1
|
Fahs HZ, Refai FS, Gopinadhan S, Moussa Y, Gan HH, Hunashal Y, Battaglia G, Cipriani PG, Ciancia C, Rahiman N, Kremb S, Xie X, Pearson YE, Butterfoss GL, Maizels RM, Esposito G, Page AP, Gunsalus KC, Piano F. A new class of natural anthelmintics targeting lipid metabolism. Nat Commun 2025; 16:305. [PMID: 39746976 PMCID: PMC11695593 DOI: 10.1038/s41467-024-54965-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 11/26/2024] [Indexed: 01/04/2025] Open
Abstract
Parasitic helminths are a major global health threat, infecting nearly one-fifth of the human population and causing significant losses in livestock and crops. Resistance to the few anthelmintic drugs is increasing. Here, we report a set of avocado fatty alcohols/acetates (AFAs) that exhibit nematocidal activity against four veterinary parasitic nematode species: Brugia pahangi, Teladorsagia circumcincta and Heligmosomoides polygyrus, as well as a multidrug resistant strain (UGA) of Haemonchus contortus. AFA shows significant efficacy in H. polygyrus infected mice. In C. elegans, AFA exposure affects all developmental stages, causing paralysis, impaired mitochondrial respiration, increased reactive oxygen species production and mitochondrial damage. In embryos, AFAs penetrate the eggshell and induce rapid developmental arrest. Genetic and biochemical tests reveal that AFAs inhibit POD-2, encoding an acetyl CoA carboxylase, the rate-limiting enzyme in lipid biosynthesis. These results uncover a new anthelmintic class affecting lipid metabolism.
Collapse
Affiliation(s)
- Hala Zahreddine Fahs
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Fathima S Refai
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates
| | - Suma Gopinadhan
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates
| | - Yasmine Moussa
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates
| | - Hin Hark Gan
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Yamanappa Hunashal
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates
| | - Gennaro Battaglia
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates
- Dipartimento di Scienze Chimiche, Università di Napoli "Federico II", 80138, Naples, Italy
| | - Patricia G Cipriani
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Claire Ciancia
- School of Infection and Immunity, University of Glasgow, Scotland, UK
| | - Nabil Rahiman
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates
| | - Stephan Kremb
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates
| | - Xin Xie
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates
| | - Yanthe E Pearson
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates
| | - Glenn L Butterfoss
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates
| | - Rick M Maizels
- School of Infection and Immunity, University of Glasgow, Scotland, UK
| | - Gennaro Esposito
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates
- Istituto Nazionale Biostrutture e Biosistemi, 00136, Rome, Italy
| | - Antony P Page
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Scotland, UK
| | - Kristin C Gunsalus
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates.
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY, USA.
| | - Fabio Piano
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates.
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY, USA.
| |
Collapse
|
2
|
Rothmann-Meyer W, Naidoo K, de Waal PJ. Spirocerca lupi draft genome, vaccine and anthelmintic targets. Mol Biochem Parasitol 2024; 259:111632. [PMID: 38834134 DOI: 10.1016/j.molbiopara.2024.111632] [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: 01/10/2024] [Revised: 05/13/2024] [Accepted: 05/23/2024] [Indexed: 06/06/2024]
Abstract
Spirocerca lupi is a parasitic nematode affecting predominantly domestic dogs. It causes spirocercosis, a disease that is often fatal. The assembled draft genome of S. lupi consists of 13,627 predicted protein-coding genes and is approximately 150 Mb in length. Several known anthelmintic gene targets such as for β-Tubulin, glutamate, and GABA receptors as well as known vaccine gene targets such as cysteine protease inhibitor and cytokines were identified in S. lupi by comparing orthologs of C. elegans anthelmintic gene targets as well as orthologs to known vaccine candidates. New anthelmintic targets were predicted through an inclusion-exclusion strategy and new vaccine targets were predicted through an immunoinformatics approach. New anthelminthic targets include DNA-directed RNA polymerases, chitin synthase, polymerases, and other enzymes. New vaccine targets include cuticle collagens. These gene targets provide a starting platform for new drug identification and vaccine design.
Collapse
Affiliation(s)
- Wiekolize Rothmann-Meyer
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa
| | - Kershney Naidoo
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa; Thermo Fisher Scientific, Hybrid Field Application Scientist & Field Service Engineer, South Africa
| | - Pamela J de Waal
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa.
| |
Collapse
|
3
|
Jung H, Zarlenga D, Martin JC, Geldhof P, Hallsworth-Pepin K, Mitreva M. The identification of small molecule inhibitors with anthelmintic activities that target conserved proteins among ruminant gastrointestinal nematodes. mBio 2024; 15:e0009524. [PMID: 38358246 PMCID: PMC10936192 DOI: 10.1128/mbio.00095-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/16/2024] Open
Abstract
Gastrointestinal nematode (GIN) infections are a major concern for the ruminant industry worldwide and result in significant production losses. Naturally occurring polyparasitism and increasing drug resistance that potentiate disease outcomes are observed among the most prevalent GINs of veterinary importance. Within the five major taxonomic clades, clade Va represents a group of GINs that predominantly affect the abomasum or small intestine of ruminants. However, the development of effective broad-spectrum anthelmintics against ruminant clade Va GINs has been challenged by a lack of comprehensive druggable genome resources. Here, we first assembled draft genomes for three clade Va species (Cooperia oncophora, Trichostrongylus colubriformis, and Ostertagia ostertagi) and compared them with closely related ruminant GINs. Genome-wide phylogenetic reconstruction showed a relationship among ruminant GINs structured by taxonomic classification. Orthogroup (OG) inference and functional enrichment analyses identified 220 clade Va-specific and Va-conserved OGs, enriched for functions related to cell cycle and cellular senescence. Further transcriptomic analysis identified 61 taxonomically and functionally conserved clade Va OGs that may function as drug targets for new broad-spectrum anthelmintics. Chemogenomic screening identified 11 compounds targeting homologs of these OGs, thus having potential anthelmintic activity. In in vitro phenotypic assays, three kinase inhibitors (digitoxigenin, K-252a, and staurosporine) exhibited broad-spectrum anthelmintic activities against clade Va GINs by obstructing the motility of exsheathed L3 (xL3) or molting of xL3 to L4. These results demonstrate valuable applications of the new ruminant GIN genomes in gaining better insights into their life cycles and offer a contemporary approach to discovering the next generation of anthelmintics.IMPORTANCEGastrointestinal nematode (GIN) infections in ruminants are caused by parasites that inhibit normal function in the digestive tract of cattle, sheep, and goats, thereby causing morbidity and mortality. Coinfection and increasing drug resistance to current therapeutic agents will continue to worsen disease outcomes and impose significant production losses on domestic livestock producers worldwide. In combination with ongoing therapeutic efforts, advancing the discovery of new drugs with novel modes of action is critical for better controlling GIN infections. The significance of this study is in assembling and characterizing new GIN genomes of Cooperia oncophora, Ostertagia ostertagi, and Trichostrongylus colubriformis for facilitating a multi-omics approach to identify novel, biologically conserved drug targets for five major GINs of veterinary importance. With this information, we were then able to demonstrate the potential of commercially available compounds as new anthelmintics.
Collapse
Affiliation(s)
- Hyeim Jung
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Dante Zarlenga
- Animal Parasitic Diseases Laboratory, U.S. Department of Agriculture, Agricultural Research Service, Beltsville, Maryland, USA
| | - John C. Martin
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Peter Geldhof
- Laboratory of Parasitology, Faculty of Veterinary Medicine, University of Ghent, Merelbeke, Belgium
| | | | - Makedonka Mitreva
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
4
|
Uthailak N, Adisakwattana P, Chienwichai P, Tipthara P, Tarning J, Thawornkuno C, Thiangtrongjit T, Reamtong O. Metabolite profiling of Trichinella spiralis adult worms and muscle larvae identifies their excretory and secretory products. Front Cell Infect Microbiol 2023; 13:1306567. [PMID: 38145042 PMCID: PMC10749202 DOI: 10.3389/fcimb.2023.1306567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 11/20/2023] [Indexed: 12/26/2023] Open
Abstract
Human trichinellosis is a parasitic infection caused by roundworms belonging to the genus Trichinella, especially Trichinella spiralis. Early and accurate clinical diagnoses of trichinellosis are required for efficacious prognosis and treatment. Current drug therapies are limited by antiparasitic resistance, poor absorption, and an inability to kill the encapsulating muscle-stage larvae. Therefore, reliable biomarkers and drug targets for novel diagnostic approaches and anthelmintic drugs are required. In this study, metabolite profiles of T. spiralis adult worms and muscle larvae were obtained using mass spectrometry-based metabolomics. In addition, metabolite-based biomarkers of T. spiralis excretory-secretory products and their related metabolic pathways were characterized. The metabolic profiling identified major, related metabolic pathways involving adenosine monophosphate (AMP)-dependent synthetase/ligase and glycolysis/gluconeogenesis in T. spiralis adult worms and muscle larvae, respectively. These pathways are potential drug targets for the treatment of the intestinal and muscular phases of infection. The metabolome of larva excretory-secretory products was characterized, with amino acid permease and carbohydrate kinase being identified as key metabolic pathways. Among six metabolites, decanoyl-l-carnitine and 2,3-dinor-6-keto prostaglandin F1α-d9 were identified as potential metabolite-based biomarkers that might be related to the host inflammatory processes. In summary, this study compared the relationships between the metabolic profiles of two T. spiralis growth stages. Importantly, the main metabolites and metabolic pathways identified may aid the development of novel clinical diagnostics and therapeutics for human trichinellosis and other related helminthic infections.
Collapse
Affiliation(s)
- Naphatsamon Uthailak
- Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Poom Adisakwattana
- Department of Helminthology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Peerut Chienwichai
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Phornpimon Tipthara
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Joel Tarning
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Charin Thawornkuno
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Tipparat Thiangtrongjit
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
5
|
Hussain R, Haider Z, Khalid H, Fatmi MQ, Carradori S, Cataldi A, Zara S. Computational medicinal chemistry applications to target Asian-prevalent strain of hepatitis C virus. RSC Adv 2023; 13:30052-30070. [PMID: 37849696 PMCID: PMC10578362 DOI: 10.1039/d3ra04622b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/04/2023] [Indexed: 10/19/2023] Open
Abstract
Hepatitis C Virus (HCV), affecting millions of people worldwide, is the leading cause of liver disorder, cirrhosis, and hepatocellular carcinoma. HCV is genetically diverse having eight genotypes and several subtypes predominant in different regions of the globe. The HCV NS3/4A protease is a primary therapeutic target for HCV with various FDA-approved antivirals and several clinical developments. However, available protease inhibitors (PIs) have lower potency against HCV genotype 3 (GT3), prevalent in South Asia. In this study, the incumbent computational tools were utilized to understand and explore interactions of the HCV GT3 receptor with the potential inhibitors after the virtual screening of one million compounds retrieved from the ZINC database. The molecular dynamics, pharmacological studies, and experimental studies uncovered the potential PIs as ZINC000224449889, ZINC000224374291, and ZINC000224374456 and the derivative of ZINC000224374456 from the ZINC library. The study revealed that these top-hit compounds exhibited good binding and better pharmacokinetics properties that might be considered the most promising compound against HCV GT3 protease. Viability test, on primary healthy Human Gingival Fibroblasts (HGFs) and cancerous AGS cell line, was also carried out to assess their safety profile after administration. In addition, Surface Plasmon Resonance (SPR) was also performed for the determination of affinity and kinetics of synthesized compounds with target proteins.
Collapse
Affiliation(s)
- Rashid Hussain
- Department of Chemistry, Forman Christian College University Lahore-54000 Pakistan
| | - Zulkarnain Haider
- Department of Chemistry, Forman Christian College University Lahore-54000 Pakistan
| | - Hira Khalid
- Department of Chemistry, Forman Christian College University Lahore-54000 Pakistan
| | - M Qaiser Fatmi
- Department of Biosciences, COMSATS University Islamabad Park Road, Chak Shahzad Islamabad 45600 Pakistan
| | - Simone Carradori
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara via dei Vestini 31 66100 Chieti Italy
| | - Amelia Cataldi
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara via dei Vestini 31 66100 Chieti Italy
| | - Susi Zara
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara via dei Vestini 31 66100 Chieti Italy
| |
Collapse
|
6
|
Arun A, Shanthi A, Raveendran M, Seenivasan N, Pushpam R, Shandeep G. An Insight into Occurrence, Biology, and Pathogenesis of Rice Root-Knot Nematode Meloidogyne graminicola. BIOLOGY 2023; 12:987. [PMID: 37508416 PMCID: PMC10376547 DOI: 10.3390/biology12070987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 07/30/2023]
Abstract
Rice (Oryza sativa L.) is one of the most widely grown crops in the world, and is a staple food for more than half of the global total population. Root-knot nematodes (RKNs), Meloidogyne spp., and especially M. graminicola, seem to be significant rice pests, which makes them the most economically important plant-parasitic nematode in this crop. RKNs develop a feeding site in galls by causing host cells to differentiate into hypertrophied, multinucleate, metabolically active cells known as giant cells. This grazing framework gives the nematode a constant food source, permitting it to develop into a fecund female and complete its life cycle inside the host root. M. graminicola effector proteins involved in nematode parasitism, including pioneer genes, were functionally characterized in earlier studies. Molecular modelling and docking studies were performed on Meloidogyne graminicola protein targets, such as β-1,4-endoglucanase, pectate lyase, phospholipase B-like protein, and G protein-coupled receptor kinase, to understand the binding affinity of Beta-D-Galacturonic Acid, 2,6,10,15,19,23-hexamethyltetracosane, (2S)-2-amino-3-phenylpropanoic acid, and 4-O-Beta-D-Galactopyranosyl-Alpha-D-Glucopyranose against ligand molecules of rice. This study discovered important molecular aspects of plant-nematode interaction and candidate effector proteins that were regulated by M. graminicola-infected rice plants. To the best of our knowledge, this is the first study to describe M. graminicola's molecular adaptation to host parasitism.
Collapse
Affiliation(s)
- Arunachalam Arun
- Department of Nematology, Tamil Nadu Agricultural University, Coimbatore 641003, Tamil Nadu, India
| | - Annaiyan Shanthi
- Department of Nematology, Tamil Nadu Agricultural University, Coimbatore 641003, Tamil Nadu, India
| | - Muthurajan Raveendran
- Directorate of Research, Tamil Nadu Agricultural University, Coimbatore 641003, Tamil Nadu, India
| | | | - Ramamoorthy Pushpam
- Department of Rice, Tamil Nadu Agricultural University, Coimbatore 641003, Tamil Nadu, India
| | - Ganeshan Shandeep
- Department of Nematology, Tamil Nadu Agricultural University, Coimbatore 641003, Tamil Nadu, India
| |
Collapse
|
7
|
Gökalp F. A Study on Natural Control against Nematodes and Whiteflies with Marigold, Known as an Antagonist Plant. J Chem Ecol 2023; 49:230-234. [PMID: 37097510 DOI: 10.1007/s10886-023-01421-6] [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: 08/19/2022] [Revised: 01/11/2023] [Accepted: 03/25/2023] [Indexed: 04/26/2023]
Abstract
The importance of finding natural solutions for the protection of our health in the fight against pests in agriculture is increasing day by day. In this study, the interaction of the active ingredients in marigolds as the great importance as a garden flower, with nematode and whitefly receptors as ligands in the fight against them, have been investigated by chemical calculation method. The inhibition effect of ligands (alpha-Terthienyl, Quercetagetin in marigold) on nematode and whitefly receptors in this plant was determined by comparing the binding energy values with reference drug active ingredients (imidacloprid, Perhexiline).This calculation method, the accuracy determined by different studies, is very important in terms of determining the most active substance in a short time, preventing time and substance loss, and will guide the experimental studies and applications to be made in this field.
Collapse
Affiliation(s)
- Faik Gökalp
- Kırıkkale University,Education Faculty, Department Of Mathematics and Science Education, Science Education, /Kırıkkale, 71450, Yahşihan, Turkey.
| |
Collapse
|
8
|
Hada A, Singh D, Banakar P, Papolu PK, Kassam R, Chatterjee M, Yadav J, Rao U. Host-delivered RNAi-mediated silencing using fusion cassettes of different functional groups of genes precludes Meloidogyne incognita multiplication in Nicotiana tabacum. PLANT CELL REPORTS 2023; 42:29-43. [PMID: 36462028 DOI: 10.1007/s00299-022-02934-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 10/04/2022] [Indexed: 06/17/2023]
Abstract
This study demonstrates multi-gene silencing approach for simultaneous silencing of several functional genes through a fusion gene strategy for protecting plants against root-knot nematode, Meloidogyne incognita. The ability of root-knot nematode (RKN), Meloidogyne incognita, to cause extensive yield decline in a wide range of cultivated crops is well-documented. Due to the inadequacies of current management approaches, the alternatively employed contemporary RNA interference (RNAi)-based host-delivered gene silencing (HD-RNAi) strategy targeting different functional effectors/genes has shown substantial potential to combat RKNs. In this direction, we have explored the possibility of simultaneous silencing of four esophageal gland genes, six plant cell-wall modifying enzymes (PCWMEs) and a serine protease gene of M. incognita using the fusion approach. In vitro RNAi showed that combinatorial gene silencing is the most effective in affecting nematode behavior in terms of reduced attraction, penetration, development, and reproduction in tomato and adzuki beans. In addition, qRT-PCR analysis of M. incognita J2s soaked in fusion-dsRNA showed perturbed expression of all the genes comprising the fusion construct confirming successful dsRNA processing which is also supported by increased mRNA abundance of five key-RNAi pathway genes. In addition, hairpin RNA expressing constructs of multi-gene fusion cassettes were developed and used for generation of Nicotiana tabacum transgenic plants. The integration of gene constructs and expression of siRNAs in transgenic events were confirmed by Southern and Northern blot analyses. Besides, bio-efficacy analyses of transgenic events, conferred up to 87% reduction in M. incognita multiplication. Correspondingly, reduced transcript accumulation of the target genes in the M. incognita females extracted from transgenic events confirmed successful gene silencing.
Collapse
Affiliation(s)
- Alkesh Hada
- Division of Nematology, ICAR-Indian Agricultural Research Institute, New Delhi, 110012, India
| | - Divya Singh
- Division of Nematology, ICAR-Indian Agricultural Research Institute, New Delhi, 110012, India
| | - Prakash Banakar
- Division of Nematology, ICAR-Indian Agricultural Research Institute, New Delhi, 110012, India.
- Department of Nematology and Centre for Bio-Nanotechnology, Chaudhary Charan Singh Haryana Agricultural University, Hisar, 125004, India.
| | - Pradeep K Papolu
- Division of Nematology, ICAR-Indian Agricultural Research Institute, New Delhi, 110012, India
| | - Rami Kassam
- Division of Nematology, ICAR-Indian Agricultural Research Institute, New Delhi, 110012, India
| | - Madhurima Chatterjee
- Division of Nematology, ICAR-Indian Agricultural Research Institute, New Delhi, 110012, India
| | - Jyoti Yadav
- Division of Nematology, ICAR-Indian Agricultural Research Institute, New Delhi, 110012, India
| | - Uma Rao
- Division of Nematology, ICAR-Indian Agricultural Research Institute, New Delhi, 110012, India.
| |
Collapse
|
9
|
Saravanan R, Saranya N, Ragapriya V, Rajaswaminathan V, Kavino M, Krishnamoorthy AS, Nakkeeran S. Nematicidal Property of Clindamycin and 5-hydroxy-2-methyl Furfural (HMF) from the Banana Endophyte Bacillus velezensis (YEBBR6) Against Banana Burrowing Nematode Radopholus similis. Indian J Microbiol 2022; 62:364-373. [PMID: 35974914 PMCID: PMC9375788 DOI: 10.1007/s12088-022-01011-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 02/17/2022] [Indexed: 11/05/2022] Open
Abstract
Radopholus similis is a burrowing nematode which causes banana toppling disease and is of major economic threat for the banana production. Bacterial endophyte Bacillus velezensis (YEBBR6) produce biomolecules like 5-hydroxy-2-methyl furfural (HMF) and clindamycin in during interaction with Fusarium oxysporum f.sp. cubense. Molecular modelling and docking studies were performed on Radopholus similis protein targets such as calreticulin, cathepsin S-like cysteine proteinase, β-1,4 -endoglucanase, reticulocalbin, venom allergen-like protein and serine carboxypeptidase to understand the mode of action of HMF and clindamycin against Radopholus similis. Structurally validated protein targets of R. similis were docked with biomolecules through AutoDock Vina module in PyRx 0.8 software to predict the binding energy of ligand and target protein. Among the chosen six targets, docking analysis revealed that clindamycin had the maximum binding affinity for β-1,4-endoglucanase (- 7.2 kcal/mol), reticulocalbin (- 7.5 kcal/mol) and serine carboxypeptidase (- 6.9 kcal/mol) in comparison with HMF and the nematicide, carbofuran 3G. Besides, clindamycin also had the maximum binding energy for the target sites calreticulin and venom allergen-like protein compared to the small molecule HMF. Novel molecule, clindamycin produced by B. velezensis served as a potential inhibitor of the target sites associated in interrupting the functions of β-1,4-endoglucanase, reticulocalbin, serine carboxypeptidase, calreticulin, cathepsin S-like cysteine proteinase, and venom allergen-like proteins. Besides, increased binding affinity of clindamycin with the protein target sites facilitated to explore it as a novel nematicidal molecule for the management of banana burrowing nematode R. similis. Thus, present investigation confirmed that, the small molecules clindamycin can be explored for nematicidal activity. Graphical Abstract Supplementary Information The online version contains supplementary material available at 10.1007/s12088-022-01011-2.
Collapse
Affiliation(s)
- R. Saravanan
- Department of Plant Pathology, Tamil Nadu Agricultural University, Coimbatore, Tamil Nadu India
| | - N. Saranya
- Department of Plant Molecular Biology and Bioinformatics, Centre for Plant Molecular Biology and Biotechnology, Tamil Nadu Agricultural University, Coimbatore, Tamil Nadu India
| | - V. Ragapriya
- Department of Plant Molecular Biology and Bioinformatics, Centre for Plant Molecular Biology and Biotechnology, Tamil Nadu Agricultural University, Coimbatore, Tamil Nadu India
| | - V. Rajaswaminathan
- Department of Plant Pathology, Tamil Nadu Agricultural University, Coimbatore, Tamil Nadu India
| | - M. Kavino
- Department of Fruit Science, Horticultural College and Research Institute, Tamil Nadu Agricultural University, Coimbatore, Tamil Nadu India
| | - A. S. Krishnamoorthy
- Department of Plant Pathology, Tamil Nadu Agricultural University, Coimbatore, Tamil Nadu India
| | - S. Nakkeeran
- Department of Plant Pathology, Tamil Nadu Agricultural University, Coimbatore, Tamil Nadu India
| |
Collapse
|
10
|
Mitreva M. Parasite OMICS, the grand challenges ahead. FRONTIERS IN PARASITOLOGY 2022; 1:995302. [PMID: 39816466 PMCID: PMC11732041 DOI: 10.3389/fpara.2022.995302] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 07/20/2022] [Indexed: 01/18/2025]
Affiliation(s)
- Makedonka Mitreva
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
11
|
Stryiński R, Mateos J, Carrera M, Jastrzębski JP, Bogacka I, Łopieńska-Biernat E. Tandem Mass Tagging (TMT) Reveals Tissue-Specific Proteome of L4 Larvae of Anisakis simplex s. s.: Enzymes of Energy and/or Carbohydrate Metabolism as Potential Drug Targets in Anisakiasis. Int J Mol Sci 2022; 23:ijms23084336. [PMID: 35457153 PMCID: PMC9027741 DOI: 10.3390/ijms23084336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 02/04/2023] Open
Abstract
Anisakis simplex s. s. is a parasitic nematode of marine mammals and causative agent of anisakiasis in humans. The cuticle and intestine of the larvae are the tissues most responsible for direct and indirect contact, respectively, of the parasite with the host. At the L4 larval stage, tissues, such as the cuticle and intestine, are fully developed and functional, in contrast to the L3 stage. As such, this work provides for the first time the tissue-specific proteome of A. simplex s. s. larvae in the L4 stage. Statistical analysis (FC ≥ 2; p-value ≤ 0.01) showed that 107 proteins were differentially regulated (DRPs) between the cuticle and the rest of the larval body. In the comparison between the intestine and the rest of the larval body at the L4 stage, 123 proteins were identified as DRPs. Comparison of the individual tissues examined revealed a total of 272 DRPs, with 133 proteins more abundant in the cuticle and 139 proteins more abundant in the intestine. Detailed functional analysis of the identified proteins was performed using bioinformatics tools. Glycolysis and the tricarboxylic acid cycle were the most enriched metabolic pathways by cuticular and intestinal proteins, respectively, in the L4 stage of A. simplex s. s. The presence of two proteins, folliculin (FLCN) and oxoglutarate dehydrogenase (OGDH), was confirmed by Western blot, and their tertiary structure was predicted and compared with other species. In addition, host–pathogen interactions were identified, and potential new allergens were predicted. The result of this manuscript shows the largest number of protein identifications to our knowledge using proteomics tools for different tissues of L4 larvae of A. simplex s. s. The identified tissue-specific proteins could serve as targets for new drugs against anisakiasis.
Collapse
Affiliation(s)
- Robert Stryiński
- Department of Biochemistry, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland
- Correspondence: (R.S.); (M.C.); (E.Ł.-B.)
| | - Jesús Mateos
- Clinical Pharmacology Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15-706 A Coruña, Spain;
| | - Mónica Carrera
- Department of Food Technology, Marine Research Institute (IIM), Spanish National Research Council (CSIC), 36-208 Vigo, Spain
- Correspondence: (R.S.); (M.C.); (E.Ł.-B.)
| | - Jan Paweł Jastrzębski
- Department of Plant Physiology, Genetics and Biotechnology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
| | - Iwona Bogacka
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
| | - Elżbieta Łopieńska-Biernat
- Department of Biochemistry, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland
- Correspondence: (R.S.); (M.C.); (E.Ł.-B.)
| |
Collapse
|
12
|
Bogucka-Kocka A, Kołodziej P, Makuch-Kocka A, Różycka D, Rykowski SK, Nekvinda J, Gruner B, Olejniczak AB. Nematicidal activity of naphthalimide-boron cluster conjugates. Chem Commun (Camb) 2022; 58:2528-2531. [DOI: 10.1039/d1cc07075d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Distinctive biological properties characterize 1,8-naphthalimides and their derivatives. This study presents and compares the activity of a series of compounds where 1,8-naphthalimide fragment was attached either to carborane or metallacarborane...
Collapse
|
13
|
Mitochondria as a potential target for the development of prophylactic and therapeutic drugs against Schistosoma mansoni infection. Antimicrob Agents Chemother 2021; 65:e0041821. [PMID: 34339272 DOI: 10.1128/aac.00418-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Emergence of parasites resistant to praziquantel, the only therapeutic agent, and its ineffectiveness as a prophylactic agent (inactive against the migratory/juvenile Schistosoma mansoni), makes the development of new antischistosomal drugs urgent. The parasite's mitochondrion is an attractive target for drug development because this organelle is essential for survival throughout the parasite's life cycle. We investigated the effects of 116 compounds against Schistosoma mansoni cercariae motility that have been reported to affect mitochondria-related processes in other organisms. Next, eight compounds plus two controls (mefloquine and praziquantel) were selected and assayed against motility of schistosomula (in vitro) and adults (ex vivo). Prophylactic and therapeutic assays were performed using infected mouse models. Inhibition of oxygen consumption rate (OCR) was assayed using Seahorse XFe24 Analyzer. All selected compounds showed excellent prophylactic activity, reducing the worm burden in the lungs to less than 15% that obtained in the vehicle control. Notably, ascofuranone showed the highest activity with a 98% reduction of the worm burden, suggesting the potential for development of ascofuranone as a prophylactic agent. The worm burden of infected mice with S. mansoni at the adult stage was reduced by more than 50% in mice treated with mefloquine, nitazoxanide, amiodarone, ascofuranone, pyrvinium pamoate, or plumbagin. Moreover, adult mitochondrial OCR was severely inhibited by ascofuranone, atovaquone, and nitazoxanide, while pyrvinium pamoate inhibited both mitochondrial and non-mitochondrial OCRs. These results demonstrate that the mitochondria of S. mansoni are feasible target for drug development.
Collapse
|
14
|
Brattig NW, Cheke RA, Garms R. Onchocerciasis (river blindness) - more than a century of research and control. Acta Trop 2021; 218:105677. [PMID: 32857984 DOI: 10.1016/j.actatropica.2020.105677] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/06/2020] [Accepted: 08/21/2020] [Indexed: 12/14/2022]
Abstract
This review summarises more than a century of research on onchocerciasis, also known as river blindness, and its control. River blindness is an infection caused by the tissue filaria Onchocerca volvulus affecting the skin, subcutaneous tissue and eyes and leading to blindness in a minority of infected persons. The parasite is transmitted by its intermediate hosts Simulium spp. which breed in rivers. Featured are history and milestones in onchocerciasis research and control, state-of-the-art data on the parasite, its endobacteria Wolbachia, on the vectors, previous and current prevalence of the infection, its diagnostics, the interaction between the parasite and its host, immune responses and the pathology of onchocerciasis. Detailed information is documented on the time course of control programmes in the afflicted countries in Africa and the Americas, a long road from previous programmes to current successes in control of the transmission of this infectious disease. By development, adjustment and optimization of the control measures, transmission by the vector has been interrupted in foci of countries in the Americas, in Uganda, in Sudan and elsewhere, followed by onchocerciasis eliminations. The current state and future perspectives for control, elimination and eradication within the next 20-30 years are described and discussed. This review contributes to a deeper comprehension of this disease by a tissue-dwelling filaria and it will be helpful in efforts to control and eliminate other filarial infections.
Collapse
|
15
|
Abstract
![]()
Helminths
represent a diverse category of parasitic organisms that
can thrive within a host for years, if not decades, in the absence
of treatment. As such, they must establish mechanisms to subsist off
their hosts, evade the immune system, and develop a niche among the
other cohabiting microbial communities. The complex interplay of biologically
small molecules (collectively known as the metabolome) derived from,
utilized by, or in response to the presence of helminths within a
host is an emerging field of study. In this Perspective, we briefly
summarize the current existing literature, categorize key host–pathogen–microbiome
interfaces that could be studied in the context of the metabolome,
and provide background on mass spectrometry-based metabolomic methodology.
Overall, we hope to provide a comprehensive guide for utilizing metabolomics
in the context of helminthic disease.
Collapse
Affiliation(s)
- Jeffrey D. Whitman
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, California 94110, United States
| | - Judy A. Sakanari
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California 94158, United States
| | - Makedonka Mitreva
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63130, United States
| |
Collapse
|
16
|
Jasmer DP, Rosa BA, Tyagi R, Mitreva M. Rapid determination of nematode cell and organ susceptibility to toxic treatments. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2020; 14:167-182. [PMID: 33125935 PMCID: PMC7593349 DOI: 10.1016/j.ijpddr.2020.10.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/16/2020] [Accepted: 10/19/2020] [Indexed: 12/28/2022]
Abstract
In research focused on the intestine of parasitic nematodes, we recently identified small molecule inhibitors toxic to intestinal cells of larval Ascaris suum (nematode intestinal toxins/toxicants; “NITs”). Some NITs had anthelmintic activity across the phylogenetic diversity of the Nematoda. The whole-worm motility inhibition assay quantified anthelmintic activity, but worm responses to NITs in relation to pathology or affected molecular pathways was not acquired. In this study we extended this research to more comprehensively determine in whole larval A. suum the cells, organ systems, molecular targets, and potential cellular pathways involved in mechanisms of toxicity leading to cell death. The experimental system utilized fluorescent nuclear probes (bisbenzimide, propidium iodide), NITs, an A. suum larval parasite culture system and transcriptional responses (RNA-seq) to NITs. The approach provides for rapid resolution of NIT-induced cell death among organ systems (e.g. intestine, excretory, esophagus, hypodermis and seam cells, and nervous), discriminates among NITs based on cell death profiles, and identifies cells and organ systems with the greatest NIT sensitivity (e.g. intestine and apparent neuronal cells adjacent to the nerve ring). Application was extended to identify cells and organs sensitive to several existing anthelmintics. This approach also resolved intestinal cell death and irreparable damage induced in adult A. suum by two NITs, establishing a new model to elucidate relevant pathologic mechanisms in adult worms. RNA-seq analysis resolved A. suum genes responsive to treatments with three NITs, identifying dihydroorotate dehydrogenase (uridine synthesis) and RAB GTPase(s) (vesicle transport) as potential targets/pathways leading to cell death. A set of genes induced by all three NITs tested suggest common stress or survival responses activated by NITs. Beyond the presented specific lines of research, elements of the overall experimental system presented in this study have broad application toward systematic development of new anthelmintics. A unique rapid cell death assay was developed for parasitic nematodes. Multiple drug-like molecules cause widespread cell death in many organs of A. suum. Multiple cell and organ systems were validated as targets for anthelmintics. Potential drug targets/pathways were implicated in activating cell death processes.
Collapse
Affiliation(s)
- Douglas P Jasmer
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, 99164, USA
| | - Bruce A Rosa
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Rahul Tyagi
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Makedonka Mitreva
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA; Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, St. Louis, MO, 63110, USA; McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri, 63108, USA.
| |
Collapse
|
17
|
Guidi A, Petrella G, Fustaino V, Saccoccia F, Lentini S, Gimmelli R, Di Pietro G, Bresciani A, Cicero DO, Ruberti G. Drug effects on metabolic profiles of Schistosoma mansoni adult male parasites detected by 1H-NMR spectroscopy. PLoS Negl Trop Dis 2020; 14:e0008767. [PMID: 33044962 PMCID: PMC7580944 DOI: 10.1371/journal.pntd.0008767] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 10/22/2020] [Accepted: 09/02/2020] [Indexed: 11/25/2022] Open
Abstract
Schistosomiasis is one of the most devastating neglected tropical parasitic diseases caused by trematodes of the genus Schistosoma. Praziquantel (PZQ) is today the only drug used in humans and animals for the treatment of schistosomiasis but unfortunately it is poorly effective on larval and juvenile stages of the parasite. Therefore, it is urgent the discovery of new drug targets and compounds. We have recently showed that the anti-anginal drug perhexiline maleate (PHX) is very active on multiple developmental stages of Schistosoma mansoni in vitro. It is well known that PHX impacts the lipid metabolism in mammals, but the final target on schistosomes still remains unknown. The aim of this study was to evaluate the ability of 1H nuclear magnetic resonance (NMR) spectroscopy in revealing metabolic perturbations due to PHX treatment of S. mansoni adult male worms. The effects of PHX were compared with the ones induced by vehicle and gambogic acid, in order to detect different metabolic profiles and specificity of the PHX action. Remarkably a list of metabolites associated to PHX-treatment was identified with enrichment in several connected metabolic pathways including also the Kennedy pathway mediating the glycerophospholipid metabolism. Our study represents the first 1H-NMR metabolomic approach to characterize the response of S. mansoni to drug treatment. The obtained "metabolic fingerprint" associated to PHX treatment could represent a strategy for displaying cellular metabolic changes for any given drug and to compare compounds targeting similar or distinct biochemical pathways.
Collapse
Affiliation(s)
- Alessandra Guidi
- Institute of Biochemistry and Cell Biology, National Research Council, Campus A. Buzzati-Traverso, Monterotondo (Rome) Italy
| | - Greta Petrella
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, Rome, Italy
| | - Valentina Fustaino
- Institute of Biochemistry and Cell Biology, National Research Council, Campus A. Buzzati-Traverso, Monterotondo (Rome) Italy
| | - Fulvio Saccoccia
- Institute of Biochemistry and Cell Biology, National Research Council, Campus A. Buzzati-Traverso, Monterotondo (Rome) Italy
| | - Sara Lentini
- Department of Translational Biology, IRBM Science Park Spa, Pomezia (Rome), Italy
| | - Roberto Gimmelli
- Institute of Biochemistry and Cell Biology, National Research Council, Campus A. Buzzati-Traverso, Monterotondo (Rome) Italy
| | - Giulia Di Pietro
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, Rome, Italy
| | - Alberto Bresciani
- Department of Translational Biology, IRBM Science Park Spa, Pomezia (Rome), Italy
| | - Daniel Oscar Cicero
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, Rome, Italy
| | - Giovina Ruberti
- Institute of Biochemistry and Cell Biology, National Research Council, Campus A. Buzzati-Traverso, Monterotondo (Rome) Italy
| |
Collapse
|
18
|
Curran DM, Grote A, Nursimulu N, Geber A, Voronin D, Jones DR, Ghedin E, Parkinson J. Modeling the metabolic interplay between a parasitic worm and its bacterial endosymbiont allows the identification of novel drug targets. eLife 2020; 9:e51850. [PMID: 32779567 PMCID: PMC7419141 DOI: 10.7554/elife.51850] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 07/14/2020] [Indexed: 12/17/2022] Open
Abstract
The filarial nematode Brugia malayi represents a leading cause of disability in the developing world, causing lymphatic filariasis in nearly 40 million people. Currently available drugs are not well-suited to mass drug administration efforts, so new treatments are urgently required. One potential vulnerability is the endosymbiotic bacteria Wolbachia-present in many filariae-which is vital to the worm. Genome scale metabolic networks have been used to study prokaryotes and protists and have proven valuable in identifying therapeutic targets, but have only been applied to multicellular eukaryotic organisms more recently. Here, we present iDC625, the first compartmentalized metabolic model of a parasitic worm. We used this model to show how metabolic pathway usage allows the worm to adapt to different environments, and predict a set of 102 reactions essential to the survival of B. malayi. We validated three of those reactions with drug tests and demonstrated novel antifilarial properties for all three compounds.
Collapse
Affiliation(s)
- David M Curran
- Program in Molecular Medicine, Hospital for Sick ChildrenTorontoCanada
| | - Alexandra Grote
- Department of Biology, Center for Genomics and Systems Biology, New York UniversityNew YorkUnited States
| | - Nirvana Nursimulu
- Program in Molecular Medicine, Hospital for Sick ChildrenTorontoCanada
- Department of Computer Science, University of TorontoTorontoCanada
| | - Adam Geber
- Department of Biology, Center for Genomics and Systems Biology, New York UniversityNew YorkUnited States
| | | | - Drew R Jones
- Department of Biochemistry and Molecular Pharmacology, New York University School of MedicineNew YorkUnited States
| | - Elodie Ghedin
- Department of Biology, Center for Genomics and Systems Biology, New York UniversityNew YorkUnited States
- Department of Epidemiology, School of Global Public Health, New York UniversityNew YorkUnited States
| | - John Parkinson
- Program in Molecular Medicine, Hospital for Sick ChildrenTorontoCanada
- Department of Computer Science, University of TorontoTorontoCanada
- Department of Biochemistry, University of TorontoTorontoCanada
- Department of Molecular Genetics, University of TorontoTorontoCanada
| |
Collapse
|
19
|
Hada A, Kumari C, Phani V, Singh D, Chinnusamy V, Rao U. Host-Induced Silencing of FMRFamide-Like Peptide Genes, flp-1 and flp-12, in Rice Impairs Reproductive Fitness of the Root-Knot Nematode Meloidogyne graminicola. FRONTIERS IN PLANT SCIENCE 2020; 11:894. [PMID: 32765539 PMCID: PMC7379849 DOI: 10.3389/fpls.2020.00894] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/02/2020] [Indexed: 05/26/2023]
Abstract
Rice (Oryza sativa L.) is one of the major staple food crops of the world. The productivity of rice is considerably affected by the root-knot nematode, Meloidogyne graminicola. Modern nematode management strategies targeting the physiological processes have established the potency of use of neuromotor genes for their management. Here, we explored the utility of two FMRFamide like peptide coding genes, Mg-flp-1 and Mg-flp-12 of M. graminicola for its management through host-induced gene silencing (HIGS) using Agrobacterium-mediated transformation of rice. The presence and integration of hairpin RNA (hpRNA) constructs in transgenic lines were confirmed by PCR, qRT-PCR, and Southern and Northern hybridization. Transgenic plants were evaluated against M. graminicola, where phenotypic effect of HIGS was pronounced with reduction in galling by 20-48% in the transgenic plants. This also led to significant decrease in total number of endoparasites by 31-50% for Mg-flp-1 and 34-51% for Mg-flp-12 transgenics. Likewise, number of egg masses per plant and eggs per egg mass also declined significantly in the transgenics, ultimately affecting the multiplication factor, when compared to the wild type plants. This study establishes the effectiveness of the two M. graminicola flp genes for its management and also for gene pyramiding.
Collapse
Affiliation(s)
- Alkesh Hada
- Division of Nematology, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | - Chanchal Kumari
- Division of Nematology, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | - Victor Phani
- Division of Nematology, ICAR-Indian Agricultural Research Institute, New Delhi, India
- Department of Agricultural Entomology, College of Agriculture, Uttar Banga Krishi Viswavidyalaya, Dakshin Dinajpur, India
| | - Divya Singh
- Division of Nematology, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | - Viswanathan Chinnusamy
- Division of Plant Physiology, ICAR—Indian Agricultural Research Institute, New Delhi, India
| | - Uma Rao
- Division of Nematology, ICAR-Indian Agricultural Research Institute, New Delhi, India
| |
Collapse
|
20
|
Guidi A, Prasanth Saraswati A, Relitti N, Gimmelli R, Saccoccia F, Sirignano C, Taglialatela-Scafati O, Campiani G, Ruberti G, Gemma S. (+)-(R)- and (-)-(S)-Perhexiline maleate: Enantioselective synthesis and functional studies on Schistosoma mansoni larval and adult stages. Bioorg Chem 2020; 102:104067. [PMID: 32663671 DOI: 10.1016/j.bioorg.2020.104067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 06/28/2020] [Indexed: 11/26/2022]
Abstract
Schistosomiasis is a neglected tropical disease mainly affecting the poorest tropical and subtropical areas of the world with the impressive number of roughly 200 million infections per year. Schistosomes are blood trematode flukes of the genus Schistosoma causing symptoms in humans and animals. Organ morbidity is caused by the accumulation of parasite eggs and subsequent development of fibrosis. If left untreated, schistosomiasis can result in substantial morbidity and even mortality. Praziquantel (PZQ) is the most effective and widely used compound for the treatment of the disease, in prevention and control programs in the last 30 years. Unfortunately, it has no effect on juvenile immature schistosomes and cannot prevent reinfection or interfere with the schistosome life cycle; moreover drug-resistance represents a serious threat. The search for an alternative or complementary treatment is urgent and drug repurposing could accelerate a solution. The anti-anginal drug perhexiline maleate (PHX) has been previously shown to be effective on larval, juvenile, and adult stages of S. mansoni and to impact egg production in vitro. Since PHX is a racemic mixture of R-(+)- and S-(-)-enantiomers, we designed and realized a stereoselective synthesis of both PHX enantiomers and developed an analytical procedure for the direct quantification of the enantiomeric excess also suitable for semipreparative separation of PHX enantiomers. We next investigated the impact of each enantiomer on viability of newly transformed schistosomula (NTS) and worm pairs of S. mansoni as well as on egg production and vitellarium morphology by in vitro studies. Our results indicate that the R-(+)-PHX is mainly driving the anti-schistosomal activity but that also the S-(-)-PHX possesses a significant activity towards S. mansoni in vitro.
Collapse
Affiliation(s)
- Alessandra Guidi
- Institute of Biochemistry and Cell Biology, National Research Council, Campus A. Buzzati-Traverso via E. Ramarini, 32 00015 Monterotondo (Rome), Italy
| | - A Prasanth Saraswati
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Nicola Relitti
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Roberto Gimmelli
- Institute of Biochemistry and Cell Biology, National Research Council, Campus A. Buzzati-Traverso via E. Ramarini, 32 00015 Monterotondo (Rome), Italy
| | - Fulvio Saccoccia
- Institute of Biochemistry and Cell Biology, National Research Council, Campus A. Buzzati-Traverso via E. Ramarini, 32 00015 Monterotondo (Rome), Italy
| | - Carmina Sirignano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy
| | - Orazio Taglialatela-Scafati
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, via Aldo Moro 2, 53100 Siena, Italy.
| | - Giovina Ruberti
- Institute of Biochemistry and Cell Biology, National Research Council, Campus A. Buzzati-Traverso via E. Ramarini, 32 00015 Monterotondo (Rome), Italy.
| | - Sandra Gemma
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, via Aldo Moro 2, 53100 Siena, Italy.
| |
Collapse
|
21
|
Chitikina SS, Buddiga P, Deb PK, Mailavaram RP, Venugopala KN, Nair AB, Al-Jaidi B, Kar S. Synthesis and anthelmintic activity of some novel (E)-2-methyl/propyl-4-(2-(substitutedbenzylidene)hydrazinyl)-5,6,7,8-tetrahydrobenzo[4,5]thieno[2,3-d]pyrimidines. Med Chem Res 2020. [DOI: 10.1007/s00044-020-02586-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
22
|
Romero N, Areche C, Cubides-Cárdenas J, Escobar N, García-Beltrán O, Simirgiotis MJ, Céspedes Á. In Vitro Anthelmintic Evaluation of Gliricidia sepium, Leucaena leucocephala, and Pithecellobium dulce: Fingerprint Analysis of Extracts by UHPLC-Orbitrap Mass Spectrometry. Molecules 2020; 25:molecules25133002. [PMID: 32630065 PMCID: PMC7412154 DOI: 10.3390/molecules25133002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/17/2020] [Accepted: 05/20/2020] [Indexed: 02/03/2023] Open
Abstract
In the present work, the anthelmintic activity (AA) of ethanolic extracts obtained from Gliricidia sepium, Leucaena leucocephala, and Pithecellobium dulce was evaluated using the third-stage-larval (L3) exsheathment inhibition test (LEIT) and egg hatch test (EHT) on Haemonchus contortus. Extracts were tested at concentrations of 0.3, 0.6, 1.2, 2.5, 5.0, 10, 20, and 40 mg/mL. The larval exsheathment inhibition (LEI) results showed that G. sepium achieved the highest average inhibition of 91.2%, compared with 44.6% for P. dulce and 41.0% for L. leucocephala at a concentration of 40 mg/mL; the corresponding IC50 values were 22.4, 41.7, and 43.3 mg/mL, respectively. The rates of egg hatching inhibition (EHI) at a concentration of 5 mg/mL were 99.5% for G. sepium, 64.2% for P. dulce, and 54% for L. leucocephala; the corresponding IC50 values were 1.9 mg/mL for G. sepium, 3.9 mg/mL for P. dulce, and 4.3 mg/mL for L. leucocephala. The species extracts studied here were also analyzed by ultra-high performance liquid chromatography and Orbitrap high resolution mass spectrometry (UHPLC-Q/Orbitrap/MS/MS), resulting in the compounds' identification associated with AA. Glycosylated flavonoids and methoxyphenols were observed in all three species: fatty acids in G. sepium and P. dulce; phenylpropanoids, anthraquinone glycosides, amino acids and glycosylated phenolic acids in G. sepium; and flavonoids in L. leucocephala. Comparatively, G. sepium presented a greater diversity of compounds potentially active against the control of gastrointestinal nematodes, which was associated with the results obtained in the applied tests.
Collapse
Affiliation(s)
- Néstor Romero
- Departamento de Sanidad Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad del Tolima, Ibagué 730001, Colombia;
- Correspondence:
| | - Carlos Areche
- Departamento de Química, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Nuñoa, Santiago 7800024, Chile;
| | - Jaime Cubides-Cárdenas
- Grupo de Investigación e Innovación en Salud y Bienestar Animal, Laboratorio de Salud Animal, Centro de Investigación Tibaitatá, Agrosavia, Mosquera 250047, Colombia;
| | - Natalia Escobar
- Facultad de Ciencias Agropecuarias, Universidad de Cundinamarca, Fusagasugá 252212, Colombia;
| | - Olimpo García-Beltrán
- Facultad de Ciencias Naturales y Matemáticas, Universidad de Ibagué, Carrera 22 Calle 67, Ibagué 730002, Colombia;
| | | | - Ángel Céspedes
- Departamento de Sanidad Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad del Tolima, Ibagué 730001, Colombia;
| |
Collapse
|
23
|
Jasmer DP, Rosa BA, Tyagi R, Bulman CA, Beerntsen B, Urban JF, Sakanari J, Mitreva M. De novo identification of toxicants that cause irreparable damage to parasitic nematode intestinal cells. PLoS Negl Trop Dis 2020; 14:e0007942. [PMID: 32453724 PMCID: PMC7274465 DOI: 10.1371/journal.pntd.0007942] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 06/05/2020] [Accepted: 05/04/2020] [Indexed: 02/06/2023] Open
Abstract
Efforts to identify new drugs for therapeutic and preventive treatments against parasitic nematodes have gained increasing interest with expanding pathogen omics databases and drug databases from which new anthelmintic compounds might be identified. Here, a novel approach focused on integrating a pan-Nematoda multi-omics data targeted to a specific nematode organ system (the intestinal tract) with evidence-based filtering and chemogenomic screening was undertaken. Based on de novo computational target prioritization of the 3,564 conserved intestine genes in A. suum, exocytosis was identified as a high priority pathway, and predicted inhibitors of exocytosis were tested using the large roundworm (Ascaris suum larval stages), a filarial worm (Brugia pahangi adult and L3), a whipworm (Trichuris muris adult), and the non-parasitic nematode Caenorhabditis elegans. 10 of 13 inhibitors were found to cause rapid immotility in A. suum L3 larvae, and five inhibitors were effective against the three phylogenetically diverse parasitic nematode species, indicating potential for a broad spectrum anthelmintics. Several distinct pathologic phenotypes were resolved related to molting, motility, or intestinal cell and tissue damage using conventional and novel histologic methods. Pathologic profiles characteristic for each inhibitor will guide future research to uncover mechanisms of the anthelmintic effects and improve on drug designs. This progress firmly validates the focus on intestinal cell biology as a useful resource to develop novel anthelmintic strategies.
Collapse
Affiliation(s)
- Douglas P Jasmer
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, United States of America
| | - Bruce A Rosa
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Rahul Tyagi
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Christina A Bulman
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, United States of America
| | - Brenda Beerntsen
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
| | - Joseph F Urban
- U.S. Department of Agriculture, Northeast Area, Agricultural Research Service, Beltsville Agricultural Research Center, Animal Parasite Diseases Laboratory and Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, Maryland, United States of America
| | - Judy Sakanari
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, United States of America
| | - Makedonka Mitreva
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America.,McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
24
|
Riaz MR, Preston GM, Mithani A. MAPPS: A Web-Based Tool for Metabolic Pathway Prediction and Network Analysis in the Postgenomic Era. ACS Synth Biol 2020; 9:1069-1082. [PMID: 32347714 DOI: 10.1021/acssynbio.9b00397] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Comparative and evolutionary analyses of metabolic networks have a wide range of applications, ranging from research into metabolic evolution through to practical applications in drug development, synthetic biology, and biodegradation. We present MAPPS: Metabolic network Analysis and Pathway Prediction Server (https://mapps.lums.edu.pk), a web-based tool to study functions and evolution of metabolic networks using traditional and 'omics data sets. MAPPS provides diverse functionalities including an interactive interface, graphical visualization of results, pathway prediction and network comparison, identification of potential drug targets, in silico metabolic engineering, host-microbe interactions, and ancestral network building. Importantly, MAPPS also allows users to upload custom data, thus enabling metabolic analyses on draft and custom genomes, and has an 'omics pipeline to filter pathway results, making it relevant in today's postgenomic era.
Collapse
Affiliation(s)
- Muhammad Rizwan Riaz
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences (LUMS), DHA, Lahore 54792, Pakistan
| | - Gail M. Preston
- Department of Plant Sciences, University of Oxford, South Parks Road, Oxford OX1 3RB, U.K
| | - Aziz Mithani
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences (LUMS), DHA, Lahore 54792, Pakistan
| |
Collapse
|
25
|
Li F, Zhang H, Li Q, Wu F, Wang Y, Wang Z, Wang X, Huang C. CDCA2 acts as an oncogene and induces proliferation of clear cell renal cell carcinoma cells. Oncol Lett 2020; 19:2466-2474. [PMID: 32194746 PMCID: PMC7039090 DOI: 10.3892/ol.2020.11322] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 12/17/2019] [Indexed: 01/20/2023] Open
Abstract
Cell division cycle-associated 2 (CDCA2) plays an important role in regulating chromosome structure during mitosis. It is highly expressed in oral squamous cell carcinoma, neuroblastoma and lung adenocarcinoma, and its upregulation is positively associated with tumor progression. However, the expression, biological function and underlying mechanisms of the role of CDCA2 in clear cell renal cell carcinoma (ccRCC) remain poorly understood. In the present study, CDCA2 was demonstrated to be upregulated in ccRCC tissues compared with normal kidney tissue, where higher expression was generally associated with the degree of malignancy. Small interfering RNA-mediated knockdown of CDCA2 expression inhibited the viability and proliferation of 786-O and CAKI-1 cells, as measured by an MTT assay, colony formation assay and flow cytometry. Furthermore, western blot analysis suggested that CDCA2 regulates cell proliferation through the cell cycle-associated proteins cyclin D1 and cyclin dependent kinase 4, and the apoptotic protein Bcl-2. In conclusion, the present study indicated that CDCA2 may be an important factor in ccRCC progression and could be a potential therapeutic target in this disease.
Collapse
Affiliation(s)
- Fang Li
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Huahua Zhang
- Medical Research and Experimental Center, Medical College, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Qian Li
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Fei Wu
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Yu Wang
- Medical Research and Experimental Center, Medical College, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Zhenzhen Wang
- Department of Prosthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004 P.R. China
| | - Xiaofei Wang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Chen Huang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China.,Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Xi'an, Shaanxi 710004 P.R. China
| |
Collapse
|
26
|
Zajíčková M, Nguyen LT, Skálová L, Raisová Stuchlíková L, Matoušková P. Anthelmintics in the future: current trends in the discovery and development of new drugs against gastrointestinal nematodes. Drug Discov Today 2019; 25:430-437. [PMID: 31883953 DOI: 10.1016/j.drudis.2019.12.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 12/06/2019] [Accepted: 12/19/2019] [Indexed: 12/21/2022]
Abstract
The control of gastrointestinal nematodes (GINs), the most abundant and serious parasites of livestock, has become difficult because of the limited number of available drugs and fast development of drug resistance. Thus, considerable efforts have been devoted to developing new anthelmintics that are efficient against nematodes, especially resistant species. Here, we summarize the most recent results using various approaches: target-based or high-throughput screening (HTS) of compound libraries; the synthesis of new derivatives or new combinations of current anthelmintics; the repurposing of drugs currently approved for other indications; and lastly, the identification of active plant products. We also evaluate the advantages and disadvantages of each of these approaches.
Collapse
Affiliation(s)
- Markéta Zajíčková
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy in Hradec Králové, Hradec Králové, Czech Republic
| | - Linh Thuy Nguyen
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy in Hradec Králové, Hradec Králové, Czech Republic
| | - Lenka Skálová
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy in Hradec Králové, Hradec Králové, Czech Republic
| | - Lucie Raisová Stuchlíková
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy in Hradec Králové, Hradec Králové, Czech Republic
| | - Petra Matoušková
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy in Hradec Králové, Hradec Králové, Czech Republic.
| |
Collapse
|
27
|
Shivakumara TN, Somvanshi VS, Phani V, Chaudhary S, Hada A, Budhwar R, Shukla RN, Rao U. Meloidogyne incognita (Nematoda: Meloidogynidae) sterol-binding protein Mi-SBP-1 as a target for its management. Int J Parasitol 2019; 49:1061-1073. [PMID: 31733196 DOI: 10.1016/j.ijpara.2019.09.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 09/16/2019] [Accepted: 09/19/2019] [Indexed: 12/16/2022]
Abstract
Meloidogyne incognita is a polyphagous plant-parasitic nematode that causes considerable yield loss in agricultural and horticultural crops. The management options available for M. incognita are extremely limited. Here we identified and characterised a M. incognita homolog of Caenorhabditis elegans sterol-binding protein (Mi-SBP-1), a transcriptional regulator of several lipogenesis pathway genes, and used RNA interference-mediated gene silencing to establish its utility as a target for the management of M. incognita. Mi-sbp-1 is predicted to be a helix-loop-helix domain containing DNA binding transcription factor, and is present in the M. incognita genome in three copies. The RNA-Seq analysis of Mi-sbp-1 silenced second stage juveniles confirmed the key role of this gene in lipogenesis regulation in M. incognita. In vitro and host-induced gene silencing of Mi-sbp-1 in M. incognita second stage juveniles resulted in loss of nematodes' ability to utilise the stored fat reserves, slower nematode development, and reduced parasitism on adzuki bean and tobacco plants. The multiplication factor for the Mi-sbp-1 silenced nematodes on adzuki bean plants was reduced by 51% compared with the control nematodes in which Mi-sbp-1 was not silenced. Transgenic expression of the double-stranded RNA construct of the Mi-sbp-1 gene in tobacco plants caused 40-45% reduction in M. incognita multiplication, 30-43.8% reduction in the number of egg masses, and 33-54% reduction in the number of eggs per egg mass compared with the wild type control plants. Our results confirm that Mi-sbp-1 is a key regulator of lipogenesis in M. incognita and suggest that it can be used as an effective target for its management. The findings of this study can be extended to develop methods to manage other economically important parasitic nematodes.
Collapse
Affiliation(s)
| | - Vishal Singh Somvanshi
- Division of Nematology, ICAR-Indian Agricultural Research Institute, New Delhi 110012, India.
| | - Victor Phani
- Division of Nematology, ICAR-Indian Agricultural Research Institute, New Delhi 110012, India
| | - Sonam Chaudhary
- Division of Nematology, ICAR-Indian Agricultural Research Institute, New Delhi 110012, India
| | - Alkesh Hada
- Division of Nematology, ICAR-Indian Agricultural Research Institute, New Delhi 110012, India
| | - Roli Budhwar
- Bionivid Technology Private Limited, 209, 4th Cross, Kasturi Nagar, Bangalore 560043, India
| | - Rohit Nandan Shukla
- Bionivid Technology Private Limited, 209, 4th Cross, Kasturi Nagar, Bangalore 560043, India
| | - Uma Rao
- Division of Nematology, ICAR-Indian Agricultural Research Institute, New Delhi 110012, India.
| |
Collapse
|
28
|
Marks ND, Winter AD, Gu HY, Maitland K, Gillan V, Ambroz M, Martinelli A, Laing R, MacLellan R, Towne J, Roberts B, Hanks E, Devaney E, Britton C. Profiling microRNAs through development of the parasitic nematode Haemonchus identifies nematode-specific miRNAs that suppress larval development. Sci Rep 2019; 9:17594. [PMID: 31772378 PMCID: PMC6879476 DOI: 10.1038/s41598-019-54154-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/04/2019] [Indexed: 02/05/2023] Open
Abstract
Parasitic nematodes transition between dramatically different free-living and parasitic stages, with correctly timed development and migration crucial to successful completion of their lifecycle. However little is known of the mechanisms controlling these transitions. microRNAs (miRNAs) negatively regulate gene expression post-transcriptionally and regulate development of diverse organisms. Here we used microarrays to determine the expression profile of miRNAs through development and in gut tissue of the pathogenic nematode Haemonchus contortus. Two miRNAs, mir-228 and mir-235, were enriched in infective L3 larvae, an arrested stage analogous to Caenorhabditis elegans dauer larvae. We hypothesized that these miRNAs may suppress development and maintain arrest. Consistent with this, inhibitors of these miRNAs promoted H. contortus development from L3 to L4 stage, while genetic deletion of C. elegans homologous miRNAs reduced dauer arrest. Epistasis studies with C. elegans daf-2 mutants showed that mir-228 and mir-235 synergise with FOXO transcription factor DAF-16 in the insulin signaling pathway. Target prediction suggests that these miRNAs suppress metabolic and transcription factor activity required for development. Our results provide novel insight into the expression and functions of specific miRNAs in regulating nematode development and identify miRNAs and their target genes as potential therapeutic targets to limit parasite survival within the host.
Collapse
Affiliation(s)
- Neil D Marks
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK
| | - Alan D Winter
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK
- West of Scotland Genetic Services, Level 2B, Laboratory Medicine, Queen Elizabeth University Hospital, Govan Road, Glasgow, G51 4TF, UK
| | - Henry Y Gu
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK
| | - Kirsty Maitland
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK
| | - Victoria Gillan
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK
| | - Martin Ambroz
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK
- Department of Biochemical Sciences, Faculty of Pharmacy, Charles University, Hradec Kralove, Czech Republic
| | - Axel Martinelli
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, N20 W10, Kita-ku, Sapporo, Japan
| | - Roz Laing
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK
| | - Rachel MacLellan
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK
| | - Jessica Towne
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK
| | - Brett Roberts
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University Avenue, Glasgow, G12 8QQ, UK
| | - Eve Hanks
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK
| | - Eileen Devaney
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK.
| | - Collette Britton
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK.
| |
Collapse
|
29
|
Cross M, York M, Długosz E, Straub JH, Biberacher S, Herath HMPD, Logan SA, Kim JS, Gasser RB, Ryan JH, Hofmann A. A suicide inhibitor of nematode trehalose-6-phosphate phosphatases. Sci Rep 2019; 9:16165. [PMID: 31700060 PMCID: PMC6838324 DOI: 10.1038/s41598-019-52593-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 08/28/2019] [Indexed: 11/12/2022] Open
Abstract
Protein-based drug discovery strategies have the distinct advantage of providing insights into the molecular mechanisms of chemical effectors. Currently, there are no known trehalose-6-phosphate phosphatase (TPP) inhibitors that possess reasonable inhibition constants and chemical scaffolds amenable to convenient modification. In the present study, we subjected recombinant TPPs to a two-tiered screening approach to evaluate several diverse compound groups with respect to their potential as TPP inhibitors. From a total of 5452 compounds tested, N-(phenylthio)phthalimide was identified as an inhibitor of nematode TPPs with apparent Ki values of 1.0 μM and 0.56 μM against the enzymes from the zoonotic roundworms Ancylostoma ceylanicum and Toxocara canis, respectively. Using site-directed mutagenesis, we demonstrate that this compound acts as a suicide inhibitor that conjugates a strictly conserved cysteine residue in the vicinity of the active site of nematode TPPs. The anthelmintic properties of N-(phenylthio)phthalimide were assessed in whole nematode assays using larvae of the ascaroids T. canis and T. cati, as well as the barber's pole worm Haemonchus contortus. The compound was particularly effective against each of the ascaroids with an IC50 value of 9.3 μM in the survival assay of T. cati larvae, whereas no bioactivity was observed against H. contortus.
Collapse
Affiliation(s)
- Megan Cross
- Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, 4111, Australia
| | - Mark York
- CSIRO Biomedical Manufacturing Program, Clayton, Victoria, 3168, Australia
| | - Ewa Długosz
- Department of Preclinical Sciences, Warsaw University of Life Sciences, 02-787, Warsaw, Poland
| | - Jan Hendrik Straub
- Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, 4111, Australia
| | - Sonja Biberacher
- Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, 4111, Australia
| | - H M P Dilrukshi Herath
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Stephanie A Logan
- CSIRO Biomedical Manufacturing Program, Clayton, Victoria, 3168, Australia
| | - Jeong-Sun Kim
- Department of Chemistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Robin B Gasser
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - John H Ryan
- CSIRO Biomedical Manufacturing Program, Clayton, Victoria, 3168, Australia
| | - Andreas Hofmann
- Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, 4111, Australia.
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria, 3010, Australia.
- Queensland Tropical Health Alliance, Smithfield, Queensland, 4878, Australia.
| |
Collapse
|
30
|
Andrade CH, Neves BJ, Melo-Filho CC, Rodrigues J, Silva DC, Braga RC, Cravo PVL. In Silico Chemogenomics Drug Repositioning Strategies for Neglected Tropical Diseases. Curr Med Chem 2019. [DOI: 10.2174/0929867325666180309114824] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Only ~1% of all drug candidates against Neglected Tropical Diseases (NTDs)
have reached clinical trials in the last decades, underscoring the need for new, safe and effective
treatments. In such context, drug repositioning, which allows finding novel indications
for approved drugs whose pharmacokinetic and safety profiles are already known,
emerging as a promising strategy for tackling NTDs. Chemogenomics is a direct descendent
of the typical drug discovery process that involves the systematic screening of chemical
compounds against drug targets in high-throughput screening (HTS) efforts, for the identification
of lead compounds. However, different to the one-drug-one-target paradigm, chemogenomics
attempts to identify all potential ligands for all possible targets and diseases. In
this review, we summarize current methodological development efforts in drug repositioning
that use state-of-the-art computational ligand- and structure-based chemogenomics approaches.
Furthermore, we highlighted the recent progress in computational drug repositioning
for some NTDs, based on curation and modeling of genomic, biological, and chemical data.
Additionally, we also present in-house and other successful examples and suggest possible solutions
to existing pitfalls.
Collapse
Affiliation(s)
- Carolina Horta Andrade
- LabMol - Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goias, Goiania, GO, 74605-170, Brazil
| | - Bruno Junior Neves
- LabMol - Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goias, Goiania, GO, 74605-170, Brazil
| | - Cleber Camilo Melo-Filho
- LabMol - Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goias, Goiania, GO, 74605-170, Brazil
| | - Juliana Rodrigues
- LabMol - Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goias, Goiania, GO, 74605-170, Brazil
| | - Diego Cabral Silva
- LabMol - Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goias, Goiania, GO, 74605-170, Brazil
| | - Rodolpho Campos Braga
- LabMol - Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goias, Goiania, GO, 74605-170, Brazil
| | - Pedro Vitor Lemos Cravo
- Laboratory of Cheminformatics, Centro Universitario de Anapolis (UniEVANGELICA), Anapolis, GO, 75083-515, Brazil
| |
Collapse
|
31
|
Identification of small molecule enzyme inhibitors as broad-spectrum anthelmintics. Sci Rep 2019; 9:9085. [PMID: 31235822 PMCID: PMC6591293 DOI: 10.1038/s41598-019-45548-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 06/06/2019] [Indexed: 11/18/2022] Open
Abstract
Targeting chokepoint enzymes in metabolic pathways has led to new drugs for cancers, autoimmune disorders and infectious diseases. This is also a cornerstone approach for discovery and development of anthelmintics against nematode and flatworm parasites. Here, we performed omics-driven knowledge-based identification of chokepoint enzymes as anthelmintic targets. We prioritized 10 of 186 phylogenetically conserved chokepoint enzymes and undertook a target class repurposing approach to test and identify new small molecules with broad spectrum anthelmintic activity. First, we identified and tested 94 commercially available compounds using an in vitro phenotypic assay, and discovered 11 hits that inhibited nematode motility. Based on these findings, we performed chemogenomic screening and tested 32 additional compounds, identifying 6 more active hits. Overall, 6 intestinal (single-species), 5 potential pan-intestinal (whipworm and hookworm) and 6 pan-Phylum Nematoda (intestinal and filarial species) small molecule inhibitors were identified, including multiple azoles, Tadalafil and Torin-1. The active hit compounds targeted three different target classes in humans, which are involved in various pathways, including carbohydrate, amino acid and nucleotide metabolism. Last, using representative inhibitors from each target class, we demonstrated in vivo efficacy characterized by negative effects on parasite fecundity in hamsters infected with hookworms.
Collapse
|
32
|
Chhiba-Govindjee VP, van der Westhuyzen CW, Bode ML, Brady D. Bacterial nitrilases and their regulation. Appl Microbiol Biotechnol 2019; 103:4679-4692. [DOI: 10.1007/s00253-019-09776-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 12/25/2022]
|
33
|
Rajasekharan SK, Lee JH, Ravichandran V, Kim JC, Park JG, Lee J. Nematicidal and insecticidal activities of halogenated indoles. Sci Rep 2019; 9:2010. [PMID: 30765810 PMCID: PMC6375993 DOI: 10.1038/s41598-019-38561-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 01/02/2019] [Indexed: 12/25/2022] Open
Abstract
Parasite death via ion channel activations is the hallmark of anthelmintic and antiparasitic drugs. Glutamate gated chloride channel (GluCl) is a prominent targets for drug selection and design in parasitology. We report several iodine-fluorine based lead activators of GluCl by computational studies and structure-activity relationship analysis. 5-Fluoro-4-iodo-1H-pyrrolo [2, 3-b] pyridine and 5-iodoindole were bioactive hits that displayed in vitro anthelmintic and insecticidal activities against Bursaphelenchus xylophilus, Meloidogyne incognita, and Tenebrio molitor. Two important findings stood out: (i) 5F4IPP induced parasite death, and interacted proficiently with Gln219 amino acid of pentameric GluCl in docking analysis, and (ii) 5-iodoindole appeared to act by forming giant vacuoles in nematodes, which led to a form of non-apoptotic death known as methuosis. The study suggests halogenated-indoles and 1H-pyrrolo [2, 3-b] pyridine derivatives be regarded potential biocides for plant-parasitic nematodes and insects, and warrants further research on the mode of actions, and field investigations.
Collapse
Affiliation(s)
| | - Jin-Hyung Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Vinothkannan Ravichandran
- Shandong University-Helmholtz Institute of Biotechnology, School of Life Science, Shandong University, Jinan, P. R. China
| | - Jin-Cheol Kim
- Department of Agricultural Chemistry, Institute of Environmentally Friendly Agriculture, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, Republic of Korea
| | - Jae Gyu Park
- Advanced Bio Convergence Center, Pohang Technopark Foundation, Pohang, 37668, Republic of Korea
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan, 38541, Republic of Korea.
| |
Collapse
|
34
|
Abstract
Parasitic nematodes (roundworms) and platyhelminths (flatworms) cause debilitating chronic infections of humans and animals, decimate crop production and are a major impediment to socioeconomic development. Here we report a broad comparative study of 81 genomes of parasitic and non-parasitic worms. We have identified gene family births and hundreds of expanded gene families at key nodes in the phylogeny that are relevant to parasitism. Examples include gene families that modulate host immune responses, enable parasite migration though host tissues or allow the parasite to feed. We reveal extensive lineage-specific differences in core metabolism and protein families historically targeted for drug development. From an in silico screen, we have identified and prioritized new potential drug targets and compounds for testing. This comparative genomics resource provides a much-needed boost for the research community to understand and combat parasitic worms.
Collapse
|
35
|
Stroehlein AJ, Gasser RB, Hall RS, Young ND. Interactive online application for the prediction, ranking and prioritisation of drug targets in Schistosoma haematobium. Parasit Vectors 2018; 11:605. [PMID: 30482220 PMCID: PMC6257948 DOI: 10.1186/s13071-018-3197-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 11/12/2018] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Human schistosomiasis is a neglected tropical disease caused by parasitic worms of the genus Schistosoma that still affects some 200 million people. The mainstay of schistosomiasis control is a single drug, praziquantel. The reliance on this drug carries a risk of resistance emerging to this anthelmintic, such that research towards alternative anti-schistosomal drugs is warranted. In this context, a number of studies have employed computational approaches to prioritise proteins for investigation as drug targets, based on extensive genomic, transcriptomic and small-molecule data now available. METHODS Here, we established a customisable, online application for the prioritisation of drug targets and applied it, for the first time, to the entire inferred proteome of S. haematobium. This application enables selection of weighted and ranked proteins representing potential drug targets, and integrates transcriptional data, orthology and gene essentiality information as well as drug-drug target associations and chemical properties of predicted ligands. RESULTS Using this application, we defined 25 potential drug targets in S. haematobium that associated with approved drugs, and 3402 targets that (although they could not be linked to any compounds) are conserved among a range of socioeconomically important flatworm species and might represent targets for new trematocides. CONCLUSIONS The online application developed here represents an interactive, customisable, expandable and reproducible drug target ranking and prioritisation approach that should be useful for the prediction of drug targets in schistosomes and other species of parasitic worms in the future. We have demonstrated the utility of this online application by predicting potential drug targets in S. haematobium that can now be evaluated using functional genomics tools and/or small molecules, to establish whether they are indeed essential for parasite survival, and to assist in the discovery of novel anti-schistosomal compounds.
Collapse
Affiliation(s)
- Andreas J. Stroehlein
- Melbourne Veterinary School, Department of Veterinary Biosciences, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Victoria 3010 Australia
| | - Robin B. Gasser
- Melbourne Veterinary School, Department of Veterinary Biosciences, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Victoria 3010 Australia
| | - Ross S. Hall
- Melbourne Veterinary School, Department of Veterinary Biosciences, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Victoria 3010 Australia
| | - Neil D. Young
- Melbourne Veterinary School, Department of Veterinary Biosciences, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Victoria 3010 Australia
| |
Collapse
|
36
|
Shanmugam G, Lee SK, Jeon J. Identification of Potential Nematicidal Compounds against the Pine Wood Nematode, Bursaphelenchus xylophilus through an In Silico Approach. Molecules 2018; 23:molecules23071828. [PMID: 30041426 PMCID: PMC6100573 DOI: 10.3390/molecules23071828] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/18/2018] [Accepted: 07/18/2018] [Indexed: 01/08/2023] Open
Abstract
Bursaphelenchus xylophilus is a destructive phytophagous nematode that mainly infects pine species and causes pine wilt disease (PWD). PWD is one of the most devastating diseases that has damaged the pine forests of eastern Asia and Portugal for the last four decades. B. xylophilus infects healthy pine trees through Monochamus beetles and its subsequent proliferation results in destruction of the infected pine trees. The poor water solubility and high cost of currently used trunk-injected chemicals such as avermectin and abamectin for the prevention of PWD are major concerns. Thus, for the identification of new compounds targeting the different targets, five proteins including cathepsin L-like cystein proteinase, peroxiredoxins, hsp90, venome allergen protein and tubulin that are known to be important for development and pathogenicity of B. xylophilus were selected. The compounds were virtually screened against five proposed targets through molecular docking into hypothetical binding sites located in a homology-built protein model. Of the fifteen nematicides screened, amocarzine, mebendazole and flubendazole were judged to bind best. For these best docked compounds, structural and electronic properties were calculated through density functional theory studies. The results emphasize that these compounds could be potential lead compounds that can be further developed into nematicidal chemical against B. xylophilus. However, further studies are required to ascertain the nematicidal activity of these compounds against phytophagous nematode.
Collapse
Affiliation(s)
- Gnanendra Shanmugam
- Department of Biotechnology, College of Life and Applied Sciences, Yeungnam University, Gyeongsan 38541, Korea.
| | - Sun Keun Lee
- Division of Forest Insect Pests and Diseases, National Institute of Forest Science, Seoul 02455, Korea.
| | - Junhyun Jeon
- Department of Biotechnology, College of Life and Applied Sciences, Yeungnam University, Gyeongsan 38541, Korea.
| |
Collapse
|
37
|
Tyagi R, Maddirala AR, Elfawal M, Fischer C, Bulman CA, Rosa BA, Gao X, Chugani R, Zhou M, Helander J, Brindley PJ, Tseng CC, Greig IR, Sakanari J, Wildman SA, Aroian R, Janetka JW, Mitreva M. Small Molecule Inhibitors of Metabolic Enzymes Repurposed as a New Class of Anthelmintics. ACS Infect Dis 2018; 4:1130-1145. [PMID: 29718656 PMCID: PMC6283408 DOI: 10.1021/acsinfecdis.8b00090] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The enormous prevalence of infections caused by parasitic nematodes worldwide, coupled to the rapid emergence of their resistance to commonly used anthelmintic drugs, presents an urgent need for the discovery of new drugs. Herein, we have identified several classes of small molecules with broad spectrum activity against these pathogens. Previously, we reported the identification of carnitine palmitoyltransferases (CPTs) as a representative class of enzymes as potential targets for metabolic chokepoint intervention that was elucidated from a combination of chemogenomic screening and experimental testing in nematodes. Expanding on these previous findings, we have discovered that several chemical classes of known small molecule inhibitors of mammalian CPTs have potent activity as anthelmintics. Cross-clade efficacy against a broad spectrum of adult parasitic nematodes was demonstrated for multiple compounds from different series. Several analogs of these initial hit compounds were designed and synthesized. The compounds we report represent a good starting point for further lead identification and optimization for development of new anthelmintic drugs with broad spectrum activity and a novel mechanism of action.
Collapse
Affiliation(s)
- Rahul Tyagi
- McDonnell Genome Institute, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, Missouri 63108, USA
| | - Amarendar Reddy Maddirala
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., Box 8231, St. Louis, Missouri 63110, USA
| | - Mostafa Elfawal
- University of Massachusetts Medical School, Suite 219 Biotech 2, 373 Plantation St., Worcester, Massachusetts 01605, USA
| | - Chelsea Fischer
- Department of Pharmaceutical Chemistry, University of California San Francisco, 1700 4th St, San Francisco, California 94158, USA
| | - Christina A. Bulman
- Department of Pharmaceutical Chemistry, University of California San Francisco, 1700 4th St, San Francisco, California 94158, USA
| | - Bruce A. Rosa
- McDonnell Genome Institute, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, Missouri 63108, USA
| | - Xin Gao
- McDonnell Genome Institute, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, Missouri 63108, USA
| | - Ryan Chugani
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., Box 8231, St. Louis, Missouri 63110, USA
| | - Mingzhou Zhou
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., Box 8231, St. Louis, Missouri 63110, USA
| | - Jon Helander
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., Box 8231, St. Louis, Missouri 63110, USA
| | - Paul J. Brindley
- Department of Microbiology, Immunology & Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine and Health Sciences, George Washington University, Ross Hall, Room 521, 2300 Eye Street, NW, Washington, DC 20037, USA
| | - Chih-Chung Tseng
- Kosterlitz Centre for Therapeutics, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, U.K
| | - Iain R. Greig
- Kosterlitz Centre for Therapeutics, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, U.K
| | - Judy Sakanari
- Department of Pharmaceutical Chemistry, University of California San Francisco, 1700 4th St, San Francisco, California 94158, USA
| | - Scott A. Wildman
- UW Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, 1111 Highland Ave., Madison, Wisconsin 53792, USA
| | - Raffi Aroian
- University of Massachusetts Medical School, Suite 219 Biotech 2, 373 Plantation St., Worcester, Massachusetts 01605, USA
| | - James W. Janetka
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., Box 8231, St. Louis, Missouri 63110, USA
| | - Makedonka Mitreva
- McDonnell Genome Institute, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, Missouri 63108, USA
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, 4523 Clayton Ave., CB 8051, St. Louis MO, 63110, USA
| |
Collapse
|
38
|
Paul A, Adnan M, Majumder M, Kar N, Meem M, Rahman MS, Rauniyar AK, Rahman N, Chy MNU, Kabir MSH. Anthelmintic activity of Piper sylvaticum Roxb. (family: Piperaceae): In vitro and in silico studies. CLINICAL PHYTOSCIENCE 2018. [DOI: 10.1186/s40816-018-0077-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
39
|
Kidane ME, Vanderloop BH, Zhou W, Thomas CD, Ramos E, Singha U, Chaudhuri M, Nes WD. Sterol methyltransferase a target for anti-amoeba therapy: towards transition state analog and suicide substrate drug design. J Lipid Res 2017; 58:2310-2323. [PMID: 29042405 PMCID: PMC5711494 DOI: 10.1194/jlr.m079418] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 10/17/2017] [Indexed: 01/18/2023] Open
Abstract
Ergosterol biosynthesis pathways essential to pathogenic protozoa growth and absent from the human host offer new chokepoint targets. Here, we present characterization and cell-based interference of Acanthamoeba spp sterol 24-/28-methylases (SMTs) that catalyze the committed step in C28- and C29-sterol synthesis. Intriguingly, our kinetic analyses suggest that 24-SMT prefers plant cycloartenol whereas 28-SMT prefers 24(28)-methylene lophenol in similar fashion to the substrate preferences of land plant SMT1 and SMT2. Transition state analog-24(R,S),25-epiminolanosterol (EL) and suicide substrate 26,27-dehydrolanosterol (DHL) differentially inhibited trophozoite growth with IC50 values of 7 nM and 6 µM, respectively, and EL yielded 20-fold higher activity than reference drug voriconazole. Against either SMT assayed with native substrate, EL exhibited tight binding ∼Ki 9 nM. Alternatively, DHL is methylated at C26 by 24-SMT that thereby, generates intermediates that complex and inactivate the enzyme, whereas DHL is not productively bound to 28-SMT. Steroidal inhibitors had no effect on human epithelial kidney cell growth or cholesterol biosynthesis at minimum amoebicidal concentrations. We hypothesize the selective inhibition of Acanthamoeba by steroidal inhibitors representing distinct chemotypes may be an efficient strategy for the development of promising compounds to combat amoeba diseases.
Collapse
Affiliation(s)
- Medhanie E Kidane
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409
| | - Boden H Vanderloop
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409
| | - Wenxu Zhou
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409
| | - Crista D Thomas
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409
| | - Emilio Ramos
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409
| | - Ujjal Singha
- Department of Microbiology and Immunology, Meharry Medical College, Nashville, TN 37208
| | - Minu Chaudhuri
- Department of Microbiology and Immunology, Meharry Medical College, Nashville, TN 37208
| | - W David Nes
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409
| |
Collapse
|
40
|
Affiliation(s)
- Sara Lustigman
- Molecular Parasitology, New York Blood Center, New York, NY, United States of America
- * E-mail:
| | - Alexandra Grote
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY, United States of America
| | - Elodie Ghedin
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY, United States of America
- College of Global Public Health, New York University, New York, NY, United States of America
| |
Collapse
|
41
|
Silva MED, Uriostegui MAM, Millán-Orozco J, Gives PMD, Hernández EL, Braga FR, Araújo JVD. Predatory activity of Butlerius nematodes and nematophagous fungi against Haemonchus contortus infective larvae. REVISTA BRASILEIRA DE PARASITOLOGIA VETERINARIA 2017; 26:92-95. [DOI: 10.1590/s1984-29612016091] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 08/23/2016] [Indexed: 01/17/2023]
Abstract
Abstract The purpose of this study was to evaluate the predatory activity of the nematode Butlerius spp. and fungal isolates of Duddingtonia flagrans, Clonostachys rosea, Arthrobotrys musiformis and Trichoderma esau against H. contortus infective larvae (L3) in grass pots. Forty-eight plastic gardening pots containing 140 g of sterile soil were used. Panicum spp. grass seeds (200 mg) were sown into each pot and individually watered with 10 mL of tap water. Twelve days after seeding, the pots were randomly divided into 6 groups (n=8). Two thousand H. contortus infective larvae (L3) were added to each group. Additionally, the following treatments were established: Group 1 – 2000 Butlerius spp. larvae; group 2 – A. musiformis (1x107 conidia); group 3 – T. esau (1x107 conidia); group 4 – C. rosea (1x107 conidia), group 5 – D. flagrans (1x107conidia) and Group 6 – no biological controller (control group). The larval population of H. contortus exposed to Butlerius spp. was reduced by 61.9%. Population reductions of 90.4, 66.7, 61.9 and 85.7% were recorded in the pots containing A. musiformis, T. esau, C. rosea and D. flagrans, respectively. The results of this study indicate that the predatory nematode Butlerius spp. and the assessed fungi display an important predatory activity can be considered suitable potential biological control agents.
Collapse
Affiliation(s)
| | | | - Jair Millán-Orozco
- Instituto Nacional de Investigaciones Forestales, Agricolas y Pecuarias, México
| | | | | | | | | |
Collapse
|
42
|
Sakata-Kato T, Wirth DF. A Novel Methodology for Bioenergetic Analysis of Plasmodium falciparum Reveals a Glucose-Regulated Metabolic Shift and Enables Mode of Action Analyses of Mitochondrial Inhibitors. ACS Infect Dis 2016; 2:903-916. [PMID: 27718558 DOI: 10.1021/acsinfecdis.6b00101] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Given that resistance to all drugs in clinical use has arisen, discovery of new antimalarial drug targets is eagerly anticipated. The Plasmodium mitochondrion has been considered a promising drug target largely based on its significant divergence from the host organelle as well as its involvement in ATP production and pyrimidine biosynthesis. However, the functions of Plasmodium mitochondrial protein complexes and associated metabolic pathways are not fully characterized. Here, we report the development of novel and robust bioenergetic assay protocols for Plasmodium falciparum asexual parasites utilizing a Seahorse Bioscience XFe24 Extracellular Flux Analyzer. These protocols allowed us to simultaneously assess the direct effects of metabolites and inhibitors on mitochondrial respiration and glycolytic activity in real-time with the readout of oxygen consumption rate and extracellular acidification rate. Using saponin-freed parasites at the schizont stage, we found that succinate, malate, glycerol-3-phosphate, and glutamate, but not pyruvate, were able to increase the oxygen consumption rate and that glycerol-3-phosphate dehydrogenase had the largest potential as an electron donor among tested mitochondrial dehydrogenases. Furthermore, we revealed the presence of a glucose-regulated metabolic shift between oxidative phosphorylation and glycolysis. We measured proton leak and reserve capacity and found bioenergetic evidence for oxidative phosphorylation in erythrocytic stage parasites but at a level much lower than that observed in mammalian cells. Lastly, we developed an assay platform for target identification and mode of action studies of mitochondria-targeting antimalarials. This study provides new insights into the bioenergetics and metabolomics of the Plasmodium mitochondria.
Collapse
Affiliation(s)
- Tomoyo Sakata-Kato
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, United States
| | - Dyann F. Wirth
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, United States
| |
Collapse
|
43
|
Guidi A, Lalli C, Perlas E, Bolasco G, Nibbio M, Monteagudo E, Bresciani A, Ruberti G. Discovery and Characterization of Novel Anti-schistosomal Properties of the Anti-anginal Drug, Perhexiline and Its Impact on Schistosoma mansoni Male and Female Reproductive Systems. PLoS Negl Trop Dis 2016; 10:e0004928. [PMID: 27518281 PMCID: PMC4982595 DOI: 10.1371/journal.pntd.0004928] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 07/26/2016] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Schistosomiasis, one of the world's greatest human neglected tropical diseases, is caused by parasitic trematodes of the genus Schistosoma. A unique feature of schistosome biology is that the induction of sexual maturation as well as the maintenance of the differentiation status of female reproductive organs and egg production, necessary for both disease transmission and pathogenesis, are strictly dependent on the male. The treatment and most control initiatives of schistosomiasis rely today on the long-term application of a single drug, praziquantel (PZQ), mostly by campaigns of mass drug administration. PZQ, while very active on adult parasites, has much lower activity against juvenile worms. Monotherapy also favors the selection of drug resistance and, therefore, new drugs are urgently needed. METHODS AND FINDINGS Following the screening of a small compound library with an ATP-based luminescent assay on Schistosoma mansoni schistosomula, we here report the identification and characterization of novel antischistosomal properties of the anti-anginal drug perhexiline maleate (PHX). By phenotypic worm survival assays and confocal microscopy studies we show that PHX, in vitro, has a marked lethal effect on all S. mansoni parasite life stages (newly transformed schistosomula, juvenile and adult worms) of the definitive host. We further demonstrate that sub-lethal doses of PHX significantly impair egg production and lipid depletion within the vitellarium of adult female worms. Moreover, we highlighted tegumental damage in adult male worms and remarkable reproductive system alterations in both female and male adult parasites. The in vivo study in S. mansoni-patent mice showed a notable variability of worm burdens in the individual experiments, with an overall minimal schistosomicidal effect upon PHX treatment. The short PHX half-life in mice, together with its very high rodent plasma proteins binding could be the cause of the modest efficacy of PHX in the schistosomiasis murine model. CONCLUSIONS/SIGNIFICANCE Overall, our data indicate that PHX could represent a promising starting point for novel schistosomicidal drug discovery programmes.
Collapse
Affiliation(s)
- Alessandra Guidi
- National Research Council, Institute of Cell Biology and Neurobiology, Campus A. Buzzati-Traverso Monterotondo, Roma, Italy
| | - Cristiana Lalli
- National Research Council, Institute of Cell Biology and Neurobiology, Campus A. Buzzati-Traverso Monterotondo, Roma, Italy
| | - Emerald Perlas
- European Molecular Biology Laboratory (EMBL), Mouse Biology Unit, Monterotondo, Italy
| | - Giulia Bolasco
- European Molecular Biology Laboratory (EMBL), Mouse Biology Unit, Monterotondo, Italy
| | - Martina Nibbio
- IRBM Science Park, Department of Preclinical Research, Pomezia, Italy
| | - Edith Monteagudo
- IRBM Science Park, Department of Preclinical Research, Pomezia, Italy
| | | | - Giovina Ruberti
- National Research Council, Institute of Cell Biology and Neurobiology, Campus A. Buzzati-Traverso Monterotondo, Roma, Italy
- * E-mail:
| |
Collapse
|
44
|
Luz AL, Lagido C, Hirschey MD, Meyer JN. In Vivo Determination of Mitochondrial Function Using Luciferase-Expressing Caenorhabditis elegans: Contribution of Oxidative Phosphorylation, Glycolysis, and Fatty Acid Oxidation to Toxicant-Induced Dysfunction. CURRENT PROTOCOLS IN TOXICOLOGY 2016; 69:25.8.1-25.8.22. [PMID: 27479364 PMCID: PMC5002950 DOI: 10.1002/cptx.10] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mitochondria are a target of many drugs and environmental toxicants; however, how toxicant-induced mitochondrial dysfunction contributes to the progression of human disease remains poorly understood. To address this issue, in vivo assays capable of rapidly assessing mitochondrial function need to be developed. Here, using the model organism Caenorhabditis elegans, we describe how to rapidly assess the in vivo role of the electron transport chain, glycolysis, or fatty acid oxidation in energy metabolism following toxicant exposure, using a luciferase-expressing ATP reporter strain. Alterations in mitochondrial function subsequent to toxicant exposure are detected by depleting steady-state ATP levels with inhibitors of the mitochondrial electron transport chain, glycolysis, or fatty acid oxidation. Differential changes in ATP following short-term inhibitor exposure indicate toxicant-induced alterations at the site of inhibition. Because a microplate reader is the only major piece of equipment required, this is a highly accessible method for studying toxicant-induced mitochondrial dysfunction in vivo. © 2016 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Anthony L. Luz
- Nicholas School of the Environment, PO Box 90328, Duke University, Durham, North Carolina, 27708
| | - Cristina Lagido
- School of Medical Sciences, Institute of Medical Sciences, Aberdeen AB25 2 ZD, UK
| | - Matthew D. Hirschey
- Department of Medicine, 500 N. Duke St., 50-201, Duke University, Durham, North Carolina, 27708
| | - Joel N. Meyer
- Nicholas School of the Environment, PO Box 90328, Duke University, Durham, North Carolina, 27708
| |
Collapse
|
45
|
Wilhelm C, Harrison OJ, Schmitt V, Pelletier M, Spencer SP, Urban JF, Ploch M, Ramalingam TR, Siegel RM, Belkaid Y. Critical role of fatty acid metabolism in ILC2-mediated barrier protection during malnutrition and helminth infection. J Exp Med 2016; 213:1409-18. [PMID: 27432938 PMCID: PMC4986525 DOI: 10.1084/jem.20151448] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 05/27/2016] [Indexed: 12/17/2022] Open
Abstract
Belkaid et al. show that type 2 innate lymphoid cells rely predominately on fatty acid metabolism during helminth infection and malnutrition. Innate lymphoid cells (ILC) play an important role in many immune processes, including control of infections, inflammation, and tissue repair. To date, little is known about the metabolism of ILC and whether these cells can metabolically adapt in response to environmental signals. Here we show that type 2 innate lymphoid cells (ILC2), important mediators of barrier immunity, predominantly depend on fatty acid (FA) metabolism during helminth infection. Further, in situations where an essential nutrient, such as vitamin A, is limited, ILC2 sustain their function and selectively maintain interleukin 13 (IL-13) production via increased acquisition and utilization of FA. Together, these results reveal that ILC2 preferentially use FAs to maintain their function in the context of helminth infection or malnutrition and propose that enhanced FA usage and FA-dependent IL-13 production by ILC2 could represent a host adaptation to maintain barrier immunity under dietary restriction.
Collapse
Affiliation(s)
- Christoph Wilhelm
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892 Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Oliver J Harrison
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Vanessa Schmitt
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Martin Pelletier
- Immunoregulation Section, Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Sean P Spencer
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892 Department of Pathology and Laboratory Medicine, Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Joseph F Urban
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD 20705
| | - Michelle Ploch
- Immunoregulation Section, Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Thirumalai R Ramalingam
- Immunopathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Richard M Siegel
- Immunoregulation Section, Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Yasmine Belkaid
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
46
|
Anthelmintic effects of a cationic toxin from a South American rattlesnake venom. Toxicon 2016; 116:49-55. [DOI: 10.1016/j.toxicon.2015.11.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 11/19/2015] [Accepted: 11/26/2015] [Indexed: 01/07/2023]
|
47
|
Gasser RB, Schwarz EM, Korhonen PK, Young ND. Understanding Haemonchus contortus Better Through Genomics and Transcriptomics. ADVANCES IN PARASITOLOGY 2016; 93:519-67. [PMID: 27238012 DOI: 10.1016/bs.apar.2016.02.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Parasitic roundworms (nematodes) cause substantial mortality and morbidity in animals globally. The barber's pole worm, Haemonchus contortus, is one of the most economically significant parasitic nematodes of small ruminants worldwide. Although this and related nematodes can be controlled relatively well using anthelmintics, resistance against most drugs in common use has become a major problem. Until recently, almost nothing was known about the molecular biology of H. contortus on a global scale. This chapter gives a brief background on H. contortus and haemonchosis, immune responses, vaccine research, chemotherapeutics and current problems associated with drug resistance. It also describes progress in transcriptomics before the availability of H. contortus genomes and the challenges associated with such work. It then reviews major progress on the two draft genomes and developmental transcriptomes of H. contortus, and summarizes their implications for the molecular biology of this worm in both the free-living and the parasitic stages of its life cycle. The chapter concludes by considering how genomics and transcriptomics can accelerate research on Haemonchus and related parasites, and can enable the development of new interventions against haemonchosis.
Collapse
Affiliation(s)
- R B Gasser
- The University of Melbourne, Parkville, VIC, Australia
| | - E M Schwarz
- The University of Melbourne, Parkville, VIC, Australia; Cornell University, Ithaca, NY, United States
| | - P K Korhonen
- The University of Melbourne, Parkville, VIC, Australia
| | - N D Young
- The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
48
|
McNulty SN, Strübe C, Rosa BA, Martin JC, Tyagi R, Choi YJ, Wang Q, Hallsworth Pepin K, Zhang X, Ozersky P, Wilson RK, Sternberg PW, Gasser RB, Mitreva M. Dictyocaulus viviparus genome, variome and transcriptome elucidate lungworm biology and support future intervention. Sci Rep 2016; 6:20316. [PMID: 26856411 PMCID: PMC4746573 DOI: 10.1038/srep20316] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 10/26/2015] [Indexed: 11/12/2022] Open
Abstract
The bovine lungworm, Dictyocaulus viviparus (order Strongylida), is an important parasite of livestock that causes substantial economic and production losses worldwide. Here we report the draft genome, variome, and developmental transcriptome of D. viviparus. The genome (161 Mb) is smaller than those of related bursate nematodes and encodes fewer proteins (14,171 total). In the first genome-wide assessment of genomic variation in any parasitic nematode, we found a high degree of sequence variability in proteins predicted to be involved host-parasite interactions. Next, we used extensive RNA sequence data to track gene transcription across the life cycle of D. viviparus, and identified genes that might be important in nematode development and parasitism. Finally, we predicted genes that could be vital in host-parasite interactions, genes that could serve as drug targets, and putative RNAi effectors with a view to developing functional genomic tools. This extensive, well-curated dataset should provide a basis for developing new anthelmintics, vaccines, and improved diagnostic tests and serve as a platform for future investigations of drug resistance and epidemiology of the bovine lungworm and related nematodes.
Collapse
Affiliation(s)
- Samantha N McNulty
- The McDonnell Genome Institute, Washington University in St Louis, MO 63108, USA
| | - Christina Strübe
- Institute for Parasitology, University of Veterinary Medicine Hannover, Hannover 30559, Germany
| | - Bruce A Rosa
- The McDonnell Genome Institute, Washington University in St Louis, MO 63108, USA
| | - John C Martin
- The McDonnell Genome Institute, Washington University in St Louis, MO 63108, USA
| | - Rahul Tyagi
- The McDonnell Genome Institute, Washington University in St Louis, MO 63108, USA
| | - Young-Jun Choi
- The McDonnell Genome Institute, Washington University in St Louis, MO 63108, USA
| | - Qi Wang
- The McDonnell Genome Institute, Washington University in St Louis, MO 63108, USA
| | | | - Xu Zhang
- The McDonnell Genome Institute, Washington University in St Louis, MO 63108, USA
| | - Philip Ozersky
- The McDonnell Genome Institute, Washington University in St Louis, MO 63108, USA
| | - Richard K Wilson
- The McDonnell Genome Institute, Washington University in St Louis, MO 63108, USA
| | - Paul W Sternberg
- HHMI, Division of Biology, California Institute of Technology, Pasadena, CA 91125, USA
| | - Robin B Gasser
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Victoria 3010, Australia
| | - Makedonka Mitreva
- The McDonnell Genome Institute, Washington University in St Louis, MO 63108, USA.,Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
49
|
Abstract
Haemonchus contortus is an important pathogen of small ruminants and is therefore a crucially important target for anthelmintic chemotherapy. Its large size and fecundity have been exploited for the development of in vitro screens for anthelmintic discovery that employ larval and adult stages in several formats. The ability of the parasite to develop to the young adult stage in Mongolian jirds (Meriones unguiculatus) provides a useful small animal model that can be used to screen compounds prior to their evaluation in infected sheep. This chapter summarizes the use of H. contortus for anthelmintic discovery, offers a perspective on current strategies in this area and suggests research challenges that could lead to improvements in the anthelmintic discovery process.
Collapse
|
50
|
Dancy BM, Brockway N, Ramadasan-Nair R, Yang Y, Sedensky MM, Morgan PG. Glutathione S-transferase mediates an ageing response to mitochondrial dysfunction. Mech Ageing Dev 2015; 153:14-21. [PMID: 26704446 DOI: 10.1016/j.mad.2015.12.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 12/03/2015] [Accepted: 12/10/2015] [Indexed: 12/30/2022]
Abstract
To understand primary mitochondrial disease, we utilized a complex I-deficient Caenorhabditis elegans mutant, gas-1. These animals strongly upregulate the expression of gst-14 (encoding a glutathione S-transferase). Knockdown of gst-14 dramatically extends the lifespan of gas-1 and increases hydroxynonenal (HNE) modified mitochondrial proteins without improving complex I function. We observed no change in reactive oxygen species levels as measured by Mitosox staining, consistent with a potential role of GST-14 in HNE clearance. The upregulation of gst-14 in gas-1 animals is specific to the pharynx. These data suggest that an HNE-mediated response in the pharynx could be beneficial for lifespan extension in the context of complex I dysfunction in C. elegans. Thus, whereas HNE is typically considered damaging, our work is consistent with recent reports of its role in signaling, and that in this case, the signal is pro-longevity in a model of mitochondrial dysfunction.
Collapse
Affiliation(s)
- Beverley M Dancy
- Center for Developmental Therapeutics, Seattle Children's Research Institute, 1900 9th Avenue, Seattle, WA 98101, USA.
| | - Nicole Brockway
- Center for Developmental Therapeutics, Seattle Children's Research Institute, 1900 9th Avenue, Seattle, WA 98101, USA.
| | - Renjini Ramadasan-Nair
- Center for Developmental Therapeutics, Seattle Children's Research Institute, 1900 9th Avenue, Seattle, WA 98101, USA.
| | - Yoing Yang
- Department of Genetics, Case Western Reserve University, Cleveland, OH 44106, USA.
| | - Margaret M Sedensky
- Center for Developmental Therapeutics, Seattle Children's Research Institute, 1900 9th Avenue, Seattle, WA 98101, USA; Department of Anesthesiology and Pain Medicine, University of Washington, 1959 NE Pacific Street, BB-1469, Seattle, WA 98195, USA.
| | - Philip G Morgan
- Center for Developmental Therapeutics, Seattle Children's Research Institute, 1900 9th Avenue, Seattle, WA 98101, USA; Department of Anesthesiology and Pain Medicine, University of Washington, 1959 NE Pacific Street, BB-1469, Seattle, WA 98195, USA.
| |
Collapse
|