1
|
Collins JB, Dilks CM, Hahnel SR, Rodriguez B, Fox BW, Redman E, Yu J, Cooke B, Sihuta K, Zamanian M, Roy PJ, Schroeder FC, Gilleard JS, Andersen EC. Naturally occurring variation in a cytochrome P450 modifies thiabendazole responses independently of beta-tubulin. PLoS Pathog 2025; 21:e1012602. [PMID: 39808673 PMCID: PMC11771912 DOI: 10.1371/journal.ppat.1012602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 01/27/2025] [Accepted: 12/29/2024] [Indexed: 01/16/2025] Open
Abstract
Widespread anthelmintic resistance has complicated the management of parasitic nematodes. Resistance to the benzimidazole (BZ) drug class is nearly ubiquitous in many species and is associated with mutations in beta-tubulin genes. However, mutations in beta-tubulin alone do not fully explain all BZ resistance. We performed a genome-wide association study using a genetically diverse panel of Caenorhabditis elegans strains to identify loci that contribute to resistance to the BZ drug thiabendazole (TBZ). We identified a quantitative trait locus (QTL) on chromosome V independent of all beta-tubulin genes and overlapping with two promising candidate genes, the cytochrome P450 gene cyp-35D1 and the nuclear hormone receptor nhr-176. Both genes were previously demonstrated to play a role in TBZ metabolism. NHR-176 binds TBZ and induces the expression of CYP-35D1, which metabolizes TBZ. We generated single gene deletions of cyp-35D1 and nhr-176 and found that both genes play a role in TBZ response. A predicted high-impact lysine-to-glutamate substitution at position 267 (K267E) in CYP-35D1 was identified in a sensitive strain, and reciprocal allele replacement strains in different genetic backgrounds were used to show that the lysine allele conferred increased TBZ resistance. Using competitive fitness assays, we found that neither allele was deleterious, but the lysine allele was selected in the presence of TBZ. Additionally, we found that the lysine allele significantly increased the rate of TBZ metabolism compared to the glutamate allele. Moreover, yeast expression assays showed that the lysine version of CYP-35D1 had twice the enzymatic activity of the glutamate allele. To connect our results to parasitic nematodes, we analyzed four Haemonchus contortus cytochrome P450 orthologs but did not find variation at the 267 position in fenbendazole-resistant populations. Overall, we confirmed that variation in this cytochrome P450 gene is the first locus independent of beta-tubulin to play a role in BZ resistance.
Collapse
Affiliation(s)
- J. B. Collins
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Clayton M. Dilks
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
- Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, Illinois, United States of America
| | - Steffen R. Hahnel
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Briana Rodriguez
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Bennett W. Fox
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, United States of America
| | - Elizabeth Redman
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, Canada
| | - Jingfang Yu
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, United States of America
| | - Brittany Cooke
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
- The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada
| | - Kateryna Sihuta
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
- The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada
| | - Mostafa Zamanian
- Department of Pathobiological Sciences, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Peter J. Roy
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
- The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Frank C. Schroeder
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, United States of America
| | - John S. Gilleard
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, Canada
| | - Erik C. Andersen
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
2
|
Shaver AO, Andersen EC. Integrating metabolomics into the diagnosis and investigation of anthelmintic resistance. Trends Parasitol 2024; 40:1097-1106. [PMID: 39572328 DOI: 10.1016/j.pt.2024.10.020] [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: 09/20/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 12/07/2024]
Abstract
Anthelmintic resistance (AR) in parasitic nematodes poses a global health problem in livestock and domestic animals and is an emerging problem in humans. Consequently, we must understand the mechanisms of AR, including target-site resistance (TSR), in which mutations affect drug binding, and non-target site resistance (NTSR), which involves alterations in drug metabolism and detoxification processes. Because much of the focus has been on TSR, NTSR has received less attention. Here, we describe how metabolomics approaches using Caenorhabditis elegans offer the ability to disentangle nematode drug metabolism, identify metabolic changes associated with resistance, uncover novel biomarkers, and enhance diagnostic methods.
Collapse
Affiliation(s)
- Amanda O Shaver
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Erik C Andersen
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
3
|
Sharma N, Au V, Martin K, Edgley ML, Moerman D, Mains PE, Gilleard JS. Multiple UDP glycosyltransferases modulate benzimidazole drug sensitivity in the nematode Caenorhabditis elegans in an additive manner. Int J Parasitol 2024; 54:535-549. [PMID: 38806068 DOI: 10.1016/j.ijpara.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 04/08/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024]
Abstract
Xenobiotic biotransformation is an important modulator of anthelmintic drug potency and a potential mechanism of anthelmintic resistance. Both the free-living nematode Caenorhabditis elegans and the ruminant parasite Haemonchus contortus biotransform benzimidazole drugs by glucose conjugation, likely catalysed by UDP-glycosyltransferase (UGT) enzymes. To identify C. elegans genes involved in benzimidazole drug detoxification, we first used a comparative phylogenetic analysis of UGTs from humans, C. elegans and H. contortus, combined with available RNAseq datasets to identify which of the 63 C. elegans ugt genes are most likely to be involved in benzimidazole drug biotransformation. RNA interference knockdown of 15 prioritized C. elegans genes identified those that sensitized animals to the benzimidazole derivative albendazole (ABZ). Genetic mutations subsequently revealed that loss of ugt-9 and ugt-11 had the strongest effects. The "ugt-9 cluster" includes these genes, together with six other closely related ugts. A CRISPR-Cas-9 deletion that removed seven of the eight ugt-9 cluster genes had greater ABZ sensitivity than the single largest-effect mutation. Furthermore, a double mutant of ugt-22 (which is not a member of the ugt-9 cluster) with the ugt-9 cluster deletion further increased ABZ sensitivity. This additivity of mutant phenotypes suggest that ugt genes act in parallel, which could have several, not mutually exclusive, explanations. ugt mutations have different effects with different benzimidazole derivatives, suggesting that enzymes with different specificities could together more efficiently detoxify drugs. Expression patterns of ugt-9, ugt-11 and ugt-22 gfp reporters differ and so likely act in different tissues which may, at least in part, explain their additive effects on drug potency. Overexpression of ugt-9 alone was sufficient to confer partial ABZ resistance, indicating increasing total UGT activity protects animals. In summary, our results suggest that the multiple UGT enzymes have overlapping but not completely redundant functions in benzimidazole drug detoxification and may represent "druggable" targets to improve benzimidazole drug potency.
Collapse
Affiliation(s)
- Nidhi Sharma
- Host-Parasite Interactions Program, Faculty of Veterinary Medicine, University of Calgary, Alberta, Canada
| | - Vinci Au
- Department of Zoology, Life Sciences Centre, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada
| | - Kiana Martin
- Department of Zoology, Life Sciences Centre, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada
| | - Mark L Edgley
- Department of Zoology, Life Sciences Centre, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada
| | - Don Moerman
- Department of Zoology, Life Sciences Centre, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada
| | - Paul E Mains
- Departments of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| | - John S Gilleard
- Host-Parasite Interactions Program, Faculty of Veterinary Medicine, University of Calgary, Alberta, Canada.
| |
Collapse
|
4
|
Collins JB, Stone SA, Koury EJ, Paredes AG, Shao F, Lovato C, Chen M, Shi R, Li AY, Candal I, Al Moutaa K, Moya ND, Andersen EC. Quantitative tests of albendazole resistance in Caenorhabditis elegans beta-tubulin mutants. Int J Parasitol Drugs Drug Resist 2024; 25:100556. [PMID: 38991432 PMCID: PMC11296247 DOI: 10.1016/j.ijpddr.2024.100556] [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: 04/11/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/13/2024]
Abstract
Benzimidazole (BZ) anthelmintics are among the most important treatments for parasitic nematode infections in the developing world. Widespread BZ resistance in veterinary parasites and emerging resistance in human parasites raise major concerns for the continued use of BZs. Knowledge of the mechanisms of resistance is necessary to make informed treatment decisions and circumvent resistance. Benzimidazole resistance has traditionally been associated with mutations and natural variants in the C. elegans beta-tubulin gene ben-1 and orthologs in parasitic species. However, variants in ben-1 alone do not explain the differences in BZ responses across parasite populations. Here, we examined the roles of five C. elegans beta-tubulin genes (tbb-1, mec-7, tbb-4, ben-1, and tbb-6) in the BZ response as well as to determine if another beta-tubulin acts redundantly with ben-1. We generated C. elegans strains with a loss of each beta-tubulin gene, as well as strains with a loss of tbb-1, mec-7, tbb-4, or tbb-6 in a genetic background that also lacks ben-1. We found that the loss of ben-1 conferred the maximum level of resistance following exposure to a single concentration of albendazole, and the loss of a second beta-tubulin gene did not alter the level of resistance. However, additional traits other than larval development could be affected by the loss of additional beta-tubulins, and the roles of other beta-tubulin genes might be revealed at different albendazole concentrations. Therefore, further work is needed to fully define the possible roles of other beta-tubulin genes in the BZ response.
Collapse
Affiliation(s)
- J B Collins
- Department of Biology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Skyler A Stone
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA
| | - Emily J Koury
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA
| | - Anna G Paredes
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA
| | - Fiona Shao
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA
| | - Crystal Lovato
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA
| | - Michael Chen
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA
| | - Richelle Shi
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA
| | - Anwyn Y Li
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA
| | - Isa Candal
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA
| | - Khadija Al Moutaa
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA
| | - Nicolas D Moya
- Department of Biology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Erik C Andersen
- Department of Biology, Johns Hopkins University, Baltimore, MD, 21218, USA.
| |
Collapse
|
5
|
Hellinga JR, Krücken J, Schulenburg H, von Samson-Himmelstjerna G. Use of Viscous medium to study anthelmintic drug action in Caenorhabditis elegans. Sci Rep 2024; 14:12756. [PMID: 38830930 PMCID: PMC11148144 DOI: 10.1038/s41598-024-63090-z] [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: 03/12/2024] [Accepted: 05/24/2024] [Indexed: 06/05/2024] Open
Abstract
Caenorhabditis elegans is an appealing tool for experimental evolution and for working with antiparasitic drugs, from understanding the molecular mechanisms of drug action and resistance to uncover new drug targets. We present a new methodology for studying the impact of antiparasitic drugs in C. elegans. Viscous medium was initially designed for C. elegans maintenance during long-term evolution experiments. Viscous medium provides a less structured environment than the standard nematode growth media agar, yet the bacteria food source remains suspended. Further, the Viscous medium offers the worm population enough support to move freely, mate, and reproduce at a rate comparable to standard agar cultures. Here, the Viscous medium was adapted for use in antiparasitic research. We observed a similar sensitivity of C. elegans to anthelmintic drugs as in standard liquid media and statistical difference to the standard agar media through a larval development assay. Using Viscous medium in C. elegans studies will considerably improve antiparasitic resistance research, and this medium could be used in studies aimed at understanding long-term multigenerational drug activity.
Collapse
Affiliation(s)
- Jacqueline R Hellinga
- Institute für Parasitologie und Tropenveterinärmedizin, Freie Universität Berlin, Robert von Ostertag Str. 7, 14163, Berlin, Germany
| | - Jürgen Krücken
- Institute für Parasitologie und Tropenveterinärmedizin, Freie Universität Berlin, Robert von Ostertag Str. 7, 14163, Berlin, Germany
| | - Hinrich Schulenburg
- Zoologisches Institut, Christian-Albrechts-Universität Zu Kiel, Am Botanischen Garten 1-9, 24118, Kiel, Germany
| | - Georg von Samson-Himmelstjerna
- Institute für Parasitologie und Tropenveterinärmedizin, Freie Universität Berlin, Robert von Ostertag Str. 7, 14163, Berlin, Germany.
| |
Collapse
|
6
|
Davie T, Serrat X, Imhof L, Snider J, Štagljar I, Keiser J, Hirano H, Watanabe N, Osada H, Fraser AG. Identification of a family of species-selective complex I inhibitors as potential anthelmintics. Nat Commun 2024; 15:3367. [PMID: 38719808 PMCID: PMC11079024 DOI: 10.1038/s41467-024-47331-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 03/28/2024] [Indexed: 05/12/2024] Open
Abstract
Soil-transmitted helminths (STHs) are major pathogens infecting over a billion people. There are few classes of anthelmintics and there is an urgent need for new drugs. Many STHs use an unusual form of anaerobic metabolism to survive the hypoxic conditions of the host gut. This requires rhodoquinone (RQ), a quinone electron carrier. RQ is not made or used by vertebrate hosts making it an excellent therapeutic target. Here we screen 480 structural families of natural products to find compounds that kill Caenorhabditis elegans specifically when they require RQ-dependent metabolism. We identify several classes of compounds including a family of species-selective inhibitors of mitochondrial respiratory complex I. These identified complex I inhibitors have a benzimidazole core and we determine key structural requirements for activity by screening 1,280 related compounds. Finally, we show several of these compounds kill adult STHs. We suggest these species-selective complex I inhibitors are potential anthelmintics.
Collapse
Affiliation(s)
- Taylor Davie
- The Donnelly Centre, University of Toronto, 160 College Street, Toronto, M5S 3E1, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Xènia Serrat
- The Donnelly Centre, University of Toronto, 160 College Street, Toronto, M5S 3E1, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Lea Imhof
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, CH-4123, Allschwil, Switzerland
- University of Basel, CH-4000, Basel, Switzerland
| | - Jamie Snider
- The Donnelly Centre, University of Toronto, 160 College Street, Toronto, M5S 3E1, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Igor Štagljar
- The Donnelly Centre, University of Toronto, 160 College Street, Toronto, M5S 3E1, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Mediterranean Institute for Life Sciences, Meštrovićevo Šetalište 45, HR-21000, Split, Croatia
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Jennifer Keiser
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, CH-4123, Allschwil, Switzerland
- University of Basel, CH-4000, Basel, Switzerland
| | - Hiroyuki Hirano
- Chemical Resource Development Research Unit, RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako Saitama, 351-0198, Japan
| | - Nobumoto Watanabe
- Chemical Resource Development Research Unit, RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako Saitama, 351-0198, Japan
| | - Hiroyuki Osada
- Chemical Resource Development Research Unit, RIKEN Center for Sustainable Resource Science, 2-1 Hirosawa, Wako Saitama, 351-0198, Japan
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo, 141-0021, Japan
| | - Andrew G Fraser
- The Donnelly Centre, University of Toronto, 160 College Street, Toronto, M5S 3E1, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
7
|
Shaver AO, Miller IR, Schaye ES, Moya ND, Collins JB, Wit J, Blanco AH, Shao FM, Andersen EJ, Khan SA, Paredes G, Andersen EC. Quantifying the fitness effects of resistance alleles with and without anthelmintic selection pressure using Caenorhabditis elegans. PLoS Pathog 2024; 20:e1012245. [PMID: 38768235 PMCID: PMC11142691 DOI: 10.1371/journal.ppat.1012245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/31/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024] Open
Abstract
Albendazole (a benzimidazole) and ivermectin (a macrocyclic lactone) are the two most commonly co-administered anthelmintic drugs in mass-drug administration programs worldwide. Despite emerging resistance, we do not fully understand the mechanisms of resistance to these drugs nor the consequences of delivering them in combination. Albendazole resistance has primarily been attributed to variation in the drug target, a beta-tubulin gene. Ivermectin targets glutamate-gated chloride channels (GluCls), but it is unknown whether GluCl genes are involved in ivermectin resistance in nature. Using Caenorhabditis elegans, we defined the fitness costs associated with loss of the drug target genes singly or in combinations of the genes that encode GluCl subunits. We quantified the loss-of-function effects on three traits: (i) multi-generational competitive fitness, (ii) fecundity, and (iii) development. In competitive fitness and development assays, we found that a deletion of the beta-tubulin gene ben-1 conferred albendazole resistance, but ivermectin resistance required the loss of two GluCl genes (avr-14 and avr-15). The fecundity assays revealed that loss of ben-1 did not provide any fitness benefit in albendazole conditions and that no GluCl deletion mutants were resistant to ivermectin. Next, we searched for evidence of multi-drug resistance across the three traits. Loss of ben-1 did not confer resistance to ivermectin, nor did loss of any single GluCl subunit or combination confer resistance to albendazole. Finally, we assessed the development of 124 C. elegans wild strains across six benzimidazoles and seven macrocyclic lactones to identify evidence of multi-drug resistance between the two drug classes and found a strong phenotypic correlation within a drug class but not across drug classes. Because each gene affects various aspects of nematode physiology, these results suggest that it is necessary to assess multiple fitness traits to evaluate how each gene contributes to anthelmintic resistance.
Collapse
Affiliation(s)
- Amanda O. Shaver
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
- Dept. of Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Isabella R. Miller
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Etta S. Schaye
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Nicolas D. Moya
- Dept. of Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - J. B. Collins
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
- Dept. of Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Janneke Wit
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Alyssa H. Blanco
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Fiona M. Shao
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Elliot J. Andersen
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Sharik A. Khan
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Gracie Paredes
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Erik C. Andersen
- Dept. of Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
8
|
Collins J, Stone SA, Koury EJ, Paredes AG, Shao F, Lovato C, Chen M, Shi R, Li AY, Candal I, Al Moutaa K, Moya N, Andersen EC. Quantitative tests of albendazole resistance in beta-tubulin mutants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.11.589070. [PMID: 38665774 PMCID: PMC11044196 DOI: 10.1101/2024.04.11.589070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Benzimidazole (BZ) anthelmintics are among the most important treatments for parasitic nematode infections in the developing world. Widespread BZ resistance in veterinary parasites and emerging resistance in human parasites raise major concerns for the continued use of BZs. Knowledge of the mechanisms of resistance is necessary to make informed treatment decisions and circumvent resistance. Benzimidazole resistance has traditionally been associated with mutations and natural variants in the C. elegans beta-tubulin gene ben-1 and orthologs in parasitic species. However, variants in ben-1 alone do not explain the differences in BZ responses across parasite populations. Here, we examine the roles of five C. elegans beta-tubulin genes (tbb-1, mec-7, tbb-4, ben-1, and tbb-6) to identify the role each gene plays in BZ response. We generated C. elegans strains with a loss of each beta-tubulin gene, as well as strains with a loss of tbb-1, mec-7, tbb-4, or tbb-6 in a genetic background that also lacks ben-1 to test beta-tubulin redundancy in BZ response. We found that only the individual loss of ben-1 conferred a substantial level of BZ resistance, although the loss of tbb-1 was found to confer a small benefit in the presence of albendazole (ABZ). The loss of ben-1 was found to confer an almost complete rescue of animal development in the presence of 30 μM ABZ, likely explaining why no additive effects caused by the loss of a second beta-tubulin were observed. We demonstrate that ben-1 is the only beta-tubulin gene in C. elegans where loss confers substantial BZ resistance.
Collapse
Affiliation(s)
- J.B. Collins
- Department of Biology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Skyler A. Stone
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Emily J. Koury
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Anna G. Paredes
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Fiona Shao
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Crystal Lovato
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Michael Chen
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Richelle Shi
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Anwyn Y. Li
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Isa Candal
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Khadija Al Moutaa
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Nicolas Moya
- Department of Biology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Erik C. Andersen
- Department of Biology, Johns Hopkins University, Baltimore, MD, 21218, USA
| |
Collapse
|
9
|
McKean EL, Grill E, Choi YJ, Mitreva M, O'Halloran DM, Hawdon JM. Altered larval activation response associated with multidrug resistance in the canine hookworm Ancylostoma caninum. Parasitology 2024; 151:271-281. [PMID: 38163962 PMCID: PMC11007283 DOI: 10.1017/s0031182023001385] [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: 07/12/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 01/03/2024]
Abstract
Parasitic gastrointestinal nematodes pose significant health risks to humans, livestock, and companion animals, and their control relies heavily on the use of anthelmintic drugs. Overuse of these drugs has led to the emergence of resistant nematode populations. Herein, a naturally occurring isolate (referred to as BCR) of the dog hookworm, Ancylostoma caninum, that is resistant to 3 major classes of anthelmintics is characterized. Various drug assays were used to determine the resistance of BCR to thiabendazole, ivermectin, moxidectin and pyrantel pamoate. When compared to a drug-susceptible isolate of A. caninum, BCR was shown to be significantly resistant to all 4 of the drugs tested. Multiple single nucleotide polymorphisms have been shown to impart benzimidazole resistance, including the F167Y mutation in the β-tubulin isotype 1 gene, which was confirmed to be present in BCR through molecular analysis. The frequency of the resistant allele in BCR was 76.3% following its first passage in the lab, which represented an increase from approximately 50% in the founding hookworm population. A second, recently described mutation in codon 134 (Q134H) was also detected at lower frequency in the BCR population. Additionally, BCR exhibits an altered larval activation phenotype compared to the susceptible isolate, suggesting differences in the signalling pathways involved in the activation process which may be associated with resistance. Further characterization of this isolate will provide insights into the mechanisms of resistance to macrocyclic lactones and tetrahydropyrimidine anthelmintics.
Collapse
Affiliation(s)
- Elise L. McKean
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC, USA
- Department of Biological Sciences, The George Washington University, Washington, DC, USA
| | - Emilia Grill
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC, USA
| | - Young-Jun Choi
- Infectious Diseases Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Makedonka Mitreva
- Infectious Diseases Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA
| | - Damien M. O'Halloran
- Department of Biological Sciences, The George Washington University, Washington, DC, USA
| | - John M. Hawdon
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC, USA
| |
Collapse
|
10
|
Shaver AO, Miller IR, Schaye ES, Moya ND, Collins J, Wit J, Blanco AH, Shao FM, Andersen EJ, Khan SA, Paredes G, Andersen EC. Quantifying the fitness effects of resistance alleles with and without anthelmintic selection pressure using Caenorhabditis elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.01.578300. [PMID: 38370666 PMCID: PMC10871296 DOI: 10.1101/2024.02.01.578300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Albendazole and ivermectin are the two most commonly co-administered anthelmintic drugs in mass-drug administration programs worldwide. Despite emerging resistance, we do not fully understand the mechanisms of resistance to these drugs nor the consequences of delivering them in combination. Albendazole resistance has primarily been attributed to variation in the drug target, a beta-tubulin gene. Ivermectin targets glutamate-gated chloride channel (GluCl) genes, but it is unknown whether these genes are involved in ivermectin resistance in nature. Using Caenorhabditis elegans, we defined the fitness costs associated with loss of the drug target genes singly or in combinations of the genes that encode GluCl subunits. We quantified the loss-of function effects on three traits: (i) multi-generational competitive fitness, (ii) fecundity, and (iii) development. In competitive fitness and development assays, we found that a deletion of the beta-tubulin gene ben-1 conferred albendazole resistance, but ivermectin resistance required loss of two GluCl genes (avr-14 and avr-15) or loss of three GluCl genes (avr-14, avr-15, and glc-1). The fecundity assays revealed that loss of ben-1 did not provide any fitness benefit in albendazole and that no GluCl deletion mutants were resistant to ivermectin. Next, we searched for evidence of multi-drug resistance across the three traits. Loss of ben-1 did not confer resistance to ivermectin, nor did loss of any single GluCl subunit or combination confer resistance to albendazole. Finally, we assessed the development of 124 C. elegans wild strains across six benzimidazoles and seven macrocyclic lactones to identify evidence of multi-drug resistance between the two drug classes and found a strong phenotypic correlation within a drug class but not across drug classes. Because each gene affects various aspects of nematode physiology, these results suggest that it is necessary to assess multiple fitness traits to evaluate how each gene contributes to anthelmintic resistance.
Collapse
Affiliation(s)
- Amanda O. Shaver
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Isabella R. Miller
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Etta S. Schaye
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Nicolas D. Moya
- Dept. of Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - J.B. Collins
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Janneke Wit
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Alyssa H. Blanco
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Fiona M. Shao
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Elliot J. Andersen
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Sharik A. Khan
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Gracie Paredes
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Erik C. Andersen
- Dept. of Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
11
|
Shanley HT, Taki AC, Nguyen N, Wang T, Byrne JJ, Ang CS, Leeming MG, Nie S, Williamson N, Zheng Y, Young ND, Korhonen PK, Hofmann A, Wells TNC, Jabbar A, Sleebs BE, Gasser RB. Structure activity relationship and target prediction for ABX464 analogues in Caenorhabditis elegans. Bioorg Med Chem 2024; 98:117540. [PMID: 38134663 DOI: 10.1016/j.bmc.2023.117540] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/20/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023]
Abstract
Global challenges with treatment failures and/or widespread resistance in parasitic worms against commercially available anthelmintics lend impetus to the development of new anthelmintics with novel mechanism(s) of action. The free-living nematode Caenorhabditis elegans is an important model organism used for drug discovery, including the screening and structure-activity investigation of new compounds, and target deconvolution. Previously, we conducted a whole-organism phenotypic screen of the 'Pandemic Response Box' (from Medicines for Malaria Venture, MMV) and identified a hit compound, called ABX464, with activity against C. elegans and a related, parasitic nematode, Haemonchus contortus. Here, we tested a series of 44 synthesized analogues to explore the pharmacophore of activity on C. elegans and revealed five compounds whose potency was similar or greater than that of ABX464, but which were not toxic to human hepatoma (HepG2) cells. Subsequently, we employed thermal proteome profiling (TPP), protein structure prediction and an in silico-docking algorithm to predict ABX464-target candidates. Taken together, the findings from this study contribute significantly to the early-stage drug discovery of a new nematocide based on ABX464. Future work is aimed at validating the ABX464-protein interactions identified here, and at assessing ABX464 and associated analogues against a panel of parasitic nematodes, towards developing a new anthelmintic with a mechanism of action that is distinct from any of the compounds currently-available commercially.
Collapse
Affiliation(s)
- Harrison T Shanley
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia; Chemical Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Aya C Taki
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Nghi Nguyen
- Chemical Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Tao Wang
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Joseph J Byrne
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Ching-Seng Ang
- Melbourne Mass Spectrometry and Proteomics Facility, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Michael G Leeming
- Melbourne Mass Spectrometry and Proteomics Facility, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Shuai Nie
- Melbourne Mass Spectrometry and Proteomics Facility, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Nicholas Williamson
- Melbourne Mass Spectrometry and Proteomics Facility, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yuanting Zheng
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Neil D Young
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Pasi K Korhonen
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Andreas Hofmann
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia; National Reference Centre for Authentic Food, Max Rubner-Institut, 95326 Kulmbach, Germany
| | - Tim N C Wells
- Medicines for Malaria Venture (MMV), 1215 Geneva, Switzerland
| | - Abdul Jabbar
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Brad E Sleebs
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia; Chemical Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.
| | - Robin B Gasser
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
12
|
Kamal M, Mukherjee S, Joshi B, Sindhu ZUD, Wangchuk P, Haider S, Ahmed N, Talukder MH, Geary TG, Yadav AK. Model nematodes as a practical innovation to promote high throughput screening of natural products for anthelmintics discovery in South Asia: Current challenges, proposed practical and conceptual solutions. Mol Biochem Parasitol 2023; 256:111594. [PMID: 37730126 DOI: 10.1016/j.molbiopara.2023.111594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/27/2023] [Accepted: 09/12/2023] [Indexed: 09/22/2023]
Abstract
With the increasing prevalence of anthelmintic resistance in animals recorded globally, and the threat of resistance in human helminths, the need for novel anthelmintic drugs is greater than ever. Most research aimed at discovering novel anthelmintic leads relies on high throughput screening (HTS) of large libraries of synthetic small molecules in industrial and academic settings in developed countries, even though it is the tropical countries that are most plagued by helminth infections. Tropical countries, however, have the advantage of possessing a rich flora that may yield natural products (NP) with promising anthelmintic activity. Focusing on South Asia, which produces one of the world's highest research outputs in NP and NP-based anthelmintic discovery, we find that limited basic research and funding, a lack of awareness of the utility of model organisms, poor industry-academia partnerships and lack of technological innovations greatly limit anthelmintics research in the region. Here we propose that utilizing model organisms including the free-living nematode Caenorhabditis elegans, that can potentially allow rapid target identification of novel anthelmintics, and Oscheius tipulae, a closely related, free-living nematode which is found abundantly in soil in hotter temperatures, could be a much-needed innovation that can enable cost-effective and efficient HTS of NPs for discovering compounds with anthelmintic/antiparasitic potential in South Asia and other tropical regions that historically have devoted limited funding for such research. Additionally, increased collaborations at the national, regional and international level between parasitologists and pharmacologists/ethnobotanists, setting up government-industry-academia partnerships to fund academic research, creating a centralized, regional collection of plant extracts or purified NPs as a dereplication strategy and HTS library, and holding regional C. elegans/O. tipulae-based anthelmintics workshops and conferences to share knowledge and resources regarding model organisms may collectively promote and foster a NP-based anthelmintics landscape in South Asia and beyond.
Collapse
Affiliation(s)
- Muntasir Kamal
- Department of Molecular Genetics, University of Toronto, Toronto, Canada.
| | - Suprabhat Mukherjee
- Department of Animal Science, Kazi Nazrul University, Asansol 713340, West Bengal, India
| | - Bishnu Joshi
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Zia-Ud-Din Sindhu
- Department of Parasitology, University of Agriculture Faisalabad, Pakistan
| | - Phurpa Wangchuk
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns Campus, QLD 4878, Australia
| | | | - Nurnabi Ahmed
- Department of Parasitology, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | | | - Timothy G Geary
- Institute of Parasitology, McGill University, Montreal, Canada; School of Biological Sciences, Queen's University-Belfast, Belfast, NI, UK
| | - Arun K Yadav
- Department of Zoology, North-Eastern Hill University, Shillong 793022, India
| |
Collapse
|
13
|
Rehborg EG, Wheeler NJ, Zamanian M. Mapping resistance-associated anthelmintic interactions in the model nematode Caenorhabditis elegans. PLoS Negl Trop Dis 2023; 17:e0011705. [PMID: 37883578 PMCID: PMC10629664 DOI: 10.1371/journal.pntd.0011705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 11/07/2023] [Accepted: 10/07/2023] [Indexed: 10/28/2023] Open
Abstract
Parasitic nematodes infect billions of people and are mainly controlled by anthelmintic mass drug administration (MDA). While there are growing efforts to better understand mechanisms of anthelmintic resistance in human and animal populations, it is unclear how resistance mechanisms that alter susceptibility to one drug affect the interactions and efficacy of drugs used in combination. Mutations that alter drug permeability across primary nematode barriers have been identified as potential resistance mechanisms using the model nematode Caenorhabditis elegans. We leveraged high-throughput assays in this model system to measure altered anthelmintic susceptibility in response to genetic perturbations of potential cuticular, amphidial, and alimentary routes of drug entry. Mutations in genes associated with these tissue barriers differentially altered susceptibility to the major anthelmintic classes (macrocyclic lactones, benzimidazoles, and nicotinic acetylcholine receptor agonists) as measured by animal development. We investigated two-way anthelmintic interactions across C. elegans genetic backgrounds that confer resistance or hypersensitivity to one or more drugs. We observe that genetic perturbations that alter susceptibility to a single drug can shift the drug interaction landscape and lead to the appearance of novel synergistic and antagonistic interactions. This work establishes a framework for investigating combinatorial therapies in model nematodes that can potentially be translated to amenable parasite species.
Collapse
Affiliation(s)
- Elena G. Rehborg
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Nicolas J. Wheeler
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Mostafa Zamanian
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
14
|
Wit J, Dilks CM, Zhang G, Guisbert KSK, Zdraljevic S, Guisbert E, Andersen EC. Praziquantel inhibits Caenorhabditis elegans development and species-wide differences might be cct-8-dependent. PLoS One 2023; 18:e0286473. [PMID: 37561720 PMCID: PMC10414639 DOI: 10.1371/journal.pone.0286473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/21/2023] [Indexed: 08/12/2023] Open
Abstract
Anthelmintic drugs are used to treat parasitic roundworm and flatworm infections in humans and other animals. Caenorhabditis elegans is an established model to investigate anthelmintics used to treat roundworms. In this study, we use C. elegans to examine the mode of action and the mechanisms of resistance against the flatworm anthelmintic drug praziquantel (PZQ), used to treat trematode and cestode infections. We found that PZQ inhibited development and that this developmental delay varies by genetic background. Interestingly, both enantiomers of PZQ are equally effective against C. elegans, but the right-handed PZQ (R-PZQ) is most effective against schistosome infections. We conducted a genome-wide association mapping with 74 wild C. elegans strains to identify a region on chromosome IV that is correlated with differential PZQ susceptibility. Five candidate genes in this region: cct-8, znf-782, Y104H12D.4, Y104H12D.2, and cox-18, might underlie this variation. The gene cct-8, a subunit of the protein folding complex TRiC, has variation that causes a putative protein coding change (G226V), which is correlated with reduced developmental delay. Gene expression analysis suggests that this variant correlates with slightly increased expression of both cct-8 and hsp-70. Acute exposure to PZQ caused increased expression of hsp-70, indicating that altered TRiC function might be involved in PZQ responses. To test if this variant affects development upon exposure to PZQ, we used CRISPR-Cas9 genome editing to introduce the V226 allele into the N2 genetic background (G226) and the G226 allele into the JU775 genetic background (V226). These experiments revealed that this variant was not sufficient to explain the effects of PZQ on development. Nevertheless, this study shows that C. elegans can be used to study PZQ mode of action and resistance mechanisms. Additionally, we show that the TRiC complex requires further evaluation for PZQ responses in C. elegans.
Collapse
Affiliation(s)
- Janneke Wit
- Molecular Biosciences, Northwestern University, Evanston, IL, United States of America
| | - Clayton M. Dilks
- Molecular Biosciences, Northwestern University, Evanston, IL, United States of America
- Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, IL, United States of America
| | - Gaotian Zhang
- Molecular Biosciences, Northwestern University, Evanston, IL, United States of America
| | - Karen S. Kim Guisbert
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL, United States of America
| | - Stefan Zdraljevic
- Molecular Biosciences, Northwestern University, Evanston, IL, United States of America
- Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, IL, United States of America
| | - Eric Guisbert
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL, United States of America
| | - Erik C. Andersen
- Molecular Biosciences, Northwestern University, Evanston, IL, United States of America
| |
Collapse
|
15
|
Collins JB, Andersen EC. The turkey ascarid, Ascaridia dissimilis, as a model genetic system. Int J Parasitol 2023; 53:405-409. [PMID: 36549442 PMCID: PMC10258144 DOI: 10.1016/j.ijpara.2022.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/02/2022] [Accepted: 10/25/2022] [Indexed: 12/23/2022]
Abstract
Parasitic nematodes cause significant effects on humans each year, with the most prevalent being Ascaris lumbricoides. Benzimidazoles (BZ) are the most widely used anthelmintic drug in humans, and although the biology of resistance to this drug class is understood in some species, resistance is poorly characterized in ascarids. Models such as Caenorhabditis elegans were essential in developing our current understanding of BZ resistance, but more closely related model nematodes are needed to understand resistance in ascarids. Here, we propose a new ascarid model species that infects turkeys, Ascaridia dissimilis, to develop a better understanding of BZ resistance.
Collapse
Affiliation(s)
- J B Collins
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Erik C Andersen
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
16
|
Haruta N, Sumiyoshi E, Honda Y, Terasawa M, Uchiyama C, Toya M, Kubota Y, Sugimoto A. A germline-specific role for unconventional components of the γ-tubulin complex in Caenorhabditis elegans. J Cell Sci 2023; 136:jcs260922. [PMID: 37313686 PMCID: PMC10657210 DOI: 10.1242/jcs.260922] [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: 12/29/2022] [Accepted: 06/07/2023] [Indexed: 06/15/2023] Open
Abstract
The γ-tubulin complex (γTuC) is a widely conserved microtubule nucleator, but some of its components, namely GCP4, GCP5 and GCP6 (also known as TUBGCP4, TUBGCP5 and TUBGCP6, respectively), have not been detected in Caenorhabditis elegans. Here, we identified two γTuC-associated proteins in C. elegans, GTAP-1 and GTAP-2, for which apparent orthologs were detected only in the genus Caenorhabditis. GTAP-1 and GTAP-2 were found to localize at centrosomes and the plasma membrane of the germline, and their centrosomal localization was interdependent. In early C. elegans embryos, whereas the conserved γTuC component MZT-1 (also known as MOZART1 and MZT1) was essential for the localization of centrosomal γ-tubulin, depletion of GTAP-1 and/or GTAP-2 caused up to 50% reduction of centrosomal γ-tubulin and precocious disassembly of spindle poles during mitotic telophase. In the adult germline, GTAP-1 and GTAP-2 contributed to efficient recruitment of the γTuC to the plasma membrane. Depletion of GTAP-1, but not of GTAP-2, severely disrupted both the microtubule array and the honeycomb-like structure of the adult germline. We propose that GTAP-1 and GTAP-2 are unconventional components of the γTuC that contribute to the organization of both centrosomal and non-centrosomal microtubules by targeting the γTuC to specific subcellular sites in a tissue-specific manner.
Collapse
Affiliation(s)
- Nami Haruta
- Laboratory of Developmental Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Eisuke Sumiyoshi
- Laboratory of Developmental Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Yu Honda
- Laboratory of Developmental Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Masahiro Terasawa
- Laboratory for Developmental Genomics, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Chihiro Uchiyama
- Laboratory of Developmental Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Mika Toya
- Laboratory for Developmental Genomics, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Yukihiko Kubota
- Laboratory of Developmental Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Asako Sugimoto
- Laboratory of Developmental Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
- Laboratory for Developmental Genomics, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| |
Collapse
|
17
|
Ríos-Valencia DG, Ambrosio J, Tirado-Mendoza R, Carrero JC, Laclette JP. What about the Cytoskeletal and Related Proteins of Tapeworms in the Host's Immune Response? An Integrative Overview. Pathogens 2023; 12:840. [PMID: 37375530 DOI: 10.3390/pathogens12060840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/09/2023] [Accepted: 06/11/2023] [Indexed: 06/29/2023] Open
Abstract
Recent advances have increased our understanding of the molecular machinery in the cytoskeleton of mammalian cells, in contrast to the case of tapeworm parasites, where cytoskeleton remains poorly characterized. The pertinence of a better knowledge of the tapeworm cytoskeleton is linked to the medical importance of these parasitic diseases in humans and animal stock. Moreover, its study could offer new possibilities for the development of more effective anti-parasitic drugs, as well as better strategies for their surveillance, prevention, and control. In the present review, we compile the results of recent experiments on the cytoskeleton of these parasites and analyze how these novel findings might trigger the development of new drugs or the redesign of those currently used in addition to supporting their use as biomarkers in cutting-edge diagnostic tests.
Collapse
Affiliation(s)
- Diana G Ríos-Valencia
- Department of Microbiology and Parasitology, School of Medicine, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México 04510, Mexico
| | - Javier Ambrosio
- Department of Microbiology and Parasitology, School of Medicine, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México 04510, Mexico
| | - Rocío Tirado-Mendoza
- Department of Microbiology and Parasitology, School of Medicine, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México 04510, Mexico
| | - Julio César Carrero
- Department of Immunology, Biomedical Research Institute, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México 04510, Mexico
| | - Juan Pedro Laclette
- Department of Immunology, Biomedical Research Institute, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México 04510, Mexico
| |
Collapse
|
18
|
Burns AR, Baker RJ, Kitner M, Knox J, Cooke B, Volpatti JR, Vaidya AS, Puumala E, Palmeira BM, Redman EM, Snider J, Marwah S, Chung SW, MacDonald MH, Tiefenbach J, Hu C, Xiao Q, Finney CAM, Krause HM, MacParland SA, Stagljar I, Gilleard JS, Cowen LE, Meyer SLF, Cutler SR, Dowling JJ, Lautens M, Zasada I, Roy PJ. Selective control of parasitic nematodes using bioactivated nematicides. Nature 2023:10.1038/s41586-023-06105-5. [PMID: 37225985 DOI: 10.1038/s41586-023-06105-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 04/20/2023] [Indexed: 05/26/2023]
Abstract
Parasitic nematodes are a major threat to global food security, particularly as the world amasses 10 billion people amid limited arable land1-4. Most traditional nematicides have been banned owing to poor nematode selectivity, leaving farmers with inadequate means of pest control4-12. Here we use the model nematode Caenorhabditis elegans to identify a family of selective imidazothiazole nematicides, called selectivins, that undergo cytochrome-p450-mediated bioactivation in nematodes. At low parts-per-million concentrations, selectivins perform comparably well with commercial nematicides to control root infection by Meloidogyne incognita, a highly destructive plant-parasitic nematode. Tests against numerous phylogenetically diverse non-target systems demonstrate that selectivins are more nematode-selective than most marketed nematicides. Selectivins are first-in-class bioactivated nematode controls that provide efficacy and nematode selectivity.
Collapse
Affiliation(s)
- Andrew R Burns
- The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.
| | - Rachel J Baker
- Davenport Research Laboratories, Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Megan Kitner
- USDA-ARS Horticultural Crops Research Laboratory, Corvallis, OR, USA
| | - Jessica Knox
- The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Brittany Cooke
- The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Jonathan R Volpatti
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Division of Neurology and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Aditya S Vaidya
- Institute for Integrative Genome Biology, University of California, Riverside, Riverside, CA, USA
- Department of Botany and Plant Sciences, University of California, Riverside, Riverside, CA, USA
| | - Emily Puumala
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Bruna M Palmeira
- Department of Comparative Biology and Experimental Medicine, Host-Parasite Interactions Program, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Elizabeth M Redman
- Department of Comparative Biology and Experimental Medicine, Host-Parasite Interactions Program, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jamie Snider
- The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Sagar Marwah
- Ajmera Transplant Centre, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology and Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Sai W Chung
- Ajmera Transplant Centre, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology and Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Margaret H MacDonald
- USDA-ARS Mycology and Nematology Genetic Diversity and Biology Laboratory, Beltsville Agricultural Research Center, Beltsville, MD, USA
| | - Jens Tiefenbach
- The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Chun Hu
- The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Qi Xiao
- Department of Biological Sciences, Host Parasite Interactions Program, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| | - Constance A M Finney
- Department of Biological Sciences, Host Parasite Interactions Program, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| | - Henry M Krause
- The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Sonya A MacParland
- Ajmera Transplant Centre, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology and Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Igor Stagljar
- The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Mediterranean Institute for Life Sciences, Split, Croatia
| | - John S Gilleard
- Department of Comparative Biology and Experimental Medicine, Host-Parasite Interactions Program, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Susan L F Meyer
- USDA-ARS Mycology and Nematology Genetic Diversity and Biology Laboratory, Beltsville Agricultural Research Center, Beltsville, MD, USA
| | - Sean R Cutler
- Institute for Integrative Genome Biology, University of California, Riverside, Riverside, CA, USA
- Department of Botany and Plant Sciences, University of California, Riverside, Riverside, CA, USA
| | - James J Dowling
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Division of Neurology and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Mark Lautens
- Davenport Research Laboratories, Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Inga Zasada
- USDA-ARS Horticultural Crops Research Laboratory, Corvallis, OR, USA
| | - Peter J Roy
- The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
19
|
Sinha A, Li Z, Poole CB, Morgan RD, Ettwiller L, Lima NF, Ferreira MU, Fombad FF, Wanji S, Carlow CKS. Genomes of the human filarial parasites Mansonella perstans and Mansonella ozzardi. FRONTIERS IN TROPICAL DISEASES 2023. [DOI: 10.3389/fitd.2023.1139343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023] Open
Abstract
The filarial parasites Mansonella ozzardi and Mansonella perstans, causative agents of mansonellosis, infect hundreds of millions of people worldwide, yet remain among the most understudied of the human filarial pathogens. M. ozzardi is highly prevalent in Latin American countries and Caribbean Islands, while M. perstans is predominantly found in sub-Saharan Africa as well as in a few areas in South America. In addition to the differences in their geographical distribution, the two parasites are transmitted by different insect vectors, as well as exhibit differences in their responses to commonly used anthelminthic drugs. The lack of genome information has hindered investigations into the biology and evolution of Mansonella parasites and understanding the molecular basis of the clinical differences between species. In the current study, high quality genomes of two independent clinical isolates of M. perstans from Cameroon and two M. ozzardi isolates one from Brazil and one from Venezuela are reported. The genomes are approximately 76 Mb in size, encode about 10,000 genes each, and are largely complete based on BUSCO scores of about 90%, similar to other completed filarial genomes. These sequences represent the first genomes from Mansonella parasites and enabled a comparative genomic analysis of the similarities and differences between Mansonella and other filarial parasites. Horizontal DNA transfers (HDT) from mitochondria (nuMTs) as well as transfers from genomes of endosymbiotic Wolbachia bacteria (nuWTs) to the host nuclear genome were identified and analyzed. Sequence comparisons and phylogenetic analysis of known targets of anti-filarial drugs diethylcarbamazine (DEC), ivermectin and mebendazole revealed that all known target genes were present in both species, except for the DEC target encoded by gon-2 gene, which is fragmented in genome assemblies from both M. ozzardi isolates. These new reference genome sequences will provide a valuable resource for further studies on biology, symbiosis, evolution and drug discovery.
Collapse
|
20
|
Shaver AO, Wit J, Dilks CM, Crombie TA, Li H, Aroian RV, Andersen EC. Variation in anthelmintic responses are driven by genetic differences among diverse C. elegans wild strains. PLoS Pathog 2023; 19:e1011285. [PMID: 37011090 PMCID: PMC10101645 DOI: 10.1371/journal.ppat.1011285] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 04/13/2023] [Accepted: 03/08/2023] [Indexed: 04/05/2023] Open
Abstract
Treatment of parasitic nematode infections in humans and livestock relies on a limited arsenal of anthelmintic drugs that have historically reduced parasite burdens. However, anthelmintic resistance (AR) is increasing, and little is known about the molecular and genetic causes of resistance for most drugs. The free-living roundworm Caenorhabditis elegans has proven to be a tractable model to understand AR, where studies have led to the identification of molecular targets of all major anthelmintic drug classes. Here, we used genetically diverse C. elegans strains to perform dose-response analyses across 26 anthelmintic drugs that represent the three major anthelmintic drug classes (benzimidazoles, macrocyclic lactones, and nicotinic acetylcholine receptor agonists) in addition to seven other anthelmintic classes. First, we found that C. elegans strains displayed similar anthelmintic responses within drug classes and significant variation across drug classes. Next, we compared the effective concentration estimates to induce a 10% maximal response (EC10) and slope estimates of each dose-response curve of each strain to the laboratory reference strain, which enabled the identification of anthelmintics with population-wide differences to understand how genetics contribute to AR. Because genetically diverse strains displayed differential susceptibilities within and across anthelmintics, we show that C. elegans is a useful model for screening potential nematicides before applications to helminths. Third, we quantified the levels of anthelmintic response variation caused by genetic differences among individuals (heritability) to each drug and observed a significant correlation between exposure closest to the EC10 and the exposure that exhibited the most heritable responses. These results suggest drugs to prioritize in genome-wide association studies, which will enable the identification of AR genes.
Collapse
Affiliation(s)
- Amanda O. Shaver
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Janneke Wit
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Clayton M. Dilks
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Timothy A. Crombie
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Hanchen Li
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Raffi V. Aroian
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Erik C. Andersen
- Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| |
Collapse
|
21
|
Multivariate chemogenomic screening prioritizes new macrofilaricidal leads. Commun Biol 2023; 6:44. [PMID: 36639423 PMCID: PMC9839782 DOI: 10.1038/s42003-023-04435-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 01/05/2023] [Indexed: 01/15/2023] Open
Abstract
Development of direct acting macrofilaricides for the treatment of human filariases is hampered by limitations in screening throughput imposed by the parasite life cycle. In vitro adult screens typically assess single phenotypes without prior enrichment for chemicals with antifilarial potential. We developed a multivariate screen that identified dozens of compounds with submicromolar macrofilaricidal activity, achieving a hit rate of >50% by leveraging abundantly accessible microfilariae. Adult assays were multiplexed to thoroughly characterize compound activity across relevant parasite fitness traits, including neuromuscular control, fecundity, metabolism, and viability. Seventeen compounds from a diverse chemogenomic library elicited strong effects on at least one adult trait, with differential potency against microfilariae and adults. Our screen identified five compounds with high potency against adults but low potency or slow-acting microfilaricidal effects, at least one of which acts through a novel mechanism. We show that the use of microfilariae in a primary screen outperforms model nematode developmental assays and virtual screening of protein structures inferred with deep learning. These data provide new leads for drug development, and the high-content and multiplex assays set a new foundation for antifilarial discovery.
Collapse
|
22
|
O'Hagan R, Avrutis A, Ramicevic E. Functions of the tubulin code in the C. elegans nervous system. Mol Cell Neurosci 2022; 123:103790. [PMID: 36368428 DOI: 10.1016/j.mcn.2022.103790] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/27/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022] Open
Abstract
Due to their elongated and polarized morphology, neurons rely on the microtubule (MT) cytoskeleton for their shape, as well as for efficient intracellular transport that maintains neuronal function, survival, and connectivity. Although all MTs are constructed from α- and β-tubulins that are highly conserved throughout eukaryotes, different MT networks within neurons exhibit different dynamics and functions. For example, molecular motors must be able to differentially recognize the axonal and dendritic MTs to deliver appropriate cargos to sensory endings and synaptic regions. The Tubulin Code hypothesis proposes that MTs can be specialized in form and function by chemical differences in their composition by inclusion of different α- and β-tubulins into the MT lattice, as well as differences in post-translational enzymatic modifications. The chemical differences encode information that allow MTs to regulate interactions with various microtubule-based molecular motors such as kinesins and dyneins as well as with structural microtubule-associated proteins (MAPs), which can, in turn, modify the function or stability of MTs. Here, we review studies involving C. elegans, a model organism with a relatively simple nervous system that is amenable to genetic analysis, that have contributed to our understanding of how the Tubulin Code can specialize neuronal MT networks to establish differences in neuronal morphology and function. Such studies have revealed molecules and mechanisms that are conserved in vertebrates and have the potential to inform our understanding of neurological diseases involving defects in the cytoskeleton and intracellular transport.
Collapse
Affiliation(s)
- Robert O'Hagan
- formerly at Biology Dept., Montclair State University, Montclair, NJ 07043, United States of America.
| | - Alexandra Avrutis
- formerly at Biology Dept., Montclair State University, Montclair, NJ 07043, United States of America
| | - Ema Ramicevic
- formerly at Biology Dept., Montclair State University, Montclair, NJ 07043, United States of America
| |
Collapse
|
23
|
Gibson SB, Ness-Cohn E, Andersen EC. Benzimidazoles cause lethality by inhibiting the function of Caenorhabditis elegans neuronal beta-tubulin. Int J Parasitol Drugs Drug Resist 2022; 20:89-96. [PMID: 36332489 PMCID: PMC9771835 DOI: 10.1016/j.ijpddr.2022.10.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 11/08/2022]
Abstract
Parasitic nematode infections cause an enormous global burden to both humans and livestock. Resistance to the limited arsenal of anthelmintic drugs used to combat these infections is widespread, including benzimidazole (BZ) compounds. Previous studies using the free-living nematode Caenorhabditis elegans to model parasitic nematode resistance have shown that loss-of-function mutations in the beta-tubulin gene ben-1 confer resistance to BZ drugs. However, the mechanism of resistance and the tissue-specific susceptibility are not well known in any nematode species. To identify in which tissue(s) ben-1 function underlies BZ susceptibility, transgenic strains that express ben-1 in different tissues, including hypodermis, muscles, neurons, intestine, and ubiquitous expression were generated. High-throughput fitness assays were performed to measure and compare the quantitative responses to BZ compounds among different transgenic lines. Significant BZ susceptibility was observed in animals expressing ben-1 in neurons, comparable to expression using the ben-1 promoter. This result suggests that ben-1 function in neurons underlies susceptibility to BZ. Subsetting neuronal expression of ben-1 based on the neurotransmitter system further restricted ben-1 function in cholinergic neurons to cause BZ susceptibility. These results better inform our current understanding of the cellular mode of action of BZs and also suggest additional treatments that might potentiate the effects of BZs in neurons.
Collapse
Affiliation(s)
- Sophia B Gibson
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA
| | - Elan Ness-Cohn
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA; Driskill Graduate Program in Life Sciences, Northwestern University, Chicago, IL, 60611, USA
| | - Erik C Andersen
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA.
| |
Collapse
|
24
|
Doyle SR, Laing R, Bartley D, Morrison A, Holroyd N, Maitland K, Antonopoulos A, Chaudhry U, Flis I, Howell S, McIntyre J, Gilleard JS, Tait A, Mable B, Kaplan R, Sargison N, Britton C, Berriman M, Devaney E, Cotton JA. Genomic landscape of drug response reveals mediators of anthelmintic resistance. Cell Rep 2022; 41:111522. [PMID: 36261007 PMCID: PMC9597552 DOI: 10.1016/j.celrep.2022.111522] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 07/11/2022] [Accepted: 09/26/2022] [Indexed: 11/18/2022] Open
Abstract
Like other pathogens, parasitic helminths can rapidly evolve resistance to drug treatment. Understanding the genetic basis of anthelmintic drug resistance in parasitic nematodes is key to tracking its spread and improving the efficacy and sustainability of parasite control. Here, we use an in vivo genetic cross between drug-susceptible and multi-drug-resistant strains of Haemonchus contortus in a natural host-parasite system to simultaneously map resistance loci for the three major classes of anthelmintics. This approach identifies new alleles for resistance to benzimidazoles and levamisole and implicates the transcription factor cky-1 in ivermectin resistance. This gene is within a locus under selection in ivermectin-resistant populations worldwide; expression analyses and functional validation using knockdown experiments support that cky-1 is associated with ivermectin survival. Our work demonstrates the feasibility of high-resolution forward genetics in a parasitic nematode and identifies variants for the development of molecular diagnostics to combat drug resistance in the field.
Collapse
Affiliation(s)
- Stephen R Doyle
- Wellcome Sanger Institute, Hinxton, Cambridgeshire CB10 1SA, UK.
| | - Roz Laing
- Institute of Biodiversity Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK.
| | - David Bartley
- Moredun Research Institute, Penicuik, Midlothian EH26 0PZ, UK
| | - Alison Morrison
- Moredun Research Institute, Penicuik, Midlothian EH26 0PZ, UK
| | - Nancy Holroyd
- Wellcome Sanger Institute, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Kirsty Maitland
- Institute of Biodiversity Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Alistair Antonopoulos
- Institute of Biodiversity Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Umer Chaudhry
- Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh EH25 9RG, UK
| | - Ilona Flis
- Institute of Biodiversity Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Sue Howell
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Jennifer McIntyre
- Institute of Biodiversity Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - John S Gilleard
- Department of Comparative Biology and Experimental Medicine, Host-Parasite Interactions Program, Faculty of Veterinary Medicine, University of Calgary, Calgary T2N 1N4, Canada
| | - Andy Tait
- Institute of Biodiversity Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Barbara Mable
- Institute of Biodiversity Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Ray Kaplan
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Neil Sargison
- Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh EH25 9RG, UK
| | - Collette Britton
- Institute of Biodiversity Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | | | - Eileen Devaney
- Institute of Biodiversity Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - James A Cotton
- Wellcome Sanger Institute, Hinxton, Cambridgeshire CB10 1SA, UK
| |
Collapse
|
25
|
Li N, Hua B, Chen Q, Teng F, Ruan M, Zhu M, Zhang L, Huo Y, Liu H, Zhuang M, Shen H, Zhu H. A sphingolipid-mTORC1 nutrient-sensing pathway regulates animal development by an intestinal peroxisome relocation-based gut-brain crosstalk. Cell Rep 2022; 40:111140. [PMID: 35905721 DOI: 10.1016/j.celrep.2022.111140] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 05/23/2022] [Accepted: 07/01/2022] [Indexed: 11/30/2022] Open
Abstract
The mTOR-dependent nutrient-sensing and response machinery is the central hub for animals to regulate their cellular and developmental programs. However, equivalently pivotal nutrient and metabolite signals upstream of mTOR and developmental-regulatory signals downstream of mTOR are not clear, especially at the organism level. We previously showed glucosylceramide (GlcCer) acts as a critical nutrient and metabolite signal for overall amino acid levels to promote development by activating the intestinal mTORC1 signaling pathway. Here, through a large-scale genetic screen, we find that the intestinal peroxisome is critical for antagonizing the GlcCer-mTORC1-mediated nutrient signal. Mechanistically, GlcCer deficiency, inactive mTORC1, or prolonged starvation relocates intestinal peroxisomes closer to the apical region in a kinesin- and microtubule-dependent manner. Those apical accumulated peroxisomes further release peroxisomal-β-oxidation-derived glycolipid hormones that target chemosensory neurons and downstream nuclear hormone receptor DAF-12 to arrest the animal development. Our data illustrate a sophisticated gut-brain axis that predominantly orchestrates nutrient-sensing-dependent development in animals.
Collapse
Affiliation(s)
- Na Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100000, China
| | - Beilei Hua
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qing Chen
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Fukang Teng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Meiyu Ruan
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Mengnan Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100000, China
| | - Li Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yinbo Huo
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100000, China
| | - Hongqin Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Min Zhuang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Huali Shen
- Institutes of Biomedical Sciences, Department of Systems Biology for Medicine and School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Huanhu Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
26
|
Pallotto LM, Dilks CM, Park YJ, Smit RB, Lu B, Gopalakrishnan C, Gilleard JS, Andersen EC, Mains PE. Interactions of C. elegans β-tubulins with the microtubule inhibitor and anthelmintic drug albendazole. Genetics 2022; 221:6613138. [PMID: 35731216 DOI: 10.1093/genetics/iyac093] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/08/2022] [Indexed: 11/14/2022] Open
Abstract
Parasitic nematodes are major human and agricultural pests, and benzimidazoles are amongst the most important broad spectrum anthelmintic drug class used for their control. Benzimidazole resistance is now widespread in many species of parasitic nematodes in livestock globally and an emerging concern for the sustainable control of human soil transmitted helminths. β-tubulin is the major benzimidazole target, although other genes may influence resistance. Among the six C. elegans β-tubulin genes, loss of ben-1 causes resistance without other apparent defects. Here, we explored the genetics of C. elegans β-tubulin genes in relation to the response to the benzimidazole derivative albendazole. The most highly expressed β-tubulin isotypes, encoded by tbb-1 and tbb-2, were known to be redundant with each other for viability, and their products are predicted not to bind benzimidazoles. We found that tbb-2 mutants, and to a lesser extent tbb-1 mutants, were hypersensitive to albendazole. The double mutant tbb-2 ben-1 is uncoordinated and short, resembling the wild type exposed to albendazole, but the tbb-1 ben-1 double mutant did not show the same phenotypes. These results suggest that tbb-2 is a modifier of ABZ sensitivity. To better understand how BEN-1 mutates to cause benzimidazole resistance, we isolated mutants resistant to albendazole and found that 15 of 16 mutations occurred in the ben-1 coding region. Mutations ranged from likely nulls to hypomorphs, and several corresponded to residues that cause resistance in other organisms. Null alleles of ben-1 are albendazole-resistant and BEN-1 shows high sequence identity with tubulins from other organisms, suggesting that many amino acid changes could cause resistance. However, our results suggest that missense mutations conferring resistance are not evenly distributed across all possible conserved sites. Independent of their roles in benzimidazole resistance, tbb-1 and tbb-2 may have specialized functions as null mutants of tbb-1 or tbb-2 were cold or heat sensitive, respectively.
Collapse
Affiliation(s)
- Linda M Pallotto
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Clayton M Dilks
- Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA.,Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, IL, 60208, USA
| | - Ye-Jean Park
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Ryan B Smit
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Brian Lu
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | | | - John S Gilleard
- Department of Comparative Biology and Experimental Medicine, Host-Parasite Interactions (HPI) Program, Faculty of Veterinary Medicine, University of Calgary, Alberta, T2N 4N1 Canada
| | - Erik C Andersen
- Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA
| | - Paul E Mains
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| |
Collapse
|
27
|
The impact of species-wide gene expression variation on Caenorhabditis elegans complex traits. Nat Commun 2022; 13:3462. [PMID: 35710766 PMCID: PMC9203580 DOI: 10.1038/s41467-022-31208-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 06/08/2022] [Indexed: 12/15/2022] Open
Abstract
Phenotypic variation in organism-level traits has been studied in Caenorhabditis elegans wild strains, but the impacts of differences in gene expression and the underlying regulatory mechanisms are largely unknown. Here, we use natural variation in gene expression to connect genetic variants to differences in organismal-level traits, including drug and toxicant responses. We perform transcriptomic analyses on 207 genetically distinct C. elegans wild strains to study natural regulatory variation of gene expression. Using this massive dataset, we perform genome-wide association mappings to investigate the genetic basis underlying gene expression variation and reveal complex genetic architectures. We find a large collection of hotspots enriched for expression quantitative trait loci across the genome. We further use mediation analysis to understand how gene expression variation could underlie organism-level phenotypic variation for a variety of complex traits. These results reveal the natural diversity in gene expression and possible regulatory mechanisms in this keystone model organism, highlighting the promise of using gene expression variation to understand how phenotypic diversity is generated.
Collapse
|
28
|
Abstract
The microtubule cytoskeleton is assembled from the α- and β-tubulin subunits of the canonical tubulin heterodimer, which polymerizes into microtubules, and a small number of other family members, such as γ-tubulin, with specialized functions. Overall, microtubule function involves the collective action of multiple α- and β-tubulin isotypes. However, despite 40 years of awareness that most eukaryotes harbor multiple tubulin isotypes, their role in the microtubule cytoskeleton has remained relatively unclear. Various model organisms offer specific advantages for gaining insight into the role of tubulin isotypes. Whereas simple unicellular organisms such as yeast provide experimental tractability that can facilitate deeper access to mechanistic details, more complex organisms, such as the fruit fly, nematode and mouse, can be used to discern potential specialized functions of tissue- and structure-specific isotypes. Here, we review the role of α- and β-tubulin isotypes in microtubule function and in associated tubulinopathies with an emphasis on the advances gained using model organisms. Overall, we argue that studying tubulin isotypes in a range of organisms can reveal the fundamental mechanisms by which they mediate microtubule function. It will also provide valuable perspectives on how these mechanisms underlie the functional and biological diversity of the cytoskeleton.
Collapse
Affiliation(s)
- Emmanuel T Nsamba
- Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011, USA
| | - Mohan L Gupta
- Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
29
|
Absence of Polymorphisms in Codons 167, 198 and 200 of All Seven β-tubulin Isotypes of Benzimidazole Susceptible and Resistant Parascaris spp. Specimens from Australia. Pathogens 2022; 11:pathogens11050490. [PMID: 35631011 PMCID: PMC9143322 DOI: 10.3390/pathogens11050490] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 11/24/2022] Open
Abstract
Benzimidazoles resistance is widespread in strongyle parasitic nematodes and associated with polym orphisms in the codons 167, 198 and 200 of isotype 1 β-tubulin (tbb-1). In ascarids, benzimidazole (BZ) resistance has rarely been reported and in none of these cases were any of these polymorphisms detected. Here, available genome and transcriptome data from WormBase ParaSite were used to compare the complete β-tubulin reservoirs of Parascaris univalens, Ascaris suum and Ascaris lumbricoides. Adult Parascaris spp. specimens collected in Australia from horses after BZ treatment (susceptible, n = 13) or surviving BZ treatment and collected after ivermectin treatment (resistant, n = 10) were genotyped regarding codons 167, 198 and 200 using Sanger sequencing. Phylogenetic analyses clearly showed that there are no one-to-one ascarid orthologs of strongyle tbb-1 genes. In the reference genomes, as well as phenotypically susceptible and resistant Parascaris spp. from Australia, six out of seven β-tubulin genes showed a BZ-susceptible genotype (F167, E198, F200). The only exception were the testis-specific β-tubulin D genes from all three ascarid species that encode tyrosine at codon 200. This was observed independently of the BZ-susceptibility phenotype of Parascaris spp. These data suggest that different mechanisms lead to BZ resistance in ascarid and strongyle nematodes.
Collapse
|
30
|
Munguía B, Saldaña J, Nieves M, Melian ME, Ferrer M, Teixeira R, Porcal W, Manta E, Domínguez L. Sensitivity of Haemonchus contortus to anthelmintics using different in vitro screening assays: a comparative study. Parasit Vectors 2022; 15:129. [PMID: 35413885 PMCID: PMC9006605 DOI: 10.1186/s13071-022-05253-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 03/24/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Helminthiasis and resistance to commercial anthelmintic compounds are major causes of economic losses for livestock producers, resulting in an urgent need for new drugs and reliable in vitro screening tests capable of detecting potentially active products. Considering this, a series of novel benzimidazole derivatives (5-methylbenzimidazole 1,2-disubstituted, 5-carboxybenzimidazole, 5-methylbenzimidazole 2-one) was screened on exsheathed L3 (xL3) and on the adult stage of Haemonchus contortus (Kirby anthelmintic-susceptible McMaster isolate). METHODS This work presents the set-up of an automated motility assay on the xL3 stage of H. contortus using an infrared tracking device (WMicrotracker One) together with a larval development test (xL3 to L4) and a motility assay on the adult stage of H. contortus. A comparative study of the sensitivity of these in vitro assays using commercial anthelmintics with different mechanisms of action was carried out, also evaluating anthelmintic activity of a series of novel benzimidazole derivatives. RESULTS The automated xL3 assay had the great advantage of being able to analyze many compounds simultaneously, but it showed the limitation of having lower sensitivity, requiring higher concentrations of the commercial anthelmintics tested compared to those needed for the adult motility or development assays. Although none of the novel 1,2,5-tri-substituted benzimidazole derivatives could significantly decrease the motility of xL3s, one of them (1e) significantly affected the development of xL3s to L4, and five new compounds (1b, 1d, 1e, 2a and 2c) reduced the motility of H. contortus adult stage. CONCLUSIONS The analysis of the results strongly suggests that the in vitro xL3 to L4 development test, particularly for the L4 stage, could be closer to the pharmacological sensitivity of the adult stage of H. contortus (target of interest) for commercial anthelmintic selected, with different mechanisms of action, and for the series of benzimidazole derivatives assayed. Therefore, an automated motility assay on L4 using the infrared tracking device is being set up. Further studies will be conducted to evaluate the in vivo anthelmintic activity of the most active novel benzimidazole derivatives.
Collapse
Affiliation(s)
- Beatriz Munguía
- Área de Farmacología, CIENFAR, Facultad de Química, Universidad de la República (Udelar), Montevideo, Uruguay
| | - Jenny Saldaña
- Área de Farmacología, CIENFAR, Facultad de Química, Universidad de la República (Udelar), Montevideo, Uruguay
| | - Magdalena Nieves
- Área de Farmacología, CIENFAR, Facultad de Química, Universidad de la República (Udelar), Montevideo, Uruguay
| | - María Elisa Melian
- Área de Farmacología, CIENFAR, Facultad de Química, Universidad de la República (Udelar), Montevideo, Uruguay
| | - Manuela Ferrer
- Área de Farmacología, CIENFAR, Facultad de Química, Universidad de la República (Udelar), Montevideo, Uruguay
| | - Ramiro Teixeira
- Área de Farmacología, CIENFAR, Facultad de Química, Universidad de la República (Udelar), Montevideo, Uruguay
| | - Williams Porcal
- Departamento de Química Orgánica, Facultad de Química, Universidad de la República (Udelar), Montevideo, Uruguay
| | - Eduardo Manta
- Departamento de Química Orgánica, Facultad de Química, Laboratorio de Química Farmacéutica, Universidad de la República (Udelar), Montevideo, Uruguay
| | - Laura Domínguez
- Área de Farmacología, CIENFAR, Facultad de Química, Universidad de la República (Udelar), Montevideo, Uruguay.
| |
Collapse
|
31
|
Lu YM, Zheng C. The Expression and Function of Tubulin Isotypes in Caenorhabditis elegans. Front Cell Dev Biol 2022; 10:860065. [PMID: 35399537 PMCID: PMC8987236 DOI: 10.3389/fcell.2022.860065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
Microtubules, made from the polymerization of the highly conserved α/β-tubulin heterodimers, serve as important components of the cytoskeleton in all eukaryotic cells. The existence of multiple tubulin isotypes in metazoan genomes and a dazzling variety of tubulin posttranslational modifications (PTMs) prompted the “tubulin code” hypothesis, which proposed that microtubule structure and functions are determined by the tubulin composition and PTMs. Evidence for the tubulin code has emerged from studies in several organisms with the characterization of specific tubulins for their expression and functions. The studies of tubulin PTMs are accelerated by the discovery of the enzymes that add or remove the PTMs. In tubulin research, the use of simple organisms, such as Caenorhabditis elegans, has been instrumental for understanding the expression and functional specialization of tubulin isotypes and the effects of their PTMs. In this review, we summarize the current understanding of the expression patterns and cellular functions of the nine α-tubulin and six β-tubulin isotypes. Expression studies are greatly facilitated by the CRISPR/Cas9-mediated endogenous GFP knock-in reporters and the organism-wide single cell transcriptomic studies. Meanwhile, functional studies benefit from the ease of genetic manipulation and precise gene replacement in C. elegans. These studies identified both ubiquitously expressed tubulin isotypes and tissue-specific isotypes. The isotypes showed functional redundancy, as well as functional specificity, which is likely caused by the subtle differences in their amino acid sequences. Many of these differences concentrate at the C-terminal tails that are subjected to several PTMs. Indeed, tubulin PTM, such as polyglutamylation, is shown to modulate microtubule organization and properties in both ciliated and non-ciliated neurons. Overall, studies from C. elegans support the distinct expression and function patterns of tubulin isotypes and the importance of their PTMs and offer the promise of cracking the tubulin code at the whole-genome and the whole-organism level.
Collapse
|
32
|
Lautens MJ, Tan JH, Serrat X, Del Borrello S, Schertzberg MR, Fraser AG. Identification of enzymes that have helminth-specific active sites and are required for Rhodoquinone-dependent metabolism as targets for new anthelmintics. PLoS Negl Trop Dis 2021; 15:e0009991. [PMID: 34843467 PMCID: PMC8659336 DOI: 10.1371/journal.pntd.0009991] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 12/09/2021] [Accepted: 11/11/2021] [Indexed: 11/18/2022] Open
Abstract
Soil transmitted helminths (STHs) are major human pathogens that infect over a billion people. Resistance to current anthelmintics is rising and new drugs are needed. Here we combine multiple approaches to find druggable targets in the anaerobic metabolic pathways STHs need to survive in their mammalian host. These require rhodoquinone (RQ), an electron carrier used by STHs and not their hosts. We identified 25 genes predicted to act in RQ-dependent metabolism including sensing hypoxia and RQ synthesis and found 9 are required. Since all 9 have mammalian orthologues, we used comparative genomics and structural modeling to identify those with active sites that differ between host and parasite. Together, we found 4 genes that are required for RQ-dependent metabolism and have different active sites. Finding these high confidence targets can open up in silico screens to identify species selective inhibitors of these enzymes as new anthelmintics.
Collapse
Affiliation(s)
- Margot J. Lautens
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - June H. Tan
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Xènia Serrat
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Andrew G. Fraser
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
33
|
Newly identified parasitic nematode beta-tubulin alleles confer resistance to benzimidazoles. Int J Parasitol Drugs Drug Resist 2021; 17:168-175. [PMID: 34637983 PMCID: PMC8503852 DOI: 10.1016/j.ijpddr.2021.09.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/06/2021] [Accepted: 09/06/2021] [Indexed: 01/08/2023]
Abstract
Infections by parasitic nematodes cause large health and economic burdens worldwide. We use anthelmintic drugs to reduce these infections. However, resistance to anthelmintic drugs is extremely common and increasing worldwide. It is essential to understand the mechanisms of resistance to slow its spread. Recently, four new parasitic nematode beta-tubulin alleles have been identified in benzimidazole (BZ) resistant parasite populations: E198I, E198K, E198T, and E198stop. These alleles have not been tested for the ability to confer resistance or for any effects that they might have on organismal fitness. We introduced these four new alleles into the sensitive C. elegans laboratory-adapted N2 strain and exposed these genome-edited strains to both albendazole and fenbendazole. We found that all four alleles conferred resistance to both BZ drugs. Additionally, we tested for fitness consequences in both control and albendazole conditions over seven generations in competitive fitness assays. We found that none of the edited alleles had deleterious effects on fitness in control conditions and that all four alleles conferred strong and equivalent fitness benefits in BZ drug conditions. Because it is unknown if previously validated alleles confer a dominant or recessive BZ resistance phenotype, we tested the phenotypes caused by five of these alleles and found that none of them conferred a dominant BZ resistance phenotype. Accurate measurements of resistance, fitness effects, and dominance caused by the resistance alleles allow for the generation of better models of population dynamics and facilitate control practices that maximize the efficacy of this critical anthelmintic drug class. Four newly identified parasitic nematode beta-tubulin alleles confer benzimidazole resistance. The four newly identified alleles do not confer deleterious fitness consequences. Five beta-tubulin alleles confer recessive benzimidazole resistance.
Collapse
|
34
|
Characterization of the β-tubulin gene family in Ascaris lumbricoides and Ascaris suum and its implication for the molecular detection of benzimidazole resistance. PLoS Negl Trop Dis 2021; 15:e0009777. [PMID: 34570778 PMCID: PMC8496844 DOI: 10.1371/journal.pntd.0009777] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 10/07/2021] [Accepted: 08/29/2021] [Indexed: 01/04/2023] Open
Abstract
Background The treatment coverage of control programs providing benzimidazole (BZ) drugs to eliminate the morbidity caused by soil-transmitted helminths (STHs) is unprecedently high. This high drug pressure may result in the development of BZ resistance in STHs and so there is an urgent need for surveillance systems detecting molecular markers associated with BZ resistance. A critical prerequisite to develop such systems is an understanding of the gene family encoding β-tubulin proteins, the principal targets of BZ drugs. Methodology and principal findings First, the β-tubulin gene families of Ascaris lumbricoides and Ascaris suum were characterized through the analysis of published genomes. Second, RNA-seq and RT-PCR analyses on cDNA were applied to determine the transcription profiles of the different gene family members. The results revealed that Ascaris species have at least seven different β-tubulin genes of which two are highly expressed during the entire lifecycle. Third, deep amplicon sequencing was performed on these two genes in more than 200 adult A. lumbricoides (Ethiopia and Tanzania) and A. suum (Belgium) worms, to investigate the intra- and inter-species genetic diversity and the presence of single nucleotide polymorphisms (SNPs) that are associated with BZ resistance in other helminth species; F167Y (TTC>TAC or TTT>TAT), E198A (GAA>GCA or GAG>GCG), E198L (GAA>TTA) and F200Y (TTC>TAC or TTT>TAT). These particular SNPs were absent in the two investigated genes in all three Ascaris populations. Significance This study demonstrated the presence of at least seven β-tubulin genes in Ascaris worms. A new nomenclature was proposed and prioritization of genes for future BZ resistance research was discussed. This is the first comprehensive description of the β-tubulin gene family in Ascaris and provides a framework to investigate the prevalence and potential role of β-tubulin sequence polymorphisms in BZ resistance in a more systematic manner than previously possible. Benzimidazole (BZ) drugs remain the standard of treatment in large-scale deworming programs that aim to control the morbidity caused by intestinal worms. As these deworming programs are expanding world-wide, there is an increasing risk of worms becoming resistant to BZ drugs, highlighting the necessity for tools to detect gene mutations associated with drug resistance. However, the development of such tools is impeded by a lack of insights into the genes that are coding for β-tubulin proteins, which are the principal targets of BZ drugs. The aim of this study was to comprehensively characterize these genes in the worm species Ascaris lumbricoides and Ascaris suum. The findings highlight that these species have at least seven β-tubulin genes. Only two genes are highly expressed throughout the different life stages of the worm, and hence are more likely to be involved in the development of BZ resistance. No mutations that have previously been associated with BZ resistance in other intestinal worms were found. This study provides a baseline towards more efficient and accurate monitoring of drug resistance in large-scale deworming programs.
Collapse
|
35
|
Martin F, Halvarsson P, Delhomme N, Höglund J, Tydén E. Exploring the β-tubulin gene family in a benzimidazole-resistant Parascaris univalens population. Int J Parasitol Drugs Drug Resist 2021; 17:84-91. [PMID: 34467878 PMCID: PMC8406161 DOI: 10.1016/j.ijpddr.2021.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/15/2021] [Accepted: 08/19/2021] [Indexed: 12/15/2022]
Abstract
Benzimidazole (BZ) drugs are frequently used to treat infections with the equine ascarid Parascaris univalens due to increasing resistance to macrocyclic lactones and pyrantel. Benzimidazole resistance is rare in ascarids in contrast to strongyle parasites where this resistance is widespread. In strongyles, single nucleotide polymorphisms (SNPs) at codons 167, 198 and 200 in a β-tubulin gene have been correlated to BZ resistance, but little is known about the β-tubulin genes and their possible involvement in BZ resistance in P. univalens and other ascarids. Previously two β-tubulin genes have been identified in P. univalens. In this study, we present five additional β-tubulin genes as well as the phylogenetic relationship of all seven genes to β-tubulins of other clade III and V nematodes. In addition, the efficacy of fenbendazole for treatment of P. univalens on a Swedish stud farm was studied in 2019 and 2020 using faecal egg count reduction test. Reductions varied from 73% to 88%, indicating the presence of a resistant P. univalens population on the farm. The emergence of BZ resistance emphasizes the need for development of molecular markers for rapid and more sensitive detection of resistant populations. We therefore investigated whether possible SNPs at positions 167, 198 or 200 in any of the β-tubulin genes could be used to distinguish between resistant and susceptible P. univalens populations. Amplicon sequencing covering the mutation sites 167, 198 and 200 in all seven β-tubulin genes revealed an absence of SNPs in both resistant and susceptible populations, suggesting that the mechanism behind BZ resistance in ascarids is different from that in strongyle nematodes and the search for a molecular marker for BZ resistance in P. univalens needs to continue. First case of fenbendazole resistance in Parascaris univalens in Europe. The P. univalens β-tubulin family contains seven genes. P. univalens β-tubulin genes cluster with β-tubulins from other clade V nematodes. No resistance associated SNPs were identified in P. univalens β-tubulin genes.
Collapse
Affiliation(s)
- Frida Martin
- Swedish University of Agricultural Sciences, Department of Biomedical Sciences and Veterinary Public Health, Section for Parasitology, Box 7036, 750 07, Uppsala, Sweden.
| | - Peter Halvarsson
- Swedish University of Agricultural Sciences, Department of Biomedical Sciences and Veterinary Public Health, Section for Parasitology, Box 7036, 750 07, Uppsala, Sweden
| | - Nicolas Delhomme
- Umeå Plant Science Centre (UPSC), Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, 901 83, Umeå, Sweden
| | - Johan Höglund
- Swedish University of Agricultural Sciences, Department of Biomedical Sciences and Veterinary Public Health, Section for Parasitology, Box 7036, 750 07, Uppsala, Sweden
| | - Eva Tydén
- Swedish University of Agricultural Sciences, Department of Biomedical Sciences and Veterinary Public Health, Section for Parasitology, Box 7036, 750 07, Uppsala, Sweden
| |
Collapse
|
36
|
Garge RK, Cha HJ, Lee C, Gollihar JD, Kachroo AH, Wallingford JB, Marcotte EM. Discovery of new vascular disrupting agents based on evolutionarily conserved drug action, pesticide resistance mutations, and humanized yeast. Genetics 2021; 219:iyab101. [PMID: 34849907 PMCID: PMC8633126 DOI: 10.1093/genetics/iyab101] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 06/15/2021] [Indexed: 12/20/2022] Open
Abstract
Thiabendazole (TBZ) is an FDA-approved benzimidazole widely used for its antifungal and antihelminthic properties. We showed previously that TBZ is also a potent vascular disrupting agent and inhibits angiogenesis at the tissue level by dissociating vascular endothelial cells in newly formed blood vessels. Here, we uncover TBZ's molecular target and mechanism of action. Using human cell culture, molecular modeling, and humanized yeast, we find that TBZ selectively targets only 1 of 9 human β-tubulin isotypes (TUBB8) to specifically disrupt endothelial cell microtubules. By leveraging epidemiological pesticide resistance data and mining chemical features of commercially used benzimidazoles, we discover that a broader class of benzimidazole compounds, in extensive use for 50 years, also potently disrupt immature blood vessels and inhibit angiogenesis. Thus, besides identifying the molecular mechanism of benzimidazole-mediated vascular disruption, this study presents evidence relevant to the widespread use of these compounds while offering potential new clinical applications.
Collapse
Affiliation(s)
- Riddhiman K Garge
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Hye Ji Cha
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Chanjae Lee
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jimmy D Gollihar
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
- US Army Research Laboratory—South, Austin, TX 78758, USA
| | - Aashiq H Kachroo
- The Department of Biology, Centre for Applied Synthetic Biology, Concordia University, Montreal, QC H4B 1R6, Canada
| | - John B Wallingford
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Edward M Marcotte
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
37
|
Gibson SB, Harper CS, Lackner LL, Andersen EC. The Caenorhabditis elegans and Haemonchus contortus beta-tubulin genes cannot substitute for loss of the Saccharomyces cerevisiae beta-tubulin gene. MICROPUBLICATION BIOLOGY 2021; 2021. [PMID: 34222836 PMCID: PMC8246273 DOI: 10.17912/micropub.biology.000411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
To better understand the mechanism of resistance caused by putative interactions between beta-tubulin and benzimidazole compounds, we sought to purify nematode-specific beta-tubulins using heterologous expression after replacement of the single Saccharomyces cerevisiae beta-tubulin gene. However, we found that haploid yeast cells containing nematode-specific beta-tubulin genes were not viable, suggesting that nematode beta-tubulin cannot substitute for the loss of the yeast beta-tubulin gene.
Collapse
Affiliation(s)
- Sophia B Gibson
- Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA
| | - Clare S Harper
- Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA.,Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, IL, 60208, USA
| | - Laura L Lackner
- Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA
| | - Erik C Andersen
- Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA
| |
Collapse
|
38
|
Nishida K, Tsuchiya K, Obinata H, Onodera S, Honda Y, Lai YC, Haruta N, Sugimoto A. Expression Patterns and Levels of All Tubulin Isotypes Analyzed in GFP Knock-In C. elegans Strains. Cell Struct Funct 2021; 46:51-64. [PMID: 33967119 PMCID: PMC10511039 DOI: 10.1247/csf.21022] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 04/28/2021] [Indexed: 11/11/2022] Open
Abstract
Most organisms have multiple α- and β-tubulin isotypes that likely contribute to the diversity of microtubule (MT) functions. To understand the functional differences of tubulin isotypes in Caenorhabditis elegans, which has nine α-tubulin isotypes and six β-tubulin isotypes, we systematically constructed null mutants and GFP-fusion strains for all tubulin isotypes with the CRISPR/Cas9 system and analyzed their expression patterns and levels in adult hermaphrodites. Four isotypes-α-tubulins TBA-1 and TBA-2 and β-tubulins TBB-1 and TBB-2-were expressed in virtually all tissues, with a distinct tissue-specific spectrum. Other isotypes were expressed in specific tissues or cell types at significantly lower levels than the broadly expressed isotypes. Four isotypes (TBA-5, TBA-6, TBA-9, and TBB-4) were expressed in different subsets of ciliated sensory neurons, and TBB-4 was inefficiently incorporated into mitotic spindle MTs. Taken together, we propose that MTs in C. elegans are mainly composed of four broadly expressed tubulin isotypes and that incorporation of a small amount of tissue-specific isotypes may contribute to tissue-specific MT properties. These newly constructed strains will be useful for further elucidating the distinct roles of tubulin isotypes.Key words: tubulin isotypes, microtubules, C. elegans.
Collapse
Affiliation(s)
- Kei Nishida
- Laboratory of Developmental Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Kenta Tsuchiya
- Laboratory of Developmental Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Hiroyuki Obinata
- Laboratory of Developmental Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Shizuka Onodera
- Laboratory of Developmental Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Yu Honda
- Laboratory of Developmental Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Yen-Cheng Lai
- Laboratory of Developmental Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Nami Haruta
- Laboratory of Developmental Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Asako Sugimoto
- Laboratory of Developmental Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| |
Collapse
|
39
|
Knox J, Joly N, Linossi EM, Carmona-Negrón JA, Jura N, Pintard L, Zuercher W, Roy PJ. A survey of the kinome pharmacopeia reveals multiple scaffolds and targets for the development of novel anthelmintics. Sci Rep 2021; 11:9161. [PMID: 33911106 PMCID: PMC8080662 DOI: 10.1038/s41598-021-88150-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 04/08/2021] [Indexed: 11/10/2022] Open
Abstract
Over one billion people are currently infected with a parasitic nematode. Symptoms can include anemia, malnutrition, developmental delay, and in severe cases, death. Resistance is emerging to the anthelmintics currently used to treat nematode infection, prompting the need to develop new anthelmintics. Towards this end, we identified a set of kinases that may be targeted in a nematode-selective manner. We first screened 2040 inhibitors of vertebrate kinases for those that impair the model nematode Caenorhabditis elegans. By determining whether the terminal phenotype induced by each kinase inhibitor matched that of the predicted target mutant in C. elegans, we identified 17 druggable nematode kinase targets. Of these, we found that nematode EGFR, MEK1, and PLK1 kinases have diverged from vertebrates within their drug-binding pocket. For each of these targets, we identified small molecule scaffolds that may be further modified to develop nematode-selective inhibitors. Nematode EGFR, MEK1, and PLK1 therefore represent key targets for the development of new anthelmintic medicines.
Collapse
Affiliation(s)
- Jessica Knox
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada.,The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Nicolas Joly
- Programme Équipe Labellisée Ligue Contre Le Cancer, Institut Jacques Monod, UMR7592, Université de Paris, CNRS, Paris, France
| | - Edmond M Linossi
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94158, USA
| | - José A Carmona-Negrón
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Natalia Jura
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94158, USA.,Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Lionel Pintard
- Programme Équipe Labellisée Ligue Contre Le Cancer, Institut Jacques Monod, UMR7592, Université de Paris, CNRS, Paris, France
| | - William Zuercher
- School of Pharmacy, UNC Eshelman, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Peter J Roy
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada. .,The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, M5S 3E1, Canada. .,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
40
|
Giunti S, Andersen N, Rayes D, De Rosa MJ. Drug discovery: Insights from the invertebrate Caenorhabditis elegans. Pharmacol Res Perspect 2021; 9:e00721. [PMID: 33641258 PMCID: PMC7916527 DOI: 10.1002/prp2.721] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/06/2021] [Indexed: 12/18/2022] Open
Abstract
Therapeutic drug development is a long, expensive, and complex process that usually takes 12-15 years. In the early phases of drug discovery, in particular, there is a growing need for animal models that ensure the reduction in both cost and time. Caenorhabditis elegans has been traditionally used to address fundamental aspects of key biological processes, such as apoptosis, aging, and gene expression regulation. During the last decade, with the advent of large-scale platforms for screenings, this invertebrate has also emerged as an essential tool in the pharmaceutical research industry to identify novel drugs and drug targets. In this review, we discuss the reasons why C. elegans has been positioned as an outstanding cost-effective option for drug discovery, highlighting both the advantages and drawbacks of this model. Particular attention is paid to the suitability of this nematode in large-scale genetic and pharmacological screenings. High-throughput screenings in C. elegans have indeed contributed to the breakthrough of a wide variety of candidate compounds involved in extensive fields including neurodegeneration, pathogen infections and metabolic disorders. The versatility of this nematode, which enables its instrumentation as a model of human diseases, is another attribute also herein underscored. As illustrative examples, we discuss the utility of C. elegans models of both human neurodegenerative diseases and parasitic nematodes in the drug discovery industry. Summing up, this review aims to demonstrate the impact of C. elegans models on the drug discovery pipeline.
Collapse
Affiliation(s)
- Sebastián Giunti
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) CCT UNS‐CONICETBahía BlancaArgentina
- Dpto de Biología, Bioquímica y FarmaciaUniversidad Nacional del SurBahía BlancaArgentina
| | - Natalia Andersen
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) CCT UNS‐CONICETBahía BlancaArgentina
- Dpto de Biología, Bioquímica y FarmaciaUniversidad Nacional del SurBahía BlancaArgentina
| | - Diego Rayes
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) CCT UNS‐CONICETBahía BlancaArgentina
- Dpto de Biología, Bioquímica y FarmaciaUniversidad Nacional del SurBahía BlancaArgentina
| | - María José De Rosa
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) CCT UNS‐CONICETBahía BlancaArgentina
- Dpto de Biología, Bioquímica y FarmaciaUniversidad Nacional del SurBahía BlancaArgentina
| |
Collapse
|
41
|
Mohammedsalih KM, Krücken J, Bashar A, Juma FR, Abdalmalaik AAH, Khalafalla A, Abakar A, Coles G, von Samson-Himmelstjerna G. Susceptible trichostrongyloid species mask presence of benzimidazole-resistant Haemonchus contortus in cattle. Parasit Vectors 2021; 14:101. [PMID: 33557939 PMCID: PMC7869217 DOI: 10.1186/s13071-021-04593-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 01/11/2021] [Indexed: 11/25/2022] Open
Abstract
Background Benzimidazole (BZ) anthelmintics are widely used to control infections with parasitic nematodes, but BZ resistance is an emerging threat among several nematode species infecting humans and animals. In Sudan, BZ-resistant Haemonchus contortus populations were recently reported in goats in South Darfur State. The objective of this study was to collect data regarding the situation of BZ resistance in cattle parasitic nematodes in South Darfur using phenotypic and molecular approaches, besides providing some epidemiological data on nematodes in cattle. Methods The faecal egg count reduction test and the egg hatch test (EHT) were used to evaluate benzimidazole efficacy in cattle nematodes in five South Darfur study areas: Beleil, Kass, Nyala, Rehed Al-Birdi and Tulus. Genomic DNA was extracted from pools of third-stage larvae (L3) (n = 40) during trials, before and after treatment, and pools of adult male Haemonchus spp. (n = 18) from abattoirs. The polymorphisms F167Y, E198A and F200Y in isotype 1 β-tubulin genes of H. contortus and H. placei were analysed using Sanger and pyrosequencing. Results Prevalence of gastro-intestinal helminths in cattle was 71% (313/443). Reduced albendazole faecal egg count reduction efficacy was detected in three study areas: Nyala (93.7%), Rehed Al-Birdi (89.7%) and Tulus (88.2%). In the EHT, EC50 values of these study areas ranged between 0.032 and 0.037 µg/ml thiabendazole. Genus-specific PCRs detected the genera Haemonchus, Trichostrongylus and Cooperia in L3 samples collected after albendazole treatment. Sanger sequencing followed by pyrosequencing assays did not detect elevated frequencies of known BZ resistance-associated alleles in codon F167Y, E198A and F200Y in isotype 1 β-tubulin gene of H. placei (≤ 11.38%). However, polymorphisms were detected in H. contortus and in samples with mixed infections with H. contortus and H. placei at codon 198, including E198L (16/58), E198V (2/58) and potentially E198Stop (1/58). All pooled L3 samples post-albendazole treatment (n = 13) were identified as H. contortus with an E198L substitution at codon 198. Conclusions To the knowledge of the authors, this is the first report of reduced albendazole efficacy in cattle in Sudan and is the first study describing an E198L substitution in phenotypically BZ-resistant nematodes collected from cattle.![]()
Collapse
Affiliation(s)
| | - Jürgen Krücken
- Institute for Parasitology and Tropical Veterinary Medicine, Freie Universität Berlin, Robert-von-Ostertag-Street 7-13, 14163, Berlin, Germany
| | - Ahmed Bashar
- Faculty of Veterinary Science, University of Nyala, P.O. Box 155, Nyala, Sudan
| | - Fathel-Rahman Juma
- Faculty of Veterinary Science, University of Nyala, P.O. Box 155, Nyala, Sudan
| | | | - Amna Khalafalla
- Faculty of Veterinary Medicine, University of Khartoum, P.O. Box 32, Khartoum North, Sudan
| | - Adam Abakar
- Faculty of Medical Laboratory Sciences, University of Gezira, P.O. Box 20, Wadmedani, Sudan
| | - Gerald Coles
- Ubley Biologics, Ubley, P.O. Box 170, Bristol, BS40 6JA, UK
| | - Georg von Samson-Himmelstjerna
- Institute for Parasitology and Tropical Veterinary Medicine, Freie Universität Berlin, Robert-von-Ostertag-Street 7-13, 14163, Berlin, Germany.
| |
Collapse
|
42
|
Wit J, Dilks CM, Andersen EC. Complementary Approaches with Free-living and Parasitic Nematodes to Understanding Anthelmintic Resistance. Trends Parasitol 2020; 37:240-250. [PMID: 33317926 DOI: 10.1016/j.pt.2020.11.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022]
Abstract
Anthelmintic drugs are the major line of defense against parasitic nematode infections, but the arsenal is limited and resistance threatens sustained efficacy of the available drugs. Discoveries of the modes of action of these drugs and mechanisms of resistance have predominantly come from studies of a related nonparasitic nematode species, Caenorhabditis elegans, and the parasitic nematode Haemonchus contortus. Here, we discuss how our understanding of anthelmintic resistance and modes of action came from the interplay of results from each of these species. We argue that this 'cycle of discovery', where results from one species inform the design of experiments in the other, can use the complementary strengths of both to understand anthelmintic modes of action and mechanisms of resistance.
Collapse
Affiliation(s)
- Janneke Wit
- Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Clayton M Dilks
- Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA; Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, IL 60208, USA
| | - Erik C Andersen
- Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
43
|
Salikin NH, Nappi J, Majzoub ME, Egan S. Combating Parasitic Nematode Infections, Newly Discovered Antinematode Compounds from Marine Epiphytic Bacteria. Microorganisms 2020; 8:E1963. [PMID: 33322253 PMCID: PMC7764037 DOI: 10.3390/microorganisms8121963] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/08/2020] [Accepted: 12/08/2020] [Indexed: 02/06/2023] Open
Abstract
Parasitic nematode infections cause debilitating diseases and impede economic productivity. Antinematode chemotherapies are fundamental to modern medicine and are also important for industries including agriculture, aquaculture and animal health. However, the lack of suitable treatments for some diseases and the rise of nematode resistance to many available therapies necessitates the discovery and development of new drugs. Here, marine epiphytic bacteria represent a promising repository of newly discovered antinematode compounds. Epiphytic bacteria are ubiquitous on marine surfaces where they are under constant pressure of grazing by bacterivorous predators (e.g., protozoans and nematodes). Studies have shown that these bacteria have developed defense strategies to prevent grazers by producing toxic bioactive compounds. Although several active metabolites against nematodes have been identified from marine bacteria, drug discovery from marine microorganisms remains underexplored. In this review, we aim to provide further insight into the need and potential for marine epiphytic bacteria to become a new source of antinematode drugs. We discuss current and emerging strategies, including culture-independent high throughput screening and the utilization of Caenorhabditis elegans as a model target organism, which will be required to advance antinematode drug discovery and development from marine microbial sources.
Collapse
Affiliation(s)
- Nor Hawani Salikin
- Centre for Marine Science and Innovation, School of Biological, Earth and Environmental Sciences, UNSW, Sydney, NSW 2052, Australia; (N.H.S.); (J.N.); (M.E.M.)
- School of Industrial Technology, Universiti Sains Malaysia, USM, 11800 Penang, Malaysia
| | - Jadranka Nappi
- Centre for Marine Science and Innovation, School of Biological, Earth and Environmental Sciences, UNSW, Sydney, NSW 2052, Australia; (N.H.S.); (J.N.); (M.E.M.)
| | - Marwan E. Majzoub
- Centre for Marine Science and Innovation, School of Biological, Earth and Environmental Sciences, UNSW, Sydney, NSW 2052, Australia; (N.H.S.); (J.N.); (M.E.M.)
| | - Suhelen Egan
- Centre for Marine Science and Innovation, School of Biological, Earth and Environmental Sciences, UNSW, Sydney, NSW 2052, Australia; (N.H.S.); (J.N.); (M.E.M.)
| |
Collapse
|
44
|
Dilks CM, Hahnel SR, Sheng Q, Long L, McGrath PT, Andersen EC. Quantitative benzimidazole resistance and fitness effects of parasitic nematode beta-tubulin alleles. Int J Parasitol Drugs Drug Resist 2020; 14:28-36. [PMID: 32858477 PMCID: PMC7473882 DOI: 10.1016/j.ijpddr.2020.08.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/04/2020] [Accepted: 08/04/2020] [Indexed: 11/17/2022]
Abstract
Infections by parasitic nematodes inflict a huge burden on the health of humans and livestock throughout the world. Anthelmintic drugs are the first line of defense against these infections. Unfortunately, resistance to these drugs is rampant and continues to spread. To improve treatment strategies, we must understand the genetics and molecular mechanisms that underlie resistance. Studies of the fungus Aspergillus nidulans and the free-living nematode Caenorhabditis elegans discovered that a beta-tubulin gene is mutated in benzimidazole (BZ) resistant strains. In parasitic nematode populations, three beta-tubulin alleles, F167Y, E198A, and F200Y, have long been correlated with resistance. Additionally, improvements in sequencing technologies have identified new alleles - E198V, E198L, E198K, E198I, and E198Stop - also correlated with BZ resistance. However, none of these alleles have been proven to cause resistance. To empirically demonstrate this point, we independently introduced the F167Y, E198A, and F200Y alleles as well as two of the newly identified alleles, E198V and E198L, into the BZ susceptible C. elegans N2 genetic background using the CRISPR-Cas9 system. These genome-edited strains were exposed to both albendazole and fenbendazole to quantitatively measure animal responses to BZs. We used a range of concentrations for each BZ compound to define response curves and found that all five of the alleles conferred resistance to BZ compounds equal to a loss of the entire beta-tubulin gene. These results prove that the parasite beta-tubulin alleles cause resistance. The E198V allele is found at low frequencies along with the E198L allele in natural parasite populations, suggesting that it could affect fitness. We performed competitive fitness assays and demonstrated that the E198V allele reduces animal health, supporting the hypothesis that this allele might be less fit in field populations. Overall, we present a powerful platform to quantitatively assess anthelmintic resistance and effects of specific resistance alleles on organismal fitness in the presence or absence of the drug.
Collapse
Affiliation(s)
- Clayton M Dilks
- Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA; Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, IL, 60208, USA
| | - Steffen R Hahnel
- Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA
| | - Qicong Sheng
- Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA
| | - Lijiang Long
- Center for Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA; Interdisciplinary Graduate Program in Quantitative Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Patrick T McGrath
- Center for Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Erik C Andersen
- Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA; Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, IL, 60208, USA.
| |
Collapse
|
45
|
Bartley DJ, Jewell NJ, Andrews LM, Mitchell S, Morrison AA. Molecular and phenotypic characterisation of fenbendazole resistance in a field-derived isolate of Ostertagia ostertagi. Vet Parasitol 2020; 289:109319. [PMID: 33249304 DOI: 10.1016/j.vetpar.2020.109319] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 10/23/2022]
Abstract
The prevalence of anthelmintic resistance in the bovine nematode Cooperia oncophora has been well documented globally but lack of efficacy against the more pathogenic nematode species Ostertagia ostertagi is less common. The sensitivity of an O. ostertagi isolate to the benzimidazole class of anthelmintic was investigated using classical parasitological techniques following apparent clinical failure of controlled release fenbendazole capsule administration in first season grazers at pasture. A controlled efficacy test (CET) was conducted in conjunction with sequencing of the β-tubulin isotype 1 gene of larvae pre- and post-fenbendazole administration. Twelve helminth-naïve calves were infected experimentally with 20,000 third stage larvae; six received oral fenbendazole (7.5 mg/kg bodyweight) 28 days post infection. Total abomasal nematode burdens were compared between treatment and control groups to determine efficacy. Fenbendazole resistance in O. ostertagi was confirmed with a total treatment failure in reducing worm burden: efficacy of 0%. Sequence analysis of the β-tubulin isotype-1 gene from forty-five infective larvae from both control and treated groups was performed. The three commonest single nucleotide polymorphisms (SNPs) associated with benzimidazole resistance, namely F167Y, E198A and F200Y, were examined. The predominant resistance-associated SNPs were F200Y (78 % control and 79 % treated groups) and F167Y (remaining genotypes) and emphasises the importance of these SNPs in clinical disease in this isolate. The development of diagnostic molecular tools based on a characterised field-derived isolate of benzimidazole-resistant Ostertagia will enable future prevalence surveys to be undertaken to assess the possible risk posed by resistance in this economically important species.
Collapse
Affiliation(s)
- Dave J Bartley
- Disease Control, Moredun Research Institute, Pentlands Science Park, Edinburgh, EH26 0PZ, United Kingdom.
| | - Natalie J Jewell
- Animal and Plant Health Agency (APHA), Job's Well Road, Carmarthen, Carmarthenshire SA31 3EZ, United Kingdom
| | - Leigh M Andrews
- Disease Control, Moredun Research Institute, Pentlands Science Park, Edinburgh, EH26 0PZ, United Kingdom
| | - Sian Mitchell
- Animal and Plant Health Agency (APHA), Job's Well Road, Carmarthen, Carmarthenshire SA31 3EZ, United Kingdom
| | - Alison A Morrison
- Disease Control, Moredun Research Institute, Pentlands Science Park, Edinburgh, EH26 0PZ, United Kingdom
| |
Collapse
|
46
|
Hahnel SR, Dilks CM, Heisler I, Andersen EC, Kulke D. Caenorhabditis elegans in anthelmintic research - Old model, new perspectives. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2020; 14:237-248. [PMID: 33249235 PMCID: PMC7704361 DOI: 10.1016/j.ijpddr.2020.09.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 09/25/2020] [Accepted: 09/26/2020] [Indexed: 12/13/2022]
Abstract
For more than four decades, the free-living nematode Caenorhabditis elegans has been extensively used in anthelmintic research. Classic genetic screens and heterologous expression in the C. elegans model enormously contributed to the identification and characterization of molecular targets of all major anthelmintic drug classes. Although these findings provided substantial insights into common anthelmintic mechanisms, a breakthrough in the treatment and control of parasitic nematodes is still not in sight. Instead, we are facing increasing evidence that the enormous diversity within the phylum Nematoda cannot be recapitulated by any single free-living or parasitic species and the development of novel broad-spectrum anthelmintics is not be a simple goal. In the present review, we summarize certain milestones and challenges of the C. elegans model with focus on drug target identification, anthelmintic drug discovery and identification of resistance mechanisms. Furthermore, we present new perspectives and strategies on how current progress in C. elegans research will support future anthelmintic research.
Collapse
Affiliation(s)
| | - Clayton M Dilks
- Northwestern University, Department of Molecular Biosciences, Evanston, IL, USA.
| | | | - Erik C Andersen
- Northwestern University, Department of Molecular Biosciences, Evanston, IL, USA.
| | | |
Collapse
|
47
|
Guest M, Kriek N, Flemming AJ. Studies of an insecticidal inhibitor of acetyl-CoA carboxylase in the nematode C. elegans. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2020; 169:104604. [PMID: 32828380 DOI: 10.1016/j.pestbp.2020.104604] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 04/24/2020] [Accepted: 04/30/2020] [Indexed: 06/11/2023]
Abstract
We have studied the mode of action of the insecticide spirotetramat in the nematode Caenorhabditis elegans. A combination of symptomology, forward genetics and genome editing show that spirotetramat acts on acetyl-CoA carboxylase (ACC) in C. elegans, as it does in insects. We found C. elegans embryos exposed to spirotetramat show a cell division defect which closely resembles the phenotype of loss-of-function mutations in the gene pod-2, which encodes ACC. We then identified two mutations in the carboxyl transferase domain of pod-2 (ACC) which confer resistance and were confirmed using CRISPR/Cas9. One of these mutations substitutes an invertebrate-specific amino acid with one ubiquitous in other taxa; this residue may, therefore, be a determinant of the selectivity of spirotetramat for invertebrates. Such a mutation may also be the target of selection for resistance in the field. Our study is a further demonstration of the utility of C. elegans in studying bioactive chemicals.
Collapse
Affiliation(s)
- M Guest
- Syngenta, Jealott's Hill International Research Centre, Bracknell, Berkshire RG42 6EY, UK
| | - N Kriek
- Syngenta, Jealott's Hill International Research Centre, Bracknell, Berkshire RG42 6EY, UK
| | - A J Flemming
- Syngenta, Jealott's Hill International Research Centre, Bracknell, Berkshire RG42 6EY, UK.
| |
Collapse
|
48
|
Tan TK, Lim YAL, Chua KH, Chai HC, Low VL, Bathmanaban P, Affendi S, Wang D, Panchadcharam C. Characterization of benzimidazole resistance in Haemonchus contortus: integration of phenotypic, genotypic and proteomic approaches. Parasitol Res 2020; 119:2851-2862. [PMID: 32651637 DOI: 10.1007/s00436-020-06790-5] [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: 06/28/2019] [Accepted: 06/28/2020] [Indexed: 11/29/2022]
Abstract
The field strain of Haemonchus contortus has a long history of anthelmintic resistance. To understand this phenomenon, the benzimidazole resistance profile was characterized from the Malaysian field-resistant strain by integrating phenotypic, genotypic and proteomic approaches. The faecal egg count reduction test (FECRT) demonstrated that benzimidazole resistance was at a critical level in the studied strain. The primary resistance mechanism was attributed to F200Y mutation in the isotype 1 β-tubulin gene as revealed by AS-PCR and direct sequencing. Furthermore, the protein response of the resistant strain towards benzimidazole (i.e., albendazole) treatment was investigated via two-dimensional difference gel electrophoresis (2D-DIGE) and tandem liquid chromatography-mass spectrometry (LC-MS/MS). These investigations illustrated an up-regulation of antioxidant (i.e., ATP-binding region and heat-shock protein 90, superoxide dismutase) and metabolic (i.e., glutamate dehydrogenase) enzymes and down-regulation of glutathione S-transferase, malate dehydrogenase, and other structural and cytoskeletal proteins (i.e., actin, troponin T). Findings from this study are pivotal in updating the current knowledge on anthelmintic resistance and providing new insights into the defence mechanisms of resistant nematodes towards drug treatment.
Collapse
Affiliation(s)
- Tiong Kai Tan
- Department of Parasitology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| | - Yvonne A L Lim
- Department of Parasitology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia. .,Centre of Excellence for Research in AIDS (CERIA), University of Malaya, Kuala Lumpur, Malaysia.
| | - Kek Heng Chua
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Hwa Chia Chai
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Van Lun Low
- Tropical Infectious Diseases Research and Education Centre (TIDREC), University of Malaya, Kuala Lumpur, Malaysia
| | | | - Sarah Affendi
- Tropical Infectious Diseases Research and Education Centre (TIDREC), University of Malaya, Kuala Lumpur, Malaysia
| | - Daryi Wang
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan
| | - Chandrawathani Panchadcharam
- Department of Veterinary Services, Ministry of Agriculture and Agro-Based Industry Malaysia, Federal Government Administrative Center, Putrajaya, Malaysia
| |
Collapse
|
49
|
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
|
50
|
Palma A, Matamoros G, Escobar D, Sánchez AL, Fontecha G. Absence of mutations associated with resistance to benzimidazole in the beta-tubulin gene of Ascaris suum. Rev Soc Bras Med Trop 2020; 53:e20190155. [PMID: 32187331 PMCID: PMC7094045 DOI: 10.1590/0037-8682-0155-2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 12/04/2019] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION: Benzimidazoles are commonly used for the control of veterinary nematodes. Resistance to benzimidazoles has been associated with three single nucleotide polymorphisms in the β-tubulin gene of common nematodes. However, these mutations are infrequent in the genus Ascaris spp. METHODS: In order to determine mutations associated with benzimidazole resistance in Ascaris suum, worms were collected from slaughtered pigs and a partial region of the β-tubulin gene was sequenced. RESULTS: All parasites showed the wildtype genotype for codons 167, 198, and 200 of the β-tubulin gene. CONCLUSIONS: This is the first report of genetic sequences associated with benzimidazole resistance in A. suum.
Collapse
Affiliation(s)
- Adalid Palma
- Microbiology Research Institute, Universidad Nacional Autónoma de Honduras (UNAH), Tegucigalpa, Honduras
| | - Gabriela Matamoros
- Microbiology Research Institute, Universidad Nacional Autónoma de Honduras (UNAH), Tegucigalpa, Honduras.,Department of Health Sciences, Brock University, St. Catharines, Ontario. Canada
| | - Denis Escobar
- Microbiology Research Institute, Universidad Nacional Autónoma de Honduras (UNAH), Tegucigalpa, Honduras
| | - Ana Lourdes Sánchez
- Department of Health Sciences, Brock University, St. Catharines, Ontario. Canada
| | - Gustavo Fontecha
- Microbiology Research Institute, Universidad Nacional Autónoma de Honduras (UNAH), Tegucigalpa, Honduras
| |
Collapse
|