1
|
Sezer G, Çetinkaya Ü. Vinpocetine increases the microsporicidal effect of albendazole on Encephalitozoon intestinalis. Med Mycol 2024; 62:myae072. [PMID: 39043448 DOI: 10.1093/mmy/myae072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/03/2024] [Accepted: 07/22/2024] [Indexed: 07/25/2024] Open
Abstract
Microsporidia are obligate, intracellular, spore-forming eukaryotic fungi that infect humans and animals. In the treatment of disseminated microsporidiosis albendazole is the choice of drug. In recent years, antiparasitic activity of phosphodiesterase (PDE) enzyme inhibitors has been demonstrated against parasites and fungi, however, there is no information on microsporidia. Vinpocetine is currently used as a cerebral vasodilator drug and also as a dietary supplement to improve cognitive functions. Vinpocetine inhibits PDE1, so we aimed to investigate whether vinpocetine alone or in combination with albendazole has any effect on the spore load of Encephalitozoon intestinalis (E. intestinalis)-infected HEK293 cells. After determining the noncytotoxic concentrations of vinpocetine and albendazole on the host cell by MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, HEK293 cells were infected with E. intestinalis spores. Then, two different concentrations of vinpocetine, albendazole, and a combination of both drugs were applied to the cells with an interval of 72 h for 15 days. Spore load of the cells was analyzed by real-time PCR. After the last treatment, spore Deoxyribonucleic Acid (DNA) load was significantly reduced only in the group treated with 14 ng/ml albendazole. It was not different from control in groups treated with 7 ng/ml albendazole and 4-20 µM vinpocetine. However, the combination of vinpocetine significantly increased the effect of albendazole at both concentrations. To our knowledge, this is the first study to investigate the microsporicidal activity of vinpocetine as well as its combinations with albendazole. However, further studies are needed to investigate the mechanism of action and also confirm in vivo conditions.
Collapse
Affiliation(s)
- Gülay Sezer
- Department of Pharmacology, Faculty of Medicine, Erciyes University, 38039 Talas/Kayseri, Türkiye
- Genkök Genome and Stem Cell Center, Erciyes University, 38039 Talas/Kayseri, Türkiye
| | - Ülfet Çetinkaya
- Genkök Genome and Stem Cell Center, Erciyes University, 38039 Talas/Kayseri, Türkiye
- Halil Bayraktar Health Vocational High School, Erciyes University, 38039 Talas/Kayseri, Türkiye
| |
Collapse
|
2
|
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
|
3
|
Jamabo M, Mahlalela M, Edkins AL, Boshoff A. Tackling Sleeping Sickness: Current and Promising Therapeutics and Treatment Strategies. Int J Mol Sci 2023; 24:12529. [PMID: 37569903 PMCID: PMC10420020 DOI: 10.3390/ijms241512529] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Human African trypanosomiasis is a neglected tropical disease caused by the extracellular protozoan parasite Trypanosoma brucei, and targeted for eradication by 2030. The COVID-19 pandemic contributed to the lengthening of the proposed time frame for eliminating human African trypanosomiasis as control programs were interrupted. Armed with extensive antigenic variation and the depletion of the B cell population during an infectious cycle, attempts to develop a vaccine have remained unachievable. With the absence of a vaccine, control of the disease has relied heavily on intensive screening measures and the use of drugs. The chemotherapeutics previously available for disease management were plagued by issues such as toxicity, resistance, and difficulty in administration. The approval of the latest and first oral drug, fexinidazole, is a major chemotherapeutic achievement for the treatment of human African trypanosomiasis in the past few decades. Timely and accurate diagnosis is essential for effective treatment, while poor compliance and resistance remain outstanding challenges. Drug discovery is on-going, and herein we review the recent advances in anti-trypanosomal drug discovery, including novel potential drug targets. The numerous challenges associated with disease eradication will also be addressed.
Collapse
Affiliation(s)
- Miebaka Jamabo
- Biotechnology Innovation Centre, Rhodes University, Makhanda 6139, South Africa; (M.J.); (M.M.)
| | - Maduma Mahlalela
- Biotechnology Innovation Centre, Rhodes University, Makhanda 6139, South Africa; (M.J.); (M.M.)
| | - Adrienne L. Edkins
- Department of Biochemistry and Microbiology, Biomedical Biotechnology Research Centre (BioBRU), Rhodes University, Makhanda 6139, South Africa;
| | - Aileen Boshoff
- Biotechnology Innovation Centre, Rhodes University, Makhanda 6139, South Africa; (M.J.); (M.M.)
| |
Collapse
|
4
|
Zheng Y, Schroeder S, Kanev GK, Botros SS, William S, Sabra ANA, Maes L, Caljon G, Gil C, Martinez A, Salado IG, Augustyns K, Edink E, Sijm M, de Heuvel E, de Esch IJP, van der Meer T, Siderius M, Sterk GJ, Brown D, Leurs R. To Target or Not to Target Schistosoma mansoni Cyclic Nucleotide Phosphodiesterase 4A? Int J Mol Sci 2023; 24:ijms24076817. [PMID: 37047792 PMCID: PMC10095301 DOI: 10.3390/ijms24076817] [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: 02/07/2023] [Revised: 03/31/2023] [Accepted: 04/01/2023] [Indexed: 04/14/2023] Open
Abstract
Schistosomiasis is a neglected tropical disease with high morbidity. Recently, the Schistosoma mansoni phosphodiesterase SmPDE4A was suggested as a putative new drug target. To support SmPDE4A targeted drug discovery, we cloned, isolated, and biochemically characterized the full-length and catalytic domains of SmPDE4A. The enzymatically active catalytic domain was crystallized in the apo-form (PDB code: 6FG5) and in the cAMP- and AMP-bound states (PDB code: 6EZU). The SmPDE4A catalytic domain resembles human PDE4 more than parasite PDEs because it lacks the parasite PDE-specific P-pocket. Purified SmPDE4A proteins (full-length and catalytic domain) were used to profile an in-house library of PDE inhibitors (PDE4NPD toolbox). This screening identified tetrahydrophthalazinones and benzamides as potential hits. The PDE inhibitor NPD-0001 was the most active tetrahydrophthalazinone, whereas the approved human PDE4 inhibitors roflumilast and piclamilast were the most potent benzamides. As a follow-up, 83 benzamide analogs were prepared, but the inhibitory potency of the initial hits was not improved. Finally, NPD-0001 and roflumilast were evaluated in an in vitro anti-S. mansoni assay. Unfortunately, both SmPDE4A inhibitors were not effective in worm killing and only weakly affected the egg-laying at high micromolar concentrations. Consequently, the results with these SmPDE4A inhibitors strongly suggest that SmPDE4A is not a suitable target for anti-schistosomiasis therapy.
Collapse
Affiliation(s)
- Yang Zheng
- Division of Medicinal Chemistry, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | | | - Georgi K Kanev
- Division of Medicinal Chemistry, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Sanaa S Botros
- Pharmacology Department, Theodor Bilharz Research Institute, Warrak El-Hadar, Imbaba, P.O. Box 30, Giza 12411, Egypt
| | - Samia William
- Parasitology Department, Theodor Bilharz Research Institute, Warrak El-Hadar, Imbaba, P.O. Box 30, Giza 12411, Egypt
| | - Abdel-Nasser A Sabra
- Pharmacology Department, Theodor Bilharz Research Institute, Warrak El-Hadar, Imbaba, P.O. Box 30, Giza 12411, Egypt
| | - Louis Maes
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Carmen Gil
- Centro de Investigaciones Biologicas (CIB-CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Ana Martinez
- Centro de Investigaciones Biologicas (CIB-CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Irene G Salado
- Medicinal Chemistry, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Koen Augustyns
- Medicinal Chemistry, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Ewald Edink
- Division of Medicinal Chemistry, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Maarten Sijm
- Division of Medicinal Chemistry, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Erik de Heuvel
- Division of Medicinal Chemistry, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Iwan J P de Esch
- Division of Medicinal Chemistry, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Tiffany van der Meer
- Division of Medicinal Chemistry, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Marco Siderius
- Division of Medicinal Chemistry, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Geert Jan Sterk
- Division of Medicinal Chemistry, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - David Brown
- School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK
| | - Rob Leurs
- Division of Medicinal Chemistry, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
5
|
Popruk S, Abu A, Ampawong S, Thiangtrongjit T, Tipthara P, Tarning J, Sreesai S, Reamtong O. Mass Spectrometry-Based Metabolomics Revealed Effects of Metronidazole on Giardia duodenalis. Pharmaceuticals (Basel) 2023; 16:ph16030408. [PMID: 36986506 PMCID: PMC10052756 DOI: 10.3390/ph16030408] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/22/2023] [Accepted: 03/02/2023] [Indexed: 03/30/2023] Open
Abstract
Giardia duodenalis is a significant protozoan that affects humans and animals. An estimated 280 million G. duodenalis diarrheal cases are recorded annually. Pharmacological therapy is crucial for controlling giardiasis. Metronidazole is the first-line therapy for treating giardiasis. Several metronidazole targets have been proposed. However, the downstream signaling pathways of these targets with respect to their antigiardial action are unclear. In addition, several giardiasis cases have demonstrated treatment failures and drug resistance. Therefore, the development of novel drugs is an urgent need. In this study, we performed a mass spectrometry-based metabolomics study to understand the systemic effects of metronidazole in G. duodenalis. A thorough analysis of metronidazole processes helps identify potential molecular pathways essential for parasite survival. The results demonstrated 350 altered metabolites after exposure to metronidazole. Squamosinin A and N-(2-hydroxyethyl)hexacosanamide were the most up-regulated and down-regulated metabolites, respectively. Proteasome and glycerophospholipid metabolisms demonstrated significant differential pathways. Comparing glycerophospholipid metabolisms of G. duodenalis and humans, the parasite glycerophosphodiester phosphodiesterase was distinct from humans. This protein is considered a potential drug target for treating giardiasis. This study improved our understanding of the effects of metronidazole and identified new potential therapeutic targets for future drug development.
Collapse
Affiliation(s)
- Supaluk Popruk
- Department of Protozoology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Amanee Abu
- Department of Protozoology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Sumate Ampawong
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Tipparat Thiangtrongjit
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Phornpimon Tipthara
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Joel Tarning
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX1 4BH, UK
| | - Suthasinee Sreesai
- Central Equipment Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
6
|
Solórzano ER, Pastore P, Dolmella A, Cazorla S, Cassará MLA, Sankaran SV, Thamotharan S, Gil DM. Importance of R-CH3⋯O tetrel bonding and vinyl⋯aryl stacking interactions in stabilizing the crystal packing of 2’,4’-dihydroxy-3’-methoxychalcone: Exploration of antileishmanial activity and molecular docking studies. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
7
|
De Araújo JS, da Silva PB, Batista MM, Peres RB, Cardoso-Santos C, Kalejaiye TD, Munday JC, De Heuvel E, Sterk GJ, Augustyns K, Salado IG, Matheeussen A, De Esch I, De Koning HP, Leurs R, Maes L, Soeiro MDNC. Evaluation of phthalazinone phosphodiesterase inhibitors with improved activity and selectivity against Trypanosoma cruzi. J Antimicrob Chemother 2021; 75:958-967. [PMID: 31860098 DOI: 10.1093/jac/dkz516] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/15/2019] [Accepted: 11/08/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Chagas' disease, caused by the protozoan parasite Trypanosoma cruzi, needs urgent alternative therapeutic options as the treatments currently available display severe limitations, mainly related to efficacy and toxicity. OBJECTIVES As phosphodiesterases (PDEs) have been claimed as novel targets against T. cruzi, our aim was to evaluate the biological aspects of 12 new phthalazinone PDE inhibitors against different T. cruzi strains and parasite forms relevant for human infection. METHODS In vitro trypanocidal activity of the inhibitors was assessed alone and in combination with benznidazole. Their effects on parasite ultrastructural and cAMP levels were determined. PDE mRNA levels from the different T. cruzi forms were measured by quantitative reverse transcription PCR. RESULTS Five TcrPDEs were found to be expressed in all parasite stages. Four compounds displayed strong effects against intracellular amastigotes. Against bloodstream trypomastigotes (BTs), three were at least as potent as benznidazole. In vitro combination therapy with one of the most active inhibitors on both parasite forms (NPD-040) plus benznidazole demonstrated a quite synergistic profile (xΣ FICI = 0.58) against intracellular amastigotes but no interaction (xΣ FICI = 1.27) when BTs were assayed. BTs treated with NPD-040 presented disrupted Golgi apparatus, a swollen flagellar pocket and signs of autophagy. cAMP measurements of untreated parasites showed that amastigotes have higher ability to efflux this second messenger than BTs. NPD-001 and NPD-040 increase the intracellular cAMP content in both BTs and amastigotes, which is also released into the extracellular milieu. CONCLUSIONS The findings demonstrate the potential of PDE inhibitors as anti-T. cruzi drug candidates.
Collapse
Affiliation(s)
| | | | - Marcos Meuser Batista
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Raiza Brandão Peres
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Camila Cardoso-Santos
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Titilola D Kalejaiye
- Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Jane C Munday
- Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Erik De Heuvel
- Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines & Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Geert Jan Sterk
- Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines & Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Koen Augustyns
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
| | - Irene G Salado
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
| | - An Matheeussen
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Iwan De Esch
- Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines & Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Harry P De Koning
- Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Rob Leurs
- Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines & Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Louis Maes
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | | |
Collapse
|
8
|
N'Dri ME, Royer L, Lavazec C. Tadalafil impacts the mechanical properties of Plasmodium falciparum gametocyte-infected erythrocytes. Mol Biochem Parasitol 2021; 244:111392. [PMID: 34171456 DOI: 10.1016/j.molbiopara.2021.111392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/03/2021] [Accepted: 06/14/2021] [Indexed: 10/21/2022]
Abstract
Plasmodium falciparum gametocytes modify the mechanical properties of their erythrocyte host to persist for several weeks in the blood circulation and to be available for mosquitoes. These changes are tightly regulated by the plasmodial phosphodiesterase delta that decreases both the stiffness and the permeability of the infected host cell. Here, we address the effect of the phosphodiesterase inhibitor tadalafil on deformability and permeability of gametocyte-infected erythrocytes. We show that this inhibitor drastically increases isosmotic lysis of gametocyte-infected erythrocytes and impairs their ability to circulate in an in vitro model for splenic retention. These findings indicate that tadalafil represents a novel drug lead potentially capable of blocking malaria parasite transmission by impacting gametocyte circulation.
Collapse
Affiliation(s)
- Marie-Esther N'Dri
- Inserm U1016, CNRS UMR8104, Université de Paris, Institut Cochin, Paris, France
| | - Ludivine Royer
- Inserm U1016, CNRS UMR8104, Université de Paris, Institut Cochin, Paris, France
| | - Catherine Lavazec
- Inserm U1016, CNRS UMR8104, Université de Paris, Institut Cochin, Paris, France.
| |
Collapse
|
9
|
Discovery of Amoebicidal Compounds by Combining Computational and Experimental Approaches. Antimicrob Agents Chemother 2021; 65:AAC.01749-20. [PMID: 33229426 DOI: 10.1128/aac.01749-20] [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: 08/13/2020] [Accepted: 11/15/2020] [Indexed: 11/20/2022] Open
Abstract
Pathogenic and opportunistic free-living amoebae such as Acanthamoeba spp. can cause keratitis (Acanthamoeba keratitis [AK]), which may ultimately lead to permanent visual impairment or blindness. Acanthamoeba can also cause rare but usually fatal granulomatous amoebic encephalitis (GAE). Current therapeutic options for AK require a lengthy treatment with nonspecific drugs that are often associated with adverse effects. Recent developments in the field led us to target cAMP pathways, specifically phosphodiesterase. Guided by computational tools, we targeted the Acanthamoeba phosphodiesterase RegA. Computational studies led to the construction and validation of a homology model followed by a virtual screening protocol guided by induced-fit docking and chemical scaffold analysis using our medicinal and biological chemistry (MBC) chemical library. Subsequently, 18 virtual screening hits were prioritized for further testing in vitro against Acanthamoeba castellanii, identifying amoebicidal hits containing piperidine and urea imidazole cores. Promising activities were confirmed in the resistant cyst form of the amoeba and in additional clinical Acanthamoeba strains, increasing their therapeutic potential. Mechanism-of-action studies revealed that these compounds produce apoptosis through reactive oxygen species (ROS)-mediated mitochondrial damage. These chemical families show promise for further optimization to produce effective antiacanthamoebal drugs.
Collapse
|
10
|
Silva DKC, Teixeira JS, Moreira DRM, da Silva TF, Barreiro EJDL, de Freitas HF, Pita SSDR, Teles ALB, Guimarães ET, Soares MBP. In Vitro, In Vivo and In Silico Effectiveness of LASSBio-1386, an N-Acyl Hydrazone Derivative Phosphodiesterase-4 Inhibitor, Against Leishmania amazonensis. Front Pharmacol 2021; 11:590544. [PMID: 33390966 PMCID: PMC7772393 DOI: 10.3389/fphar.2020.590544] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 11/03/2020] [Indexed: 11/16/2022] Open
Abstract
Leishmaniasis are group of neglected diseases with worldwide distribution that affect about 12 million people. The current treatment is limited and may cause severe adverse effects, and thus, the search for new drugs more effective and less toxic is relevant. We have previously investigated the immunomodulatory effects of LASSBio-1386, an N-acylhydrazone derivative. Here we investigated the in vitro and in vivo activity of LASSBio-1386 against L. amazonensis. LASSBio-1386 inhibited the proliferation of promastigotes of L. amazonensis (EC50 = 2.4 ± 0.48 µM), while presenting low cytotoxicity to macrophages (CC50 = 74.1 ± 2.9 µM). In vitro incubation with LASSBio-1386 reduced the percentage of Leishmania-infected macrophages and the number of intracellular parasites (EC50 = 9.42 ± 0.64 µM). Also, in vivo treatment of BALB/c mice infected with L. amazonensis resulted in a decrease of lesion size, parasitic load and caused histopathological alterations, when compared to vehicle-treated control. Moreover, LASSBio-1386 caused ultrastructural changes, arrested cell cycle in G0/G1 phase and did not alter the membrane mitochondrial potential of L. amazonensis. Aiming to its possible molecular interactions, we performed docking and molecular dynamics studies on Leishmania phosphodiesterase B1 (PDB code: 2R8Q) and LASSBio-1386. The computational analyses suggest that LASSBio-1386 acts against Leishmania through the modulation of leishmanial PDE activity. In conclusion, our results indicate that LASSBio-1386 is a promising candidate for the development of new leishmaniasis treatment.
Collapse
Affiliation(s)
- Dahara Keyse Carvalho Silva
- Departamento de Ciências da Vida, Núcleo de Estudo e Pesquisa em Histopatologia, Universidade Estadual da Bahia (UNEB), Salvador, Brazil.,Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
| | - Jessicada Silva Teixeira
- Departamento de Ciências da Vida, Núcleo de Estudo e Pesquisa em Histopatologia, Universidade Estadual da Bahia (UNEB), Salvador, Brazil.,Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
| | - Diogo Rodrigo Magalhães Moreira
- Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
| | - Tiago Fernandes da Silva
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio®), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Eliezer Jesus de Lacerda Barreiro
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio®), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Humberto Fonseca de Freitas
- Laboratório de Bioinformática e Modelagem Molecular (LaBiMM), Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Brazil
| | - Samuel Silva da Rocha Pita
- Laboratório de Bioinformática e Modelagem Molecular (LaBiMM), Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Brazil
| | - André Lacerda Braga Teles
- Departamento de Ciências da Vida, Laboratório de Modelagem Molecular Medicinal e Toxicológica, Universidade Estadual da Bahia (UNEB), Salvador, Brazil
| | - Elisalva Teixeira Guimarães
- Departamento de Ciências da Vida, Núcleo de Estudo e Pesquisa em Histopatologia, Universidade Estadual da Bahia (UNEB), Salvador, Brazil.,Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
| | - Milena Botelho Pereira Soares
- Laboratório de Engenharia Tecidual e Imunofarmacologia, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil.,Instituto Senai de Inovação em Sistemas Avançados em Saúde, Senai/Cimatec, Salvador, Brazil
| |
Collapse
|
11
|
Munday JC, Kunz S, Kalejaiye TD, Siderius M, Schroeder S, Paape D, Alghamdi AH, Abbasi Z, Huang SX, Donachie AM, William S, Sabra AN, Sterk GJ, Botros SS, Brown DG, Hoffman CS, Leurs R, de Koning HP. Cloning and functional complementation of ten Schistosoma mansoni phosphodiesterases expressed in the mammalian host stages. PLoS Negl Trop Dis 2020; 14:e0008447. [PMID: 32730343 PMCID: PMC7430754 DOI: 10.1371/journal.pntd.0008447] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/17/2020] [Accepted: 06/02/2020] [Indexed: 01/29/2023] Open
Abstract
Only a single drug against schistosomiasis is currently available and new drug development is urgently required but very few drug targets have been validated and characterised. However, regulatory systems including cyclic nucleotide metabolism are emerging as primary candidates for drug discovery. Here, we report the cloning of ten cyclic nucleotide phosphodiesterase (PDE) genes of S. mansoni, out of a total of 11 identified in its genome. We classify these PDEs by homology to human PDEs. Male worms displayed higher expression levels for all PDEs, in mature and juvenile worms, and schistosomula. Several functional complementation approaches were used to characterise these genes. We constructed a Trypanosoma brucei cell line in which expression of a cAMP-degrading PDE complements the deletion of TbrPDEB1/B2. Inhibitor screens of these cells expressing only either SmPDE4A, TbrPDEB1 or TbrPDEB2, identified highly potent inhibitors of the S. mansoni enzyme that elevated the cellular cAMP concentration. We further expressed most of the cloned SmPDEs in two pde1Δ/pde2Δ strains of Saccharomyces cerevisiae and some also in a specialised strain of Schizosacharomyces pombe. Five PDEs, SmPDE1, SmPDE4A, SmPDE8, SmPDE9A and SmPDE11 successfully complemented the S. cerevisiae strains, and SmPDE7var also complemented to a lesser degree, in liquid culture. SmPDE4A, SmPDE8 and SmPDE11 were further assessed in S. pombe for hydrolysis of cAMP and cGMP; SmPDE11 displayed considerable preferrence for cGMP over cAMP. These results and tools enable the pursuit of a rigorous drug discovery program based on inhibitors of S. mansoni PDEs.
Collapse
Affiliation(s)
- Jane C. Munday
- Institute of Infection, Immunity and inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
| | - Stefan Kunz
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, The Netherlands
| | - Titilola D. Kalejaiye
- Institute of Infection, Immunity and inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
| | - Marco Siderius
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, The Netherlands
| | | | - Daniel Paape
- Institute of Infection, Immunity and inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
| | - Ali H. Alghamdi
- Institute of Infection, Immunity and inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
| | - Zainab Abbasi
- Biology Department, Boston College, Chestnut Hill, Massachusetts, United States of America
| | - Sheng Xiang Huang
- Biology Department, Boston College, Chestnut Hill, Massachusetts, United States of America
| | - Anne-Marie Donachie
- Institute of Infection, Immunity and inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
| | - Samia William
- Department of Pharmacology, Theodor Bilharz Research Institute, Warrak El-Hadar, Imbaba, Egypt
| | - Abdel Nasser Sabra
- Department of Pharmacology, Theodor Bilharz Research Institute, Warrak El-Hadar, Imbaba, Egypt
| | - Geert Jan Sterk
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, The Netherlands
| | - Sanaa S. Botros
- Department of Pharmacology, Theodor Bilharz Research Institute, Warrak El-Hadar, Imbaba, Egypt
| | - David G. Brown
- School of Biosciences, University of Kent, United Kingdom
| | - Charles S. Hoffman
- Biology Department, Boston College, Chestnut Hill, Massachusetts, United States of America
| | - Rob Leurs
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, The Netherlands
| | - Harry P. de Koning
- Institute of Infection, Immunity and inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
| |
Collapse
|
12
|
Salado IG, Singh AK, Moreno-Cinos C, Sakaine G, Siderius M, Van der Veken P, Matheeussen A, van der Meer T, Sadek P, Gul S, Maes L, Sterk GJ, Leurs R, Brown D, Augustyns K. Lead Optimization of Phthalazinone Phosphodiesterase Inhibitors as Novel Antitrypanosomal Compounds. J Med Chem 2020; 63:3485-3507. [PMID: 32196340 DOI: 10.1021/acs.jmedchem.9b00985] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Human African trypanosomiasis is causing thousands of deaths every year in the rural areas of Africa. In this manuscript we describe the optimization of a family of phtalazinone derivatives. Phosphodiesterases have emerged as attractive molecular targets for a novel treatment for a variety of neglected parasitic diseases. Compound 1 resulted in being a potent TbrPDEB1 inhibitor with interesting activity against T. brucei in a phenotypic screen. Derivative 1 was studied in an acute in vivo mouse disease model but unfortunately showed no efficacy due to low metabolic stability. We report structural modifications to achieve compounds with an improved metabolic stability while maintaining high potency against TbrPDEB1 and T. brucei. Compound 14 presented a good microsomal stability in mouse and human microsomes and provides a good starting point for future efforts.
Collapse
Affiliation(s)
- Irene G Salado
- Laboratory of Medicinal Chemistry, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - Abhimanyu K Singh
- School of Biosciences, University of Kent, Canterbury, Kent CT2 7NJ, United Kingdom
| | - Carlos Moreno-Cinos
- Laboratory of Medicinal Chemistry, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - Guna Sakaine
- Laboratory of Medicinal Chemistry, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - Marco Siderius
- Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines & Systems, Faculty of Science, Vrije Universiteit Amsterdam, 1081 Amsterdam, The Netherlands
| | - Pieter Van der Veken
- Laboratory of Medicinal Chemistry, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - An Matheeussen
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - Tiffany van der Meer
- Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines & Systems, Faculty of Science, Vrije Universiteit Amsterdam, 1081 Amsterdam, The Netherlands
| | - Payman Sadek
- Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines & Systems, Faculty of Science, Vrije Universiteit Amsterdam, 1081 Amsterdam, The Netherlands
| | - Sheraz Gul
- Fraunhofer-IME SP, Schnackenburgallee 114, Hamburg 22525, Germany
| | - Louis Maes
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - Geert-Jan Sterk
- Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines & Systems, Faculty of Science, Vrije Universiteit Amsterdam, 1081 Amsterdam, The Netherlands
| | - Rob Leurs
- Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines & Systems, Faculty of Science, Vrije Universiteit Amsterdam, 1081 Amsterdam, The Netherlands
| | - David Brown
- School of Biosciences, University of Kent, Canterbury, Kent CT2 7NJ, United Kingdom
| | - Koen Augustyns
- Laboratory of Medicinal Chemistry, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| |
Collapse
|
13
|
Botros SS, El-Lakkany NM, Seif el-Din SH, William S, Sabra AN, Hammam OA, de Koning HP. The phosphodiesterase-4 inhibitor roflumilast impacts Schistosoma mansoni ovipositing in vitro but displays only modest antischistosomal activity in vivo. Exp Parasitol 2020; 208:107793. [DOI: 10.1016/j.exppara.2019.107793] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/14/2019] [Accepted: 11/07/2019] [Indexed: 12/30/2022]
|
14
|
Discovery of novel Schistosoma mansoni PDE4A inhibitors as potential agents against schistosomiasis. Future Med Chem 2019; 11:1703-1720. [PMID: 31370708 DOI: 10.4155/fmc-2018-0592] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Aim: Due to the urgent need for effective drugs to treat schistosomiasis that act through a known molecular mechanism of action, we focused on a target-based approach with the aim to discover inhibitors of a cyclic nucleotide phosphodiesterase from Schistosoma mansoni (SmPDE4A). Materials & methods: To discover new inhibitors of SmPDE4A homology models of the enzyme structure were constructed based on known human and protozoan homologs. The best two models were selected for subsequent virtual screening of our in-house chemical library. Results & conclusion: A total of 25 library compounds were selected for experimental confirmation as SmPDE4A inhibitors and after dose-response experiments, three top hits were identified. The results presented validate the virtual screening approach to identify new inhibitors for clinically relevant phosphodiesterases.
Collapse
|
15
|
Ochoa R, García E, Robledo SM, Cardona G W. Virtual and experimental screening of phenylfuranchalcones as potential anti-Leishmania candidates. J Mol Graph Model 2019; 91:164-171. [PMID: 31252366 DOI: 10.1016/j.jmgm.2019.06.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/30/2019] [Accepted: 06/18/2019] [Indexed: 12/12/2022]
Abstract
Discovery of novel or repurposed chemical treatments for leishmaniasis is a priority given the limited number of therapeutic alternatives available. One way to accelerate the finding is by implementing virtual screening methodologies using structural information, with subsequent experimental validations. Here we tested a library of 48 phenylfuranchalcones as anti-Leishmania agents that can be associated to the potential inhibition of a protein target within the parasite. For that purpose, a list of 43 protein structures from different Leishmania species was prepared to dock the virtual compound library. The protein with the best predicted scores was used as reference to select a subset of previously synthesized compounds for in vitro validation of their cytotoxicity and anti-Leishmania activity. We found a set of active compounds (EC50 < 25 μM) that were compared with the computational results using Spearman correlations. The analysis allowed us to propose the inhibition of a phosphodiesterase enzyme as the potential mechanism of action.
Collapse
Affiliation(s)
- Rodrigo Ochoa
- PECET-Medical Research Institute, School of Medicine, Universidad de Antioquia-UdeA, Calle 70 No. 52-21, A.A 1226, Medellín, Colombia
| | - Elisa García
- Chemistry of Colombian Plants, Institute of Chemistry, Exact and Natural Sciences School, Universidad de Antioquia-UdeA, Calle 70 No. 52-21, A.A 1226, Medellín, Colombia
| | - Sara M Robledo
- PECET-Medical Research Institute, School of Medicine, Universidad de Antioquia-UdeA, Calle 70 No. 52-21, A.A 1226, Medellín, Colombia
| | - Wilson Cardona G
- Chemistry of Colombian Plants, Institute of Chemistry, Exact and Natural Sciences School, Universidad de Antioquia-UdeA, Calle 70 No. 52-21, A.A 1226, Medellín, Colombia.
| |
Collapse
|
16
|
Alkynamide phthalazinones as a new class of TbrPDEB1 inhibitors. Bioorg Med Chem 2019; 27:3998-4012. [PMID: 31327675 DOI: 10.1016/j.bmc.2019.06.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/21/2018] [Accepted: 06/14/2019] [Indexed: 12/26/2022]
Abstract
Several 3',5'-cyclic nucleotide phosphodiesterases (PDEs) have been validated as good drug targets for a large variety of diseases. Trypanosoma brucei PDEB1 (TbrPDEB1) has been designated as a promising drug target for the treatment of human African trypanosomiasis. Recently, the first class of selective nanomolar TbrPDEB1 inhibitors was obtained by targeting the parasite specific P-pocket. However, these biphenyl-substituted tetrahydrophthalazinone-based inhibitors did not show potent cellular activity against Trypanosoma brucei (T. brucei) parasites, leaving room for further optimization. Herein, we report the discovery of a new class of potent TbrPDEB1 inhibitors that display improved activities against T. brucei parasites. Exploring different linkers between the reported tetrahydrophthalazinone core scaffold and the amide tail group resulted in the discovery of alkynamide phthalazinones as new TbrPDEB1 inhibitors, which exhibit submicromolar activities versus T. brucei parasites and no cytotoxicity to human MRC-5 cells. Elucidation of the crystal structure of alkynamide 8b (NPD-048) bound to the catalytic domain of TbrPDEB1 shows a bidentate interaction with the key-residue Gln874 and good directionality towards the P-pocket. Incubation of trypanosomes with alkynamide 8b results in an increase of intracellular cAMP, validating a PDE-mediated effect in vitro and providing a new interesting compound series for further studies towards selective TbrPDEB1 inhibitors with potent phenotypic activity.
Collapse
|
17
|
Pharmacological and molecular dynamics analyses of differences in inhibitor binding to human and nematode PDE4: Implications for management of parasitic nematodes. PLoS One 2019; 14:e0214554. [PMID: 30917179 PMCID: PMC6436744 DOI: 10.1371/journal.pone.0214554] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 03/14/2019] [Indexed: 11/19/2022] Open
Abstract
Novel chemical controls are needed that selectively target human, animal, and plant parasitic nematodes with reduced adverse effects on the host or the environment. We hypothesize that the phosphodiesterase (PDE) enzyme family represents a potential target for development of novel nematicides and anthelmintics. To test this, we identified six PDE families present in the nematode phylum that are orthologous to six of the eleven human PDE families. We characterized the binding interactions of family-selective PDE inhibitors with human and C. elegans PDE4 in conjunction with molecular dynamics (MD) simulations to evaluate differences in binding interactions of these inhibitors within the PDE4 catalytic domain. We observed that roflumilast (human PDE4-selective inhibitor) and zardaverine (selective for human PDE3 and PDE4) were 159- and 77-fold less potent, respectively, in inhibiting C. elegans PDE4. The pan-specific PDE inhibitor isobutyl methyl xanthine (IBMX) had similar affinity for nematode and human PDE4. Of 32 residues within 5 Å of the ligand binding site, five revealed significant differences in non-bonded interaction energies (van der Waals and electrostatic interaction energies) that could account for the differential binding affinities of roflumilast and zardaverine. One site (Phe506 in the human PDE4D3 amino acid sequence corresponding to Tyr253 in C. elegans PDE4) is predicted to alter the binding conformation of roflumilast and zardaverine (but not IBMX) into a less energetically favorable state for the nematode enzyme. The pharmacological differences in sensitivity to PDE4 inhibitors in conjunction with differences in the amino acids comprising the inhibitor binding sites of human and C. elegans PDE4 catalytic domains together support the feasibility of designing the next generation of anthelmintics/nematicides that could selectively bind to nematode PDEs.
Collapse
|
18
|
Veale CGL. Unpacking the Pathogen Box-An Open Source Tool for Fighting Neglected Tropical Disease. ChemMedChem 2019; 14:386-453. [PMID: 30614200 DOI: 10.1002/cmdc.201800755] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Indexed: 12/13/2022]
Abstract
The Pathogen Box is a 400-strong collection of drug-like compounds, selected for their potential against several of the world's most important neglected tropical diseases, including trypanosomiasis, leishmaniasis, cryptosporidiosis, toxoplasmosis, filariasis, schistosomiasis, dengue virus and trichuriasis, in addition to malaria and tuberculosis. This library represents an ensemble of numerous successful drug discovery programmes from around the globe, aimed at providing a powerful resource to stimulate open source drug discovery for diseases threatening the most vulnerable communities in the world. This review seeks to provide an in-depth analysis of the literature pertaining to the compounds in the Pathogen Box, including structure-activity relationship highlights, mechanisms of action, related compounds with reported activity against different diseases, and, where appropriate, discussion on the known and putative targets of compounds, thereby providing context and increasing the accessibility of the Pathogen Box to the drug discovery community.
Collapse
Affiliation(s)
- Clinton G L Veale
- School of Chemistry and Physics, Pietermaritzburg Campus, University of KwaZulu-Natal, Private Bag X01, Scottsville, 3209, South Africa
| |
Collapse
|
19
|
Pobsuk N, Paracha TU, Chaichamnong N, Salaloy N, Suphakun P, Hannongbua S, Choowongkomon K, Pekthong D, Chootip K, Ingkaninan K, Gleeson MP. Design, synthesis and evaluation of N2,N4-diaminoquinazoline based inhibitors of phosphodiesterase type 5. Bioorg Med Chem Lett 2019; 29:267-270. [DOI: 10.1016/j.bmcl.2018.11.043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/19/2018] [Accepted: 11/20/2018] [Indexed: 12/23/2022]
|
20
|
Bioinformatics Analysis and Functional Prediction of Transmembrane Proteins in Entamoeba histolytica. Genes (Basel) 2018; 9:genes9100499. [PMID: 30332795 PMCID: PMC6209943 DOI: 10.3390/genes9100499] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/09/2018] [Accepted: 09/12/2018] [Indexed: 12/18/2022] Open
Abstract
Entamoeba histolytica is an invasive, pathogenic parasite causing amoebiasis. Given that proteins involved in transmembrane (TM) transport are crucial for the adherence, invasion, and nutrition of the parasite, we conducted a genome-wide bioinformatics analysis of encoding proteins to functionally classify and characterize all the TM proteins in E. histolytica. In the present study, 692 TM proteins have been identified, of which 546 are TM transporters. For the first time, we report a set of 141 uncharacterized proteins predicted as TM transporters. The percentage of TM proteins was found to be lower in comparison to the free-living eukaryotes, due to the extracellular nature and functional diversification of the TM proteins. The number of multi-pass proteins is larger than the single-pass proteins; though both have their own significance in parasitism, multi-pass proteins are more extensively required as these are involved in acquiring nutrition and for ion transport, while single-pass proteins are only required at the time of inciting infection. Overall, this intestinal parasite implements multiple mechanisms for establishing infection, obtaining nutrition, and adapting itself to the new host environment. A classification of the repertoire of TM transporters in the present study augments several hints on potential methods of targeting the parasite for therapeutic benefits.
Collapse
|
21
|
Sebastián-Pérez V, Hendrickx S, Munday JC, Kalejaiye T, Martínez A, Campillo NE, de Koning H, Caljon G, Maes L, Gil C. Cyclic Nucleotide-Specific Phosphodiesterases as Potential Drug Targets for Anti-Leishmania Therapy. Antimicrob Agents Chemother 2018; 62:e00603-18. [PMID: 30104270 PMCID: PMC6153811 DOI: 10.1128/aac.00603-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 08/05/2018] [Indexed: 01/22/2023] Open
Abstract
The available treatments for leishmaniasis are less than optimal due to inadequate efficacy, toxic side effects, and the emergence of resistant strains, clearly endorsing the urgent need for discovery and development of novel drug candidates. Ideally, these should act via an alternative mechanism of action to avoid cross-resistance with the current drugs. As cyclic nucleotide-specific phosphodiesterases (PDEs) of Leishmania major have been postulated as putative drug targets, a series of potential inhibitors of Leishmania PDEs were explored. Several displayed potent and selective in vitro activity against L. infantum intracellular amastigotes. One imidazole derivative, compound 35, was shown to reduce the parasite loads in vivo and to increase the cellular cyclic AMP (cAMP) level at in a dose-dependent manner at just 2× and 5× the 50% inhibitory concentration (IC50), indicating a correlation between antileishmanial activity and increased cellular cAMP levels. Docking studies and molecular dynamics simulations pointed to imidazole 35 exerting its activity through PDE inhibition. This study establishes for the first time that inhibition of cAMP PDEs can potentially be exploited for new antileishmanial chemotherapy.
Collapse
Affiliation(s)
| | - Sarah Hendrickx
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Jane C Munday
- Institute of Infection, Inflammation and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Titilola Kalejaiye
- Institute of Infection, Inflammation and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Ana Martínez
- Centro de Investigaciones Biológicas (CIB, CSIC), Madrid, Spain
| | | | - Harry de Koning
- Institute of Infection, Inflammation and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Guy Caljon
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Louis Maes
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Carmen Gil
- Centro de Investigaciones Biológicas (CIB, CSIC), Madrid, Spain
| |
Collapse
|
22
|
Blaazer AR, Singh AK, de Heuvel E, Edink E, Orrling KM, Veerman JJN, van den Bergh T, Jansen C, Balasubramaniam E, Mooij WJ, Custers H, Sijm M, Tagoe DNA, Kalejaiye TD, Munday JC, Tenor H, Matheeussen A, Wijtmans M, Siderius M, de Graaf C, Maes L, de Koning HP, Bailey DS, Sterk GJ, de Esch IJP, Brown DG, Leurs R. Targeting a Subpocket in Trypanosoma brucei Phosphodiesterase B1 (TbrPDEB1) Enables the Structure-Based Discovery of Selective Inhibitors with Trypanocidal Activity. J Med Chem 2018; 61:3870-3888. [PMID: 29672041 PMCID: PMC5949723 DOI: 10.1021/acs.jmedchem.7b01670] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
![]()
Several trypanosomatid
cyclic nucleotide phosphodiesterases (PDEs)
possess a unique, parasite-specific cavity near the ligand-binding
region that is referred to as the P-pocket. One of these enzymes, Trypanosoma brucei PDE B1 (TbrPDEB1), is considered a drug
target for the treatment of African sleeping sickness. Here, we elucidate
the molecular determinants of inhibitor binding and reveal that the
P-pocket is amenable to directed design. By iterative cycles of design,
synthesis, and pharmacological evaluation and by elucidating the structures
of inhibitor-bound TbrPDEB1, hPDE4B, and hPDE4D complexes, we have
developed 4a,5,8,8a-tetrahydrophthalazinones as the first selective
TbrPDEB1 inhibitor series. Two of these, 8 (NPD-008)
and 9 (NPD-039), were potent (Ki = 100 nM) TbrPDEB1 inhibitors with antitrypanosomal effects
(IC50 = 5.5 and 6.7 μM, respectively). Treatment
of parasites with 8 caused an increase in intracellular
cyclic adenosine monophosphate (cAMP) levels and severe disruption
of T. brucei cellular organization, chemically validating
trypanosomal PDEs as therapeutic targets in trypanosomiasis.
Collapse
Affiliation(s)
- Antoni R Blaazer
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems , Vrije Universiteit Amsterdam , 1081 HZ Amsterdam , The Netherlands
| | - Abhimanyu K Singh
- School of Biosciences , University of Kent , Canterbury CT2 7NJ , U.K
| | - Erik de Heuvel
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems , Vrije Universiteit Amsterdam , 1081 HZ Amsterdam , The Netherlands
| | - Ewald Edink
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems , Vrije Universiteit Amsterdam , 1081 HZ Amsterdam , The Netherlands
| | - Kristina M Orrling
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems , Vrije Universiteit Amsterdam , 1081 HZ Amsterdam , The Netherlands
| | | | | | - Chimed Jansen
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems , Vrije Universiteit Amsterdam , 1081 HZ Amsterdam , The Netherlands
| | | | - Wouter J Mooij
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems , Vrije Universiteit Amsterdam , 1081 HZ Amsterdam , The Netherlands
| | - Hans Custers
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems , Vrije Universiteit Amsterdam , 1081 HZ Amsterdam , The Netherlands
| | - Maarten Sijm
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems , Vrije Universiteit Amsterdam , 1081 HZ Amsterdam , The Netherlands
| | - Daniel N A Tagoe
- Institute of Infection, Immunity and Inflammation , University of Glasgow , Glasgow G12 8TA , U.K
| | - Titilola D Kalejaiye
- Institute of Infection, Immunity and Inflammation , University of Glasgow , Glasgow G12 8TA , U.K
| | - Jane C Munday
- Institute of Infection, Immunity and Inflammation , University of Glasgow , Glasgow G12 8TA , U.K
| | | | - An Matheeussen
- Laboratory for Microbiology, Parasitology and Hygiene , University of Antwerp , 2610 Wilrijk , Belgium
| | - Maikel Wijtmans
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems , Vrije Universiteit Amsterdam , 1081 HZ Amsterdam , The Netherlands
| | - Marco Siderius
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems , Vrije Universiteit Amsterdam , 1081 HZ Amsterdam , The Netherlands
| | - Chris de Graaf
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems , Vrije Universiteit Amsterdam , 1081 HZ Amsterdam , The Netherlands
| | - Louis Maes
- Laboratory for Microbiology, Parasitology and Hygiene , University of Antwerp , 2610 Wilrijk , Belgium
| | - Harry P de Koning
- Institute of Infection, Immunity and Inflammation , University of Glasgow , Glasgow G12 8TA , U.K
| | | | - Geert Jan Sterk
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems , Vrije Universiteit Amsterdam , 1081 HZ Amsterdam , The Netherlands
| | - Iwan J P de Esch
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems , Vrije Universiteit Amsterdam , 1081 HZ Amsterdam , The Netherlands
| | - David G Brown
- School of Biosciences , University of Kent , Canterbury CT2 7NJ , U.K
| | - Rob Leurs
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems , Vrije Universiteit Amsterdam , 1081 HZ Amsterdam , The Netherlands
| |
Collapse
|
23
|
Svensson F, Bender A, Bailey D. Fragment-Based Drug Discovery of Phosphodiesterase Inhibitors. J Med Chem 2017; 61:1415-1424. [DOI: 10.1021/acs.jmedchem.7b00404] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Fredrik Svensson
- IOTA Pharmaceuticals, St Johns
Innovation Centre, Cowley Road, Cambridge CB4 0WS, U.K
- Centre
for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
| | - Andreas Bender
- Centre
for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
| | - David Bailey
- IOTA Pharmaceuticals, St Johns
Innovation Centre, Cowley Road, Cambridge CB4 0WS, U.K
| |
Collapse
|
24
|
Long T, Rojo-Arreola L, Shi D, El-Sakkary N, Jarnagin K, Rock F, Meewan M, Rascón AA, Lin L, Cunningham KA, Lemieux GA, Podust L, Abagyan R, Ashrafi K, McKerrow JH, Caffrey CR. Phenotypic, chemical and functional characterization of cyclic nucleotide phosphodiesterase 4 (PDE4) as a potential anthelmintic drug target. PLoS Negl Trop Dis 2017; 11:e0005680. [PMID: 28704396 PMCID: PMC5526615 DOI: 10.1371/journal.pntd.0005680] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 07/25/2017] [Accepted: 06/04/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Reliance on just one drug to treat the prevalent tropical disease, schistosomiasis, spurs the search for new drugs and drug targets. Inhibitors of human cyclic nucleotide phosphodiesterases (huPDEs), including PDE4, are under development as novel drugs to treat a range of chronic indications including asthma, chronic obstructive pulmonary disease and Alzheimer's disease. One class of huPDE4 inhibitors that has yielded marketed drugs is the benzoxaboroles (Anacor Pharmaceuticals). METHODOLOGY/PRINCIPAL FINDINGS A phenotypic screen involving Schistosoma mansoni and 1,085 benzoxaboroles identified a subset of huPDE4 inhibitors that induced parasite hypermotility and degeneration. To uncover the putative schistosome PDE4 target, we characterized four PDE4 sequences (SmPDE4A-D) in the parasite's genome and transcriptome, and cloned and recombinantly expressed the catalytic domain of SmPDE4A. Among a set of benzoxaboroles and catechol inhibitors that differentially inhibit huPDE4, a relationship between the inhibition of SmPDE4A, and parasite hypermotility and degeneration, was measured. To validate SmPDE4A as the benzoxaborole molecular target, we first generated Caenorhabditis elegans lines that express a cDNA for smpde4a on a pde4(ce268) mutant (hypermotile) background: the smpde4a transgene restored mutant worm motility to that of the wild type. We then showed that benzoxaborole inhibitors of SmPDE4A that induce hypermotility in the schistosome also elicit a hypermotile response in the C. elegans lines that express the smpde4a transgene, thereby confirming SmPDE4A as the relevant target. CONCLUSIONS/SIGNIFICANCE The orthogonal chemical, biological and genetic strategies employed identify SmPDE4A's contribution to parasite motility and degeneration, and its potential as a drug target. Transgenic C. elegans is highlighted as a potential screening tool to optimize small molecule chemistries to flatworm molecular drug targets.
Collapse
Affiliation(s)
- Thavy Long
- Center for Discovery and Innovation in Parasitic Diseases, University of California San Francisco, San Francisco, California, United States of America
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
| | - Liliana Rojo-Arreola
- Center for Discovery and Innovation in Parasitic Diseases, University of California San Francisco, San Francisco, California, United States of America
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
| | - Da Shi
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Nelly El-Sakkary
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Kurt Jarnagin
- Anacor Pharmaceuticals, Inc., Palo Alto, California, United States of America
| | - Fernando Rock
- Anacor Pharmaceuticals, Inc., Palo Alto, California, United States of America
| | - Maliwan Meewan
- Anacor Pharmaceuticals, Inc., Palo Alto, California, United States of America
| | - Alberto A. Rascón
- Center for Discovery and Innovation in Parasitic Diseases, University of California San Francisco, San Francisco, California, United States of America
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
| | - Lin Lin
- Department of Physiology, University of California San Francisco, San Francisco, California, United States of America
| | - Katherine A. Cunningham
- Department of Physiology, University of California San Francisco, San Francisco, California, United States of America
| | - George A. Lemieux
- Department of Physiology, University of California San Francisco, San Francisco, California, United States of America
| | - Larissa Podust
- Center for Discovery and Innovation in Parasitic Diseases, University of California San Francisco, San Francisco, California, United States of America
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
| | - Ruben Abagyan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Kaveh Ashrafi
- Department of Physiology, University of California San Francisco, San Francisco, California, United States of America
| | - James H. McKerrow
- Center for Discovery and Innovation in Parasitic Diseases, University of California San Francisco, San Francisco, California, United States of America
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
| | - Conor R. Caffrey
- Center for Discovery and Innovation in Parasitic Diseases, University of California San Francisco, San Francisco, California, United States of America
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
25
|
Soni R, Sharma D, Rai P, Sharma B, Bhatt TK. Signaling Strategies of Malaria Parasite for Its Survival, Proliferation, and Infection during Erythrocytic Stage. Front Immunol 2017; 8:349. [PMID: 28400771 PMCID: PMC5368685 DOI: 10.3389/fimmu.2017.00349] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 03/10/2017] [Indexed: 12/22/2022] Open
Abstract
Irrespective of various efforts, malaria persist the most debilitating effect in terms of morbidity and mortality. Moreover, the existing drugs are also vulnerable to the emergence of drug resistance. To explore the potential targets for designing the most effective antimalarial therapies, it is required to focus on the facts of biochemical mechanism underlying the process of parasite survival and disease pathogenesis. This review is intended to bring out the existing knowledge about the functions and components of the major signaling pathways such as kinase signaling, calcium signaling, and cyclic nucleotide-based signaling, serving the various aspects of the parasitic asexual stage and highlighted the Toll-like receptors, glycosylphosphatidylinositol-mediated signaling, and molecular events in cytoadhesion, which elicit the host immune response. This discussion will facilitate a look over essential components for parasite survival and disease progression to be implemented in discovery of novel antimalarial drugs and vaccines.
Collapse
Affiliation(s)
- Rani Soni
- Department of Biotechnology, School of Life sciences, Central University of Rajasthan , Ajmer , India
| | - Drista Sharma
- Department of Biotechnology, School of Life sciences, Central University of Rajasthan , Ajmer , India
| | - Praveen Rai
- Department of Biotechnology, School of Life sciences, Central University of Rajasthan , Ajmer , India
| | - Bhaskar Sharma
- Department of Biotechnology, School of Life sciences, Central University of Rajasthan , Ajmer , India
| | - Tarun K Bhatt
- Department of Biotechnology, School of Life sciences, Central University of Rajasthan , Ajmer , India
| |
Collapse
|
26
|
Abstract
INTRODUCTION Despite the fact that diseases caused by protozoan parasites represent serious challenges for public health, animal production and welfare, only a limited panel of drugs has been marketed for clinical applications. AREAS COVERED Herein, the authors investigate two strategies, namely whole organism screening and target-based drug design. The present pharmacopoeia has resulted from whole organism screening, and the mode of action and targets of selected drugs are discussed. However, the more recent extensive genome sequencing efforts and the development of dry and wet lab genomics and proteomics that allow high-throughput screening of interactions between micromolecules and recombinant proteins has resulted in target-based drug design as the predominant focus in anti-parasitic drug development. Selected examples of target-based drug design studies are presented, and calcium-dependent protein kinases, important drug targets in apicomplexan parasites, are discussed in more detail. EXPERT OPINION Despite the enormous efforts in target-based drug development, this approach has not yet generated market-ready antiprotozoal drugs. However, whole-organism screening approaches, comprising of both in vitro and in vivo investigations, should not be disregarded. The repurposing of already approved and marketed drugs could be a suitable strategy to avoid fastidious approval procedures, especially in the case of neglected or veterinary parasitoses.
Collapse
Affiliation(s)
- Joachim Müller
- a Institute of Parasitology, Vetsuisse Faculty , University of Bern , Bern , Switzerland
| | - Andrew Hemphill
- a Institute of Parasitology, Vetsuisse Faculty , University of Bern , Bern , Switzerland
| |
Collapse
|
27
|
Qvit N, Schechtman D, Pena DA, Berti DA, Soares CO, Miao Q, Liang LA, Baron LA, Teh-Poot C, Martínez-Vega P, Ramirez-Sierra MJ, Churchill E, Cunningham AD, Malkovskiy AV, Federspiel NA, Gozzo FC, Torrecilhas AC, Manso Alves MJ, Jardim A, Momar N, Dumonteil E, Mochly-Rosen D. Scaffold proteins LACK and TRACK as potential drug targets in kinetoplastid parasites: Development of inhibitors. Int J Parasitol Drugs Drug Resist 2016; 6:74-84. [PMID: 27054066 PMCID: PMC4805777 DOI: 10.1016/j.ijpddr.2016.02.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 02/04/2016] [Accepted: 02/08/2016] [Indexed: 01/15/2023]
Abstract
Parasitic diseases cause ∼ 500,000 deaths annually and remain a major challenge for therapeutic development. Using a rational design based approach, we developed peptide inhibitors with anti-parasitic activity that were derived from the sequences of parasite scaffold proteins LACK (Leishmania's receptor for activated C-kinase) and TRACK (Trypanosoma receptor for activated C-kinase). We hypothesized that sequences in LACK and TRACK that are conserved in the parasites, but not in the mammalian ortholog, RACK (Receptor for activated C-kinase), may be interaction sites for signaling proteins that are critical for the parasites' viability. One of these peptides exhibited leishmanicidal and trypanocidal activity in culture. Moreover, in infected mice, this peptide was also effective in reducing parasitemia and increasing survival without toxic effects. The identified peptide is a promising new anti-parasitic drug lead, as its unique features may limit toxicity and drug-resistance, thus overcoming central limitations of most anti-parasitic drugs.
Collapse
Affiliation(s)
- Nir Qvit
- Department of Chemical and Systems Biology, Stanford University, School of Medicine, Stanford, CA 94305, USA.
| | - Deborah Schechtman
- Department of Chemical and Systems Biology, Stanford University, School of Medicine, Stanford, CA 94305, USA; Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, SP, Brazil
| | | | | | | | - Qianqian Miao
- National Reference Centre for Parasitology, Research Institute of the McGill University, Montreal, Canada
| | - Liying Annie Liang
- National Reference Centre for Parasitology, Research Institute of the McGill University, Montreal, Canada
| | - Lauren A Baron
- Laboratorio de Parasitología, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, Yucatán, Mexico
| | - Christian Teh-Poot
- Laboratorio de Parasitología, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, Yucatán, Mexico
| | - Pedro Martínez-Vega
- Laboratorio de Parasitología, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, Yucatán, Mexico
| | - Maria Jesus Ramirez-Sierra
- Laboratorio de Parasitología, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, Yucatán, Mexico
| | - Eric Churchill
- Department of Chemical and Systems Biology, Stanford University, School of Medicine, Stanford, CA 94305, USA
| | - Anna D Cunningham
- Department of Chemical and Systems Biology, Stanford University, School of Medicine, Stanford, CA 94305, USA
| | - Andrey V Malkovskiy
- Biomaterials and Advanced Drug Delivery Laboratory, Stanford University, Stanford, CA 94305, USA
| | - Nancy A Federspiel
- Department of Chemical and Systems Biology, Stanford University, School of Medicine, Stanford, CA 94305, USA
| | - Fabio Cesar Gozzo
- Institute of Chemistry, University of Campinas, Campinas, SP, Brazil
| | | | | | - Armando Jardim
- Institute of Parasitology and Centre for Host-Parasite Interactions, McGill University, Québec, Canada
| | - Ndao Momar
- National Reference Centre for Parasitology, Research Institute of the McGill University, Montreal, Canada
| | - Eric Dumonteil
- Laboratorio de Parasitología, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, Yucatán, Mexico
| | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology, Stanford University, School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
28
|
Jansen C, Kooistra AJ, Kanev GK, Leurs R, de Esch IJP, de Graaf C. PDEStrIAn: A Phosphodiesterase Structure and Ligand Interaction Annotated Database As a Tool for Structure-Based Drug Design. J Med Chem 2016; 59:7029-65. [DOI: 10.1021/acs.jmedchem.5b01813] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Chimed Jansen
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute
of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Albert J. Kooistra
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute
of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Georgi K. Kanev
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute
of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Rob Leurs
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute
of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Iwan J. P. de Esch
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute
of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Chris de Graaf
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute
of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
29
|
Veerman J, van den Bergh T, Orrling KM, Jansen C, Cos P, Maes L, Chatelain E, Ioset JR, Edink EE, Tenor H, Seebeck T, de Esch I, Leurs R, Sterk GJ. Synthesis and evaluation of analogs of the phenylpyridazinone NPD-001 as potent trypanosomal TbrPDEB1 phosphodiesterase inhibitors and in vitro trypanocidals. Bioorg Med Chem 2016; 24:1573-81. [PMID: 26935942 DOI: 10.1016/j.bmc.2016.02.032] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/21/2016] [Accepted: 02/24/2016] [Indexed: 12/16/2022]
Abstract
Trypanosomal phosphodiesterases B1 and B2 (TbrPDEB1 and TbrPDEB2) play an important role in the life cycle of Trypanosoma brucei, the causative parasite of human African trypanosomiasis (HAT), also known as African sleeping sickness. Knock down of both enzymes leads to cell cycle arrest and is lethal to the parasite. Recently, we reported the phenylpyridazinone, NPD-001, with low nanomolar IC50 values on both TbrPDEB1 (IC50: 4nM) and TbrPDEB2 (IC50: 3nM) (J. Infect. Dis.2012, 206, 229). In this study, we now report on the first structure activity relationships of a series of phenylpyridazinone analogs as TbrPDEB1 inhibitors. A selection of compounds was also shown to be anti-parasitic. Importantly, a good correlation between TbrPDEB1 IC50 and EC50 against the whole parasite was observed. Preliminary analysis of the SAR of selected compounds on TbrPDEB1 and human PDEs shows large differences which shows the potential for obtaining parasite selective PDE inhibitors. The results of these studies support the pharmacological validation of the Trypanosome PDEB family as novel therapeutic approach for HAT and provide as well valuable information for the design of potent TbrPDEB1 inhibitors that could be used for the treatment of this disease.
Collapse
Affiliation(s)
- Johan Veerman
- Mercachem, PO Box 6747, 6503 GE Nijmegen, The Netherlands
| | | | - Kristina M Orrling
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines & Systems (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - Chimed Jansen
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines & Systems (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - Paul Cos
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Groenenborgerlaan 171, 2020 Wilrijk, Belgium
| | - Louis Maes
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Groenenborgerlaan 171, 2020 Wilrijk, Belgium
| | - Eric Chatelain
- DNDi (Drugs for Neglected Diseases initiative), 15 Chemin Louis Dunant, 1202 Geneva, Switzerland
| | - Jean-Robert Ioset
- DNDi (Drugs for Neglected Diseases initiative), 15 Chemin Louis Dunant, 1202 Geneva, Switzerland
| | - Ewald E Edink
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines & Systems (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - Hermann Tenor
- Takeda, Takeda Pharmaceuticals International GmbH, Thurgauerstrasse 130, 8152 Glattpark-Opfikon, Zurich, Switzerland
| | - Thomas Seebeck
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Iwan de Esch
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines & Systems (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - Rob Leurs
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines & Systems (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - Geert Jan Sterk
- Mercachem, PO Box 6747, 6503 GE Nijmegen, The Netherlands; Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines & Systems (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands.
| |
Collapse
|
30
|
Tagoe DNA, Kalejaiye TD, de Koning HP. The ever unfolding story of cAMP signaling in trypanosomatids: vive la difference! Front Pharmacol 2015; 6:185. [PMID: 26441645 PMCID: PMC4561360 DOI: 10.3389/fphar.2015.00185] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 08/17/2015] [Indexed: 12/25/2022] Open
Abstract
Kinetoplastids are unicellular, eukaryotic, flagellated protozoans containing the eponymous kinetoplast. Within this order, the family of trypanosomatids are responsible for some of the most serious human diseases, including Chagas disease (Trypanosoma cruzi), sleeping sickness (Trypanosoma brucei spp.), and leishmaniasis (Leishmania spp). Although cAMP is produced during the life cycle stages of these parasites, its signaling pathways are very different from those of mammals. The absence of G-protein-coupled receptors, the presence of structurally different adenylyl cyclases, the paucity of known cAMP effector proteins and the stringent need for regulation of cAMP in the small kinetoplastid cells all suggest a significantly different biochemical pathway and likely cell biology. However, each of the main kinetoplastid parasites express four class 1-type cyclic nucleotide-specific phosphodiesterases (PDEA-D), which have highly similar catalytic domains to that of human PDEs. To date, only TbrPDEB, expressed as two slightly different isoforms TbrPDEB1 and B2, has been found to be essential when ablated. Although the genomes contain reasonably well conserved genes for catalytic and regulatory domains of protein kinase A, these have been shown to have varied structural and functional roles in the different species. Recent discovery of a role of cAMP/AMP metabolism in a quorum-sensing signaling pathway in T. brucei, and the identification of downstream cAMP Response Proteins (CARPs) whose expression levels correlate with sensitivity to PDE inhibitors, suggests a complex signaling cascade. The interplay between the roles of these novel CARPs and the quorum-sensing signaling pathway on cell division and differentiation makes for intriguing cell biology and a new paradigm in cAMP signal transduction, as well as potential targets for trypanosomatid-specific cAMP pathway-based therapeutics.
Collapse
Affiliation(s)
- Daniel N A Tagoe
- Wellcome Trust Centre for Molecular Parasitology, University of Glasgow , Glasgow, UK ; Institute of Infection, Inflammation and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow , Glasgow, UK ; Department of Laboratory Technology, Division of Medical Laboratory Technology, University of Cape Coast , Cape Coast, Ghana
| | - Titilola D Kalejaiye
- Institute of Infection, Inflammation and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow , Glasgow, UK
| | - Harry P de Koning
- Institute of Infection, Inflammation and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow , Glasgow, UK
| |
Collapse
|
31
|
Kaiser M, Mäser P, Tadoori LP, Ioset JR, Brun R. Antiprotozoal Activity Profiling of Approved Drugs: A Starting Point toward Drug Repositioning. PLoS One 2015; 10:e0135556. [PMID: 26270335 PMCID: PMC4535766 DOI: 10.1371/journal.pone.0135556] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 07/23/2015] [Indexed: 11/23/2022] Open
Abstract
Neglected tropical diseases cause significant morbidity and mortality and are a source of poverty in endemic countries. Only a few drugs are available to treat diseases such as leishmaniasis, Chagas’ disease, human African trypanosomiasis and malaria. Since drug development is lengthy and expensive, a drug repurposing strategy offers an attractive fast-track approach to speed up the process. A set of 100 registered drugs with drug repositioning potential for neglected diseases was assembled and tested in vitro against four protozoan parasites associated with the aforementioned diseases. Several drugs and drug classes showed in vitro activity in those screening assays. The results are critically reviewed and discussed in the perspective of a follow-up drug repositioning strategy where R&D has to be addressed with limited resources.
Collapse
Affiliation(s)
- Marcel Kaiser
- Parasite Chemotherapy, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
- * E-mail:
| | - Pascal Mäser
- Parasite Chemotherapy, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | | | | | - Reto Brun
- Parasite Chemotherapy, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| |
Collapse
|
32
|
Ramdani G, Naissant B, Thompson E, Breil F, Lorthiois A, Dupuy F, Cummings R, Duffier Y, Corbett Y, Mercereau-Puijalon O, Vernick K, Taramelli D, Baker DA, Langsley G, Lavazec C. cAMP-Signalling Regulates Gametocyte-Infected Erythrocyte Deformability Required for Malaria Parasite Transmission. PLoS Pathog 2015; 11:e1004815. [PMID: 25951195 PMCID: PMC4423841 DOI: 10.1371/journal.ppat.1004815] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 03/13/2015] [Indexed: 12/31/2022] Open
Abstract
Blocking Plasmodium falciparum transmission to mosquitoes has been designated a strategic objective in the global agenda of malaria elimination. Transmission is ensured by gametocyte-infected erythrocytes (GIE) that sequester in the bone marrow and at maturation are released into peripheral blood from where they are taken up during a mosquito blood meal. Release into the blood circulation is accompanied by an increase in GIE deformability that allows them to pass through the spleen. Here, we used a microsphere matrix to mimic splenic filtration and investigated the role of cAMP-signalling in regulating GIE deformability. We demonstrated that mature GIE deformability is dependent on reduced cAMP-signalling and on increased phosphodiesterase expression in stage V gametocytes, and that parasite cAMP-dependent kinase activity contributes to the stiffness of immature gametocytes. Importantly, pharmacological agents that raise cAMP levels in transmissible stage V gametocytes render them less deformable and hence less likely to circulate through the spleen. Therefore, phosphodiesterase inhibitors that raise cAMP levels in P. falciparum infected erythrocytes, such as sildenafil, represent new candidate drugs to block transmission of malaria parasites. Malaria transmission is ensured by deformable mature gametocyte-infected erythrocytes being taken up when a mosquito bites. Non-deformable immature gametocyte stages are sequestered in the bone marrow, as their lack of deformability would lead to their splenic clearance. In the present study, we apply nano-filtration technology to mimic splenic retention and demonstrate that deformability of transmissible mature stage V gametocytes is regulated by parasite cyclic AMP-dependent kinase signalling. Importantly, when we used drugs to raise cAMP levels we render transmissible mature gametocytes as stiff as non-transmissible gametocytes. In contrast, when we inhibit the cAMP-dependent kinase we render immature gametocytes more deformable. Thus, by two different approaches we confirm that the drop in cAMP levels in mature gametocytes leads to an increase in their deformability and hence more likely to circulate through the spleen. Our molecular observations have the potential to be translated into therapies for blocking malaria transmission by demonstrating that raising cAMP levels with sildenafil also known as “Viagra” renders mature gametocytes rigid. These findings provide the proof of principle that deformability of circulating gametocytes is targetable by pharmacological agents and as such, it provides a novel approach to prevent the spread of parasites. PDE inhibitors therefore represent novel drug leads potentially capable of blocking transmission and improving the worldwide fight to eliminate malaria from the human population.
Collapse
Affiliation(s)
- Ghania Ramdani
- Laboratoire de Biologie Cellulaire Comparative des Apicomplexes, Faculté de Médicine, Université Paris Descartes—Sorbonne Paris Cité, Paris, France
- Inserm U1016, CNRS UMR8104, Institut Cochin, Paris, France
| | - Bernina Naissant
- Inserm U1016, CNRS UMR8104, Institut Cochin, Paris, France
- Laboratoire de Biologie de la Transmission de Plasmodium, Faculté de Médicine, Université Paris Descartes—Sorbonne Paris Cité, Paris, France
| | - Eloise Thompson
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Florence Breil
- Institut Pasteur, Unité de Génétique et Génomique des Insectes Vecteurs, CNRS URA 3012, Paris, France
| | - Audrey Lorthiois
- Inserm U1016, CNRS UMR8104, Institut Cochin, Paris, France
- Laboratoire de Biologie de la Transmission de Plasmodium, Faculté de Médicine, Université Paris Descartes—Sorbonne Paris Cité, Paris, France
- Institut Pasteur, Unité de Génétique et Génomique des Insectes Vecteurs, CNRS URA 3012, Paris, France
| | - Florian Dupuy
- Inserm U1016, CNRS UMR8104, Institut Cochin, Paris, France
- Laboratoire de Biologie de la Transmission de Plasmodium, Faculté de Médicine, Université Paris Descartes—Sorbonne Paris Cité, Paris, France
| | - Ross Cummings
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Yoann Duffier
- Inserm U1016, CNRS UMR8104, Institut Cochin, Paris, France
- Laboratoire de Biologie de la Transmission de Plasmodium, Faculté de Médicine, Université Paris Descartes—Sorbonne Paris Cité, Paris, France
| | - Yolanda Corbett
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università di Milano, Milano, Italy
| | | | - Kenneth Vernick
- Institut Pasteur, Unité de Génétique et Génomique des Insectes Vecteurs, CNRS URA 3012, Paris, France
| | - Donatella Taramelli
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università di Milano, Milano, Italy
| | - David A. Baker
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Gordon Langsley
- Laboratoire de Biologie Cellulaire Comparative des Apicomplexes, Faculté de Médicine, Université Paris Descartes—Sorbonne Paris Cité, Paris, France
- Inserm U1016, CNRS UMR8104, Institut Cochin, Paris, France
- * E-mail: (GL); (CL)
| | - Catherine Lavazec
- Inserm U1016, CNRS UMR8104, Institut Cochin, Paris, France
- Laboratoire de Biologie de la Transmission de Plasmodium, Faculté de Médicine, Université Paris Descartes—Sorbonne Paris Cité, Paris, France
- Institut Pasteur, Unité de Génétique et Génomique des Insectes Vecteurs, CNRS URA 3012, Paris, France
- * E-mail: (GL); (CL)
| |
Collapse
|
33
|
Abstract
UNLABELLED The protozoan parasite Trypanosoma brucei engages in surface-induced social behavior, termed social motility, characterized by single cells assembling into multicellular groups that coordinate their movements in response to extracellular signals. Social motility requires sensing and responding to extracellular signals, but the underlying mechanisms are unknown. Here we report that T. brucei social motility depends on cyclic AMP (cAMP) signaling systems in the parasite's flagellum (synonymous with cilium). Pharmacological inhibition of cAMP-specific phosphodiesterase (PDE) completely blocks social motility without impacting the viability or motility of individual cells. Using a fluorescence resonance energy transfer (FRET)-based sensor to monitor cAMP dynamics in live cells, we demonstrate that this block in social motility correlates with an increase in intracellular cAMP levels. RNA interference (RNAi) knockdown of the flagellar PDEB1 phenocopies pharmacological PDE inhibition, demonstrating that PDEB1 is required for social motility. Using parasites expressing distinct fluorescent proteins to monitor individuals in a genetically heterogeneous community, we found that the social motility defect of PDEB1 knockdowns is complemented by wild-type parasites in trans. Therefore, PDEB1 knockdown cells are competent for social motility but appear to lack a necessary factor that can be provided by wild-type cells. The combined data demonstrate that the role of cyclic nucleotides in regulating microbial social behavior extends to African trypanosomes and provide an example of transcomplementation in parasitic protozoa. IMPORTANCE In bacteria, studies of cell-cell communication and social behavior have profoundly influenced our understanding of microbial physiology, signaling, and pathogenesis. In contrast, mechanisms underlying social behavior in protozoan parasites are mostly unknown. Here we show that social behavior in the protozoan parasite Trypanosoma brucei is governed by cyclic-AMP signaling systems in the flagellum, with intriguing parallels to signaling systems that control bacterial social behavior. We also generated a T. brucei social behavior mutant and found that the mutant phenotype is complemented by wild-type cells grown in the same culture. Our findings open new avenues for dissecting social behavior and signaling in protozoan parasites and illustrate the capacity of these organisms to influence each other's behavior in mixed communities.
Collapse
|
34
|
Peña I, Pilar Manzano M, Cantizani J, Kessler A, Alonso-Padilla J, Bardera AI, Alvarez E, Colmenarejo G, Cotillo I, Roquero I, de Dios-Anton F, Barroso V, Rodriguez A, Gray DW, Navarro M, Kumar V, Sherstnev A, Drewry DH, Brown JR, Fiandor JM, Julio Martin J. New compound sets identified from high throughput phenotypic screening against three kinetoplastid parasites: an open resource. Sci Rep 2015; 5:8771. [PMID: 25740547 PMCID: PMC4350103 DOI: 10.1038/srep08771] [Citation(s) in RCA: 181] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 01/28/2015] [Indexed: 12/11/2022] Open
Abstract
Using whole-cell phenotypic assays, the GlaxoSmithKline high-throughput screening (HTS) diversity set of 1.8 million compounds was screened against the three kinetoplastids most relevant to human disease, i.e. Leishmania donovani, Trypanosoma cruzi and Trypanosoma brucei. Secondary confirmatory and orthogonal intracellular anti-parasiticidal assays were conducted, and the potential for non-specific cytotoxicity determined. Hit compounds were chemically clustered and triaged for desirable physicochemical properties. The hypothetical biological target space covered by these diversity sets was investigated through bioinformatics methodologies. Consequently, three anti-kinetoplastid chemical boxes of ~200 compounds each were assembled. Functional analyses of these compounds suggest a wide array of potential modes of action against kinetoplastid kinases, proteases and cytochromes as well as potential host–pathogen targets. This is the first published parallel high throughput screening of a pharma compound collection against kinetoplastids. The compound sets are provided as an open resource for future lead discovery programs, and to address important research questions.
Collapse
Affiliation(s)
- Imanol Peña
- Molecular Discovery Research, Tres Cantos Medicines Development Campus, GlaxoSmithKline, Tres Cantos, Spain
| | - M Pilar Manzano
- Diseases of the Developing World (DDW), Tres Cantos Medicines Development Campus, GlaxoSmithKline, Tres Cantos, Spain
| | - Juan Cantizani
- Diseases of the Developing World (DDW), Tres Cantos Medicines Development Campus, GlaxoSmithKline, Tres Cantos, Spain
| | - Albane Kessler
- Diseases of the Developing World (DDW), Tres Cantos Medicines Development Campus, GlaxoSmithKline, Tres Cantos, Spain
| | - Julio Alonso-Padilla
- Department of Microbiology, Division of Parasitology, New York University School of Medicine, New York, NY, USA
| | - Ana I Bardera
- Molecular Discovery Research, Tres Cantos Medicines Development Campus, GlaxoSmithKline, Tres Cantos, Spain
| | - Emilio Alvarez
- Molecular Discovery Research, Tres Cantos Medicines Development Campus, GlaxoSmithKline, Tres Cantos, Spain
| | - Gonzalo Colmenarejo
- Molecular Discovery Research, Tres Cantos Medicines Development Campus, GlaxoSmithKline, Tres Cantos, Spain
| | - Ignacio Cotillo
- Diseases of the Developing World (DDW), Tres Cantos Medicines Development Campus, GlaxoSmithKline, Tres Cantos, Spain
| | - Irene Roquero
- Molecular Discovery Research, Tres Cantos Medicines Development Campus, GlaxoSmithKline, Tres Cantos, Spain
| | - Francisco de Dios-Anton
- Molecular Discovery Research, Tres Cantos Medicines Development Campus, GlaxoSmithKline, Tres Cantos, Spain
| | - Vanessa Barroso
- Molecular Discovery Research, Tres Cantos Medicines Development Campus, GlaxoSmithKline, Tres Cantos, Spain
| | - Ana Rodriguez
- Department of Microbiology, Division of Parasitology, New York University School of Medicine, New York, NY, USA
| | - David W Gray
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, UK
| | - Miguel Navarro
- Instituto de Parasitología y Biomedicina "López-Neyra" Consejo Superior de Investigaciones Cientificas, Granada, Spain
| | - Vinod Kumar
- Computational Biology, Quantitative Sciences, GlaxoSmithKline, Collegeville, PA, USA
| | - Alexander Sherstnev
- Computational Biology, Quantitative Sciences, GlaxoSmithKline, Medicines Research Center, Stevenage, Hertfordshire, UK
| | - David H Drewry
- Chemical Sciences, Molecular Discovery Research, GlaxoSmithKline, Research Triangle Park, NC, USA
| | - James R Brown
- Computational Biology, Quantitative Sciences, GlaxoSmithKline, Collegeville, PA, USA
| | - Jose M Fiandor
- Diseases of the Developing World (DDW), Tres Cantos Medicines Development Campus, GlaxoSmithKline, Tres Cantos, Spain
| | - J Julio Martin
- Molecular Discovery Research, Tres Cantos Medicines Development Campus, GlaxoSmithKline, Tres Cantos, Spain
| |
Collapse
|
35
|
Nagle A, Khare S, Kumar AB, Supek F, Buchynskyy A, Mathison CJN, Chennamaneni N, Pendem N, Buckner FS, Gelb M, Molteni V. Recent developments in drug discovery for leishmaniasis and human African trypanosomiasis. Chem Rev 2014; 114:11305-47. [PMID: 25365529 PMCID: PMC4633805 DOI: 10.1021/cr500365f] [Citation(s) in RCA: 243] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Indexed: 02/08/2023]
Affiliation(s)
- Advait
S. Nagle
- Genomics
Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Shilpi Khare
- Genomics
Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Arun Babu Kumar
- Departments of Chemistry, Biochemistry, and Medicine, University
of Washington, Seattle, Washington 98195, United States
| | - Frantisek Supek
- Genomics
Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Andriy Buchynskyy
- Departments of Chemistry, Biochemistry, and Medicine, University
of Washington, Seattle, Washington 98195, United States
| | - Casey J. N. Mathison
- Genomics
Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Naveen
Kumar Chennamaneni
- Departments of Chemistry, Biochemistry, and Medicine, University
of Washington, Seattle, Washington 98195, United States
| | - Nagendar Pendem
- Departments of Chemistry, Biochemistry, and Medicine, University
of Washington, Seattle, Washington 98195, United States
| | - Frederick S. Buckner
- Departments of Chemistry, Biochemistry, and Medicine, University
of Washington, Seattle, Washington 98195, United States
| | - Michael
H. Gelb
- Departments of Chemistry, Biochemistry, and Medicine, University
of Washington, Seattle, Washington 98195, United States
| | - Valentina Molteni
- Genomics
Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| |
Collapse
|
36
|
Identification and characterization of hundreds of potent and selective inhibitors of Trypanosoma brucei growth from a kinase-targeted library screening campaign. PLoS Negl Trop Dis 2014; 8:e3253. [PMID: 25340575 PMCID: PMC4207660 DOI: 10.1371/journal.pntd.0003253] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 09/08/2014] [Indexed: 01/07/2023] Open
Abstract
In the interest of identification of new kinase-targeting chemotypes for target and pathway analysis and drug discovery in Trypanosomal brucei, a high-throughput screen of 42,444 focused inhibitors from the GlaxoSmithKline screening collection was performed against parasite cell cultures and counter-screened against human hepatocarcinoma (HepG2) cells. In this way, we have identified 797 sub-micromolar inhibitors of T. brucei growth that are at least 100-fold selective over HepG2 cells. Importantly, 242 of these hit compounds acted rapidly in inhibiting cellular growth, 137 showed rapid cidality. A variety of in silico and in vitro physicochemical and drug metabolism properties were assessed, and human kinase selectivity data were obtained, and, based on these data, we prioritized three compounds for pharmacokinetic assessment and demonstrated parasitological cure of a murine bloodstream infection of T. brucei rhodesiense with one of these compounds (NEU-1053). This work represents a successful implementation of a unique industrial-academic collaboration model aimed at identification of high quality inhibitors that will provide the parasitology community with chemical matter that can be utilized to develop kinase-targeting tool compounds. Furthermore these results are expected to provide rich starting points for discovery of kinase-targeting tool compounds for T. brucei, and new HAT therapeutics discovery programs. Human African trypanosomiasis, or sleeping sickness, affects 10,000 patients annually, yet current drugs for this disease are poor, with high toxicity and inconvenient dosing requirements. Trypanosoma brucei, the parasite that causes sleeping sickness, is sensitive to a class of compounds called kinase inhibitors, and our project was aimed at identifying kinase-targeting compounds that rapidly and irreversibly inhibit parasite growth. This was accomplished by high-throughput screening of over 42,000 compounds, which resulted in identification of 797 potent inhibitors of parasite growth that are non-toxic to human cells. These inhibitors were studied for the speed of their effects and reversibility of growth inhibition, and were grouped on the basis of chemical structure similarity. One compound was shown to cure mice from a bloodstream of infection of T. brucei. These compounds can now be utilized by the research community as starting points for new drug discovery, and also as tool compounds for understanding the function of kinases in T. brucei.
Collapse
|
37
|
Vij A, Biswas A, Bhattacharya A, Das PK. A soluble phosphodiesterase in Leishmania donovani negatively regulates cAMP signaling by inhibiting protein kinase A through a two way process involving catalytic as well as non-catalytic sites. Int J Biochem Cell Biol 2014; 57:197-206. [PMID: 25310904 DOI: 10.1016/j.biocel.2014.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 09/09/2014] [Accepted: 10/02/2014] [Indexed: 10/24/2022]
Abstract
Intracellular cAMP level and cAMP mediated responses are elevated when Leishmania are exposed to macrophage phagolysosome conditions (37 °C and pH 5.5). Phosphodiesterases play major role in cAMP regulation and in the present study we have cloned and characterized a 2.1 kb cytosolic isoform of phosphodiesterase from Leishmania donovani (LdPDED) which plays important role in cAMP homeostasis when the promastigotes are exposed to macrophage phagolysome conditions for converting to axenic amastigotes. Domain characterization suggested the presence of two pseudo-substrate sites similar to the ones present in the regulatory subunit of cAMP-dependent protein kinase A (PKA) and a putative PKA phosphorylation site at T(708) of C-terminus of LdPDED. Deletion constructs and site directed mutagenesis revealed the ability of LdPDED to interact with L. donovani PKA catalytic subunits (LdPKAC1 and LdPKAC2) resulting in inhibition of kinase activity in one hand and increase of phosphodiesterase activity through PKA mediated phosphorylation at putative phosphorylation site on the other hand. This study therefore identifies a unique phosphodiesterase in L. donovani which appears to regulate cAMP-dependent PKA signaling through a two way process.
Collapse
Affiliation(s)
- Amit Vij
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Arunima Biswas
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | | | - Pijush K Das
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India.
| |
Collapse
|
38
|
Denny PW, Steel PG. Yeast as a potential vehicle for neglected tropical disease drug discovery. ACTA ACUST UNITED AC 2014; 20:56-63. [PMID: 25121554 DOI: 10.1177/1087057114546552] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
High-throughput screening (HTS) efforts for neglected tropical disease (NTD) drug discovery have recently received increased attention because several initiatives have begun to attempt to reduce the deficit in new and clinically acceptable therapies for this spectrum of infectious diseases. HTS primarily uses two basic approaches, cell-based and in vitro target-directed screening. Both of these approaches have problems; for example, cell-based screening does not reveal the target or targets that are hit, whereas in vitro methodologies lack a cellular context. Furthermore, both can be technically challenging, expensive, and difficult to miniaturize for ultra-HTS [(u)HTS]. The application of yeast-based systems may overcome some of these problems and offer a cost-effective platform for target-directed screening within a eukaryotic cell context. Here, we review the advantages and limitations of the technologies that may be used in yeast cell-based, target-directed screening protocols, and we discuss how these are beginning to be used in NTD drug discovery.
Collapse
Affiliation(s)
- P W Denny
- Biophysical Sciences Institute, Department of Chemistry and School of Biological Sciences, University Science Laboratories, Durham, UK School of Medicine, Pharmacy and Health, Durham University, Durham, UK
| | - P G Steel
- Biophysical Sciences Institute, Department of Chemistry and School of Biological Sciences, University Science Laboratories, Durham, UK
| |
Collapse
|
39
|
Woodring JL, Pollastri MP. Inhibitors of Protozoan Phosphodiesterases as Potential Therapeutic Approaches for Tropical Diseases. ACTA ACUST UNITED AC 2014. [DOI: 10.1002/9783527682348.ch12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
40
|
Njoroge M, Njuguna NM, Mutai P, Ongarora DSB, Smith PW, Chibale K. Recent approaches to chemical discovery and development against malaria and the neglected tropical diseases human African trypanosomiasis and schistosomiasis. Chem Rev 2014; 114:11138-63. [PMID: 25014712 DOI: 10.1021/cr500098f] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
| | | | | | | | - Paul W Smith
- Novartis Institute for Tropical Diseases , Singapore 138670, Singapore
| | | |
Collapse
|
41
|
Ogungbe IV, Erwin WR, Setzer WN. Antileishmanial phytochemical phenolics: molecular docking to potential protein targets. J Mol Graph Model 2014; 48:105-17. [PMID: 24463105 DOI: 10.1016/j.jmgm.2013.12.010] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Revised: 12/21/2013] [Accepted: 12/30/2013] [Indexed: 11/25/2022]
Abstract
A molecular docking analysis has been carried out to examine potential Leishmania protein targets of antiprotozoal plant-derived polyphenolic compounds. A total of 352 phenolic phytochemicals, including 10 aurones, six cannabinoids, 34 chalcones, 20 chromenes, 52 coumarins, 92 flavonoids, 41 isoflavonoids, 52 lignans, 25 quinones, eight stilbenoids, nine xanthones, and three miscellaneous phenolic compounds, were used in the virtual screening study using 24 Leishmania enzymes (52 different protein structures from the Protein Data Bank). Noteworthy protein targets were Leishmania dihydroorotate dehydrogenase, N-myristoyl transferase, phosphodiesterase B1, pteridine reductase, methionyl-tRNA synthetase, tyrosyl-tRNA synthetase, uridine diphosphate-glucose pyrophosphorylase, nicotinamidase, and glycerol-3-phosphate dehydrogenase. Based on in-silico analysis of antiparasitic polyphenolics in this study, two aurones, one chalcone, five coumarins, six flavonoids, one isoflavonoid, three lignans, and one stilbenoid, can be considered to be promising drug leads worthy of further investigation.
Collapse
Affiliation(s)
- Ifedayo Victor Ogungbe
- Department of Chemistry & Biochemistry, Jackson State University, Jackson, MS 39217, USA.
| | - William R Erwin
- Department of Chemistry, University of Alabama in Huntsville, Huntsville, AL 35899, USA
| | - William N Setzer
- Department of Chemistry, University of Alabama in Huntsville, Huntsville, AL 35899, USA.
| |
Collapse
|
42
|
The utility of yeast as a tool for cell-based, target-directed high-throughput screening. Parasitology 2013; 141:8-16. [PMID: 23611102 DOI: 10.1017/s0031182013000425] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Many Neglected Tropical Diseases (NTDs) have recently been subject of increased focus, particularly with relation to high-throughput screening (HTS) initiatives. These vital endeavours largely rely of two approaches, in vitro target-directed screening using biochemical assays or cell-based screening which takes no account of the target or targets being hit. Despite their successes both of these approaches have limitations; for example, the production of soluble protein and a lack of cellular context or the problems and expense of parasite cell culture. In addition, both can be challenging to miniaturize for ultra (u)HTS and expensive to utilize. Yeast-based systems offer a cost-effective approach to study and screen protein targets in a direct-directed manner within a eukaryotic cellular context. In this review, we examine the utility and limitations of yeast cell-based, target-directed screening. In particular we focus on the currently under-explored possibility of using such formats in uHTS screening campaigns for NTDs.
Collapse
|
43
|
Jansen C, Wang H, Kooistra AJ, de Graaf C, Orrling KM, Tenor H, Seebeck T, Bailey D, de Esch IJP, Ke H, Leurs R. Discovery of novel Trypanosoma brucei phosphodiesterase B1 inhibitors by virtual screening against the unliganded TbrPDEB1 crystal structure. J Med Chem 2013; 56:2087-96. [PMID: 23409953 DOI: 10.1021/jm3017877] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Trypanosoma brucei cyclic nucleotide phosphodiesterase B1 (TbrPDEB1) and TbrPDEB2 have recently been validated as new therapeutic targets for human African trypanosomiasis by both genetic and pharmacological means. In this study we report the crystal structure of the catalytic domain of the unliganded TbrPDEB1 and its use for the in silico screening for new TbrPDEB1 inhibitors with novel scaffolds. The TbrPDEB1 crystal structure shows the characteristic folds of human PDE enzymes but also contains the parasite-specific P-pocket found in the structures of Leishmania major PDEB1 and Trypanosoma cruzi PDEC. The unliganded TbrPDEB1 X-ray structure was subjected to a structure-based in silico screening approach that combines molecular docking simulations with a protein-ligand interaction fingerprint (IFP) scoring method. This approach identified six novel TbrPDEB1 inhibitors with IC50 values of 10-80 μM, which may be further optimized as potential selective TbrPDEB inhibitors.
Collapse
Affiliation(s)
- Chimed Jansen
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), VU University Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Müller J, Hemphill A. New approaches for the identification of drug targets in protozoan parasites. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 301:359-401. [PMID: 23317822 DOI: 10.1016/b978-0-12-407704-1.00007-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Antiparasitic chemotherapy is an important issue for drug development. Traditionally, novel compounds with antiprotozoan activities have been identified by screening of compound libraries in high-throughput systems. More recently developed approaches employ target-based drug design supported by genomics and proteomics of protozoan parasites. In this chapter, the drug targets in protozoan parasites are reviewed. The gene-expression machinery has been among the first targets for antiparasitic drugs and is still under investigation as a target for novel compounds. Other targets include cytoskeletal proteins, proteins involved in intracellular signaling, membranes, and enzymes participating in intermediary metabolism. In apicomplexan parasites, the apicoplast is a suitable target for established and novel drugs. Some drugs act on multiple subcellular targets. Drugs with nitro groups generate free radicals under anaerobic growth conditions, and drugs with peroxide groups generate radicals under aerobic growth conditions, both affecting multiple cellular pathways. Mefloquine and thiazolides are presented as examples for antiprotozoan compounds with multiple (side) effects. The classic approach of drug discovery employing high-throughput physiological screenings followed by identification of drug targets has yielded the mainstream of current antiprotozoal drugs. Target-based drug design supported by genomics and proteomics of protozoan parasites has not produced any antiparasitic drug so far. The reason for this is discussed and a synthesis of both methods is proposed.
Collapse
Affiliation(s)
- Joachim Müller
- Institute of Parasitology, University of Berne, Berne, Switzerland.
| | | |
Collapse
|
45
|
Orrling KM, Jansen C, Vu XL, Balmer V, Bregy P, Shanmugham A, England P, Bailey D, Cos P, Maes L, Adams E, van den Bogaart E, Chatelain E, Ioset JR, van de Stolpe A, Zorg S, Veerman J, Seebeck T, Sterk GJ, de Esch IJP, Leurs R. Catechol pyrazolinones as trypanocidals: fragment-based design, synthesis, and pharmacological evaluation of nanomolar inhibitors of trypanosomal phosphodiesterase B1. J Med Chem 2012; 55:8745-56. [PMID: 22963052 DOI: 10.1021/jm301059b] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Trypanosomal phosphodiesterases B1 and B2 (TbrPDEB1 and TbrPDEB2) play an important role in the life cycle of Trypanosoma brucei, the causative parasite of human African trypanosomiasis (HAT), also known as African sleeping sickness. We used homology modeling and docking studies to guide fragment growing into the parasite-specific P-pocket in the enzyme binding site. The resulting catechol pyrazolinones act as potent TbrPDEB1 inhibitors with IC₅₀ values down to 49 nM. The compounds also block parasite proliferation (e.g., VUF13525 (20b): T. brucei rhodesiense IC₅₀ = 60 nM, T. brucei brucei IC₅₀ = 520 nM, T. cruzi = 7.6 μM), inducing a typical multiple nuclei and kinetoplast phenotype without being generally cytotoxic. The mode of action of 20b was investigated with recombinantly engineered trypanosomes expressing a cAMP-sensitive FRET sensor, confirming a dose-response related increase of intracellular cAMP levels in trypanosomes. Our findings further validate the TbrPDEB family as antitrypanosomal target.
Collapse
Affiliation(s)
- Kristina M Orrling
- Leiden/Amsterdam Centre of Drug Research-LACDR, Amsterdam Institute of Molecules, Medicines and Systems- AIMMS, Division of Medicinal Chemistry, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Interview: Interview with Future Medicinal Chemistry’s US Senior Editor, Iwao Ojima. Future Med Chem 2012; 4:2019-22. [DOI: 10.4155/fmc.12.151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Professor Iwao Ojima studied at the University of Tokyo (Japan) before being appointed as a Senior Research Fellow and Group Leader at the Sagami Institute of Chemical Research. He is now Director of the Institute of Chemical Biology and Drug Discovery at State University of New York (USA) and has been a visiting professor in European, North American and Asian academic institutions. Professor Ojima agreed to serve as the US Senior Editor of Future Medicinal Chemistry when it launched in 2009 and continues to provide his expertise to the journal. Professor Ojima spoke to Future Medicinal Chemistry about why medicinal chemistry is such an exciting field to work in, the state of the pharmaceutical industry, and what features and issues make this journal unique. Interview conducted by Isaac Bruce, Commissioning Editor.
Collapse
|
47
|
Wang H, Kunz S, Chen G, Seebeck T, Wan Y, Robinson H, Martinelli S, Ke H. Biological and structural characterization of Trypanosoma cruzi phosphodiesterase C and Implications for design of parasite selective inhibitors. J Biol Chem 2012; 287:11788-97. [PMID: 22356915 PMCID: PMC3320927 DOI: 10.1074/jbc.m111.326777] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Revised: 02/06/2012] [Indexed: 12/12/2022] Open
Abstract
Trypanosoma cruzi phosphodiesterase C (TcrPDEC) is a potential new drug target for the treatment of Chagas disease but has not been well studied. This study reports the enzymatic properties of various kinetoplastid PDECs and the crystal structures of the unliganded TcrPDEC1 catalytic domain and its complex with an inhibitor. Mutations of PDEC during the course of evolution led to inactivation of PDEC in Trypanosoma brucei/Trypanosoma evansi/Trypanosoma congolense, whereas the enzyme is active in all other kinetoplastids. The TcrPDEC1 catalytic domain hydrolyzes both cAMP and cGMP with a K(m) of 23.8 μm and a k(cat) of 31 s(-1) for cAMP and a K(m) of 99.1 μm and a k(cat) of 17 s(-1) for cGMP, thus confirming its dual specificity. The crystal structures show that the N-terminal fragment wraps around the TcrPDEC catalytic domain and may thus regulate its enzymatic activity via direct interactions with the active site residues. A PDE5 selective inhibitor that has an IC(50) of 230 nm for TcrPDEC1 binds to TcrPDEC1 in an orientation opposite to that of sildenafil. This observation, together with the screen of the inhibitory potency of human PDE inhibitors against TcrPDEC, implies that the scaffold of some human PDE inhibitors might be used as the starting model for design of parasite PDE inhibitors. The structural study also identified a unique parasite pocket that neighbors the active site and may thus be valuable for the design of parasite-specific inhibitors.
Collapse
Affiliation(s)
- Huanchen Wang
- From the Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599-7260
| | - Stefan Kunz
- the Institute for Cell Biology, University of Bern, CH-3012 Bern, Switzerland
| | - Gong Chen
- the School of Chemistry and Chemical Engineering, Sun Yat-sen University, Guangzhou 510275, China
| | - Thomas Seebeck
- the Institute for Cell Biology, University of Bern, CH-3012 Bern, Switzerland
| | - Yiqian Wan
- the School of Chemistry and Chemical Engineering, Sun Yat-sen University, Guangzhou 510275, China
| | - Howard Robinson
- the Biology Department, Brookhaven National Laboratory, Upton, New York 11973-5000, and
| | - Sibylla Martinelli
- the Department of Pathology, University of Bern, CH-3012 Bern, Switzerland
| | - Hengming Ke
- From the Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599-7260
| |
Collapse
|
48
|
Ochiana SO, Gustafson A, Bland ND, Wang C, Russo MJ, Campbell RK, Pollastri MP. Synthesis and evaluation of human phosphodiesterases (PDE) 5 inhibitor analogs as trypanosomal PDE inhibitors. Part 2. Tadalafil analogs. Bioorg Med Chem Lett 2012; 22:2582-4. [PMID: 22377518 PMCID: PMC3307956 DOI: 10.1016/j.bmcl.2012.01.118] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 01/25/2012] [Accepted: 01/30/2012] [Indexed: 12/19/2022]
Abstract
In this Letter we describe our ongoing target repurposing efforts focused on discovery of inhibitors of the essential trypanosomal phosphodiesterase TbrPDEB1. This enzyme has been implicated in virulence of Trypanosoma brucei, the causative agent of human African trypanosomiasis (HAT). We outline the synthesis and biological evaluation of analogs of tadalafil, a human PDE5 inhibitor currently utilized for treatment of erectile dysfunction, and report that these analogs are weak inhibitors of TbrPDEB1.
Collapse
Affiliation(s)
- Stefan O Ochiana
- Northeastern University Department of Chemistry and Chemical Biology, 417 Egan Research Center, 360 Huntington Avenue, Boston, MA 02115, United States
| | | | | | | | | | | | | |
Collapse
|
49
|
Wang C, Ashton TD, Gustafson A, Bland ND, Ochiana SO, Campbell RK, Pollastri MP. Synthesis and evaluation of human phosphodiesterases (PDE) 5 inhibitor analogs as trypanosomal PDE inhibitors. Part 1. Sildenafil analogs. Bioorg Med Chem Lett 2012; 22:2579-81. [PMID: 22370268 PMCID: PMC3307826 DOI: 10.1016/j.bmcl.2012.01.119] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 01/25/2012] [Accepted: 01/30/2012] [Indexed: 12/20/2022]
Abstract
Parasitic diseases, such as African sleeping sickness, have a significant impact on the health and well-being in the poorest regions of the world. Pragmatic drug discovery efforts are needed to find new therapeutic agents. In this Letter we describe target repurposing efforts focused on trypanosomal phosphodiesterases. We outline the synthesis and biological evaluation of analogs of sildenafil (1), a human PDE5 inhibitor, for activities against trypanosomal PDEB1 (TbrPDEB1). We find that, while low potency analogs can be prepared, this chemical class is a sub-optimal starting point for further development of TbrPDE inhibitors.
Collapse
Affiliation(s)
- Cuihua Wang
- Northeastern University Department of Chemistry & Chemical Biology, 417 Egan Research Center, 360 Huntington Avenue, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
de Koning HP, Gould MK, Sterk GJ, Tenor H, Kunz S, Luginbuehl E, Seebeck T. Pharmacological validation of Trypanosoma brucei phosphodiesterases as novel drug targets. J Infect Dis 2012; 206:229-37. [PMID: 22291195 PMCID: PMC3379837 DOI: 10.1093/infdis/jir857] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The development of drugs for neglected infectious diseases often uses parasite-specific enzymes as targets. We here demonstrate that parasite enzymes with highly conserved human homologs may represent a promising reservoir of new potential drug targets. The cyclic nucleotide-specific phosphodiesterases (PDEs) of Trypanosoma brucei, causative agent of the fatal human sleeping sickness, are essential for the parasite. The highly conserved human homologs are well-established drug targets. We here describe what is to our knowledge the first pharmacological validation of trypanosomal PDEs as drug targets. High-throughput screening of a proprietary compound library identified a number of potent hits. One compound, the tetrahydrophthalazinone compound A (Cpd A), was further characterized. It causes a dramatic increase of intracellular cyclic adenosine monophosphate (cAMP). Short-term cell viability is not affected, but cell proliferation is inhibited immediately, and cell death occurs within 3 days. Cpd A prevents cytokinesis, resulting in multinucleated, multiflagellated cells that eventually lyse. These observations pharmacologically validate the highly conserved trypanosomal PDEs as potential drug targets.
Collapse
Affiliation(s)
- Harry P de Koning
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
| | | | | | | | | | | | | |
Collapse
|