1
|
Gómez-Escobedo R, Méndez-Álvarez D, Vázquez C, Saavedra E, Vázquez K, Alcántara-Farfán V, Cordero-Martínez J, Gonzalez-Gonzalez A, Rivera G, Nogueda-Torres B. Molecular Docking-Based Virtual Screening of FDA-Approved Drugs Using Trypanothione Reductase Identified New Trypanocidal Agents. Molecules 2024; 29:3796. [PMID: 39202874 PMCID: PMC11357579 DOI: 10.3390/molecules29163796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024] Open
Abstract
American trypanosomiasis or Chagas disease, caused by Trypanosoma cruzi (T. cruzi), affects approximately 6-7 million people worldwide. However, its pharmacological treatment causes several uncomfortable side effects, causing patients' treatment abandonment. Therefore, there is a need for new and better treatments. In this work, the molecular docking of nine hundred twenty-four FDA-approved drugs on three different sites of trypanothione reductase of T. cruzi (TcTR) was carried out to find potential trypanocidal agents. Finally, biological evaluations in vitro and in vivo were conducted with the selected FDA-approved drugs. Digoxin, alendronate, flucytosine, and dihydroergotamine showed better trypanocidal activity than the reference drugs benznidazole and nifurtimox in the in vitro evaluation against the trypomastigotes form. Further, these FDA-approved drugs were able to reduce 20-50% parasitemia in a short time in an in vivo model, although with less efficiency than benznidazole. Therefore, the results suggest a combined therapy of repurposed and canonical drugs against T. cruzi infection.
Collapse
Affiliation(s)
- Rogelio Gómez-Escobedo
- Departamento de Parasitología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 07738, Mexico;
| | - Domingo Méndez-Álvarez
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico; (D.M.-Á.); (A.G.-G.)
| | - Citlali Vázquez
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico; (C.V.); (E.S.)
| | - Emma Saavedra
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico; (C.V.); (E.S.)
| | - Karina Vázquez
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Nuevo León, General Escobedo 66050, Mexico;
| | - Verónica Alcántara-Farfán
- Laboratorio de Bioquímica Farmacológica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 07738, Mexico; (V.A.-F.); (J.C.-M.)
| | - Joaquín Cordero-Martínez
- Laboratorio de Bioquímica Farmacológica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 07738, Mexico; (V.A.-F.); (J.C.-M.)
| | - Alonzo Gonzalez-Gonzalez
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico; (D.M.-Á.); (A.G.-G.)
| | - Gildardo Rivera
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico; (D.M.-Á.); (A.G.-G.)
| | - Benjamín Nogueda-Torres
- Departamento de Parasitología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 07738, Mexico;
| |
Collapse
|
2
|
González-Matos M, Aguado ME, Izquierdo M, Monzote L, González-Bacerio J. Compounds with potentialities as novel chemotherapeutic agents in leishmaniasis at preclinical level. Exp Parasitol 2024; 260:108747. [PMID: 38518969 DOI: 10.1016/j.exppara.2024.108747] [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: 12/15/2023] [Revised: 02/27/2024] [Accepted: 03/19/2024] [Indexed: 03/24/2024]
Abstract
Leishmaniasis are neglected infectious diseases caused by kinetoplastid protozoan parasites from the genus Leishmania. These sicknesses are present mainly in tropical regions and almost 1 million new cases are reported each year. The absence of vaccines, as well as the high cost, toxicity or resistance to the current drugs determines the necessity of new treatments against these pathologies. In this review, several compounds with potentialities as new antileishmanial drugs are presented. The discussion is restricted to the preclinical level and molecules are organized according to their chemical nature, source and molecular targets. In this manner, we present antimicrobial peptides, flavonoids, withanolides, 8-aminoquinolines, compounds from Leish-Box, pyrazolopyrimidines, and inhibitors of tubulin polymerization/depolymerization, topoisomerase IB, proteases, pteridine reductase, N-myristoyltransferase, as well as enzymes involved in polyamine metabolism, response against oxidative stress, signaling pathways, and sterol biosynthesis. This work is a contribution to the general knowledge of these compounds as antileishmanial agents.
Collapse
Affiliation(s)
- Maikel González-Matos
- Center for Protein Studies, Faculty of Biology, University of Havana, Calle 25 #455 Entre I y J, Vedado, La Habana, Cuba
| | - Mirtha Elisa Aguado
- Center for Protein Studies, Faculty of Biology, University of Havana, Calle 25 #455 Entre I y J, Vedado, La Habana, Cuba
| | - Maikel Izquierdo
- Center for Protein Studies, Faculty of Biology, University of Havana, Calle 25 #455 Entre I y J, Vedado, La Habana, Cuba
| | - Lianet Monzote
- Department of Parasitology, Center for Research, Diagnosis and Reference, Tropical Medicine Institute "Pedro Kourí", Autopista Novia Del Mediodía Km 6½, La Lisa, La Habana, Cuba.
| | - Jorge González-Bacerio
- Center for Protein Studies, Faculty of Biology, University of Havana, Calle 25 #455 Entre I y J, Vedado, La Habana, Cuba; Department of Biochemistry, Faculty of Biology, University of Havana, Calle 25 #455 Entre I y J, Vedado, La Habana, Cuba.
| |
Collapse
|
3
|
Marín M, López M, Gallego-Yerga L, Álvarez R, Peláez R. Experimental structure based drug design (SBDD) applications for anti-leishmanial drugs: A paradigm shift? Med Res Rev 2024; 44:1055-1120. [PMID: 38142308 DOI: 10.1002/med.22005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 11/14/2023] [Accepted: 11/27/2023] [Indexed: 12/25/2023]
Abstract
Leishmaniasis is a group of neglected tropical diseases caused by at least 20 species of Leishmania protozoa, which are spread by the bite of infected sandflies. There are three main forms of the disease: cutaneous leishmaniasis (CL, the most common), visceral leishmaniasis (VL, also known as kala-azar, the most serious), and mucocutaneous leishmaniasis. One billion people live in areas endemic to leishmaniasis, with an annual estimation of 30,000 new cases of VL and more than 1 million of CL. New treatments for leishmaniasis are an urgent need, as the existing ones are inefficient, toxic, and/or expensive. We have revised the experimental structure-based drug design (SBDD) efforts applied to the discovery of new drugs against leishmaniasis. We have grouped the explored targets according to the metabolic pathways they belong to, and the key achieved advances are highlighted and evaluated. In most cases, SBDD studies follow high-throughput screening campaigns and are secondary to pharmacokinetic optimization, due to the majoritarian belief that there are few validated targets for SBDD in leishmaniasis. However, some SBDD strategies have significantly contributed to new drug candidates against leishmaniasis and a bigger number holds promise for future development.
Collapse
Affiliation(s)
- Miguel Marín
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Marta López
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Laura Gallego-Yerga
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Raquel Álvarez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Rafael Peláez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| |
Collapse
|
4
|
Exertier C, Salerno A, Antonelli L, Fiorillo A, Ocello R, Seghetti F, Caciolla J, Uliassi E, Masetti M, Fiorentino E, Orsini S, Di Muccio T, Ilari A, Bolognesi ML. Fragment Merging, Growing, and Linking Identify New Trypanothione Reductase Inhibitors for Leishmaniasis. J Med Chem 2024; 67:402-419. [PMID: 38164929 PMCID: PMC10788915 DOI: 10.1021/acs.jmedchem.3c01439] [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: 08/06/2023] [Revised: 10/30/2023] [Accepted: 11/27/2023] [Indexed: 01/03/2024]
Abstract
Trypanothione reductase (TR) is a suitable target for drug discovery approaches against leishmaniasis, although the identification of potent inhibitors is still challenging. Herein, we harnessed a fragment-based drug discovery (FBDD) strategy to develop new TR inhibitors. Previous crystallographic screening identified fragments 1-3, which provided ideal starting points for a medicinal chemistry campaign. In silico investigations revealed critical hotspots in the TR binding site, guiding our structure- and ligand-based structure-actvity relationship (SAR) exploration that yielded fragment-derived compounds 4-14. A trend of improvement in Leishmania infantum TR inhibition was detected along the optimization and confirmed by the crystal structures of 9, 10, and 14 in complex with Trypanosoma brucei TR. Compound 10 showed the best TR inhibitory profile (Ki = 0.2 μM), whereas 9 was the best one in terms of in vitro and ex vivo activity. Although further fine-tuning is needed to improve selectivity, we demonstrated the potentiality of FBDD on a classic but difficult target for leishmaniasis.
Collapse
Affiliation(s)
- Cécile Exertier
- Institute of Molecular Biology and Pathology (IBPM) of the National Research Council of Italy (CNR), c/o Department of Biochemical Sciences, Sapienza University of Rome, Piazzale A. Moro 5, Roma 00185, Italy
| | - Alessandra Salerno
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum─University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Lorenzo Antonelli
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, Roma 00185, Italy
| | - Annarita Fiorillo
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, Roma 00185, Italy
| | - Riccardo Ocello
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum─University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, via Morego 30, Genova 16163, Italy
| | - Francesca Seghetti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum─University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Jessica Caciolla
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum─University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Elisa Uliassi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum─University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Matteo Masetti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum─University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Eleonora Fiorentino
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, Roma 00161, Italy
| | - Stefania Orsini
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, Roma 00161, Italy
| | - Trentina Di Muccio
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, Roma 00161, Italy
| | - Andrea Ilari
- Institute of Molecular Biology and Pathology (IBPM) of the National Research Council of Italy (CNR), c/o Department of Biochemical Sciences, Sapienza University of Rome, Piazzale A. Moro 5, Roma 00185, Italy
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum─University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| |
Collapse
|
5
|
Barrera-Téllez FJ, Prieto-Martínez FD, Hernández-Campos A, Martínez-Mayorga K, Castillo-Bocanegra R. In Silico Exploration of the Trypanothione Reductase (TryR) of L. mexicana. Int J Mol Sci 2023; 24:16046. [PMID: 38003236 PMCID: PMC10671491 DOI: 10.3390/ijms242216046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/23/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
Human leishmaniasis is a neglected tropical disease which affects nearly 1.5 million people every year, with Mexico being an important endemic region. One of the major defense mechanisms of these parasites is based in the polyamine metabolic pathway, as it provides the necessary compounds for its survival. Among the enzymes in this route, trypanothione reductase (TryR), an oxidoreductase enzyme, is crucial for the Leishmania genus' survival against oxidative stress. Thus, it poses as an attractive drug target, yet due to the size and features of its catalytic pocket, modeling techniques such as molecular docking focusing on that region is not convenient. Herein, we present a computational study using several structure-based approaches to assess the druggability of TryR from L. mexicana, the predominant Leishmania species in Mexico, beyond its catalytic site. Using this consensus methodology, three relevant pockets were found, of which the one we call σ-site promises to be the most favorable one. These findings may help the design of new drugs of trypanothione-related diseases.
Collapse
Affiliation(s)
- Francisco J. Barrera-Téllez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Fernando D. Prieto-Martínez
- Instituto de Química, Unidad Mérida, Universidad Nacional Autónoma de México, Carretera Mérida-Tetiz, Km. 4.5, Ucú 97357, Mexico
| | - Alicia Hernández-Campos
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Karina Martínez-Mayorga
- Instituto de Investigaciones en Matemáticas Aplicadas y en Sistemas, Unidad Mérida, Universidad Nacional Autónoma de México, Sierra Papacal, Mérida 97302, Mexico
| | - Rafael Castillo-Bocanegra
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| |
Collapse
|
6
|
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: 1.0] [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
|
7
|
Fiorillo A, Colotti G, Exertier C, Liuzzi A, Seghetti F, Salerno A, Caciolla J, Ilari A. Innovative Approach for a Classic Target: Fragment Screening on Trypanothione Reductase Reveals New Opportunities for Drug Design. Front Mol Biosci 2022; 9:900882. [PMID: 35860359 PMCID: PMC9289546 DOI: 10.3389/fmolb.2022.900882] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/19/2022] [Indexed: 11/22/2022] Open
Abstract
Trypanothione reductase (TR) is a key factor in the redox homeostasis of trypanosomatid parasites, critical for survival in the hostile oxidative environment generated by the host to fight infection. TR is considered an attractive target for the development of new trypanocidal agents as it is essential for parasite survival but has no close homolog in humans. However, the high efficiency and turnover of TR challenging targets since only potent inhibitors, with nanomolar IC50, can significantly affect parasite redox state and viability. To aid the design of effective compounds targeting TR, we performed a fragment-based crystal screening at the Diamond Light Source XChem facility using a library optimized for follow-up synthesis steps. The experiment, allowing for testing over 300 compounds, resulted in the identification of 12 new ligands binding five different sites. Interestingly, the screening revealed the existence of an allosteric pocket close to the NADPH binding site, named the “doorstop pocket” since ligands binding at this site interfere with TR activity by hampering the “opening movement” needed to allow cofactor binding. The second remarkable site, known as the Z-site, identified by the screening, is located within the large trypanothione cavity but corresponds to a region not yet exploited for inhibition. The fragments binding to this site are close to each other and have some remarkable features making them ideal for follow-up optimization as a piperazine moiety in three out of five fragments.
Collapse
Affiliation(s)
- Annarita Fiorillo
- Institute of Molecular Biology and Pathology, Italian National Research Council, IBPM-CNR, Rome, Italy
- Department of Biochemical Sciences, Sapienza University, Rome, Italy
- *Correspondence: Annarita Fiorillo,
| | - Gianni Colotti
- Institute of Molecular Biology and Pathology, Italian National Research Council, IBPM-CNR, Rome, Italy
- Department of Biochemical Sciences, Sapienza University, Rome, Italy
| | - Cécile Exertier
- Institute of Molecular Biology and Pathology, Italian National Research Council, IBPM-CNR, Rome, Italy
| | - Anastasia Liuzzi
- Institute of Molecular Biology and Pathology, Italian National Research Council, IBPM-CNR, Rome, Italy
| | - Francesca Seghetti
- Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Alessandra Salerno
- Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Jessica Caciolla
- Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Andrea Ilari
- Institute of Molecular Biology and Pathology, Italian National Research Council, IBPM-CNR, Rome, Italy
| |
Collapse
|
8
|
Ali V, Behera S, Nawaz A, Equbal A, Pandey K. Unique thiol metabolism in trypanosomatids: Redox homeostasis and drug resistance. ADVANCES IN PARASITOLOGY 2022; 117:75-155. [PMID: 35878950 DOI: 10.1016/bs.apar.2022.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Trypanosomatids are mainly responsible for heterogeneous parasitic diseases: Leishmaniasis, Sleeping sickness, and Chagas disease and control of these diseases implicates serious challenges due to the emergence of drug resistance. Redox-active biomolecules are the endogenous substances in organisms, which play important role in the regulation of redox homeostasis. The redox-active substances like glutathione, trypanothione, cysteine, cysteine persulfides, etc., and other inorganic intermediates (hydrogen peroxide, nitric oxide) are very useful as defence mechanism. In the present review, the suitability of trypanothione and other essential thiol molecules of trypanosomatids as drug targets are described in Leishmania and Trypanosoma. We have explored the role of tryparedoxin, tryparedoxin peroxidase, ascorbate peroxidase, superoxide dismutase, and glutaredoxins in the anti-oxidant mechanism and drug resistance. Up-regulation of some proteins in trypanothione metabolism helps the parasites in survival against drug pressure (sodium stibogluconate, Amphotericin B, etc.) and oxidative stress. These molecules accept electrons from the reduced trypanothione and donate their electrons to other proteins, and these proteins reduce toxic molecules, neutralize reactive oxygen, or nitrogen species; and help parasites to cope with oxidative stress. Thus, a better understanding of the role of these molecules in drug resistance and redox homeostasis will help to target metabolic pathway proteins to combat Leishmaniasis and trypanosomiases.
Collapse
Affiliation(s)
- Vahab Ali
- Laboratory of Molecular Biochemistry and Cell Biology, Department of Biochemistry, ICMR-Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Patna, Bihar, India.
| | - Sachidananda Behera
- Laboratory of Molecular Biochemistry and Cell Biology, Department of Biochemistry, ICMR-Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Patna, Bihar, India
| | - Afreen Nawaz
- Laboratory of Molecular Biochemistry and Cell Biology, Department of Biochemistry, ICMR-Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Patna, Bihar, India
| | - Asif Equbal
- Laboratory of Molecular Biochemistry and Cell Biology, Department of Biochemistry, ICMR-Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Patna, Bihar, India; Department of Botany, Araria College, Purnea University, Purnia, Bihar, India
| | - Krishna Pandey
- Department of Clinical Medicine, ICMR-Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Patna, Bihar, India
| |
Collapse
|
9
|
Docampo R, Vercesi AE. Mitochondrial Ca 2+ and Reactive Oxygen Species in Trypanosomatids. Antioxid Redox Signal 2022; 36:969-983. [PMID: 34218689 PMCID: PMC9125514 DOI: 10.1089/ars.2021.0058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/31/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023]
Abstract
Significance: Millions of people are infected with trypanosomatids and new therapeutic approaches are needed. Trypanosomatids possess one mitochondrion per cell and its study has led to discoveries of general biological interest. These mitochondria, as in their animal counterparts, generate reactive oxygen species (ROS) and have evolved enzymatic and nonenzymatic defenses against them. Mitochondrial calcium ion (Ca2+) overload leads to generation of ROS and its study could lead to relevant information on the biology of trypanosomatids and to novel drug targets. Recent Advances: Mitochondrial Ca2+ is normally involved in maintaining the bioenergetics of trypanosomes, but when Ca2+ overload occurs, it is associated with cell death. Trypanosomes lack key players in the mechanism of cell death described in mammalian cells, although mitochondrial Ca2+ overload results in collapse of their membrane potential, production of ROS, and cytochrome c release. They are also very resistant to mitochondrial permeability transition, and cell death after mitochondrial Ca2+ overload depends on generation of ROS. Critical Issues: In this review, we consider the mechanisms of mitochondrial oxidant generation and removal and the involvement of Ca2+ in trypanosome cell death. Future Directions: More studies are required to determine the reactions involved in generation of ROS by the mitochondria of trypanosomatids, their enzymatic and nonenzymatic defenses against ROS, and the occurrence and composition of a mitochondrial permeability transition pore. Antioxid. Redox Signal. 36, 969-983.
Collapse
Affiliation(s)
- Roberto Docampo
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA
| | | |
Collapse
|
10
|
Examination of multiple Trypanosoma cruzi targets in a new drug discovery approach for Chagas disease. Bioorg Med Chem 2022; 58:116577. [DOI: 10.1016/j.bmc.2021.116577] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/10/2021] [Accepted: 12/10/2021] [Indexed: 12/21/2022]
|
11
|
Li M, Gaussmann S, Tippler B, Ott J, Popowicz GM, Schliebs W, Sattler M, Erdmann R, Kalel VC. Novel Trypanocidal Inhibitors that Block Glycosome Biogenesis by Targeting PEX3-PEX19 Interaction. Front Cell Dev Biol 2022; 9:737159. [PMID: 34988071 PMCID: PMC8721105 DOI: 10.3389/fcell.2021.737159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/15/2021] [Indexed: 12/02/2022] Open
Abstract
Human pathogenic trypanosomatid parasites harbor a unique form of peroxisomes termed glycosomes that are essential for parasite viability. We and others previously identified and characterized the essential Trypanosoma brucei ortholog TbPEX3, which is the membrane-docking factor for the cytosolic receptor PEX19 bound to the glycosomal membrane proteins. Knockdown of TbPEX3 expression leads to mislocalization of glycosomal membrane and matrix proteins, and subsequent cell death. As an early step in glycosome biogenesis, the PEX3–PEX19 interaction is an attractive drug target. We established a high-throughput assay for TbPEX3–TbPEX19 interaction and screened a compound library for small-molecule inhibitors. Hits from the screen were further validated using an in vitro ELISA assay. We identified three compounds, which exhibit significant trypanocidal activity but show no apparent toxicity to human cells. Furthermore, we show that these compounds lead to mislocalization of glycosomal proteins, which is toxic to the trypanosomes. Moreover, NMR-based experiments indicate that the inhibitors bind to PEX3. The inhibitors interfering with glycosomal biogenesis by targeting the TbPEX3–TbPEX19 interaction serve as starting points for further optimization and anti-trypanosomal drug development.
Collapse
Affiliation(s)
- Mengqiao Li
- Department of Systems Biochemistry, Faculty of Medicine, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Stefan Gaussmann
- Institute of Structural Biology, Helmholtz Zentrum München, Neuherberg, Germany.,Department of Chemistry, Bavarian NMR Center, Technical University of Munich, Garching, Germany
| | - Bettina Tippler
- Department of Systems Biochemistry, Faculty of Medicine, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Julia Ott
- Department of Systems Biochemistry, Faculty of Medicine, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Grzegorz M Popowicz
- Institute of Structural Biology, Helmholtz Zentrum München, Neuherberg, Germany.,Department of Chemistry, Bavarian NMR Center, Technical University of Munich, Garching, Germany
| | - Wolfgang Schliebs
- Department of Systems Biochemistry, Faculty of Medicine, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Michael Sattler
- Institute of Structural Biology, Helmholtz Zentrum München, Neuherberg, Germany.,Department of Chemistry, Bavarian NMR Center, Technical University of Munich, Garching, Germany
| | - Ralf Erdmann
- Department of Systems Biochemistry, Faculty of Medicine, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Vishal C Kalel
- Department of Systems Biochemistry, Faculty of Medicine, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
12
|
Ramu D, Singh S. Potential molecular targets of Leishmania pathways in developing novel antileishmanials. Future Microbiol 2021; 17:41-57. [PMID: 34877877 DOI: 10.2217/fmb-2021-0094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The illness known as leishmaniasis has not become a household name like malaria, although it stands as the second-largest parasitic disease, surpassed only by malaria. As no licensed vaccine is available, treatment for leishmaniasis mostly relies on chemotherapy. Inefficiency and drug resistance are the major impediments in current therapeutics. In this scenario, identification of novel molecular drug candidates is indispensable to develop robust antileishmanials. The exploration of structure-based drugs to target enzymes/molecules of Leishmania which differ structurally/functionally from their equivalents in mammalian hosts not only helps in developing a new class of antileishmanials, but also paves the way to understand Leishmania biology. This review provides a comprehensive overview on possible drug candidates relating to various Leishmania molecular pathways.
Collapse
Affiliation(s)
- Dandugudumula Ramu
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, 201314, India
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| |
Collapse
|
13
|
Turcano L, Battista T, De Haro ET, Missineo A, Alli C, Paonessa G, Colotti G, Harper S, Fiorillo A, Ilari A, Bresciani A. Spiro-containing derivatives show antiparasitic activity against Trypanosoma brucei through inhibition of the trypanothione reductase enzyme. PLoS Negl Trop Dis 2020; 14:e0008339. [PMID: 32437349 PMCID: PMC7269337 DOI: 10.1371/journal.pntd.0008339] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 06/03/2020] [Accepted: 04/30/2020] [Indexed: 11/19/2022] Open
Abstract
Trypanothione reductase (TR) is a key enzyme that catalyzes the reduction of trypanothione, an antioxidant dithiol that protects Trypanosomatid parasites from oxidative stress induced by mammalian host defense systems. TR is considered an attractive target for the development of novel anti-parasitic agents as it is essential for parasite survival but has no close homologue in humans. We report here the identification of spiro-containing derivatives as inhibitors of TR from Trypanosoma brucei (TbTR), the parasite responsible for Human African Trypanosomiasis. The hit series, identified by high throughput screening, was shown to bind TbTR reversibly and to compete with the trypanothione (TS2) substrate. The prototype compound 1 from this series was also found to impede the growth of Trypanosoma brucei parasites in vitro. The X-ray crystal structure of TbTR in complex with compound 1 solved at 1.98 Å allowed the identification of the hydrophobic pocket where the inhibitor binds, placed close to the catalytic histidine (His 461’) and lined by Trp21, Val53, Ile106, Tyr110 and Met113. This new inhibitor is specific for TbTR and no activity was detected against the structurally similar human glutathione reductase (hGR). The central spiro scaffold is known to be suitable for brain active compounds in humans thus representing an attractive starting point for the future treatment of the central nervous system stage of T. brucei infections. Trypanosoma brucei is a parasite responsible for neglected pathologies such as human African trypanosomiasis, also known as sleeping sickness. This disease is endemic in sub-Saharan Africa, with 70 million people at risk of infection. Current treatments for this type of disease are limited by their toxicity, administration in endemic countries and treatment resistance. Therapies against infectious diseases typically rely on targeting one or more components of the parasite that are not present in humans to ensure the best possible therapeutic window. In this case we aimed at targeting the Trypanosoma brucei trypanothione reductase (TR), one enzyme that synthesize the reduced trypanothione a key molecule for preserving the parasite redox balance. This enzyme does not exist in humans that have glutathione instead of trypanothione. Past attempts to identify novel inhibitors of this target has failed to generate drug-like molecules. To overcome this limitation we employed a recent, higher quality, TR activity assay to test a collection of compounds previously reported to be active against these parasites. This approach led to the identification and validation of a new chemotype with a unique mode of inhibition of TR. This chemical series is a drug-like starting point, in fact its core (spiro) is present in drugs approved for human use.
Collapse
Affiliation(s)
- Lorenzo Turcano
- Department of Translational and Discovery Research, Pomezia (Roma) Italy
| | - Theo Battista
- Dipartimento di Scienze Biochimiche, Sapienza Università di Roma, Roma, Italy
| | | | - Antonino Missineo
- Department of Translational and Discovery Research, Pomezia (Roma) Italy
| | - Cristina Alli
- Department of Translational and Discovery Research, Pomezia (Roma) Italy
| | - Giacomo Paonessa
- Department of Translational and Discovery Research, Pomezia (Roma) Italy
| | - Gianni Colotti
- Istituto di Biologia e Patologia Molecolari del CNR c/o Dipartimento di Scienze Biochimiche, Sapienza Università di Roma, Roma, Italy
| | | | - Annarita Fiorillo
- Dipartimento di Scienze Biochimiche, Sapienza Università di Roma, Roma, Italy
| | - Andrea Ilari
- Istituto di Biologia e Patologia Molecolari del CNR c/o Dipartimento di Scienze Biochimiche, Sapienza Università di Roma, Roma, Italy
- * E-mail: (AI); (AB)
| | - Alberto Bresciani
- Department of Translational and Discovery Research, Pomezia (Roma) Italy
- * E-mail: (AI); (AB)
| |
Collapse
|
14
|
Talevi A, Carrillo C, Comini M. The Thiol-polyamine Metabolism of Trypanosoma cruzi: Molecular Targets and Drug Repurposing Strategies. Curr Med Chem 2019; 26:6614-6635. [PMID: 30259812 DOI: 10.2174/0929867325666180926151059] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/23/2018] [Accepted: 09/10/2018] [Indexed: 12/18/2022]
Abstract
Chagas´ disease continues to be a challenging and neglected public health problem in many American countries. The etiologic agent, Trypanosoma cruzi, develops intracellularly in the mammalian host, which hinders treatment efficacy. Progress in the knowledge of parasite biology and host-pathogen interaction has not been paralleled by the development of novel, safe and effective therapeutic options. It is then urgent to seek for novel therapeutic candidates and to implement drug discovery strategies that may accelerate the discovery process. The most appealing targets for pharmacological intervention are those essential for the pathogen and, whenever possible, absent or significantly different from the host homolog. The thiol-polyamine metabolism of T. cruzi offers interesting candidates for a rational design of selective drugs. In this respect, here we critically review the state of the art of the thiolpolyamine metabolism of T. cruzi and the pharmacological potential of its components. On the other hand, drug repurposing emerged as a valid strategy to identify new biological activities for drugs in clinical use, while significantly shortening the long time and high cost associated with de novo drug discovery approaches. Thus, we also discuss the different drug repurposing strategies available with a special emphasis in their applications to the identification of drug candidates targeting essential components of the thiol-polyamine metabolism of T. cruzi.
Collapse
Affiliation(s)
- Alan Talevi
- Medicinal Chemistry, Department of Biological Sciences, Faculty of Exact Sciences, University of La Plata, La Plata, Argentina
| | - Carolina Carrillo
- Instituto de Ciencias y Tecnología Dr. César Milstein (ICT Milstein) - CONICET. Ciudad Autónoma de Buenos Aires, Argentina
| | - Marcelo Comini
- Institut Pasteur de Montevideo, Mataojo 2020, Montevideo 11400, Uruguay
| |
Collapse
|
15
|
do Carmo Santos N, da Paixão VG, da Rocha Pita SS. New Trypanosoma cruzi Trypanothione Reductase Inhibitors Identification using the Virtual Screening in Database of Nucleus Bioassay, Biosynthesis and Ecophysiology (NuBBE). ACTA ACUST UNITED AC 2019. [DOI: 10.2174/2211352516666180928130031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Background:
American trypanosomiasis, also known as Chagas disease, is caused by
the protozoan Trypanosoma cruzi (T. cruzi) and affects approximately 10 to 12 million, primarily
in Latin America. Since its discovery in 1909, there is no effective treatment for its chronic phase,
with benzonidazole being the only anti-trypanosoma drug used in Brazil, despite the absence of
conclusive evidence to prove its efficacy and safety. Thus, it is necessary to develop new drugs that
are more effective and selective against Trypanosoma cruzi.
Methods:
The T. cruzi enzyme Trypanothione Reductase (TcTR) is a validated target for the discovery
of new antiprotozoal compounds and we employed the Virtual Screening technique on the
database of Nucleus of Bioassays, Biosynthesis and Ecophysiology (NuBBE), aiming to search for
new chemical moieties against T. cruzi. From these we selected the 10 best ligand energies interactions
and verified their interaction profile with the main TcTR sites through the AuPosSOM server
(https://www.biomedicale.univ-paris5.fr/aupossom).
Results and Conclusion:
Finally, we analyzed some pharmacokinetics and toxicological information
through the servers Aggregator Advisor (http://advisor.bkslab.org), Pred-hERG 4.0
(http://labmol.com.br/predherg) and pkCSM (http://biosig.unimelb.edu.au/pkcsm/prediction) which
we expect will be useful in in vitro preclinical trials.</P>
Collapse
Affiliation(s)
- Nelcí do Carmo Santos
- Bioinformatics and Molecular Modeling Laboratory (LaBiMM), Pharmacy College, Federal University of Bahia, Salvador-BA, Brazil
| | - Vinícius G. da Paixão
- Bioinformatics and Molecular Modeling Laboratory (LaBiMM), Pharmacy College, Federal University of Bahia, Salvador-BA, Brazil
| | - Samuel S. da Rocha Pita
- Bioinformatics and Molecular Modeling Laboratory (LaBiMM), Pharmacy College, Federal University of Bahia, Salvador-BA, Brazil
| |
Collapse
|
16
|
da Paixão VG, Pita SSDR. In silico identification and evaluation of new Trypanosoma cruzi trypanothione reductase (TcTR) inhibitors obtained from natural products database of the Bahia semi-arid region (NatProDB). Comput Biol Chem 2019; 79:36-47. [PMID: 30710804 DOI: 10.1016/j.compbiolchem.2019.01.009] [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: 11/26/2018] [Revised: 01/14/2019] [Accepted: 01/17/2019] [Indexed: 11/17/2022]
Abstract
Trypanosoma cruzi Trypanothione Reductase (TcTR) is one of the therapeutic targets studied in the development of new drugs against Chagas' disease. Due to its biodiversity, Brazil has several compounds of natural origin that were not yet properly explored in drug discovery. Therefore, we employed the Virtual Screening against TcTR aiming to discover new inhibitors from the Natural Products Database of the Bahia Semi-Arid region (NatProDB). This database has a wide chemical diversity favoring the discovery of new chemical entities. Subsequently, we analyzed the best docking conformations using self-organizing maps (AuPosSOM) aiming to verify their interaction sites at TcTR. Finally, the Pred-hERG, the Aggregator Advisor, the FAF-DRUGS and the pkCSM results allowed us to evaluate, respectively, the cardiotoxicity, aggregation capacity, presence of false positives (PAINS) and its toxicity. Thus, we selected three molecules that could be tested in in vitro assays in the hope that the computational results reported here would favor the development of new anti-chagasic drugs.
Collapse
Affiliation(s)
- Vinícius Guimarães da Paixão
- Laboratório de Bioinformática e Modelagem Molecular (LaBiMM), Universidade Federal da Bahia, Av. Barão de Jeremoabo, 147, Faculdade de Farmácia, Ondina, 40170-115, Salvador, BA, Brazil
| | - Samuel Silva da Rocha Pita
- Laboratório de Bioinformática e Modelagem Molecular (LaBiMM), Universidade Federal da Bahia, Av. Barão de Jeremoabo, 147, Faculdade de Farmácia, Ondina, 40170-115, Salvador, BA, Brazil.
| |
Collapse
|
17
|
Ilari A, Genovese I, Fiorillo F, Battista T, De Ionna I, Fiorillo A, Colotti G. Toward a Drug Against All Kinetoplastids: From LeishBox to Specific and Potent Trypanothione Reductase Inhibitors. Mol Pharm 2018; 15:3069-3078. [PMID: 29897765 DOI: 10.1021/acs.molpharmaceut.8b00185] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Leishmaniasis, Chagas disease, and sleeping sickness affect millions of people worldwide and lead to the death of about 50 000 humans per year. These diseases are caused by the kinetoplastids Leishmania, Trypanosoma cruzi, and Trypanosoma brucei, respectively. These parasites share many general features, including gene conservation, high amino acid identity among proteins, the presence of subcellular structures as glycosomes and the kinetoplastid, and genome architecture, that may make drug development family specific, rather than species-specific, i.e., on the basis of the inhibition of a common, conserved parasite target. However, no optimal molecular targets or broad-spectrum drugs have been identified to date to cure these diseases. Here, the LeishBox from GlaxoSmithKline high-throughput screening, a 192-molecule set of best antileishmanial compounds, based on 1.8 million compounds, was used to identify specific inhibitors of a validated Leishmania target, trypanothione reductase (TR), while analyzing in parallel the homologous human enzyme glutathione reductase (GR). We identified three specific highly potent TR inhibitors and performed docking on the TR solved structure, thereby elucidating the putative molecular basis of TR inhibition. Since TRs from kinetoplastids are well conserved, and these compounds inhibit the growth of Leishmania, Trypanosoma cruzi, and Trypanosoma brucei, the identification of a common validated target may lead to the development of potent antikinetoplastid drugs.
Collapse
Affiliation(s)
- Andrea Ilari
- Institute of Molecular Biology and Pathology, Italian National Research Council (IBPM CNR), Department of Biochemical Sciences , Sapienza University , P.le A. Moro 5 , 00185 Rome , Italy
| | - Ilaria Genovese
- Department of Biochemical Sciences , Sapienza University , P.le A. Moro 5 , 00185 Rome , Italy
| | - Fabiana Fiorillo
- Department of Biochemical Sciences , Sapienza University , P.le A. Moro 5 , 00185 Rome , Italy
| | - Theo Battista
- Department of Biochemical Sciences , Sapienza University , P.le A. Moro 5 , 00185 Rome , Italy
| | - Ilenia De Ionna
- Department of Biochemical Sciences , Sapienza University , P.le A. Moro 5 , 00185 Rome , Italy
| | - Annarita Fiorillo
- Department of Biochemical Sciences , Sapienza University , P.le A. Moro 5 , 00185 Rome , Italy
| | - Gianni Colotti
- Institute of Molecular Biology and Pathology, Italian National Research Council (IBPM CNR), Department of Biochemical Sciences , Sapienza University , P.le A. Moro 5 , 00185 Rome , Italy
| |
Collapse
|
18
|
Tiwari N, Tanwar N, Munde M. Molecular insights into trypanothione reductase-inhibitor interaction: A structure-based review. Arch Pharm (Weinheim) 2018; 351:e1700373. [DOI: 10.1002/ardp.201700373] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 03/20/2018] [Accepted: 03/23/2018] [Indexed: 11/06/2022]
Affiliation(s)
- Neha Tiwari
- School of Physical Sciences; Jawaharlal Nehru University; New Delhi India
| | - Neetu Tanwar
- School of Physical Sciences; Jawaharlal Nehru University; New Delhi India
| | - Manoj Munde
- School of Physical Sciences; Jawaharlal Nehru University; New Delhi India
| |
Collapse
|
19
|
Charlton RL, Rossi-Bergmann B, Denny PW, Steel PG. Repurposing as a strategy for the discovery of new anti-leishmanials: the-state-of-the-art. Parasitology 2018; 145:219-236. [PMID: 28805165 PMCID: PMC5964475 DOI: 10.1017/s0031182017000993] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 05/23/2017] [Accepted: 05/25/2017] [Indexed: 12/17/2022]
Abstract
Leishmaniasis is a vector-borne neglected tropical disease caused by protozoan parasites of the genus Leishmania for which there is a paucity of effective viable non-toxic drugs. There are 1·3 million new cases each year causing considerable socio-economic hardship, best measured in 2·4 million disability adjusted life years, with greatest impact on the poorest communities, which means that desperately needed new antileishmanial treatments have to be both affordable and accessible. Established medicines with cheaper and faster development times may hold the cure for this neglected tropical disease. This concept of using old drugs for new diseases may not be novel but, with the ambitious target of controlling or eradicating tropical diseases by 2020, this strategy is still an important one. In this review, we will explore the current state-of-the-art of drug repurposing strategies in the search for new treatments for leishmaniasis.
Collapse
Affiliation(s)
- Rebecca L Charlton
- Department of Chemistry,University Science Laboratories,South Road,Durham DH1 3LE,UK
| | - Bartira Rossi-Bergmann
- Instituto de Biofísica Carlos Chagas Filho,Universidade Federal do Rio de Janeiro,Ilha do Fundão,CEP 21·949-900 Rio de Janeiro,RJ,Brazil
| | - Paul W Denny
- Department of Biosciences,University Science Laboratories,South Road,Durham DH1 3LE,UK
| | - Patrick G Steel
- Department of Chemistry,University Science Laboratories,South Road,Durham DH1 3LE,UK
| |
Collapse
|
20
|
Khan MOF. Trypanothione Reductase: A Viable Chemotherapeutic Target for Antitrypanosomal and Antileishmanial Drug Design. Drug Target Insights 2017. [DOI: 10.1177/117739280700200007] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- M. Omar F. Khan
- College of Pharmacy, Southwestern Oklahoma State University, 100 Campus Drive, Weatherford, OK 73096, U.S.A
| |
Collapse
|
21
|
Rodríguez-Becerra J, Cáceres-Jensen L, Hernández-Ramos J, Barrientos L. Identification of potential trypanothione reductase inhibitors among commercially available
$$\upbeta $$
β
-carboline derivatives using chemical space, lead-like and drug-like filters, pharmacophore models and molecular docking. Mol Divers 2017; 21:697-711. [DOI: 10.1007/s11030-017-9747-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 04/16/2017] [Indexed: 10/19/2022]
|
22
|
Abstract
In trypanosomatids, polyamine and trypanothione pathways can be considered as a whole unique metabolism, where most enzymes are essential for parasitic survival and infectivity. Leishmania parasites and all the other members of the Trypanosomatids family depend on polyamines for growth and survival: the enzymes involved in the synthesis and utilization of spermidine and trypanothione, i.e., arginase, ornithine decarboxylase, S-adenosylmethionine decarboxylase, spermidine synthase and in particular trypanothione synthetase-amidase, trypanothione reductase and tryparedoxin-dependent peroxidase are promising targets for drug development. This review deals with recent structure-based studies on these enzymes, aimed at the discovery of inhibitors of this pathway.
Collapse
|
23
|
Daunes S, Yardley V, Croft SL, D'Silva C. Antiprotozoal glutathione derivatives with flagellar membrane binding activity against T. brucei rhodesiense. Bioorg Med Chem 2016; 25:1329-1340. [PMID: 28131508 DOI: 10.1016/j.bmc.2016.12.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 12/05/2016] [Accepted: 12/10/2016] [Indexed: 12/16/2022]
Abstract
A new series of N-substituted S-(2,4-dinitrophenyl)glutathione dibutyl diesters were synthesized to improve in vitro anti-protozoal activity against the pathogenic parasites Trypanosoma brucei rhodesiense, Trypanosoma cruzi and Leishmania donovani. The results obtained indicate that N-substituents enhance the inhibitory properties of glutathione diesters whilst showing reduced toxicity against KB cells as in the cases of compounds 5, 9, 10, 16, 18 and 19. We suggest that the interaction of N-substituted S-(2,4-dinitrophenyl) glutathione dibutyl diesters with T. b. brucei occurs mainly by weak hydrophobic interactions such as London and van der Waals forces. A QSAR study indicated that the inhibitory activity of the peptide is associated negatively with the average number of C atoms, NC and positively to SZX, the ZX shadow a geometric descriptor related to molecular size and orientation of the compound. HPLC-UV studies in conjunction with optical microscopy indicate that the observed selectivity of inhibition of these compounds against bloodstream form T. b. brucei parasites in comparison to L. donovani under the same conditions is due to intracellular uptake via endocytosis in the flagellar pocket.
Collapse
Affiliation(s)
- Sylvie Daunes
- School of Chemistry and Environmental Sciences, The Manchester Metropolitan University, Faculty of Science and Engineering, John Dalton Building, Chester Street, Manchester M1 5GD, UK
| | - Vanessa Yardley
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Simon L Croft
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Claudius D'Silva
- School of Chemistry and Environmental Sciences, The Manchester Metropolitan University, Faculty of Science and Engineering, John Dalton Building, Chester Street, Manchester M1 5GD, UK; School of Chemistry, Central University of Rajasthan, Bandar Sindari, Jaipur-Ajmer Highway (Rajasthan), India.
| |
Collapse
|
24
|
Investigation of Antileishmanial Activities of Acridines Derivatives against Promastigotes and Amastigotes Form of Parasites Using Quantitative Structure Activity Relationship Analysis. ACTA ACUST UNITED AC 2016. [DOI: 10.1155/2016/5137289] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In a search of newer and potent antileishmanial (against promastigotes and amastigotes form of parasites) drug, a series of 60 variously substituted acridines derivatives were subjected to a quantitative structure activity relationship (QSAR) analysis for studying, interpreting, and predicting activities and designing new compounds by using multiple linear regression and artificial neural network (ANN) methods. The used descriptors were computed with Gaussian 03, ACD/ChemSketch, Marvin Sketch, and ChemOffice programs. The QSAR models developed were validated according to the principles set up by the Organisation for Economic Co-operation and Development (OECD). The principal component analysis (PCA) has been used to select descriptors that show a high correlation with activities. The univariate partitioning (UP) method was used to divide the dataset into training and test sets. The multiple linear regression (MLR) method showed a correlation coefficient of 0.850 and 0.814 for antileishmanial activities against promastigotes and amastigotes forms of parasites, respectively. Internal and external validations were used to determine the statistical quality of QSAR of the two MLR models. The artificial neural network (ANN) method, considering the relevant descriptors obtained from the MLR, showed a correlation coefficient of 0.933 and 0.918 with 7-3-1 and 6-3-1 ANN models architecture for antileishmanial activities against promastigotes and amastigotes forms of parasites, respectively. The applicability domain of MLR models was investigated using simple and leverage approaches to detect outliers and outsides compounds. The effects of different descriptors in the activities were described and used to study and design new compounds with higher activities compared to the existing ones.
Collapse
|
25
|
Koelewijn JM, Lutz M, Detz RJ, Reek JNH. Anode Preparation Strategies for the Electrocatalytic Oxidation of Water Based on Strong Interactions between Multiwalled Carbon Nanotubes and Cationic Acetylammonium Pyrene Moieties in Aqueous Solutions. Chempluschem 2016; 81:1098-1106. [DOI: 10.1002/cplu.201600235] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Indexed: 01/21/2023]
Affiliation(s)
- Jacobus M. Koelewijn
- Van ‘t Hoff Institute for Molecular Sciences; University of Amsterdam; Science Park 904 1098 XH Amsterdam The Netherlands
| | - Martin Lutz
- Crystal and Structural Chemistry Bijvoet Center for Biomolecular Research; Utrecht University; Padualaan 8 3584 CH Utrecht The Netherlands
| | - Remko J. Detz
- Van ‘t Hoff Institute for Molecular Sciences; University of Amsterdam; Science Park 904 1098 XH Amsterdam The Netherlands
| | - Joost N. H. Reek
- Van ‘t Hoff Institute for Molecular Sciences; University of Amsterdam; Science Park 904 1098 XH Amsterdam The Netherlands
| |
Collapse
|
26
|
Wu S, Mao G, Kirsebom LA. Inhibition of Bacterial RNase P RNA by Phenothiazine Derivatives. Biomolecules 2016; 6:biom6030038. [PMID: 27618117 PMCID: PMC5039424 DOI: 10.3390/biom6030038] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 08/24/2016] [Accepted: 08/26/2016] [Indexed: 12/19/2022] Open
Abstract
There is a need to identify novel scaffolds and targets to develop new antibiotics. Methylene blue is a phenothiazine derivative, and it has been shown to possess anti-malarial and anti-trypanosomal activities. Here, we show that different phenothiazine derivatives and pyronine G inhibited the activities of three structurally different bacterial RNase P RNAs (RPRs), including that from Mycobacterium tuberculosis, with Ki values in the lower μM range. Interestingly, three antipsychotic phenothiazines (chlorpromazine, thioridazine, and trifluoperazine), which are known to have antibacterial activities, also inhibited the activity of bacterial RPRs, albeit with higher Ki values than methylene blue. Phenothiazines also affected lead(II)-induced cleavage of bacterial RPR and inhibited yeast tRNA(Phe), indicating binding of these drugs to functionally important regions. Collectively, our findings provide the first experimental data showing that long, noncoding RNAs could be targeted by different phenothiazine derivatives.
Collapse
Affiliation(s)
- Shiying Wu
- Department of Cell and Molecular Biology, Box 596, Biomedical Centre, Uppsala SE-751 24, Sweden.
| | - Guanzhong Mao
- Department of Cell and Molecular Biology, Box 596, Biomedical Centre, Uppsala SE-751 24, Sweden.
| | - Leif A Kirsebom
- Department of Cell and Molecular Biology, Box 596, Biomedical Centre, Uppsala SE-751 24, Sweden.
| |
Collapse
|
27
|
Mechanistic insight into thermal 1,3- and 1,5-sulfonyl migrations of N-arenesulfonylphenothiazines and N-arenesulfonylphenoxazines. MONATSHEFTE FUR CHEMIE 2016. [DOI: 10.1007/s00706-016-1661-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
28
|
Argüelles AJ, Cordell GA, Maruenda H. Molecular Docking and Binding Mode Analysis of Plant Alkaloids as in Vitro and in silico Inhibitors of Trypanothione Reductase from Trypanosoma cruzi. Nat Prod Commun 2016. [DOI: 10.1177/1934578x1601100118] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Trypanothione reductase (TryR) is a key enzyme in the metabolism of Trypanosoma cruzi, the parasite responsible for Chagas disease. The available repertoire of TryR inhibitors relies heavily on synthetic substrates of limited structural diversity, and less on plant-derived natural products. In this study, a molecular docking procedure using a Lamarckian Genetic Algorithm was implemented to examine the protein-ligand binding interactions of strong in vitro inhibitors for which no X-ray data is available. In addition, a small, skeletally diverse, set of natural alkaloids was assessed computationally against T. cruzi TryR in search of new scaffolds for lead development. The preferential binding mode (low number of clusters, high cluster population), together with the deduced binding interactions were used to discriminate among the virtual inhibitors. This study confirms the prior in vitro data and proposes quebrachamine, cephalotaxine, cryptolepine, (22 S,25 S)-tomatidine, (22 R,25 S)-solanidine, and (22 R,25 R)-solasodine as new alkaloid scaffold leads in the search for more potent and selective TryR inhibitors.
Collapse
Affiliation(s)
- Alonso J. Argüelles
- Pontificia Universidad Católica del Perú, Departamento de Ciencias - Sección Química, Lima, Perú
| | - Geoffrey A. Cordell
- Pontificia Universidad Católica del Perú, Departamento de Ciencias - Sección Química, Lima, Perú
- Natural Products Inc., Evanston, IL 60203, USA
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Helena Maruenda
- Pontificia Universidad Católica del Perú, Departamento de Ciencias - Sección Química, Lima, Perú
| |
Collapse
|
29
|
Tang J, Xu B, Mao X, Yang H, Wang X, Lv X. One-Pot Synthesis of Pyrrolo[3,2,1-kl]phenothiazines through Copper-Catalyzed Tandem Coupling/Double Cyclization Reaction. J Org Chem 2015; 80:11108-14. [DOI: 10.1021/acs.joc.5b01745] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Jiaming Tang
- Department of Chemistry,
College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua 321004, People’s Republic of China
| | - Bingqing Xu
- Department of Chemistry,
College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua 321004, People’s Republic of China
| | - Xi Mao
- Department of Chemistry,
College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua 321004, People’s Republic of China
| | - Hongyan Yang
- Department of Chemistry,
College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua 321004, People’s Republic of China
| | - Xiaoxia Wang
- Department of Chemistry,
College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua 321004, People’s Republic of China
| | - Xin Lv
- Department of Chemistry,
College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua 321004, People’s Republic of China
| |
Collapse
|
30
|
O’Sullivan MC, Durham TB, Valdes HE, Dauer KL, Karney NJ, Forrestel AC, Bacchi CJ, Baker JF. Dibenzosuberyl substituted polyamines and analogs of clomipramine as effective inhibitors of trypanothione reductase; molecular docking, and assessment of trypanocidal activities. Bioorg Med Chem 2015; 23:996-1010. [DOI: 10.1016/j.bmc.2015.01.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 01/04/2015] [Accepted: 01/09/2015] [Indexed: 12/15/2022]
|
31
|
Sánchez-Murcia PA, Ruiz-Santaquiteria M, Toro MA, de Lucio H, Jiménez MÁ, Gago F, Jiménez-Ruiz A, Camarasa MJ, Velázquez S. Comparison of hydrocarbon-and lactam-bridged cyclic peptides as dimerization inhibitors of Leishmania infantum trypanothione reductase. RSC Adv 2015. [DOI: 10.1039/c5ra06853c] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Helical peptides stabilizedviaall-hydrocarbon or lactam side-chain bridging were investigated as disruptors ofLeishmania infantumtrypanothione reductase.
Collapse
Affiliation(s)
| | | | - Miguel A. Toro
- Departamento de Biología de Sistemas
- Universidad de Alcalá
- Madrid
- Spain
| | - Héctor de Lucio
- Departamento de Biología de Sistemas
- Universidad de Alcalá
- Madrid
- Spain
| | | | - Federico Gago
- Departamento de Ciencias Biomédicas
- Unidad Asociada al CSIC
- Universidad de Alcalá
- Madrid
- Spain
| | | | | | | |
Collapse
|
32
|
Saha D, Sharma A. Docking-based screening of natural product database in quest for dual site inhibitors of Trypanosoma cruzi trypanothione reductase (TcTR). Med Chem Res 2014. [DOI: 10.1007/s00044-014-1122-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
33
|
Persch E, Bryson S, Todoroff NK, Eberle C, Thelemann J, Dirdjaja N, Kaiser M, Weber M, Derbani H, Brun R, Schneider G, Pai EF, Krauth-Siegel RL, Diederich F. Binding to large enzyme pockets: small-molecule inhibitors of trypanothione reductase. ChemMedChem 2014; 9:1880-91. [PMID: 24788386 DOI: 10.1002/cmdc.201402032] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Indexed: 01/16/2023]
Abstract
The causative agents of the parasitic disease human African trypanosomiasis belong to the family of trypanosomatids. These parasitic protozoa exhibit a unique thiol redox metabolism that is based on the flavoenzyme trypanothione reductase (TR). TR was identified as a potential drug target and features a large active site that allows a multitude of possible ligand orientations, which renders rational structure-based inhibitor design highly challenging. Herein we describe the synthesis, binding properties, and kinetic analysis of a new series of small-molecule inhibitors of TR. The conjunction of biological activities, mutation studies, and virtual ligand docking simulations led to the prediction of a binding mode that was confirmed by crystal structure analysis. The crystal structures revealed that the ligands bind to the hydrophobic wall of the so-called "mepacrine binding site". The binding conformation and potency of the inhibitors varied for TR from Trypanosoma brucei and T. cruzi.
Collapse
Affiliation(s)
- Elke Persch
- Laboratorium für Organische Chemie, ETH Zürich, Vladimir-Prelog-Weg 3, 8093 Zurich (Switzerland)
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
The potential of flavoproteins as targets of pharmacological treatments is immense. In this review we present an overview of the current research progress on medical interventions based on flavoproteins with a special emphasis on cancer, infectious diseases, and neurological disorders.
Collapse
Affiliation(s)
- Esther Jortzik
- Interdisciplinary Research Center, Justus Liebig University, Giessen, Germany
| | | | | | | |
Collapse
|
35
|
The receptor-dependent LQTA-QSAR: application to a set of trypanothione reductase inhibitors. J Comput Aided Mol Des 2012; 26:1055-65. [PMID: 22972559 DOI: 10.1007/s10822-012-9598-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 08/29/2012] [Indexed: 11/27/2022]
Abstract
A new Receptor-Dependent LQTA-QSAR approach, RD-LQTA-QSAR, is proposed as a new 4D-QSAR method. It is an evolution of receptor independent LQTA-QSAR. This approach uses the free GROMACS package to carry out molecular dynamics simulations and generates a conformational ensemble profile for each compound. Such an ensemble is used to build molecular interaction field-based QSAR models, as in CoMFA. To show the potential of this methodology, a set of 38 phenothiazine derivatives that are specific competitive T. cruzi trypanothione reductase inhibitors, was chosen. Using a combination of molecular docking and molecular dynamics simulations, the binding mode of the phenotiazine derivatives was evaluated in a simulated induced fit approach. The ligands alignments were performed using both ligand and binding site atoms, enabling unbiased alignment. The models obtained were extensively validated by leave-N-out cross-validation and y-randomization techniques to test for their robustness and absence of chance correlation. The final model presented Q(2) LOO of 0.87 and R² of 0.92 and a suitable external prediction of [Formula: see text]= 0.78. The adapted binding site obtained is useful to perform virtual screening and ligand structure-based design and the descriptors in the final model can aid in the design new inhibitors.
Collapse
|
36
|
Abstract
SIGNIFICANCE Parasitic diseases affect hundreds of millions of people worldwide and represent major health problems. Treatment is becoming extremely difficult due to the emergence of drug resistance, the absence of effective vaccines, and the spread of insecticide-resistant vectors. Thus, identification of affordable and readily available drugs against resistant parasites is of global demand. RECENT ADVANCES Susceptibility of many parasites to oxidative stress is a well-known phenomenon. Therefore, generation of reactive oxygen species (ROS) or inhibition of endogenous antioxidant enzymes would be a novel therapeutic approach to develop antiparasitic drugs. This article highlights the unique metabolic pathways along with redox enzymes of unicellular (Plasmodium falciparum, Trypanosoma cruzi, Trypanosoma brucei, Leishmania donovani, Entamoeba histolytica, and Trichomonas vaginalis) and multicellular parasites (Schistosoma mansoni), which could be utilized to promote ROS-mediated toxicity. CRITICAL ISSUES Enzymes involved in various vital redox reactions could be potential targets for drug development. FUTURE DIRECTIONS The identification of redox-active antiparasitic drugs along with their mode of action will help researchers around the world in designing novel drugs in the future.
Collapse
Affiliation(s)
- Chinmay Pal
- Department of Infectious Diseases and Immunology, Indian Institute of Chemical Biology, Kolkata, India
| | | |
Collapse
|
37
|
Da Rocha Pita SS, Batista PR, Albuquerque MG, Pascutti PG. Molecular Dynamics Simulations of Peptide Inhibitors Complexed WithTrypanosoma cruziTrypanothione Reductase. Chem Biol Drug Des 2012; 80:561-71. [DOI: 10.1111/j.1747-0285.2012.01429.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
38
|
Alptuzun V, Cakiroglu G, Limoncu ME, Erac B, Hosgor-Limoncu M, Erciyas E. Synthesis and antileishmanial activity of novel pyridinium-hydrazone derivatives. J Enzyme Inhib Med Chem 2012; 28:960-7. [DOI: 10.3109/14756366.2012.697058] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Vildan Alptuzun
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ege University,
Bornova, Izmir, Turkey
| | - Gokcer Cakiroglu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ege University,
Bornova, Izmir, Turkey
| | - M. Emin Limoncu
- Vocational School of Health Services, Celal Bayar University,
Manisa, Turkey
| | - Bayri Erac
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Ege University,
Bornova, Izmir, Turkey
| | - Mine Hosgor-Limoncu
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Ege University,
Bornova, Izmir, Turkey
| | - Ercin Erciyas
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ege University,
Bornova, Izmir, Turkey
| |
Collapse
|
39
|
Venkatesan SK, Dubey VK. Footprinting of inhibitor interactions of in silico identified inhibitors of trypanothione reductase of Leishmania parasite. ScientificWorldJournal 2012; 2012:963658. [PMID: 22550471 PMCID: PMC3322522 DOI: 10.1100/2012/963658] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 11/02/2011] [Indexed: 11/17/2022] Open
Abstract
Structure-based virtual screening of NCI Diversity set II compounds was performed to indentify novel inhibitor scaffolds of trypanothione reductase (TR) from Leishmania infantum. The top 50 ranked hits were clustered using the AuPoSOM tool. Majority of the top-ranked compounds were Tricyclic. Clustering of hits yielded four major clusters each comprising varying number of subclusters differing in their mode of binding and orientation in the active site. Moreover, for the first time, we report selected alkaloids and dibenzothiazepines as inhibitors of Leishmania infantum TR. The mode of binding observed among the clusters also potentiates the probable in vitro inhibition kinetics and aids in defining key interaction which might contribute to the inhibition of enzymatic reduction of T[S] 2. The method provides scope for automation and integration into the virtual screening process employing docking softwares, for clustering the small molecule inhibitors based upon protein-ligand interactions.
Collapse
Affiliation(s)
- Santhosh K Venkatesan
- Department of Biotechnology, Indian Institute of Technology Guwahati, Assam 781039, India
| | | |
Collapse
|
40
|
da Rocha Pita SS, Albuquerque MG, Rodrigues CR, Castro HC, Hopfinger AJ. Receptor-dependent 4D-QSAR analysis of peptidemimetic inhibitors of Trypanosoma cruzi trypanothione reductase with receptor-based alignment. Chem Biol Drug Des 2012; 79:740-8. [PMID: 22269140 DOI: 10.1111/j.1747-0285.2012.01338.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Receptor-dependent four-dimensional quantitative structure-activity relationship (RD-4D-QSAR) studies were applied on a series of 21 peptides reversible inhibitors of Trypanosoma cruzi trypanothione reductase (TR) (Amino Acids, 20, 2001, 145). The RD-4D-QSAR (J Chem Inform Comp Sci, 43, 2003, 1591) approach can evaluate multiple conformations from molecular dynamics simulation and several superposition structure alignments inside a box composed by unitary cubic cells. The descriptors are the occupancy frequency of the atoms types inside the grid cells. We could develop 3D-QSAR models that were highly predictive (q(2) above 0.71). The 3D-QSAR models can be visualized as a spatial map of atom types that are important on the comprehension of the ligand-enzyme interaction mechanism, pointing main pharmacophoric groups and TR subsites described in the literature. We were able also to identify some TR subsites for further development in the drug discovery process against tropical diseases not yet studied.
Collapse
|
41
|
Duyzend MH, Clark CT, Simmons SL, Johnson WB, Larson AM, Leconte AM, Wills AW, Ginder-Vogel M, Wilhelm AK, Czechowicz JA, Alberg DG. Synthesis and evaluation of substrate analogue inhibitors of trypanothione reductase. J Enzyme Inhib Med Chem 2011; 27:784-94. [PMID: 22085139 DOI: 10.3109/14756366.2011.604319] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Trypanothione reductase (TR) is found in the trypanosomatid parasites, where it catalyses the NADPH-dependent reduction of the glutathione analogue, trypanothione, and is a key player in the parasite's defenses against oxidative stress. TR is a promising target for the development of antitrypanosomal drugs; here, we report our synthesis and evaluation of compounds 3-5 as low micromolar Trypanosoma cruzi TR inhibitors. Although 4 and 5 were designed as potential irreversible inhibitors, these compounds, as well as 3, displayed reversible competitive inhibition. Compound 3 proved to be the most potent inhibitor, with a K(i) = 2 µM.
Collapse
Affiliation(s)
- Michael H Duyzend
- Department of Chemistry, Carleton College, Northfield, MN 55057, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Patterson S, Alphey MS, Jones DC, Shanks EJ, Street IP, Frearson JA, Wyatt PG, Gilbert IH, Fairlamb AH. Dihydroquinazolines as a novel class of Trypanosoma brucei trypanothione reductase inhibitors: discovery, synthesis, and characterization of their binding mode by protein crystallography. J Med Chem 2011; 54:6514-30. [PMID: 21851087 PMCID: PMC3188286 DOI: 10.1021/jm200312v] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Trypanothione reductase (TryR) is a genetically validated drug target in the parasite Trypanosoma brucei , the causative agent of human African trypanosomiasis. Here we report the discovery, synthesis, and development of a novel series of TryR inhibitors based on a 3,4-dihydroquinazoline scaffold. In addition, a high resolution crystal structure of TryR, alone and in complex with substrates and inhibitors from this series, is presented. This represents the first report of a high resolution complex between a noncovalent ligand and this enzyme. Structural studies revealed that upon ligand binding the enzyme undergoes a conformational change to create a new subpocket which is occupied by an aryl group on the ligand. Therefore, the inhibitor, in effect, creates its own small binding pocket within the otherwise large, solvent exposed active site. The TryR-ligand structure was subsequently used to guide the synthesis of inhibitors, including analogues that challenged the induced subpocket. This resulted in the development of inhibitors with improved potency against both TryR and T. brucei parasites in a whole cell assay.
Collapse
Affiliation(s)
- Stephen Patterson
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee , Dow Street, Dundee DD1 5EH, U.K
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Aguilera-Venegas B, Olea-Azar C, Norambuena E, Arán VJ, Mendizábal F, Lapier M, Maya JD, Kemmerling U, López-Muñoz R. ESR, electrochemical, molecular modeling and biological evaluation of 4-substituted and 1,4-disubstituted 7-nitroquinoxalin-2-ones as potential anti-Trypanosoma cruzi agents. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2011; 78:1004-1012. [PMID: 21239218 DOI: 10.1016/j.saa.2010.12.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 10/28/2010] [Accepted: 12/09/2010] [Indexed: 05/30/2023]
Abstract
Electrochemical and ESR studies were carried out in this work with the aim of characterizing the reduction mechanisms of 4-substituted and 1,4-disubstituted 7-nitroquinoxalin-2-ones by means of cyclic voltammetry in DMSO as aprotic solvent. Two reduction mechanisms were found for these compounds: the first, for compounds bearing a labile hydrogen by following a self-protonation mechanism (ECE steps), and the second, for compounds without labile hydrogen, based on a purely electrochemical reduction mechanism (typical of nitroheterocycles). The electrochemical results were corroborated using ESR spectroscopy allowing us to propose the hyperfine splitting pattern of the nitro-radical, which was later corroborated by the ESR simulation spectra. All these compounds were assayed as growth inhibitors against Trypanosoma cruzi: first, on the non-proliferative (and infective) form of the parasite (trypomastigote stage), and then, the ones that displayed activity, were assayed on the non-infective form (epimastigote stage). Thus, we found four new compounds highly active against T. cruzi. Finally, molecular modeling studies suggest the inhibition of the trypanothione reductase like one of the possible mechanisms involved in the trypanocidal action.
Collapse
Affiliation(s)
- Benjamín Aguilera-Venegas
- Departamento de Química Inorgánica y Analítica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Venkatesan SK, Shukla AK, Dubey VK. Molecular docking studies of selected tricyclic and quinone derivatives on trypanothione reductase of Leishmania infantum. J Comput Chem 2011; 31:2463-75. [PMID: 20340105 DOI: 10.1002/jcc.21538] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Visceral leishmaniasis, most lethal form of Leishmaniasis, is caused by Leishmania infantum in the Old world. Current therapeutics for the disease is associated with a risk of high toxicity and development of drug resistant strains. Thiol-redox metabolism involving trypanothione and trypanothione reductase, key for survival of Leishmania, is a validated target for rational drug design. Recently published structure of trypanothione reductase (TryR) from L. infantum, in oxidized and reduced form along with Sb(III), provides vital clues on active site of the enzyme. In continuation with our attempts to identify potent inhibitors of TryR, we have modeled binding modes of selected tricyclic compounds and quinone derivatives, using AutoDock4. Here, we report a unique binding mode for quinone derivatives and 9-aminoacridine derivatives, at the FAD binding domain. A conserved hydrogen bonding pattern was observed in all these compounds with residues Thr335, Lys60, His461. With the fact that these residues aid in the orientation of FAD towards the active site forming the core of the FAD binding domain, designing selective and potent compounds that could replace FAD in vivo during the synthesis of Trypanothione reductase can be deployed as an effective strategy in designing new drugs towards Leishmaniasis. We also report the binding of Phenothiazine and 9-aminoacridine derivatives at the Z site of the protein. The biological significance and possible mode of inhibition by quinone derivatives, which binds to FAD binding domain, along with other compounds are discussed.
Collapse
|
45
|
|
46
|
Eberle C, Lauber BS, Fankhauser D, Kaiser M, Brun R, Krauth-Siegel RL, Diederich F. Improved Inhibitors of Trypanothione Reductase by Combination of Motifs: Synthesis, Inhibitory Potency, Binding Mode, and Antiprotozoal Activities. ChemMedChem 2010; 6:292-301. [DOI: 10.1002/cmdc.201000420] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2010] [Indexed: 11/05/2022]
|
47
|
Urbina JA. Specific chemotherapy of Chagas disease: relevance, current limitations and new approaches. Acta Trop 2010; 115:55-68. [PMID: 19900395 DOI: 10.1016/j.actatropica.2009.10.023] [Citation(s) in RCA: 326] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2009] [Revised: 10/22/2009] [Accepted: 10/26/2009] [Indexed: 01/31/2023]
Abstract
A critical review of the development of specific chemotherapeutic approaches for the management of American Trypanosomiasis or Chagas disease is presented, including controversies on the pathogenesis of the disease, the initial efforts that led to the development of currently available drugs (nifurtimox and benznidazole), limitations of these therapies and novel approaches for the development of anti-Trypanosoma cruzi drugs, based on our growing understanding of the biology of this parasite. Among the later, the most promising approaches are ergosterol biosynthesis inhibitors such as posaconazole and ravuconazole, poised to enter clinical trials for chronic Chagas disease in the short term; inhibitors of cruzipain, the main cysteine protease of T. cruzi, essential for its survival and proliferation in vitro and in vivo; bisphosphonates, metabolic stable pyrophosphate analogs that have trypanocidal activity through the inhibition of the parasite's farnesyl-pyrophosphate synthase or hexokinase; inhibitors of trypanothione synthesis and redox metabolism and inhibitors of hypoxanthine-guanine phosphoribosyl-transferase, an essential enzyme for purine salvage in T. cruzi and related organisms. Finally, the economic and political challenges faced by development of drugs for the treatment of neglected tropical diseases, which afflict almost exclusively poor populations in developing countries, are analyzed and recent potential solutions for this conundrum are discussed.
Collapse
|
48
|
Pandey VP, Bisht SS, Mishra M, Kumar A, Siddiqi MI, Verma A, Mittal M, Sane SA, Gupta S, Tripathi RP. Synthesis and molecular docking studies of 1-phenyl-4-glycosyl-dihydropyridines as potent antileishmanial agents. Eur J Med Chem 2010; 45:2381-8. [DOI: 10.1016/j.ejmech.2010.02.018] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Revised: 02/03/2010] [Accepted: 02/04/2010] [Indexed: 11/27/2022]
|
49
|
Spinks D, Shanks EJ, Cleghorn LAT, McElroy S, Jones D, James D, Fairlamb AH, Frearson JA, Wyatt PG, Gilbert IH. Investigation of trypanothione reductase as a drug target in Trypanosoma brucei. ChemMedChem 2010; 4:2060-9. [PMID: 19924760 PMCID: PMC2855869 DOI: 10.1002/cmdc.200900262] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
There is an urgent need for new drugs for the treatment of tropical parasitic diseases such as human African trypanosomiasis, which is caused by Trypanosoma brucei. The enzyme trypanothione reductase (TryR) is a potential drug target within these organisms. Herein we report the screening of a 62,000 compound library against T. brucei TryR. Further work was undertaken to optimise potency and selectivity of two novel-compound series arising from the enzymatic and whole parasite screens and mammalian cell counterscreens. Both of these series, containing either a quinoline or pyrimidinopyrazine scaffold, yielded low micromolar inhibitors of the enzyme and growth of the parasite. The challenges of inhibiting TryR with druglike molecules is discussed.
Collapse
Affiliation(s)
- Daniel Spinks
- Drug Discovery Unit, College of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Assaying phenothiazine derivatives as trypanothione reductase and glutathione reductase inhibitors by theoretical docking and Molecular Dynamics studies. J Mol Graph Model 2009; 28:371-81. [DOI: 10.1016/j.jmgm.2009.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Revised: 07/31/2009] [Accepted: 09/15/2009] [Indexed: 11/22/2022]
|