1
|
Gomes R, D. Polêto M, Verli H, Almeida VM, Marana SR, Bender A, Godoi BF, Rodrigues VL, da S. Emery F, Trossini GHG. Fragment Screening Reveals Novel Scaffolds against Sirtuin-2-Related Protein 1 from Trypanosoma brucei. ACS OMEGA 2025; 10:3808-3819. [PMID: 39926558 PMCID: PMC11799985 DOI: 10.1021/acsomega.4c09231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/03/2024] [Accepted: 12/09/2024] [Indexed: 02/11/2025]
Abstract
Sirtuin-2 (Sir2) is a histone deacetylase recognized as an antitrypanosomal target, yet there is limited knowledge regarding their potent inhibitors. This investigation employs the fragment-based drug discovery (FBDD) framework to identify novel inhibitors against Trypanosoma brucei Sir2-related protein 1. Initially, frequent residue-ligand interactions extracted from the crystallographic structures of human Sir2 and key features of human and parasitic Sir2 active sites were utilized to curate a targeted fragment library. Screening identified ten fragment hits, which introduced nine novel substructures compared to known Sir2 inhibitors. Among these, fragment 1 was the most potent, with an IC50 value of 17.8 μM and a ligand efficiency of 0.41. Further chemical space exploration of 30 compounds from the two most promising hits confirmed fragment 1 as the most potent. This study underscores the effectiveness of FBDD in discovering chemically distinct starting points with favorable ligand efficiency against protein targets in infectious diseases.
Collapse
Affiliation(s)
- Renan
A. Gomes
- Departamento
de Farmácia, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, Avenue. Lineu Prestes, 580, Cidade Universitária,
São Paulo, São Paulo 05508-000, Brazil
| | - Marcelo D. Polêto
- Centro
de Biotecnologia, Universidade Federal do
Rio Grande do Sul, Avenue
Bento Gonçalves, 9500, Porto Alegre, Rio Grande do Sul 91500-970, Brazil
| | - Hugo Verli
- Centro
de Biotecnologia, Universidade Federal do
Rio Grande do Sul, Avenue
Bento Gonçalves, 9500, Porto Alegre, Rio Grande do Sul 91500-970, Brazil
| | - Vitor M. Almeida
- Departamento
de Bioquímica, Instituto de Química, Universidade de São Paulo, Avenue Prof. Lineu Prestes, 748, São Paulo, São Paulo 05508-000, Brazil
| | - Sandro R. Marana
- Departamento
de Bioquímica, Instituto de Química, Universidade de São Paulo, Avenue Prof. Lineu Prestes, 748, São Paulo, São Paulo 05508-000, Brazil
| | - Andreas Bender
- Centre
for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K.
| | - Bruna F. Godoi
- Centre
for Research and Advancement in Fragments and Molecular Targets (CRAFT),
Departamento de Ciências Farmacêuticas, Faculdade de
Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Av. Prof. Zeferino Vaz, Campus USP, Ribeirão Preto, São Paulo 14040-903, Brazil
| | - Vinícius
T. L. Rodrigues
- Centre
for Research and Advancement in Fragments and Molecular Targets (CRAFT),
Departamento de Ciências Farmacêuticas, Faculdade de
Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Av. Prof. Zeferino Vaz, Campus USP, Ribeirão Preto, São Paulo 14040-903, Brazil
| | - Flavio da S. Emery
- Centre
for Research and Advancement in Fragments and Molecular Targets (CRAFT),
Departamento de Ciências Farmacêuticas, Faculdade de
Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Av. Prof. Zeferino Vaz, Campus USP, Ribeirão Preto, São Paulo 14040-903, Brazil
| | - Gustavo H. G. Trossini
- Departamento
de Farmácia, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, Avenue. Lineu Prestes, 580, Cidade Universitária,
São Paulo, São Paulo 05508-000, Brazil
| |
Collapse
|
2
|
Gaona-López C, Vazquez-Jimenez LK, Gonzalez-Gonzalez A, Delgado-Maldonado T, Ortiz-Pérez E, Nogueda-Torres B, Moreno-Rodríguez A, Vázquez K, Saavedra E, Rivera G. Advances in Protozoan Epigenetic Targets and Their Inhibitors for the Development of New Potential Drugs. Pharmaceuticals (Basel) 2023; 16:ph16040543. [PMID: 37111300 PMCID: PMC10143871 DOI: 10.3390/ph16040543] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/29/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023] Open
Abstract
Protozoan parasite diseases cause significant mortality and morbidity worldwide. Factors such as climate change, extreme poverty, migration, and a lack of life opportunities lead to the propagation of diseases classified as tropical or non-endemic. Although there are several drugs to combat parasitic diseases, strains resistant to routinely used drugs have been reported. In addition, many first-line drugs have adverse effects ranging from mild to severe, including potential carcinogenic effects. Therefore, new lead compounds are needed to combat these parasites. Although little has been studied regarding the epigenetic mechanisms in lower eukaryotes, it is believed that epigenetics plays an essential role in vital aspects of the organism, from controlling the life cycle to the expression of genes involved in pathogenicity. Therefore, using epigenetic targets to combat these parasites is foreseen as an area with great potential for development. This review summarizes the main known epigenetic mechanisms and their potential as therapeutics for a group of medically important protozoal parasites. Different epigenetic mechanisms are discussed, highlighting those that can be used for drug repositioning, such as histone post-translational modifications (HPTMs). Exclusive parasite targets are also emphasized, including the base J and DNA 6 mA. These two categories have the greatest potential for developing drugs to treat or eradicate these diseases.
Collapse
Affiliation(s)
- Carlos Gaona-López
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico
| | - Lenci K Vazquez-Jimenez
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico
| | - Alonzo Gonzalez-Gonzalez
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico
| | - Timoteo Delgado-Maldonado
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico
| | - Eyrá Ortiz-Pérez
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico
| | - Benjamín Nogueda-Torres
- Departamento de Parasitología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Adriana Moreno-Rodríguez
- Laboratorio de Estudios Epidemiológicos, Clínicos, Diseños Experimentales e Investigación, Facultad de Ciencias Químicas, Universidad Autónoma "Benito Juárez" de Oaxaca, Avenida Universidad S/N, Ex Hacienda Cinco Señores, Oaxaca 68120, Mexico
| | - Karina Vázquez
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Nuevo León, Francisco Villa 20, General Escobedo 66054, Mexico
| | - Emma Saavedra
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Gildardo Rivera
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico
| |
Collapse
|
3
|
Chu KB, Lee HA, Pflieger M, Fischer F, Asfaha Y, Alves Avelar LA, Skerhut A, Kassack MU, Hansen FK, Schöler A, Kurz T, Kim MJ, Moon EK, Quan FS. Antiproliferation and Antiencystation Effect of Class II Histone Deacetylase Inhibitors on Acanthamoeba castellanii. ACS Infect Dis 2022; 8:271-279. [PMID: 34994538 DOI: 10.1021/acsinfecdis.1c00390] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Acanthamoeba is a ubiquitous and free-living protozoan pathogen responsible for causing Acanthamoeba keratitis (AK), a severe corneal infection inflicting immense pain that can result in permanent blindness. A drug-based treatment of AK has remained arduous because Acanthamoeba trophozoites undergo encystment to become highly drug-resistant cysts upon exposure to harsh environmental conditions such as amoebicidal agents (e.g., polyhexanide, chloroquine, and chlorohexidine). As such, drugs that block the Acanthamoeba encystation process could result in a successful AK treatment. Histone deacetylase inhibitors (HDACi) have recently emerged as novel therapeutic options for treating various protozoan and parasitic diseases. Here, we investigated whether novel HDACi suppress the proliferation and encystation of Acanthamoeba. Synthetic class II HDACi FFK29 (IIa selective) and MPK576 (IIb selective) dose-dependently decreased the viability of Acanthamoeba trophozoites. While these HDACi demonstrated a negligible effect on the viability of mature cysts, Acanthamoeba encystation was significantly inhibited by these HDACi. Apoptosis was slightly increased in trophozoites after a treatment with these HDACi, whereas cysts were unaffected by the HDACi exposure. The viability of human corneal cells was not affected by HDACi concentrations up to 10 μmol/L. In conclusion, these synthetic HDACi demonstrated potent amoebicidal effects and inhibited the growth and encystation of Acanthamoeba, thus highlighting their enormous potential for further development.
Collapse
Affiliation(s)
- Ki-Back Chu
- Department of Biomedical Science, Graduate School, Kyung Hee University, 02447 Seoul, South Korea
| | - Hae-Ahm Lee
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Kyung Hee University, 02447 Seoul, South Korea
| | - Marc Pflieger
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, 40225 Dusseldorf, Germany
| | - Fabian Fischer
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, 40225 Dusseldorf, Germany
| | - Yodita Asfaha
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, 40225 Dusseldorf, Germany
| | - Leandro A. Alves Avelar
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, 40225 Dusseldorf, Germany
| | - Alexander Skerhut
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, 40225 Dusseldorf, Germany
| | - Matthias U. Kassack
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, 40225 Dusseldorf, Germany
| | - Finn K Hansen
- Pharmaceutical and Cell Biological Chemistry, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Andrea Schöler
- Institute for Drug Discovery, Medical Faculty, Leipzig University, Brüderstraße 34, 04103 Leipzig, Germany
| | - Thomas Kurz
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, 40225 Dusseldorf, Germany
| | - Min-Jeong Kim
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Kyung Hee University, 02447 Seoul, South Korea
| | - Eun-Kyung Moon
- Department of Medical Zoology, Kyung Hee University, School of Medicine, 02447 Seoul, South Korea
| | - Fu-Shi Quan
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Kyung Hee University, 02447 Seoul, South Korea
- Department of Medical Zoology, Kyung Hee University, School of Medicine, 02447 Seoul, South Korea
| |
Collapse
|
4
|
Ghazy E, Abdelsalam M, Robaa D, Pierce RJ, Sippl W. Histone Deacetylase (HDAC) Inhibitors for the Treatment of Schistosomiasis. Pharmaceuticals (Basel) 2022; 15:ph15010080. [PMID: 35056137 PMCID: PMC8779837 DOI: 10.3390/ph15010080] [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: 12/20/2021] [Revised: 01/06/2022] [Accepted: 01/08/2022] [Indexed: 12/18/2022] Open
Abstract
Schistosomiasis is a major neglected parasitic disease that affects more than 240 million people worldwide and for which the control strategy consists of mass treatment with the only available drug, praziquantel. Schistosomes display morphologically distinct stages during their life cycle and the transformations between stages are controlled by epigenetic mechanisms. The targeting of epigenetic actors might therefore represent the parasites’ Achilles’ heel. Specifically, histone deacetylases have been recently characterized as drug targets for the treatment of schistosomiasis. This review focuses on the recent development of inhibitors for schistosome histone deacetylases. In particular, advances in the development of inhibitors of Schistosoma mansoni histone deacetylase 8 have indicated that targeting this enzyme is a promising approach for the treatment of this infection.
Collapse
Affiliation(s)
- Ehab Ghazy
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, 06120 Halle (Saale), Germany; (E.G.); (M.A.); (D.R.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Mohamed Abdelsalam
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, 06120 Halle (Saale), Germany; (E.G.); (M.A.); (D.R.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Dina Robaa
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, 06120 Halle (Saale), Germany; (E.G.); (M.A.); (D.R.)
| | - Raymond J. Pierce
- Centre d’Infection et d’Immunité de Lille, U1019—UMR9017—CIIL, Institute Pasteur de Lille, CNRS, Inserm, CHU Lille, Univ. Lille, F-59000 Lille, France;
| | - Wolfgang Sippl
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, 06120 Halle (Saale), Germany; (E.G.); (M.A.); (D.R.)
- Correspondence:
| |
Collapse
|
5
|
Marek M, Ramos-Morales E, Picchi-Constante GFA, Bayer T, Norström C, Herp D, Sales-Junior PA, Guerra-Slompo EP, Hausmann K, Chakrabarti A, Shaik TB, Merz A, Troesch E, Schmidtkunz K, Goldenberg S, Pierce RJ, Mourão MM, Jung M, Schultz J, Sippl W, Zanchin NIT, Romier C. Species-selective targeting of pathogens revealed by the atypical structure and active site of Trypanosoma cruzi histone deacetylase DAC2. Cell Rep 2021; 37:110129. [PMID: 34936867 DOI: 10.1016/j.celrep.2021.110129] [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: 07/06/2021] [Revised: 10/26/2021] [Accepted: 11/23/2021] [Indexed: 01/12/2023] Open
Abstract
Writing and erasing of posttranslational modifications are crucial to phenotypic plasticity and antigenic variation of eukaryotic pathogens. Targeting pathogens' modification machineries, thus, represents a valid approach to fighting parasitic diseases. However, identification of parasitic targets and the development of selective anti-parasitic drugs still represent major bottlenecks. Here, we show that the zinc-dependent histone deacetylases (HDACs) of the protozoan parasite Trypanosoma cruzi are key regulators that have significantly diverged from their human counterparts. Depletion of T. cruzi class I HDACs tcDAC1 and tcDAC2 compromises cell-cycle progression and division, leading to cell death. Notably, tcDAC2 displays a deacetylase activity essential to the parasite and shows major structural differences with human HDACs. Specifically, tcDAC2 harbors a modular active site with a unique subpocket targeted by inhibitors showing substantial anti-parasitic effects in cellulo and in vivo. Thus, the targeting of the many atypical HDACs in pathogens can enable anti-parasitic selective chemical impairment.
Collapse
Affiliation(s)
- Martin Marek
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, UMR 7104, U 1258, 67404 Illkirch, France; IGBMC, Department of Integrated Structural Biology, 1 rue Laurent Fries, B.P. 10142, 67404 Illkirch Cedex, France
| | - Elizabeth Ramos-Morales
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, UMR 7104, U 1258, 67404 Illkirch, France; IGBMC, Department of Integrated Structural Biology, 1 rue Laurent Fries, B.P. 10142, 67404 Illkirch Cedex, France
| | | | - Theresa Bayer
- Institute of Pharmacy, Martin-Luther-Universität Halle-Wittenberg, Wolfgang-Langenbeck-Straße 4, 06120 Halle/Saale, Germany
| | | | - Daniel Herp
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-Universität Freiburg, Albertstraße 25, 79104 Freiburg, Germany
| | - Policarpo A Sales-Junior
- Instituto René Rachou, Fundação Oswaldo Cruz, Avenida Augusto de Lima, 1715, 30190-002 Belo Horizonte, Brazil
| | | | - Kristin Hausmann
- Institute of Pharmacy, Martin-Luther-Universität Halle-Wittenberg, Wolfgang-Langenbeck-Straße 4, 06120 Halle/Saale, Germany
| | - Alokta Chakrabarti
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-Universität Freiburg, Albertstraße 25, 79104 Freiburg, Germany
| | - Tajith B Shaik
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, UMR 7104, U 1258, 67404 Illkirch, France; IGBMC, Department of Integrated Structural Biology, 1 rue Laurent Fries, B.P. 10142, 67404 Illkirch Cedex, France
| | - Annika Merz
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-Universität Freiburg, Albertstraße 25, 79104 Freiburg, Germany
| | - Edouard Troesch
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, UMR 7104, U 1258, 67404 Illkirch, France; IGBMC, Department of Integrated Structural Biology, 1 rue Laurent Fries, B.P. 10142, 67404 Illkirch Cedex, France
| | - Karin Schmidtkunz
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-Universität Freiburg, Albertstraße 25, 79104 Freiburg, Germany
| | - Samuel Goldenberg
- Instituto Carlos Chagas, Fiocruz Paraná, Curitiba, Paraná 81350-010, Brazil
| | - Raymond J Pierce
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL -Centre d'Infection et d'Immunité de Lille, 59000 Lille, France
| | - Marina M Mourão
- Instituto René Rachou, Fundação Oswaldo Cruz, Avenida Augusto de Lima, 1715, 30190-002 Belo Horizonte, Brazil
| | - Manfred Jung
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-Universität Freiburg, Albertstraße 25, 79104 Freiburg, Germany
| | - Johan Schultz
- Kancera AB, Nanna Svartz Väg 4, SE-17165 Solna, Sweden
| | - Wolfgang Sippl
- Institute of Pharmacy, Martin-Luther-Universität Halle-Wittenberg, Wolfgang-Langenbeck-Straße 4, 06120 Halle/Saale, Germany
| | - Nilson I T Zanchin
- Instituto Carlos Chagas, Fiocruz Paraná, Curitiba, Paraná 81350-010, Brazil.
| | - Christophe Romier
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, UMR 7104, U 1258, 67404 Illkirch, France; IGBMC, Department of Integrated Structural Biology, 1 rue Laurent Fries, B.P. 10142, 67404 Illkirch Cedex, France.
| |
Collapse
|
6
|
Abstract
Infections caused by protozoans remain a public health issue, especially in tropical countries. Serious adverse events, lack of efficacy at the different stages of the infection and routes of administration that have a negative impact on treatment adherence are some of the problems with currently available therapy against these diseases. Here we describe an epigenetic target, sirtuin 2 and its related proteins, that is promising given the results in phenotypic assays and in vivo models against Sir2 of Plasmodium falciparum, Leishmania donovani, Leishmania infantum, Schistosoma mansoni, Trypanosoma brucei and Trypanosoma cruzi parasites. The results we present highlight how this target can be extensively explored and how its inhibitors might be employed in the clinic.
Collapse
|
7
|
Ortore G, Poli G, Martinelli A, Tuccinardi T, Rizzolio F, Caligiuri I. From Anti-infective Agents to Cancer Therapy: a Drug Repositioning Study Revealed a New Use for Nitrofuran Derivatives. Med Chem 2021; 18:249-259. [PMID: 33992059 DOI: 10.2174/1573406417666210511001241] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 02/06/2021] [Accepted: 02/07/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The progression of ovarian cancer seems to be related to HDAC1, HDAC3 and HDAC6 activity. A possible strategy for improving therapies for treating ovarian carcinoma, minimizing the preclinical screenings, is the repurposing of already approved pharmaceutical products as inhibitors of these enzymes. OBJECTIVE This work was aimed to implement a computational strategy for identifying new HDAC inhibitors for ovarian carcinoma treatment among approved drugs. METHOD The CHEMBL database was used to construct training, test and decoys sets for performing and validating HDAC1, HDAC3 and HDAC6 3D-QSAR models obtained by using FLAP program. Docking and MD simulations were used in combination with the generated models to identify novel potential HDAC inhibitors. Cell viability assays and Western blot analyses were performed on normal and cancer cells for a direct evaluation of the anti-proliferative activity and an in vitro estimation of HDAC inhibition of the compounds selected through in silico screening. RESULT The best quantitative prediction was obtained for the HDAC6 3D-QSAR model. The screening of approved drugs highlighted a new potential use as HDAC inhibitors for some compounds, in particular nitrofuran derivatives, usually known for their antibacterial activity, and frequently used as antimicrobial adjuvant therapy in cancer treatment. Experimental evaluation of these derivatives highlighted a significant antiproliferative activity against cancer cell lines overexpressing HDAC6, and an increase in acetylated alpha-tubulin levels. CONCLUSION Experimental results support the hypothesis of a potential direct interaction of nitrofuran derivatives with HDACs. In addition to the possible repurposing of already approved drugs, this work suggests the nitro group as a new zinc binding group, able to interact with the catalytic zinc ion of HDACs.
Collapse
Affiliation(s)
| | - Giulio Poli
- Department of Pharmacy, Pisa University, Pisa, Italy
| | | | | | - Flavio Rizzolio
- Pathology Unit, Centro di Riferimento Oncologico (CRO) IRCCS, Aviano, Italy
| | - Isabella Caligiuri
- Pathology Unit, Centro di Riferimento Oncologico (CRO) IRCCS, Aviano, Italy
| |
Collapse
|
8
|
Vaca HR, Celentano AM, Toscanini MA, Heimburg T, Ghazy E, Zeyen P, Hauser AT, Oliveira G, Elissondo MC, Jung M, Sippl W, Camicia F, Rosenzvit MC. The potential for histone deacetylase (HDAC) inhibitors as cestocidal drugs. PLoS Negl Trop Dis 2021; 15:e0009226. [PMID: 33657105 PMCID: PMC7959350 DOI: 10.1371/journal.pntd.0009226] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 03/15/2021] [Accepted: 02/10/2021] [Indexed: 11/18/2022] Open
Abstract
Background Echinococcosis and cysticercosis are neglected tropical diseases caused by cestode parasites (family Taeniidae). Not only there is a small number of approved anthelmintics for the treatment of these cestodiases, but also some of them are not highly effective against larval stages, such that identifying novel drug targets and their associated compounds is critical. Histone deacetylase (HDAC) enzymes are validated drug targets in cancers and other diseases, and have been gaining relevance for developing new potential anti-parasitic treatments in the last years. Here, we present the anthelmintic profile for a panel of recently developed HDAC inhibitors against the model cestode Mesocestoides vogae (syn. M. corti). Methodology/Principal findings Phenotypic screening was performed on M. vogae by motility measurements and optical microscopic observations. Some HDAC inhibitors showed potent anthelmintic activities; three of them -entinostat, TH65, and TH92- had pronounced anthelmintic effects, reducing parasite viability by ~100% at concentrations of ≤ 20 μM. These compounds were selected for further characterization and showed anthelmintic effects in the micromolar range and in a time- and dose-dependent manner. Moreover, these compounds induced major alterations on the morphology and ultrastructural features of M. vogae. The potencies of these compounds were higher than albendazole and the anthelmintic effects were irreversible. Additionally, we evaluated pairwise drug combinations of these HDAC inhibitors and albendazole. The results suggested a positive interaction in the anthelmintic effect for individual pairs of compounds. Due to the maximum dose approved for entinostat, adjustments in the dose regime and/or combinations with currently-used anthelmintic drugs are needed, and the selectivity of TH65 and TH92 towards parasite targets should be assessed. Conclusion, significance The results presented here suggest that HDAC inhibitors represent novel and potent drug candidates against cestodes and pave the way to understanding the roles of HDACs in these parasites. Neglected tropical diseases, such as echinococcosis and cysticercosis, which are caused by taeniid cestodes (tapeworms), represent serious public health problems in many countries around the world. Given that there is only a small number of approved anthelmintics for the treatment of cestodiases, and that most of them are not highly effective against larval stages, identifying novel drug targets and their associated compounds is critical. Histone deacetylases (HDACs) are enzymes that produce epigenetic modifications of chromatin, thus modifying cellular gene expression. In this study, we evaluate and characterize a number of HDAC inhibitors on the model cestode Mesocestoides vogae and report the anthelmintic profile of these compounds. Some of the HDAC inhibitors tested showed potent anthelmintic effects, particularly entinostat, TH65 and TH92. These compounds were selected as the most promising candidates due to their high potencies, which were superior to the commercially-available anthelmintic drug albendazole. We also evaluated pairwise drug combinations of HDAC inhibitors and albendazole. The findings of this study provide a starting point for the development of new HDAC-based cestocidal compounds.
Collapse
Affiliation(s)
- Hugo Rolando Vaca
- Instituto de Microbiología y Parasitología Médica, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Tecnológicas (IMPaM, UBA-CONICET). Facultad de Medicina, Paraguay 2155, piso 13, C1121ABG, Buenos Aires, Argentina
- Laboratorio de Zoonosis Parasitarias, Instituto de Investigaciones en Producción, Sanidad y Ambiente (IIPROSAM), Facultad de Ciencias Exactas y Naturales (FCEyN), Universidad Nacional de Mar del Plata (UNMdP), Mar del Plata, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Ana María Celentano
- Instituto de Microbiología y Parasitología Médica, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Tecnológicas (IMPaM, UBA-CONICET). Facultad de Medicina, Paraguay 2155, piso 13, C1121ABG, Buenos Aires, Argentina
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires, Argentina
| | - María Agustina Toscanini
- Instituto de Microbiología y Parasitología Médica, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Tecnológicas (IMPaM, UBA-CONICET). Facultad de Medicina, Paraguay 2155, piso 13, C1121ABG, Buenos Aires, Argentina
| | - Tino Heimburg
- Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Halle(Saale), Germany
| | - Ehab Ghazy
- Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Halle(Saale), Germany
| | - Patrik Zeyen
- Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Halle(Saale), Germany
| | | | | | - María Celina Elissondo
- Laboratorio de Zoonosis Parasitarias, Instituto de Investigaciones en Producción, Sanidad y Ambiente (IIPROSAM), Facultad de Ciencias Exactas y Naturales (FCEyN), Universidad Nacional de Mar del Plata (UNMdP), Mar del Plata, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Manfred Jung
- Institute of Pharmaceutical Sciences, University of Freiburg, Freiburg, Germany
| | - Wolfgang Sippl
- Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Halle(Saale), Germany
| | - Federico Camicia
- Instituto de Microbiología y Parasitología Médica, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Tecnológicas (IMPaM, UBA-CONICET). Facultad de Medicina, Paraguay 2155, piso 13, C1121ABG, Buenos Aires, Argentina
- * E-mail: (FC); (MCR)
| | - Mara Cecilia Rosenzvit
- Instituto de Microbiología y Parasitología Médica, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Tecnológicas (IMPaM, UBA-CONICET). Facultad de Medicina, Paraguay 2155, piso 13, C1121ABG, Buenos Aires, Argentina
- * E-mail: (FC); (MCR)
| |
Collapse
|
9
|
Murakoshi F, Bando H, Sugi T, Adeyemi OS, Nonaka M, Nakaya T, Kato K. Nullscript inhibits Cryptosporidium and Toxoplasma growth. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2020; 14:159-166. [PMID: 33120250 PMCID: PMC7593347 DOI: 10.1016/j.ijpddr.2020.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 10/05/2020] [Accepted: 10/08/2020] [Indexed: 11/26/2022]
Abstract
Cryptosporidium and Toxoplasma are parasites that have caused problems worldwide. Cryptosporidium causes severe watery diarrhoea and may be fatal in immunocompromised patients and in infants. Nitazoxanide is the only agent currently approved by the FDA, but its efficacy is limited. Toxoplasmosis is also a problem in the immunocompromised, as currently available treatment options have limited efficacy and patient tolerance can be poor. In the present investigation, we screened libraries of epigenetic compounds to identify those that inhibited C. parvum growth. Nullscript was identified as a compound with an inhibitory effect on C. parvum and T. gondii growth, and was less toxic to host cells. Nullscript was also able to significantly decrease oocyst excretion in C. parvum-infected SCID mice. A library of epigenetic compounds was used to screen for compounds effective against Cryptosporiium parvum. Nullscript has been shown to be effective in inhibiting the growth of C. parvum and T. gondii. Nullscript significantly decreased oocyst excretion in C. parvum-infected SCID mice.
Collapse
Affiliation(s)
- Fumi Murakoshi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan; Department of Infectious Diseases, Kyoto Prefectural University of Medicine, 465, Kawaramachi-hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan; Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, Yomogida 232-3, Naruko-onsen, Osaki, Miyagi, 989-6711, Japan
| | - Hironori Bando
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, Yomogida 232-3, Naruko-onsen, Osaki, Miyagi, 989-6711, Japan
| | - Tatsuki Sugi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan
| | - Oluyomi Stephen Adeyemi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan; Medicinal Biochemistry, Nanomedicine & Toxicology Laboratory, Department of Biochemistry, Landmark University, PMB 1001, Omu-Aran, 251101, Nigeria
| | - Motohiro Nonaka
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan
| | - Takaaki Nakaya
- Department of Infectious Diseases, Kyoto Prefectural University of Medicine, 465, Kawaramachi-hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Kentaro Kato
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan; Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, Yomogida 232-3, Naruko-onsen, Osaki, Miyagi, 989-6711, Japan.
| |
Collapse
|
10
|
Elmezayen AD, Yelekçi K. Homology modeling and in silico design of novel and potential dual-acting inhibitors of human histone deacetylases HDAC5 and HDAC9 isozymes. J Biomol Struct Dyn 2020; 39:6396-6414. [PMID: 32715940 DOI: 10.1080/07391102.2020.1798812] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Histone deacetylases (HDACs) are a group of enzymes that have prominent and crucial effect on various biological systems, mainly by their suppressive effect on transcription. Searching for inhibitors targeting their respective isoforms without affecting other targets is greatly needed. Some histone deacetylases have no crystal structures, such as HDAC5 and HDAC9. Lacking proper and suitable crystal structure is obstructing the designing of appropriate isoform selective inhibitors. Here in this study, we constructed human HDAC5 and HDAC9 protein models using human HDAC4 (PDB:2VQM_A) as a template by the means of homology modeling approach. Based on the Z-score of the built models, model M0014 of HDAC5 and model M0020 of HDAC9 were selected. The models were verified by MODELLER and validated using the Web-based PROCHECK server. All selected known inhibitors displayed reasonable binding modes and equivalent predicted Ki values in comparison to the experimental binding affinities (Ki/IC50). The known inhibitor Rac26 showed the best binding affinity for HDAC5, while TMP269 showed the best binding affinity for HDAC9. The best two compounds, CHEMBL2114980 and CHEMBL217223, had relatively similar inhibition constants against HDAC5 and HDAC9. The built models and their complexes were subjected to molecular dynamic simulations (MD) for 100 ns. Examining the MD simulation results of all studied structures, including the RMSD, RMSF, radius of gyration and potential energy suggested the stability and reliability of the built models. Accordingly, the results obtained in this study could be used for designing de novo inhibitors against HDAC5 and HDAC9. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ammar D Elmezayen
- Department of Bioinformatics and Genetics, Faculty of Engineering and Natural Sciences, Kadir Has University, Istanbul, Turkey
| | - Kemal Yelekçi
- Department of Bioinformatics and Genetics, Faculty of Engineering and Natural Sciences, Kadir Has University, Istanbul, Turkey
| |
Collapse
|
11
|
Wahab S, Saettone A, Nabeel-Shah S, Dannah N, Fillingham J. Exploring the Histone Acetylation Cycle in the Protozoan Model Tetrahymena thermophila. Front Cell Dev Biol 2020; 8:509. [PMID: 32695779 PMCID: PMC7339932 DOI: 10.3389/fcell.2020.00509] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 05/28/2020] [Indexed: 12/22/2022] Open
Abstract
The eukaryotic histone acetylation cycle is composed of three classes of proteins, histone acetyltransferases (HATs) that add acetyl groups to lysine amino acids, bromodomain (BRD) containing proteins that are one of the most characterized of several protein domains that recognize acetyl-lysine (Kac) and effect downstream function, and histone deacetylases (HDACs) that catalyze the reverse reaction. Dysfunction of selected proteins of these three classes is associated with human disease such as cancer. Additionally, the HATs, BRDs, and HDACs of fungi and parasitic protozoa present potential drug targets. Despite their importance, the function and mechanisms of HATs, BRDs, and HDACs and how they relate to chromatin remodeling (CR) remain incompletely understood. Tetrahymena thermophila (Tt) provides a highly tractable single-celled free-living protozoan model for studying histone acetylation, featuring a massively acetylated somatic genome, a property that was exploited in the identification of the first nuclear/type A HAT Gcn5 in the 1990s. Since then, Tetrahymena remains an under-explored model for the molecular analysis of HATs, BRDs, and HDACs. Studies of HATs, BRDs, and HDACs in Tetrahymena have the potential to reveal the function of HATs and BRDs relevant to both fundamental eukaryotic biology and to the study of disease mechanisms in parasitic protozoa.
Collapse
Affiliation(s)
| | | | | | | | - Jeffrey Fillingham
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
| |
Collapse
|
12
|
Divsalar DN, Simoben CV, Schonhofer C, Richard K, Sippl W, Ntie-Kang F, Tietjen I. Novel Histone Deacetylase Inhibitors and HIV-1 Latency-Reversing Agents Identified by Large-Scale Virtual Screening. Front Pharmacol 2020; 11:905. [PMID: 32625097 PMCID: PMC7311767 DOI: 10.3389/fphar.2020.00905] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 06/03/2020] [Indexed: 12/16/2022] Open
Abstract
Current antiretroviral therapies used for HIV management do not target latent viral reservoirs in humans. The experimental “shock-and-kill” therapeutic approach involves use of latency-reversal agents (LRAs) that reactivate HIV expression in reservoir-containing cells, followed by infected cell elimination through viral or host immune cytopathic effects. Several LRAs that function as histone deacetylase (HDAC) inhibitors are reported to reverse HIV latency in cells and in clinical trials; however, none to date have consistently reduced viral reservoirs in humans, prompting a need to identify new LRAs. Toward this goal, we describe here a virtual screening (VS) approach which uses 14 reported HDAC inhibitors to probe PubChem and identifies 60 LRA candidates. We then show that four screening “hits” including (S)-N-Hydroxy-4-(3-methyl-2-phenylbutanamido)benzamide (compound 15), N-(4-Aminophenyl)heptanamide (16), N-[4-(Heptanoylamino)phenyl]heptanamide (17), and 4-(1,3-Dioxo-1H-benzo[de]isoquinolin-2(3H)-yl)-N-(2-hydroxyethyl)butanamide (18) inhibit HDAC activity and/or reverse HIV latency in vitro. This study demonstrates and supports that VS-based approaches can readily identify novel HDAC inhibitors and LRAs, which in turn may help toward inhibitor design and chemical optimization efforts for improved HIV shock-and-kill-based efforts.
Collapse
Affiliation(s)
- Donya Naz Divsalar
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Conrad Veranso Simoben
- Department of Chemistry, University of Buea, Buea, Cameroon.,Instutite of Pharmacy, Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Cole Schonhofer
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Khumoekae Richard
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Wolfgang Sippl
- Instutite of Pharmacy, Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Fidele Ntie-Kang
- Department of Chemistry, University of Buea, Buea, Cameroon.,Instutite of Pharmacy, Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Ian Tietjen
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada.,The Wistar Institute, Philadelphia, PA, United States
| |
Collapse
|
13
|
Histone deacetylase inhibitors with high in vitro activities against Plasmodium falciparum isolates collected from Gabonese children and adults. Sci Rep 2019; 9:17336. [PMID: 31758015 PMCID: PMC6874535 DOI: 10.1038/s41598-019-53912-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 11/04/2019] [Indexed: 01/25/2023] Open
Abstract
Histone deacetylase (HDAC) enzymes are targets for the development of antimalarial drugs with a different mode of action to established antimalarials. Broad-spectrum HDAC-inhibitors show high potency against Plasmodium falciparum, but displayed some toxicity towards human cells. Inhibitors of human HDAC6 are new drug candidates with supposed reduced toxicity to human cells and favorable activities against laboratory P. falciparum strains. We investigated the potency of 12 peptoid-based HDAC-inhibitors against asexual stages of P. falciparum clinical isolates. Parasites representing different genetic backgrounds were isolated from adults and children with uncomplicated malaria in Gabon. Clinical studies on (non-HDAC-inhibitors) antimalarials, moreover, found lower drug efficacy in children, mainly attributed to acquired immunity with age in endemic areas. Therefore, we compared the in vitro sensitivity profiles of adult- and child-derived isolates to antimalarials (HDAC and standard drugs). All HDAC-inhibitors showed 50% inhibitory concentrations at nanomolar ranges with higher activities than the FDA approved reference HDAC-inhibitor SAHA. We propose peptoid-based HDAC6-inhibitors to be lead structures for further development as antimalarial chemotherapeutics. Our results further suggest no differences in activity of the tested antimalarials between P. falciparum parasites isolated from children and adults.
Collapse
|
14
|
A Tiny Change Makes a Big Difference in the Anti-Parasitic Activities of an HDAC Inhibitor. Int J Mol Sci 2019; 20:ijms20122973. [PMID: 31216674 PMCID: PMC6627744 DOI: 10.3390/ijms20122973] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 06/14/2019] [Accepted: 06/15/2019] [Indexed: 01/08/2023] Open
Abstract
We previously synthesized an hydroxamate derivative (N-hydroxy-4-[2-(3-methoxyphenyl)acetamido]benzamide) named 363 with potent anti-Toxoplasma gondii activity and histone deacetylase inhibitor (HDACi) effects. Here we show that 1-N-hydroxy-4-N-[(2-methoxyphenyl)methyl]benzene-1,4-dicarboxamide, a 363 isomer, does not have antiparasitic potency and has a 13-fold decrease in HDACi activity. The in silico modeling of T. gondii HDACs of the type II strain discloses identity varying from 25% to 62% on more than 250 residues for S8EP32_TOXG and A0A125YPH4_TOXGM. We observed a high conservation degree with the human HDAC2 (53% and 64% identity, respectively) and a moderate one with the human HDAC8 (30–40%). Two other TgHDACs, S8F6L4_TOXGM and S8GEI3_TOXGM, were identified as displaying a higher similarity with some bacterial orthologs (~35%) than with the human enzymes (~25%). The docking in parallel of the two compounds on the models generated allowed us to gain insights on the docking of these hydroxamate derivatives that guide their specificity and potency against T. gondii histone deacetylase. This information would constitute the rationale from which more specific derivatives can be synthetized.
Collapse
|
15
|
Nanduri R, Kalra R, Bhagyaraj E, Chacko AP, Ahuja N, Tiwari D, Kumar S, Jain M, Parkesh R, Gupta P. AutophagySMDB: a curated database of small molecules that modulate protein targets regulating autophagy. Autophagy 2019; 15:1280-1295. [PMID: 30669929 DOI: 10.1080/15548627.2019.1571717] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Macroautophagy/autophagy is a complex self-degradative mechanism responsible for clearance of non functional organelles and proteins. A range of factors influences the autophagic process, and disruptions in autophagy-related mechanisms lead to disease states, and further exacerbation of disease. Despite in-depth research into autophagy and its role in pathophysiological processes, the resources available to use it for therapeutic purposes are currently lacking. Herein we report the Autophagy Small Molecule Database (AutophagySMDB; http://www.autophagysmdb.org/ ) of small molecules and their cognate protein targets that modulate autophagy. Presently, AutophagySMDB enlists ~10,000 small molecules which regulate 71 target proteins. All entries are comprised of information such as EC50 (half maximal effective concentration), IC50 (half maximal inhibitory concentration), Kd (dissociation constant) and Ki (inhibition constant), IUPAC name, canonical SMILE, structure, molecular weight, QSAR (quantitative structure activity relationship) properties such as hydrogen donor and acceptor count, aromatic rings and XlogP. AutophagySMDB is an exhaustive, cross-platform, manually curated database, where either the cognate targets for small molecule or small molecules for a target can be searched. This database is provided with different search options including text search, advanced search and structure search. Various computational tools such as tree tool, cataloging tools, and clustering tools have also been implemented for advanced analysis. Data and the tools provided in this database helps to identify common or unique scaffolds for designing novel drugs or to improve the existing ones for autophagy small molecule therapeutics. The approach to multitarget drug discovery by identifying common scaffolds has been illustrated with experimental validation. Abbreviations: AMPK: AMP-activated protein kinase; ATG: autophagy related; AutophagySMDB: autophagy small molecule database; BCL2: BCL2, apoptosis regulator; BECN1: beclin 1; CAPN: calpain; MTOR: mechanistic target of rapamycin kinase; PPARG: peroxisome proliferator activated receptor gamma; SMILES: simplified molecular input line entry system; SQSTM1: sequestosome 1; STAT3: signal transducer and activator of transcription.
Collapse
Affiliation(s)
- Ravikanth Nanduri
- a Department of Molecular Biology , CSIR-Institute of Microbial Technology , Chandigarh , India
| | - Rashi Kalra
- a Department of Molecular Biology , CSIR-Institute of Microbial Technology , Chandigarh , India
| | - Ella Bhagyaraj
- a Department of Molecular Biology , CSIR-Institute of Microbial Technology , Chandigarh , India
| | - Anuja P Chacko
- a Department of Molecular Biology , CSIR-Institute of Microbial Technology , Chandigarh , India
| | - Nancy Ahuja
- a Department of Molecular Biology , CSIR-Institute of Microbial Technology , Chandigarh , India
| | - Drishti Tiwari
- a Department of Molecular Biology , CSIR-Institute of Microbial Technology , Chandigarh , India
| | - Sumit Kumar
- a Department of Molecular Biology , CSIR-Institute of Microbial Technology , Chandigarh , India
| | - Monika Jain
- a Department of Molecular Biology , CSIR-Institute of Microbial Technology , Chandigarh , India
| | - Raman Parkesh
- a Department of Molecular Biology , CSIR-Institute of Microbial Technology , Chandigarh , India
| | - Pawan Gupta
- a Department of Molecular Biology , CSIR-Institute of Microbial Technology , Chandigarh , India
| |
Collapse
|
16
|
Ruzic D, Petkovic M, Agbaba D, Ganesan A, Nikolic K. Combined Ligand and Fragment‐based Drug Design of Selective Histone Deacetylase – 6 Inhibitors. Mol Inform 2019; 38:e1800083. [DOI: 10.1002/minf.201800083] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 12/08/2018] [Indexed: 12/16/2022]
Affiliation(s)
- Dusan Ruzic
- Department of Pharmaceutical Chemistry, Faculty of PharmacyUniversity of Belgrade Vojvode Stepe 450 11000 Belgrade Serbia
| | - Milos Petkovic
- Department of Organic Chemistry, Faculty of PharmacyUniversity of Belgrade Vojvode Stepe 450 11000 Belgrade Serbia
| | - Danica Agbaba
- Department of Pharmaceutical Chemistry, Faculty of PharmacyUniversity of Belgrade Vojvode Stepe 450 11000 Belgrade Serbia
| | - A. Ganesan
- School of PharmacyUniversity of East Anglia Norwich Research Park NR4 7TJ Norwich United Kingdom
| | - Katarina Nikolic
- Department of Pharmaceutical Chemistry, Faculty of PharmacyUniversity of Belgrade Vojvode Stepe 450 11000 Belgrade Serbia
- Department of Pharmaceutical Chemistry, Faculty of PharmacyUniversity of Belgrade Vojvode Stepe 450 11000 Belgrade Serbia
| |
Collapse
|
17
|
A nanodelivered Vorinostat derivative is a promising oral compound for the treatment of visceral leishmaniasis. Pharmacol Res 2019; 139:375-383. [DOI: 10.1016/j.phrs.2018.11.039] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/11/2018] [Accepted: 11/28/2018] [Indexed: 12/21/2022]
|
18
|
Synthesis, spectral characterization, docking studies and biological activity of urea, thiourea, sulfonamide and carbamate derivatives of imatinib intermediate. Mol Divers 2018; 23:723-738. [DOI: 10.1007/s11030-018-9906-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 12/11/2018] [Indexed: 10/27/2022]
|
19
|
Yuan Y, Hu Z, Bao M, Sun R, Long X, Long L, Li J, Wu C, Bao J. Screening of novel histone deacetylase 7 inhibitors through molecular docking followed by a combination of molecular dynamics simulations and ligand-based approach. J Biomol Struct Dyn 2018; 37:4092-4103. [PMID: 30417746 DOI: 10.1080/07391102.2018.1541141] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Histone acetylation/deacetylation is a key mechanism for transcription regulation which plays an important role in control of gene expression, tissue growth, and development. In particular, histone deacetylase 7 (HDAC7), a member of class IIa HDACs, is crucial to maintain cell homeostasis, and HDAC7 has emerged as a new target for cancer therapy. In this study, molecular docking was applied to screen candidate inhibitors and 21 compounds were found. Following the 50 ns molecular dynamics simulations and binding free energy calculation, ZINC00156160, ZINC01703144, ZINC04293665, and ZINC13900201 were identified as potential HDAC7 inhibitors, which would provide a sound starting point for further studies involving molecular modeling coupled with biochemical experiments. Meanwhile, similarity computation and substructure search were combined, and then we found that compounds sharing common backbone "CC(=O)N[C@@H](CSc1ccccc1)C(=O)O" could be efficient to inhibit the bioactivity of HDAC7. Then comparative molecular similarity indices analysis (CoMSIA) techniques were implemented to investigate the relationship between properties of the substituent group and bioactivities of small molecules. The CoMSIA model exhibited powerful predictivity, with satisfactory statistical parameters such as q2 of 0.659, R2 of 0.952, and F of 268.448. Contour maps of the CoMSIA model gave insight into the feature requirements of the common backbone for the HDAC7 inhibitory activity. Finally, details of designing novel HDAC7 inhibitors were confirmed by a combination of receptor-based docking and ligand-based structure-activity relationship. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Yuan Yuan
- a Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University , Chengdu , Sichuan , P.R. China
| | - Zongyue Hu
- a Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University , Chengdu , Sichuan , P.R. China
| | - Minyue Bao
- b State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University , Chengdu , Sichuan , China
| | - Rong Sun
- a Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University , Chengdu , Sichuan , P.R. China
| | - Xin Long
- a Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University , Chengdu , Sichuan , P.R. China
| | - Li Long
- a Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University , Chengdu , Sichuan , P.R. China
| | - Jianzong Li
- a Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University , Chengdu , Sichuan , P.R. China
| | - Chuanfang Wu
- a Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University , Chengdu , Sichuan , P.R. China
| | - Jinku Bao
- a Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University , Chengdu , Sichuan , P.R. China.,b State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University , Chengdu , Sichuan , China
| |
Collapse
|
20
|
Histone deacetylases as targets for antitrypanosomal drugs. Future Sci OA 2018; 4:FSO325. [PMID: 30271613 PMCID: PMC6153458 DOI: 10.4155/fsoa-2018-0037] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/17/2018] [Indexed: 12/29/2022] Open
Abstract
Parasitic protozoa comprise several species that are causative agents of important diseases. These diseases are distributed throughout the world and include leishmaniasis, Chagas disease and sleeping sickness, malaria and toxoplasmosis. Treatment is based on drugs that were developed many years ago, which have side effects and produce resistant parasites. One approach for the development of new drugs is the identification of new molecular targets. We summarize the data on histone deacetylases, a class of enzymes that act on histones, which are closely associated with DNA and its regulation. These enzymes may constitute new targets for the development of antiparasitic protozoa drugs. Although several protozoan species are mentioned, members of the Trypanosomatidae family are the main focus of this short review. Parasitic protozoa comprise species that are causative agents of important diseases distributed throughout the world. The available drugs for treatment were developed many years ago, might cause side effects and produce resistant parasites. The identification of new molecular targets is required for the development of new drugs. Histone deacetylases act on histones, are closely associated with DNA and thus may constitute new targets for antiparasitic therapy, especially that against trypanosomatid protozoa.
Collapse
|
21
|
Loeuillet C, Touquet B, Oury B, Eddaikra N, Pons JL, Guichou JF, Labesse G, Sereno D. Synthesis of aminophenylhydroxamate and aminobenzylhydroxamate derivatives and in vitro screening for antiparasitic and histone deacetylase inhibitory activity. Int J Parasitol Drugs Drug Resist 2018; 8:59-66. [PMID: 29414107 PMCID: PMC6114082 DOI: 10.1016/j.ijpddr.2018.01.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/10/2018] [Accepted: 01/16/2018] [Indexed: 12/21/2022]
Abstract
A series of aminophenylhydroxamates and aminobenzylhydroxamates were synthesized and screened for their antiparasitic activity against Leishmania, Trypanosoma, and Toxoplasma. Their anti-histone deacetylase (HDAC) potency was determined. Moderate to no antileishmanial or antitrypanosomal activity was found (IC50 > 10 μM) that contrast with the highly efficient anti-Toxoplasma activity (IC50 < 1.0 μM) of these compounds. The antiparasitic activity of the synthetized compounds correlates well with their HDAC inhibitory activity. The best-performing compound (named 363) express a high anti-HDAC6 inhibitory activity (IC50 of 0.045 ± 0.015 μM) a moderate cytotoxicity and a high anti-Toxoplasma activity in the range of known anti-Toxoplasma compounds (IC50 of 0.35-2.25 μM). The calculated selectivity index (10-300 using different human cell lines) of the compound 363 makes it a lead compound for the future development of anti-Toxoplasma molecules.
Collapse
Affiliation(s)
- C Loeuillet
- Univ. Grenoble Alpes, CNRS, CHU Grenoble Alpes, Grenoble INP, TIMC-IMAG, F-38000 Grenoble, France; IRD, Univ Montpellier, MiVegec, Montpellier, France
| | - B Touquet
- Institute for Advanced Biosciences (IAB), Team Host-Pathogen Interactions & Immunity to Infection, INSERM U 1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France
| | - B Oury
- IRD, Univ Montpellier, InterTryp, Montpellier, France; IRD, Univ Montpellier, MiVegec, Montpellier, France
| | - N Eddaikra
- Laboratoire d'Eco-épidemiologie Parasitaire et Génétique des Populations, Institut Pasteur d'Alger, Route du Petit Staoueli, Dely Brahim, Alger, Algeria; Laboratoire de Biochimie Analytique et Biotechnologies, Université Mouloud Mammeri de Tizi Ouzou, Algeria
| | - J L Pons
- Centre de Biochimie Structurale (CBS), INSERM, CNRS, Université de Montpellier, France
| | - J F Guichou
- Centre de Biochimie Structurale (CBS), INSERM, CNRS, Université de Montpellier, France
| | - G Labesse
- Centre de Biochimie Structurale (CBS), INSERM, CNRS, Université de Montpellier, France
| | - D Sereno
- IRD, Univ Montpellier, InterTryp, Montpellier, France; IRD, Univ Montpellier, MiVegec, Montpellier, France.
| |
Collapse
|
22
|
Hailu GS, Robaa D, Forgione M, Sippl W, Rotili D, Mai A. Lysine Deacetylase Inhibitors in Parasites: Past, Present, and Future Perspectives. J Med Chem 2017; 60:4780-4804. [DOI: 10.1021/acs.jmedchem.6b01595] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Gebremedhin S. Hailu
- Dipartimento
di Chimica e Tecnologie del Farmaco “Sapienza” Università di Roma, 00185 Rome, Italy
| | - Dina Robaa
- Institute of Pharmacy, Martin-Luther-Universitat Halle-Wittenberg, Halle, Germany
| | - Mariantonietta Forgione
- Dipartimento
di Chimica e Tecnologie del Farmaco “Sapienza” Università di Roma, 00185 Rome, Italy
- Center
for Life Nano Science@Sapienza, Italian Institute of Technology, Viale Regina Elena 291, 00161 Rome, Italy
| | - Wolfgang Sippl
- Institute of Pharmacy, Martin-Luther-Universitat Halle-Wittenberg, Halle, Germany
| | - Dante Rotili
- Dipartimento
di Chimica e Tecnologie del Farmaco “Sapienza” Università di Roma, 00185 Rome, Italy
| | - Antonello Mai
- Dipartimento
di Chimica e Tecnologie del Farmaco “Sapienza” Università di Roma, 00185 Rome, Italy
- Istituto
Pasteur, Fondazione Cenci-Bolognetti, “Sapienza” Università di Roma, 00185 Rome, Italy
| |
Collapse
|
23
|
A safe and selective method for reduction of 2-nitrophenylacetic acid systems to N-aryl hydroxamic acids using continuous flow hydrogenation. Tetrahedron Lett 2017. [DOI: 10.1016/j.tetlet.2017.01.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
24
|
Comparative effects of histone deacetylases inhibitors and resveratrol on Trypanosoma cruzi replication, differentiation, infectivity and gene expression. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2016; 7:23-33. [PMID: 28038431 PMCID: PMC5199159 DOI: 10.1016/j.ijpddr.2016.12.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 12/07/2016] [Accepted: 12/07/2016] [Indexed: 12/22/2022]
Abstract
Histone post-translational modification, mediated by histone acetyltransferases and deacetylases, is one of the most studied factors affecting gene expression. Recent data showing differential histone acetylation states during the Trypanosoma cruzi cell cycle suggest a role for epigenetics in the control of this process. As a starting point to study the role of histone deacetylases in the control of gene expression and the consequences of their inhibition and activation in the biology of T. cruzi, two inhibitors for different histone deacetylases: trichostatin A for class I/II and sirtinol for class III and the activator resveratrol for class III, were tested on proliferative and infective forms of this parasite. The two inhibitors tested caused histone hyperacetylation whereas resveratrol showed the opposite effect on both parasite forms, indicating that a biologically active in vivo level of these compounds was achieved. Histone deacetylase inhibitors caused life stage-specific effects, increasing trypomastigotes infectivity and blocking metacyclogenesis. Moreover, these inhibitors affected specific transcript levels, with sirtinol causing the most pronounced change. On the other hand, resveratrol showed strong anti-parasitic effects. This compound diminished epimastigotes growth, promoted metacyclogenesis, reduced in vitro infection and blocked differentiation and/or replication of intracellular amastigotes. In conclusion, the data presented here supports the notion that these compounds can modulate T. cruzi gene expression, differentiation, infection and histones deacetylase activity. Furthermore, among the compounds tested in this study, the results point to Resveratrol as promising trypanocidal drug candidate. HDACis and resveratrol caused opposite changes on histones acetylation. HDACis and resveratrol affected parasites metacyclogenesis, growth and infection. Different HDACis caused opposite effects on transcript levels.
Collapse
|
25
|
Sola I, Artigas A, Taylor MC, Pérez-Areales FJ, Viayna E, Clos MV, Pérez B, Wright CW, Kelly JM, Muñoz-Torrero D. Synthesis and biological evaluation of N-cyanoalkyl-, N-aminoalkyl-, and N-guanidinoalkyl-substituted 4-aminoquinoline derivatives as potent, selective, brain permeable antitrypanosomal agents. Bioorg Med Chem 2016; 24:5162-5171. [PMID: 27591008 PMCID: PMC5080452 DOI: 10.1016/j.bmc.2016.08.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 08/19/2016] [Accepted: 08/20/2016] [Indexed: 11/30/2022]
Abstract
Current drugs against human African trypanosomiasis (HAT) suffer from several serious drawbacks. The search for novel, effective, brain permeable, safe, and inexpensive antitrypanosomal compounds is therefore an urgent need. We have recently reported that the 4-aminoquinoline derivative huprine Y, developed in our group as an anticholinesterasic agent, exhibits a submicromolar potency against Trypanosoma brucei and that its homo- and hetero-dimerization can result in to up to three-fold increased potency and selectivity. As an alternative strategy towards more potent smaller molecule anti-HAT agents, we have explored the introduction of ω-cyanoalkyl, ω-aminoalkyl, or ω-guanidinoalkyl chains at the primary amino group of huprine or the simplified 4-aminoquinoline analogue tacrine. Here, we describe the evaluation of a small in-house library and a second generation of newly synthesized derivatives, which has led to the identification of 13 side chain modified 4-aminoquinoline derivatives with submicromolar potencies against T. brucei. Among these compounds, the guanidinononyltacrine analogue 15e exhibits a 5-fold increased antitrypanosomal potency, 10-fold increased selectivity, and 100-fold decreased anticholinesterasic activity relative to the parent huprine Y. Its biological profile, lower molecular weight relative to dimeric compounds, reduced lipophilicity, and ease of synthesis, make it an interesting anti-HAT lead, amenable to further optimization to eliminate its remaining anticholinesterasic activity.
Collapse
Affiliation(s)
- Irene Sola
- Laboratory of Pharmaceutical Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - Albert Artigas
- Laboratory of Pharmaceutical Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - Martin C Taylor
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - F Javier Pérez-Areales
- Laboratory of Pharmaceutical Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - Elisabet Viayna
- Laboratory of Pharmaceutical Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - M Victòria Clos
- Department of Pharmacology, Therapeutics and Toxicology, Institute of Neurosciences, Autonomous University of Barcelona, E-08193, Bellaterra, Barcelona, Spain
| | - Belén Pérez
- Department of Pharmacology, Therapeutics and Toxicology, Institute of Neurosciences, Autonomous University of Barcelona, E-08193, Bellaterra, Barcelona, Spain
| | - Colin W Wright
- Bradford School of Pharmacy, University of Bradford, West Yorkshire BD7 1 DP, United Kingdom
| | - John M Kelly
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Diego Muñoz-Torrero
- Laboratory of Pharmaceutical Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain.
| |
Collapse
|
26
|
Repurposing strategies for tropical disease drug discovery. Bioorg Med Chem Lett 2016; 26:2569-76. [PMID: 27080183 DOI: 10.1016/j.bmcl.2016.03.103] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/21/2016] [Accepted: 03/29/2016] [Indexed: 12/22/2022]
Abstract
Neglected tropical diseases (NTDs) and other diseases of the developing world, such as malaria, attract research investments that are disproportionately low compared to their impact on human health worldwide. Therefore, pragmatic methods for launching new drug discovery programs have emerged that repurpose existing chemical matter as new drugs or new starting points for optimization. In this Digest we describe applications of different repurposing approaches for NTDs, and provide a means by which these approaches may be differentiated from each other. These include drug repurposing, target repurposing, target class repurposing, and lead repurposing.
Collapse
|
27
|
Di Pietro O, Vicente-García E, Taylor MC, Berenguer D, Viayna E, Lanzoni A, Sola I, Sayago H, Riera C, Fisa R, Clos MV, Pérez B, Kelly JM, Lavilla R, Muñoz-Torrero D. Multicomponent reaction-based synthesis and biological evaluation of tricyclic heterofused quinolines with multi-trypanosomatid activity. Eur J Med Chem 2015; 105:120-37. [PMID: 26479031 PMCID: PMC4638191 DOI: 10.1016/j.ejmech.2015.10.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 09/28/2015] [Accepted: 10/06/2015] [Indexed: 11/28/2022]
Abstract
Human African trypanosomiasis (HAT), Chagas disease and leishmaniasis, which are caused by the trypanosomatids Trypanosoma brucei, Trypanosoma cruzi and Leishmania species, are among the most deadly neglected tropical diseases. The development of drugs that are active against several trypanosomatids is appealing from a clinical and economic viewpoint, and seems feasible, as these parasites share metabolic pathways and hence might be treatable by common drugs. From benzonapthyridine 1, an inhibitor of acetylcholinesterase (AChE) for which we have found a remarkable trypanocidal activity, we have designed and synthesized novel benzo[h][1,6]naphthyridines, pyrrolo[3,2-c]quinolines, azepino[3,2-c]quinolines, and pyrano[3,2-c]quinolines through 2–4-step sequences featuring an initial multicomponent Povarov reaction as the key step. To assess the therapeutic potential of the novel compounds, we have evaluated their in vitro activity against T. brucei, T. cruzi, and Leishmania infantum, as well as their brain permeability, which is of specific interest for the treatment of late-stage HAT. To assess their potential toxicity, we have determined their cytotoxicity against rat myoblast L6 cells and their AChE inhibitory activity. Several tricyclic heterofused quinoline derivatives were found to display an interesting multi-trypanosomatid profile, with one-digit micromolar potencies against two of these parasites and two-digit micromolar potency against the other. Pyranoquinoline 39, which displays IC50 values of 1.5 μM, 6.1 μM and 29.2 μM against T. brucei, L. infantum and T. cruzi, respectively, brain permeability, better drug-like properties (lower lipophilicity and molecular weight and higher CNS MPO desirability score) than hit 1, and the lowest AChE inhibitory activity of the series (IC50 > 30 μM), emerges as an interesting multi-trypanosomatid lead, amenable to further optimization particularly in terms of its selectivity index over mammalian cells. Novel classes of tricyclic heterofused quinolines have been synthesized. Their 2–4-step syntheses involve a multicomponent Povarov reaction as the key step. Some compounds exhibit single digit micromolar potencies against 2 trypanosomatids. All compounds with multi-trypanosomatid activity can cross the blood–brain barrier. Most compounds with multi-trypanosomatid activity have drug like properties.
Collapse
Affiliation(s)
- Ornella Di Pietro
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Facultat de Farmàcia, and Institut de Biomedicina (IBUB), Universitat de Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | | | - Martin C Taylor
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Diana Berenguer
- Laboratori de Parasitologia, Departament de Microbiologia i Parasitologia Sanitàries, Facultat de Farmàcia, Universitat de Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Elisabet Viayna
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Facultat de Farmàcia, and Institut de Biomedicina (IBUB), Universitat de Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Anna Lanzoni
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Facultat de Farmàcia, and Institut de Biomedicina (IBUB), Universitat de Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Irene Sola
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Facultat de Farmàcia, and Institut de Biomedicina (IBUB), Universitat de Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Helena Sayago
- Barcelona Science Park, Baldiri Reixac, 10-12, E-08028, Barcelona, Spain
| | - Cristina Riera
- Laboratori de Parasitologia, Departament de Microbiologia i Parasitologia Sanitàries, Facultat de Farmàcia, Universitat de Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Roser Fisa
- Laboratori de Parasitologia, Departament de Microbiologia i Parasitologia Sanitàries, Facultat de Farmàcia, Universitat de Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - M Victòria Clos
- Departament de Farmacologia, de Terapèutica i de Toxicologia, Institut de Neurociències, Universitat Autònoma de Barcelona, E-08193, Bellaterra, Barcelona, Spain
| | - Belén Pérez
- Departament de Farmacologia, de Terapèutica i de Toxicologia, Institut de Neurociències, Universitat Autònoma de Barcelona, E-08193, Bellaterra, Barcelona, Spain
| | - John M Kelly
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Rodolfo Lavilla
- Barcelona Science Park, Baldiri Reixac, 10-12, E-08028, Barcelona, Spain; Laboratori de Química Orgànica, Facultat de Farmàcia, Universitat de Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Diego Muñoz-Torrero
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Facultat de Farmàcia, and Institut de Biomedicina (IBUB), Universitat de Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain.
| |
Collapse
|
28
|
Wang Q, Rosa BA, Nare B, Powell K, Valente S, Rotili D, Mai A, Marshall GR, Mitreva M. Targeting Lysine Deacetylases (KDACs) in Parasites. PLoS Negl Trop Dis 2015; 9:e0004026. [PMID: 26402733 PMCID: PMC4581690 DOI: 10.1371/journal.pntd.0004026] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 08/02/2015] [Indexed: 12/30/2022] Open
Abstract
Due to an increasing problem of drug resistance among almost all parasites species ranging from protists to worms, there is an urgent need to explore new drug targets and their inhibitors to provide new and effective parasitic therapeutics. In this regard, there is growing interest in exploring known drug leads of human epigenetic enzymes as potential starting points to develop novel treatments for parasitic diseases. This approach of repurposing (starting with validated targets and inhibitors) is quite attractive since it has the potential to reduce the expense of drug development and accelerate the process of developing novel drug candidates for parasite control. Lysine deacetylases (KDACs) are among the most studied epigenetic drug targets of humans, and a broad range of small-molecule inhibitors for these enzymes have been reported. In this work, we identify the KDAC protein families in representative species across important classes of parasites, screen a compound library of 23 hydroxamate- or benzamide-based small molecules KDAC inhibitors, and report their activities against a range of parasitic species, including the pathogen of malaria (Plasmodium falciparum), kinetoplastids (Trypanosoma brucei and Leishmania donovani), and nematodes (Brugia malayi, Dirofilaria immitis and Haemonchus contortus). Compound activity against parasites is compared to that observed against the mammalian cell line (L929 mouse fibroblast) in order to determine potential parasite-versus-host selectivity). The compounds showed nanomolar to sub-nanomolar potency against various parasites, and some selectivity was observed within the small panel of compounds tested. The possible binding modes of the active compounds at the different protein target sites within different species were explored by docking to homology models to help guide the discovery of more selective, parasite-specific inhibitors. This current work supports previous studies that explored the use of KDAC inhibitors in targeting Plasmodium to develop new anti-malarial treatments, and also pioneers experiments with these KDAC inhibitors as potential new anthelminthics. The selectivity observed begins to address the challenges of targeting specific parasitic diseases while limiting host toxicity. Due to pandemic drug resistance in the treatment of parasitic infections, there is an urgent need to identify novel drug targets and their associated drug compounds. Although “drug repurposing”, i.e. the application of known drugs and compounds to new indications such as infectious diseases, provides a cost effective approach in the development of novel therapeutics, selectivity is one of the major obstacles to overcome in getting such compounds into clinical trials as anti-parasitic drugs. Using the lysine deacetylases (KDACs) as an example, we explored the activities of a panel of known inhibitors against the KDAC targets in a range of parasitic organisms. The computational study of their binding modes to the targets (by docking the compounds to the homology models within different organisms in comparison with the human proteins) helps to rationalize the different activities observed and provide insight on the optimization of lead compounds to improve selectivity. Our work provides support of “drug repurposing” in the treatment of parasitic diseases, and demonstrates the necessity of optimizing these leads for the ultimate goal of preparing them for clinical use.
Collapse
Affiliation(s)
- Qi Wang
- The Genome Institute, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Bruce A. Rosa
- The Genome Institute, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Bakela Nare
- SCYNEXIS, Inc, Research Triangle Park, North Carolina, United States of America
| | - Kerrie Powell
- SCYNEXIS, Inc, Research Triangle Park, North Carolina, United States of America
| | - Sergio Valente
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, Università degli Studi di Roma “La Sapienza”, Roma, Italy
| | - Dante Rotili
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, Università degli Studi di Roma “La Sapienza”, Roma, Italy
| | - Antonello Mai
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, Università degli Studi di Roma “La Sapienza”, Roma, Italy
| | - Garland R. Marshall
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Makedonka Mitreva
- The Genome Institute, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Departments of Genetics and of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
29
|
Engel JA, Jones AJ, Avery VM, Sumanadasa SDM, Ng SS, Fairlie DP, Skinner-Adams T, Andrews KT. Profiling the anti-protozoal activity of anti-cancer HDAC inhibitors against Plasmodium and Trypanosoma parasites. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2015. [PMID: 26199860 PMCID: PMC4506969 DOI: 10.1016/j.ijpddr.2015.05.004] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Histone deacetylase (HDAC) enzymes work together with histone acetyltransferases (HATs) to reversibly acetylate both histone and non-histone proteins. As a result, these enzymes are involved in regulating chromatin structure and gene expression as well as other important cellular processes. HDACs are validated drug targets for some types of cancer, with four HDAC inhibitors clinically approved. However, they are also showing promise as novel drug targets for other indications, including malaria and other parasitic diseases. In this study the in vitro activity of four anti-cancer HDAC inhibitors was examined against parasites that cause malaria and trypanosomiasis. Three of these inhibitors, suberoylanilide hydroxamic acid (SAHA; vorinostat®), romidepsin (Istodax®) and belinostat (Beleodaq®), are clinically approved for the treatment of T-cell lymphoma, while the fourth, panobinostat, has recently been approved for combination therapy use in certain patients with multiple myeloma. All HDAC inhibitors were found to inhibit the growth of asexual-stage Plasmodium falciparum malaria parasites in the nanomolar range (IC50 10–200 nM), while only romidepsin was active at sub-μM concentrations against bloodstream form Trypanosoma brucei brucei parasites (IC50 35 nM). The compounds were found to have some selectivity for malaria parasites compared with mammalian cells, but were not selective for trypanosome parasites versus mammalian cells. All compounds caused hyperacetylation of histone and non-histone proteins in P. falciparum asexual stage parasites and inhibited deacetylase activity in P. falciparum nuclear extracts in addition to recombinant PfHDAC1 activity. P. falciparum histone hyperacetylation data indicate that HDAC inhibitors may differentially affect the acetylation profiles of histone H3 and H4. Four clinically approved anti-cancer HDAC inhibitors potently inhibited P. falciparum. Only one, Romidepsin, was active against T. b. brucei parasites. All compounds hyperacetylated histone and non-histone proteins in P. falciparum. Some differential effects on Plasmodium histone acetylation were observed. All compounds inhibited Plasmodium nuclear deacetylase activity and PfHDAC1.
Collapse
Affiliation(s)
- Jessica A Engel
- Eskitis Institute for Drug Discovery, Griffith University, Queensland, Australia
| | - Amy J Jones
- Eskitis Institute for Drug Discovery, Griffith University, Queensland, Australia
| | - Vicky M Avery
- Eskitis Institute for Drug Discovery, Griffith University, Queensland, Australia
| | | | - Susanna S Ng
- Eskitis Institute for Drug Discovery, Griffith University, Queensland, Australia
| | - David P Fairlie
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Q4072, Australia
| | - Tina Skinner-Adams
- Eskitis Institute for Drug Discovery, Griffith University, Queensland, Australia
| | - Katherine T Andrews
- Eskitis Institute for Drug Discovery, Griffith University, Queensland, Australia
| |
Collapse
|
30
|
HDAC8: a multifaceted target for therapeutic interventions. Trends Pharmacol Sci 2015; 36:481-92. [PMID: 26013035 DOI: 10.1016/j.tips.2015.04.013] [Citation(s) in RCA: 200] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 04/27/2015] [Accepted: 04/28/2015] [Indexed: 02/08/2023]
Abstract
Histone deacetylase 8 (HDAC8) is a class I histone deacetylase implicated as a therapeutic target in various diseases, including cancer, X-linked intellectual disability, and parasitic infections. It is a structurally well-characterized enzyme that also deacetylates nonhistone proteins. In cancer, HDAC8 is a major 'epigenetic player' that is linked to deregulated expression or interaction with transcription factors critical to tumorigenesis. In the parasite Schistosoma mansoni and in viral infections, HDAC8 is a novel target to subdue infection. The current challenge remains in the development of potent selective inhibitors that would specifically target HDAC8 with fewer adverse effects compared with pan-HDAC inhibitors. Here, we review HDAC8 as a drug target and discuss inhibitors with respect to their structural features and therapeutic interventions.
Collapse
|