thousands of compounds with anti-T. cruzi activity, notably the nitroheterocyclic compounds, fexinidazole and its metabolites. In addition, drug repurposing, drug combinations, re-dosing regimens and the development of new formulations have been evaluated. The CYP51 antifungal azoles, as posaconazole, ravuconazole and its prodrug fosravuconazole presented promising results in experimental Chagas disease. Drug combinations of nitroheterocyclic and azoles were able to induce cure in murine infection. New treatment schemes using BNZ showed efficacy in the experimental chronic stage, including against dormant forms of T. cruzi. And finally, sesquiterpene lactone formulated in nanocarriers displayed outstanding efficacy against different strains of T. cruzi, susceptible or resistant to BNZ, the reference drug. These pre-clinical results are encouraging and provide interesting evidence to improve the treatment of patients with Chagas disease.
Collapse
Affiliation(s)
- Ana Lia Mazzeti
- Laboratório de Desenvolvimento Galênico e Nanotecnologia, Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil.,Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, 21040-360, Brazil.,Laboratório de Doenças Parasitárias, Escola de Medicina & Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Patricia Capelari-Oliveira
- Laboratório de Desenvolvimento Galênico e Nanotecnologia, Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Maria Terezinha Bahia
- Laboratório de Doenças Parasitárias, Escola de Medicina & Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Vanessa Carla Furtado Mosqueira
- Laboratório de Desenvolvimento Galênico e Nanotecnologia, Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| |
Collapse
|
24
|
Bhutani P, Joshi G, Raja N, Bachhav N, Rajanna PK, Bhutani H, Paul AT, Kumar R. U.S. FDA Approved Drugs from 2015-June 2020: A Perspective. J Med Chem 2021; 64:2339-2381. [PMID: 33617716 DOI: 10.1021/acs.jmedchem.0c01786] [Citation(s) in RCA: 265] [Impact Index Per Article: 88.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the present work, we report compilation and analysis of 245 drugs, including small and macromolecules approved by the U.S. FDA from 2015 until June 2020. Nearly 29% of the drugs were approved for the treatment of various types of cancers. Other major therapeutic areas of focus were infectious diseases (14%); neurological conditions (12%); and genetic, metabolic, and cardiovascular disorders (7-8% each). Itemization of the approved drugs according to the year of approval, sponsor, target, chemical class, major drug-metabolizing enzyme(s), route of administration/elimination, and drug-drug interaction liability (perpetrator or/and victim) is presented and discussed. An effort has been made to analyze the pharmacophores to identify the structural (e.g., aromatic, heterocycle, and aliphatic), elemental (e.g., boron, sulfur, fluorine, phosphorus, and deuterium), and functional group (e.g., nitro drugs) diversity among the approved drugs. Further, descriptor-based chemical space analysis of FDA approved drugs and several strategies utilized for optimizing metabolism leading to their discoveries have been emphasized. Finally, an analysis of drug-likeness for the approved drugs is presented.
Collapse
Affiliation(s)
- Priyadeep Bhutani
- Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb R&D Centre, Syngene International Limited, Bangalore 560099, India.,Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Rajasthan 333031, India
| | - Gaurav Joshi
- Laboratory for Drug Design and Synthesis, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda 151001, India
| | - Nivethitha Raja
- Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb R&D Centre, Syngene International Limited, Bangalore 560099, India
| | - Namrata Bachhav
- 1015 E Cozza Drive # 12, Spokane Washington 99208, United States
| | - Prabhakar K Rajanna
- Pharmaceutical Candidate Optimization, Biocon Bristol-Myers Squibb R&D Centre, Syngene International Limited, Bangalore 560099, India
| | - Hemant Bhutani
- Pharmaceutical Development, Biocon Bristol-Myers Squibb R&D Centre, Bristol-Myers Squibb India Private Limited, Bangalore 560099, India
| | - Atish T Paul
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Pilani Campus, Rajasthan 333031, India
| | - Raj Kumar
- Laboratory for Drug Design and Synthesis, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda 151001, India
| |
Collapse
|
25
|
Ward AI, Olmo F, Atherton RL, Taylor MC, Kelly JM. Trypanosoma cruzi amastigotes that persist in the colon during chronic stage murine infections have a reduced replication rate. Open Biol 2020; 10:200261. [PMID: 33321060 PMCID: PMC7776577 DOI: 10.1098/rsob.200261] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Chronic Trypanosoma cruzi infections are typically lifelong, with small numbers of parasites surviving in restricted tissue sites, which include the gastrointestinal tract. There is considerable debate about the replicative status of these persistent parasites and whether there is a role for dormancy in long-term infection. Here, we investigated T. cruzi proliferation in the colon of chronically infected mice using 5-ethynyl-2′deoxyuridine incorporation into DNA to provide ‘snapshots’ of parasite replication status. Highly sensitive imaging of the extremely rare infection foci, at single-cell resolution, revealed that parasites are three times more likely to be in S-phase during the acute stage than during the chronic stage. By implication, chronic infections of the colon are associated with a reduced rate of parasite replication. Despite this, very few host cells survived infection for more than 14 days, suggesting that T. cruzi persistence continues to involve regular cycles of replication, host cell lysis and re-infection. We could find no evidence for wide-spread dormancy in parasites that persist in this tissue reservoir.
Collapse
Affiliation(s)
- Alexander I Ward
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Francisco Olmo
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Richard L Atherton
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Martin C Taylor
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - John M Kelly
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
26
|
Maldonado E, Rojas DA, Morales S, Miralles V, Solari A. Dual and Opposite Roles of Reactive Oxygen Species (ROS) in Chagas Disease: Beneficial on the Pathogen and Harmful on the Host. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8867701. [PMID: 33376582 PMCID: PMC7746463 DOI: 10.1155/2020/8867701] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/22/2020] [Accepted: 11/25/2020] [Indexed: 11/18/2022]
Abstract
Chagas disease is a neglected tropical disease, which affects an estimate of 6-7 million people worldwide. Chagas disease is caused by Trypanosoma cruzi, which is a eukaryotic flagellate unicellular organism. At the primary infection sites, these parasites are phagocytized by macrophages, which produce reactive oxygen species (ROS) in response to the infection with T. cruzi. The ROS produce damage to the host tissues; however, macrophage-produced ROS is also used as a signal for T. cruzi proliferation. At the later stages of infection, mitochondrial ROS is produced by the infected cardiomyocytes that contribute to the oxidative damage, which persists at the chronic stage of the disease. The oxidative damage leads to a functional impairment of the heart. In this review article, we will discuss the mechanisms by which T. cruzi is able to deal with the oxidative stress and how this helps the parasite growth at the acute phase of infection and how the oxidative stress affects the cardiomyopathy at the chronic stage of the Chagas disease. We will describe the mechanisms used by the parasite to deal with ROS and reactive nitrogen species (RNS) through the trypanothione and the mechanisms used to repair the damaged DNA. Also, a description of the events produced by ROS at the acute and chronic stages of the disease is presented. Lastly, we discuss the benefits of ROS for T. cruzi growth and proliferation and the possible mechanisms involved in this phenomenon. Hypothesis is put forward to explain the molecular mechanisms by which ROS triggers parasite growth and proliferation and how ROS is able to produce a long persisting damage on cardiomyocytes even in the absence of the parasite.
Collapse
Affiliation(s)
- Edio Maldonado
- Programa Biología Celular y Molecular, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Diego A. Rojas
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Sebastian Morales
- Programa Biología Celular y Molecular, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Vicente Miralles
- Departamento de Bioquímica y Biología Molecular, Universidad de Valencia, Valencia, Spain
| | - Aldo Solari
- Programa Biología Celular y Molecular, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
27
|
Losada Galván I, Alonso-Padilla J, Cortés-Serra N, Alonso-Vega C, Gascón J, Pinazo MJ. Benznidazole for the treatment of Chagas disease. Expert Rev Anti Infect Ther 2020; 19:547-556. [PMID: 33043726 DOI: 10.1080/14787210.2021.1834849] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Chagas disease affects 6-7 million people, mainly in the Americas, and benznidazole is one of the two therapeutic options available. Trypanocide treatment aims to eliminate the parasite from the body to prevent the establishment or progression of visceral damage, mainly cardiac and/or digestive. Remarkably, it helps interrupt vertical transmission when administered to women of childbearing age. AREAS COVERED We discuss the basic and scarce data regarding chemical, pharmacokinetic, and pharmacodynamic structure. We also collect the most important data from previous phase II and III studies, as well as studies currently underway and upcoming. We reflect on the main indications for treatment and its challenges, such as the profile of adverse effects in adults, the pharmaceutical formulations, the search for reliable biomarkers, as well as regulatory aspects and access barriers. Alternative strategies such as shorter regimens, lower doses, and fixed doses are currently being evaluated to improve access and the safety profile of this treatment. EXPERT OPINION Benznidazole is likely to continue to be the drug of choice for Chagas disease in the coming years. However, it would probably be with a different treatment scheme.
Collapse
Affiliation(s)
| | | | | | | | - Joaquim Gascón
- ISGlobal, Hospital Clínic - Universitat De Barcelona, Barcelona, Spain
| | | |
Collapse
|
28
|
de Moura Ferraz LR, Tabosa AÉGA, da Silva Nascimento DDS, Ferreira AS, de Albuquerque Wanderley Sales V, Silva JYR, Júnior SA, Rolim LA, de Souza Pereira JJ, Rolim-Neto PJ. ZIF-8 as a promising drug delivery system for benznidazole: development, characterization, in vitro dialysis release and cytotoxicity. Sci Rep 2020; 10:16815. [PMID: 33033328 PMCID: PMC7545170 DOI: 10.1038/s41598-020-73848-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/24/2020] [Indexed: 12/20/2022] Open
Abstract
Chagas disease (CD), caused by the flagellate protozoan Trypanosoma cruzi, is one of the major public health problems in developing countries. Benznidazole (BNZ) is the only drug available for CD treatment in most countries, however, it presents high toxicity and low bioavailability. To address these problems this study used Zeolitic Imidazolate Framework-8 (ZIF-8), which has garnered considerable attention due to its potential applications, enabling the controlled delivery of drugs. The present work developed and characterized a BNZ@ZIF-8 system, and the modulation of BNZ release from the ZIF-8 framework was evaluated through the in vitro dialysis release method under sink conditions at different pH values. Moreover, the in vitro evaluation of cell viability and cytotoxicity by MTT assay were also performed. The dissolution studies corroborated that a pH sensitive Drug Delivery System capable of vectorizing the release of BNZ was developed, may leading to the improvement in the bioavailability of BNZ. The MTT assay showed that no statistically significant toxic effects occurred in the developed system, nor significant effects on cell viability.
Collapse
Affiliation(s)
- Leslie Raphael de Moura Ferraz
- Laboratório de Tecnologia Dos Medicamentos, Department of Pharmaceutical Sciences, Federal University of Pernambuco, Av. Prof. Arthur de Sá, s/n, Cidade Universitária, Recife, PE, 50740-521, Brazil.
| | - Alinne Élida Gonçalves Alves Tabosa
- Laboratório de Tecnologia Dos Medicamentos, Department of Pharmaceutical Sciences, Federal University of Pernambuco, Av. Prof. Arthur de Sá, s/n, Cidade Universitária, Recife, PE, 50740-521, Brazil
| | - Débora Dolores Souza da Silva Nascimento
- Laboratório de Tecnologia Dos Medicamentos, Department of Pharmaceutical Sciences, Federal University of Pernambuco, Av. Prof. Arthur de Sá, s/n, Cidade Universitária, Recife, PE, 50740-521, Brazil
| | - Aline Silva Ferreira
- Laboratório de Tecnologia Dos Medicamentos, Department of Pharmaceutical Sciences, Federal University of Pernambuco, Av. Prof. Arthur de Sá, s/n, Cidade Universitária, Recife, PE, 50740-521, Brazil
| | - Victor de Albuquerque Wanderley Sales
- Laboratório de Tecnologia Dos Medicamentos, Department of Pharmaceutical Sciences, Federal University of Pernambuco, Av. Prof. Arthur de Sá, s/n, Cidade Universitária, Recife, PE, 50740-521, Brazil
| | - José Yago Rodrigues Silva
- Laboratório de Terras Raras, Departamento de Química Fundamental, Federal University of Pernambuco, Av. Jornalista Aníbal Fernandes, s/n - Cidade Universitária, Recife, PE, 50740-560, Brazil
| | - Severino Alves Júnior
- Laboratório de Terras Raras, Departamento de Química Fundamental, Federal University of Pernambuco, Av. Jornalista Aníbal Fernandes, s/n - Cidade Universitária, Recife, PE, 50740-560, Brazil
| | - Larissa Araújo Rolim
- Central de Análise de Fármacos, Medicamentos e Alimentos, Federal University of Vale Do São Francisco, Av. José de Sá Maniçoba, s/n, Centro, Petrolina, PE, 56304-917, Brazil
| | | | - Pedro José Rolim-Neto
- Laboratório de Tecnologia Dos Medicamentos, Department of Pharmaceutical Sciences, Federal University of Pernambuco, Av. Prof. Arthur de Sá, s/n, Cidade Universitária, Recife, PE, 50740-521, Brazil
| |
Collapse
|
29
|
Andrade LDC, Santi AMM, Alves CL, Ferreira WRR, de Assis AV, Oliveira E, Machado CR, Murta SMF. The heterologous expression of Escherichia coli MutT enzyme is involved in the protection against oxidative stress in Leishmania braziliensis. Mem Inst Oswaldo Cruz 2020; 115:e190469. [PMID: 32638832 PMCID: PMC7337112 DOI: 10.1590/0074-02760190469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 06/02/2020] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Oxidative stress is responsible for generating DNA lesions and the 8-oxoguanine (8-oxoG) is the most commonly lesion found in DNA damage. When this base is incorporated during DNA replication, it could generate double-strand DNA breaks and cellular death. MutT enzyme hydrolyzes the 8-oxoG from the nucleotide pool, preventing its incorporation during DNA replication. OBJECTIVES To investigate the importance of 8-oxoG in Leishmania infantum and L. braziliensis, in this study we analysed the impact of heterologous expression of Escherichia coli MutT (EcMutT) enzyme in drug-resistance phenotype and defense against oxidative stress. METHODS Comparative analysis of L. braziliensis and L. infantum H2O2 tolerance and cell cycle profile were performed. Lines of L. braziliensis and L. infantum expressing EcMutT were generated and evaluated using susceptibility tests to H2O2 and SbIII, cell cycle analysis, γH2A western blotting, and BrdU native detection assay. FINDINGS Comparative analysis of tolerance to oxidative stress generated by H2O2 showed that L. infantum is more tolerant to exogenous H2O2 than L. braziliensis. In addition, cell cycle analysis showed that L. infantum, after treatment with H2O2, remains in G1 phase, returning to its normal growth rate after 72 h. In contrast, after treatment with H2O2, L. braziliensis parasites continue to move to the next stages of the cell cycle. Expression of the E. coli MutT gene in L. braziliensis and L. infantum does not interfere in parasite growth or in susceptibility to SbIII. Interestingly, we observed that L. braziliensis EcMutT-expressing clones were more tolerant to H2O2 treatment, presented lower activation of γH2A, a biomarker of genotoxic stress, and lower replication stress than its parental non-transfected parasites. In contrast, the EcMutT is not involved in protection against oxidative stress generated by H2O2 in L. infantum. MAIN CONCLUSIONS Our results showed that 8-oxoG clearance in L. braziliensis is important to avoid misincorporation during DNA replication after oxidative stress generated by H2O2.
Collapse
Affiliation(s)
| | | | - Ceres Luciana Alves
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Wesley Roger Rodrigues Ferreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Antônio Vinícius de Assis
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Edward Oliveira
- Instituto René Rachou, Fundação Oswaldo Cruz-Fiocruz, Belo Horizonte, MG, Brazil
| | - Carlos Renato Machado
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | |
Collapse
|
30
|
Francisco AF, Jayawardhana S, Olmo F, Lewis MD, Wilkinson SR, Taylor MC, Kelly JM. Challenges in Chagas Disease Drug Development. Molecules 2020; 25:E2799. [PMID: 32560454 PMCID: PMC7355550 DOI: 10.3390/molecules25122799] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 02/08/2023] Open
Abstract
The protozoan parasite Trypanosoma cruzi causes Chagas disease, an important public health problem throughout Latin America. Current therapeutic options are characterised by limited efficacy, long treatment regimens and frequent toxic side-effects. Advances in this area have been compromised by gaps in our knowledge of disease pathogenesis, parasite biology and drug activity. Nevertheless, several factors have come together to create a more optimistic scenario. Drug-based research has become more systematic, with increased collaborations between the academic and commercial sectors, often within the framework of not-for-profit consortia. High-throughput screening of compound libraries is being widely applied, and new technical advances are helping to streamline the drug development pipeline. In addition, drug repurposing and optimisation of current treatment regimens, informed by laboratory research, are providing a basis for new clinical trials. Here, we will provide an overview of the current status of Chagas disease drug development, highlight those areas where progress can be expected, and describe how fundamental research is helping to underpin the process.
Collapse
Affiliation(s)
- Amanda F. Francisco
- Department of Infection Biology, London School of Hygiene and Tropical Medicine Keppel Street, London WC1E 7HT, UK; (A.F.F.); (S.J.); (F.O.); (M.D.L.); (M.C.T.)
| | - Shiromani Jayawardhana
- Department of Infection Biology, London School of Hygiene and Tropical Medicine Keppel Street, London WC1E 7HT, UK; (A.F.F.); (S.J.); (F.O.); (M.D.L.); (M.C.T.)
| | - Francisco Olmo
- Department of Infection Biology, London School of Hygiene and Tropical Medicine Keppel Street, London WC1E 7HT, UK; (A.F.F.); (S.J.); (F.O.); (M.D.L.); (M.C.T.)
| | - Michael D. Lewis
- Department of Infection Biology, London School of Hygiene and Tropical Medicine Keppel Street, London WC1E 7HT, UK; (A.F.F.); (S.J.); (F.O.); (M.D.L.); (M.C.T.)
| | - Shane R. Wilkinson
- School of Biological and Chemical Sciences, Queen Mary University of London Mile End Road, London E1 4NS, UK;
| | - Martin C. Taylor
- Department of Infection Biology, London School of Hygiene and Tropical Medicine Keppel Street, London WC1E 7HT, UK; (A.F.F.); (S.J.); (F.O.); (M.D.L.); (M.C.T.)
| | - John M. Kelly
- Department of Infection Biology, London School of Hygiene and Tropical Medicine Keppel Street, London WC1E 7HT, UK; (A.F.F.); (S.J.); (F.O.); (M.D.L.); (M.C.T.)
| |
Collapse
|
31
|
Rose E, Carvalho JL, Hecht M. Mechanisms of DNA repair in Trypanosoma cruzi: What do we know so far? DNA Repair (Amst) 2020; 91-92:102873. [PMID: 32505694 DOI: 10.1016/j.dnarep.2020.102873] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/27/2020] [Accepted: 05/22/2020] [Indexed: 12/13/2022]
Abstract
Trypanosoma cruzi is the etiological agent of Chagas Disease, which affects 6-7 million people worldwide. Since the early stages of infection and throughout its life cycle, the parasite is exposed to several genotoxic agents. Furthermore, DNA damage is also part of the mechanism of action of at least a few trypanocidal drugs, including Benznidazole. Thus, it is paramount for the parasite to count on an efficient DNA repair machinery to guarantee genome integrity and survival. The present work provides an up-to-date review of both the conserved and peculiar DNA repair mechanisms described in T. cruzi against oxidative stress, ultraviolet and ionizing radiation, DNA adduct-inducing agents, and Benznidazole. The comprehension of the DNA repair mechanisms of the parasite may shed light on the parasite evolution and possibly pave the way for the development of novel and more effective trypanocidal drugs.
Collapse
Affiliation(s)
- Ester Rose
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, Brasília, Brazil.
| | - Juliana Lott Carvalho
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, Brasília, Brazil; Genomic Sciences and Biotechnology Program, Catholic University of Brasília, Brasília, Brazil
| | - Mariana Hecht
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasília, Brasília, Brazil
| |
Collapse
|
32
|
Pardali V, Giannakopoulou E, Balourdas DI, Myrianthopoulos V, Taylor MC, Šekutor M, Mlinarić-Majerski K, Kelly JM, Zoidis G. Lipophilic Guanylhydrazone Analogues as Promising Trypanocidal Agents: An Extended SAR Study. Curr Pharm Des 2020; 26:838-866. [DOI: 10.2174/1381612826666200210150127] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 11/19/2019] [Indexed: 12/18/2022]
Abstract
In this report, we extend the SAR analysis of a number of lipophilic guanylhydrazone analogues with
respect to in vitro growth inhibition of Trypanosoma brucei and Trypanosoma cruzi. Sleeping sickness and Chagas
disease, caused by the tropical parasites T. brucei and T. cruzi, constitute a significant socioeconomic burden
in low-income countries of sub-Saharan Africa and Latin America, respectively. Drug development is underfunded.
Moreover, current treatments are outdated and difficult to administer, while drug resistance is an emerging
concern. The synthesis of adamantane-based compounds that have potential as antitrypanosomal agents is
extensively reviewed. The critical role of the adamantane ring was further investigated by synthesizing and testing
a number of novel lipophilic guanylhydrazones. The introduction of hydrophobic bulky substituents onto the
adamantane ring generated the most active analogues, illustrating the synergistic effect of the lipophilic character
of the C1 side chain and guanylhydrazone moiety on trypanocidal activity. The n-decyl C1-substituted compound
G8 proved to be the most potent adamantane derivative against T. brucei with activity in the nanomolar range
(EC50=90 nM). Molecular simulations were also performed to better understand the structure-activity relationships
between the studied guanylhydrazone analogues and their potential enzyme target.
Collapse
Affiliation(s)
- Vasiliki Pardali
- School of Health Sciences, Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, GR-15771 Athens, Greece
| | - Erofili Giannakopoulou
- School of Health Sciences, Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, GR-15771 Athens, Greece
| | - Dimitrios-Ilias Balourdas
- School of Health Sciences, Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, GR-15771 Athens, Greece
| | - Vassilios Myrianthopoulos
- School of Health Sciences, Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, GR-15771 Athens, Greece
| | - Martin C. Taylor
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Marina Šekutor
- Department of Organic Chemistry and Biochemistry, Ruder Boskovic Institute, Bijenicka cesta 54, 10 000 Zagreb, Croatia
| | - Kata Mlinarić-Majerski
- Department of Organic Chemistry and Biochemistry, Ruder Boskovic Institute, Bijenicka cesta 54, 10 000 Zagreb, Croatia
| | - John M. Kelly
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Grigoris Zoidis
- School of Health Sciences, Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, GR-15771 Athens, Greece
| |
Collapse
|
33
|
Repolês BM, Machado CR, Florentino PTV. DNA lesions and repair in trypanosomatids infection. Genet Mol Biol 2020; 43:e20190163. [PMID: 32236391 PMCID: PMC7197992 DOI: 10.1590/1678-4685-gmb-2019-0163] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 10/21/2019] [Indexed: 12/22/2022] Open
Abstract
Pathological processes such as bacterial, viral and parasitic infections can generate a plethora of responses such as, but not restricted to, oxidative stress that can be harmful to the host and the pathogen. This stress occurs when there is an imbalance between reactive oxygen species produced and antioxidant factors produced in response to the infection. This imbalance can lead to DNA lesions in both infected cells as well as in the pathogen. The effects of the host response on the parasite lead to several kinds of DNA damage, causing alterations in the parasite's metabolism; the reaction and sensitivity of the parasite to these responses are related to the DNA metabolism and life cycle of each parasite. The present review will discuss the survival strategies developed by host cells and Trypanosoma cruzi, focusing on the DNA repair mechanisms of these organisms throughout infection including the relationship between DNA damage, stress response features, and the unique characteristics of these diseases.
Collapse
Affiliation(s)
- Bruno M Repolês
- Universidade Federal de Minas Gerais, Departamento de Bioquímica e Imunologia, Belo Horizonte MG, Brazil
| | - Carlos Renato Machado
- Universidade Federal de Minas Gerais, Departamento de Bioquímica e Imunologia, Belo Horizonte MG, Brazil
| | | |
Collapse
|
34
|
Sykes ML, Hilko DH, Kung LI, Poulsen SA, Avery VM. Investigation of pyrimidine nucleoside analogues as chemical probes to assess compound effects on the proliferation of Trypanosoma cruzi intracellular parasites. PLoS Negl Trop Dis 2020; 14:e0008068. [PMID: 32163414 PMCID: PMC7112222 DOI: 10.1371/journal.pntd.0008068] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 04/01/2020] [Accepted: 01/15/2020] [Indexed: 02/05/2023] Open
Abstract
Trypanosoma cruzi parasites utilise de novo pyrimidine biosynthesis to produce DNA and survive within mammalian host cells. This pathway can be hijacked to assess the replication of intracellular parasites with the exogenous addition of a DNA specific probe. To identify suitable probe compounds for this application, a collection of pyrimidine nucleoside analogues was assessed for incorporation into T. cruzi intracellular amastigote DNA using image-based technology and script-based analysis. Associated mammalian cell toxicity of these compounds was also determined against both the parasite host cells (3T3 cells) and HEK293 cells. Incorporation of 5-ethynyl-2′-deoxyuridine (EdU) into parasite DNA was the most effective of the probes tested, with minimal growth inhibition observed following either two or four hours EdU exposure. EdU was subsequently utilised as a DNA probe, followed by visualisation with click chemistry to a fluorescent azide, to assess the impact of drugs and compounds with previously demonstrated activity against T. cruzi parasites, on parasite replication. The inhibitory profiles of these molecules highlight the benefit of this approach for identifying surviving parasites post-treatment in vitro and classifying compounds as either fast or slow-acting. F-ara-EdU resulted in <50% activity observed against T. cruzi amastigotes following 48 hours incubation, at 73 μM. Collectively, this supports the further development of pyrimidine nucleosides as chemical probes to investigate replication of the parasite T. cruzi. Chagas disease occurs within 21 countries in the Americas, causes over 10, 000 deaths per year and a further 25 million people are at risk of being infected. The cause of Chagas disease is Trypanosoma cruzi, a single celled protozoan parasite, which enters the bloodstream of a host by the bite of a “kissing bug”. In advanced disease stages, the parasite hides in heart and gut tissue and is difficult to treat. Identifying the replicative ability of these parasites is important to understanding Chagas disease progression and the effectiveness of compounds and drugs for treatment. By testing a panel of nucleoside analogues that may incorporate into DNA during synthesis, we developed an image-based method with a fluorescently-labelled DNA probe to identify replicating parasites. This method has effectively shown that drugs used to treat the parasite are able to clear intracellular infection, whilst a compound that was not efficacious in clinical trials leaves replicating T. cruzi behind. This methodology can be used to understand the action of further compounds and supports the identification of new, less toxic probes to assess intracellular parasite replication.
Collapse
Affiliation(s)
- Melissa Louise Sykes
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia
| | - David Hugh Hilko
- Chemical Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia
| | - Livia Isabella Kung
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia.,Institute of Molecular Health Sciences, ETH Zurich, Switzerland
| | - Sally-Ann Poulsen
- Chemical Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia
| | - Vicky Marie Avery
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia
| |
Collapse
|
35
|
Petravicius PO, Costa-Martins AG, Silva MN, Reis-Cunha JL, Bartholomeu DC, Teixeira MM, Zingales B. Mapping benznidazole resistance in trypanosomatids and exploring evolutionary histories of nitroreductases and ABCG transporter protein sequences. Acta Trop 2019; 200:105161. [PMID: 31494121 DOI: 10.1016/j.actatropica.2019.105161] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/22/2019] [Accepted: 09/03/2019] [Indexed: 12/24/2022]
Abstract
The nitro-heterocyclic compound benznidazole (BZ) is the first-line drug for the treatment of Chagas disease, caused by the protozoan Trypanosoma cruzi. However, therapeutic failures are common for reasons that include the influences of parasite and host genetics, the effects of toxicity on adherence to treatment, and difficulties in demonstrating parasitological cure. To obtain information on the origin of the resistance to BZ and eliminate from the scenery the participation of the host, initially we mapped the susceptibility to the drug in thirteen species of seven genera of the family Trypanosomatidae. We verified that all Trypanosoma species are sensitive to low concentrations of the drug (IC50 2.7 to 25 µM) while Non-Trypanosoma species are highly resistant to these concentrations. The two groups of parasites correspond to the major phylogenetic lineages of trypanosomatids. Next, we searched in the trypanosomatid genome databases homologs of two type-I nitroreductases (NTR-1 and OYE) and an ABC transporter (ABCG1) that have been associated with BZ resistance in T. cruzi. The predicted proteins were characterized regarding domains and used for phylogenetic analyses. Homologous NTR-1 genes were found in all trypanosomatids investigated and the structural characteristics of the enzyme suggest that it may be functional. OYE genes were absent in BZ-sensitive African trypanosomes, which excludes the participation of this enzyme in BZ bio-activation. Two copies of ABCG1 genes were observed in most BZ resistant species, while Trypanosoma species exhibit only one copy per haploid genome. Functional studies are required to verify the involvement of these genes in BZ resistance. In addition, since multiple mechanisms can contribute to BZ susceptibility, our study poses a range of organisms highly resistant to BZ in which these aspects can be investigated. Preliminary studies on BZ uptake indicate marked differences between BZ-sensitive and BZ-resistant species.
Collapse
|
36
|
Screening diagnostic candidates from Leishmania infantum proteins for human visceral leishmaniasis using an immunoproteomics approach. Parasitology 2019; 146:1467-1476. [PMID: 31142384 DOI: 10.1017/s0031182019000714] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
There is no suitable vaccine against human visceral leishmaniasis (VL) and available drugs are toxic and/or present high cost. In this context, diagnostic tools should be improved for clinical management and epidemiological evaluation of disease. However, the variable sensitivity and/or specificity of the used antigens are limitations, showing the necessity to identify new molecules to be tested in a more sensitive and specific serology. In the present study, an immunoproteomics approach was performed in Leishmania infantum promastigotes and amastigotes employing sera samples from VL patients. Aiming to avoid undesired cross-reactivity in the serological assays, sera from Chagas disease patients and healthy subjects living in the endemic region of disease were also used in immunoblottings. The most reactive spots for VL samples were selected, and 29 and 21 proteins were identified in the promastigote and amastigote extracts, respectively. Two of them, endonuclease III and GTP-binding protein, were cloned, expressed, purified and tested in ELISA experiments against a large serological panel, and results showed high sensitivity and specificity values for the diagnosis of disease. In conclusion, the identified proteins could be considered in future studies as candidate antigens for the serodiagnosis of human VL.
Collapse
|
37
|
Crespillo-Andújar C, Venanzi-Rullo E, López-Vélez R, Monge-Maillo B, Norman F, López-Polín A, Pérez-Molina JA. Safety Profile of Benznidazole in the Treatment of Chronic Chagas Disease: Experience of a Referral Centre and Systematic Literature Review with Meta-Analysis. Drug Saf 2019; 41:1035-1048. [PMID: 30006773 DOI: 10.1007/s40264-018-0696-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Benznidazole is the preferred drug for treatment of Chagas disease. However, it is toxic and of limited value in chronic infection. OBJECTIVE We aimed to estimate the rates of and factors related to adverse reactions (ARs) to benznidazole and treatment discontinuations (TDs). METHODS A meta-analysis was performed using an electronic search of the published literature with no language restrictions until June 2017. Prospective studies were included of chronically infected patients in which at least one treatment arm included benznidazole. Data were added from a prospective cohort of patients with Chagas disease at our centre (January 2007-June 2017). Weighted rates of ARs and TDs were estimated, and potentially related factors were analysed. RESULTS Some 413 studies were found, from which we chose 42 (nine clinical trials and 33 observational studies, including ours), comprising data for 7822 patients. The weighted rate of ARs to benznidazole was 44.1% (95% confidence interval [CI] 37.2-51.2). ARs were more frequent in adults than in children (51.6 vs. 24.5%), with the most common being skin reactions (34%), gastrointestinal complaints (12.6%) and neurological symptoms (11.5%). Grade 4 ARs were recorded in 3% of cases. The weighted rate of TDs was 11.4% (95% CI 8.5-14.5); TDs were more frequent in adults than in children (14.2 vs. 3.8%). In our cohort, only female sex was related to an increased rate of ARs but not to TDs. CONCLUSION Benznidazole had a poor tolerability profile, with a high incidence of TDs, especially in adult patients and women. Optimised dosing schedules and/or new drugs are urgently needed.
Collapse
Affiliation(s)
- Clara Crespillo-Andújar
- National Referral Unit for Tropical and Travel Medicine, Department of Internal Medicine, Hospital Universitario La Paz-Carlos III, IdiPAZ, Madrid, Spain
| | - Emmanuele Venanzi-Rullo
- Division of Infectious Diseases, University of Messina, Polyclinic "G. Martino", Messina, Italy
| | - Rogelio López-Vélez
- National Referral Centre for Tropical Diseases, Infectious Diseases Department, Hospital Universitario Ramón y Cajal, IRYCIS, Carretera de Colmenar Km 9, 1, 28034, Madrid, Spain
| | - Begoña Monge-Maillo
- National Referral Centre for Tropical Diseases, Infectious Diseases Department, Hospital Universitario Ramón y Cajal, IRYCIS, Carretera de Colmenar Km 9, 1, 28034, Madrid, Spain
| | - Francesca Norman
- National Referral Centre for Tropical Diseases, Infectious Diseases Department, Hospital Universitario Ramón y Cajal, IRYCIS, Carretera de Colmenar Km 9, 1, 28034, Madrid, Spain
| | - Ana López-Polín
- National Referral Centre for Tropical Diseases, Infectious Diseases Department, Hospital Universitario Ramón y Cajal, IRYCIS, Carretera de Colmenar Km 9, 1, 28034, Madrid, Spain
| | - José A Pérez-Molina
- National Referral Centre for Tropical Diseases, Infectious Diseases Department, Hospital Universitario Ramón y Cajal, IRYCIS, Carretera de Colmenar Km 9, 1, 28034, Madrid, Spain.
| |
Collapse
|
38
|
Flick AC, Leverett CA, Ding HX, McInturff E, Fink SJ, Helal CJ, O’Donnell CJ. Synthetic Approaches to the New Drugs Approved During 2017. J Med Chem 2019; 62:7340-7382. [DOI: 10.1021/acs.jmedchem.9b00196] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Andrew C. Flick
- Seattle Genetics, Inc. 21823 30th Drive SE, Bothell, Washington 98021, United States
| | - Carolyn A. Leverett
- Groton Laboratories, Pfizer Worldwide Research and Development, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Hong X. Ding
- Pharmacodia (Beijing) Co., Ltd., Beijing, 100085, China
| | - Emma McInturff
- Groton Laboratories, Pfizer Worldwide Research and Development, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Sarah J. Fink
- BioDuro, 11011 Torreyana Road, San Diego, California 92121, United States
| | - Christopher J. Helal
- Groton Laboratories, Pfizer Worldwide Research and Development, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Christopher J. O’Donnell
- Groton Laboratories, Pfizer Worldwide Research and Development, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
39
|
Piacenza L, Trujillo M, Radi R. Reactive species and pathogen antioxidant networks during phagocytosis. J Exp Med 2019; 216:501-516. [PMID: 30792185 PMCID: PMC6400530 DOI: 10.1084/jem.20181886] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/04/2019] [Accepted: 02/04/2019] [Indexed: 11/23/2022] Open
Abstract
The generation of phagosomal cytotoxic reactive species (i.e., free radicals and oxidants) by activated macrophages and neutrophils is a crucial process for the control of intracellular pathogens. The chemical nature of these species, the reactions they are involved in, and the subsequent effects are multifaceted and depend on several host- and pathogen-derived factors that influence their production rates and catabolism inside the phagosome. Pathogens rely on an intricate and synergistic antioxidant armamentarium that ensures their own survival by detoxifying reactive species. In this review, we discuss the generation, kinetics, and toxicity of reactive species generated in phagocytes, with a focus on the response of macrophages to internalized pathogens and concentrating on Mycobacterium tuberculosis and Trypanosoma cruzi as examples of bacterial and parasitic infection, respectively. The ability of pathogens to deal with host-derived reactive species largely depends on the competence of their antioxidant networks at the onset of invasion, which in turn can tilt the balance toward pathogen survival, proliferation, and virulence over redox-dependent control of infection.
Collapse
Affiliation(s)
- Lucía Piacenza
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Madia Trujillo
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Rafael Radi
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
40
|
The in vivo and in vitro roles of Trypanosoma cruzi Rad51 in the repair of DNA double strand breaks and oxidative lesions. PLoS Negl Trop Dis 2018; 12:e0006875. [PMID: 30422982 PMCID: PMC6258567 DOI: 10.1371/journal.pntd.0006875] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 11/27/2018] [Accepted: 09/26/2018] [Indexed: 11/22/2022] Open
Abstract
In Trypanosoma cruzi, the etiologic agent of Chagas disease, Rad51 (TcRad51) is a central enzyme for homologous recombination. Here we describe the different roles of TcRad51 in DNA repair. Epimastigotes of T. cruzi overexpressing TcRAD51 presented abundant TcRad51-labeled foci before gamma irradiation treatment, and a faster growth recovery when compared to single-knockout epimastigotes for RAD51. Overexpression of RAD51 also promoted increased resistance against hydrogen peroxide treatment, while the single-knockout epimastigotes for RAD51 exhibited increased sensitivity to this oxidant agent, which indicates a role for this gene in the repair of DNA oxidative lesions. In contrast, TcRad51 was not involved in the repair of crosslink lesions promoted by UV light and cisplatin treatment. Also, RAD51 single-knockout epimastigotes showed a similar growth rate to that exhibited by wild-type ones after treatment with hydroxyurea, but an increased sensitivity to methyl methane sulfonate. Besides its role in epimastigotes, TcRad51 is also important during mammalian infection, as shown by increased detection of T. cruzi cells overexpressing RAD51, and decreased detection of single-knockout cells for RAD51, in both fibroblasts and macrophages infected with amastigotes. Besides that, RAD51-overexpressing parasites infecting mice also presented increased infectivity and higher resistance against benznidazole. We thus show that TcRad51 is involved in the repair of DNA double strands breaks and oxidative lesions in two different T. cruzi developmental stages, possibly playing an important role in the infectivity of this parasite. Trypanosoma cruzi is the causative agent of Chagas disease, a tropical neglected illness that affects 6 million people worldwide, mostly in Latin America. Our research group focuses on the elucidation of DNA repair and metabolism of T. cruzi, and on the possible implications of those mechanisms in both parasite genetic diversity and Chagas disease development. In this work, we investigated the involvement of homologous recombination in the oxidation-induced damage response, and in DNA repair, in T. cruzi cells after gamma radiation exposure. We also examined whether TcRad51 –a key protein for homologous recombination–could be an important factor for T. cruzi’s infectivity. We generated genetically-modified T. cruzi cells for RAD51 and studied the in vitro and in vivo infectivity of these mutant cells. Our results indicate that TcRad51 is a key protein involved in the repair of oxidative and DNA double-strand breaks lesions in two crucial developmental forms of T. cruzi.
Collapse
|
41
|
Moreno ÉM, Leal SM, Stashenko EE, García LT. Induction of programmed cell death in Trypanosoma cruzi by Lippia alba essential oils and their major and synergistic terpenes (citral, limonene and caryophyllene oxide). BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 18:225. [PMID: 30053848 PMCID: PMC6062979 DOI: 10.1186/s12906-018-2293-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 07/18/2018] [Indexed: 11/10/2022]
Abstract
Background Chagas Disease caused by Trypanosoma cruzi infection, is one of the most important neglected tropical diseases (NTD), without an effective therapy for the successful parasite eradication or for the blocking of the disease’s progression, in its advanced stages. Due to their low toxicity, wide pharmacologic spectrum, and potential synergies, medicinal plants as Lippia alba, offer a promising reserve of bioactive molecules. The principal goal of this work is to characterize the inhibitory properties and cellular effects of the Citral and Carvone L. alba chemotype essential oils (EOs) and their main bioactive terpenes (and the synergies among them) on T. cruzi forms. Methods Twelve L. alba EOs, produced under diverse environmental conditions, were extracted by microwave assisted hydrodistillation, and chemically characterized using gas chromatography coupled mass spectrometry. Trypanocidal activity and cytotoxicity were determined for each oil, and their major compounds, on epimastigotes (Epi), trypomastigotes (Tryp), amastigotes (Amas), and Vero cells. Pharmacologic interactions were defined by a matrix of combinations among the most trypanocidal terpenes (limonene, carvone; citral and caryophyllene oxide). The treated cell phenotype was assessed by fluorescent and optic microscopy, flow cytometry, and DNA electrophoresis assays. Results The L. alba EOs displayed significant differences in their chemical composition and trypanocidal performance (p = 0.0001). Citral chemotype oils were more trypanocidal than Carvone EOs, with Inhibitory Concentration 50 (IC50) of 14 ± 1.5 μg/mL, 22 ± 1.4 μg/mL and 74 ± 4.4 μg/mL, on Epi, Tryp and Amas, respectively. Limonene exhibited synergistic interaction with citral, caryophyllene oxide and Benznidazole (decreasing by 17 times its IC50) and was the most effective and selective treatment. The cellular analysis suggested that these oils or their bioactive terpenes (citral, caryophyllene oxide and limonene) could be inducing T. cruzi cell death by an apoptotic-like mechanism. Conclusions EOs extracted from L. alba Citral chemotype demonstrated significant trypanocidal activity on the three forms of T. cruzi studied, and their composition and trypanocidal performance were influenced by production parameters. Citral, caryophyllene oxide, and limonene showed a possible induction of an apoptotic-like phenotype. The best selective anti-T. cruzi activity was achieved by limonene, the effects of which were also synergic with citral, caryophyllene oxide and benznidazole.
Collapse
|
42
|
Leite DI, Fontes FDV, Bastos MM, Hoelz LVB, Bianco MDCAD, de Oliveira AP, da Silva PB, da Silva CF, Batista DDGJ, da Gama ANS, Peres RB, Villar JDF, Soeiro MDNC, Boechat N. New 1,2,3-triazole-based analogues of benznidazole for use against Trypanosoma cruzi infection: In vitro and in vivo evaluations. Chem Biol Drug Des 2018; 92:1670-1682. [PMID: 29745048 DOI: 10.1111/cbdd.13333] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/09/2018] [Accepted: 04/21/2018] [Indexed: 12/18/2022]
Abstract
Chagas disease has spread throughout the world mainly because of the migration of infected individuals. In Brazil, only benznidazole (Bnz) is used; however, it is toxic and not active in the chronic phase, and cases of resistance are described. This work aimed at the synthesis and the trypanocidal evaluation in vitro and in vivo of six new Bnz analogues (3-8). They were designed by exploring the bioisosteric substitution between the amide group contained in Bnz and the 1,2,3-triazole ring. All the compounds were synthesized in good yields. With the exception of compound 7, the in vitro biological evaluation shows that all Bnz analogues were active against the amastigote form, whereas only compounds 3, 4, 5, and 8 were active against trypomastigote. Compounds 4 and 5 showed the most promising activities in vitro against the form of trypomastigote, being more active than Bnz. In vivo evaluation of compounds, 3-8 showed lower potency and higher toxicity than Bnz. Although the 1,2,3-triazole ring has been described in the literature as an amide bioisostere, its substitution here has reduced the activity of the compounds and made them more toxic. Thus, further molecular optimization could provide novel therapeutic agents for Chagas' disease.
Collapse
Affiliation(s)
- Débora Inácio Leite
- Departamento de Sintese de Farmacos, Instituto de Tecnologia em Farmacos, Farmanguinhos - Fiocruz, Fundacao Oswaldo Cruz, Rio de Janeiro, Brazil.,Programa de Pos-Graduação em Farmacologia e Quimica Medicinal do Instituto de Ciencias Biomedicas - ICB- UFRJ, Centro de Ciências da Saúde - CCS, Rio de Janeiro, Brasil
| | - Fábio de Vasconcellos Fontes
- Departamento de Sintese de Farmacos, Instituto de Tecnologia em Farmacos, Farmanguinhos - Fiocruz, Fundacao Oswaldo Cruz, Rio de Janeiro, Brazil.,Departamento de Ciencia e Tecnologia, Praça General Tiburcio, Instituto Militar de Engenharia, Rio de Janeiro, Brasil
| | - Monica Macedo Bastos
- Departamento de Sintese de Farmacos, Instituto de Tecnologia em Farmacos, Farmanguinhos - Fiocruz, Fundacao Oswaldo Cruz, Rio de Janeiro, Brazil.,Programa de Pos-Graduação em Farmacologia e Quimica Medicinal do Instituto de Ciencias Biomedicas - ICB- UFRJ, Centro de Ciências da Saúde - CCS, Rio de Janeiro, Brasil
| | - Lucas Villas Boas Hoelz
- Departamento de Sintese de Farmacos, Instituto de Tecnologia em Farmacos, Farmanguinhos - Fiocruz, Fundacao Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Maria da Conceição Avelino Dias Bianco
- Departamento de Sintese de Farmacos, Instituto de Tecnologia em Farmacos, Farmanguinhos - Fiocruz, Fundacao Oswaldo Cruz, Rio de Janeiro, Brazil.,Programa de Pos-Graduação em Farmacologia e Quimica Medicinal do Instituto de Ciencias Biomedicas - ICB- UFRJ, Centro de Ciências da Saúde - CCS, Rio de Janeiro, Brasil
| | - Andressa Paula de Oliveira
- Departamento de Sintese de Farmacos, Instituto de Tecnologia em Farmacos, Farmanguinhos - Fiocruz, Fundacao Oswaldo Cruz, Rio de Janeiro, Brazil.,Programa de Pos-Graduação em Farmacologia e Quimica Medicinal do Instituto de Ciencias Biomedicas - ICB- UFRJ, Centro de Ciências da Saúde - CCS, Rio de Janeiro, Brasil.,PROBIN - Abeu - Centro Universitario UNIABEU, Belford Roxo, Rio de Janeiro, Brasil
| | | | - Cristiane França da Silva
- Laboratorio de Biologia Celular, Instituto Oswaldo Cruz - IOC, Fundacao Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Denise da Gama Jean Batista
- Laboratorio de Biologia Celular, Instituto Oswaldo Cruz - IOC, Fundacao Oswaldo Cruz, Rio de Janeiro, Brazil
| | | | - Raiza Brandão Peres
- Laboratorio de Biologia Celular, Instituto Oswaldo Cruz - IOC, Fundacao Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Jose Daniel Figueroa Villar
- Departamento de Ciencia e Tecnologia, Praça General Tiburcio, Instituto Militar de Engenharia, Rio de Janeiro, Brasil
| | | | - Nubia Boechat
- Departamento de Sintese de Farmacos, Instituto de Tecnologia em Farmacos, Farmanguinhos - Fiocruz, Fundacao Oswaldo Cruz, Rio de Janeiro, Brazil.,Programa de Pos-Graduação em Farmacologia e Quimica Medicinal do Instituto de Ciencias Biomedicas - ICB- UFRJ, Centro de Ciências da Saúde - CCS, Rio de Janeiro, Brasil
| |
Collapse
|
43
|
Crespillo-Andújar C, Calbacho Robles M, Norman FF, Pérez-Molina JA. Severe immune thrombocytopaenia in a patient taking benznidazole for chronic Chagas disease. BMJ Case Rep 2018; 2018:bcr-2017-223788. [PMID: 29588298 PMCID: PMC5878288 DOI: 10.1136/bcr-2017-223788] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2018] [Indexed: 12/27/2022] Open
Abstract
Chagas disease is a parasitic disease that mostly affects Latin American countries, but it has currently become a worldwide epidemic due to migration. Both drugs marketed for its treatment (benznidazole and nifurtimox) are associated with a high rate of adverse reactions. Benznidazole is preferred initially because of its more favourable toxicity profile and perceived greater efficacy. Hypersensitivity dermatological reactions, gastrointestinal and neurological disturbances represent the most common drug-related adverse events. General symptoms such as fever, arthralgia, myalgia or bone marrow depression (leucopaenia) are seen less frequently. We describe the case of a 33-year-old woman with chronic Chagas disease who presented with acute gingival bleeding and severe thrombocytopaenia, probably related to benznidazole treatment. Temporal association with drug initiation and recovery after treatment withdrawal were demonstrated. Clinicians should be aware of the possible association between immune thrombocytopaenia and benznidazole, even though the pathogenesis remains unclear at present.
Collapse
Affiliation(s)
- Clara Crespillo-Andújar
- National Referral Unit for Tropical and Travel Medicine, Department of Internal Medicine, Hospital Universitario La Paz-Carlos III, IdiPAZ, Madrid, Spain
| | - María Calbacho Robles
- Department of Hematology, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Francesca F Norman
- National Referral Centre for Tropical Diseases, Infectious Diseases Department, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Jose A Pérez-Molina
- National Referral Centre for Tropical Diseases, Infectious Diseases Department, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| |
Collapse
|
44
|
da Silva Lara L, Andrade-Lima L, Magalhães Calvet C, Borsoi J, Lopes Alberto Duque T, Henriques-Pons A, Souza Pereira MC, Veiga Pereira L. Trypanosoma cruzi infection of human induced pluripotent stem cell-derived cardiomyocytes: an in vitro model for drug screening for Chagas disease. Microbes Infect 2018; 20:312-316. [PMID: 29577969 DOI: 10.1016/j.micinf.2018.03.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/23/2018] [Accepted: 03/13/2018] [Indexed: 01/28/2023]
Abstract
Chagas disease, caused by Trypanosoma cruzi, is an important global public health problem which, despite partial efficacy of benznidazole (Bz) in acute phase, urgently needs an effective treatment. Cardiotoxicity is a major safety concern for conduction of more accurate preclinical drug screening platforms. Human induced pluripotent stem cells derived cardiomyocytes (hiPSC-CM) are a reliable model to study genetic and infectious cardiac alterations and may improve drug development. Herein, we introduce hiPSC-CM as a suitable model to study T. cruzi heart infection and to predict the safety and efficacy of anti-T. cruzi drugs.
Collapse
Affiliation(s)
- Leonardo da Silva Lara
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fiocruz, Av. Brasil 4365, Manguinhos, RJ, 21045-900, Brazil
| | - Leonardo Andrade-Lima
- National Laboratory for Embryonic Stem Cells (LaNCE), Institute of Biosciences, University of São Paulo, SP, 05508-090, Brazil
| | - Claudia Magalhães Calvet
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fiocruz, Av. Brasil 4365, Manguinhos, RJ, 21045-900, Brazil
| | - Juliana Borsoi
- National Laboratory for Embryonic Stem Cells (LaNCE), Institute of Biosciences, University of São Paulo, SP, 05508-090, Brazil
| | | | - Andrea Henriques-Pons
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo cruz, Fiocruz - Av. Brasil 4365, Manguinhos, RJ, 21045-900, Brazil
| | - Mirian Claudia Souza Pereira
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fiocruz, Av. Brasil 4365, Manguinhos, RJ, 21045-900, Brazil.
| | - Lygia Veiga Pereira
- National Laboratory for Embryonic Stem Cells (LaNCE), Institute of Biosciences, University of São Paulo, SP, 05508-090, Brazil.
| |
Collapse
|
45
|
Rojas DA, Urbina F, Moreira-Ramos S, Castillo C, Kemmerling U, Lapier M, Maya JD, Solari A, Maldonado E. Endogenous overexpression of an active phosphorylated form of DNA polymerase β under oxidative stress in Trypanosoma cruzi. PLoS Negl Trop Dis 2018; 12:e0006220. [PMID: 29432450 PMCID: PMC5825160 DOI: 10.1371/journal.pntd.0006220] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 02/23/2018] [Accepted: 01/08/2018] [Indexed: 01/09/2023] Open
Abstract
Trypanosoma cruzi is exposed during its life to exogenous and endogenous oxidative stress, leading to damage of several macromolecules such as DNA. There are many DNA repair pathways in the nucleus and mitochondria (kinetoplast), where specific protein complexes detect and eliminate damage to DNA. One group of these proteins is the DNA polymerases. In particular, Tc DNA polymerase β participates in kinetoplast DNA replication and repair. However, the mechanisms which control its expression under oxidative stress are still unknown. Here we describe the effect of oxidative stress on the expression and function of Tc DNA polymerase β To this end parasite cells (epimastigotes and trypomastigotes) were exposed to peroxide during short periods of time. Tc DNA polymerase β which was associated physically with kinetoplast DNA, showed increased protein levels in response to peroxide damage in both parasite forms analyzed. Two forms of DNA polymerase β were identified and overexpressed after peroxide treatment. One of them was phosphorylated and active in DNA synthesis after renaturation on polyacrylamide electrophoresis gel. This phosphorylated form showed 3-4-fold increase in both parasite forms. Our findings indicate that these increments in protein levels are not under transcriptional control because the level of Tc DNA polymerase β mRNA is maintained or slightly decreased during the exposure to oxidative stress. We propose a mechanism where a DNA repair pathway activates a cascade leading to the increment of expression and phosphorylation of Tc DNA polymerase β in response to oxidative damage, which is discussed in the context of what is known in other trypanosomes which lack transcriptional control. Exposure of Trypanosome cruzi to oxidative stress leads to damage of several macromolecules such as DNA. DNA polymerases play a very important role in DNA repair after oxidative damage. One of them is Tc DNA polymerase β. In this work, two form of this DNA polymerase were identified and overexpressed in T. cruzi cells after hydrogen peroxide treatment been one of them a phosphorylated and highly active form. The increment of Tc DNA polymerase β was not correlated with changes in mRNA levels, indicating absence of transcriptional control. We propose a mechanism where hydrogen peroxide treatment activates a pathway leading to expression and phosphorylation of Tc DNA polymerase β in response to oxidative damage.
Collapse
Affiliation(s)
- Diego A. Rojas
- Microbiology and Micology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Fabiola Urbina
- Cellular and Molecular Biology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Sandra Moreira-Ramos
- Cellular and Molecular Biology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Christian Castillo
- Anatomy and Developmental Biology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Ulrike Kemmerling
- Anatomy and Developmental Biology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Michel Lapier
- Molecular and Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Juan Diego Maya
- Molecular and Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Aldo Solari
- Cellular and Molecular Biology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Edio Maldonado
- Cellular and Molecular Biology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
- * E-mail:
| |
Collapse
|
46
|
Franco J, Scarone L, Comini MA. Drugs and Drug Resistance in African and American Trypanosomiasis. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2018. [DOI: 10.1016/bs.armc.2018.08.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
47
|
Genome-wide mutagenesis and multi-drug resistance in American trypanosomes induced by the front-line drug benznidazole. Sci Rep 2017; 7:14407. [PMID: 29089615 PMCID: PMC5663738 DOI: 10.1038/s41598-017-14986-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 10/18/2017] [Indexed: 12/28/2022] Open
Abstract
Chagas disease is caused by the protozoan parasite Trypanosoma cruzi and affects 5–8 million people in Latin America. Although the nitroheterocyclic compound benznidazole has been the front-line drug for several decades, treatment failures are common. Benznidazole is a pro-drug and is bio-activated within the parasite by the mitochondrial nitroreductase TcNTR-1, leading to the generation of reactive metabolites that have trypanocidal activity. To better assess drug action and resistance, we sequenced the genomes of T. cruzi Y strain (35.5 Mb) and three benznidazole-resistant clones derived from a single drug-selected population. This revealed the genome-wide accumulation of mutations in the resistant parasites, in addition to variations in DNA copy-number. We observed mutations in DNA repair genes, linked with increased susceptibility to DNA alkylating and inter-strand cross-linking agents. Stop-codon-generating mutations in TcNTR-1 were associated with cross-resistance to other nitroheterocyclic drugs. Unexpectedly, the clones were also highly resistant to the ergosterol biosynthesis inhibitor posaconazole, a drug proposed for use against T. cruzi infections, in combination with benznidazole. Our findings therefore identify the highly mutagenic activity of benznidazole metabolites in T. cruzi, demonstrate that this can result in multi-drug resistance, and indicate that vigilance will be required if benznidazole is used in combination therapy.
Collapse
|
48
|
González L, García-Huertas P, Triana-Chávez O, García GA, Murta SMF, Mejía-Jaramillo AM. Aldo-keto reductase and alcohol dehydrogenase contribute to benznidazole natural resistance in Trypanosoma cruzi. Mol Microbiol 2017; 106:704-718. [PMID: 28884498 DOI: 10.1111/mmi.13830] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2017] [Indexed: 12/16/2022]
Abstract
The improvement of Chagas disease treatment is focused not only on the development of new drugs but also in understanding mechanisms of action and resistance to drugs conventionally used. Thus, some strategies aim to detect specific changes in proteins between sensitive and resistant parasites and to evaluate the role played in these processes by functional genomics. In this work, we used a natural Trypanosoma cruzi population resistant to benznidazole, which has clones with different susceptibilities to this drug without alterations in the NTR I gene. Using 2DE-gel electrophoresis, the aldo-keto reductase and the alcohol dehydrogenase proteins were found up regulated in the natural resistant clone and therefore their possible role in the resistance to benznidazole and glyoxal was investigated. Both genes were overexpressed in a drug sensitive T. cruzi clone and the biological changes in response to these compounds were evaluated. The results showed that the overexpression of these proteins enhances resistance to benznidazole and glyoxal in T. cruzi. Moreover, a decrease in mitochondrial and cell membrane damage was observed, accompanied by a drop in the intracellular concentration of reactive oxygen species after treatment. Our results suggest that these proteins are involved in the mechanism of action of benznidazole.
Collapse
Affiliation(s)
- Laura González
- Grupo Biología y Control de Enfermedades Infecciosas-BCEI, Universidad de Antioquia, UdeA, Medellín, Colombia
| | - Paola García-Huertas
- Grupo Biología y Control de Enfermedades Infecciosas-BCEI, Universidad de Antioquia, UdeA, Medellín, Colombia
| | - Omar Triana-Chávez
- Grupo Biología y Control de Enfermedades Infecciosas-BCEI, Universidad de Antioquia, UdeA, Medellín, Colombia
| | - Gabriela Andrea García
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chaben"- ANLIS "Dr. Carlos G. Malbrán", Buenos Aires, Argentina
| | | | - Ana M Mejía-Jaramillo
- Grupo Biología y Control de Enfermedades Infecciosas-BCEI, Universidad de Antioquia, UdeA, Medellín, Colombia
| |
Collapse
|
49
|
Zulfiqar B, Jones AJ, Sykes ML, Shelper TB, Davis RA, Avery VM. Screening a Natural Product-Based Library against Kinetoplastid Parasites. Molecules 2017; 22:E1715. [PMID: 29023425 PMCID: PMC6151456 DOI: 10.3390/molecules22101715] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/04/2017] [Accepted: 10/04/2017] [Indexed: 01/06/2023] Open
Abstract
Kinetoplastid parasites cause vector-borne parasitic diseases including leishmaniasis, human African trypanosomiasis (HAT) and Chagas disease. These Neglected Tropical Diseases (NTDs) impact on some of the world's lowest socioeconomic communities. Current treatments for these diseases cause severe toxicity and have limited efficacy, highlighting the need to identify new treatments. In this study, the Davis open access natural product-based library was screened against kinetoplastids (Leishmania donovani DD8, Trypanosoma brucei brucei and Trypanosoma cruzi) using phenotypic assays. The aim of this study was to identify hit compounds, with a focus on improved efficacy, selectivity and potential to target several kinetoplastid parasites. The IC50 values of the natural products were obtained for L. donovani DD8, T. b. brucei and T. cruzi in addition to cytotoxicity against the mammalian cell lines, HEK-293, 3T3 and THP-1 cell lines were determined to ascertain parasite selectivity. Thirty-one compounds were identified with IC50 values of ≤ 10 µM against the kinetoplastid parasites tested. Lissoclinotoxin E (1) was the only compound identified with activity across all three investigated parasites, exhibiting IC50 values < 5 µM. In this study, natural products with the potential to be new chemical starting points for drug discovery efforts for kinetoplastid diseases were identified.
Collapse
Affiliation(s)
- Bilal Zulfiqar
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland 4111, Australia.
| | - Amy J Jones
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland 4111, Australia.
| | - Melissa L Sykes
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland 4111, Australia.
| | - Todd B Shelper
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland 4111, Australia.
| | - Rohan A Davis
- Natural Product Chemistry, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland 4111, Australia.
| | - Vicky M Avery
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland 4111, Australia.
| |
Collapse
|
50
|
Synthesis, crystal structure, catalytic and anti-Trypanosoma cruzi activity of a new chromium(III) complex containing bis(3,5-dimethylpyrazol-1-yl)methane. J Mol Struct 2017. [DOI: 10.1016/j.molstruc.2017.06.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|