1
|
de Oliveira GB, Santos Costa ÉC, Severina do Monte Z, de Almeida GC, da Silva Falcão EP, Scotti L, Tullius Scotti M, Oliveira Silva R, de Sousa Oliveira DS, Ademar Sales Junior P, Alves Pereira VR, José de Melo S. Structure-based Virtual Screening and Drug Design Development of Leishmanicidal Pyrimidines. Chem Biodivers 2025:e202402881. [PMID: 39814686 DOI: 10.1002/cbdv.202402881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 01/18/2025]
Abstract
Leishmaniasis is a neglected disease caused by parasites of the genus Leishmania sp. that causes approximately 1 million cases and 650,000 deaths annually worldwide. Its treatment has several limitations mainly due to high toxicity and clinical resistance, and the search for alternatives is highly desirable. The present work aimed to design new antileishmanial compounds through a virtual screening of a small in-house library of pyrimidine compounds, never tested against Leishmania, using the active site of trypanothione reductase (TR) as a target model. The compounds showed favorable affinity with the amino acid residues of the active site of TR. Pyr 1-9 were synthesized and tested against Leishmania amazonensis strain. Four derivatives demonstrated activity against promastigote (IC50 value between 11.23 and 91.5 µM) and three other compounds demonstrated discreet activity against amastigote, IC50 value between 81.29 and 153.21 µM. Based on the results obtained in the screening, three new pyrimidines Pyr 10-12 were designed to optimize activity, cytotoxicity, and selectivity. Pyr 10 and Pyr 11 demonstrated good activity against promastigotes, with IC50 of 11.38 ± 9.7 and 20.01 ± 13.55 µM, respectively, and improved cytotoxicity and selectivity. No activity was obtained against amastigotes. Thus, this study contributes important information for the development of new pyrimidines active against Leishmania.
Collapse
Affiliation(s)
- Gerliny Bezerra de Oliveira
- Department of Pharmaceutical Sciences, Postgraduate Program in Pharmaceutical Sciences, Federal University of Pernambuco/UFPE, Recife, Brazil
| | - Érick Caique Santos Costa
- Department of Biosciences, Postgraduate Program in Biological Sciences, Federal University of Pernambuco/UFPE, Recife, Brazil
| | - Zenaide Severina do Monte
- Department of Pharmaceutical Sciences, Postgraduate Program in Pharmaceutical Sciences, Federal University of Pernambuco/UFPE, Recife, Brazil
| | - Gleybson Correia de Almeida
- Department of Pharmaceutical Sciences, Postgraduate Program in Pharmaceutical Sciences, Federal University of Pernambuco/UFPE, Recife, Brazil
| | | | - Luciana Scotti
- Department of Chemistry, Health Sciences Center, Laboratory of Cheminformatics, Program of Natural and Synthetic Bioactive Products (PgPNSB), Federal University of Paraiba, Joao Pessoa, Brazil
| | - Marcus Tullius Scotti
- Department of Chemistry, Health Sciences Center, Laboratory of Cheminformatics, Program of Natural and Synthetic Bioactive Products (PgPNSB), Federal University of Paraiba, Joao Pessoa, Brazil
| | | | - Daniele Santana de Sousa Oliveira
- Department of Immunology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Federal University of Pernambuco, Recife, Brazil
| | - Policarpo Ademar Sales Junior
- Department of Immunology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Federal University of Pernambuco, Recife, Brazil
| | - Valéria Rêgo Alves Pereira
- Department of Immunology, Aggeu Magalhães Research Center, Oswaldo Cruz Foundation, Federal University of Pernambuco, Recife, Brazil
| | - Sebastião José de Melo
- Department of Pharmaceutical Sciences, Postgraduate Program in Pharmaceutical Sciences, Federal University of Pernambuco/UFPE, Recife, Brazil
- Department of Biosciences, Postgraduate Program in Biological Sciences, Federal University of Pernambuco/UFPE, Recife, Brazil
| |
Collapse
|
2
|
Giraudo A, Bolchi C, Pallavicini M, Di Santo R, Costi R, Saccoliti F. Uncovering the Mechanism of Action of Antiprotozoal Agents: A Survey on Photoaffinity Labeling Strategy. Pharmaceuticals (Basel) 2024; 18:28. [PMID: 39861091 PMCID: PMC11768348 DOI: 10.3390/ph18010028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
Plasmodium, Leishmania, and Trypanosoma parasites are responsible for infectious diseases threatening millions of people worldwide. Despite more recent efforts devoted to the search for new antiprotozoal agents, efficacy, safety, and resistance issues still hinder the development of suited therapeutic options. The lack of robustly validated targets and the complexity of parasite's diseases have made phenotypic screening a preferential drug discovery strategy for the identification of new chemical entities. However, via this approach, no information on biological target(s) and mechanisms of action of compounds are provided. Among the target deconvolution strategies useful to fill this gap, photoaffinity labeling (PAL) has emerged as one of most suited to enable investigation in a complex cellular environment. More recently, PAL has been exploited to unravel the molecular basis of bioactive compounds' function in live parasites, allowing elucidation of the mechanism of action of both approved drugs and new chemical entities. Besides highlighting new potential drug targets, PAL can provide valuable information on efficacy and liabilities of small molecules at the molecular level, which could be exploited to greatly facilitate the rational optimization of compounds in terms of potency and safety. In this review, we will report the most recent studies that have leveraged PAL to disclose the biological targets and mechanism of action of phenotypically active compounds targeting kinetoplastid diseases (i.e., human African trypanosomiasis, leishmaniasis, and Chagas disease) and malaria. Moreover, we will comment on potential perspectives that this innovative approach can provide in aiding the discovery and development of new antiprotozoal drugs.
Collapse
Affiliation(s)
- Alessandro Giraudo
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, I-20133 Milano, Italy
| | - Cristiano Bolchi
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, I-20133 Milano, Italy
| | - Marco Pallavicini
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, I-20133 Milano, Italy
| | - Roberto Di Santo
- Dipartimento di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, “Sapienza” Università di Roma, p.le Aldo Moro 5, I-00185 Rome, Italy
| | - Roberta Costi
- Dipartimento di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, “Sapienza” Università di Roma, p.le Aldo Moro 5, I-00185 Rome, Italy
| | - Francesco Saccoliti
- Dipartimento di Scienze della Vita, della Salute e delle Professioni Sanitarie, Università degli Studi “Link Campus University”, Via del Casale di S. Pio V 44, I-00165 Rome, Italy
| |
Collapse
|
3
|
Bhattacharya I, Pyne N, Paul S. In vitro and in silico approaches manifest the anti-leishmanial activity of wild edible mushroom Amanita princeps. In Silico Pharmacol 2024; 13:6. [PMID: 39726904 PMCID: PMC11668711 DOI: 10.1007/s40203-024-00287-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 11/09/2024] [Indexed: 12/28/2024] Open
Abstract
Visceral Leishmaniasis, caused by Leishmania donovani, is the second most deadly parasitic disease, causing over 65,000 deaths annually. Synthetic drugs available in the market, to combat this disease, have numerous side effects. In this backdrop, we aim to find safer antileishmanial alternatives with minimal side effects from mushrooms, which harbour various secondary metabolites with promising efficacy. Robust screening of sixteen extracts from eight different wild mushrooms reveals that the hydroalcoholic extract of Amanita princeps has outstanding antileishmanial activity against Leishmania donovani. Metabolomic profiling of this lead extract identifies 50 bioactive mycocompounds and among them, 10 were selected for in-silico study against five major targets-arginase, spermidine synthase, ornithine decarboxylase, trypanothione reductase and SOD, crucial for thiol-redox balance in parasites in the polyamine synthesis pathway. Molecular docking analysis against our prioritised targets identified two mycompounds Ergosterol and Taraxacolide 1-O-b-D-glucopyranoside from Amanita princeps having the highest binding affinity of -15.8 and -11.8 kcal/mol respectively against the ornithine decarboxylase of polyamine synthesis pathway. However, MD simulations and free energy calculation using MM-GBSA analysis revealed the better stability of ergosterol with PASP receptors suggesting its promising role as an anti-leishmanial compound. Further results of in vitro arginase, SOD, and NO enzyme assays also corroborated with in-silico findings, reinforcing the anti-leishmanial efficacy of the Amanita princeps extract. Thus, both in silico and in vitro analyses suggest the efficacy of both Ergosterol and Taraxacolide 1-O-b-D-glucopyranoside compounds resourced from Amanita princeps as potent antileishmanial agents. Graphical abstract
Collapse
Affiliation(s)
- Ishita Bhattacharya
- Laboratory of Cell and Molecular Biology, Department of Botany, Centre of Advanced Study, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019 India
| | - Nibedita Pyne
- Laboratory of Cell and Molecular Biology, Department of Botany, Centre of Advanced Study, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019 India
| | - Santanu Paul
- Laboratory of Cell and Molecular Biology, Department of Botany, Centre of Advanced Study, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019 India
| |
Collapse
|
4
|
Nketia PB, Gasu EN, Mensah JO, Borquaye LS. In silico identification of α-bisabolol and letestuianin C as potential inhibitors of Trypanosoma brucei trypanothione reductase. J Biomol Struct Dyn 2024; 42:8660-8672. [PMID: 37584491 DOI: 10.1080/07391102.2023.2247084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/05/2023] [Indexed: 08/17/2023]
Abstract
Despite the recent advances in drug research, finding a safe, effective, and easy to use chemotherapy for human African trypanosomiasis (HAT) remains a challenging task. Trypanosomatids have developed resistance mechanisms towards melarsoprol (the current drug of choice), and the fact that it is poisonous is problematic. Therefore, a search for alternative therapeutics against the parasite is urgently needed. Natural products offer potential for drug discovery, but little or nothing is known about the target of inhibition or possible mode of inhibition. Therefore, this study aimed to use molecular docking and molecular dynamics simulations to evaluate 30 antitrypanosomal natural products as potential inhibitors of trypanothione reductase, a key protein necessary for the survival of the Trypanosoma brucei. The study also assessed the pharmacokinetic properties of the most promising compounds. Of the compounds evaluated, α-bisabolol, letestuianin C, waltherione, and mexicanin E were found to bind at the active site of TR and interact with Met115, Tyr112, and Trp23, which are essential for enzyme functioning. Molecular dynamic simulations revealed the sustained binding of α-bisabolol and letestuianin C throughout the simulation period, potentially obstructing the binding of the substrate (T[S]2) and impeding catalysis. The binding of these compounds to TR led to the presence of solvent molecules in the enzyme's active site, and this could potentially lead to protein aggregation. Furthermore, α-bisabolol and letestuianin C exhibited promising safety profiles. Consequently, α-bisabolol and letestuianin C have been shown to be viable candidates for targeting trypanothione reductase in the fight against human African trypanosomiasis.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Prisca Baah Nketia
- Department of Chemistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Edward Ntim Gasu
- Department of Chemistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Central Laboratory, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | | | - Lawrence Sheringham Borquaye
- Department of Chemistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Central Laboratory, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| |
Collapse
|
5
|
González-Montero MC, Andrés-Rodríguez J, García-Fernández N, Pérez-Pertejo Y, Reguera RM, Balaña-Fouce R, García-Estrada C. Targeting Trypanothione Metabolism in Trypanosomatids. Molecules 2024; 29:2214. [PMID: 38792079 PMCID: PMC11124245 DOI: 10.3390/molecules29102214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Infectious diseases caused by trypanosomatids, including African trypanosomiasis (sleeping sickness), Chagas disease, and different forms of leishmaniasis, are Neglected Tropical Diseases affecting millions of people worldwide, mainly in vulnerable territories of tropical and subtropical areas. In general, current treatments against these diseases are old-fashioned, showing adverse effects and loss of efficacy due to misuse or overuse, thus leading to the emergence of resistance. For these reasons, searching for new antitrypanosomatid drugs has become an urgent necessity, and different metabolic pathways have been studied as potential drug targets against these parasites. Considering that trypanosomatids possess a unique redox pathway based on the trypanothione molecule absent in the mammalian host, the key enzymes involved in trypanothione metabolism, trypanothione reductase and trypanothione synthetase, have been studied in detail as druggable targets. In this review, we summarize some of the recent findings on the molecules inhibiting these two essential enzymes for Trypanosoma and Leishmania viability.
Collapse
Affiliation(s)
- María-Cristina González-Montero
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (M.-C.G.-M.); (J.A.-R.); (N.G.-F.); (Y.P.-P.); (R.M.R.)
| | - Julia Andrés-Rodríguez
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (M.-C.G.-M.); (J.A.-R.); (N.G.-F.); (Y.P.-P.); (R.M.R.)
| | - Nerea García-Fernández
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (M.-C.G.-M.); (J.A.-R.); (N.G.-F.); (Y.P.-P.); (R.M.R.)
| | - Yolanda Pérez-Pertejo
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (M.-C.G.-M.); (J.A.-R.); (N.G.-F.); (Y.P.-P.); (R.M.R.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Rosa M. Reguera
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (M.-C.G.-M.); (J.A.-R.); (N.G.-F.); (Y.P.-P.); (R.M.R.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Rafael Balaña-Fouce
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (M.-C.G.-M.); (J.A.-R.); (N.G.-F.); (Y.P.-P.); (R.M.R.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Carlos García-Estrada
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain; (M.-C.G.-M.); (J.A.-R.); (N.G.-F.); (Y.P.-P.); (R.M.R.)
- Instituto de Biomedicina (IBIOMED), Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Sharma V, Madia VN, Tudino V, Nguyen JV, Debnath A, Messore A, Ialongo D, Patacchini E, Palenca I, Basili Franzin S, Seguella L, Esposito G, Petrucci R, Di Matteo P, Bortolami M, Saccoliti F, Di Santo R, Scipione L, Costi R, Podust LM. Miconazole-like Scaffold is a Promising Lead for Naegleria fowleri-Specific CYP51 Inhibitors. J Med Chem 2023; 66:17059-17073. [PMID: 38085955 PMCID: PMC10758121 DOI: 10.1021/acs.jmedchem.3c01898] [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: 10/10/2023] [Revised: 11/17/2023] [Accepted: 11/22/2023] [Indexed: 12/29/2023]
Abstract
Developing drugs for brain infection by Naegleria fowleri is an unmet medical need. We used a combination of cheminformatics, target-, and phenotypic-based drug discovery methods to identify inhibitors that target an essential N. fowleri enzyme, sterol 14-demethylase (NfCYP51). A total of 124 compounds preselected in silico were tested against N. fowleri. Nine primary hits with EC50 ≤ 10 μM were phenotypically identified. Cocrystallization with NfCYP51 focused attention on one primary hit, miconazole-like compound 2a. The S-enantiomer of 2a produced a 1.74 Å cocrystal structure. A set of analogues was then synthesized and evaluated to confirm the superiority of the S-configuration over the R-configuration and the advantage of an ether linkage over an ester linkage. The two compounds, S-8b and S-9b, had an improved EC50 and KD compared to 2a. Importantly, both were readily taken up into the brain. The brain-to-plasma distribution coefficient of S-9b was 1.02 ± 0.12, suggesting further evaluation as a lead for primary amoebic meningoencephalitis.
Collapse
Affiliation(s)
- Vandna Sharma
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, Center for Discovery
and Innovation in Parasitic Diseases, University
of California San Diego, La Jolla, California 92093, United States
| | - Valentina Noemi Madia
- Dipartimento
di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci
Bolognetti, “Sapienza” Università
di Roma, p.le Aldo Moro 5, Rome I-00185, Italy
| | - Valeria Tudino
- Dipartimento
di Biotecnologie, Università degli
Studi di Siena, Chimica e Farmacia via Aldo Moro 2, Siena 53100, Italy
| | - Jennifer V. Nguyen
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, Center for Discovery
and Innovation in Parasitic Diseases, University
of California San Diego, La Jolla, California 92093, United States
| | - Anjan Debnath
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, Center for Discovery
and Innovation in Parasitic Diseases, University
of California San Diego, La Jolla, California 92093, United States
| | - Antonella Messore
- Dipartimento
di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci
Bolognetti, “Sapienza” Università
di Roma, p.le Aldo Moro 5, Rome I-00185, Italy
| | - Davide Ialongo
- Dipartimento
di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci
Bolognetti, “Sapienza” Università
di Roma, p.le Aldo Moro 5, Rome I-00185, Italy
| | - Elisa Patacchini
- Dipartimento
di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci
Bolognetti, “Sapienza” Università
di Roma, p.le Aldo Moro 5, Rome I-00185, Italy
| | - Irene Palenca
- Department
of Physiology and Pharmacology “V. Erspamer”, “Sapienza″ Università di Roma, p.le Aldo Moro 5, Rome I-00185, Italy
| | - Silvia Basili Franzin
- Department
of Physiology and Pharmacology “V. Erspamer”, “Sapienza″ Università di Roma, p.le Aldo Moro 5, Rome I-00185, Italy
| | - Luisa Seguella
- Department
of Physiology and Pharmacology “V. Erspamer”, “Sapienza″ Università di Roma, p.le Aldo Moro 5, Rome I-00185, Italy
| | - Giuseppe Esposito
- Department
of Physiology and Pharmacology “V. Erspamer”, “Sapienza″ Università di Roma, p.le Aldo Moro 5, Rome I-00185, Italy
| | - Rita Petrucci
- Dipartimento
di Scienze di Base e Applicate per l’Ingegneria, “Sapienza” Università di Roma, Via Castro Laurenziano 7, Rome 00161, Italy
| | - Paola Di Matteo
- Dipartimento
di Scienze di Base e Applicate per l’Ingegneria, “Sapienza” Università di Roma, Via Castro Laurenziano 7, Rome 00161, Italy
| | - Martina Bortolami
- Dipartimento
di Scienze di Base e Applicate per l’Ingegneria, “Sapienza” Università di Roma, Via Castro Laurenziano 7, Rome 00161, Italy
| | - Francesco Saccoliti
- D3 PharmaChemistry, Italian
Institute of Technology, Via Morego 30, Genova 16163, Italy
| | - Roberto Di Santo
- Dipartimento
di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci
Bolognetti, “Sapienza” Università
di Roma, p.le Aldo Moro 5, Rome I-00185, Italy
| | - Luigi Scipione
- Dipartimento
di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci
Bolognetti, “Sapienza” Università
di Roma, p.le Aldo Moro 5, Rome I-00185, Italy
| | - Roberta Costi
- Dipartimento
di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci
Bolognetti, “Sapienza” Università
di Roma, p.le Aldo Moro 5, Rome I-00185, Italy
| | - Larissa M. Podust
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, Center for Discovery
and Innovation in Parasitic Diseases, University
of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
9
|
Dutra Barroso Gomes N, Paula Magalhães E, Rodrigues Ribeiro L, Cavalcante JW, Morais Gomes Maia M, Cunha da Silva FR, Ali A, Machado Marinho M, Silva Marinho E, Silva Dos Santos H, Costa Martins AM, Róseo Paula Pessoa Bezerra de Menezes R. Trypanocidal potential of synthetic p-aminochalcones: In silico and in vitro evaluation. Bioorg Chem 2023; 141:106931. [PMID: 37879182 DOI: 10.1016/j.bioorg.2023.106931] [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: 07/19/2023] [Revised: 10/10/2023] [Accepted: 10/19/2023] [Indexed: 10/27/2023]
Abstract
Chagas disease (CD) is a neglected tropical disease of worldwide health concern, caused by the flagellate protozoan Trypanosoma cruzi (T. cruzi), endemic in Latin America and present in North America and Europe. The WHO recommended drug for CD, benznidazole has low safety profile and several limitations. Therefore, an entity with better therapeutic potential to treat CD is required. Chalcones are an important class of compounds, which have shown antichagasic potential. Thus, the objective of this study was to evaluate the activity of synthetic p-aminochalcones against T. cruzi. Chalcones 1 and 2 were synthesized by Claisen-Schmidt condensation and characterized by both spectroscopic and theoretical methods. Initially, they were submitted to molecular docking simulations using cruzain and trypanothione reductase (TR) enzymes. It was expected to observe the possible interactions of chalcones with the catalytic site and other important regions of these main pharmacological targets of T. cruzi. Their cytotoxicity within host cells were assessed by MTT reduction assay using LLC-MK2 cells, with CC50 = 85.6 ± 9.2 µM and 1115 ± 381.7 µM for chalcones 1 and 2, respectively. These molecules were also tested against epimastigote and trypomastigote life forms of T. cruzi, causing reduction in the number of viable parasites. For the evaluation of the effect on intracellular amastigotes, infected LLC-MK2 cells were incubated with the chalcones for 24 h, causing reduction in the percentage of infected cells and the number of amastigotes/100 cells. Finally, flow cytometry assays were performed for analyzing cell death mechanisms (7-AAD/AxPE labelling), cytoplasmic ROS accumulation (DCFH-DA assay) and mitochondrial transmembrane potential disruption (Rho123 assay). Both chalcones (1 and 2) caused membrane damage, ROS accumulation and mitochondrial depolarization. In conclusion, the synthetic p-aminochalcones presented trypanocidal effect, causing membrane damage and oxidative stress. Their mechanism of action may be related to cruzain and TR inhibition.
Collapse
Affiliation(s)
| | - Emanuel Paula Magalhães
- Post-Graduate Program in Pharmaceutical Sciences, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Lyanna Rodrigues Ribeiro
- Post-Graduate Program in Pharmaceutical Sciences, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | | | | | - Arif Ali
- Post-Graduate Program in Pharmacology, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Márcia Machado Marinho
- Theoretical and Eletrochemical Chemistry Research Group, State University of Ceará, Limoeiro do Norte, CE, Brazil; State University of Vale do Acaraú, Center for Exact Sciences and Technology, Sobral, CE, Brazil
| | - Emmanuel Silva Marinho
- Theoretical and Eletrochemical Chemistry Research Group, State University of Ceará, Limoeiro do Norte, CE, Brazil
| | - Hélcio Silva Dos Santos
- State University of Vale do Acaraú, Center for Exact Sciences and Technology, Sobral, CE, Brazil
| | - Alice Maria Costa Martins
- Department of Clinical and Toxicological Analysis, Federal University of Ceará, Fortaleza, CE, Brazil
| | | |
Collapse
|
10
|
Abirami M, Karan Kumar B, Dey S, Johri S, Reguera RM, Balaña-Fouce R, Gowri Chandra Sekhar KV, Sankaranarayanan M. Molecular-level strategic goals and repressors in Leishmaniasis - Integrated data to accelerate target-based heterocyclic scaffolds. Eur J Med Chem 2023; 257:115471. [PMID: 37257213 DOI: 10.1016/j.ejmech.2023.115471] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 06/02/2023]
Abstract
Leishmaniasis is a complex of neglected tropical diseases caused by various species of leishmanial parasites that primarily affect the world's poorest people. A limited number of standard medications are available for this disease that has been used for several decades, these drugs have many drawbacks such as resistance, higher cost, and patient compliance, making it difficult to reach the poor. The search for novel chemical entities to treat leishmaniasis has led to target-based scaffold research. Among several identified potential molecular targets, enzymes involved in the purine salvage pathway include polyamine biosynthetic process, such as arginase, ornithine decarboxylase, S-adenosylmethionine decarboxylase, spermidine synthase, trypanothione reductase as well as enzymes in the DNA cell cycle, such as DNA topoisomerases I and II plays vital role in the life cycle survival of leishmanial parasite. This review mainly focuses on various heterocyclic scaffolds, and their specific inhibitory targets against leishmaniasis, particularly those from the polyamine biosynthesis pathway and DNA topoisomerases with estimated activity studies of various heterocyclic analogs in terms of their IC50 or EC50 value, reported molecular docking analysis from available published literatures.
Collapse
Affiliation(s)
- M Abirami
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani, 333031, India
| | - Banoth Karan Kumar
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani, 333031, India; Department of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, AP, India
| | - Sanchita Dey
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani, 333031, India
| | - Samridhi Johri
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani, 333031, India
| | - Rosa M Reguera
- Department of Biomedical Sciences, University of León, 24071, León, Spain
| | | | - Kondapalli Venkata Gowri Chandra Sekhar
- Department of Chemistry, Birla Institute of Technology and Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Hyderabad, 500078, Telangana, India
| | - Murugesan Sankaranarayanan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani, 333031, India.
| |
Collapse
|
11
|
Henriquez-Figuereo A, Morán-Serradilla C, Angulo-Elizari E, Sanmartín C, Plano D. Small molecules containing chalcogen elements (S, Se, Te) as new warhead to fight neglected tropical diseases. Eur J Med Chem 2023; 246:115002. [PMID: 36493616 DOI: 10.1016/j.ejmech.2022.115002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/21/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022]
Abstract
Neglected tropical diseases (NTDs) encompass a group of infectious diseases with a protozoan etiology, high incidence, and prevalence in developing countries. As a result, economic factors constitute one of the main obstacles to their management. Endemic countries have high levels of poverty, deprivation and marginalization which affect patients and limit their access to proper medical care. As a matter of fact, statistics remain uncollected in some affected areas due to non-reporting cases. World Health Organization and other organizations proposed a plan for the eradication and control of the vector, although many of these plans were halted by the COVID-19 pandemic. Despite of the available drugs to treat these pathologies, it exists a lack of effectiveness against several parasite strains. Treatment protocols for diseases such as American trypanosomiasis (Chagas disease), leishmaniasis, and human African trypanosomiasis (HAT) have not achieved the desired results. Unfortunately, these drugs present limitations such as side effects, toxicity, teratogenicity, renal, and hepatic impairment, as well as high costs that have hindered the control and eradication of these diseases. This review focuses on the analysis of a collection of scientific shreds of evidence with the aim of identifying novel chalcogen-derived molecules with biological activity against Chagas disease, leishmaniasis and HAT. Compounds illustrated in each figure share the distinction of containing at least one chalcogen element. Sulfur (S), selenium (Se), and tellurium (Te) have been grouped and analyzed in accordance with their design strategy, chemical synthesis process and biological activity. After an exhaustive revision of the related literature on S, Se, and Te compounds, 183 compounds presenting excellent biological performance were gathered against the different causative agents of CD, leishmaniasis and HAT.
Collapse
Affiliation(s)
- Andreina Henriquez-Figuereo
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Technology and Chemistry, Irunlarrea 1, 31008, Pamplona, Spain; Institute of Tropical Health, University of Navarra, Irunlarrea 1, 31008, Pamplona, Spain.
| | - Cristina Morán-Serradilla
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Technology and Chemistry, Irunlarrea 1, 31008, Pamplona, Spain
| | - Eduardo Angulo-Elizari
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Technology and Chemistry, Irunlarrea 1, 31008, Pamplona, Spain
| | - Carmen Sanmartín
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Technology and Chemistry, Irunlarrea 1, 31008, Pamplona, Spain; Institute of Tropical Health, University of Navarra, Irunlarrea 1, 31008, Pamplona, Spain.
| | - Daniel Plano
- University of Navarra, School of Pharmacy and Nutrition, Department of Pharmaceutical Technology and Chemistry, Irunlarrea 1, 31008, Pamplona, Spain; Institute of Tropical Health, University of Navarra, Irunlarrea 1, 31008, Pamplona, Spain.
| |
Collapse
|
12
|
de Sousa NF, da Silva Souza HD, de Menezes RPB, da Silva Alves F, Acevedo CAH, de Lima Nunes TA, Sessions ZL, Scotti L, Muratov EN, Mendonça-Junior FJB, da Franca Rodrigues KA, de Athayde Filho PF, Scotti MT. Selene-Ethylenelacticamides and N-Aryl-Propanamides as Broad-Spectrum Leishmanicidal Agents. Pathogens 2023; 12:136. [PMID: 36678484 PMCID: PMC9860784 DOI: 10.3390/pathogens12010136] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/20/2022] [Accepted: 12/26/2022] [Indexed: 01/18/2023] Open
Abstract
The World Health Organization classifies Leishmania as one of the 17 “neglected diseases” that burden tropical and sub-tropical climate regions with over half a million diagnosed cases each year. Despite this, currently available anti-leishmania drugs have high toxicity and the potential to be made obsolete by parasite drug resistance. We chose to analyze organoselenides for leishmanicidal potential given the reduced toxicity inherent to selenium and the displayed biological activity of organoselenides against Leishmania. Thus, the biological activities of 77 selenoesters and their N-aryl-propanamide derivatives were predicted using robust in silico models of Leishmania infantum, Leishmania amazonensis, Leishmania major, and Leishmania (Viannia) braziliensis. The models identified 28 compounds with >60% probability of demonstrating leishmanicidal activity against L. infantum, and likewise, 26 for L. amazonesis, 25 for L. braziliensis, and 23 for L. major. The in silico prediction of ADMET properties suggests high rates of oral absorption and good bioavailability for these compounds. In the in silico toxicity evaluation, only seven compounds showed signs of toxicity in up to one or two parameters. The methodology was corroborated with the ensuing experimental validation, which evaluated the inhibition of the Promastigote form of the Leishmania species under study. The activity of the molecules was determined by the IC50 value (µM); IC50 values < 20 µM indicated better inhibition profiles. Sixteen compounds were synthesized and tested for their activity. Eight molecules presented IC50 values < 20 µM for at least one of the Leishmania species under study, with compound NC34 presenting the strongest parasite inhibition profile. Furthermore, the methodology used was effective, as many of the compounds with the highest probability of activity were confirmed by the in vitro tests performed.
Collapse
Affiliation(s)
- Natália Ferreira de Sousa
- Post-Graduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil
| | | | | | - Francinara da Silva Alves
- Post-Graduate Program in Chemistry, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil
| | - Chonny Alexander Herrera Acevedo
- Post-Graduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil
| | - Thaís Amanda de Lima Nunes
- Infectious Diseases Laboratory, Federal University of Delta of Parnaíba, Av. São Sebastião, nº 2819-Nossa Sra. de Fátima, Parnaíba 64202-020, PI, Brazil
| | - Zoe L. Sessions
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Luciana Scotti
- Post-Graduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil
| | - Eugene N. Muratov
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | - Klinger Antônio da Franca Rodrigues
- Infectious Diseases Laboratory, Federal University of Delta of Parnaíba, Av. São Sebastião, nº 2819-Nossa Sra. de Fátima, Parnaíba 64202-020, PI, Brazil
| | | | - Marcus Tullius Scotti
- Post-Graduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil
| |
Collapse
|
13
|
Soto-Vásquez MR, Alvarado-García PAA, Osorio EH, Tallini LR, Bastida J. Antileishmanial Activity of Clinanthus milagroanthus S. Leiva & Meerow (Amaryllidaceae) Collected in Peru. PLANTS (BASEL, SWITZERLAND) 2023; 12:322. [PMID: 36679035 PMCID: PMC9866881 DOI: 10.3390/plants12020322] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 06/17/2023]
Abstract
Leishmaniasis is a worldwide infectious parasitic disease caused by different species of protozoa of the genus Leishmania, which are transmitted to animals and humans through the bite of insects of the Psychodidae family. In the present work, the antileishmanial activity of an alkaloid extract of the bulbs of Clinanthus milagroanthus S. Leiva & Meerow (Amaryllidaceae) was evaluated in vitro, in vivo, and in silico against the parasite Leishmania braziliensis, and the chemical profile of the sample was determined by GC-MS analysis. At concentrations of 1, 10, and 100 µg·mL−1, the alkaloid extract presented inhibition percentages of 8.7%, 23.1%, and 98.8%, respectively, against L. braziliensis with a p < 0.05, and IC50 values of 18.5 ± 0.3 µg·mL−1. Furthermore, at a dose of 1.0 mg·kg−1, a greater decrease in lesion size was observed (90%) for in vivo assays, as well as a decrease in infection (96%), finding no significant differences (p > 0.05) in comparison with amphotericin B (92% and 98%, respectively). Eleven alkaloids were identified in C. milagroanthus bulbs: galanthamine, vittatine/crinine, 8-O-demethylmaritidine, anhydrolycorine, 11,12-dehydroanhydrolycorine, hippamine, lycorine, 2-hydroxyanhydrolycorine, 7-hydroxyclivonine, 2α-hydroxyhomolycorine, and 7-hydroxyclivonine isomer. A molecular model of Leishmania braziliensis trypanothione reductase (TRLb) was built using computational experiments to evaluate in silico the potential of the Amaryllidaceae alkaloid identified in C. milagroanthus toward this enzyme. The structures galanthamine, 7-hydroxyclivonine isomer, and crinine showed better estimated free energy of binding than the reference compound, amphotericin B. In conclusion, this is the first in vitro, in vivo, and in silico report about the antileishmanial potential and alkaloid profiling of the extract of C. milagroanthus bulbs, which could become an interesting source of bioactive molecules.
Collapse
Affiliation(s)
- Marilú Roxana Soto-Vásquez
- Facultad de Farmacia y Bioquímica, Universidad Nacional de Trujillo, Av. Juan Pablo II s/n, Trujillo 13011, Peru
| | | | - Edison H. Osorio
- Facultad de Ciencias Naturales y Matemáticas, Universidad de Ibagué, Carrera 22 Calle 67, Ibagué 730001, Colombia
| | - Luciana R. Tallini
- Departament de Biologia, Sanitat i Medi Ambient, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, Av. Joan XXIII 27–31, 08028 Barcelona, Spain
| | - Jaume Bastida
- Departament de Biologia, Sanitat i Medi Ambient, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, Av. Joan XXIII 27–31, 08028 Barcelona, Spain
| |
Collapse
|
14
|
Inhibition of Leishmania infantum Trypanothione Reductase by New Aminopropanone Derivatives Interacting with the NADPH Binding Site. Molecules 2023; 28:molecules28010338. [PMID: 36615531 PMCID: PMC9823735 DOI: 10.3390/molecules28010338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/15/2022] [Accepted: 12/28/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND As a result of the paucity of treatment, Leishmaniasis continues to provoke about 60,000 deaths every year worldwide. New molecules are needed, and drug discovery research is oriented toward targeting proteins crucial for parasite survival. Among them, trypanothione reductase (TR) is of remarkable interest owing to its vital role in Leishmania species protozoan parasite life. Our previously identified compound 1 is a novel chemotype endowed with a unique mode of TR inhibition thanks to its binding to a formerly unknown but druggable site at the entrance of the NADPH binding cavity, absent in human glutathione reductase (hGR). METHODS We designed and synthesized new 3-amino-1-arylpropan-1-one derivatives structurally related to compound 1 and evaluated their potential inhibition activity on TR from Leishmania infantum (LiTR). Cluster docking was performed to assess the binding poses of the compounds. RESULTS The newly synthesized compounds were screened at a concentration of 100 μM in in vitro assays and all of them proved to be active with residual activity percentages lower than 75%. CONCLUSIONS Compounds 2a and 2b were the most potent inhibitors found, suggesting that an additional aromatic ring might be promising for enzymatic inhibition. Further structure-activity relationships are needed to optimize our compounds activity.
Collapse
|
15
|
Jain S, Sahu U, Kumar A, Khare P. Metabolic Pathways of Leishmania Parasite: Source of Pertinent Drug Targets and Potent Drug Candidates. Pharmaceutics 2022; 14:pharmaceutics14081590. [PMID: 36015216 PMCID: PMC9416627 DOI: 10.3390/pharmaceutics14081590] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 02/04/2023] Open
Abstract
Leishmaniasis is a tropical disease caused by a protozoan parasite Leishmania that is transmitted via infected female sandflies. At present, leishmaniasis treatment mainly counts on chemotherapy. The currently available drugs against leishmaniasis are costly, toxic, with multiple side effects, and limitations in the administration route. The rapid emergence of drug resistance has severely reduced the potency of anti-leishmanial drugs. As a result, there is a pressing need for the development of novel anti-leishmanial drugs with high potency, low cost, acceptable toxicity, and good pharmacokinetics features. Due to the availability of preclinical data, drug repurposing is a valuable approach for speeding up the development of effective anti-leishmanial through pointing to new drug targets in less time, having low costs and risk. Metabolic pathways of this parasite play a crucial role in the growth and proliferation of Leishmania species during the various stages of their life cycle. Based on available genomics/proteomics information, known pathways-based (sterol biosynthetic pathway, purine salvage pathway, glycolysis, GPI biosynthesis, hypusine, polyamine biosynthesis) Leishmania-specific proteins could be targeted with known drugs that were used in other diseases, resulting in finding new promising anti-leishmanial therapeutics. The present review discusses various metabolic pathways of the Leishmania parasite and some drug candidates targeting these pathways effectively that could be potent drugs against leishmaniasis in the future.
Collapse
Affiliation(s)
- Surbhi Jain
- Department of Microbiology, All India Institute of Medical Sciences, Bhopal 462026, Madhya Pradesh, India; (S.J.); (U.S.)
| | - Utkarsha Sahu
- Department of Microbiology, All India Institute of Medical Sciences, Bhopal 462026, Madhya Pradesh, India; (S.J.); (U.S.)
- Division of Synthetic Biology, Absolute Foods, Plot 68, Sector 44, Gurugram 122003, Haryana, India
| | - Awanish Kumar
- Department of Biotechnology, National Institute of Technology, Raipur 492010, Chhattisgarh, India
- Correspondence: or (A.K.); (P.K.)
| | - Prashant Khare
- Department of Microbiology, All India Institute of Medical Sciences, Bhopal 462026, Madhya Pradesh, India; (S.J.); (U.S.)
- Division of Synthetic Biology, Absolute Foods, Plot 68, Sector 44, Gurugram 122003, Haryana, India
- Correspondence: or (A.K.); (P.K.)
| |
Collapse
|
16
|
Battista T, Federico S, Brogi S, Pozzetti L, Khan T, Butini S, Ramunno A, Fiorentino E, Orsini S, Di Muccio T, Fiorillo A, Exertier C, Di Risola D, Colotti G, Gemma S, Ilari A, Campiani G. Optimization of Potent and Specific Trypanothione Reductase Inhibitors: A Structure-Based Drug Discovery Approach. ACS Infect Dis 2022; 8:1687-1699. [PMID: 35880849 DOI: 10.1021/acsinfecdis.2c00325] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Leishmania spp. are responsible for up to 1 million new cases each year. The current therapeutic arsenal against Leishmania is largely inadequate, and there is an urgent need for better drugs. Trypanothione reductase (TR) represents a druggable target since it is essential for the parasite and not shared by the human host. Here, we report the optimization of a novel class of potent and selective LiTR inhibitors realized through a concerted effort involving X-ray crystallography, synthesis, structure-activity relationship (SAR) investigation, molecular modeling, and in vitro phenotypic assays. 5-Nitrothiophene-2-carboxamides 3, 6e, and 8 were among the most potent and selective TR inhibitors identified in this study. 6e and 8 displayed leishmanicidal activity in the low micromolar range coupled to SI > 50. Our studies could pave the way for the use of TR inhibitors not only against leishmaniasis but also against other trypanosomatidae due to the structural similarity of TR enzymes.
Collapse
Affiliation(s)
- Theo Battista
- 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, 00185 Roma, Italy.,Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via L. Giorgieri, 1, 34127 Trieste, Italy
| | - Stefano Federico
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Simone Brogi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Luca Pozzetti
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Tuhina Khan
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Stefania Butini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Anna Ramunno
- Department of Pharmacy/DIFARMA, University of Salerno, via Giovanni Paolo II 132, 84084 Fisciano, Salerno, Italy
| | - Eleonora Fiorentino
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Stefania Orsini
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Trentina Di Muccio
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Annarita Fiorillo
- 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, 00185 Roma, Italy.,Department of Biochemical Sciences, Sapienza University of Rome, Piazzale A. Moro 5, 00185 Roma, Italy
| | - 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, 00185 Roma, Italy
| | - Daniel Di Risola
- Department of Biochemical Sciences, Sapienza University of Rome, Piazzale A. Moro 5, 00185 Roma, Italy
| | - Gianni Colotti
- 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, 00185 Roma, Italy
| | - Sandra Gemma
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, 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, 00185 Roma, Italy
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| |
Collapse
|
17
|
Bisht S, Kumar L, Kaul G, Akhir A, Saxena D, Chopra S, Karthik R, Goyal N, Batra S. Synthesis and Biological Evaluation of Substituted 3‐Isoxazolethioethers as Antileishmanial and Antibacterial Agents. ChemistrySelect 2022. [DOI: 10.1002/slct.202201664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Shweta Bisht
- Medicinal and Process Chemistry Division CSIR-Central Drug Research Institute Sector 10, Jankipuram Extension Sitapur Road Lucknow 226031 UP India
| | - Lalan Kumar
- Medicinal and Process Chemistry Division CSIR-Central Drug Research Institute Sector 10, Jankipuram Extension Sitapur Road Lucknow 226031 UP India
| | - Grace Kaul
- Molecular Microbiology and Immunology Division CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension Sitapur Road Lucknow 226031 India
- Academy of Scientific and Innovative Research CSIR- Human Resource Development Centre CSIR-HRDC) Campus, Sector 19, Kamla Nehru Nagar Ghaziabad 201002 UP India
| | - Abdul Akhir
- Molecular Microbiology and Immunology Division CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension Sitapur Road Lucknow 226031 India
- Academy of Scientific and Innovative Research CSIR- Human Resource Development Centre CSIR-HRDC) Campus, Sector 19, Kamla Nehru Nagar Ghaziabad 201002 UP India
| | - Deepanshi Saxena
- Molecular Microbiology and Immunology Division CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension Sitapur Road Lucknow 226031 India
- Academy of Scientific and Innovative Research CSIR- Human Resource Development Centre CSIR-HRDC) Campus, Sector 19, Kamla Nehru Nagar Ghaziabad 201002 UP India
| | - Sidharth Chopra
- Molecular Microbiology and Immunology Division CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension Sitapur Road Lucknow 226031 India
- Academy of Scientific and Innovative Research CSIR- Human Resource Development Centre CSIR-HRDC) Campus, Sector 19, Kamla Nehru Nagar Ghaziabad 201002 UP India
| | - R. Karthik
- Biochemistry Division CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension Sitapur Road Lucknow 226031 India
- Academy of Scientific and Innovative Research CSIR- Human Resource Development Centre CSIR-HRDC) Campus, Sector 19, Kamla Nehru Nagar Ghaziabad 201002 UP India
| | - Neena Goyal
- Biochemistry Division CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension Sitapur Road Lucknow 226031 India
- Academy of Scientific and Innovative Research CSIR- Human Resource Development Centre CSIR-HRDC) Campus, Sector 19, Kamla Nehru Nagar Ghaziabad 201002 UP India
| | - Sanjay Batra
- Medicinal and Process Chemistry Division CSIR-Central Drug Research Institute Sector 10, Jankipuram Extension Sitapur Road Lucknow 226031 UP India
- Academy of Scientific and Innovative Research CSIR- Human Resource Development Centre CSIR-HRDC) Campus, Sector 19, Kamla Nehru Nagar Ghaziabad 201002 UP India
| |
Collapse
|
18
|
Panda SK, Gazim ZC, Swain SS, Bento MCVDA, Sena JDS, Mukazayire MJ, Van Puyvelde L, Luyten W. Ethnomedicinal, Phytochemical and Pharmacological Investigations of Tetradenia riparia (Hochst.) Codd (Lamiaceae). Front Pharmacol 2022; 13:896078. [PMID: 35721148 PMCID: PMC9201335 DOI: 10.3389/fphar.2022.896078] [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: 03/14/2022] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
Tetradenia riparia Hochsteter codd. (Lamiaceae) in its native African continent, is considered one of the most popular aromatic medicinal plants. In folk medicine it may be used as an infusion to treat respiratory problems, cough, headache, stomach pain, diarrhea, fever, malaria, and dengue; and in the form of compresses it is applied for the relief of headaches and toothaches. The species T. riparia has been researched for decades to isolate and identify chemical constituents present in extracts or essential oil obtained from the leaves, floral buds, or stems of this plant. The present study reviews the scientific literature on ethnomedicinal, phytochemical, and pharmacological aspects of T. riparia. We discuss issues related to the botanical and geographical description of the species, ethnobotanical uses, phytochemical studies on its essential oil and extracts, and biological activities of T. riparia. Several compounds have already been isolated from leaves, such as ibozol, 7α-hydroxyroileanone, 1',2'-dideacetylboronolide, 8(14),15-sandaracopimaradiene-7α,18-diol; 5,6-dihydro-α-pyrone and α-pyrone. Terpenes predominated in the essential oil, comprising monoterpenes, sesquiterpenes, diterpenes, hydrocarbons, and oxygenates. Most phytocompounds were isolated from the leaves and flower buds, namely fenchone, 14-hydroxy-9-epi (E)-caryophyllene, 9β, 13β-epoxy-7-abietene, and 6,7-dehydroroileanone. These compounds provide the species a high pharmacological potential, with antimicrobial, antioxidant, antitumor, analgesic, anti-leishmania, anti-tuberculosis, and anti-parasitic activities. Therefore, this species is a promising herbal medicine.
Collapse
Affiliation(s)
- Sujogya Kumar Panda
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Leuven, Belgium.,Centre of Environment Climate Change and Public Health, RUSA, Utkal University, Bhubaneswar, India
| | - Zilda Cristiani Gazim
- Chemistry Laboratory of Natural Products, Graduate Program in Animal Science and Biotechnology Applied to Agriculture, Paranaense University, Umuarama, Brazil
| | - Shasank S Swain
- Division of Microbiology and NCDs, ICMR-Regional Medical Research Centre, Bhubaneswar, India
| | - Marisa Cassia Vieira de Araujo Bento
- Chemistry Laboratory of Natural Products, Graduate Program in Animal Science and Biotechnology Applied to Agriculture, Paranaense University, Umuarama, Brazil
| | - Jéssica da Silva Sena
- Chemistry Laboratory of Natural Products, Graduate Program in Animal Science and Biotechnology Applied to Agriculture, Paranaense University, Umuarama, Brazil
| | - Marie Jeanne Mukazayire
- Department of Pharmacy, School of Pharmacy and Medicine, College of Medicine and Health Sciences, University of Rwanda, Huye, Rwanda
| | - Luc Van Puyvelde
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Leuven, Belgium
| | - Walter Luyten
- Department of Biology, Animal Physiology and Neurobiology Section, KU Leuven, Leuven, Belgium
| |
Collapse
|
19
|
Kumar A, Nimsarkar P, Singh S. Probing the Interactions Responsible for the Structural Stability of Trypanothione Reductase Through Computer Simulation and Biophysical Characterization. Protein J 2022; 41:230-244. [PMID: 35364760 DOI: 10.1007/s10930-022-10052-x] [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] [Accepted: 03/22/2022] [Indexed: 11/26/2022]
Abstract
With the necessity to develop antileishmanial drugs with substrate specificity, trypanothione reductase (TryR) has gained popularity in parasitology. TryR is unique to be present only in trypanosomatids and is functionally similar to glutathione in mammals. It protects against oxidative stress exerted by the host defense mechanism. The TryR enzyme is essential for the survival of Leishmania parasites in the host as it reduces trypanothione and aids in neutralizing hydrogen peroxide produced by the host macrophages during infection. Henceforth, it becomes vital to decipher their functional stability and behaviour in the presence of denaturants. Our study is focused on structural, functional and behavioural stability aspects of TryR with different concentrations of Urea, Guanidinium chloride, alcohol based compounds followed by extensive molecular dynamics simulations in a lipid bilayer system. The results obtained from the study reveal an interesting insight into the possible mechanisms of modulation of the structure, function and stability of the TryR protein.
Collapse
Affiliation(s)
- Anurag Kumar
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune, 411007, India
| | - Prajakta Nimsarkar
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune, 411007, India
| | - Shailza Singh
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune, 411007, India.
| |
Collapse
|
20
|
Febrifugine dihydrochloride as a new oral chemotherapeutic agent against visceral leishmaniasis infection. Exp Parasitol 2022; 236-237:108250. [DOI: 10.1016/j.exppara.2022.108250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 03/09/2022] [Accepted: 03/23/2022] [Indexed: 11/19/2022]
|
21
|
Pyne N, Paul S. Screening of medicinal plants unraveled the leishmanicidal credibility of Garcinia cowa; highlighting Norcowanin, a novel anti-leishmanial phytochemical through in-silico study. J Parasit Dis 2022; 46:202-214. [PMID: 35299910 PMCID: PMC8901847 DOI: 10.1007/s12639-021-01441-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 08/07/2021] [Indexed: 02/04/2023] Open
Abstract
Leishmaniasis, one of the most prevalent yet neglected parasitic causes of death, yearns for therapeutic control and treatment. Severely toxic and inefficient modern-day pentavalent antimonials, caters the search for naturally derived drugs, as efficient alternatives for disease treatment. The anti-promastigote activity of ten different plants selected for their ethnomedicinal properties revealed significant leishmanicidal capacity; the most potent being Garcinia cowa methanolic extract with an IC50 value of 21.4 µg/ml. Garcinia cowa, a plant endemic to North-Eastern India that is of the Clusiaceae family, is replete with such medicinal qualities as antimicrobial, antiviral, antiparasitic, and antiproliferative activities. Computational biology with its tools such as molecular docking has opened new horizons aimed at a better understanding of biological systems, complexes, and their interactions, and subsequently drug discovery via in silico techniques. Therefore, an in-silico study was designed to evaluate the binding capability of six phytochemicals- cowanin, cowanol, cowaxanthone, norcowanin, rubraxanthone, and a basic xanthone, found in Garcinia cowa against Pentamidine, a synthetic anti-leishmanial drug. The active sites of three characteristic enzymes belonging to the Leishmania donovani parasite: O-acetylserine sulfhydrylase (OASS), Trypanothione reductase (TryR), and N-Myristoyltransferase (NMT) were chosen as target proteins. Results revealed lower binding energies and higher affinities, of nearly all the phytochemicals with respect to Pentamidine, indicating their leishmanicidal potential. Norcowanin showed the lowest average binding of - 9.8 kcal/mol against all the three enzymes under study.
Collapse
Affiliation(s)
- Nibedita Pyne
- Laboratory of Cell and Molecular Biology, Department of Botany, Centre of Advanced Study, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019 India
| | - Santanu Paul
- Laboratory of Cell and Molecular Biology, Department of Botany, Centre of Advanced Study, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019 India
| |
Collapse
|
22
|
Almeida FS, Sousa GLS, Rocha JC, Ribeiro FF, de Oliveira MR, de Lima Grisi TCS, Araújo DAM, de C Nobre MS, Castro RN, Amaral IPG, Keesen TSL, de Moura RO. In vitro anti-Leishmania activity and molecular docking of spiro-acridine compounds as potential multitarget agents against Leishmania infantum. Bioorg Med Chem Lett 2021; 49:128289. [PMID: 34311084 DOI: 10.1016/j.bmcl.2021.128289] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/13/2021] [Accepted: 07/20/2021] [Indexed: 11/28/2022]
Abstract
Leishmaniasis is an infectious disease with several limitations regarding treatment schemes. This work reports the anti-Leishmania activity of spiroacridine compounds against the promastigote (IC50 = 1.1 to 6.0 µg / mL) and amastigote forms of the best compounds (EC50 = 4.9 and 0.9 µg / mL) inLeishmania (L.) infantumand proposes an in-silico study with possible selective therapeutic targets for L. infantum. The substituted dimethyl-amine compound (AMTAC 11) showed the best leishmanicidal activity in vitro, and was found to interact with TryRandLdTopoI. comparisons with standard inhibitors were performed, and its main interactions were elucidated. Based on the biological assessment and the structure-activity relationship study, the spiroacridine compounds appear to be promisinganti-leishmaniachemotherapeutic agents to be explored.
Collapse
Affiliation(s)
- Fernanda S Almeida
- Programa de Doutorado em Biotecnologia, Rede Nordeste de Biotecnologia, Universidade Federal da Paraíba, João Pessoa, PB 58051-900, Brazil; Laboratório de Imunologia das Doenças Infeciosas, Universidade Federal da Paraíba, João Pessoa, PB 58051-900, Brazil
| | - Gleyton L S Sousa
- Programa de Doutorado em Química, Universidade Federal Rural do Rio de Janeiro, Seropédica, RJ 23897-000, Brazil
| | - Juliana C Rocha
- Laboratório de Imunologia das Doenças Infeciosas, Universidade Federal da Paraíba, João Pessoa, PB 58051-900, Brazil
| | - Frederico F Ribeiro
- Centro de Ciências Exatas e da Natureza, Universidade Federal da Paraíba, João Pessoa, PB 58059-900, Brazil
| | - Márcia Rosa de Oliveira
- Departamento de Biologia Molecular, Centro de Ciências Exatas e da Natureza, Universidade Federal da Paraíba, Joao Pessoa, Paraíba CEP 58059-900, Brazil
| | | | - Demetrius A M Araújo
- Departamento de Biologia Celular e Molecular, Universidade Federal da Paraíba, João Pessoa, PB 58059-900, Brazil
| | - Michelangela S de C Nobre
- Programa de Doutorado em Ciências Farmacêuticas, Universidade Federal de Pernambuco, Recife, PE 50670-901, Brazil
| | - Rosane N Castro
- Programa de Doutorado em Química, Universidade Federal Rural do Rio de Janeiro, Seropédica, RJ 23897-000, Brazil
| | - Ian P G Amaral
- Departamento de Biologia Celular e Molecular, Universidade Federal da Paraíba, João Pessoa, PB 58059-900, Brazil
| | - Tatjana S L Keesen
- Departamento de Biologia Celular e Molecular, Universidade Federal da Paraíba, João Pessoa, PB 58059-900, Brazil; Laboratório de Imunologia das Doenças Infeciosas, Universidade Federal da Paraíba, João Pessoa, PB 58051-900, Brazil
| | - Ricardo Olímpio de Moura
- Centro de Ciências Biológicas e da Saúde, Universidade Estadual da Paraíba, Campina Grande, PB 58429-500, Brazil.
| |
Collapse
|
23
|
Druggable hot spots in trypanothione reductase: novel insights and opportunities for drug discovery revealed by DRUGpy. J Comput Aided Mol Des 2021; 35:871-882. [PMID: 34181199 DOI: 10.1007/s10822-021-00403-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/18/2021] [Indexed: 10/21/2022]
Abstract
Assessment of target druggability guided by search and characterization of hot spots is a pivotal step in early stages of drug-discovery. The raw output of FTMap provides the data to perform this task, but it relies on manual intervention to properly combine different sets of consensus sites, therefore allowing identification of hot spots and evaluation of strength, shape and distance among them. Thus, the user's previous experience on the target and the software has a direct impact on how data generated by FTMap server can be explored. DRUGpy plugin was developed to overcome this limitation. By automatically assembling and scoring all possible combinations of consensus sites, DRUGpy plugin provides FTMap users a straight-forward method to identify and characterize hot spots in protein targets. DRUGpy is available in all operating systems that support PyMOL software. DRUGpy promptly identifies and characterizes pockets that are predicted by FTMap to bind druglike molecules with high-affinity (druggable sites) or low-affinity (borderline sites) and reveals how protein conformational flexibility impacts on the target's druggability. The use of DRUGpy on the analysis of trypanothione reductases (TR), a validated drug target against trypanosomatids, showcases the usefulness of the plugin, and led to the identification of a druggable pocket in the conserved dimer interface present in this class of proteins, opening new perspectives to the design of selective inhibitors.
Collapse
|
24
|
Identification of 3-Methoxycarpachromene and Masticadienonic Acid as New Target Inhibitors against Trypanothione Reductase from Leishmania Infantum Using Molecular Docking and ADMET Prediction. Molecules 2021; 26:molecules26113335. [PMID: 34206087 PMCID: PMC8199445 DOI: 10.3390/molecules26113335] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/25/2021] [Accepted: 05/27/2021] [Indexed: 11/17/2022] Open
Abstract
Polyphenolic and Terpenoids are potent natural antiparasitic compounds. This study aimed to identify new drug against Leishmania parasites, leishmaniasis’s causal agent. A new in silico analysis was accomplished using molecular docking, with the Autodock vina program, to find the binding affinity of two important phytochemical compounds, Masticadienonic acid and the 3-Methoxycarpachromene, towards the trypanothione reductase as target drugs, responsible for the defense mechanism against oxidative stress and virulence of these parasites. There were exciting and new positive results: the molecular docking results show as elective binding profile for ligands inside the active site of this crucial enzyme. The ADMET study suggests that the 3-Methoxycarpachromene has the highest probability of human intestinal absorption. Through this work, 3-Methoxycarpachromene and Masticadienonic acid are shown to be potentially significant in drug discovery, especially in treating leishmaniasis. Hence, drug development should be completed with promising results.
Collapse
|
25
|
da Silva MA, Fokoue HH, Fialho SN, Dos Santos APDA, Rossi NRDLP, Gouveia ADJ, Ferreira AS, Passarini GM, Garay AFG, Alfonso JJ, Soares AM, Zanchi FB, Kato MJ, Teles CBG, Kuehn CC. Antileishmanial activity evaluation of a natural amide and its synthetic analogs against Leishmania (V.) braziliensis: an integrated approach in vitro and in silico. Parasitol Res 2021; 120:2199-2218. [PMID: 33963899 DOI: 10.1007/s00436-021-07169-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 04/21/2021] [Indexed: 10/21/2022]
Abstract
Leishmaniasis is considered a neglected disease, which makes it an unattractive market for the pharmaceutical industry; hence, efforts in the search for biologically active substances are hampered by this lack of financial motivation. Thus, in the present study, we report the leishmanicidal activity and the possible mechanisms of action of compounds with promising activity against the species Leishmania (V.) braziliensis, the causative agent of the skin disease leishmaniasis. The natural compound 1a (piplartine) and the analog 2a were the most potent against promastigote forms with growth inhibition values for 50% of the parasite population (IC50) = 8.58 and 11.25 μM, respectively. For amastigote forms, the ICa50 values were 1.46 and 16.7 μM, respectively. In the molecular docking study, piplartine showed favorable binding energy (-7.13 kcal/mol) and with 50% inhibition of trypanothione reductase (IC50) = 91.1 μM. Preliminary investigations of the mechanism of action indicate that piplartine increased ROS levels, induced loss of cell membrane integrity, and caused accumulation of lipid bodies after 24 h of incubation at its lowest effective concentration (IC50), which was not observed for the synthetic analog 2a. The mode of action for the leishmanicidal activity of piplartine (1a) was assigned to involve affinity for the trypanothione reductase of Leishmania (V.) braziliensis TR.
Collapse
Affiliation(s)
- Minelly A da Silva
- Federal Institute of Education, Science and Technology of Rondônia - IFRO, Porto Velho, Rondônia, Brazil.
- Federal University of Rondônia - UNIR, Porto Velho, Rondônia, Brazil.
- Instituto Federal de Rondônia - Porto Velho-Calama, Av. Calama, 4985 - Flodoaldo Pontes Pinto, Porto Velho, RO, 76820-441, Brazil.
| | - Harold H Fokoue
- Fundação Oswaldo Cruz, Instituto de Tecnologia em Fármacos, Farmanguinhos - FIOCRUZ/RJ, Rio de Janeiro, Brazil
| | - Saara N Fialho
- Fundação Oswaldo Cruz - Rondônia - FIOCRUZ/RO, Porto Velho, Rondônia, Brazil
- Rede de Biodiversidade e Biotecnologia da Amazônia Legal - BIONORTE, Porto Velho, Rondônia, Brazil
| | | | - Norton R D L P Rossi
- Federal University of Rondônia - UNIR, Porto Velho, Rondônia, Brazil
- Fundação Oswaldo Cruz - Rondônia - FIOCRUZ/RO, Porto Velho, Rondônia, Brazil
| | | | - Amália S Ferreira
- Fundação Oswaldo Cruz - Rondônia - FIOCRUZ/RO, Porto Velho, Rondônia, Brazil
| | - Guilherme M Passarini
- Federal University of Rondônia - UNIR, Porto Velho, Rondônia, Brazil
- Fundação Oswaldo Cruz - Rondônia - FIOCRUZ/RO, Porto Velho, Rondônia, Brazil
| | - Ana F G Garay
- Fundação Oswaldo Cruz - Rondônia - FIOCRUZ/RO, Porto Velho, Rondônia, Brazil
- Centro para el Desarrollo de la Investigación Científica - CEDIC, Asunción, Paraguay
| | - Jorge J Alfonso
- Fundação Oswaldo Cruz - Rondônia - FIOCRUZ/RO, Porto Velho, Rondônia, Brazil
- Centro para el Desarrollo de la Investigación Científica - CEDIC, Asunción, Paraguay
| | - Andreimar M Soares
- Federal University of Rondônia - UNIR, Porto Velho, Rondônia, Brazil
- Fundação Oswaldo Cruz - Rondônia - FIOCRUZ/RO, Porto Velho, Rondônia, Brazil
- Rede de Biodiversidade e Biotecnologia da Amazônia Legal - BIONORTE, Porto Velho, Rondônia, Brazil
- National Institute of Science and Technology of Epidemiology in the Western Amazon - INCT-EpiAmO, Rondônia, Brazil
| | - Fernando B Zanchi
- Federal University of Rondônia - UNIR, Porto Velho, Rondônia, Brazil
- Fundação Oswaldo Cruz - Rondônia - FIOCRUZ/RO, Porto Velho, Rondônia, Brazil
- Rede de Biodiversidade e Biotecnologia da Amazônia Legal - BIONORTE, Porto Velho, Rondônia, Brazil
| | - Massuo J Kato
- Institute of Chemistry, University of São Paulo - USP, São Paulo, Brazil
| | - Carolina B G Teles
- Federal University of Rondônia - UNIR, Porto Velho, Rondônia, Brazil
- Fundação Oswaldo Cruz - Rondônia - FIOCRUZ/RO, Porto Velho, Rondônia, Brazil
- Rede de Biodiversidade e Biotecnologia da Amazônia Legal - BIONORTE, Porto Velho, Rondônia, Brazil
- National Institute of Science and Technology of Epidemiology in the Western Amazon - INCT-EpiAmO, Rondônia, Brazil
| | - Christian C Kuehn
- Federal University of Rondônia - UNIR, Porto Velho, Rondônia, Brazil
| |
Collapse
|
26
|
Madia VN, Nicolai A, Messore A, De Leo A, Ialongo D, Tudino V, Saccoliti F, De Vita D, Scipione L, Artico M, Taurone S, Taglieri L, Di Santo R, Scarpa S, Costi R. Design, Synthesis and Biological Evaluation of New Pyrimidine Derivatives as Anticancer Agents. Molecules 2021; 26:molecules26030771. [PMID: 33540875 PMCID: PMC7867324 DOI: 10.3390/molecules26030771] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 02/06/2023] Open
Abstract
Background: Anticancer drug resistance is a challenging phenomenon of growing concern which arises from alteration in drug targets. Despite the fast speed of new chemotherapeutic agent design, the increasing prevalence of this phenomenon requires further research and treatment development. Recently, we reported a new aminopyrimidine compound—namely RDS 344—as a potential innovative anticancer agent. Methods: Herein, we report the design, synthesis, and anti-proliferative activity of new aminopyrimidine derivatives structurally related to RDS 3442 obtained by carrying out substitutions at position 6 of the pyrimidine core and/or on the 2-aniline ring of our hit. The ability to inhibit cell proliferation was evaluated on different types of tumors, glioblastoma, triple-negative breast cancer, oral squamous cell carcinomas and colon cancer plus on human dermal fibroblasts chosen as control of normal cells. Results: The most interesting compound was the N-benzyl counterpart of RDS 3442, namely 2a, that induced a significant decrease in cell viability in all the tested tumor cell lines, with EC50s ranging from 4 and 8 μM, 4–13 times more active of hit. Conclusions: These data suggest a potential role for this class of molecules as promising tool for new approaches in treating cancers of different histotype.
Collapse
Affiliation(s)
- Valentina Noemi Madia
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, “Sapienza” Università di Roma, p.le Aldo Moro 5, 00185 Rome, Italy; (V.N.M.); (A.D.L.); (D.I.); (V.T.); (L.S.); (R.D.S.); (R.C.)
| | - Alice Nicolai
- Department of Sensory Organs, “Sapienza” University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (A.N.); (M.A.); (S.T.)
- Department of Experimental Medicine, “Sapienza” University of Rome, Viale Regina Elena 324, I-00161 Rome, Italy; (L.T.); (S.S.)
| | - Antonella Messore
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, “Sapienza” Università di Roma, p.le Aldo Moro 5, 00185 Rome, Italy; (V.N.M.); (A.D.L.); (D.I.); (V.T.); (L.S.); (R.D.S.); (R.C.)
- Correspondence: ; Tel.: +39-06-4991-3965
| | - Alessandro De Leo
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, “Sapienza” Università di Roma, p.le Aldo Moro 5, 00185 Rome, Italy; (V.N.M.); (A.D.L.); (D.I.); (V.T.); (L.S.); (R.D.S.); (R.C.)
| | - Davide Ialongo
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, “Sapienza” Università di Roma, p.le Aldo Moro 5, 00185 Rome, Italy; (V.N.M.); (A.D.L.); (D.I.); (V.T.); (L.S.); (R.D.S.); (R.C.)
| | - Valeria Tudino
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, “Sapienza” Università di Roma, p.le Aldo Moro 5, 00185 Rome, Italy; (V.N.M.); (A.D.L.); (D.I.); (V.T.); (L.S.); (R.D.S.); (R.C.)
| | - Francesco Saccoliti
- D3 PharmaChemistry, Italian Institute of Technology, Via Morego 30, I-16163 Genova, Italy;
| | - Daniela De Vita
- Department of Environmental Biology, “Sapienza” University of Rome, p.le Aldo Moro 5, I-00185, Rome, Italy;
| | - Luigi Scipione
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, “Sapienza” Università di Roma, p.le Aldo Moro 5, 00185 Rome, Italy; (V.N.M.); (A.D.L.); (D.I.); (V.T.); (L.S.); (R.D.S.); (R.C.)
| | - Marco Artico
- Department of Sensory Organs, “Sapienza” University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (A.N.); (M.A.); (S.T.)
| | - Samanta Taurone
- Department of Sensory Organs, “Sapienza” University of Rome, Viale del Policlinico 155, 00161 Rome, Italy; (A.N.); (M.A.); (S.T.)
| | - Ludovica Taglieri
- Department of Experimental Medicine, “Sapienza” University of Rome, Viale Regina Elena 324, I-00161 Rome, Italy; (L.T.); (S.S.)
| | - Roberto Di Santo
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, “Sapienza” Università di Roma, p.le Aldo Moro 5, 00185 Rome, Italy; (V.N.M.); (A.D.L.); (D.I.); (V.T.); (L.S.); (R.D.S.); (R.C.)
| | - Susanna Scarpa
- Department of Experimental Medicine, “Sapienza” University of Rome, Viale Regina Elena 324, I-00161 Rome, Italy; (L.T.); (S.S.)
| | - Roberta Costi
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, “Sapienza” Università di Roma, p.le Aldo Moro 5, 00185 Rome, Italy; (V.N.M.); (A.D.L.); (D.I.); (V.T.); (L.S.); (R.D.S.); (R.C.)
| |
Collapse
|
27
|
Piñeyro MD, Arias D, Parodi-Talice A, Guerrero S, Robello C. Trypanothione Metabolism as Drug Target for Trypanosomatids. Curr Pharm Des 2021; 27:1834-1846. [PMID: 33308115 DOI: 10.2174/1381612826666201211115329] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 10/01/2020] [Accepted: 10/08/2020] [Indexed: 11/22/2022]
Abstract
Chagas Disease, African sleeping sickness, and leishmaniasis are neglected diseases caused by pathogenic trypanosomatid parasites, which have a considerable impact on morbidity and mortality in poor countries. The available drugs used as treatment have high toxicity, limited access, and can cause parasite drug resistance. Long-term treatments, added to their high toxicity, result in patients that give up therapy. Trypanosomatids presents a unique trypanothione based redox system, which is responsible for maintaining the redox balance. Therefore, inhibition of these essential and exclusive parasite's metabolic pathways, absent from the mammalian host, could lead to the development of more efficient and safe drugs. The system contains different redox cascades, where trypanothione and tryparedoxins play together a central role in transferring reduced power to different enzymes, such as 2-Cys peroxiredoxins, non-selenium glutathione peroxidases, ascorbate peroxidases, glutaredoxins and methionine sulfoxide reductases, through NADPH as a source of electrons. There is sufficient evidence that this complex system is essential for parasite survival and infection. In this review, we explore what is known in terms of essentiality, kinetic and structural data, and the development of inhibitors of enzymes from this trypanothione-based redox system. The recent advances and limitations in the development of lead inhibitory compounds targeting these enzymes have been discussed. The combination of molecular biology, bioinformatics, genomics, and structural biology is fundamental since the knowledge of unique features of the trypanothione-dependent system will provide tools for rational drug design in order to develop better treatments for these diseases.
Collapse
Affiliation(s)
| | - Diego Arias
- Instituto de Agrobiotecnologia del Litoral y Facultad de Bioquimica y Ciencias Biologicas, CONICET-UNL, Santa F, Argentina
| | | | - Sergio Guerrero
- Instituto de Agrobiotecnologia del Litoral y Facultad de Bioquimica y Ciencias Biologicas, CONICET-UNL, Santa F, Argentina
| | - Carlos Robello
- Unidad de Biologia Molecular, Instituto Pasteur Montevideo, Montevideo, Uruguay
| |
Collapse
|
28
|
Saccoliti F, Di Santo R, Costi R. Recent Advancement in the Search of Innovative Antiprotozoal Agents Targeting Trypanothione Metabolism. ChemMedChem 2020; 15:2420-2435. [PMID: 32805075 DOI: 10.1002/cmdc.202000325] [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: 05/12/2020] [Revised: 08/13/2020] [Indexed: 01/28/2023]
Abstract
Leishmania and Trypanosoma parasites are responsible for the challenging neglected tropical diseases leishmaniases, Chagas disease, and human African trypanosomiasis, which account for up to 40,000 deaths annually mainly in developing countries. Current chemotherapy relies on drugs with significant limitations in efficacy and safety, prompting the urgent need to explore innovative approaches to improve the drug discovery pipeline. The unique trypanothione-based redox pathway, which is absent in human hosts, is vital for all trypanosomatids and offers valuable opportunities to guide the rational development of specific, broad-spectrum and innovative anti-trypanosomatid agents. Major efforts focused on the key metabolic enzymes trypanothione synthetase-amidase and trypanothione reductase, whose inhibition should affect the entire pathway and, finally, parasite survival. Herein, we will report and comment on the most recent studies in the search for enzyme inhibitors, underlining the promising opportunities that have emerged so far to drive the exploration of future successful therapeutic approaches.
Collapse
Affiliation(s)
- Francesco Saccoliti
- D3 PharmaChemistry, Italian Institute of Technology, Via Morego 30, 16163, Genova, Italy
| | - Roberto Di Santo
- Dipartimento di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, "Sapienza" Università di Roma, P. le Aldo Moro 5, 00185, Roma, Italy
| | - Roberta Costi
- Dipartimento di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, "Sapienza" Università di Roma, P. le Aldo Moro 5, 00185, Roma, Italy
| |
Collapse
|
29
|
Raj S, Sasidharan S, Balaji SN, Saudagar P. An overview of biochemically characterized drug targets in metabolic pathways of Leishmania parasite. Parasitol Res 2020; 119:2025-2037. [DOI: 10.1007/s00436-020-06736-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 05/28/2020] [Indexed: 12/18/2022]
|
30
|
Crentsil JA, Yamthe LRT, Anibea BZ, Broni E, Kwofie SK, Tetteh JKA, Osei-Safo D. Leishmanicidal Potential of Hardwickiic Acid Isolated From Croton sylvaticus. Front Pharmacol 2020; 11:753. [PMID: 32523532 PMCID: PMC7261830 DOI: 10.3389/fphar.2020.00753] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 05/06/2020] [Indexed: 01/31/2023] Open
Abstract
Leishmania is a parasitic protozoon responsible for the neglected tropical disease Leishmaniasis. Approximately, 350 million people are susceptible and close to 70,000 death cases globally are reported annually. The lack of effective leishmanicides, the emergence of drug resistance and toxicity concerns necessitate the pursuit for effective antileishmanial drugs. Natural compounds serve as reservoirs for discovering new drugs due to their chemical diversity. Hardwickiic acid (HA) isolated from the stembark of Croton sylvaticus was evaluated for its leishmanicidal potential against Leishmania donovani and L. major promastigotes. The susceptibility of the promastigotes to HA was determined using the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide/phenazine methosulfate colorimetric assay with Amphotericin B serving as positive control. HA showed a significant antileishmanial activity on L. donovani promastigotes with an IC50 value of 31.57± 0.06 µM with respect to the control drug, amphotericin B with IC50 of 3.35 ± 0.14 µM). The cytotoxic activity was observed to be CC50 = 247.83 ± 6.32 µM against 29.99 ± 2.82 µM for curcumin, the control, resulting in a selectivity index of SI = 7.85. Molecular modeling, docking and dynamics simulations of selected drug targets corroborated the observed antileishmanial activity of HA. Novel insights into the mechanisms of binding were obtained for trypanothione reductase (TR), pteridine reductase 1 (PTR1), and glutamate cysteine ligase (GCL). The binding affinity of HA to the drug targets LmGCL, LmPTR1, LdTR, LmTR, LdGCL, and LdPTR1 were obtained as -8.0, -7.8, -7.6, -7.5, -7.4 and -7.1 kcal/mol, respectively. The role of Lys16, Ser111, and Arg17 as critical residues required for binding to LdPTR1 was reinforced. HA was predicted as a Caspase-3 stimulant and Caspase-8 stimulant, implying a possible role in apoptosis, which was shown experimentally that HA induced parasite death by loss of membrane integrity. HA was also predicted as antileishmanial molecule corroborating the experimental activity. Therefore, HA is a promising antileishmanial molecule worthy of further development as a biotherapeutic agent.
Collapse
Affiliation(s)
- Justice Afrifa Crentsil
- Department of Chemistry, School of Physical and Mathematical Sciences, College of Basic and Applied Sciences (CBAS), University of Ghana, Accra, Ghana
| | - Lauve Rachel Tchokouaha Yamthe
- Institute for Medical Research and Medicinal Plants Studies, Yaoundé, Cameroon.,Department of Parasitology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana.,Antimicrobial and Biocontrol Agents Unit, Laboratory for Phytobiochemistry and Medicinal Plants Studies, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
| | - Barbara Zenabu Anibea
- Department of Chemistry, School of Physical and Mathematical Sciences, College of Basic and Applied Sciences (CBAS), University of Ghana, Accra, Ghana
| | - Emmanuel Broni
- Department of Biomedical Engineering, School of Engineering Sciences, CBAS, University of Ghana, Accra, Ghana
| | - Samuel Kojo Kwofie
- Department of Biomedical Engineering, School of Engineering Sciences, CBAS, University of Ghana, Accra, Ghana.,West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, CBAS, University of Ghana, Accra, Ghana.,Department of Medicine, Loyola University Medical Center, Maywood, IL, United States.,Department of Physics and Engineering Science, Coastal Carolina University, Conway, SC, United States
| | - John Kweku Amissah Tetteh
- Department of Immunology, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Dorcas Osei-Safo
- Department of Chemistry, School of Physical and Mathematical Sciences, College of Basic and Applied Sciences (CBAS), University of Ghana, Accra, Ghana
| |
Collapse
|
31
|
Battista T, Colotti G, Ilari A, Fiorillo A. Targeting Trypanothione Reductase, a Key Enzyme in the Redox Trypanosomatid Metabolism, to Develop New Drugs against Leishmaniasis and Trypanosomiases. Molecules 2020; 25:E1924. [PMID: 32326257 PMCID: PMC7221613 DOI: 10.3390/molecules25081924] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 01/21/2023] Open
Abstract
The protozoans Leishmania and Trypanosoma, belonging to the same Trypanosomatidae family, are the causative agents of Leishmaniasis, Chagas disease, and human African trypanosomiasis. Overall, these infections affect millions of people worldwide, posing a serious health issue as well as socio-economical concern. Current treatments are inadequate, mainly due to poor efficacy, toxicity, and emerging resistance; therefore, there is an urgent need for new drugs.
Collapse
Affiliation(s)
- Theo Battista
- 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, c/o Department of Biochemical Sciences, Sapienza University, P.le A.Moro 5, 00185 Rome, Italy; (G.C.); (A.I.)
| | - Andrea Ilari
- Institute of Molecular Biology and Pathology, Italian National Research Council, IBPM-CNR, c/o Department of Biochemical Sciences, Sapienza University, P.le A.Moro 5, 00185 Rome, Italy; (G.C.); (A.I.)
| | - Annarita Fiorillo
- Department of Biochemical Sciences, Sapienza University, P.le A.Moro 5, 00185 Rome, Italy;
| |
Collapse
|
32
|
Agostinelli E. Biochemical and pathophysiological properties of polyamines. Amino Acids 2020; 52:111-117. [PMID: 32072296 DOI: 10.1007/s00726-020-02821-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 01/27/2020] [Indexed: 02/07/2023]
Affiliation(s)
- Enzo Agostinelli
- Department of Biochemical Sciences, A. Rossi Fanelli', Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy. .,International Polyamines Foundation 'ETS-ONLUS', Via del Forte Tiburtino 98, 00159, Rome, Italy.
| |
Collapse
|
33
|
Kuldeep J, R. K, Kaur P, Goyal N, Siddiqi MI. Identification of potential anti-leishmanial agents using computational investigation and biological evaluation against trypanothione reductase. J Biomol Struct Dyn 2020; 39:960-969. [DOI: 10.1080/07391102.2020.1721330] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Jitendra Kuldeep
- Molecular & Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Karthik R.
- Biochemistry Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Pavneet Kaur
- Biochemistry Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Neena Goyal
- Biochemistry Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Mohammad Imran Siddiqi
- Molecular & Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
| |
Collapse
|
34
|
Kwofie SK, Broni E, Dankwa B, Enninful KS, Kwarko GB, Darko L, Durvasula R, Kempaiah P, Rathi B, Miller Iii WA, Yaya A, Wilson MD. Outwitting an Old Neglected Nemesis: A Review on Leveraging Integrated Data-Driven Approaches to Aid in Unraveling of Leishmanicides of Therapeutic Potential. Curr Top Med Chem 2020; 20:349-366. [PMID: 31994465 DOI: 10.2174/1568026620666200128160454] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 08/20/2019] [Accepted: 09/12/2019] [Indexed: 11/22/2022]
Abstract
The global prevalence of leishmaniasis has increased with skyrocketed mortality in the past decade. The causative agent of leishmaniasis is Leishmania species, which infects populations in almost all the continents. Prevailing treatment regimens are consistently inefficient with reported side effects, toxicity and drug resistance. This review complements existing ones by discussing the current state of treatment options, therapeutic bottlenecks including chemoresistance and toxicity, as well as drug targets. It further highlights innovative applications of nanotherapeutics-based formulations, inhibitory potential of leishmanicides, anti-microbial peptides and organometallic compounds on leishmanial species. Moreover, it provides essential insights into recent machine learning-based models that have been used to predict novel leishmanicides and also discusses other new models that could be adopted to develop fast, efficient, robust and novel algorithms to aid in unraveling the next generation of anti-leishmanial drugs. A plethora of enriched functional genomic, proteomic, structural biology, high throughput bioassay and drug-related datasets are currently warehoused in both general and leishmania-specific databases. The warehoused datasets are essential inputs for training and testing algorithms to augment the prediction of biotherapeutic entities. In addition, we demonstrate how pharmacoinformatics techniques including ligand-, structure- and pharmacophore-based virtual screening approaches have been utilized to screen ligand libraries against both modeled and experimentally solved 3D structures of essential drug targets. In the era of data-driven decision-making, we believe that highlighting intricately linked topical issues relevant to leishmanial drug discovery offers a one-stop-shop opportunity to decipher critical literature with the potential to unlock implicit breakthroughs.
Collapse
Affiliation(s)
- Samuel K Kwofie
- Department of Biomedical Engineering, School of Engineering Sciences, College of Basic & Applied Sciences, University of Ghana, PMB LG 77, Legon, Accra, Ghana.,West African Center for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana.,Department of Medicine, Loyola University Chicago, Loyola University Medical Center, Maywood, IL 60153, United States
| | - Emmanuel Broni
- Department of Biomedical Engineering, School of Engineering Sciences, College of Basic & Applied Sciences, University of Ghana, PMB LG 77, Legon, Accra, Ghana
| | - Bismark Dankwa
- Department of Parasitology, Noguchi Memorial Institute for Medical Research (NMIMR), College of Health Sciences (CHS), University of Ghana, Legon, Accra, Ghana
| | - Kweku S Enninful
- Department of Parasitology, Noguchi Memorial Institute for Medical Research (NMIMR), College of Health Sciences (CHS), University of Ghana, Legon, Accra, Ghana
| | - Gabriel B Kwarko
- West African Center for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Louis Darko
- Department of Biomedical Engineering, School of Engineering Sciences, College of Basic & Applied Sciences, University of Ghana, PMB LG 77, Legon, Accra, Ghana
| | - Ravi Durvasula
- Department of Medicine, Loyola University Chicago, Loyola University Medical Center, Maywood, IL 60153, United States
| | - Prakasha Kempaiah
- Department of Medicine, Loyola University Chicago, Loyola University Medical Center, Maywood, IL 60153, United States
| | - Brijesh Rathi
- Department of Medicine, Loyola University Chicago, Loyola University Medical Center, Maywood, IL 60153, United States.,Department of Chemistry, Hansraj College University Enclave, University of Delhi, Delhi, 110007, India
| | - Whelton A Miller Iii
- Department of Medicine, Loyola University Chicago, Loyola University Medical Center, Maywood, IL 60153, United States.,Department of Chemistry, Physics, & Engineering, Lincoln University, Lincoln University, PA 19352, United States.,Department of Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Abu Yaya
- Department of Materials Science and Engineering, College of Basic & Applied Sciences, University of Ghana, Legon, Ghana
| | - Michael D Wilson
- Department of Medicine, Loyola University Chicago, Loyola University Medical Center, Maywood, IL 60153, United States.,Department of Parasitology, Noguchi Memorial Institute for Medical Research (NMIMR), College of Health Sciences (CHS), University of Ghana, Legon, Accra, Ghana
| |
Collapse
|
35
|
Lee SM, Kim MS, Hayat F, Shin D. Recent Advances in the Discovery of Novel Antiprotozoal Agents. Molecules 2019; 24:E3886. [PMID: 31661934 PMCID: PMC6864685 DOI: 10.3390/molecules24213886] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 10/15/2019] [Accepted: 10/23/2019] [Indexed: 11/16/2022] Open
Abstract
Parasitic diseases have serious health, social, and economic impacts, especially in the tropical regions of the world. Diseases caused by protozoan parasites are responsible for considerable mortality and morbidity, affecting more than 500 million people worldwide. Globally, the burden of protozoan diseases is increasing and is been exacerbated because of a lack of effective medication due to the drug resistance and toxicity of current antiprotozoal agents. These limitations have prompted many researchers to search for new drugs against protozoan parasites. In this review, we have compiled the latest information (2012-2017) on the structures and pharmacological activities of newly developed organic compounds against five major protozoan diseases, giardiasis, leishmaniasis, malaria, trichomoniasis, and trypanosomiasis, with the aim of showing recent advances in the discovery of new antiprotozoal drugs.
Collapse
Affiliation(s)
- Seong-Min Lee
- College of Pharmacy, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Korea.
| | - Min-Sun Kim
- College of Pharmacy, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Korea.
| | - Faisal Hayat
- College of Pharmacy, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Korea.
| | - Dongyun Shin
- College of Pharmacy, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Korea.
| |
Collapse
|
36
|
Matadamas-Martínez F, Hernández-Campos A, Téllez-Valencia A, Vázquez-Raygoza A, Comparán-Alarcón S, Yépez-Mulia L, Castillo R. Leishmania mexicana Trypanothione Reductase Inhibitors: Computational and Biological Studies. MOLECULES (BASEL, SWITZERLAND) 2019; 24:molecules24183216. [PMID: 31487860 PMCID: PMC6767256 DOI: 10.3390/molecules24183216] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/24/2019] [Accepted: 08/31/2019] [Indexed: 12/27/2022]
Abstract
Leishmanicidal drugs have many side effects, and drug resistance to all of them has been documented. Therefore, the development of new drugs and the identification of novel therapeutic targets are urgently needed. Leishmania mexicana trypanothione reductase (LmTR), a NADPH-dependent flavoprotein oxidoreductase important to thiol metabolism, is essential for parasite viability. Its absence in the mammalian host makes this enzyme an attractive target for the development of new anti-Leishmania drugs. Herein, a tridimensional model of LmTR was constructed and the molecular docking of 20 molecules from a ZINC database was performed. Five compounds (ZINC04684558, ZINC09642432, ZINC12151998, ZINC14970552, and ZINC11841871) were selected (docking scores -10.27 kcal/mol to -5.29 kcal/mol and structurally different) and evaluated against recombinant LmTR (rLmTR) and L. mexicana promastigote. Additionally, molecular dynamics simulation of LmTR-selected compound complexes was achieved. The five selected compounds inhibited rLmTR activity in the range of 32.9% to 40.1%. The binding of selected compounds to LmTR involving different hydrogen bonds with distinct residues of the molecule monomers A and B is described. Compound ZINC12151998 (docking score -10.27 kcal/mol) inhibited 32.9% the enzyme activity (100 µM) and showed the highest leishmanicidal activity (IC50 = 58 µM) of all the selected compounds. It was more active than glucantime, and although its half-maximal cytotoxicity concentration (CC50 = 53 µM) was higher than that of the other four compounds, it was less cytotoxic than amphotericin B. Therefore, compound ZINC12151998 provides a promising starting point for a hit-to-lead process in our search for new anti-Leishmania drugs that are more potent and less cytotoxic.
Collapse
Affiliation(s)
- Félix Matadamas-Martínez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, Unidad Médica de Alta Especialidad-Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Alicia Hernández-Campos
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Alfredo Téllez-Valencia
- Facultad de Medicina y Nutrición, Universidad Juárez del Estado de Durango Av. Universidad y Fanny Anitúa S/N, Durango 34000, Mexico
| | - Alejandra Vázquez-Raygoza
- Facultad de Medicina y Nutrición, Universidad Juárez del Estado de Durango Av. Universidad y Fanny Anitúa S/N, Durango 34000, Mexico
| | - Sandra Comparán-Alarcón
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, Unidad Médica de Alta Especialidad-Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Lilián Yépez-Mulia
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, Unidad Médica de Alta Especialidad-Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico.
| | - Rafael Castillo
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico.
| |
Collapse
|
37
|
De Gasparo R, Halgas O, Harangozo D, Kaiser M, Pai EF, Krauth‐Siegel RL, Diederich F. Targeting a Large Active Site: Structure‐Based Design of Nanomolar Inhibitors of
Trypanosoma brucei
Trypanothione Reductase. Chemistry 2019; 25:11416-11421. [DOI: 10.1002/chem.201901664] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/03/2019] [Indexed: 01/16/2023]
Affiliation(s)
- Raoul De Gasparo
- Laboratorium für Organische ChemieETH Zurich Vladimir-Prelog-Weg 3 8093 Zurich Switzerland
| | - Ondrej Halgas
- Departments of Biochemistry and Medical BiophysicsUniversity of Toronto Medical Sciences Building, 5318, 1 King's College Circle Toronto ON M5S 1A8 Canada
- The Campbell Family Institute for Cancer ResearchUniversity Health Network 101 College Street Toronto ON M5G 1L7 Canada
| | - Dora Harangozo
- Laboratorium für Organische ChemieETH Zurich Vladimir-Prelog-Weg 3 8093 Zurich Switzerland
| | - Marcel Kaiser
- Swiss Tropical and Public Health Institute Socinstrasse 57 4002 Basel Switzerland
- University of Basel Petersplatz 1 4003 Basel Switzerland
| | - Emil F. Pai
- Departments of Biochemistry and Medical BiophysicsUniversity of Toronto Medical Sciences Building, 5318, 1 King's College Circle Toronto ON M5S 1A8 Canada
- The Campbell Family Institute for Cancer ResearchUniversity Health Network 101 College Street Toronto ON M5G 1L7 Canada
| | - R. Luise Krauth‐Siegel
- Biochemie-Zentrum Heidelberg (BZH)Universität Heidelberg Im Neuenheimer Feld 328 69120 Heidelberg Germany
| | - François Diederich
- Laboratorium für Organische ChemieETH Zurich Vladimir-Prelog-Weg 3 8093 Zurich Switzerland
| |
Collapse
|
38
|
Majid Shah S, Ullah F, Ayaz M, Sadiq A, Hussain S, Ali Shah AUH, Adnan Ali Shah S, Wadood A, Nadhman A. β-Sitosterol from Ifloga spicata (Forssk.) Sch. Bip. as potential anti-leishmanial agent against leishmania tropica: Docking and molecular insights. Steroids 2019; 148:56-62. [PMID: 31085212 DOI: 10.1016/j.steroids.2019.05.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 04/27/2019] [Accepted: 05/08/2019] [Indexed: 01/20/2023]
Abstract
The current study was aimed to evaluate the anti-leishmanial potentials of β-sitosterol isolated from Ifloga spicata. The anti-leishmanial potential of β-sitosterol is well documented against Leishmania donovani and Leishmania amazonensis but unexplored against Leishmania tropica. Structure of the compound was elucidated by FT-IR, mass spectrometry and multinuclear (1H and 13C) magnetic resonance spectroscopy. The compound was evaluated for its anti-leishmanial potentials against L. tropica KWH23 using in vitro anti-promastigote, DNA interaction, apoptosis, docking studies against leishmanolysin (GP63) and trypanothione reductase (TR) receptors using MOE 2016 software. β-sitosterol exhibited significant activity against leishmania promastigotes with IC50 values of 9.2 ± 0.06 μg/mL. The standard drug glucantaime showed IC50 of 5.33 ± 0.07 µg/mL. Further mechanistic studies including DNA targeting and apoptosis induction via acridine orange assay exhibited promising anti-leishmanial potentials for β-sitosterol. Molecular docking with leishmanolysin (GP63) and trypanothione reductase (TR) receptors displayed the binding scores of β-sitosterol with targets TR and GP63 were -7.659 and -6.966 respectively. The low binding energies -61.54 (for TR) and -33.24 (for GP63) indicate that it strongly bind to the active sites of target receptors. The results confirmed that β-sitosterol have considerable anti-leishmanial potentials and need further studies as potential natural anti-leishmanial agent against L. tropica.
Collapse
Affiliation(s)
- Syed Majid Shah
- Department of Pharmacy, University of Malakand, Khyber Pakhtunkhwa 18800, Pakistan; Department of Pharmacy, Kohat University of Science & Technology, Kohat 26000, Khyber Pakhtunkhwa, Pakistan
| | - Farhat Ullah
- Department of Pharmacy, University of Malakand, Khyber Pakhtunkhwa 18800, Pakistan.
| | - Muhammad Ayaz
- Department of Pharmacy, University of Malakand, Khyber Pakhtunkhwa 18800, Pakistan.
| | - Abdul Sadiq
- Department of Pharmacy, University of Malakand, Khyber Pakhtunkhwa 18800, Pakistan
| | - Sajid Hussain
- Department of Pharmacy, University of Malakand, Khyber Pakhtunkhwa 18800, Pakistan; Department of Pharmacy, Kohat University of Science & Technology, Kohat 26000, Khyber Pakhtunkhwa, Pakistan
| | - Azhar-Ul-Haq Ali Shah
- Department of Chemistry, Kohat University of Science & Technology, Kohat 26000, Khyber Pakhtunkhwa Pakistan
| | - Syed Adnan Ali Shah
- Faculty of Pharmacy, Universiti Teknologi MARA Cawangan Selangor Kampus Puncak Alam, Bandar Puncak Alam, Selangor 42300, Malaysia; Atta-ur-Rahman Institute for Natural Products Discovery (AuRIns), Universiti Teknologi MARA Cawangan Selangor Kampus Puncak Alam, Bandar Puncak Alam, Selangor 42300, Malaysia
| | - Abdul Wadood
- Department of Biochemistry, UCS, Shankar Abdul Wali Khan University, Mardan 23200, Pakistan.
| | - Akhtar Nadhman
- Institute of Integrative Biosciences IIB, CECOS University, Peshawar Pakistan
| |
Collapse
|
39
|
Colotti G, Saccoliti F, Gramiccia M, Di Muccio T, Prakash J, Yadav S, Dubey VK, Vistoli G, Battista T, Mocci S, Fiorillo A, Bibi A, Madia VN, Messore A, Costi R, Di Santo R, Ilari A. Structure-guided approach to identify a novel class of anti-leishmaniasis diaryl sulfide compounds targeting the trypanothione metabolism. Amino Acids 2019; 52:247-259. [PMID: 31037461 DOI: 10.1007/s00726-019-02731-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/03/2019] [Indexed: 01/03/2023]
Abstract
Leishmania protozoans are the causative agent of leishmaniasis, a neglected tropical disease consisting of three major clinical forms: visceral leishmaniasis (VL), cutaneous leishmaniasis, and mucocutaneous leishmaniasis. VL is caused by Leishmania donovani in East Africa and the Indian subcontinent and by Leishmania infantum in Europe, North Africa, and Latin America, and causes an estimated 60,000 deaths per year. Trypanothione reductase (TR) is considered to be one of the best targets to find new drugs against leishmaniasis. This enzyme is fundamental for parasite survival in the human host since it reduces trypanothione, a molecule used by the tryparedoxin/tryparedoxin peroxidase system of Leishmania to neutralize the hydrogen peroxide produced by host macrophages during infection. Recently, we solved the X-ray structure of TR in complex with the diaryl sulfide compound RDS 777 (6-(sec-butoxy)-2-((3-chlorophenyl)thio)pyrimidin-4-amine), which impairs the parasite defense against the reactive oxygen species by inhibiting TR with high efficiency. The compound binds to the catalytic site and engages in hydrogen bonds the residues more involved in the catalysis, namely Glu466', Cys57 and Cys52, thereby inhibiting the trypanothione binding. On the basis of the RDS 777-TR complex, we synthesized structurally related diaryl sulfide analogs as TR inhibitors able to compete for trypanothione binding to the enzyme and to kill the promastigote in the micromolar range. One of the most active among these compounds (RDS 562) was able to reduce the trypanothione concentration in cell of about 33% via TR inhibition. RDS 562 inhibits selectively Leishmania TR, while it does not inhibit the human homolog glutathione reductase.
Collapse
Affiliation(s)
- Gianni Colotti
- Istituto di Biologia e Patologia Molecolari, Consiglio Nazionale delle Ricerche, IBPM-CNR, c/o Dip. Scienze Biochimiche Università Sapienza, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Francesco Saccoliti
- Dip. Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, Università Sapienza, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Marina Gramiccia
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità Viale Regina Elena, 299, 00161, Rome, Italy
| | - Trentina Di Muccio
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità Viale Regina Elena, 299, 00161, Rome, Italy
| | - Jay Prakash
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, 981039, India
| | - Sunita Yadav
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University) Varanasi, Varanasi, 221005, India
| | - Vikash Kumar Dubey
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University) Varanasi, Varanasi, 221005, India
| | - Giulio Vistoli
- Dip. di Scienze Farmaceutiche, Università degli Studi di Milano, via Mangiagalli 25, 20133, Milan, Italy
| | - Theo Battista
- Dip. Scienze Biochimiche, Università Sapienza, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Stefano Mocci
- Dip. Scienze Biochimiche, Università Sapienza, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Annarita Fiorillo
- Dip. Scienze Biochimiche, Università Sapienza, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Aasia Bibi
- Dip. Scienze Biochimiche, Università Sapienza, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Valentina Noemi Madia
- Dip. Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, Università Sapienza, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Antonella Messore
- Dip. Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, Università Sapienza, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Roberta Costi
- Dip. Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, Università Sapienza, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Roberto Di Santo
- Dip. Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, Università Sapienza, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Andrea Ilari
- Istituto di Biologia e Patologia Molecolari, Consiglio Nazionale delle Ricerche, IBPM-CNR, c/o Dip. Scienze Biochimiche Università Sapienza, P.le Aldo Moro 5, 00185, Rome, Italy.
| |
Collapse
|
40
|
Synthesis and Leishmanicidal Activity of Novel Urea, Thiourea, and Selenourea Derivatives of Diselenides. Antimicrob Agents Chemother 2019; 63:AAC.02200-18. [PMID: 30782984 DOI: 10.1128/aac.02200-18] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 02/13/2019] [Indexed: 11/20/2022] Open
Abstract
A novel series of thirty-one N-substituted urea, thiourea, and selenourea derivatives containing diphenyldiselenide entities were synthesized, fully characterized by spectroscopic and analytical methods, and screened for their in vitro leishmanicidal activities. The cytotoxic activity of these derivatives was tested against Leishmania infantum axenic amastigotes, and selectivity was assessed in human THP-1 cells. Thirteen of the synthesized compounds showed a significant antileishmanial activity, with 50% effective concentration (EC50) values lower than that for the reference drug miltefosine (EC50, 2.84 μM). In addition, the derivatives 9, 11, 42, and 47, with EC50 between 1.1 and 1.95 μM, also displayed excellent selectivity (selectivity index ranged from 12.4 to 22.7) and were tested against infected macrophages. Compound 11, a derivative with a cyclohexyl chain, exhibited the highest activity against intracellular amastigotes, with EC50 values similar to those observed for the standard drug edelfosine. Structure-activity relationship analyses revealed that N-aliphatic substitution in urea and selenourea is recommended for the leishmanicidal activity of these analogs. Preliminary studies of the mechanism of action for the hit compounds was carried out by measuring their ability to inhibit trypanothione reductase. Even though the obtained results suggest that this enzyme is not the target for most of these derivatives, their activity comparable to that of the standards and lack of toxicity in THP-1 cells highlight the potential of these compounds to be optimized for leishmaniasis treatment.
Collapse
|
41
|
Taglieri L, Saccoliti F, Nicolai A, Peruzzi G, Madia VN, Tudino V, Messore A, Di Santo R, Artico M, Taurone S, Salvati M, Costi R, Scarpa S. Discovery of a pyrimidine compound endowed with antitumor activity. Invest New Drugs 2019; 38:39-49. [PMID: 30900116 DOI: 10.1007/s10637-019-00762-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 03/08/2019] [Indexed: 02/07/2023]
Abstract
Recently, some synthetic nitrogen-based heterocyclic molecules, such as PJ34, have shown pronounced antitumor activity. Therefore, we designed and synthesized new derivatives characterized by a nitrogen-containing scaffold and evaluated their antiproliferative properties in tumor cells. We herein report the effects of three newly synthesized compounds on cell lines from three different human cancers: triple-negative breast cancer, colon carcinoma and glioblastoma. We found that two of these compounds did not affect proliferation, while the third significantly inhibited replication of the three cell lines. Moreover, this third molecule at 20 μM led to the upregulation of p21 and p27 and blockage of the cell cycle at G0/G1; in addition, it induced apoptosis in all three cell lines when used at higher concentrations (30-50 μM). The results demonstrate that this compound is a potent inhibitor of replication, an inducer of apoptosis and a negative regulator of cell cycle progression for cancer cells of different histotypes. Our data suggest a potential role for this new molecule as an interesting and powerful tool for new approaches in treating various cancers.
Collapse
Affiliation(s)
- Ludovica Taglieri
- Department of Experimental Medicine, Sapienza University, Viale Regina Elena 324, 00161, Rome, Italy
| | - Francesco Saccoliti
- Dipartimento di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Alice Nicolai
- Department of Experimental Medicine, Sapienza University, Viale Regina Elena 324, 00161, Rome, Italy
- Department of Sensory Organs, Sapienza University, Viale del Policlinico 155, 00161, Rome, Italy
| | - Giovanna Peruzzi
- Italian Institute of Technology, Center for Life Nanoscience@Sapienza, Viale Regina Elena 324, 00161, Rome, Italy
| | - Valentina Noemi Madia
- Dipartimento di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Valeria Tudino
- Dipartimento di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Antonella Messore
- Dipartimento di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Roberto Di Santo
- Dipartimento di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Marco Artico
- Department of Sensory Organs, Sapienza University, Viale del Policlinico 155, 00161, Rome, Italy
| | - Samanta Taurone
- Department of Sensory Organs, Sapienza University, Viale del Policlinico 155, 00161, Rome, Italy
| | - Maurizio Salvati
- Department of Human Neurosciences, Sapienza University, Viale Regina Elena 324, 00161, Rome, Italy
| | - Roberta Costi
- Dipartimento di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy.
| | - Susanna Scarpa
- Department of Experimental Medicine, Sapienza University, Viale Regina Elena 324, 00161, Rome, Italy
| |
Collapse
|
42
|
Kumar A, Chauhan N, Singh S. Understanding the Cross-Talk of Redox Metabolism and Fe-S Cluster Biogenesis in Leishmania Through Systems Biology Approach. Front Cell Infect Microbiol 2019; 9:15. [PMID: 30778378 PMCID: PMC6369582 DOI: 10.3389/fcimb.2019.00015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/17/2019] [Indexed: 11/13/2022] Open
Abstract
Leishmania parasites possess an exceptional oxidant and chemical defense mechanism, involving a very unique small molecular weight thiol, trypanothione (T[SH]2), that helps the parasite to manage its survival inside the host macrophage. The reduced state of T[SH]2 is maintained by NADPH-dependent trypanothione reductase (TryR) by recycling trypanothione disulfide (TS2). Along with its most important role as central reductant, T[SH]2 have also been assumed to regulate the activation of iron-sulfur cluster proteins (Fe/S). Fe/S clusters are versatile cofactors of various proteins and execute a much broader range of essential biological processes viz., TCA cycle, redox homeostasis, etc. Although, several Fe/S cluster proteins and their roles have been identified in Leishmania, some of the components of how T[SH]2 is involved in the regulation of Fe/S proteins remains to be explored. In pursuit of this aim, a systems biology approach was undertaken to get an insight into the overall picture to unravel how T[SH]2 synthesis and reduction is linked with the regulation of Fe/S cluster proteins and controls the redox homeostasis at a larger scale. In the current study, we constructed an in silico kinetic model of T[SH]2 metabolism. T[SH]2 reduction reaction was introduced with a perturbation in the form of its inhibition to predict the overall behavior of the model. The main control of reaction fluxes were exerted by TryR reaction rate that affected almost all the important reactions in the model. It was observed that the model was more sensitive to the perturbation introduced in TryR reaction, 5 to 6-fold. Furthermore, due to inhibition, the T[SH]2 synthesis rate was observed to be gradually decreased by 8 to 14-fold. This has also caused an elevated level of free radicals which apparently affected the activation of Fe/S cluster proteins. The present kinetic model has demonstrated the importance of T[SH]2 in leishmanial cellular redox metabolism. Hence, we suggest that, by designing highly potent and specific inhibitors of TryR enzyme, inhibition of T[SH]2 reduction and overall inhibition of most of the downstream pathways including Fe/S protein activation reactions, can be accomplished.
Collapse
|
43
|
Saccoliti F, Madia VN, Tudino V, De Leo A, Pescatori L, Messore A, De Vita D, Scipione L, Brun R, Kaiser M, Mäser P, Calvet CM, Jennings GK, Podust LM, Pepe G, Cirilli R, Faggi C, Di Marco A, Battista MR, Summa V, Costi R, Di Santo R. Design, Synthesis, and Biological Evaluation of New 1-(Aryl-1 H-pyrrolyl)(phenyl)methyl-1 H-imidazole Derivatives as Antiprotozoal Agents. J Med Chem 2019; 62:1330-1347. [PMID: 30615444 DOI: 10.1021/acs.jmedchem.8b01464] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We have designed and synthesized a series of new imidazole-based compounds structurally related to an antiprotozoal agent with nanomolar activity which we identified recently. The new analogues possess micromolar activities against Trypanosoma brucei rhodesiense and Leishmania donovani and nanomolar potency against Plasmodium falciparum. Most of the analogues displayed IC50 within the low nanomolar range against Trypanosoma cruzi, with very high selectivity toward the parasite. Discussion of structure-activity relationships and in vitro biological data for the new compounds are provided against a number of different protozoa. The mechanism of action for the most potent derivatives (5i, 6a-c, and 8b) was assessed by a target-based assay using recombinant T. cruzi CYP51. Bioavailability and efficacy of selected hits were assessed in a T. cruzi mouse model, where 6a and 6b reduced parasitemia in animals >99% following intraperitoneal administration of 25 mg/kg/day dose for 4 consecutive days.
Collapse
Affiliation(s)
- Francesco Saccoliti
- Istituto Pasteur-Fondazione Cenci Bolognetti , Dipartimento di Chimica e Tecnologie del Farmaco, "Sapienza" Università di Roma , p. le Aldo Moro 5 , I-00185 Rome , Italy
| | - Valentina Noemi Madia
- Istituto Pasteur-Fondazione Cenci Bolognetti , Dipartimento di Chimica e Tecnologie del Farmaco, "Sapienza" Università di Roma , p. le Aldo Moro 5 , I-00185 Rome , Italy
| | - Valeria Tudino
- Istituto Pasteur-Fondazione Cenci Bolognetti , Dipartimento di Chimica e Tecnologie del Farmaco, "Sapienza" Università di Roma , p. le Aldo Moro 5 , I-00185 Rome , Italy
| | - Alessandro De Leo
- Istituto Pasteur-Fondazione Cenci Bolognetti , Dipartimento di Chimica e Tecnologie del Farmaco, "Sapienza" Università di Roma , p. le Aldo Moro 5 , I-00185 Rome , Italy
| | - Luca Pescatori
- Istituto Pasteur-Fondazione Cenci Bolognetti , Dipartimento di Chimica e Tecnologie del Farmaco, "Sapienza" Università di Roma , p. le Aldo Moro 5 , I-00185 Rome , Italy
| | - Antonella Messore
- Istituto Pasteur-Fondazione Cenci Bolognetti , Dipartimento di Chimica e Tecnologie del Farmaco, "Sapienza" Università di Roma , p. le Aldo Moro 5 , I-00185 Rome , Italy
| | - Daniela De Vita
- Istituto Pasteur-Fondazione Cenci Bolognetti , Dipartimento di Chimica e Tecnologie del Farmaco, "Sapienza" Università di Roma , p. le Aldo Moro 5 , I-00185 Rome , Italy
| | - Luigi Scipione
- Istituto Pasteur-Fondazione Cenci Bolognetti , Dipartimento di Chimica e Tecnologie del Farmaco, "Sapienza" Università di Roma , p. le Aldo Moro 5 , I-00185 Rome , Italy
| | - Reto Brun
- Swiss Tropical and Public Health Institute , Socinstrasse 57 , CH-4002 Basel , Switzerland
| | - Marcel Kaiser
- Swiss Tropical and Public Health Institute , Socinstrasse 57 , CH-4002 Basel , Switzerland
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute , Socinstrasse 57 , CH-4002 Basel , Switzerland
| | - Claudia M Calvet
- Skaggs School of Pharmacy and Pharmaceutical Sciences , University of California San Diego , La Jolla , California 92093 , United States.,Laboratório de Ultraestrutura Celular , Instituto Oswaldo Cruz (IOC), FIOCRUZ, Rio de Janeiro , Rio de Janeiro 21040-360 , Brazil
| | - Gareth K Jennings
- Skaggs School of Pharmacy and Pharmaceutical Sciences , University of California San Diego , La Jolla , California 92093 , United States
| | - Larissa M Podust
- Skaggs School of Pharmacy and Pharmaceutical Sciences , University of California San Diego , La Jolla , California 92093 , United States
| | - Giacomo Pepe
- Dipartimento di Farmacia , Università di Salerno , Via Giovanni Paolo II 132 , I-84084 Fisciano , Salerno , Italy
| | - Roberto Cirilli
- Centro Nazionale per il Controllo e la Valutazione dei Farmaci , Istituto Superiore di Sanita , Viale Regina Elena 299 , I-00161 Rome , Italy
| | - Cristina Faggi
- Dipartimento di Chimica , Università degli studi di Firenze , Via della Lastruccia 13 , I-50019 , Sesto Fiorentino , Florence , Italy
| | - Annalise Di Marco
- Drug Discovery , IRBM Science Park , Via Pontina km 30,600 , Pomezia, Rome 00071 , Italy
| | - Maria Rosaria Battista
- Drug Discovery , IRBM Science Park , Via Pontina km 30,600 , Pomezia, Rome 00071 , Italy
| | - Vincenzo Summa
- Drug Discovery , IRBM Science Park , Via Pontina km 30,600 , Pomezia, Rome 00071 , Italy
| | - Roberta Costi
- Istituto Pasteur-Fondazione Cenci Bolognetti , Dipartimento di Chimica e Tecnologie del Farmaco, "Sapienza" Università di Roma , p. le Aldo Moro 5 , I-00185 Rome , Italy
| | - Roberto Di Santo
- Istituto Pasteur-Fondazione Cenci Bolognetti , Dipartimento di Chimica e Tecnologie del Farmaco, "Sapienza" Università di Roma , p. le Aldo Moro 5 , I-00185 Rome , Italy
| |
Collapse
|
44
|
Identification and binding mode of a novel Leishmania Trypanothione reductase inhibitor from high throughput screening. PLoS Negl Trop Dis 2018; 12:e0006969. [PMID: 30475811 PMCID: PMC6283646 DOI: 10.1371/journal.pntd.0006969] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 12/06/2018] [Accepted: 11/03/2018] [Indexed: 11/30/2022] Open
Abstract
Trypanothione reductase (TR) is considered to be one of the best targets to find new drugs against Leishmaniasis. This enzyme is fundamental for parasite survival in the host since it reduces trypanothione, a molecule used by the tryparedoxin/tryparedoxin peroxidase system of Leishmania to neutralize hydrogen peroxide produced by host macrophages during infection. In order to identify new lead compounds against Leishmania we developed and validated a new luminescence-based high-throughput screening (HTS) assay that allowed us to screen a library of 120,000 compounds. We identified a novel chemical class of TR inhibitors, able to kill parasites with an IC50 in the low micromolar range. The X-ray crystal structure of TR in complex with a compound from this class (compound 3) allowed the identification of its binding site in a pocket at the entrance of the NADPH binding site. Since the binding site of compound 3 identified by the X-ray structure is unique, and is not present in human homologs such as glutathione reductase (hGR), it represents a new target for drug discovery efforts. Human leishmaniasis is one of the most diffused neglected vector-borne diseases and causes 60,000 deaths annually, a rate surpassed only by malaria among parasitic diseases. Anti-Leishmania treatments are unsatisfactory in terms of their safety and efficacy and there is an urgent need to find treatments. Compounds targeting proteins that are essential for parasite survival but that are not present in the human host are of especial interest with a view to developing selective and non-toxic drugs. Leishmania uses trypanothione as its main detoxifying molecule, allowing the parasite to neutralize the reactive oxygen species produced by macrophages during the infection. Trypanothione is activated by Trypanothione reductase (TR), an enzyme that is absent in man but that is essential for parasite survival, and is therefore considered an attractive target. The new luminescence-based high-throughput screening assay that we have developed and validated allowed us to identify new TR inhibitors by screening a collection of 120,000 compounds. Hit follow-up led to a prototype molecule, compound 3, that we have shown is able to bind in a cavity at the entrance of the NADPH binding site, thereby inhibiting TR. Compound 3 is not able to inhibit the human homolog glutathione reductase (hGR) since the residues lining its NADPH binding cavity are not conserved with respect to TR. Based on their mechanism of action, compounds from the class represented by compound 3 are useful leads for the design new drugs against leishmaniasis.
Collapse
|
45
|
Saccoliti F, Madia VN, Tudino V, De Leo A, Pescatori L, Messore A, De Vita D, Scipione L, Brun R, Kaiser M, Mäser P, Calvet CM, Jennings GK, Podust LM, Costi R, Di Santo R. Biological evaluation and structure-activity relationships of imidazole-based compounds as antiprotozoal agents. Eur J Med Chem 2018; 156:53-60. [PMID: 30006174 DOI: 10.1016/j.ejmech.2018.06.063] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/25/2018] [Accepted: 06/27/2018] [Indexed: 11/19/2022]
Abstract
We discovered a series of azole antifungal compounds as effective antiprotozoal agents. They displayed promising inhibitory activities within the micromolar-submicromolar range against P. falciparum, L. donovani, and T. b. rhodesiense. Moreover, most of such compounds showed excellent nanomolar IC50 against T. cruzi, showing also very low cytotoxicity. Discussion of structure-activity relationships and biological data for these compounds are provided against the different parasites. To assess the mechanism of action against T. cruzi we proved that the most potent compounds (3b, 3j-l) inhibited the T. cruzi CYP51. Moreover, the most active derivative 3j dramatically reduced parasitemia in T. cruzi mouse model without acute toxicity.
Collapse
Affiliation(s)
- Francesco Saccoliti
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, "Sapienza" Università di Roma, p.le Aldo Moro 5, I-00185, Rome, Italy.
| | - Valentina Noemi Madia
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, "Sapienza" Università di Roma, p.le Aldo Moro 5, I-00185, Rome, Italy.
| | - Valeria Tudino
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, "Sapienza" Università di Roma, p.le Aldo Moro 5, I-00185, Rome, Italy.
| | - Alessandro De Leo
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, "Sapienza" Università di Roma, p.le Aldo Moro 5, I-00185, Rome, Italy.
| | - Luca Pescatori
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, "Sapienza" Università di Roma, p.le Aldo Moro 5, I-00185, Rome, Italy.
| | - Antonella Messore
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, "Sapienza" Università di Roma, p.le Aldo Moro 5, I-00185, Rome, Italy.
| | - Daniela De Vita
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, "Sapienza" Università di Roma, p.le Aldo Moro 5, I-00185, Rome, Italy.
| | - Luigi Scipione
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, "Sapienza" Università di Roma, p.le Aldo Moro 5, I-00185, Rome, Italy.
| | - Reto Brun
- Swiss Tropical and Public Health Institute, Socinstrasse 57, CH-4002, Basel, Switzerland.
| | - Marcel Kaiser
- Swiss Tropical and Public Health Institute, Socinstrasse 57, CH-4002, Basel, Switzerland.
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute, Socinstrasse 57, CH-4002, Basel, Switzerland.
| | - Claudia Magalhaes Calvet
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, 92093, USA; Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, RJ, 21040-360, Brazil.
| | - Gareth K Jennings
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Larissa M Podust
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Roberta Costi
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, "Sapienza" Università di Roma, p.le Aldo Moro 5, I-00185, Rome, Italy.
| | - Roberto Di Santo
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, "Sapienza" Università di Roma, p.le Aldo Moro 5, I-00185, Rome, Italy.
| |
Collapse
|
46
|
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
|
47
|
Ortalli M, Ilari A, Colotti G, De Ionna I, Battista T, Bisi A, Gobbi S, Rampa A, Di Martino RMC, Gentilomi GA, Varani S, Belluti F. Identification of chalcone-based antileishmanial agents targeting trypanothione reductase. Eur J Med Chem 2018; 152:527-541. [PMID: 29758517 DOI: 10.1016/j.ejmech.2018.04.057] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/27/2018] [Accepted: 04/28/2018] [Indexed: 12/11/2022]
Abstract
All currently used first-line and second-line drugs for the treatment of leishmaniasis exhibit several drawbacks including toxicity, high costs and route of administration. Furthermore, some drugs are associated with the emergence of drug resistance. Thus, the development of new treatments for leishmaniasis is a priority in the field of neglected tropical diseases. The present work highlights the use of natural derived products, i.e. chalcones, as potential source of antileishmanial agents. Thirty-one novel chalcone compounds have been synthesized and their activity has been evaluated against promastigotes of Leishmania donovani; 16 compounds resulted active against L. donovani in a range from 3.0 to 21.5 μM, showing low toxicity against mammalian cells. Among these molecules, 6 and 16 showed good inhibitory activity on both promastigotes and intracellular amastigotes, coupled with an high selectivity index. Furthermore, compounds 6 and 16 inhibited the promastigote growth of other leishmanial species, including L. tropica, L. major and L. infantum. Finally, 6 and 16 interacted with high affinity with trypanothione reductase (TR), an essential enzyme for the leishmanial parasite and compound 6 inhibited TR with sub-micromolar potency. Thus, the effective inhibitory activity against Leishmania, the lack of toxicity on mammalian cells and the ability to block a crucial parasite's enzyme, highlight the potential for compound 6 to be optimized as novel drug candidate against leishmaniasis.
Collapse
Affiliation(s)
- Margherita Ortalli
- Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Andrea Ilari
- CNR-Institute of Molecular Biology and Pathology, Rome, Italy
| | - Gianni Colotti
- CNR-Institute of Molecular Biology and Pathology, Rome, Italy
| | - Ilenia De Ionna
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University, Rome, Italy
| | - Theo Battista
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University, Rome, Italy
| | - Alessandra Bisi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Silvia Gobbi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Angela Rampa
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Rita M C Di Martino
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Giovanna A Gentilomi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Stefania Varani
- Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum University of Bologna, Bologna, Italy; Unit of Clinical Microbiology, Regional Reference Centre for Microbiological Emergencies (CRREM), St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Federica Belluti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, Bologna, Italy.
| |
Collapse
|
48
|
Vijayakumar S, Das P. Recent progress in drug targets and inhibitors towards combating leishmaniasis. Acta Trop 2018; 181:95-104. [PMID: 29452111 DOI: 10.1016/j.actatropica.2018.02.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 01/24/2018] [Accepted: 02/11/2018] [Indexed: 12/22/2022]
Abstract
Lesihmaniasis is one of the major neglected tropical disease caused by the parasite of the genus Leishmania. The disease has more than one clinical forms and the visceral form is considered fatal. With the lack of potential vaccine, chemotherapy is the major treatment source considered for the control of the disease in the infected people. Drugs including amphotericin B and miltefosine are widely used for the treatment, however, development of resistance by the parasite towards the administered drug and high-toxicity of the drug are of major concern. Hence, more attention has been shown on identifying new targets, effective inhibitors, and better drug delivery system against the disease. This review deals with recent studies on drug targets and exploring their essentiality for the survival of Leishmania. Further, new inhibitors for those targets, novel anti-leishmanial peptides and vaccines against leishmaniasis were discussed. We believe that this pool of information will ease the researchers to gain knowledge and help in choosing right targets and design of new inhibitors against Leishmaniasis.
Collapse
|
49
|
De Gasparo R, Brodbeck-Persch E, Bryson S, Hentzen NB, Kaiser M, Pai EF, Krauth-Siegel RL, Diederich F. Biological Evaluation and X-ray Co-crystal Structures of Cyclohexylpyrrolidine Ligands for Trypanothione Reductase, an Enzyme from the Redox Metabolism of Trypanosoma. ChemMedChem 2018; 13:957-967. [PMID: 29624890 DOI: 10.1002/cmdc.201800067] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Indexed: 01/02/2023]
Abstract
The tropical diseases human African trypanosomiasis, Chagas disease, and the various forms of leishmaniasis are caused by parasites of the family of trypanosomatids. These protozoa possess a unique redox metabolism based on trypanothione and trypanothione reductase (TR), making TR a promising drug target. We report the optimization of properties and potency of cyclohexylpyrrolidine inhibitors of TR by structure-based design. The best inhibitors were freely soluble and showed competitive inhibition constants (Ki ) against Trypanosoma (T.) brucei TR and T. cruzi TR and in vitro activities (half-maximal inhibitory concentration, IC50 ) against these parasites in the low micromolar range, with high selectivity against human glutathione reductase. X-ray co-crystal structures confirmed the binding of the ligands to the hydrophobic wall of the "mepacrine binding site" with the new, solubility-providing vectors oriented toward the surface of the large active site.
Collapse
Affiliation(s)
- Raoul De Gasparo
- Laboratorium für Organische Chemie, ETH Zürich, Vladimir-Prelog-Weg 3, 8093, Zürich, Switzerland
| | - Elke Brodbeck-Persch
- Laboratorium für Organische Chemie, ETH Zürich, Vladimir-Prelog-Weg 3, 8093, Zürich, Switzerland
| | - Steve Bryson
- Departments of Biochemistry, Medical Biophysics, and Molecular Genetics, University of Toronto, Medical Sciences Building, #5358, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.,The Campbell Family Institute for Cancer Research, University Health Network, 101 College Street, Toronto, ON, M5G 1L7, Canada
| | - Nina B Hentzen
- Laboratorium für Organische Chemie, ETH Zürich, Vladimir-Prelog-Weg 3, 8093, Zürich, Switzerland
| | - Marcel Kaiser
- Swiss Tropical and Public Health Institute, Socinstrasse 57, 4002, Basel, Switzerland.,University of Basel, Petersplatz 1, 4003, Basel, Switzerland
| | - Emil F Pai
- Departments of Biochemistry, Medical Biophysics, and Molecular Genetics, University of Toronto, Medical Sciences Building, #5358, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.,The Campbell Family Institute for Cancer Research, University Health Network, 101 College Street, Toronto, ON, M5G 1L7, Canada
| | - R Luise Krauth-Siegel
- Biochemie-Zentrum Heidelberg (BZH), Universität Heidelberg, Im Neuenheimer Feld 328, 69120, Heidelberg, Germany
| | - François Diederich
- Laboratorium für Organische Chemie, ETH Zürich, Vladimir-Prelog-Weg 3, 8093, Zürich, Switzerland
| |
Collapse
|
50
|
Borsari C, Quotadamo A, Ferrari S, Venturelli A, Cordeiro-da-Silva A, Santarem N, Costi MP. Scaffolds and Biological Targets Avenue to Fight Against Drug Resistance in Leishmaniasis. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2018. [DOI: 10.1016/bs.armc.2018.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|