1
|
Mao EY, Page SW, Sleebs BE, Gancheva MR, Wilson DW. A review of natural products as a source of next-generation drugs against apicomplexan parasites. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:51. [PMID: 40481189 PMCID: PMC12144137 DOI: 10.1038/s44259-025-00119-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 05/12/2025] [Indexed: 06/11/2025]
Abstract
Despite the substantial global health and economic burden of apicomplexan parasites in humans and livestock, treatment options remain limited. Natural products have long played an important role in combating these diseases, offering diverse chemical structures and bioactive compounds. This review summarises past and present natural-product-based therapies for six economically significant apicomplexans and explores the potential of revisiting natural products as a source of next-generation treatments.
Collapse
Affiliation(s)
- Emma Y Mao
- Research Centre for Infectious Diseases, School of Biological Sciences, The University of Adelaide, Adelaide, SA, 5005, Australia.
- ARC Training Centre for Environmental and Agricultural Solutions to Antimicrobial Resistance (CEAStAR), St Lucia, Queensland, Australia.
- Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, 5005, SA, Australia.
| | | | - Brad E Sleebs
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Maria R Gancheva
- Research Centre for Infectious Diseases, School of Biological Sciences, The University of Adelaide, Adelaide, SA, 5005, Australia
- ARC Training Centre for Environmental and Agricultural Solutions to Antimicrobial Resistance (CEAStAR), St Lucia, Queensland, Australia
- Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, 5005, SA, Australia
| | - Danny W Wilson
- Research Centre for Infectious Diseases, School of Biological Sciences, The University of Adelaide, Adelaide, SA, 5005, Australia.
- ARC Training Centre for Environmental and Agricultural Solutions to Antimicrobial Resistance (CEAStAR), St Lucia, Queensland, Australia.
- Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, 5005, SA, Australia.
- Burnet Institute, Melbourne, 3004, Victoria, Australia.
| |
Collapse
|
2
|
Samarelli R, Pugliese N, Saleh M, Prioletti M, Cordon R, Cavicchio P, Salierno D, Crescenzo G, Circella E, Camarda A. Treatment of avian malaria in captive African penguins ( Spheniscus demersus) by the combination of atovaquone and proguanil hydrochloride. Int J Vet Sci Med 2025; 13:1-8. [PMID: 40007641 PMCID: PMC11852231 DOI: 10.1080/23144599.2025.2460919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 02/27/2025] Open
Abstract
Avian malaria, a vector-borne disease caused by Plasmodium spp., poses significant threats to various bird populations, particularly captive penguins like the endangered African penguin (Spheniscus demersus). Penguins, originating from regions with low malaria prevalence, are highly susceptible when housed in malaria-permissive areas. This study evaluates the efficacy of an atovaquone/proguanil hydrochloride treatment protocol to manage avian malaria in a captive African penguin colony in an Italian zoo. The study involved 30 penguins monitored over 3 years. Thirteen penguins tested positive for Plasmodium spp., with 11 undergoing treatment. The treatment protocol consisted of atovaquone/proguanil hydrochloride (10/4 mg/kg) administered orally for 3 days, repeated after a week. Post-treatment monitoring at 7, 30, and 60 days, and follow-ups up to 2 years, showed that all but one penguin cleared the infection. The treatment was well tolerated, with no adverse effects observed. The findings suggest that this protocol is effective as a treatment of avian malaria and could be a valuable tool in avian malaria management, particularly for endangered species in captivity. However, the persistence of Plasmodium relictum in one case highlights the need for careful post-treatment monitoring to prevent recurrence or reinfection. The study underscores the importance of developing tailored antimalarial protocols for captive birds to enhance conservation efforts and mitigate the risks posed by avian malaria.
Collapse
Affiliation(s)
- Rossella Samarelli
- Dipartimento di Medicina Veterinaria, Università degli Studi di Bari, Valenzano, Italy
| | - Nicola Pugliese
- Dipartimento di Medicina Veterinaria, Università degli Studi di Bari, Valenzano, Italy
| | - Medhat Saleh
- Dipartimento di Medicina Veterinaria, Università degli Studi di Bari, Valenzano, Italy
- Department of Animal Production, Faculty of Agriculture, Benha University, Benha, Egypt
| | - Michela Prioletti
- Dipartimento di Medicina Veterinaria, Università degli Studi di Bari, Valenzano, Italy
| | | | | | - Dalila Salierno
- Dipartimento di Medicina Veterinaria, Università degli Studi di Bari, Valenzano, Italy
| | - Giuseppe Crescenzo
- Dipartimento di Medicina Veterinaria, Università degli Studi di Bari, Valenzano, Italy
| | - Elena Circella
- Dipartimento di Medicina Veterinaria, Università degli Studi di Bari, Valenzano, Italy
| | - Antonio Camarda
- Dipartimento di Medicina Veterinaria, Università degli Studi di Bari, Valenzano, Italy
| |
Collapse
|
3
|
Bhargava S, Deshmukh R, Dewangan HK. Recent Advancement in Drug Development for Treating Malaria using Herbal Medicine and Nanotechnological Approach. Curr Pharm Des 2025; 31:203-218. [PMID: 39279710 DOI: 10.2174/0113816128321468240828103439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/05/2024] [Accepted: 07/12/2024] [Indexed: 09/18/2024]
Abstract
More than two hundred million people around the world are infected with malaria, a blood-borne disease that poses a significant risk to human life. Single medications, such as lumefantrine, primaquine, and chloroquine, as well as combinations of these medications with artemisinin or its derivatives, are currently being used as therapies. In addition, due to rising antimalarial drug resistance, other therapeutic options are needed immediately. Furthermore, due to anti-malarial medication failures, a new drug is required. Medication discovery and development are costly and time-consuming. Many malaria treatments have been developed however, most treatments have low water solubility and bioavailability. They may also cause drugresistant parasites, which would increase malaria cases and fatalities. Nanotechnology may offer a safer, more effective malaria therapy and control option. Nanoparticles' high loading capacity, concentrated drug delivery, biocompatibility, and low toxicity make them an attractive alternative to traditional therapy. Nanotechnologybased anti-malarial chemotherapeutic medications outperform conventional therapies in therapeutic benefits, safety, and cost. This improves patient treatment compliance. The limitations of malaria treatments and the importance of nanotechnological approaches to the treatment of malaria were also topics that were covered in this review. The most recent advancements in nanomaterials and the advantages they offer in terms of medication delivery are discussed in this article. The prospective therapy for malaria is also discussed. Additionally, the limitations of malaria therapies and the importance of nanotechnology-based approaches to the treatment of malaria were explored.
Collapse
Affiliation(s)
- Sarvesh Bhargava
- Institute of Pharmaceutical Research, GLA University, Mathura, U.P., India
| | - Rohitas Deshmukh
- Institute of Pharmaceutical Research, GLA University, Mathura, U.P., India
| | - Hitesh Kumar Dewangan
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
| |
Collapse
|
4
|
Ouji M, Reyser T, Yamaryo-Botté Y, Nguyen M, Rengel D, Dutreuil A, Marcellin M, Burlet-Schiltz O, Augereau JM, Riscoe MK, Paloque L, Botté C, Benoit-Vical F. In artemisinin-resistant falciparum malaria parasites, mitochondrial metabolic pathways are essential for survival but not those of apicoplast. Int J Parasitol Drugs Drug Resist 2024; 26:100565. [PMID: 39332236 PMCID: PMC11466614 DOI: 10.1016/j.ijpddr.2024.100565] [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: 03/13/2024] [Revised: 09/13/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024]
Abstract
Emergence and spread of parasite resistance to artemisinins, the first-line antimalarial therapy, threaten the malaria eradication policy. To identify therapeutic targets to eliminate artemisinin-resistant parasites, the functioning of the apicoplast and the mitochondrion was studied, focusing on the fatty acid synthesis type II (FASII) pathway in the apicoplast and the electron transfer chain in the mitochondrion. A significant enrichment of the FASII pathway among the up-regulated genes in artemisinin-resistant parasites under dihydroartemisinin treatment was found, in agreement with published transcriptomic data. However, using GC-MS analyzes of fatty acids, we demonstrated for the first time that the FASII pathway is non-functional, ruling out the use of FASII inhibitors to target artemisinin-resistant parasites. Conversely, by assessing the modulation of the oxygen consumption rate, we evidenced that mitochondrial respiration remains functional and flexible in artemisinin-resistant parasites and even at the quiescent stage. Two novel compounds targeting electron transport chain (ELQ300, ELQ400) efficiently killed quiescent artemisinin-resistant parasites. Therefore, mitochondrial respiration represents a key target for the elimination of artemisinin-resistant persistent Plasmodium falciparum parasites.
Collapse
Affiliation(s)
- Manel Ouji
- LCC-CNRS, Laboratoire de Chimie de Coordination, Université de Toulouse, CNRS, Toulouse, France; MAAP, New Antimalarial Molecules and Pharmacological Approaches, Inserm ERL 1289, Toulouse, France; Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France
| | - Thibaud Reyser
- LCC-CNRS, Laboratoire de Chimie de Coordination, Université de Toulouse, CNRS, Toulouse, France; MAAP, New Antimalarial Molecules and Pharmacological Approaches, Inserm ERL 1289, Toulouse, France; Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France
| | - Yoshiki Yamaryo-Botté
- ApicoLipid Team, Institute for Advanced Biosciences, CNRS UMR5309, Université Grenoble Alpes, INSERM U1209, Grenoble, France
| | - Michel Nguyen
- LCC-CNRS, Laboratoire de Chimie de Coordination, Université de Toulouse, CNRS, Toulouse, France; MAAP, New Antimalarial Molecules and Pharmacological Approaches, Inserm ERL 1289, Toulouse, France; Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France
| | - David Rengel
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France
| | - Axelle Dutreuil
- LCC-CNRS, Laboratoire de Chimie de Coordination, Université de Toulouse, CNRS, Toulouse, France; MAAP, New Antimalarial Molecules and Pharmacological Approaches, Inserm ERL 1289, Toulouse, France; Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France
| | - Marlène Marcellin
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France; Infrastructure nationale de Protéomique, ProFI, FR 2048, Toulouse, France
| | - Odile Burlet-Schiltz
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France; Infrastructure nationale de Protéomique, ProFI, FR 2048, Toulouse, France
| | - Jean-Michel Augereau
- LCC-CNRS, Laboratoire de Chimie de Coordination, Université de Toulouse, CNRS, Toulouse, France; MAAP, New Antimalarial Molecules and Pharmacological Approaches, Inserm ERL 1289, Toulouse, France; Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France
| | - Michael K Riscoe
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA; Department of Molecular Microbiology and Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Lucie Paloque
- LCC-CNRS, Laboratoire de Chimie de Coordination, Université de Toulouse, CNRS, Toulouse, France; MAAP, New Antimalarial Molecules and Pharmacological Approaches, Inserm ERL 1289, Toulouse, France; Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France
| | - Cyrille Botté
- ApicoLipid Team, Institute for Advanced Biosciences, CNRS UMR5309, Université Grenoble Alpes, INSERM U1209, Grenoble, France
| | - Françoise Benoit-Vical
- LCC-CNRS, Laboratoire de Chimie de Coordination, Université de Toulouse, CNRS, Toulouse, France; MAAP, New Antimalarial Molecules and Pharmacological Approaches, Inserm ERL 1289, Toulouse, France; Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France.
| |
Collapse
|
5
|
Gholam GM, Mahendra FR, Irsal RAP, Dwicesaria MA, Ariefin M, Kristiadi M, Rizki AFM, Azmi WA, Artika IM, Siregar JE. Computational exploration of compounds in Xylocarpus granatum as a potential inhibitor of Plasmodium berghei using docking, molecular dynamics, and DFT studies. Biochem Biophys Res Commun 2024; 733:150684. [PMID: 39293331 DOI: 10.1016/j.bbrc.2024.150684] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 09/20/2024]
Abstract
Malaria remains a global health concern, with the emergence of resistance to the antimalarial drug atovaquone through cytochrome b (cyt b) being well-documented. This study was prompted by the presence of this mutation in cyt b to enable new drug candidates capable of overcoming drug resistance. Our objective was to identify potential drug candidates from compounds of Xylocarpus granatum by computationally assessing their interactions with Plasmodium berghei cyt b. Using computational methods, we modeled cyt b (GenBank: AF146076.1), identified the binding cavity, and analyzed the Ramachandran plot against cyt b. Additionally, we conducted drug-likeness and absorption, distribution, metabolism, excretion, and toxicity (ADMET) studies, along with density functional theory (DFT) analysis of the compounds. Molecular docking and molecular dynamics simulation (MDS) were used to evaluate the binding energy and stability of the cyt b-ligand complex. Notably, our investigation highlighted kaempferol as a promising compound due to its high binding energy of 7.67 kcal/mol among all X. granatum compounds, coupled with favorable pharmacological properties (ADMET) and antiprotozoal properties at Pa 0.345 > Pi 0.009 (PASS value). DFT analysis showed that kaempferol has an energy gap of 4.514 eV. MDS indicated that all tested ligands caused changes in bonding and affected the structural conformation of cyt b, as observed before MDS (0 ns) and after MDS (100 ns). The most notable differences were observed in the types of hydrogen bonds between 0 and 100 ns. Nevertheles, MDS results from a 100 ns simulation revealed consistent behavior for kaempferol across various parameters including root mean square deviation (RMSD), root mean square fluctuation (RMSF), radius of gyration (Rg), solvent-accessible surface area (SASA), molecular mechanics-Poisson Boltzmann surface area (MM-PBSA), and hydrogen bonds. The cyt b-kaempferol complex demonstrated favorable energy stability, as supported by the internal energy distribution values observed in principal component analysis (PCA), which closely resembled those of the atovaquone control. Additionally, trajectory stability analysis indicated structural stability, with a cumulative eigenvalue of 24.7 %. Dynamic cross-correlation matrix (DCCM) analysis revealed a positive correlation among catalytic cytochrome residues within the amino acid residues range 119-268. The results of our research indicate that the structure of kaempferol holds promise as a potential candidate against Plasmodium.
Collapse
Affiliation(s)
- Gusnia Meilin Gholam
- Department of Biochemistry, Faculty of Mathematics and Natural Sciences, Bogor Agricultural University, Dramaga Campus, Bogor 16680, Indonesia; Bioinformatics Research Center, Indonesian Institute of Bioinformatics (INBIO Indonesia), Malang, East Java, 65145, Indonesia.
| | - Fachrur Rizal Mahendra
- Department of Biochemistry, Faculty of Mathematics and Natural Sciences, Bogor Agricultural University, Dramaga Campus, Bogor 16680, Indonesia; Bioinformatics Research Center, Indonesian Institute of Bioinformatics (INBIO Indonesia), Malang, East Java, 65145, Indonesia.
| | - Riyan Alifbi Putera Irsal
- Department of Biochemistry, Faculty of Mathematics and Natural Sciences, Bogor Agricultural University, Dramaga Campus, Bogor 16680, Indonesia.
| | - Maheswari Alfira Dwicesaria
- Department of Biochemistry, Faculty of Mathematics and Natural Sciences, Bogor Agricultural University, Dramaga Campus, Bogor 16680, Indonesia; Bioinformatics Research Center, Indonesian Institute of Bioinformatics (INBIO Indonesia), Malang, East Java, 65145, Indonesia.
| | - Mokhamat Ariefin
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, University of Palangka Raya, Indonesia.
| | - Mikael Kristiadi
- Department of Biochemistry, Faculty of Mathematics and Natural Sciences, Bogor Agricultural University, Dramaga Campus, Bogor 16680, Indonesia.
| | - Andita Fitri Mutiara Rizki
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Jalan Raya Bogor Km. 46, Cibinong, Bogor 16911, Indonesia.
| | - Wihda Aisarul Azmi
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Jalan Raya Bogor Km. 46, Cibinong, Bogor 16911, Indonesia.
| | - I Made Artika
- Department of Biochemistry, Faculty of Mathematics and Natural Sciences, Bogor Agricultural University, Dramaga Campus, Bogor 16680, Indonesia.
| | - Josephine Elizabeth Siregar
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Jalan Raya Bogor Km. 46, Cibinong, Bogor 16911, Indonesia.
| |
Collapse
|
6
|
Daniel L, Karam A, Franco CHJ, Conde C, Sacramento de Morais A, Mosnier J, Fonta I, Villarreal W, Pradines B, Moreira DRM, Navarro M. Metal(triphenylphosphine)-atovaquone Complexes: Synthesis, Antimalarial Activity, and Suppression of Heme Detoxification. Inorg Chem 2024; 63:17087-17099. [PMID: 39185932 PMCID: PMC11409218 DOI: 10.1021/acs.inorgchem.4c02751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
To ascertain the bioinorganic chemistry of metals conjugated with quinones, the complexes [Ag(ATV)(PPh3)2] (1), [Au(ATV)(PPh3)]·2H2O (2), and [Cu(ATV)(PPh3)2] (3) were synthesized by the coordination of the antimalarial naphthoquinone atovaquone (ATV) to the starting materials [Ag(PPh3)2]NO3, [Au(PPh3)Cl], and [Cu(PPh3)2NO3], respectively. These complexes were characterized by analytical and spectroscopical techniques. X-ray diffraction of single crystals precisely confirmed the coordination mode of ATV to the metals, which was monodentate or bidentate, depending on the metal center. Both coordination modes showed high stability in the solid state and in solution. All three complexes showed negative log D values at pH 5, but at pH 7.4, while complex 2 continued to have a negative log D value, complexes 1 and 3 displayed positive values, indicating a more hydrophilic character. ATV and complexes 1-3 could bind to ferriprotoporphyrin IX (FePPIX); however, only complexes 1-3 could inhibit β-hematin crystal formation. Phenotype-based activity revealed that all three metal complexes are able to inhibit the growth of P. falciparum with potency and selectivity comparable to those of ATV, while the starting materials lack this activity. The outcomes of this chemical design may provide significant insights into structure-activity relationships for the development of new antimalarial agents.
Collapse
Affiliation(s)
- Luana Daniel
- Laboratório de Química Bioinorgânica e Catalise, Departamento Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais 36036-900, Brazil
| | - Arquímedes Karam
- Laboratório de Química Bioinorgânica e Catalise, Departamento Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais 36036-900, Brazil
| | - Chris Hebert J Franco
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, 1049-001, Portugal
| | - Camila Conde
- Laboratório de Química Bioinorgânica e Catalise, Departamento Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais 36036-900, Brazil
| | | | - Joel Mosnier
- Unité Parasitologie et Entomologie, Institut de Recherche Biomédicale des Armées, Marseille, 13005, France
- Aix-Marseille Univ, SSA, AP-HM, RITMES, Marseille, 13005, France
- IHU Méditerranée Infection, 19-21 Boulevard Jean Moulin, Marseille, 13005, France
- Centre National de Référence du Paludisme, Marseille, 13005, France
| | - Isabelle Fonta
- Unité Parasitologie et Entomologie, Institut de Recherche Biomédicale des Armées, Marseille, 13005, France
- Aix-Marseille Univ, SSA, AP-HM, RITMES, Marseille, 13005, France
- IHU Méditerranée Infection, 19-21 Boulevard Jean Moulin, Marseille, 13005, France
- Centre National de Référence du Paludisme, Marseille, 13005, France
| | - Wilmer Villarreal
- Grupo de Química Inorgânica Medicinal e Reações Aplicadas, Instituto de Química, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 91501-970, Brazil
| | - Bruno Pradines
- Unité Parasitologie et Entomologie, Institut de Recherche Biomédicale des Armées, Marseille, 13005, France
- Aix-Marseille Univ, SSA, AP-HM, RITMES, Marseille, 13005, France
- IHU Méditerranée Infection, 19-21 Boulevard Jean Moulin, Marseille, 13005, France
- Centre National de Référence du Paludisme, Marseille, 13005, France
| | | | - Maribel Navarro
- Laboratório de Química Bioinorgânica e Catalise, Departamento Química, Instituto de Ciências Exatas, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais 36036-900, Brazil
| |
Collapse
|
7
|
Schäfer TM, Pessanha de Carvalho L, Inoue J, Kreidenweiss A, Held J. The problem of antimalarial resistance and its implications for drug discovery. Expert Opin Drug Discov 2024; 19:209-224. [PMID: 38108082 DOI: 10.1080/17460441.2023.2284820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/14/2023] [Indexed: 12/19/2023]
Abstract
INTRODUCTION Malaria remains a devastating infectious disease with hundreds of thousands of casualties each year. Antimalarial drug resistance has been a threat to malaria control and elimination for many decades and is still of concern today. Despite the continued effectiveness of current first-line treatments, namely artemisinin-based combination therapies, the emergence of drug-resistant parasites in Southeast Asia and even more alarmingly the occurrence of resistance mutations in Africa is of great concern and requires immediate attention. AREAS COVERED A comprehensive overview of the mechanisms underlying the acquisition of drug resistance in Plasmodium falciparum is given. Understanding these processes provides valuable insights that can be harnessed for the development and selection of novel antimalarials with reduced resistance potential. Additionally, strategies to mitigate resistance to antimalarial compounds on the short term by using approved drugs are discussed. EXPERT OPINION While employing strategies that utilize already approved drugs may offer a prompt and cost-effective approach to counter antimalarial drug resistance, it is crucial to recognize that only continuous efforts into the development of novel antimalarial drugs can ensure the successful treatment of malaria in the future. Incorporating resistance propensity assessment during this developmental process will increase the likelihood of effective and enduring malaria treatments.
Collapse
Affiliation(s)
| | | | - Juliana Inoue
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Andrea Kreidenweiss
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- German Center for Infection Research (DZIF), Tübingen, Germany
| | - Jana Held
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- German Center for Infection Research (DZIF), Tübingen, Germany
| |
Collapse
|
8
|
Huang L, Sun Y, Huo DD, Xu M, Xia LY, Yang N, Hong W, Huang L, Nie WM, Liao RH, Zhang MZ, Zhu DY, Li Y, Ma HC, Zhang X, Li YG, Huang XA, Wang JY, Cao WC, Wang FS, Jiang JF. Successful treatment with doxycycline monotherapy for human infection with Babesia venatorum (Babesiidae, Sporozoa) in China: a case report and proposal for a clinical regimen. Infect Dis Poverty 2023; 12:67. [PMID: 37443058 PMCID: PMC10339522 DOI: 10.1186/s40249-023-01111-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/31/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND Human babesiosis is a worldwide disease caused by intraerythrocytic protozoa of the genus Babesia. It is transmitted by bites from ixodid ticks, and mechanically transmitted by blood transfusion. It is primarily treated with quinine and/or atovaquone, which are not readily available in China. In this study, we developed a novel treatment regimen involving doxycycline monotherapy in a patient with severe Babesia venatorum infection as an alternative therapeutic medication. The aim of our study is to provide a guidance for clinical practice treatment of human babesiosis. CASE PRESENTATION A 73-year-old man who had undergone splenectomy and blood transfusion 8 years prior, presented with an unexplained fever, headache, and thrombocytopenia, and was admitted to the Fifth Medical Center of the PLA General Hospital. He was diagnosed with B. venatorum infection by morphological review of thin peripheral blood smears, which was confirmed by multi-gene polymerase chain reaction (PCR), and sequencing of the entire 18s rRNA and partial β-tubulin encoding genes, as well as isolation by animal inoculation. The doxycycline monotherapy regimen (peros, 0.1 g bisindie) was administered following pharmacological guidance and an effective outcome was observed. The patient recovered rapidly following the doxycycline monotherapy. The protozoan load in peripheral blood samples decreased by 88% in hematocrit counts after 8 days, and negative PCR results were obtained after 90 days of follow-up at the hospital. The treatment lasted for 3 months without any side effects or sequelae. The nine-month follow-up survey of the patient did not reveal any signs of recrudescence or anti-babesial tolerance. CONCLUSIONS We have reported a clinical case of successful doxycycline monotherapy for human babesiosis caused by B. venatorum, which provides an optional medical intervention for human babesiosis.
Collapse
Affiliation(s)
- Lei Huang
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, People's Republic of China
| | - Yi Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, People's Republic of China
| | - Dan-Dan Huo
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, People's Republic of China
| | - Ming Xu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, People's Republic of China
- Inner Mongolia Medical University, Hohhot, 010059, People's Republic of China
| | - Luo-Yuan Xia
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, People's Republic of China
- School of Public Health, Shandong University, Jinan, 250100, People's Republic of China
| | - Ning Yang
- The Center for Clinical Laboratory, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, People's Republic of China
| | - Wei Hong
- The Center for Clinical Laboratory, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, People's Republic of China
| | - Lin Huang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, People's Republic of China
| | - Wei-Min Nie
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, People's Republic of China
| | - Ru-He Liao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, People's Republic of China
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510000, People's Republic of China
| | - Ming-Zhu Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, People's Republic of China
| | - Dai-Yun Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, People's Republic of China
| | - Yan Li
- The Center for Clinical Laboratory, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, People's Republic of China
| | - He-Cheng Ma
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, People's Republic of China
| | - Xin Zhang
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, People's Republic of China
| | - Yong-Gang Li
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, People's Republic of China
| | - Xin-An Huang
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510000, People's Republic of China
| | - Jing-Yuan Wang
- School of Public Health, Shandong University, Jinan, 250100, People's Republic of China
| | - Wu-Chun Cao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, People's Republic of China.
- School of Public Health, Shandong University, Jinan, 250100, People's Republic of China.
| | - Fu-Sheng Wang
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, People's Republic of China.
| | - Jia-Fu Jiang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, People's Republic of China.
| |
Collapse
|
9
|
Gujjari L, Kalani H, Pindiprolu SK, Arakareddy BP, Yadagiri G. Current challenges and nanotechnology-based pharmaceutical strategies for the treatment and control of malaria. Parasite Epidemiol Control 2022; 17:e00244. [PMID: 35243049 PMCID: PMC8866151 DOI: 10.1016/j.parepi.2022.e00244] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/12/2021] [Accepted: 02/13/2022] [Indexed: 12/19/2022] Open
Abstract
Malaria is one of the prevalent tropical diseases caused by the parasitic protozoan of the genus Plasmodium spp. With an estimated 228 million cases, it is a major public health concern with high incidence of morbidity and mortality worldwide. The emergence of drug-resistant parasites, inadequate vector control measures, and the non-availability of effective vaccine(s) against malaria pose a serious challenge to malaria eradication especially in underdeveloped and developing countries. Malaria treatment and control comprehensively relies on chemical compounds, which encompass various complications, including severe toxic effects, emergence of drug resistance, and high cost of therapy. To overcome the clinical failures of anti-malarial chemotherapy, a new drug development is of an immediate need. However, the drug discovery and development process is expensive and time consuming. In such a scenario, nanotechnological strategies may offer promising alternative approach for the treatment and control of malaria, with improved efficacy and safety. Nanotechnology based formulations of existing anti-malarial chemotherapeutic agents prove to exceed the limitations of existing therapies in relation to optimum therapeutic benefits, safety, and cost effectiveness, which indeed advances the patient's compliance in treatment. In this review, the shortcomings of malaria therapeutics and necessity of nanotechnological strategies for treating malaria were discussed.
Collapse
Affiliation(s)
- Lohitha Gujjari
- Centre of Infectious Diseases, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S. A. S. Nagar, Punjab 160 062, India
- Department of Entomology, The Ohio State University, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691, USA
| | - Hamed Kalani
- Infectious Diseases Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Sai Kiran Pindiprolu
- Department of Pharmacology, School of Pharmaceutical Sciences and Technologies, Jawaharlal Nehru Technological University, Kakinada, Andhra Pradesh 533003, India
| | | | - Ganesh Yadagiri
- Department of Pharmacology, School of Pharmaceutical Sciences and Technologies, Jawaharlal Nehru Technological University, Kakinada, Andhra Pradesh 533003, India
- Centre for Food Animal Health, The Ohio State University, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH 44691, USA
| |
Collapse
|
10
|
Simwela NV, Waters AP. Current status of experimental models for the study of malaria. Parasitology 2022; 149:1-22. [PMID: 35357277 PMCID: PMC9378029 DOI: 10.1017/s0031182021002134] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 01/09/2023]
Abstract
Infection by malaria parasites (Plasmodium spp.) remains one of the leading causes of morbidity and mortality, especially in tropical regions of the world. Despite the availability of malaria control tools such as integrated vector management and effective therapeutics, these measures have been continuously undermined by the emergence of vector resistance to insecticides or parasite resistance to frontline antimalarial drugs. Whilst the recent pilot implementation of the RTS,S malaria vaccine is indeed a remarkable feat, highly effective vaccines against malaria remain elusive. The barriers to effective vaccines result from the complexity of both the malaria parasite lifecycle and the parasite as an organism itself with consequent major gaps in our understanding of their biology. Historically and due to the practical and ethical difficulties of working with human malaria infections, research into malaria parasite biology has been extensively facilitated by animal models. Animals have been used to study disease pathogenesis, host immune responses and their (dys)regulation and further disease processes such as transmission. Moreover, animal models remain at the forefront of pre-clinical evaluations of antimalarial drugs (drug efficacy, mode of action, mode of resistance) and vaccines. In this review, we discuss commonly used animal models of malaria, the parasite species used and their advantages and limitations which hinder their extrapolation to actual human disease. We also place into this context the most recent developments such as organoid technologies and humanized mice.
Collapse
Affiliation(s)
- Nelson V. Simwela
- Institute of Infection, Immunity & Inflammation, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| | - Andrew P. Waters
- Institute of Infection, Immunity & Inflammation, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| |
Collapse
|
11
|
Usey MM, Huet D. Parasite powerhouse: A review of the Toxoplasma gondii mitochondrion. J Eukaryot Microbiol 2022; 69:e12906. [PMID: 35315174 PMCID: PMC9490983 DOI: 10.1111/jeu.12906] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Toxoplasma gondii is a member of the apicomplexan phylum, a group of single-celled eukaryotic parasites that cause significant human morbidity and mortality around the world. T. gondii harbors two organelles of endosymbiotic origin: a non-photosynthetic plastid, known as the apicoplast, and a single mitochondrion derived from the ancient engulfment of an α-proteobacterium. Due to excitement surrounding the novelty of the apicoplast, the T. gondii mitochondrion was, to a certain extent, overlooked for about two decades. However, recent work has illustrated that the mitochondrion is an essential hub of apicomplexan-specific biology. Development of novel techniques, such as cryo-electron microscopy, complexome profiling, and next-generation sequencing have led to a renaissance in mitochondrial studies. This review will cover what is currently known about key features of the T. gondii mitochondrion, ranging from its genome to protein import machinery and biochemical pathways. Particular focus will be given to mitochondrial features that diverge significantly from the mammalian host, along with discussion of this important organelle as a drug target.
Collapse
Affiliation(s)
- Madelaine M. Usey
- Department of Cellular BiologyUniversity of GeorgiaAthensGeorgiaUSA,Center for Tropical and Emerging Global DiseasesUniversity of GeorgiaAthensGeorgiaUSA
| | - Diego Huet
- Center for Tropical and Emerging Global DiseasesUniversity of GeorgiaAthensGeorgiaUSA,Department of Pharmaceutical and Biomedical SciencesUniversity of GeorgiaAthensGeorgiaUSA
| |
Collapse
|
12
|
Tuvshintulga B, Sivakumar T, Nugraha AB, Ahedor B, Batmagnai E, Otgonsuren D, Liu MM, Xuan X, Igarashi I, Yokoyama N. A combination of clofazimine‒atovaquone as a potent therapeutic regimen for the radical cure of Babesia microti infection in immunocompromised hosts. J Infect Dis 2021; 225:238-242. [PMID: 34664651 DOI: 10.1093/infdis/jiab537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/18/2021] [Indexed: 12/21/2022] Open
Abstract
Human babesiosis caused by Babesia microti can be fatal in immunocompromised patients, and the currently used drugs are often ineffective. A recent study found that clofazimine clears B. microti Munich strain in immunocompromised mice. In the present study, we investigated the efficacies of clofazimine and two-drug combinations involving clofazimine, atovaquone, and azithromycin against B. microti Peabody mjr strain in immunocompromised mice. Treatment with clofazimine alone, clofazimine plus azithromycin, and atovaquone plus azithromycin was ineffective and failed to eliminate the parasites completely, while a 44-day treatment with clofazimine plus atovaquone was highly effective and resulted in a radical cure.
Collapse
Affiliation(s)
- Bumduuren Tuvshintulga
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Thillaiampalam Sivakumar
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Arifin Budiman Nugraha
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Believe Ahedor
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Enkhbaatar Batmagnai
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Davaajav Otgonsuren
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Ming Ming Liu
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Xuenan Xuan
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Ikuo Igarashi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Naoaki Yokoyama
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| |
Collapse
|
13
|
Renard I, Ben Mamoun C. Treatment of Human Babesiosis: Then and Now. Pathogens 2021; 10:pathogens10091120. [PMID: 34578153 PMCID: PMC8469882 DOI: 10.3390/pathogens10091120] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/23/2021] [Accepted: 08/27/2021] [Indexed: 12/26/2022] Open
Abstract
Babesiosis is an emerging tick-borne disease caused by apicomplexan parasites of the genus Babesia. With its increasing incidence worldwide and the risk of human-to-human transmission through blood transfusion, babesiosis is becoming a rising public health concern. The current arsenal for the treatment of human babesiosis is limited and consists of combinations of atovaquone and azithromycin or clindamycin and quinine. These combination therapies were not designed based on biological criteria unique to Babesia parasites, but were rather repurposed based on their well-established efficacy against other apicomplexan parasites. However, these compounds are associated with mild or severe adverse events and a rapid emergence of drug resistance, thus highlighting the need for new therapeutic strategies that are specifically tailored to Babesia parasites. Herein, we review ongoing babesiosis therapeutic and management strategies and their limitations, and further review current efforts to develop new, effective, and safer therapies for the treatment of this disease.
Collapse
|
14
|
Amrane D, Primas N, Arnold CS, Hutter S, Louis B, Sanz-Serrano J, Azqueta A, Amanzougaghene N, Tajeri S, Mazier D, Verhaeghe P, Azas N, Botté C, Vanelle P. Antiplasmodial 2-thiophenoxy-3-trichloromethyl quinoxalines target the apicoplast of Plasmodium falciparum. Eur J Med Chem 2021; 224:113722. [PMID: 34364164 DOI: 10.1016/j.ejmech.2021.113722] [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: 06/16/2021] [Revised: 07/22/2021] [Accepted: 07/25/2021] [Indexed: 10/20/2022]
Abstract
The identification of a plant-like Achille's Heel relict, i.e. the apicoplast, that is essential for Plasmodium spp., the causative agent of malaria lead to an attractive drug target for new antimalarials with original mechanism of action. Although it is not photosynthetic, the apicoplast retains several anabolic pathways that are indispensable for the parasite. Based on previously identified antiplasmodial hit-molecules belonging to the 2-trichloromethylquinazoline and 3-trichloromethylquinoxaline series, we report herein an antiplasmodial Structure-Activity Relationships (SAR) study at position two of the quinoxaline ring of 16 newly synthesized compounds. Evaluation of their activity toward the multi-resistant K1 Plasmodium falciparum strain and cytotoxicity on the human hepatocyte HepG2 cell line revealed a hit compound (3k) with a PfK1 EC50 value of 0.3 μM and a HepG2 CC50 value of 56.0 μM (selectivity index = 175). Moreover, hit-compound 3k was not cytotoxic on VERO or CHO cell lines and was not genotoxic in the in vitro comet assay. Activity cliffs were observed when the trichloromethyl group was replaced by CH3, CF3 or H, showing that this group played a key role in the antiplasmodial activity. Biological investigations performed to determine the target and mechanism of action of the compound 3k strongly suggest that the apicoplast is the putative target as showed by severe alteration of apicoplaste biogenesis and delayed death response. Considering that there are very few molecules that affect the Plasmodium apicoplast, our work provides, for the first time, evidence of the biological target of trichloromethylated derivatives.
Collapse
Affiliation(s)
- Dyhia Amrane
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, 13385, Marseille Cedex 05, France
| | - Nicolas Primas
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, 13385, Marseille Cedex 05, France; APHM, Hôpital Conception, Service Central de la Qualité et de l'Information Pharmaceutiques, 13005, Marseille, France.
| | | | - Sébastien Hutter
- Aix Marseille Univ, IHU Méditerranée Infection, UMR VITROME, IRD, SSA, Mycology & Tropical Eucaryotic Pathogens, 13005, Marseille Cedex 05, France
| | - Béatrice Louis
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, 13385, Marseille Cedex 05, France
| | - Julen Sanz-Serrano
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Nutrition, University of Navarra, C/ Irunlarrea 1, CP 31008, Pamplona, Navarra, Spain
| | - Amaya Azqueta
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Nutrition, University of Navarra, C/ Irunlarrea 1, CP 31008, Pamplona, Navarra, Spain; Navarra Institute for Health Research, IdiSNA, Irunlarrea 3, 31008, Pamplona, Spain
| | - Nadia Amanzougaghene
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI, 75013, Paris, France
| | - Shahin Tajeri
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI, 75013, Paris, France
| | - Dominique Mazier
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI, 75013, Paris, France
| | - Pierre Verhaeghe
- LCC-CNRS Université de Toulouse, CNRS, UPS, 31400, Toulouse, France; CHU de Toulouse, Service Pharmacie, 330 Avenue de Grande-Bretagne, 31059, Toulouse Cedex 9, France
| | - Nadine Azas
- Aix Marseille Univ, IHU Méditerranée Infection, UMR VITROME, IRD, SSA, Mycology & Tropical Eucaryotic Pathogens, 13005, Marseille Cedex 05, France
| | - Cyrille Botté
- ApicoLipid Team, Institute for Advanced Biosciences, Université Grenoble Alpes, La Tronche, France.
| | - Patrice Vanelle
- Aix Marseille Univ, CNRS, ICR UMR 7273, Equipe Pharmaco-Chimie Radicalaire, Faculté de Pharmacie, 13385, Marseille Cedex 05, France; APHM, Hôpital Conception, Service Central de la Qualité et de l'Information Pharmaceutiques, 13005, Marseille, France.
| |
Collapse
|
15
|
Chiu JE, Renard I, George S, Pal AC, Alday PH, Narasimhan S, Riscoe MK, Doggett JS, Ben Mamoun C. Cytochrome b Drug Resistance Mutation Decreases Babesia Fitness in the Tick Stages But Not the Mammalian Erythrocytic Cycle. J Infect Dis 2021; 225:135-145. [PMID: 34139755 PMCID: PMC8730496 DOI: 10.1093/infdis/jiab321] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/15/2021] [Indexed: 12/28/2022] Open
Abstract
Human babesiosis is an emerging tick-borne malaria-like illness caused by Babesia parasites following their development in erythrocytes. Here, we show that a mutation in the Babesia microti mitochondrial cytochrome b (Cytb) that confers resistance to the antibabesial drug ELQ-502 decreases parasite fitness in the arthropod vector. Interestingly, whereas the mutant allele does not affect B. microti fitness during the mammalian blood phase of the parasite life cycle and is genetically stable as parasite burden increases, ELQ-502-resistant mutant parasites developing in the tick vector are genetically unstable with a high rate of the wild-type allele emerging during the nymphal stage. Furthermore, we show that B. microti parasites with this mutation are transmitted from the tick to the host, raising the possibility that the frequency of Cytb resistance mutations may be decreased by passage through the tick vector, but could persist in the environment if present when ticks feed.
Collapse
Affiliation(s)
- Joy E Chiu
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Isaline Renard
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Santosh George
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Anasuya C Pal
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Sukanya Narasimhan
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | - Choukri Ben Mamoun
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Correspondence: Choukri Ben Mamoun, PhD, Yale School of Medicine, Departments of Medicine and Microbial Pathogenesis, Section of Infectious Diseases, 300 Cedar Street, New Haven, CT 06520 ()
| |
Collapse
|
16
|
Exploring Ubiquinone Biosynthesis Inhibition as a Strategy for Improving Atovaquone Efficacy in Malaria. Antimicrob Agents Chemother 2021; 65:AAC.01516-20. [PMID: 33495230 DOI: 10.1128/aac.01516-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 01/16/2021] [Indexed: 11/20/2022] Open
Abstract
Atovaquone (AV) acts on the malaria parasite by competing with ubiquinol (UQH2) for its union to the mitochondrial bc1 complex, preventing the ubiquinone-8 and ubiquinone-9 (UQ-8 and UQ-9) redox recycling, which is a necessary step in pyrimidine biosynthesis. This study focused on UQ biosynthesis in Plasmodium falciparum and adopted proof-of-concept research to better elucidate the mechanism of action of AV and improve its efficacy. Initially, UQ biosynthesis was evaluated using several radioactive precursors and chromatographic techniques. This methodology was suitable for studying the biosynthesis of both UQ homologs and its redox state. Additionally, the composition of UQ was investigated in parasites cultivated at different oxygen saturations or in the presence of AV. AV affected the redox states of both UQ-8 and UQ-9 homologs by increasing the levels of the respective reduced forms. Conversely, low-oxygen environments specifically inhibited UQ-9 biosynthesis and increased the antimalarial efficacy of AV. These findings encouraged us to investigate the biological importance and the potential of UQ biosynthesis as a drug target based on its inhibition by 4-nitrobenzoate (4-NB), a 4-hydroxybenzoate (4-HB) analog. 4-NB effectively inhibits UQ biosynthesis and enhances the effects of AV on parasitic growth and respiration rate. Although 4-NB itself exhibits poor antimalarial activity, its 50% inhibitory concentration (IC50) value increased significantly in the presence of a soluble UQ analog, p-aminobenzoic acid (pABA), or 4-HB. These results indicate the potential of AV combined with 4-NB as a novel therapy for malaria and other diseases caused by AV-sensitive pathogens.
Collapse
|
17
|
Cortez-Maya S, Moreno-Herrera A, Palos I, Rivera G. Old Antiprotozoal Drugs: Are They Still Viable Options for Parasitic Infections or New Options for Other Diseases? Curr Med Chem 2020; 27:5403-5428. [DOI: 10.2174/0929867326666190628163633] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 05/31/2019] [Accepted: 06/10/2019] [Indexed: 01/16/2023]
Abstract
Parasitic diseases, caused by helminths (ascariasis, hookworm, trichinosis, and schistosomiasis)
and protozoa (chagas, leishmaniasis, and amebiasis), are considered a serious public
health problem in developing countries. Additionally, there is a limited arsenal of anti-parasitic
drugs in the current pipeline and growing drug resistance. Therefore, there is a clear need for the
discovery and development of new compounds that can compete and replace these drugs that have
been controlling parasitic infections over the last decades. However, this approach is highly resource-
intensive, expensive and time-consuming. Accordingly, a drug repositioning strategy of the
existing drugs or drug-like molecules with known pharmacokinetics and safety profiles is alternatively
being used as a fast approach towards the identification of new treatments. The artemisinins,
mefloquine, tribendimidine, oxantel pamoate and doxycycline for the treatment of helminths, and
posaconazole and hydroxymethylnitrofurazone for the treatment of protozoa are promising candidates.
Therefore, traditional antiprotozoal drugs, which were developed in some cases decades ago,
are a valid solution. Herein, we review the current status of traditional anti-helminthic and antiprotozoal
drugs in terms of drug targets, mode of action, doses, adverse effects, and parasite resistance
to define their suitability for repurposing strategies. Current antiparasitic drugs are not only
still viable for the treatment of helminth and protozoan infections but are also important candidates
for new pharmacological treatments.
Collapse
Affiliation(s)
- Sandra Cortez-Maya
- Instituto de Quimica, Universidad Nacional Autonoma de Mexico, Cd. Universitaria, Circuito Exterior, Coyoacan, 04510 Ciudad de Mexico, Mexico
| | - Antonio Moreno-Herrera
- Laboratorio de Biotecnologia Farmaceutica, Centro de Biotecnologia Genomica, Instituto Politecnico Nacional, 88710 Reynosa, Mexico
| | - Isidro Palos
- Unidad AcadEmica Multidisciplinaria Reynosa-Rodhe, Universidad AutOnoma de Tamaulipas, 88710 Reynosa, Mexico
| | - Gildardo Rivera
- Laboratorio de Biotecnologia Farmaceutica, Centro de Biotecnologia Genomica, Instituto Politecnico Nacional, 88710 Reynosa, Mexico
| |
Collapse
|
18
|
Chugh A, Kumar A, Verma A, Kumar S, Kumar P. A review of antimalarial activity of two or three nitrogen atoms containing heterocyclic compounds. Med Chem Res 2020. [DOI: 10.1007/s00044-020-02604-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
19
|
Ling L, Mulaka M, Munro J, Dass S, Mather MW, Riscoe MK, Llinás M, Zhou J, Ke H. Genetic ablation of the mitoribosome in the malaria parasite Plasmodium falciparum sensitizes it to antimalarials that target mitochondrial functions. J Biol Chem 2020; 295:7235-7248. [PMID: 32273345 PMCID: PMC7247301 DOI: 10.1074/jbc.ra120.012646] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 04/04/2020] [Indexed: 02/05/2023] Open
Abstract
The mitochondrion of malaria parasites contains several clinically validated drug targets. Within Plasmodium spp., the causative agents of malaria, the mitochondrial DNA (mtDNA) is only 6 kb long, being the smallest mitochondrial genome among all eukaryotes. The mtDNA encodes only three proteins of the mitochondrial electron transport chain and ∼27 small, fragmented rRNA genes having lengths of 22-195 nucleotides. The rRNA fragments are thought to form a mitochondrial ribosome (mitoribosome), together with ribosomal proteins imported from the cytosol. The mitoribosome of Plasmodium falciparum is essential for maintenance of the mitochondrial membrane potential and parasite viability. However, the role of the mitoribosome in sustaining the metabolic status of the parasite mitochondrion remains unclear. The small ribosomal subunit in P. falciparum has 14 annotated mitoribosomal proteins, and employing a CRISPR/Cas9-based conditional knockdown tool, here we verified the location and tested the essentiality of three candidates (PfmtRPS12, PfmtRPS17, and PfmtRPS18). Using immuno-EM, we provide evidence that the P. falciparum mitoribosome is closely associated with the mitochondrial inner membrane. Upon knockdown of the mitoribosome, parasites became hypersensitive to inhibitors targeting mitochondrial Complex III (bc1), dihydroorotate dehydrogenase (DHOD), and the F1F0-ATP synthase complex. Furthermore, the mitoribosome knockdown blocked the pyrimidine biosynthesis pathway and reduced the cellular pool of pyrimidine nucleotides. These results suggest that disruption of the P. falciparum mitoribosome compromises the metabolic capacity of the mitochondrion, rendering the parasite hypersensitive to a panel of inhibitors that target mitochondrial functions.
Collapse
Affiliation(s)
- Liqin Ling
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129; Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Maruthi Mulaka
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129
| | - Justin Munro
- Department of Chemistry and Huck Center for Malaria Research, Pennsylvania State University, University Park, Pennsylvania 16802
| | - Swati Dass
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129
| | - Michael W Mather
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129
| | - Michael K Riscoe
- Portland Veterans Affairs Medical Center, Portland, Oregon 97239
| | - Manuel Llinás
- Department of Chemistry and Huck Center for Malaria Research, Pennsylvania State University, University Park, Pennsylvania 16802; Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania 16802
| | - Jing Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Hangjun Ke
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129.
| |
Collapse
|
20
|
Smilkstein MJ, Pou S, Krollenbrock A, Bleyle LA, Dodean RA, Frueh L, Hinrichs DJ, Li Y, Martinson T, Munar MY, Winter RW, Bruzual I, Whiteside S, Nilsen A, Koop DR, Kelly JX, Kappe SHI, Wilder BK, Riscoe MK. ELQ-331 as a prototype for extremely durable chemoprotection against malaria. Malar J 2019; 18:291. [PMID: 31455339 PMCID: PMC6712883 DOI: 10.1186/s12936-019-2921-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 08/17/2019] [Indexed: 12/02/2022] Open
Abstract
Background The potential benefits of long-acting injectable chemoprotection (LAI-C) against malaria have been recently recognized, prompting a call for suitable candidate drugs to help meet this need. On the basis of its known pharmacodynamic and pharmacokinetic profiles after oral dosing, ELQ-331, a prodrug of the parasite mitochondrial electron transport inhibitor ELQ-300, was selected for study of pharmacokinetics and efficacy as LAI-C in mice. Methods Four trials were conducted in which mice were injected with a single intramuscular dose of ELQ-331 or other ELQ-300 prodrugs in sesame oil with 1.2% benzyl alcohol; the ELQ-300 content of the doses ranged from 2.5 to 30 mg/kg. Initial blood stage challenges with Plasmodium yoelii were used to establish the model, but the definitive study measure of efficacy was outcome after sporozoite challenge with a luciferase-expressing P. yoelii, assessed by whole-body live animal imaging. Snapshot determinations of plasma ELQ-300 concentration ([ELQ-300]) were made after all prodrug injections; after the highest dose of ELQ-331 (equivalent to 30 mg/kg ELQ-300), both [ELQ-331] and [ELQ-300] were measured at a series of timepoints from 6 h to 5½ months after injection. Results A single intramuscular injection of ELQ-331 outperformed four other ELQ-300 prodrugs and, at a dose equivalent to 30 mg/kg ELQ-300, protected mice against challenge with P. yoelii sporozoites for at least 4½ months. Pharmacokinetic evaluation revealed rapid and essentially complete conversion of ELQ-331 to ELQ-300, a rapidly achieved (< 6 h) and sustained (4–5 months) effective plasma ELQ-300 concentration, maximum ELQ-300 concentrations far below the estimated threshold for toxicity, and a distinctive ELQ-300 concentration versus time profile. Pharmacokinetic modeling indicates a high-capacity, slow-exchange tissue compartment which serves to accumulate and then slowly redistribute ELQ-300 into blood, and this property facilitates an extremely long period during which ELQ-300 concentration is sustained above a minimum fully-protective threshold (60–80 nM). Conclusions Extrapolation of these results to humans predicts that ELQ-331 should be capable of meeting and far-exceeding currently published duration-of-effect goals for anti-malarial LAI-C. Furthermore, the distinctive pharmacokinetic profile of ELQ-300 after treatment with ELQ-331 may facilitate durable protection and enable protection for far longer than 3 months. These findings suggest that ELQ-331 warrants consideration as a leading prototype for LAI-C.
Collapse
Affiliation(s)
- Martin J Smilkstein
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA.
| | - Sovitj Pou
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA
| | - Alina Krollenbrock
- Department of Physiology and Pharmacology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Lisa A Bleyle
- Bioanalytical Shared Resource Core Pharmacokinetics, Department of Physiology and Pharmacology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, L334, Portland, OR, 97239, USA
| | - Rozalia A Dodean
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA.,Department of Chemistry, Portland State University, PO Box 751, Portland, OR, 97207, USA
| | - Lisa Frueh
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA
| | - David J Hinrichs
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA
| | - Yuexin Li
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA
| | - Thomas Martinson
- Vaccine & Gene Therapy Institute (VGTI), Oregon Health and Science University (West Campus), 505 NW 185th Avenue, #1, Beaverton, OR, 97006, USA
| | - Myrna Y Munar
- Oregon State University/Oregon Health and Science University College of Pharmacy, 2730 SW Moody Avenue, CL5CP, Portland, OR, 97201, USA
| | - Rolf W Winter
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA.,Department of Chemistry, Portland State University, PO Box 751, Portland, OR, 97207, USA
| | - Igor Bruzual
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA
| | - Samantha Whiteside
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Ave N., Suite 500, Seattle, WA, USA
| | - Aaron Nilsen
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA
| | - Dennis R Koop
- Bioanalytical Shared Resource Core Pharmacokinetics, Department of Physiology and Pharmacology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, L334, Portland, OR, 97239, USA
| | - Jane X Kelly
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA.,Department of Chemistry, Portland State University, PO Box 751, Portland, OR, 97207, USA
| | - Stefan H I Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Ave N., Suite 500, Seattle, WA, USA
| | - Brandon K Wilder
- Vaccine & Gene Therapy Institute (VGTI), Oregon Health and Science University (West Campus), 505 NW 185th Avenue, #1, Beaverton, OR, 97006, USA
| | - Michael K Riscoe
- VA Portland Health Care System Research and Development Service, 3710 SW US Veterans Hospital Road, RD-33, Portland, OR, 97239, USA.,Department of Molecular Microbiology and Immunology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| |
Collapse
|
21
|
Bed Nets, Insecticides, and Antimalarials: Where to Next? Trends Parasitol 2019; 35:668-670. [PMID: 31303446 DOI: 10.1016/j.pt.2019.06.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 06/19/2019] [Indexed: 11/22/2022]
Abstract
Insecticide-impregnated bed nets have saved millions from fatal malaria, but their effectiveness is waning due to mosquito insecticide resistance. A new strategy (Paton et al., Nature, 2019) to deliver parasiticidal compounds into mosquitoes to kill transmission-stage parasites could enhance the effectiveness of bed nets and get around the perennial problems of resistance.
Collapse
|
22
|
Nyagwange J, Awino E, Tijhaar E, Svitek N, Pelle R, Nene V. Leveraging the Medicines for Malaria Venture malaria and pathogen boxes to discover chemical inhibitors of East Coast fever. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2019; 9:80-86. [PMID: 30771616 PMCID: PMC6376154 DOI: 10.1016/j.ijpddr.2019.01.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/14/2019] [Accepted: 01/14/2019] [Indexed: 11/17/2022]
Abstract
Chemotherapy of East Coast fever, a lymphoproliferative cancer-like disease of cattle causing significant economic losses in Africa, is largely dependent on the use of buparvaquone, a drug that was developed in the late 1980's. The disease is caused by the tick-borne protozoan pathogen Theileria parva. Buparvaquone can be used prophylactically and it is also active against tropical theileriosis, caused by the related parasite Theileria annulata. Recently, drug resistance was reported in T. annulata, and could occur in T. parva. Using a 3H-thymidine incorporation assay we screened 796 open source compounds from the Medicines for Malaria Venture (MMV) to discover novel chemicals with potential inhibitory activity to T. parva. We identified nine malaria box compounds and eight pathogen box compounds that inhibited the proliferation of F100TpM, a T. parva infected lymphocyte cell line. However, only two compounds, MMV008212 and MMV688372 represent promising leads with IC50 values of 0.78 and 0.61 μM, respectively, and CC50 values > 5 μM. The remaining compounds exhibited a high degree of toxicity (CC50 values < 1.09 μM) on the proliferation of bovine peripheral blood mononuclear cells stimulated with concanavalin A. We also tested the anti-cancer drug, dasatinib, used in the chemotherapy of some leukemias. Dasatinib was as active and safe as buparvaquone in vitro, with an IC50 of 5 and 4.2 nM, respectively, and CC50 > 10 μM. Our preliminary data suggest that it may be possible to repurpose compounds from the cancer field as well as MMV as novel anti-T. parva molecules. 17 of 796 MMV compounds inhibited proliferation of T parva infected lymphocytes. 15 of the 17 hit compounds also inhibited proliferation of control cells. MMV008212 and MMV688372 represent promising leads, therapeutic index >8. Dasatinib was as active and safe as buparvaquone in vitro, therapeutic index >2000.
Collapse
Affiliation(s)
- James Nyagwange
- International Livestock Research Institute, P. O. Box 30709, Nairobi, Kenya; Cell Biology and Immunology Group, Wageningen University, the Netherlands
| | - Elias Awino
- International Livestock Research Institute, P. O. Box 30709, Nairobi, Kenya
| | - Edwin Tijhaar
- Cell Biology and Immunology Group, Wageningen University, the Netherlands
| | - Nicholas Svitek
- International Livestock Research Institute, P. O. Box 30709, Nairobi, Kenya
| | - Roger Pelle
- Biosciences Eastern and Central Africa, International Livestock Research Institute (BecA-ILRI) Hub, P. O. Box 30709, Nairobi, Kenya
| | - Vishvanath Nene
- International Livestock Research Institute, P. O. Box 30709, Nairobi, Kenya.
| |
Collapse
|
23
|
Runtuwene LR, Tuda JSB, Mongan AE, Makalowski W, Frith MC, Imwong M, Srisutham S, Nguyen Thi LA, Tuan NN, Eshita Y, Maeda R, Yamagishi J, Suzuki Y. Nanopore sequencing of drug-resistance-associated genes in malaria parasites, Plasmodium falciparum. Sci Rep 2018; 8:8286. [PMID: 29844487 PMCID: PMC5974085 DOI: 10.1038/s41598-018-26334-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 05/10/2018] [Indexed: 11/09/2022] Open
Abstract
Here, we report the application of a portable sequencer, MinION, for genotyping the malaria parasite Plasmodium falciparum. In the present study, an amplicon mixture of nine representative genes causing resistance to anti-malaria drugs is diagnosed. First, we developed the procedure for four laboratory strains (3D7, Dd2, 7G8, and K1), and then applied the developed procedure to ten clinical samples. We sequenced and re-sequenced the samples using the obsolete flow cell R7.3 and the most recent flow cell R9.4. Although the average base-call accuracy of the MinION sequencer was 74.3%, performing >50 reads at a given position improves the accuracy of the SNP call, yielding a precision and recall rate of 0.92 and 0.8, respectively, with flow cell R7.3. These numbers increased significantly with flow cell R9.4, in which the precision and recall are 1 and 0.97, respectively. Based on the SNP information, the drug resistance status in ten clinical samples was inferred. We also analyzed K13 gene mutations from 54 additional clinical samples as a proof of concept. We found that a novel amino-acid changing variation is dominant in this area. In addition, we performed a small population-based analysis using 3 and 5 cases (K13) and 10 and 5 cases (PfCRT) from Thailand and Vietnam, respectively. We identified distinct genotypes from the respective regions. This approach will change the standard methodology for the sequencing diagnosis of malaria parasites, especially in developing countries.
Collapse
Affiliation(s)
- Lucky R Runtuwene
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba, 277-8562, Japan
| | - Josef S B Tuda
- Faculty of Medicine, Sam Ratulangi University, Kampus Unsrat, Bahu Manado, 95115, Indonesia
| | - Arthur E Mongan
- Faculty of Medicine, Sam Ratulangi University, Kampus Unsrat, Bahu Manado, 95115, Indonesia
| | - Wojciech Makalowski
- Institute of Bioinformatics, Faculty of Medicine, University of Münster, Niels-Stensen Strasse 14, Münster, 48149, Germany
| | - Martin C Frith
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba, 277-8562, Japan.,Artificial Intelligence Research Center, Advanced Industrial Science and Technology, 2-3-26, Aomi, Koto-ku, Tokyo, 135-0064, Japan.,AIST-Waseda CBBD-OIL, 3-4-1 Ookubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Mallika Imwong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Thung Phaya Thai, Ratchathewi, Bangkok, 10400, Thailand
| | - Suttipat Srisutham
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Thung Phaya Thai, Ratchathewi, Bangkok, 10400, Thailand
| | - Lan Anh Nguyen Thi
- National Institute of Hygiene and Epidemiology, 1 Yersin Street, Hanoi, 112800, Vietnam
| | - Nghia Nguyen Tuan
- National Institute of Hygiene and Epidemiology, 1 Yersin Street, Hanoi, 112800, Vietnam
| | - Yuki Eshita
- Division of Collaboration and Education, Research Center for Zoonosis Control, Hokkaido University, North 20, West 10 Kita-ku, Sapporo, Hokkaido, 001-0020, Japan.,Department of Medical Entomology, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Thung Phaya Thai, Ratchathewi, Bangkok, 10400, Thailand
| | - Ryuichiro Maeda
- Division of Biomedical Science, Department of Basic Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Nishi 2 Sen-11 Inadacho, Obihiro, Hokkaido, 080-0834, Japan
| | - Junya Yamagishi
- Global Station for Zoonosis Control, GI-CoRE, Hokkaido University, North 20, West 10 Kita-ku, Sapporo, Hokkaido, 001-0020, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba, 277-8562, Japan.
| |
Collapse
|
24
|
Ke H, Dass S, Morrisey JM, Mather MW, Vaidya AB. The mitochondrial ribosomal protein L13 is critical for the structural and functional integrity of the mitochondrion in Plasmodium falciparum. J Biol Chem 2018; 293:8128-8137. [PMID: 29626096 DOI: 10.1074/jbc.ra118.002552] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/02/2018] [Indexed: 12/22/2022] Open
Abstract
The phylum Apicomplexa contains a group of protozoa causing diseases in humans and livestock. Plasmodium spp., the causative agent of malaria, contains a mitochondrion that is very divergent from that of their hosts. The malarial mitochondrion is a clinically validated target for the antimalarial drug atovaquone, which specifically blocks the electron transfer activity of the bc1 complex of the mitochondrial electron transport chain (mtETC). Most mtETC proteins are nuclear-encoded and imported from the cytosol, but three key protein subunits are encoded in the Plasmodium mitochondrial genome: cyt b, COXI, and COXIII. They are translated inside the mitochondrion by mitochondrial ribosomes (mitoribosomes). Here, we characterize the function of one large mitoribosomal protein in Plasmodium falciparum, PfmRPL13. We found that PfmRPL13 localizes to the parasite mitochondrion and is refractory to genetic knockout. Ablation of PfmRPL13 using a conditional knockdown system (TetR-DOZI-aptamer) caused a series of adverse events in the parasite, including mtETC deficiency, loss of mitochondrial membrane potential (Δψm), and death. The PfmRPL13 knockdown parasite also became hypersensitive to proguanil, a drug proposed to target an alternative process for maintaining Δψm Surprisingly, transmission EM revealed that PfmRPL13 disruption also resulted in an unusually elongated and branched mitochondrion. The growth arrest of the knockdown parasite could be rescued with a second copy of PfmRPL13, but not by supplementation with decylubiquinone or addition of a yeast dihydroorotate dehydrogenase gene. In summary, we provide first and direct evidence that mitoribosomes are essential for malaria parasites to maintain the structural and functional integrity of the mitochondrion.
Collapse
Affiliation(s)
- Hangjun Ke
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129.
| | - Swati Dass
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129
| | - Joanne M Morrisey
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129
| | - Michael W Mather
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129
| | - Akhil B Vaidya
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129
| |
Collapse
|
25
|
Atovaquone oral bioavailability enhancement using electrospraying technology. Eur J Pharm Sci 2018; 111:195-204. [PMID: 28974387 DOI: 10.1016/j.ejps.2017.09.051] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 09/21/2017] [Accepted: 09/29/2017] [Indexed: 11/22/2022]
Abstract
Atovaquone in combination with proguanil hydrochloride, marketed as Malarone® tablets by GlaxoSmithKline (GSK), is prescribed for the treatment of malaria. High dose and poor bioavailability are the main hurdles associated with atovaquone oral therapy. The present study reports development of atovaquone nanoparticles, using in house designed and fabricated electrospraying equipment, and the assessment of bioavailability and therapeutic efficacy of the nanoparticles after oral administration. Solid nanoparticles of atovaquone were successfully produced by electrospraying and were characterized for particle size and flow properties. Differential Scanning Calorimetry, X-ray Diffraction, Fourier Transform Infrared Spectroscopy studies were also carried out. Atovaquone nanoparticles along with proguanil hydrochloride and a suitable wetting agent were filled in size 2 hard gelatin capsules. The formulation was compared with Malarone® tablets (GSK) and Mepron® suspension (GSK) in terms of in vitro release profile and in vivo pharmacokinetic studies. It showed 2.9-fold and 1.8-fold improved bioavailability in rats compared to Malarone® tablets and Mepron® suspension respectively. Therapeutic efficacy of the formulation was determined using modified Peter's 4-day suppressive tests and clinical simulation studies using Plasmodium berghei ANKA infected Swiss mice and compared to Malarone®. The developed formulation showed a 128-fold dose reduction in the modified Peter's 4-day suppressive tests and 32-fold dose reduction in clinical simulation studies. Given that only one capsule a day of developed formulation is required to be administered orally compared to 4 Malarone® tablets once a day and that too at a significantly reduced dose, this nanoparticle formulation will definitely reduce the side-effects of the treatment and is also likely to increase patient compliance.
Collapse
|
26
|
Lack of mitochondrial MutS homolog 1 in Toxoplasma gondii disrupts maintenance and fidelity of mitochondrial DNA and reveals metabolic plasticity. PLoS One 2017; 12:e0188040. [PMID: 29141004 PMCID: PMC5687708 DOI: 10.1371/journal.pone.0188040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 10/31/2017] [Indexed: 11/19/2022] Open
Abstract
The importance of maintaining the fidelity of the mitochondrial genome is underscored by the presence of various repair pathways within this organelle. Presumably, the repair of mitochondrial DNA would be of particular importance in organisms that possess only a single mitochondrion, like the human pathogens Plasmodium falciparum and Toxoplasma gondii. Understanding the machinery that maintains mitochondrial DNA in these parasites is of particular relevance, as mitochondrial function is a validated and effective target for anti-parasitic drugs. We previously determined that the Toxoplasma MutS homolog TgMSH1 localizes to the mitochondrion. MutS homologs are key components of the nuclear mismatch repair system in mammalian cells, and both yeast and plants possess MutS homologs that localize to the mitochondria where they regulate DNA stability. Here we show that the lack of TgMSH1 results in accumulation of single nucleotide variations in mitochondrial DNA and a reduction in mitochondrial DNA content. Additionally, parasites lacking TgMSH1 function can survive treatment with the cytochrome b inhibitor atovaquone. While the Tgmsh1 knockout strain has several missense mutations in cytochrome b, none affect amino acids known to be determinants of atovaquone sensitivity and atovaquone is still able to inhibit electron transport in the Tgmsh1 mutants. Furthermore, culture of Tgmsh1 mutant in the presence atovaquone leads to parasites with enhanced atovaquone resistance and complete shutdown of respiration. Thus, parasites lacking TgMSH1 overcome the disruption of mitochondrial DNA by adapting their physiology allowing them to forgo the need for oxidative phosphorylation. Consistent with this idea, the Tgmsh1 mutant is resistant to mitochondrial inhibitors with diverse targets and exhibits reduced ability to grow in the absence of glucose. This work shows TgMSH1 as critical for the maintenance and fidelity of the mitochondrial DNA in Toxoplasma, reveals a novel mechanism for atovaquone resistance, and exposes the physiological plasticity of this important human pathogen.
Collapse
|
27
|
Recent advances in use of silver nanoparticles as antimalarial agents. Int J Pharm 2017; 526:254-270. [PMID: 28450172 DOI: 10.1016/j.ijpharm.2017.04.042] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 04/17/2017] [Accepted: 04/18/2017] [Indexed: 11/23/2022]
Abstract
Malaria is one of the most common infectious diseases, which has become a great public health problem all over the world. Ineffectiveness of available antimalarial treatment is the main reason behind its menace. The failure of current treatment strategies is due to emergence of drug resistance in Plasmodium falciparum and drug toxicity in human beings. Therefore, the development of novel and effective antimalarial drugs is the need of the hour. Considering the huge biomedical applications of nanotechnology, it can be potentially used for the malarial treatment. Silver nanoparticles (AgNPs) have demonstrated significant activity against malarial parasite (P. falciparum) and vector (female Anopheles mosquito). It is believed that AgNPs will be a solution for the control of malaria. This review emphasizes the pros- and cons of existing antimalarial treatments and in depth discussion on application of AgNPs for treatment of malaria. The role of nanoparticles for site specific drug delivery and toxicological issues have also been discussed.
Collapse
|
28
|
Atovaquone and ELQ-300 Combination Therapy as a Novel Dual-Site Cytochrome bc1 Inhibition Strategy for Malaria. Antimicrob Agents Chemother 2016; 60:4853-9. [PMID: 27270285 DOI: 10.1128/aac.00791-16] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 05/24/2016] [Indexed: 12/31/2022] Open
Abstract
Antimalarial combination therapies play a crucial role in preventing the emergence of drug-resistant Plasmodium parasites. Although artemisinin-based combination therapies (ACTs) comprise the majority of these formulations, inhibitors of the mitochondrial cytochrome bc1 complex (cyt bc1) are among the few compounds that are effective for both acute antimalarial treatment and prophylaxis. There are two known sites for inhibition within cyt bc1: atovaquone (ATV) blocks the quinol oxidase (Qo) site of cyt bc1, while some members of the endochin-like quinolone (ELQ) family, including preclinical candidate ELQ-300, inhibit the quinone reductase (Qi) site and retain full potency against ATV-resistant Plasmodium falciparum strains with Qo site mutations. Here, we provide the first in vivo comparison of ATV, ELQ-300, and combination therapy consisting of ATV plus ELQ-300 (ATV:ELQ-300), using P. yoelii murine models of malaria. In our monotherapy assessments, we found that ATV functioned as a single-dose curative compound in suppressive tests whereas ELQ-300 demonstrated a unique cumulative dosing effect that successfully blocked recrudescence even in a high-parasitemia acute infection model. ATV:ELQ-300 therapy was highly synergistic, and the combination was curative with a single combined dose of 1 mg/kg of body weight. Compared to the ATV:proguanil (Malarone) formulation, ATV:ELQ-300 was more efficacious in multiday, acute infection models and was equally effective at blocking the emergence of ATV-resistant parasites. Ultimately, our data suggest that dual-site inhibition of cyt bc1 is a valuable strategy for antimalarial combination therapy and that Qi site inhibitors such as ELQ-300 represent valuable partner drugs for the clinically successful Qo site inhibitor ATV.
Collapse
|
29
|
Lawres LA, Garg A, Kumar V, Bruzual I, Forquer IP, Renard I, Virji AZ, Boulard P, Rodriguez EX, Allen AJ, Pou S, Wegmann KW, Winter RW, Nilsen A, Mao J, Preston DA, Belperron AA, Bockenstedt LK, Hinrichs DJ, Riscoe MK, Doggett JS, Ben Mamoun C. Radical cure of experimental babesiosis in immunodeficient mice using a combination of an endochin-like quinolone and atovaquone. J Exp Med 2016; 213:1307-18. [PMID: 27270894 PMCID: PMC4925016 DOI: 10.1084/jem.20151519] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 04/25/2016] [Indexed: 01/24/2023] Open
Abstract
Human babesiosis is a tick-borne multisystem disease caused by Babesia species of the apicomplexan phylum. Most clinical cases and fatalities of babesiosis are caused by Babesia microti Current treatment for human babesiosis consists of two drug combinations, atovaquone + azithromycin or quinine + clindamycin. These treatments are associated with adverse side effects and a significant rate of drug failure. Here, we provide evidence for radical cure of experimental babesiosis in immunodeficient mice using a combination of an endochin-like quinolone (ELQ) prodrug and atovaquone. In vivo efficacy studies in mice using ELQ-271, ELQ-316, and the ELQ-316 prodrug, ELQ-334, demonstrated excellent growth inhibitory activity against the parasite, with potency equal to that of orally administered atovaquone at 10 mg/kg. Analysis of recrudescent parasites after ELQ or atovaquone monotherapy identified genetic substitutions in the Qi or Qo sites, respectively, of the cytochrome bc1 complex. Impressively, a combination of ELQ-334 and atovaquone, at doses as low as 5.0 mg/kg each, resulted in complete clearance of the parasite with no recrudescence up to 122 d after discontinuation of therapy. These results will set the stage for future clinical evaluation of ELQ and atovaquone combination therapy for treatment of human babesiosis.
Collapse
Affiliation(s)
- Lauren A Lawres
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT 06520
| | - Aprajita Garg
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT 06520
| | - Vidya Kumar
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT 06520
| | - Igor Bruzual
- Veterans Affairs Medical Center, Portland, OR 97239
| | | | - Isaline Renard
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT 06520
| | - Azan Z Virji
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT 06520
| | - Pierre Boulard
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT 06520
| | - Eduardo X Rodriguez
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT 06520
| | - Alexander J Allen
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT 06520
| | - Sovitj Pou
- Veterans Affairs Medical Center, Portland, OR 97239
| | | | | | - Aaron Nilsen
- Veterans Affairs Medical Center, Portland, OR 97239
| | - Jialing Mao
- Department of Internal Medicine, Section of Rheumatology, Yale School of Medicine, New Haven, CT 06520
| | | | - Alexia A Belperron
- Department of Internal Medicine, Section of Rheumatology, Yale School of Medicine, New Haven, CT 06520
| | - Linda K Bockenstedt
- Department of Internal Medicine, Section of Rheumatology, Yale School of Medicine, New Haven, CT 06520
| | | | | | | | - Choukri Ben Mamoun
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT 06520
| |
Collapse
|
30
|
Vincent IM, Barrett MP. Metabolomic-based strategies for anti-parasite drug discovery. ACTA ACUST UNITED AC 2014; 20:44-55. [PMID: 25281738 DOI: 10.1177/1087057114551519] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Metabolomics-based studies are proving of great utility in the analysis of modes of action (MOAs) and resistance mechanisms of drugs in parasitic protozoa. They have helped to determine the MOA of eflornithine, half of the gold standard combination therapy in use against human African trypanosomiasis (HAT), as well as the mechanism of resistance to this drug. In Leishmania, metabolomics has also given insight into the MOA of miltefosine, an alkylphospholipid. Several studies on antimony resistance in Leishmania have been conducted, analyzing the metabolic content of resistant lines, offering clues as to the MOA of this class of drugs. A study of chloroquine resistance in Plasmodium falciparum combined metabolomics techniques with other genetic and proteomic techniques to offer new insight into the role of the PfCRT protein. The MOA and mechanism of resistance to a group of halogenated pyrimidines in Trypanosoma brucei have also recently been elucidated. Effective as metabolomics techniques are, care must be taken in the design and implementation of these experiments, to ensure the resulting data are meaningful. This review outlines the steps required to conduct a metabolomics experiment as well as provide an overview of metabolomics-based drug research in protozoa to date.
Collapse
Affiliation(s)
- Isabel M Vincent
- The Glasgow Polyomics Facility and Wellcome Trust Centre for Molecular Parasitology, University of Glasgow, Glasgow, UK
| | - Michael P Barrett
- The Glasgow Polyomics Facility and Wellcome Trust Centre for Molecular Parasitology, University of Glasgow, Glasgow, UK
| |
Collapse
|
31
|
de Souza NB, de Andrade IM, Carneiro PF, Jardim GAM, de Melo IMM, da Silva EN, Krettli AU. Blood shizonticidal activities of phenazines and naphthoquinoidal compounds against Plasmodium falciparum in vitro and in mice malaria studies. Mem Inst Oswaldo Cruz 2014; 109:546-52. [PMID: 25099332 PMCID: PMC4156448 DOI: 10.1590/0074-0276130603] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 05/05/2014] [Indexed: 11/28/2022] Open
Abstract
Due to the recent advances of atovaquone, a naphthoquinone, through clinical trials as treatment for malarial infection, 19 quinone derivatives with previously reported structures were also evaluated for blood schizonticide activity against the malaria parasite Plasmodium falciparum. These compounds include 2-hydroxy-3-methylamino naphthoquinones (2-9), lapachol (10), nor-lapachol (11), iso-lapachol (12), phthiocol (13) and phenazines (12-20). Their cytotoxicities were also evaluated against human hepatoma and normal monkey kidney cell lines. Compounds 2 and 5 showed the highest activity against P. falciparum chloroquine-resistant blood-stage parasites (clone W2), indicated by their low inhibitory concentration for 50% (IC50) of parasite growth. The therapeutic potential of the active compounds was evaluated according to the selectivity index, which is a ratio of the cytotoxicity minimum lethal dose which eliminates 50% of cells and the in vitro IC50. Naphthoquinones 2 and 5, with activities similar to the reference antimalarial chloroquine, were also active against malaria in mice and suppressed parasitaemia by more than 60% in contrast to compound 11 which was inactive. Based on their in vitro and in vivo activities, compounds 2 and 5 are considered promising molecules for antimalarial treatment and warrant further study.
Collapse
Affiliation(s)
| | - Isabel M de Andrade
- Laboratório de Malária, Centro de Pesquisas René-Rachou-Fiocruz, Belo
Horizonte, MG, Brasil
| | - Paula F Carneiro
- Núcleo de Pesquisas de Produtos Naturais, Universidade Federal do Rio de
Janeiro, Rio de Janeiro, RJ, Brasil
| | - Guilherme AM Jardim
- Laboratório de Química Sintética e Heterocíclica, Departamento de
Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo
Horizonte, MG, Brasil
| | - Isadora MM de Melo
- Laboratório de Química Sintética e Heterocíclica, Departamento de
Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo
Horizonte, MG, Brasil
| | - Eufrânio N da Silva
- Laboratório de Química Sintética e Heterocíclica, Departamento de
Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo
Horizonte, MG, Brasil
| | | |
Collapse
|
32
|
A whole cell pathway screen reveals seven novel chemosensitizers to combat chloroquine resistant malaria. Sci Rep 2014; 3:1734. [PMID: 23615863 PMCID: PMC3635055 DOI: 10.1038/srep01734] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 04/11/2013] [Indexed: 11/09/2022] Open
Abstract
Due to the widespread prevalence of resistant parasites, chloroquine (CQ) was removed from front-line antimalarial chemotherapy in the 1990s despite its initial promise of disease eradication. Since then, resistance-conferring mutations have been identified in transporters such as the PfCRT, that allow for the efflux of CQ from its primary site of action, the parasite digestive vacuole. Chemosensitizing/chemoreversing compounds interfere with the function of these transporters thereby sensitizing parasites to CQ once again. However, compounds identified thus far have disappointing in vivo efficacy and screening for alternative candidates is required to revive this strategy. In this study, we propose a simple and direct means to rapidly screen for such compounds using a fluorescent-tagged CQ molecule. When this screen was applied to a small library, seven novel chemosensitizers (octoclothepin, methiothepin, metergoline, loperamide, chlorprothixene, L-703,606 and mibefradil) were quickly elucidated, including two which showed greater potency than the classical chemosensitizers verapamil and desipramine.
Collapse
|
33
|
Rosenthal PJ. The interplay between drug resistance and fitness in malaria parasites. Mol Microbiol 2013; 89:1025-38. [PMID: 23899091 DOI: 10.1111/mmi.12349] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2013] [Indexed: 12/01/2022]
Abstract
Controlling the spread of antimalarial drug resistance, especially resistance of Plasmodium falciparum to artemisinin-based combination therapies, is a high priority. Available data indicate that, as with other microorganisms, the spread of drug-resistant malaria parasites is limited by fitness costs that frequently accompany resistance. Resistance-mediating polymorphisms in malaria parasites have been identified in putative drug transporters and in target enzymes. The impacts of these polymorphisms on parasite fitness have been characterized in vitro and in animal models. Additional insights have come from analyses of samples from clinical studies, both evaluating parasites under different selective pressures and determining the clinical consequences of infection with different parasites. With some exceptions, resistance-mediating polymorphisms lead to malaria parasites that, compared with wild type, grow less well in culture and in animals, and are replaced by wild type when drug pressure diminishes in the clinical setting. In some cases, the fitness costs of resistance may be offset by compensatory mutations that increase virulence or changes that enhance malaria transmission. However, not enough is known about effects of resistance mediators on parasite fitness. A better appreciation of the costs of fitness-mediating mutations will facilitate the development of optimal guidelines for the treatment and prevention of malaria.
Collapse
Affiliation(s)
- Philip J Rosenthal
- Department of Medicine, University of California, San Francisco, CA, 94143, USA
| |
Collapse
|
34
|
Pharmacogenomics of Cytauxzoon felis cytochrome b: implications for atovaquone and azithromycin therapy in domestic cats with cytauxzoonosis. J Clin Microbiol 2013; 51:3066-9. [PMID: 23784135 DOI: 10.1128/jcm.01407-13] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cytauxzoon felis, an emerging virulent protozoan parasite that infects domestic cats, is treated with atovaquone and azithromycin (A&A). Atovaquone targets parasite cytochrome b. We characterized the C. felis cytochrome b gene (cytb) in cats with cytauxzoonosis and found a cytb genotype that was associated with survival in A&A-treated cats.
Collapse
|
35
|
Exploration of 4(1H)-pyridones as a novel family of potent antimalarial inhibitors of the plasmodial cytochrome bc1. Future Med Chem 2013; 4:2311-23. [PMID: 23234553 DOI: 10.4155/fmc.12.177] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
A novel family of antimalarials based on the 4(1H)-pyridone scaffold is described. The compounds display potent antimalarial activity against Plasmodium falciparum in vitro and in vivo. Like atovaquone, 4(1H)-pyridones exert their antimalarial action by inhibiting selectively the electron-transport chain in P. falciparum at the cytochrome bc1 level (complex III). However, despite the similar mechanism of action, no cross-resistance with atovaquone has been found, suggesting that the binding mode of 4(1H)-pyridones might be different from that of atovaquone. The medicinal chemistry program, focused on improving potency and physicochemical properties, ultimately led to the discovery of GSK932121, which was progressed efficiently into first time in human studies. However, progression of GSK932121 was terminated when new toxicology results were obtained in the rat with a soluble phosphate prodrug of the candidate, indicating a potentially narrow therapeutic index.
Collapse
|
36
|
Wurtz N, Pascual A, Marin-Jauffre A, Bouchiba H, Benoit N, Desbordes M, Martelloni M, Pommier de Santi V, Richa G, Taudon N, Pradines B, Briolant S. Early treatment failure during treatment of Plasmodium falciparum malaria with atovaquone-proguanil in the Republic of Ivory Coast. Malar J 2012; 11:146. [PMID: 22551095 PMCID: PMC3447648 DOI: 10.1186/1475-2875-11-146] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 05/02/2012] [Indexed: 11/29/2022] Open
Abstract
The increased spread of drug-resistant malaria highlights the need for alternative drugs for treatment and chemoprophylaxis. The combination of atovaquone‐proguanil (Malarone®) has shown high efficacy against Plasmodium falciparum with only mild side-effects. Treatment failures have been attributed to suboptimal dosages or to parasite resistance resulting from a point mutation in the cytochrome b gene. In this paper, a case of early treatment failure was reported in a patient treated with atovaquone-proguanil; this failure was not associated with a mutation in the parasite cytochrome b gene, with impaired drug bioavailability, or with re-infection.
Collapse
Affiliation(s)
- Nathalie Wurtz
- Unité de Parasitologie - Unité de Recherche pour les Maladies Infectieuses et Tropicales Emergentes - UMR 6236, Institut de Recherche Biomédicale des Armées, Marseille, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Goldberg DE, Siliciano RF, Jacobs WR. Outwitting evolution: fighting drug-resistant TB, malaria, and HIV. Cell 2012; 148:1271-83. [PMID: 22424234 PMCID: PMC3322542 DOI: 10.1016/j.cell.2012.02.021] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Indexed: 11/20/2022]
Abstract
Although caused by vastly different pathogens, the world's three most serious infectious diseases, tuberculosis, malaria, and HIV-1 infection, share the common problem of drug resistance. The pace of drug development has been very slow for tuberculosis and malaria and rapid for HIV-1. But for each disease, resistance to most drugs has appeared quickly after the introduction of the drug. Learning how to manage and prevent resistance is a major medical challenge that requires an understanding of the evolutionary dynamics of each pathogen. This Review summarizes the similarities and differences in the evolution of drug resistance for these three pathogens.
Collapse
Affiliation(s)
- Daniel E Goldberg
- Department of Medicine and Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | |
Collapse
|
38
|
Song W, Guo J, Ding F, Hu W, Li Z, Gao M. Study on acute toxicity and structure-activity relationship of Daphnia magna exposed to naphthoquinones. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2011; 32:102-106. [PMID: 21787735 DOI: 10.1016/j.etap.2011.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2010] [Revised: 03/31/2011] [Accepted: 04/09/2011] [Indexed: 05/31/2023]
Abstract
The acute toxicities of the six naphthoquinone compounds on Daphnia magna (D. magna) at 48 h were classified as harmful, toxic, and very toxic. The results indicated that logP played an important part in the toxicity of compounds to organism. And 1-carbonyl or the other hydrophobic substituents of the naphthoquinone compounds are likely to mediate the binding of the compound to the target via hydrogen bonding and hydrophobic interactions. Our results provided a foundation for further investigation using 3D-QSAR and HQSAR to evaluate the aquatic ecological risk and the mechanism of toxicity of naphthoquinones.
Collapse
Affiliation(s)
- Wenhua Song
- School of Environmental and Chemical Engineering, Tianjin Polytechnic University, No. 63 Chenglin Road, Hedong District, Tianjin 300160, China.
| | | | | | | | | | | |
Collapse
|
39
|
Martins VDP, Dinamarco TM, Curti C, Uyemura SA. Classical and alternative components of the mitochondrial respiratory chain in pathogenic fungi as potential therapeutic targets. J Bioenerg Biomembr 2011; 43:81-8. [PMID: 21271279 DOI: 10.1007/s10863-011-9331-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The frequency of opportunistic fungal infection has increased drastically, mainly in patients who are immunocompromised due to organ transplant, leukemia or HIV infection. In spite of this, only a few classes of drugs with a limited array of targets, are available for antifungal therapy. Therefore, more specific and less toxic drugs with new molecular targets is desirable for the treatment of fungal infections. In this context, searching for differences between mitochondrial mammalian hosts and fungi in the classical and alternative components of the mitochondrial respiratory chain may provide new potential therapeutic targets for this purpose.
Collapse
Affiliation(s)
- Vicente de Paulo Martins
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | | | | | | |
Collapse
|
40
|
Kathpal A, Patakottu BR, Patankar S. FORS-D Analysis in P. falciparum can Differentiate Classes of Genes Under Selection. Evol Bioinform Online 2011; 7:21-9. [PMID: 21487534 PMCID: PMC3072625 DOI: 10.4137/ebo.s6609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
FORS-D is a measure of the contribution of base order to the stem loop potential of a nucleic acid sequence and can also give information on evolutionary pressures on sequences to move away from secondary structure. Negative FORS-D values in a gene are associated with exons and nucleotide substitutions such as SNPs. An analysis of P. falciparum genes under selection pressure shows a correlation between negative FORS-D values and SNP density for genes that drug targets but not for drug transporters or antigenic variation genes. Analysis of the dhfr gene shows that a majority of rare mutations that associate with drug resistance also fall into regions with negative FORS-D values. These data suggest that FORS-D values might be predictors for drug target genes and drug resistance mutations in these genes.
Collapse
Affiliation(s)
- Atish Kathpal
- Birla Institute of Technology and Science, Pilani, Goa Campus, Zuarinagar 403726, Goa, India
| | | | | |
Collapse
|
41
|
Antao T, Hastings IM. Environmental, pharmacological and genetic influences on the spread of drug-resistant malaria. Proc Biol Sci 2010; 278:1705-12. [PMID: 21084349 DOI: 10.1098/rspb.2010.1907] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Plasmodium falciparum malaria is subject to artificial selection from antimalarial drugs that select for drug-resistant parasites. We describe and apply a flexible new approach to investigate how epistasis, inbreeding, selection heterogeneity and multiple simultaneous drug deployments interact to influence the spread of drug-resistant malaria. This framework recognizes that different human 'environments' within which treatment may occur (such as semi- and non-immune humans taking full or partial drug courses) influence the genetic interactions between parasite loci involved in resistance. Our model provides an explanation for how the rate of spread varies according to different malaria transmission intensities, why resistance might stabilize at intermediate frequencies and also identifies several factors that influence the decline of resistance after a drug is removed. Results suggest that studies based on clinical outcomes might overestimate the spread of resistant parasites, especially in high-transmission areas. We show that when transmission decreases, prevalence might decrease without a corresponding change in frequency of resistance and that this relationship is heavily influenced by the extent of linkage disequilibrium between loci. This has important consequences on the interpretation of data from areas where control is being successful and suggests that reducing transmission might have less impact on the spread of resistance than previously expected.
Collapse
Affiliation(s)
- Tiago Antao
- Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK.
| | | |
Collapse
|
42
|
Afonso A, Neto Z, Castro H, Lopes D, Alves AC, Tomás AM, Rosário VD. Plasmodium chabaudi chabaudi malaria parasites can develop stable resistance to atovaquone with a mutation in the cytochrome b gene. Malar J 2010; 9:135. [PMID: 20492669 PMCID: PMC2881937 DOI: 10.1186/1475-2875-9-135] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2010] [Accepted: 05/21/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Plasmodium falciparum, has developed resistance to many of the drugs in use. The recommended treatment policy is now to use drug combinations. The atovaquone-proguanil (AP) drug combination, is one of the treatment and prophylaxis options. Atovaquone (ATQ) exerts its action by inhibiting plasmodial mitochondria electron transport at the level of the cytochrome bc1 complex. Plasmodium falciparum in vitro resistance to ATQ has been associated with specific point mutations in the region spanning codons 271-284 of the cytochrome b gene. ATQ -resistant Plasmodium yoelii and Plasmodium berghei lines have been obtained and resistant lines have amino acid mutations in their CYT b protein sequences. Plasmodium chabaudi model for studying drug-responses and drug-resistance selection is a very useful rodent malaria model but no ATQ resistant parasites have been reported so far. The aim of this study was to determine the ATQ sensitivity of the P. chabaudi clones, to select a resistant parasite line and to perform genotypic characterization of the cytb gene of these clones. METHODS To select for ATQ resistance, Plasmodium. chabaudi chabaudi clones were exposed to gradually increasing concentrations of ATQ during several consecutive passages in mice. Plasmodium chabaudi cytb gene was amplified and sequenced. RESULTS ATQ resistance was selected from the clone AS-3CQ. In order to confirm whether an heritable genetic mutation underlies the response of AS-ATQ to ATQ, the stability of the drug resistance phenotype in this clone was evaluated by measuring drug responses after (i) multiple blood passages in the absence of the drug, (ii) freeze/thawing of parasites in liquid nitrogen and (iii) transmission through a mosquito host, Anopheles stephensi. ATQ resistance phenotype of the drug-selected parasite clone kept unaltered. Therefore, ATQ resistance in clone AS-ATQ is genetically encoded. The Minimum Curative Dose of AS-ATQ showed a six-fold increase in MCD to ATQ relative to AS-3CQ. CONCLUSIONS A mutation was found on the P. chabaudi cytb gene from the AS-ATQ sample a substitution at the residue Tyr268 for an Asn, this mutation is homologous to the one found in P. falciparum isolates resistant to ATQ.
Collapse
Affiliation(s)
- Ana Afonso
- Unit of Medical Parasitology and Microbiology (UPMM)/IHMT Rua da Junqueira 100, 1349-008 Lisbon, Portugal.
| | | | | | | | | | | | | |
Collapse
|
43
|
Hughes LM, Covian R, Gribble GW, Trumpower BL. Probing binding determinants in center P of the cytochrome bc(1) complex using novel hydroxy-naphthoquinones. BIOCHIMICA ET BIOPHYSICA ACTA 2010; 1797:38-43. [PMID: 19660431 PMCID: PMC2787711 DOI: 10.1016/j.bbabio.2009.07.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Revised: 07/23/2009] [Accepted: 07/23/2009] [Indexed: 11/28/2022]
Abstract
Atovaquone is a substituted 2-hydroxy-naphthoquinone used therapeutically against Plasmodium falciparum (malaria) and Pneumocystis pathogens. It acts by inhibiting the cytochrome bc(1) complex via interactions with the Rieske iron-sulfur protein and cytochrome b in the ubiquinol oxidation pocket. As the targeted pathogens have developed resistance to this drug there is an urgent need for new alternatives. To better understand the determinants of inhibitor binding in the ubiquinol oxidation pocket of the bc(1) complex we synthesized a series of hydroxy-naphthoquinones bearing a methyl group on the benzene ring that is predicted to interact with the nuclear encoded Rieske iron-sulfur protein. We have also attempted to overcome the metabolic instability of a potent cytochrome bc(1) complex inhibitor, a 2-hydroxy-naphthoquinone with a branched side chain, by fluorinating the terminal methyl group. We have tested these new 2-hydroxy-naphthoquinones against yeast and bovine cytochrome bc(1) complexes to model the interaction with pathogen and human enzymes and determine parameters that affect efficacy of binding of these inhibitors. We identified a hydroxy-naphthoquinone with a trifluoromethyl function that has potential for development as an anti-fungal and anti-parasitic therapeutic.
Collapse
Affiliation(s)
- Louise M. Hughes
- Department of Biochemistry, Dartmouth Medical School, 7200 Vail, Hanover, NH 03755, USA
| | - Raul Covian
- Department of Biochemistry, Dartmouth Medical School, 7200 Vail, Hanover, NH 03755, USA
| | | | - Bernard L. Trumpower
- Department of Biochemistry, Dartmouth Medical School, 7200 Vail, Hanover, NH 03755, USA
| |
Collapse
|
44
|
Sakuma M, Setoguchi A, Endo Y. Possible Emergence of Drug-Resistant Variants ofBabesia gibsoniin Clinical Cases Treated with Atovaquone and Azithromycin. J Vet Intern Med 2009; 23:493-8. [DOI: 10.1111/j.1939-1676.2009.0300.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
45
|
Smilkstein MJ, Forquer I, Kanazawa A, Kelly JX, Winter RW, Hinrichs DJ, Kramer DM, Riscoe MK. A drug-selected Plasmodium falciparum lacking the need for conventional electron transport. Mol Biochem Parasitol 2008; 159:64-8. [PMID: 18308406 PMCID: PMC2396451 DOI: 10.1016/j.molbiopara.2008.01.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2007] [Revised: 01/11/2008] [Accepted: 01/15/2008] [Indexed: 10/22/2022]
Abstract
Mitochondrial electron transport is essential for survival in Plasmodium falciparum, making the cytochrome (cyt) bc(1) complex an attractive target for antimalarial drug development. Here we report that P. falciparum cultivated in the presence of a novel cyt bc(1) inhibitor underwent a fundamental transformation in biochemistry to a phenotype lacking a requirement for electron transport through the cyt bc(1) complex. Growth of the drug-selected parasite clone (SB1-A6) is robust in the presence of diverse cyt bc(1) inhibitors, although electron transport is fully inhibited by these same agents. This transformation defies expected molecular-based concepts of drug resistance, has important implications for the study of cyt bc(1) as an antimalarial drug target, and may offer a glimpse into the evolutionary future of Plasmodium.
Collapse
Affiliation(s)
- Martin J Smilkstein
- Medical Research Service, Department of Veterans Affairs Medical Center, Portland, OR 97239, USA.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Ekala MT, Khim N, Legrand E, Randrianarivelojosia M, Jambou R, Fandeur T, Menard D, Assi SB, Henry MC, Rogier C, Bouchier C, Mercereau-Puijalon O. Sequence analysis of Plasmodium falciparum cytochrome b in multiple geographic sites. Malar J 2007; 6:164. [PMID: 18086297 PMCID: PMC2228307 DOI: 10.1186/1475-2875-6-164] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Accepted: 12/17/2007] [Indexed: 11/29/2022] Open
Abstract
Background The antimalarial drug atovaquone specifically targets Plasmodium falciparum cytochrome b (Pfcytb), a mitochondrial gene with uniparental inheritance. Cases of resistance to atovaquone associated with mutant Pfcytb have been reported, justifying efforts to better document the natural polymorphism of this gene. To this end, a large molecular survey was conducted in several malaria endemic areas where atovaquone was not yet in regular use. Methods The polymorphism of the Pfcytb was analysed by direct sequencing of PCR products corresponding to the full length coding region. Sequence was generated for 671 isolates originating from three continents: Africa (Senegal, Ivory Coast, Central African Republic and Madagascar), Asia (Cambodia) and South America (French Guiana). Results Overall, 11 polymorphic sites were observed, of which eight were novel mutations. There was a large disparity in the geographic distribution of the mutants. All isolates from Senegal, Central African Republic and Madagascar displayed a Camp/3D7 wild type Pfcytb sequence, as did most samples originating from Cambodia and Ivory Coast. One synonymous (t759a at codon V253V) and two non-synonymous (t553g and a581g at codons F185V and H194R, respectively) singletons were detected in Ivory Coast. Likewise, two synonymous (a126t and c793t at codons -T42T and L265L, respectively) singletons were observed in Cambodia. In contrast, seven mutated sites, affecting seven codons and defining four mutant haplotypes were observed in French Guiana. The wild type allele was observed in only 14% of the French Guiana isolates. The synonymous c688t mutation at position L230L was highly prevalent; the most frequent allele was the c688t single mutant, observed in 84% of the isolates. The other alleles were singletons (a126t/a165c, a4g/a20t/a1024c and a20t/t341c/c688t corresponding to T42T/S55S, N2D/N71I/I342L, N71I/L114S/L230L, respectively" please replace with ' corresponding to T42T/S55S, N2D/N71I/I342L and N71I/L114S/L230L, respectively). The codon 268 polymorphisms associated with atovaquone resistance were not observed in the panel the isolates studied. Overall, the wild type PfCYTb protein isoform was highly predominant in all study areas, including French Guiana, suggesting stringent functional constraints. Conclusion These data along with previously identified Pfcytb field polymorphisms indicate a clustering of molecular signatures, suggesting different ancestral types in South America and other continents. The absence of mutations associated with most atovaquone-proguanil clinical failures indicates that the atovaquone-proguanil association is an interesting treatment option in the study areas.
Collapse
Affiliation(s)
- Marie-Thérèse Ekala
- Immunologie Moléculaire des Parasites, CNRS URA 2581, Institut Pasteur, 25 rue du Dr ROUX, 75724 Paris Cedex 15, Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Painter HJ, Morrisey JM, Mather MW, Vaidya AB. Specific role of mitochondrial electron transport in blood-stage Plasmodium falciparum. Nature 2007; 446:88-91. [PMID: 17330044 DOI: 10.1038/nature05572] [Citation(s) in RCA: 365] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2006] [Accepted: 01/05/2007] [Indexed: 11/08/2022]
Abstract
The origin of all mitochondria can be traced to the symbiotic arrangement that resulted in the emergence of eukaryotes in a world that was exclusively populated by prokaryotes. This arrangement, however, has been in continuous genetic flux: the varying degrees of gene loss and transfer from the mitochondrial genome in different eukaryotic lineages seem to signify an ongoing 'conflict' between the host and the symbiont. Eukaryotic parasites belonging to the phylum Apicomplexa provide an excellent example to support this view. These organisms contain the smallest mitochondrial genomes known, with an organization that differs among various genera; one genus, Cryptosporidium, seems to have lost the entire mitochondrial genome. Here we show that erythrocytic stages of the human malaria parasite Plasmodium falciparum seem to maintain an active mitochondrial electron transport chain to serve just one metabolic function: regeneration of ubiquinone required as the electron acceptor for dihydroorotate dehydrogenase, an essential enzyme for pyrimidine biosynthesis. Transgenic P. falciparum parasites expressing Saccharomyces cerevisiae dihydroorotate dehydrogenase, which does not require ubiquinone as an electron acceptor, were completely resistant to inhibitors of mitochondrial electron transport. Maintenance of mitochondrial membrane potential, however, was essential in these parasites, as indicated by their hypersensitivity to proguanil, a drug that collapsed the membrane potential in the presence of electron transport inhibitors. Thus, acquisition of just one enzyme can render mitochondrial electron transport nonessential in erythrocytic stages of P. falciparum.
Collapse
Affiliation(s)
- Heather J Painter
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129, USA
| | | | | | | |
Collapse
|
48
|
Abstract
Protein kinases (PKs) are prime targets for drug discovery in a variety of diseases, including cancer and neurodegenerative pathologies. The characterisation of the kinome of the human malaria parasite Plasmodium falciparum has revealed profound divergences, at several levels, between PKs of the parasite and those of its host. Here, the authors review the major issues and recent advances regarding the development of Plasmodium-selective PK inhibitors, with emphasis on target identification and validation, and on structure-based design. The authors also discuss the possibility of interfering with: i) Plasmodium PKs regulating transmission to the mosquito vector; and ii) host PKs that may be required for parasite survival.
Collapse
Affiliation(s)
- Christian Doerig
- INSERM U609, Wellcome Centre for Molecular Parasitology, Biomedical Research Centre, University of Glasgow, 120 University Place, Glasgow, UK.
| | | |
Collapse
|
49
|
Baniecki ML, Wirth DF, Clardy J. High-throughput Plasmodium falciparum growth assay for malaria drug discovery. Antimicrob Agents Chemother 2006; 51:716-23. [PMID: 17116676 PMCID: PMC1797774 DOI: 10.1128/aac.01144-06] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
New therapeutic agents for the treatment of malaria, the world's most deadly parasitic disease, are urgently needed. Malaria afflicts 300 to 500 million people and results in 1 to 2 million deaths annually, and more than 85% of all malaria-related mortality involves young children and pregnant women in sub-Saharan Africa. The emergence of multidrug-resistant parasites, especially in Plasmodium falciparum, has eroded the efficacy of almost all currently available therapeutic agents. The discovery of new drugs, including drugs with novel cellular targets, could be accelerated with a whole-organism high-throughput screen (HTS) of structurally diverse small-molecule libraries. The standard whole-organism screen is based on incorporation of [3H]hypoxanthine and has liabilities, such as limited throughput, high cost, multiple labor-intensive steps, and disposal of radioactive waste. Recently, screens have been reported that do not use radioactive incorporation, but their reporter signal is not robust enough for HTS. We report a P. falciparum growth assay that is technically simple, robust, and compatible with the automation necessary for HTS. The assay monitors DNA content by addition of the fluorescent dye 4',6-diamidino-2-phenylindole (DAPI) as a reporter of blood-stage parasite growth. This DAPI P. falciparum growth assay was used to measure the 50% inhibitory concentrations (IC50s) of a diverse set of known antimalarials. The resultant IC50s compared favorably with those obtained in the [3H]hypoxanthine incorporation assay. Over 79,000 small molecules have been tested for antiplasmodial activity using the DAPI P. falciparum growth assay, and 181 small molecules were identified as highly active against multidrug-resistant parasites.
Collapse
Affiliation(s)
- Mary Lynn Baniecki
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Avenue, C-643, Boston, MA 02115, USA
| | | | | |
Collapse
|
50
|
Nyakeriga AM, Perlmann H, Hagstedt M, Berzins K, Troye-Blomberg M, Zhivotovsky B, Perlmann P, Grandien A. Drug-induced death of the asexual blood stages of Plasmodium falciparum occurs without typical signs of apoptosis. Microbes Infect 2006; 8:1560-8. [PMID: 16702009 DOI: 10.1016/j.micinf.2006.01.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2005] [Revised: 01/13/2006] [Accepted: 01/16/2006] [Indexed: 11/24/2022]
Abstract
There is clear evidence that most antimalarial drugs, while acting through different mechanisms, are associated with parasite growth/development inhibition and eventual parasite death. However, the exact mode of parasite death remains unclear. In the present study, we investigated the ability of various drugs, including two antimalarial drugs (chloroquine and atovaquone), a topoisemerase II inhibitor (etoposide) and a nitric oxide donor (S-nitro-N-acetyl-D, L-penicillamine), to induce apoptosis in a laboratory strain of Plasmodium falciparum. Results obtained from flow cytometric analysis showed a significant reduction in the percent of parasitemia and parasite growth in all drug-treated parasite cultures, including those treated with etoposide and S-nitro-N-acetyl-D, L-penicillamine. For further investigation, we used various biochemical approaches including the terminal dUTP nick-end labeling assay, determination of mitochondrial membrane integrity and DNA degradation/fragmentation, to analyze the changes occurring during parasite-drug interactions and eventual death. We observed that loss of membrane potential was induced in parasite cultures treated with atovaquone, while S-nitro-N-acetyl-D, L-penicillamine induced abnormal parasite forms, "crisis forms", and minor DNA degradation. However, these features were not observed in the parasite cultures treated with chloroquine nor were other features of apoptosis-like death associated with any of the drugs used in this study. The death resulting from the various drug treatments is atypical of apotosis. More studies will be needed to define the precise mode of death exhibited by P. falciparum.
Collapse
Affiliation(s)
- Alice M Nyakeriga
- Department of Immunology Wenner-Gren Institute, Stockholm University, Stockholm, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|