1
|
Koike A, Brindley PJ. CRISPR/Cas genome editing, functional genomics, and diagnostics for parasitic helminths. Int J Parasitol 2025:S0020-7519(25)00092-X. [PMID: 40348052 DOI: 10.1016/j.ijpara.2025.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/30/2025] [Accepted: 05/04/2025] [Indexed: 05/14/2025]
Abstract
Functional genomics using CRISPR (Clustered Regulatory Interspaced Short Palindromic Repeats)/Cas (CRISPR-associated endonuclease)-based approaches has revolutionized biomedical sciences. Gene editing is also widespread in parasitology generally and its use is increasing in studies on helminths including flatworm and roundworm parasites. Here, we survey the progress, specifically with experimental CRISPR-facilitated functional genomics to investigate helminth biology and pathogenesis, and also with the burgeoning use of CRISPR-based methods to assist in diagnosis of helminth infections. We also provide an historical timeline of the introduction and uses of CRISPR in helminth species to date.
Collapse
Affiliation(s)
- Akito Koike
- Department of Microbiology, Immunology & Tropical Medicine, School of Medicine & Health Sciences, George Washington University, Washington, D.C. 20037, USA
| | - Paul J Brindley
- Department of Microbiology, Immunology & Tropical Medicine, School of Medicine & Health Sciences, George Washington University, Washington, D.C. 20037, USA.
| |
Collapse
|
2
|
Allen N, Huson KM, Prchal L, Robinson MW, Brophy PM, Morphew RM. Detoxome Capacity of the Adult Rumen Fluke Calicophoron daubneyi Extends into Its Secreted Extracellular Vesicles. J Proteome Res 2025; 24:624-638. [PMID: 39829022 PMCID: PMC11812014 DOI: 10.1021/acs.jproteome.4c00615] [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: 08/27/2024] [Revised: 12/09/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025]
Abstract
Helminth parasites have long adapted to survive hostile host environments and can likely adapt against the chemical anthelmintic challenge. One proposed adaptation route is via Phase I and II xenobiotic metabolizing enzymes (XMEs). For successful Helminth pharmacotherapy discovery programs, a working understanding of Helminth-derived chemical detoxification, the Helminth detoxome, is a must. At present, the detoxome of a newly emerging Helminth parasite, the rumen fluke Calicophoron daubneyi, remains unexplored. Thus, a combined bioinformatics, sub-, and global-proteomic approach has been employed to examine the detoxome of adult C. daubneyi. Transcriptome analysis revealed a complement of Phase I (cytochrome P450s and monoamine oxygenases) and Phase II (glutathione transferases [GSTs] and sulfotransferases) XMEs. Affinity-led subproteomic exploration of the GSTs revealed six GST isoforms in adult rumen fluke (CdGST-Mu1-2, S1, and S3-5), with global approaches identifying additional GSTs (CdGST-O1-2, Z1, and S2) and a unique egg-specific variant (CdGST-S6). Examination of C. daubneyi extracellular vesicles revealed a GST profile replicating that of the adult with the absence of two isoforms (CdGST-S2 and S4), with an additional identification of a sulfotransferase. These data represent the first exploration into the complete rumen fluke detoxification capacity and will provide direction for future anthelmintic discovery programs.
Collapse
Affiliation(s)
- Nathan
Rhys Allen
- Department
of Life Sciences, Aberystwyth University, Aberystwyth, Wales SY23 3DA, U.K.
| | - Kathryn M. Huson
- School
of Biological Sciences, Queen’s University
Belfast, Belfast, Northern Ireland BT9 5BY, U.K.
| | - Lukas Prchal
- Department
of Life Sciences, Aberystwyth University, Aberystwyth, Wales SY23 3DA, U.K.
| | - Mark W. Robinson
- School
of Biological Sciences, Queen’s University
Belfast, Belfast, Northern Ireland BT9 5BY, U.K.
| | - Peter M. Brophy
- Department
of Life Sciences, Aberystwyth University, Aberystwyth, Wales SY23 3DA, U.K.
| | - Russell M. Morphew
- Department
of Life Sciences, Aberystwyth University, Aberystwyth, Wales SY23 3DA, U.K.
| |
Collapse
|
3
|
McIntyre J, Morrison A, Maitland K, Berger D, Price DRG, Dougan S, Grigoriadis D, Tracey A, Holroyd N, Bull K, Rose Vineer H, Glover MJ, Morgan ER, Nisbet AJ, McNeilly TN, Bartley Y, Sargison N, Bartley D, Berriman M, Cotton JA, Devaney E, Laing R, Doyle SR. Chromosomal genome assembly resolves drug resistance loci in the parasitic nematode Teladorsagia circumcincta. PLoS Pathog 2025; 21:e1012820. [PMID: 39913358 PMCID: PMC11801625 DOI: 10.1371/journal.ppat.1012820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 12/09/2024] [Indexed: 02/11/2025] Open
Abstract
The parasitic nematode Teladorsagia circumcincta is one of the most important pathogens of sheep and goats in temperate climates worldwide and can rapidly evolve resistance to drugs used to control it. To understand the genetics of drug resistance, we have generated a highly contiguous genome assembly for the UK T. circumcincta isolate, MTci2. Assembly using PacBio long-reads and Hi-C long-molecule scaffolding together with manual curation resulted in a 573 Mb assembly (N50 = 84 Mb, total scaffolds = 1,286) with five autosomal and one sex-linked chromosomal-scale scaffolds consistent with its karyotype. The genome resource was further improved via annotation of 22,948 genes, with manual curation of over 3,200 of these, resulting in a robust and near complete resource (96.3% complete protein BUSCOs) to support basic and applied research on this important veterinary pathogen. Genome-wide analyses of drug resistance, combining evidence from three distinct experiments, identified selection around known candidate genes for benzimidazole, levamisole and ivermectin resistance, as well as novel regions associated with ivermectin and moxidectin resistance. These insights into contemporary and historic genetic selection further emphasise the importance of contiguous genome assemblies in interpreting genome-wide genetic variation associated with drug resistance and identifying key loci to prioritise in developing diagnostic markers of anthelmintic resistance to support parasite control.
Collapse
Affiliation(s)
- Jennifer McIntyre
- School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Garscube Campus, Glasgow, United Kingdom
| | - Alison Morrison
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, United Kingdom
| | - Kirsty Maitland
- School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Garscube Campus, Glasgow, United Kingdom
| | - Duncan Berger
- Wellcome Sanger Institute, Hinxton, Cambridgeshire, United Kingdom
| | - Daniel R. G. Price
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, United Kingdom
| | - Sam Dougan
- Wellcome Sanger Institute, Hinxton, Cambridgeshire, United Kingdom
| | - Dionysis Grigoriadis
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, Cambridgeshire, United Kingdom
| | - Alan Tracey
- Wellcome Sanger Institute, Hinxton, Cambridgeshire, United Kingdom
| | - Nancy Holroyd
- Wellcome Sanger Institute, Hinxton, Cambridgeshire, United Kingdom
| | - Katie Bull
- Veterinary Parasitology and Ecology Group, University of Bristol, Bristol, United Kingdom
| | - Hannah Rose Vineer
- Veterinary Parasitology and Ecology Group, University of Bristol, Bristol, United Kingdom
- University of Liverpool, Institute of Infection, Veterinary and Ecological Sciences, Leahurst Campus, Neston, Cheshire, United Kingdom
| | - Mike J. Glover
- Torch Farm & Equine Ltd., Veterinary Surgeons, South Molton, Devon, United Kingdom
| | - Eric R. Morgan
- Veterinary Parasitology and Ecology Group, University of Bristol, Bristol, United Kingdom
- Queen’s University Belfast, School of Biological Sciences, Belfast, United Kingdom
| | - Alasdair J. Nisbet
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, United Kingdom
| | - Tom N. McNeilly
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, United Kingdom
| | - Yvonne Bartley
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, United Kingdom
| | - Neil Sargison
- Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Dave Bartley
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, United Kingdom
| | - Matt Berriman
- Wellcome Sanger Institute, Hinxton, Cambridgeshire, United Kingdom
| | - James A. Cotton
- Wellcome Sanger Institute, Hinxton, Cambridgeshire, United Kingdom
| | - Eileen Devaney
- School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Garscube Campus, Glasgow, United Kingdom
| | - Roz Laing
- School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Garscube Campus, Glasgow, United Kingdom
| | - Stephen R. Doyle
- Wellcome Sanger Institute, Hinxton, Cambridgeshire, United Kingdom
| |
Collapse
|
4
|
Villamizar-Monsalve MA, Sánchez-Montejo J, López-Abán J, Vicente B, Marín M, Fernández-Ceballos N, Peláez R, Muro A. Development and Application of an In Vitro Drug Screening Assay for Schistosoma mansoni Schistosomula Using YOLOv5. Biomedicines 2024; 12:2894. [PMID: 39767801 PMCID: PMC11727284 DOI: 10.3390/biomedicines12122894] [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: 11/29/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Schistosomiasis impacts over 230 million people globally, with 251.4 million needing treatment. The disease causes intestinal and urinary symptoms, such as hepatic fibrosis, hepatomegaly, splenomegaly, and bladder calcifications. While praziquantel (PZQ) is the primary treatment, its effectiveness against juvenile stages (schistosomula) is limited, highlighting the need for new therapeutic agents, repurposed drugs, or reformulated compounds. Existing microscopy methods for assessing schistosomula viability are labor-intensive, subjective, and time-consuming. METHODS An artificial intelligence (AI)-assisted culture system using YOLOv5 was developed to evaluate compounds against Schistosoma mansoni schistosomula. The AI model, based on object detection, was trained on 4390 images distinguishing between healthy and damaged schistosomula. The system was externally validated against human counters, and a small-scale assay was performed to demonstrate its potential for larger-scale assays in the future. RESULTS The AI model exhibited high accuracy, achieving a mean average precision (mAP) of 0.966 (96.6%) and effectively differentiating between healthy and damaged schistosomula. External validation demonstrated significantly improved accuracy and counting time compared to human counters. A small-scale assay was conducted to validate the system, identifying 28 potential compounds with schistosomicidal activity against schistosomula in vitro and providing their preliminary LC50 values. CONCLUSIONS This AI-powered method significantly improves accuracy and time efficiency compared to traditional microscopy. It enables the evaluation of compounds for potential schistosomiasis drugs without the need for dyes or specialized equipment, facilitating more efficient drug assessment.
Collapse
Affiliation(s)
- María Alejandra Villamizar-Monsalve
- Infectious and Tropical Diseases Research Group (e-INTRO), Biomedical Research Institute of Salamanca, Research Centre for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), Faculty of Pharmacy, University of Salamanca, 37007 Salamanca, Spain; (M.A.V.-M.); (J.S.-M.); (B.V.); (A.M.)
| | - Javier Sánchez-Montejo
- Infectious and Tropical Diseases Research Group (e-INTRO), Biomedical Research Institute of Salamanca, Research Centre for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), Faculty of Pharmacy, University of Salamanca, 37007 Salamanca, Spain; (M.A.V.-M.); (J.S.-M.); (B.V.); (A.M.)
| | - Julio López-Abán
- Infectious and Tropical Diseases Research Group (e-INTRO), Biomedical Research Institute of Salamanca, Research Centre for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), Faculty of Pharmacy, University of Salamanca, 37007 Salamanca, Spain; (M.A.V.-M.); (J.S.-M.); (B.V.); (A.M.)
| | - Belén Vicente
- Infectious and Tropical Diseases Research Group (e-INTRO), Biomedical Research Institute of Salamanca, Research Centre for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), Faculty of Pharmacy, University of Salamanca, 37007 Salamanca, Spain; (M.A.V.-M.); (J.S.-M.); (B.V.); (A.M.)
| | - Miguel Marín
- Organic and Pharmaceutical Chemistry Department, Biomedical Research Institute of Salamanca Research (IBSAL), Faculty of Pharmacy, University of Salamanca, 37007 Salamanca, Spain; (M.M.); (N.F.-C.); (R.P.)
| | - Noelia Fernández-Ceballos
- Organic and Pharmaceutical Chemistry Department, Biomedical Research Institute of Salamanca Research (IBSAL), Faculty of Pharmacy, University of Salamanca, 37007 Salamanca, Spain; (M.M.); (N.F.-C.); (R.P.)
| | - Rafael Peláez
- Organic and Pharmaceutical Chemistry Department, Biomedical Research Institute of Salamanca Research (IBSAL), Faculty of Pharmacy, University of Salamanca, 37007 Salamanca, Spain; (M.M.); (N.F.-C.); (R.P.)
| | - Antonio Muro
- Infectious and Tropical Diseases Research Group (e-INTRO), Biomedical Research Institute of Salamanca, Research Centre for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), Faculty of Pharmacy, University of Salamanca, 37007 Salamanca, Spain; (M.A.V.-M.); (J.S.-M.); (B.V.); (A.M.)
| |
Collapse
|
5
|
Shoko R, Mazadza A. Computer-Aided Discovery of Abrus precatorius Compounds With Anti-Schistosomal Potential. Biomed Eng Comput Biol 2024; 15:11795972241294112. [PMID: 39530083 PMCID: PMC11552047 DOI: 10.1177/11795972241294112] [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: 05/25/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Schistosomiasis, which causes over 200 000 deaths annually, has since the 1970s been controlled by praziquintel. The reliance on a single drug to combat schistosomiasis, and reports of laboratory resistance to the drug, has created an urgent need in the scientific community to develop new chemotherapies to complement or supplement praziquantel. Medicinal plants are a potential reservoir of compounds with schistosomicidal activity. In the current study, we carried out computer-aided screening of Abrus precatorius compounds to discover compounds with potential to inhibit Schistosoma mansoni purine nucleoside phosphorylase (SmPNP). Thus, 99 compounds retrieved from Lotus Natural Compounds Database were docked into the active site of SmPNP. The top-ranked compounds were subjected to Lipinski's druglikeness and toxicity risk predictions. Three lead compounds, abrusogenin, cirsimaritin and hispidulin, were identified as having high binding affinities, favourable interactions with SmPNP active site residues and good toxicity risk prediction results. Molecular dynamics (MD) simulations were used to assess the stability of the interactions of these lead compounds with SmPNP. Collectively, analyses of the MD trajectories confirms that the lead compounds bound and interacted stably with active site residues of SmPNP. We conclude that abrusogenin, cirsimaritin and hispidulin could serve as hit compounds for the development of new antischistosomal drugs, based on plant-derived natural products. However, experimental studies are required to further evaluate the potentials of these compounds as possible therapeutics against schistosomiasis.
Collapse
Affiliation(s)
- Ryman Shoko
- Department of Biology, School of Natural Sciences and Mathematics, Chinhoyi University of Technology, Chinhoyi, Zimbabwe
| | - Allen Mazadza
- Department of Biology, School of Natural Sciences and Mathematics, Chinhoyi University of Technology, Chinhoyi, Zimbabwe
| |
Collapse
|
6
|
JUTZELER KS, PLATT RN, DIAZ R, MORALES M, LE CLEC’H W, CHEVALIER FD, ANDERSON TJ. Abundant genetic variation is retained in many laboratory schistosome populations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.21.619418. [PMID: 39484487 PMCID: PMC11526883 DOI: 10.1101/2024.10.21.619418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Schistosomes are obligately sexual blood flukes that can be maintained in the laboratory using freshwater snails as intermediate and rodents as definitive hosts. The genetic composition of laboratory schistosome populations is poorly understood: whether genetic variation has been purged due to serial inbreeding or retained is unclear. We sequenced 19 - 24 parasites from each of five laboratory Schistosoma mansoni populations and compared their genomes with published exome data from four S. mansoni field populations. We found abundant genomic variation (0.897 - 1.22 million variants) within laboratory populations: these retained on average 49% (π = 3.27e-04 - 8.94e-04) of the nucleotide diversity observed in the four field parasite populations (π = 1.08e-03 - 2.2e-03). However, the pattern of variation was very different in laboratory and field populations. Tajima's D was positive in all laboratory populations except SmBRE, indicative of recent population bottlenecks, but negative in all field populations. Current effective population size estimates of laboratory populations were lower (2 - 258) compared to field populations (3,174 - infinity). The distance between markers at which linkage disequilibrium (LD) decayed to 0.5 was longer in laboratory populations (59 bp - 180 kb) compared to field populations (9 bp - 9.5 kb). SmBRE was the least variable; this parasite also shows low fitness across the lifecycle, consistent with inbreeding depression. The abundant genetic variation present in most laboratory schistosome populations has several important implications: (i) measurement of parasite phenotypes, such as drug resistance, using laboratory parasite populations will determine average values and underestimate trait variation; (ii) genome-wide association studies (GWAS) can be conducted in laboratory schistosome populations by measuring phenotypes and genotypes of individual worms; (iii) genetic drift may lead to divergence in schistosome populations maintained in different laboratories. We conclude that the abundant genetic variation retained within many laboratory schistosome populations can provide valuable, untapped opportunities for schistosome research.
Collapse
Affiliation(s)
- Kathrin S. JUTZELER
- Host parasite Interaction Program, Texas Biomedical Research Institute, P.O. Box 760549, 78245 San Antonio, Texas, USA
- UT Health, Microbiology, Immunology & Molecular Genetics, San Antonio, TX 78229
| | - Roy N. PLATT
- Disease Intervention and Prevention program, Texas Biomedical Research Institute, P.O. Box 760549, 78245 San Antonio, Texas, USA
| | - Robbie DIAZ
- Texas Biomedical Research Institute, P.O. Box 760549, 78245 San Antonio, Texas, USA
| | - Madison MORALES
- Texas Biomedical Research Institute, P.O. Box 760549, 78245 San Antonio, Texas, USA
| | - Winka LE CLEC’H
- Host parasite Interaction Program, Texas Biomedical Research Institute, P.O. Box 760549, 78245 San Antonio, Texas, USA
| | - Frédéric D. CHEVALIER
- Host parasite Interaction Program, Texas Biomedical Research Institute, P.O. Box 760549, 78245 San Antonio, Texas, USA
| | - Timothy J.C. ANDERSON
- Disease Intervention and Prevention program, Texas Biomedical Research Institute, P.O. Box 760549, 78245 San Antonio, Texas, USA
| |
Collapse
|
7
|
Ekloh W, Asafu-Adjaye A, Tawiah-Mensah CNL, Ayivi-Tosuh SM, Quartey NKA, Aiduenu AF, Gayi BK, Koudonu JAM, Basing LA, Yamoah JAA, Dofuor AK, Osei JHN. A comprehensive exploration of schistosomiasis: Global impact, molecular characterization, drug discovery, artificial intelligence and future prospects. Heliyon 2024; 10:e33070. [PMID: 38988508 PMCID: PMC11234110 DOI: 10.1016/j.heliyon.2024.e33070] [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: 02/02/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024] Open
Abstract
Schistosomiasis, one of the neglected tropical diseases which affects both humans and animals, is caused by trematode worms of the genus Schistosoma. The disease is caused by several species of Schistosoma which affect several organs such as urethra, liver, bladder, intestines, skin and bile ducts. The life cycle of the disease involves an intermediate host (snail) and a mammalian host. It affects people who are in close proximity to water bodies where the intermediate host is abundant. Common clinical manifestations of the disease at various stages include fever, chills, headache, cough, dysuria, hyperplasia and hydronephrosis. To date, most of the control strategies are dependent on effective diagnosis, chemotherapy and public health education on the biology of the vectors and parasites. Microscopy (Kato-Katz) is considered the golden standard for the detection of the parasite, while praziquantel is the drug of choice for the mass treatment of the disease since no vaccines have yet been developed. Most of the previous reviews on schistosomiasis have concentrated on epidemiology, life cycle, diagnosis, control and treatment. Thus, a comprehensive review that is in tune with modern developments is needed. Here, we extend this domain to cover historical perspectives, global impact, symptoms and detection, biochemical and molecular characterization, gene therapy, current drugs and vaccine status. We also discuss the prospects of using plants as potential and alternative sources of novel anti-schistosomal agents. Furthermore, we highlight advanced molecular techniques, imaging and artificial intelligence that may be useful in the future detection and treatment of the disease. Overall, the proper detection of schistosomiasis using state-of-the-art tools and techniques, as well as development of vaccines or new anti-schistosomal drugs may aid in the elimination of the disease.
Collapse
Affiliation(s)
- William Ekloh
- Department of Biochemistry, School of Biological Sciences, College of Agriculture and Natural Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Andy Asafu-Adjaye
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Christopher Nii Laryea Tawiah-Mensah
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | | | - Naa Kwarley-Aba Quartey
- Department of Food Science and Technology, Faculty of Biosciences, College of Science, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Albert Fynn Aiduenu
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Accra, Ghana
| | - Blessing Kwabena Gayi
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Accra, Ghana
| | | | - Laud Anthony Basing
- Department of Medical Diagnostics, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Jennifer Afua Afrifa Yamoah
- Animal Health Division, Council for Scientific and Industrial Research-Animal Research Institute, Adenta-Frafraha, Accra, Ghana
| | - Aboagye Kwarteng Dofuor
- Department of Biological Sciences, School of Natural and Environmental Sciences, University of Environment and Sustainable Development, Somanya, Ghana
| | - Joseph Harold Nyarko Osei
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| |
Collapse
|
8
|
Jutzeler KS, Le Clec'h W, Chevalier FD, Anderson TJC. Contribution of parasite and host genotype to immunopathology of schistosome infections. Parasit Vectors 2024; 17:203. [PMID: 38711063 PMCID: PMC11073996 DOI: 10.1186/s13071-024-06286-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/18/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND The role of pathogen genotype in determining disease severity and immunopathology has been studied intensively in microbial pathogens including bacteria, fungi, protozoa and viruses but is poorly understood in parasitic helminths. The medically important blood fluke Schistosoma mansoni is an excellent model system to study the impact of helminth genetic variation on immunopathology. Our laboratory has demonstrated that laboratory schistosome populations differ in sporocyst growth and cercarial production in the intermediate snail host and worm establishment and fecundity in the vertebrate host. Here, we (i) investigate the hypothesis that schistosome genotype plays a significant role in immunopathology and related parasite life history traits in the vertebrate mouse host and (ii) quantify the relative impact of parasite and host genetics on infection outcomes. METHODS We infected BALB/c and C57BL/6 mice with four different laboratory schistosome populations from Africa and the Americas. We quantified disease progression in the vertebrate host by measuring body weight and complete blood count (CBC) with differential over a 12-week infection period. On sacrifice, we assessed parasitological (egg and worm counts, fecundity), immunopathological (organ measurements and histopathology) and immunological (CBC with differential and cytokine profiles) characteristics to determine the impact of parasite and host genetics. RESULTS We found significant variation between parasite populations in worm numbers, fecundity, liver and intestine egg counts, liver and spleen weight, and fibrotic area but not in granuloma size. Variation in organ weight was explained by egg burden and intrinsic parasite factors independent of egg burden. We found significant variation between infected mouse lines in cytokine levels (IFN-γ, TNF-α), eosinophils, lymphocytes and monocyte counts. CONCLUSIONS This study showed that both parasite and host genotype impact the outcome of infection. While host genotype explains most of the variation in immunological traits, parasite genotype explains most of the variation in parasitological traits, and both host and parasite genotypes impact immunopathology outcomes.
Collapse
Affiliation(s)
- Kathrin S Jutzeler
- Host Parasite Interaction Program, Texas Biomedical Research Institute, P.O. Box 760549, San Antonio, TX, 78245, USA.
- UT Health, Microbiology, Immunology & Molecular Genetics, San Antonio, TX, 78229, USA.
| | - Winka Le Clec'h
- Host Parasite Interaction Program, Texas Biomedical Research Institute, P.O. Box 760549, San Antonio, TX, 78245, USA
| | - Frédéric D Chevalier
- Host Parasite Interaction Program, Texas Biomedical Research Institute, P.O. Box 760549, San Antonio, TX, 78245, USA
| | - Timothy J C Anderson
- Disease Intervention and Prevention Program, Texas Biomedical Research Institute, P.O. Box 760549, San Antonio, TX, 78245, USA.
| |
Collapse
|
9
|
Moreira BP, Gava SG, Haeberlein S, Gueye S, Santos ESS, Weber MHW, Abramyan TM, Grevelding CG, Mourão MM, Falcone FH. Identification of potent schistosomicidal compounds predicted as type II-kinase inhibitors against Schistosoma mansoni c-Jun N-terminal kinase SMJNK. FRONTIERS IN PARASITOLOGY 2024; 3:1394407. [PMID: 39817168 PMCID: PMC11732180 DOI: 10.3389/fpara.2024.1394407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 04/10/2024] [Indexed: 01/18/2025]
Abstract
Introduction Schistosomiasis has for many years relied on a single drug, praziquantel (PZQ) for treatment of the disease. Immense efforts have been invested in the discovery of protein kinase (PK) inhibitors; however, given that the majority of PKs are still not targeted by an inhibitor with a useful level of selectivity, there is a compelling need to expand the chemical space available for synthesizing new, potent, and selective PK inhibitors. Small-molecule inhibitors targeting the ATP pocket of the catalytic domain of PKs have the potential to become drugs devoid of (major) side effects, particularly if they bind selectively. This is the case for type II PK inhibitors, which cause PKs to adopt the so-called DFG-out conformation, corresponding to the inactive state of the enzyme. Methods The goal was to perform a virtual screen against the ATP pocket of the inactive JNK protein kinase. After virtually screening millions of compounds, Atomwise provided 85 compounds predicted to target c-Jun N-terminal kinase (JNK) as type II inhibitors. Selected compounds were screened in vitro against larval stage (schistosomula) of S. mansoni using the XTT assay. Adult worms were assessed for motility, attachment, and pairing stability. Active compounds were further analyzed by molecular docking against SmJNK. Results In total, 33 compounds were considered active in at least one of the assays, and two compounds were active in every in vitro screening assay. The two most potent compounds presented strong effects against both life stages of the parasite, and microscopy analysis showed phenotypic alterations on the tegument, in the gonads, and impairment of cell proliferation. Conclusion The approach to screen type II kinase inhibitors resulted in the identification of active compounds that will be further developed against schistosomiasis.
Collapse
Affiliation(s)
- Bernardo P. Moreira
- Institut für Parasitologie, Biomedizinisches Forschungszentrum Seltersberg (BFS), Justus Liebig Universitaet Giessen, Giessen, Germany
| | - Sandra G. Gava
- Grupo de Pesquisa em Helmintologia e Malacologia Médica, Instituto René Rachou, Fundação Oswaldo Cruz – Fiocruz, Belo Horizonte, Brazil
| | - Simone Haeberlein
- Institut für Parasitologie, Biomedizinisches Forschungszentrum Seltersberg (BFS), Justus Liebig Universitaet Giessen, Giessen, Germany
| | - Sophie Gueye
- Polytech Angers, Université d’Angers, Angers, France
| | - Ester S. S. Santos
- Grupo de Pesquisa em Helmintologia e Malacologia Médica, Instituto René Rachou, Fundação Oswaldo Cruz – Fiocruz, Belo Horizonte, Brazil
| | | | | | - Christoph G. Grevelding
- Institut für Parasitologie, Biomedizinisches Forschungszentrum Seltersberg (BFS), Justus Liebig Universitaet Giessen, Giessen, Germany
| | - Marina M. Mourão
- Grupo de Pesquisa em Helmintologia e Malacologia Médica, Instituto René Rachou, Fundação Oswaldo Cruz – Fiocruz, Belo Horizonte, Brazil
| | - Franco H. Falcone
- Institut für Parasitologie, Biomedizinisches Forschungszentrum Seltersberg (BFS), Justus Liebig Universitaet Giessen, Giessen, Germany
| |
Collapse
|
10
|
Villamizar-Monsalve MA, López-Abán J, Vicente B, Peláez R, Muro A. Current drug strategies for the treatment and control of schistosomiasis. Expert Opin Pharmacother 2024; 25:409-420. [PMID: 38511392 DOI: 10.1080/14656566.2024.2333372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 03/18/2024] [Indexed: 03/22/2024]
Abstract
INTRODUCTION Schistosomiasis, one of the current Neglected Tropical Diseases (NTDs) affects over 230 million people globally, with nearly 700 million at risk in more than 74 countries. Praziquantel (PZQ) has served as the primary treatment for the past four decades; however, its effectiveness is limited as it solely eliminates adult worms. In regions where infections are frequent, PZQ exhibits only temporary efficacy and has restricted potential to disrupt the prolonged transmission of the disease. AREAS COVERED A comprehensive exploration using the PubMed database was conducted to review current pharmacotherapy approaches for schistosomiasis. This review also encompasses recent research findings related to potential novel therapeutics and the repurposing of existing drugs. EXPERT OPINION Current schistosoma treatment strategies, primarily relying on PZQ, face challenges like temporary effectiveness and limited impact on disease transmission. Drug repurposing, due to economic constraints, is decisive for NTDs. Despite PZQ's efficacy, its failure to prevent reinfection highlights the need for complementary strategies, especially in regions with persistent environmental foci. Integrating therapies against diverse schistosome stages boosts efficacy and impedes resistance. Uncovering novel agents is essential to address resistance concerns in tackling this neglected tropical disease. Integrated strategies present a comprehensive approach to navigate the complex challenges.
Collapse
Affiliation(s)
- María Alejandra Villamizar-Monsalve
- Infectious and Tropical Diseases Research Group (e-INTRO), Biomedical Research Institute of Salamanca Research Centre for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), Salamanca, Spain
| | - Julio López-Abán
- Infectious and Tropical Diseases Research Group (e-INTRO), Biomedical Research Institute of Salamanca Research Centre for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), Salamanca, Spain
| | - Belén Vicente
- Infectious and Tropical Diseases Research Group (e-INTRO), Biomedical Research Institute of Salamanca Research Centre for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), Salamanca, Spain
| | - Rafael Peláez
- Organic and Pharmaceutical Chemistry Department, Biomedical Research Institute of Salamanca Research Centre for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), Salamanca, Spain
| | - Antonio Muro
- Infectious and Tropical Diseases Research Group (e-INTRO), Biomedical Research Institute of Salamanca Research Centre for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), Salamanca, Spain
| |
Collapse
|
11
|
Rinaldi G, Loukas A, Sotillo J. Trematode Genomics and Proteomics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1454:507-539. [PMID: 39008274 DOI: 10.1007/978-3-031-60121-7_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Trematode infections stand out as one of the frequently overlooked tropical diseases, despite their wide global prevalence and remarkable capacity to parasitize diverse host species and tissues. Furthermore, these parasites hold significant socio-economic, medical, veterinary and agricultural implications. Over the past decades, substantial strides have been taken to bridge the information gap concerning various "omic" tools, such as proteomics and genomics, in this field. In this edition of the book, we highlight recent progress in genomics and proteomics concerning trematodes with a particular focus on the advances made in the past 5 years. Additionally, we present insights into cutting-edge technologies employed in studying trematode biology and shed light on the available resources for exploring the molecular facets of this particular group of parasitic helminths.
Collapse
Affiliation(s)
- Gabriel Rinaldi
- Department of Life Sciences, Aberystwyth University, Aberystwyth, UK
| | - Alex Loukas
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Javier Sotillo
- Laboratorio de Referencia e Investigación en Parasitología, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Spain.
| |
Collapse
|
12
|
LoVerde PT. Schistosomiasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1454:75-105. [PMID: 39008264 DOI: 10.1007/978-3-031-60121-7_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Schistosomiasis is a major cause of morbidity in the world and almost 800 million people worldwide are at risk for schistosomiasis; it is second only to malaria as a major infectious disease. Globally, it is estimated that the disease affects more than 250 million people in 78 countries of the world and is responsible for some 280,000-500,000 deaths each year. The three major schistosomes infecting humans are Schistosoma mansoni, S. japonicum, and S. haematobium. This chapter covers a wide range of aspects of schistosomiasis, including basic biology of the parasites, epidemiology, immunopathology, treatment, control, vaccines, and genomics/proteomics. In this chapter, the reader will understand the significant toll this disease takes in terms of mortality and morbidity. A description of the various life stages of schistosomes is presented, which will be informative for both those unfamiliar with the disease and experienced scientists. Clinical and public health aspects are addressed that cover acute and chronic disease, diagnosis, current treatment regimens and alternative drugs, and schistosomiasis control programs. A brief overview of genomics and proteomics is included that details recent advances in the field that will help scientists investigate the molecular biology of schistosomes. The reader will take away an appreciation for general aspects of schistosomiasis and the current research advances.
Collapse
Affiliation(s)
- Philip T LoVerde
- Department of Biochemistry and Structural Biology, University of Texas Health, San Antonio, TX, USA.
| |
Collapse
|
13
|
Mukherjee A, Kar I, Patra AK. Understanding anthelmintic resistance in livestock using "omics" approaches. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:125439-125463. [PMID: 38015400 DOI: 10.1007/s11356-023-31045-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/08/2023] [Indexed: 11/29/2023]
Abstract
Widespread and improper use of various anthelmintics, genetic, and epidemiological factors has resulted in anthelmintic-resistant (AR) helminth populations in livestock. This is currently quite common globally in different livestock animals including sheep, goats, and cattle to gastrointestinal nematode (GIN) infections. Therefore, the mechanisms underlying AR in parasitic worm species have been the subject of ample research to tackle this challenge. Current and emerging technologies in the disciplines of genomics, transcriptomics, metabolomics, and proteomics in livestock species have advanced the understanding of the intricate molecular AR mechanisms in many major parasites. The technologies have improved the identification of possible biomarkers of resistant parasites, the ability to find actual causative genes, regulatory networks, and pathways of parasites governing the AR development including the dynamics of helminth infection and host-parasite infections. In this review, various "omics"-driven technologies including genome scan, candidate gene, quantitative trait loci, transcriptomic, proteomic, and metabolomic approaches have been described to understand AR of parasites of veterinary importance. Also, challenges and future prospects of these "omics" approaches are also discussed.
Collapse
Affiliation(s)
- Ayan Mukherjee
- Department of Animal Biotechnology, West Bengal University of Animal and Fishery Sciences, Nadia, Mohanpur, West Bengal, India
| | - Indrajit Kar
- Department of Avian Sciences, West Bengal University of Animal and Fishery Sciences, Nadia, Mohanpur, West Bengal, India
| | - Amlan Kumar Patra
- American Institute for Goat Research, Langston University, Oklahoma, 73050, USA.
| |
Collapse
|
14
|
Alwan SN, Taylor AB, Rhodes J, Tidwell M, McHardy SF, LoVerde PT. Oxamniquine derivatives overcome Praziquantel treatment limitations for Schistosomiasis. PLoS Pathog 2023; 19:e1011018. [PMID: 37428793 PMCID: PMC10359000 DOI: 10.1371/journal.ppat.1011018] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 07/20/2023] [Accepted: 05/31/2023] [Indexed: 07/12/2023] Open
Abstract
Human schistosomiasis is a neglected tropical disease caused by Schistosoma mansoni, S. haematobium, and S. japonicum. Praziquantel (PZQ) is the method of choice for treatment. Due to constant selection pressure, there is an urgent need for new therapies for schistosomiasis. Previous treatment of S. mansoni included the use of oxamniquine (OXA), a drug that is activated by a schistosome sulfotransferase (SULT). Guided by data from X-ray crystallography and Schistosoma killing assays more than 350 OXA derivatives were designed, synthesized, and tested. We were able to identify CIDD-0150610 and CIDD-0150303 as potent derivatives in vitro that kill (100%) of all three Schistosoma species at a final concentration of 71.5 μM. We evaluated the efficacy of the best OXA derivates in an in vivo model after treatment with a single dose of 100 mg/kg by oral gavage. The highest rate of worm burden reduction was achieved by CIDD -150303 (81.8%) against S. mansoni, CIDD-0149830 (80.2%) against S. haematobium and CIDD-066790 (86.7%) against S. japonicum. We have also evaluated the ability of the derivatives to kill immature stages since PZQ does not kill immature schistosomes. CIDD-0150303 demonstrated (100%) killing for all life stages at a final concentration of 143 μM in vitro and effective reduction in worm burden in vivo against S. mansoni. To understand how OXA derivatives fit in the SULT binding pocket, X-ray crystal structures of CIDD-0150303 and CIDD-0150610 demonstrate that the SULT active site will accommodate further modifications to our most active compounds as we fine tune them to increase favorable pharmacokinetic properties. Treatment with a single dose of 100 mg/kg by oral gavage with co-dose of PZQ + CIDD-0150303 reduced the worm burden of PZQ resistant parasites in an animal model by 90.8%. Therefore, we conclude that CIDD-0150303, CIDD-0149830 and CIDD-066790 are novel drugs that overcome some of PZQ limitations, and CIDD-0150303 can be used with PZQ in combination therapy.
Collapse
Affiliation(s)
- Sevan N. Alwan
- Departments of Biochemistry and Structural Biology, University of Texas Health at San Antonio; San Antonio, Texas, Unites States of America
| | - Alexander B. Taylor
- Biology Core Facilities, University of Texas Health at San Antonio; San Antonio, Texas, Unites States of America
| | - Jayce Rhodes
- Center for Innovative Drug Discovery, Department of Chemistry, University of Texas at San Antonio; San Antonio, Texas, Unites States of America
| | - Michael Tidwell
- Center for Innovative Drug Discovery, Department of Chemistry, University of Texas at San Antonio; San Antonio, Texas, Unites States of America
| | - Stanton F. McHardy
- Center for Innovative Drug Discovery, Department of Chemistry, University of Texas at San Antonio; San Antonio, Texas, Unites States of America
| | - Philip T. LoVerde
- Departments of Biochemistry and Structural Biology, University of Texas Health at San Antonio; San Antonio, Texas, Unites States of America
| |
Collapse
|
15
|
Padalino G, Coghlan A, Pagliuca G, Forde-Thomas JE, Berriman M, Hoffmann KF. Using ChEMBL to Complement Schistosome Drug Discovery. Pharmaceutics 2023; 15:1359. [PMID: 37242601 PMCID: PMC10220823 DOI: 10.3390/pharmaceutics15051359] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Schistosomiasis is one of the most important neglected tropical diseases. Until an effective vaccine is registered for use, the cornerstone of schistosomiasis control remains chemotherapy with praziquantel. The sustainability of this strategy is at substantial risk due to the possibility of praziquantel insensitive/resistant schistosomes developing. Considerable time and effort could be saved in the schistosome drug discovery pipeline if available functional genomics, bioinformatics, cheminformatics and phenotypic resources are systematically leveraged. Our approach, described here, outlines how schistosome-specific resources/methodologies, coupled to the open-access drug discovery database ChEMBL, can be cooperatively used to accelerate early-stage, schistosome drug discovery efforts. Our process identified seven compounds (fimepinostat, trichostatin A, NVP-BEP800, luminespib, epoxomicin, CGP60474 and staurosporine) with ex vivo anti-schistosomula potencies in the sub-micromolar range. Three of those compounds (epoxomicin, CGP60474 and staurosporine) also demonstrated potent and fast-acting ex vivo effects on adult schistosomes and completely inhibited egg production. ChEMBL toxicity data were also leveraged to provide further support for progressing CGP60474 (as well as luminespib and TAE684) as a novel anti-schistosomal compound. As very few compounds are currently at the advanced stages of the anti-schistosomal pipeline, our approaches highlight a strategy by which new chemical matter can be identified and quickly progressed through preclinical development.
Collapse
Affiliation(s)
- Gilda Padalino
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff CF10 3NB, UK
| | - Avril Coghlan
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK;
| | | | | | - Matthew Berriman
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK;
| | - Karl F. Hoffmann
- The Department of Life Sciences (DLS), Aberystwyth University, Aberystwyth SY23 3DA, UK;
| |
Collapse
|
16
|
Toth K, Alwan S, Khan S, McHardy SF, LoVerde PT, Cameron MD. Addressing the oxamniquine in vitro-in vivo paradox to facilitate a new generation of anti-schistosome treatments. Int J Parasitol Drugs Drug Resist 2023; 21:65-73. [PMID: 36758271 PMCID: PMC9929523 DOI: 10.1016/j.ijpddr.2023.01.003] [Citation(s) in RCA: 2] [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: 11/23/2022] [Revised: 01/09/2023] [Accepted: 01/17/2023] [Indexed: 02/03/2023]
Abstract
The antischistosomal drug oxamniquine, OXA, requires activation by a sulfotransferase within the parasitic worm to enable killing. Examination of the pharmacokinetic/pharmacodynamic (PK/PD) relationship for OXA identified an in vitro-in vivo paradox with the maximal clinical plasma concentrations five-to ten-times lower than the efficacious concentration for in vitro schistosomal killing. The parasite resides in the vasculature between the intestine and the liver, and modeling the PK data to determine portal concentrations fits with in vitro studies and explains the required human dose. In silico models were used to predict murine dosing to recapitulate human conditions for OXA portal concentration and time course. Follow-up PK studies verified in mice that a 50-100 mg/kg oral gavage dose of OXA formulated in acetate buffer recapitulates the 20-40 mg/kg dose common in patients. OXA was rapidly cleared through a combination of metabolism and excretion into bile. OXA absorbance and tissue distribution were similar in wild-type and P-gp efflux transporter knockout mice. The incorporation of in vitro efficacy data and portal concentration was demonstrated for an improved OXA-inspired analog that has been shown to kill S. mansoni, S. haematobium, and S. japonicum, whereas OXA is only effective against S. mansoni. Second-generation OXA analogs should optimize both in vitro killing and physiochemical properties to achieve high portal concentration via rapid oral absorption, facilitated by favorable solubility, permeability, and minimal intestinal metabolism.
Collapse
Affiliation(s)
- Katalin Toth
- Department of Molecular Medicine, UF Scripps Biomedical Research, Jupiter, FL, 33458, United States
| | - Sevan Alwan
- Department of Biochemistry and Structural Biology, UT Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, United States
| | - Susan Khan
- Department of Molecular Medicine, UF Scripps Biomedical Research, Jupiter, FL, 33458, United States
| | - Stanton F McHardy
- Center for Innovative Drug Discovery, University of Texas at San Antonio, Department of Chemistry, One UTSA Circle, San Antonio, TX, 78249, United States
| | - Philip T LoVerde
- Department of Biochemistry and Structural Biology, UT Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, United States
| | - Michael D Cameron
- Department of Molecular Medicine, UF Scripps Biomedical Research, Jupiter, FL, 33458, United States.
| |
Collapse
|
17
|
Thorn CS, Maness RW, Hulke JM, Delmore KE, Criscione CD. Population genomics of helminth parasites. J Helminthol 2023; 97:e29. [PMID: 36927601 DOI: 10.1017/s0022149x23000123] [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] [Indexed: 03/18/2023]
Abstract
Next generation sequencing technologies have facilitated a shift from a few targeted loci in population genetic studies to whole genome approaches. Here, we review the types of questions and inferences regarding the population biology and evolution of parasitic helminths being addressed within the field of population genomics. Topics include parabiome, hybridization, population structure, loci under selection and linkage mapping. We highlight various advances, and note the current trends in the field, particularly a focus on human-related parasites despite the inherent biodiversity of helminth species. We conclude by advocating for a broader application of population genomics to reflect the taxonomic and life history breadth displayed by helminth parasites. As such, our basic knowledge about helminth population biology and evolution would be enhanced while the diversity of helminths in itself would facilitate population genomic comparative studies to address broader ecological and evolutionary concepts.
Collapse
Affiliation(s)
- C S Thorn
- Department of Biology, Texas A&M University, 3258 TAMU, College Station, TX, 77843, USA
| | - R W Maness
- Department of Biology, Texas A&M University, 3258 TAMU, College Station, TX, 77843, USA
| | - J M Hulke
- Department of Biology, Texas A&M University, 3258 TAMU, College Station, TX, 77843, USA
| | - K E Delmore
- Department of Biology, Texas A&M University, 3258 TAMU, College Station, TX, 77843, USA
| | - C D Criscione
- Department of Biology, Texas A&M University, 3258 TAMU, College Station, TX, 77843, USA
| |
Collapse
|
18
|
Beesley NJ, Cwiklinski K, Allen K, Hoyle RC, Spithill TW, La Course EJ, Williams DJL, Paterson S, Hodgkinson JE. A major locus confers triclabendazole resistance in Fasciola hepatica and shows dominant inheritance. PLoS Pathog 2023; 19:e1011081. [PMID: 36701396 PMCID: PMC9904461 DOI: 10.1371/journal.ppat.1011081] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 02/07/2023] [Accepted: 12/22/2022] [Indexed: 01/27/2023] Open
Abstract
Fasciola hepatica infection is responsible for substantial economic losses in livestock worldwide and poses a threat to human health in endemic areas. The mainstay of control in livestock and the only drug licenced for use in humans is triclabendazole (TCBZ). TCBZ resistance has been reported on every continent and threatens effective control of fasciolosis in many parts of the world. To date, understanding the genetic mechanisms underlying TCBZ resistance has been limited to studies of candidate genes, based on assumptions of their role in drug action. Taking an alternative approach, we combined a genetic cross with whole-genome sequencing to localise a ~3.2Mbp locus within the 1.2Gbp F. hepatica genome that confers TCBZ resistance. We validated this locus independently using bulk segregant analysis of F. hepatica populations and showed that it is the target of drug selection in the field. We genotyped individual parasites and tracked segregation and reassortment of SNPs to show that TCBZ resistance exhibits Mendelian inheritance and is conferred by a dominant allele. We defined gene content within this locus to pinpoint genes involved in membrane transport, (e.g. ATP-binding cassette family B, ABCB1), transmembrane signalling and signal transduction (e.g. GTP-Ras-adenylyl cyclase and EGF-like protein), DNA/RNA binding and transcriptional regulation (e.g. SANT/Myb-like DNA-binding domain protein) and drug storage and sequestration (e.g. fatty acid binding protein, FABP) as prime candidates for conferring TCBZ resistance. This study constitutes the first experimental cross and genome-wide approach for any heritable trait in F. hepatica and is key to understanding the evolution of drug resistance in Fasciola spp. to inform deployment of efficacious anthelmintic treatments in the field.
Collapse
Affiliation(s)
- Nicola J Beesley
- Veterinary Parasitology, Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Krystyna Cwiklinski
- Veterinary Parasitology, Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Katherine Allen
- Veterinary Parasitology, Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Rebecca C Hoyle
- Veterinary Parasitology, Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Terry W Spithill
- Department of Animal, Plant and Soil Sciences and Centre for AgriBioscience, La Trobe University, Bundoora, Australia
| | | | - Diana J L Williams
- Veterinary Parasitology, Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Steve Paterson
- Centre for Genomic Research, Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Jane E Hodgkinson
- Veterinary Parasitology, Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
19
|
Mordvinov V, Pakharukova M. Xenobiotic-Metabolizing Enzymes in Trematodes. Biomedicines 2022; 10:biomedicines10123039. [PMID: 36551794 PMCID: PMC9775572 DOI: 10.3390/biomedicines10123039] [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: 10/31/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/26/2022] Open
Abstract
Trematode infections occur worldwide causing considerable deterioration of human health and placing a substantial financial burden on the livestock industry. The hundreds of millions of people afflicted with trematode infections rely entirely on only two drugs (praziquantel and triclabendazole) for treatment. An understanding of anthelmintic biotransformation pathways in parasites should clarify factors that can modulate therapeutic potency of anthelmintics currently in use and may lead to the discovery of synergistic compounds for combination treatments. Despite the pronounced epidemiological significance of trematodes, there is still no adequate understanding of the functionality of their metabolic systems, including xenobiotic-metabolizing enzymes. The review is focused on the structure and functional significance of the xenobiotic-metabolizing system in trematodes. Knowledge in this field can solve practical problems related to the search for new targets for antiparasitic therapy based on a focused action on certain elements of the parasite's metabolic system. Knowledge of the functionality of this system is required to understand the adaptation of the biochemical processes of parasites residing in the host and mechanisms of drug resistance development, as well as to select a promising molecular target for the discovery and development of new anthelmintic drugs.
Collapse
Affiliation(s)
- Viatcheslav Mordvinov
- Laboratory of Molecular Mechanisms of Pathological Processes, Institute of Cytology and Genetics SB RAS, 10 Akad. Lavrentiev Ave., Novosibirsk 630090, Russia
| | - Maria Pakharukova
- Laboratory of Molecular Mechanisms of Pathological Processes, Institute of Cytology and Genetics SB RAS, 10 Akad. Lavrentiev Ave., Novosibirsk 630090, Russia
- Department of Natural Sciences, Novosibirsk State University, 2 Pirogov Str., Novosibirsk 630090, Russia
- Correspondence: ; Tel.: +7-(913)-394-6669
| |
Collapse
|
20
|
Men P, Geng C, Zhang X, Zhang W, Xie L, Feng D, Du S, Wang M, Huang X, Lu X. Biosynthesis mechanism, genome mining and artificial construction of echinocandin O-sulfonation. Metab Eng 2022; 74:160-167. [DOI: 10.1016/j.ymben.2022.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/19/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022]
|
21
|
Nikolakis ZL, Adams RH, Wade KJ, Lund AJ, Carlton EJ, Castoe TA, Pollock DD. Prospects for genomic surveillance for selection in schistosome parasites. FRONTIERS IN EPIDEMIOLOGY 2022; 2:932021. [PMID: 38455290 PMCID: PMC10910990 DOI: 10.3389/fepid.2022.932021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/12/2022] [Indexed: 03/09/2024]
Abstract
Schistosomiasis is a neglected tropical disease caused by multiple parasitic Schistosoma species, and which impacts over 200 million people globally, mainly in low- and middle-income countries. Genomic surveillance to detect evidence for natural selection in schistosome populations represents an emerging and promising approach to identify and interpret schistosome responses to ongoing control efforts or other environmental factors. Here we review how genomic variation is used to detect selection, how these approaches have been applied to schistosomes, and how future studies to detect selection may be improved. We discuss the theory of genomic analyses to detect selection, identify experimental designs for such analyses, and review studies that have applied these approaches to schistosomes. We then consider the biological characteristics of schistosomes that are expected to respond to selection, particularly those that may be impacted by control programs. Examples include drug resistance, host specificity, and life history traits, and we review our current understanding of specific genes that underlie them in schistosomes. We also discuss how inherent features of schistosome reproduction and demography pose substantial challenges for effective identification of these traits and their genomic bases. We conclude by discussing how genomic surveillance for selection should be designed to improve understanding of schistosome biology, and how the parasite changes in response to selection.
Collapse
Affiliation(s)
- Zachary L. Nikolakis
- Department of Biology, University of Texas at Arlington, Arlington, TX, United States
| | - Richard H. Adams
- Department of Biological and Environmental Sciences, Georgia College and State University, Milledgeville, GA, United States
| | - Kristen J. Wade
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO, United States
| | - Andrea J. Lund
- Department of Environmental and Occupational Health, Colorado School of Public Health, University of Colorado, Anschutz, Aurora, CO, United States
| | - Elizabeth J. Carlton
- Department of Environmental and Occupational Health, Colorado School of Public Health, University of Colorado, Anschutz, Aurora, CO, United States
| | - Todd A. Castoe
- Department of Biology, University of Texas at Arlington, Arlington, TX, United States
| | - David D. Pollock
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
22
|
Wit J, Workentine ML, Redman E, Laing R, Stevens L, Cotton JA, Chaudhry U, Ali Q, Andersen EC, Yeaman S, Wasmuth JD, Gilleard JS. Genomic signatures of selection associated with benzimidazole drug treatments in Haemonchus contortus field populations. Int J Parasitol 2022; 52:677-689. [PMID: 36113620 DOI: 10.1016/j.ijpara.2022.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/19/2022] [Accepted: 07/19/2022] [Indexed: 11/22/2022]
Abstract
Genome-wide methods offer a powerful approach to detect signatures of drug selection. However, limited availability of suitable reference genomes and the difficulty of obtaining field populations with well-defined, distinct drug treatment histories mean there is little information on the signatures of selection in parasitic nematodes and on how best to detect them. This study addresses these knowledge gaps by using field populations of Haemonchus contortus with well-defined benzimidazole treatment histories, leveraging a recently completed chromosomal-scale reference genome assembly. We generated a panel of 49,393 genomic markers to genotype 20 individual adult worms from each of four H. contortus populations: two from closed sheep flocks with an approximate 20 year history of frequent benzimidazole treatment, and two populations with a history of little or no treatment. Sampling occurred in the same geographical region to limit genetic differentiation and maximise the detection sensitivity. A clear signature of selection was detected on chromosome I, centred on the isotype-1 β-tubulin gene. Two additional, but weaker, signatures of selection were detected; one near the middle of chromosome I spanning 3.75 Mbp and 259 annotated genes, and one on chromosome II spanning a region of 3.3 Mbp and 206 annotated genes, including the isotype-2 β-tubulin locus. We also assessed how sensitivity was impacted by sequencing depth, worm number, and pooled versus individual worm sequence data. This study provides the first known direct genome-wide evidence for any parasitic nematode, that the isotype-1 β-tubulin gene is quantitatively the single most important benzimidazole resistance locus. It also identified two additional genomic regions that likely contain benzimidazole resistance loci of secondary importance. This study provides an experimental framework to maximise the power of genome-wide approaches to detect signatures of selection driven by anthelmintic drug treatments in field populations of parasitic nematodes.
Collapse
Affiliation(s)
- Janneke Wit
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada; Host-Parasite Interactions (HPI) Program, University of Calgary, Calgary, Alberta, Canada
| | | | - Elizabeth Redman
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Roz Laing
- Institute of Biodiversity Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Garscube Campus, Glasgow, UK
| | - Lewis Stevens
- Tree of Life, Wellcome Sanger Institute, Cambridge, UK
| | - James A Cotton
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, UK
| | - Umer Chaudhry
- University of Edinburgh, Roslin Institute, Easter Bush Veterinary Centre, Roslin, Midlothian, UK
| | - Qasim Ali
- Department of Parasitology FVAS, University of Agriculture, D.I. Khan, Pakistan
| | - Erik C Andersen
- Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Samuel Yeaman
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - James D Wasmuth
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada; Host-Parasite Interactions (HPI) Program, University of Calgary, Calgary, Alberta, Canada
| | - John S Gilleard
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada; Host-Parasite Interactions (HPI) Program, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
23
|
Naidoo P, Mkhize-Kwitshana ZL. Clustered Regularly Interspaced Short Palindromic Repeats/ CRISPR associated protein 9-mediated editing of Schistosoma mansoni genes: Identifying genes for immunologically potent drug and vaccine development. Rev Soc Bras Med Trop 2022; 55:e0131. [PMID: 35976333 PMCID: PMC9405935 DOI: 10.1590/0037-8682-0131-2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 07/08/2022] [Indexed: 12/26/2022] Open
Abstract
Schistosomiasis is a neglected acute and chronic tropical disease caused by intestinal (Schistosoma mansoni and Schistosoma japonicum) and urogenital (Schistosoma haematobium) helminth parasites (blood flukes or digenetic trematodes). It afflicts over 250 million people worldwide, the majority of whom reside in impoverished tropical and subtropical regions in sub-Saharan Africa. Schistosomiasis is the second most common devastating parasitic disease in the world after malaria and causes over 200,000 deaths annually. Currently, there is no effective and approved vaccine available for human use, and treatment strongly relies on praziquantel drug therapy, which is ineffective in killing immature larval schistosomula stages and eggs already lodged in the tissues. The Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR associated protein 9 (CRISPR/Cas9)-mediated gene editing tool is used to deactivate a gene of interest to scrutinize its role in health and disease, and to identify genes for vaccine and drug targeting. The present review aims to summarize the major findings from the current literature reporting the usage of CRISPR/Cas9-mediated gene editing to inactivate genes in S. mansoni (acetylcholinesterase (AChE), T2 ribonuclease omega-1 (ω1), sulfotransferase oxamniquine resistance protein (SULT-OR), and α-N-acetylgalactosaminidase (SmNAGAL)), and freshwater gastropod snails, Biomphalaria glabrata (allograft inflammatory factor (BgAIF)), an obligatory component of the life cycle of S. mansoni, to identify their roles in the pathogenesis of schistosomiasis, and to highlight the importance of such studies in identifying and developing drugs and vaccines with high therapeutic efficacy.
Collapse
Affiliation(s)
- Pragalathan Naidoo
- University of KwaZulu-Natal, College of Health Sciences, Department of Medical Microbiology, Durban, KwaZulu-Natal, South Africa.,South African Medical Research Council (SAMRC), Division of Research Capacity Development, Cape Town, Western Cape, South Africa
| | - Zilungile Lynette Mkhize-Kwitshana
- University of KwaZulu-Natal, College of Health Sciences, Department of Medical Microbiology, Durban, KwaZulu-Natal, South Africa.,South African Medical Research Council (SAMRC), Division of Research Capacity Development, Cape Town, Western Cape, South Africa
| |
Collapse
|
24
|
Rennar GA, Gallinger TL, Mäder P, Lange-Grünweller K, Haeberlein S, Grünweller A, Grevelding CG, Schlitzer M. Disulfiram and dithiocarbamate analogues demonstrate promising antischistosomal effects. Eur J Med Chem 2022; 242:114641. [PMID: 36027862 DOI: 10.1016/j.ejmech.2022.114641] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 07/14/2022] [Accepted: 07/27/2022] [Indexed: 11/03/2022]
Abstract
Schistosomiasis is a neglected tropical disease with more than 200 million new infections per year. It is caused by parasites of the genus Schistosoma and can lead to death if left untreated. Currently, only two drugs are available to combat schistosomiasis: praziquantel and, to a limited extent, oxamniquine. However, the intensive use of these two drugs leads to an increased probability of the emergence of resistance. Thus, the search for new active substances and their targeted development are mandatory. In this study the substance class of "dithiocarbamates" and their potential as antischistosomal agents is highlighted. These compounds are derived from the basic structure of the human aldehyde dehydrogenase inhibitor disulfiram (tetraethylthiuram disulfide, DSF) and its metabolites. Our compounds revealed promising activity (in vitro) against adults of Schistosoma mansoni, such as the reduction of egg production, pairing stability, vitality, and motility. Moreover, tegument damage as well as gut dilatations or even the death of the parasite were observed. We performed detailed structure-activity relationship studies on both sides of the dithiocarbamate core leading to a library of approximately 300 derivatives (116 derivatives shown here). Starting with 100 μm we improved antischistosomal activity down to 25 μm by substitution of the single bonded sulfur atom for example with different benzyl moieties and integration of the two residues on the nitrogen atom into a cyclic structure like piperazine. Its derivatization at the 4-nitrogen with a sulfonyl group or an acyl group led to the most active derivatives of this study which were active at 10 μm. In light of this SAR study, we identified 17 derivatives that significantly reduced motility and induced several other phenotypes at 25 μm, and importantly five of them have antischistosomal activity also at 10 μm. These derivatives were found to be non-cytotoxic in two human cell lines at 100 μm. Therefore, dithiocarbamates seem to be interesting new candidates for further antischistosomal drug development.
Collapse
Affiliation(s)
- Georg A Rennar
- Department of Pharmaceutical Chemistry, Philipps-Universität Marburg, Marbacher, Weg 6, 35032, Marburg, Germany
| | - Tom L Gallinger
- Department of Pharmaceutical Chemistry, Philipps-Universität Marburg, Marbacher, Weg 6, 35032, Marburg, Germany
| | - Patrick Mäder
- Department of Pharmaceutical Chemistry, Philipps-Universität Marburg, Marbacher, Weg 6, 35032, Marburg, Germany
| | - Kerstin Lange-Grünweller
- Department of Pharmaceutical Chemistry, Philipps-Universität Marburg, Marbacher, Weg 6, 35032, Marburg, Germany
| | - Simone Haeberlein
- BFS, Institute of Parasitology, Justus-Liebig-Universität Gießen, Schubertstraße 81, 35392, Gießen, Germany
| | - Arnold Grünweller
- Department of Pharmaceutical Chemistry, Philipps-Universität Marburg, Marbacher, Weg 6, 35032, Marburg, Germany
| | - Christoph G Grevelding
- BFS, Institute of Parasitology, Justus-Liebig-Universität Gießen, Schubertstraße 81, 35392, Gießen, Germany.
| | - Martin Schlitzer
- Department of Pharmaceutical Chemistry, Philipps-Universität Marburg, Marbacher, Weg 6, 35032, Marburg, Germany.
| |
Collapse
|
25
|
Vianney TJ, Berger DJ, Doyle SR, Sankaranarayanan G, Serubanja J, Nakawungu PK, Besigye F, Sanya RE, Holroyd N, Allan F, Webb EL, Elliott AM, Berriman M, Cotton JA. Genome-wide analysis of Schistosoma mansoni reveals limited population structure and possible praziquantel drug selection pressure within Ugandan hot-spot communities. PLoS Negl Trop Dis 2022; 16:e0010188. [PMID: 35981002 PMCID: PMC9426917 DOI: 10.1371/journal.pntd.0010188] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 08/30/2022] [Accepted: 07/05/2022] [Indexed: 12/23/2022] Open
Abstract
Populations within schistosomiasis control areas, especially those in Africa, are recommended to receive regular mass drug administration (MDA) with praziquantel (PZQ) as the main strategy for controlling the disease. The impact of PZQ treatment on schistosome genetics remains poorly understood, and is limited by a lack of high-resolution genetic data on the population structure of parasites within these control areas. We generated whole-genome sequence data from 174 individual miracidia collected from both children and adults from fishing communities on islands in Lake Victoria in Uganda that had received either annual or quarterly MDA with PZQ over four years, including samples collected immediately before and four weeks after treatment. Genome variation within and between samples was characterised and we investigated genomic signatures of natural selection acting on these populations that could be due to PZQ treatment. The parasite population on these islands was more diverse than found in nearby villages on the lake shore. We saw little or no genetic differentiation between villages, or between the groups of villages with different treatment intensity, but slightly higher genetic diversity within the pre-treatment compared to post-treatment parasite populations. We identified classes of genes significantly enriched within regions of the genome with evidence of recent positive selection among post-treatment and intensively treated parasite populations. The differential selection observed in post-treatment and pre-treatment parasite populations could be linked to any reduced susceptibility of parasites to praziquantel treatment.
Collapse
Affiliation(s)
- Tushabe John Vianney
- Parasites and Microbes, Wellcome Sanger Institute, Hinxton, United Kingdom
- Immunomodulation and Vaccines Programme, Medical Research Council/Uganda Virus Research Institute and the London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Duncan J. Berger
- Parasites and Microbes, Wellcome Sanger Institute, Hinxton, United Kingdom
| | - Stephen R. Doyle
- Parasites and Microbes, Wellcome Sanger Institute, Hinxton, United Kingdom
| | | | - Joel Serubanja
- Immunomodulation and Vaccines Programme, Medical Research Council/Uganda Virus Research Institute and the London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Prossy Kabuubi Nakawungu
- Immunomodulation and Vaccines Programme, Medical Research Council/Uganda Virus Research Institute and the London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Fred Besigye
- Immunomodulation and Vaccines Programme, Medical Research Council/Uganda Virus Research Institute and the London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Richard E. Sanya
- Immunomodulation and Vaccines Programme, Medical Research Council/Uganda Virus Research Institute and the London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Health and Systems for Health Unit, African Population and Health Research Center, Nairobi, Kenya
| | - Nancy Holroyd
- Parasites and Microbes, Wellcome Sanger Institute, Hinxton, United Kingdom
| | - Fiona Allan
- Department of Life Sciences, Natural History Museum, London, United Kingdom
| | - Emily L. Webb
- MRC International Statistics and Epidemiology Group, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Alison M. Elliott
- Immunomodulation and Vaccines Programme, Medical Research Council/Uganda Virus Research Institute and the London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Matthew Berriman
- Parasites and Microbes, Wellcome Sanger Institute, Hinxton, United Kingdom
| | - James A. Cotton
- Parasites and Microbes, Wellcome Sanger Institute, Hinxton, United Kingdom
| |
Collapse
|
26
|
Wei TY, Zheng Y, Wan M, Yang S, Tang J, Wu Y, Li J, Chen SX. Analysis of FR901379 Biosynthetic Genes in Coleophoma empetri by Clustered Regularly Interspaced Short Palindromic Repeats/Cas9-Based Genomic Manipulation. ACS Chem Biol 2022; 17:2130-2141. [PMID: 35822391 DOI: 10.1021/acschembio.2c00250] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The compound FR901379, a sulfated echinocandin produced by the filamentous fungus Coleophoma empetri F-11899, is an important intermediate for the synthesis of the antifungal drug micafungin. In this study, we established an efficient clustered regularly interspaced short palindromic repeats/Cas9-based gene editing tool for the industrial production strain C. empetri SIPI1284. With this method, the efficiency of gene mutagenesis in the target locus is up to 84%, which enables the rapid gene disruption for the analysis of FR901379 biosynthetic genes. Next, we verified the putative functional genes of the FR901379 biosynthetic gene cluster via gene disruption and gene complementation in vivo. These core functional genes included the nonribosomal peptide synthetase gene (CEnrps), the fatty-acyl-AMP ligase gene (CEligase) responsible for the formation of the activated form of palmitic acid and its transfer to CEnrps, four nonheme mononuclear iron oxygenase genes (CEoxy1, CEoxy2, CEoxy3, and CEoxy4) responsible for the synthesis of nonproteinogenic amino acids, l-homotyrosine biosynthesis genes (CEhtyA-D), two cytochrome P450 enzyme genes (CEp450-1 and CEp450-2), and a transcription regulator gene (CEhyp). In addition, by screening the whole genome, we identified two unknown genes (CEp450-3 and CEsul) responsible for the sulfonyloxy group of FR901379, which were separated from the core FR901379 biosynthetic cluster. Furthermore, during gene disruptions in the research, we obtained a series of FR901379 analogues and elucidated the relationship between the groups and antifungal activities.
Collapse
Affiliation(s)
- Teng-Yun Wei
- State Key Laboratory of New Drug and Pharmaceutical Process, China State Institute of Pharmaceutical Industry, Shanghai Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Yan Zheng
- State Key Laboratory of New Drug and Pharmaceutical Process, China State Institute of Pharmaceutical Industry, Shanghai Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Miyang Wan
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Songbai Yang
- State Key Laboratory of New Drug and Pharmaceutical Process, China State Institute of Pharmaceutical Industry, Shanghai Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Jiawei Tang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yuanjie Wu
- State Key Laboratory of New Drug and Pharmaceutical Process, China State Institute of Pharmaceutical Industry, Shanghai Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Jiyang Li
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shao-Xin Chen
- State Key Laboratory of New Drug and Pharmaceutical Process, China State Institute of Pharmaceutical Industry, Shanghai Institute of Pharmaceutical Industry, Shanghai 201203, China
| |
Collapse
|
27
|
Guzman MA, Rugel A, Alwan SN, Tarpley R, Taylor AB, Chevalier FD, Wendt GR, Collins JJ, Anderson TJC, McHardy SF, LoVerde PT. Schistosome Sulfotransferases: Mode of Action, Expression and Localization. Pharmaceutics 2022; 14:1416. [PMID: 35890311 PMCID: PMC9323829 DOI: 10.3390/pharmaceutics14071416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 11/24/2022] Open
Abstract
Oxamniquine (OXA) is a prodrug activated by a sulfotransferase (SULT) that was only active against Schistosoma mansoni. We have reengineered OXA to be effective against S. haematobium and S. japonicum. Three derivatives stand out, CIDD-0066790, CIDD-0072229, and CIDD-0149830 as they kill all three major human schistosome species. However, questions remain. Is the OXA mode of action conserved in derivatives? RNA-interference experiments demonstrate that knockdown of the SmSULT, ShSULT, and SjSULT results in resistance to CIDD-0066790. Confirming that the OXA-derivative mode of action is conserved. Next is the level of expression of the schistosome SULTs in each species, as well as changes in SULT expression throughout development in S. mansoni. Using multiple tools, our data show that SmSULT has higher expression compared to ShSULT and SjSULT. Third, is the localization of SULT in the adult, multicellular eucaryotic schistosome species. We utilized fluorescence in situ hybridization and uptake of radiolabeled OXA to determine that multiple cell types throughout the adult schistosome worm express SULT. Thus, we hypothesize the ability of many cells to express the sulfotransferase accounts for the ability of the OXA derivatives to kill adult worms. Our studies demonstrate that the OXA derivatives are able to kill all three human schistosome species and thus will be a useful complement to PZQ.
Collapse
Affiliation(s)
- Meghan A. Guzman
- Department of Microbiology and Immunology, University of Texas Health, San Antonio, TX 78229, USA; (M.A.G.); (A.R.)
- Department of Biochemistry and Structural Biology, University of Texas Health, San Antonio, TX 78229, USA; (S.N.A.); (A.B.T.)
| | - Anastasia Rugel
- Department of Microbiology and Immunology, University of Texas Health, San Antonio, TX 78229, USA; (M.A.G.); (A.R.)
- Department of Biochemistry and Structural Biology, University of Texas Health, San Antonio, TX 78229, USA; (S.N.A.); (A.B.T.)
| | - Sevan N. Alwan
- Department of Biochemistry and Structural Biology, University of Texas Health, San Antonio, TX 78229, USA; (S.N.A.); (A.B.T.)
| | - Reid Tarpley
- Center for Innovative Drug Discovery, University of Texas at San Antonio, San Antonio, TX 78249, USA; (R.T.); (S.F.M.)
| | - Alexander B. Taylor
- Department of Biochemistry and Structural Biology, University of Texas Health, San Antonio, TX 78229, USA; (S.N.A.); (A.B.T.)
| | - Frédéric D. Chevalier
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA;
| | - George R. Wendt
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA; (G.R.W.); (J.J.C.III)
| | - James J. Collins
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA; (G.R.W.); (J.J.C.III)
| | - Timothy J. C. Anderson
- Disease Intervention & Prevention, Texas Biomedical Research Institute, San Antonio, TX 78227, USA;
| | - Stanton F. McHardy
- Center for Innovative Drug Discovery, University of Texas at San Antonio, San Antonio, TX 78249, USA; (R.T.); (S.F.M.)
| | - Philip T. LoVerde
- Department of Biochemistry and Structural Biology, University of Texas Health, San Antonio, TX 78229, USA; (S.N.A.); (A.B.T.)
- Department of Pathology and Laboratory Medicine, University of Texas Health, San Antonio, TX 78229, USA
| |
Collapse
|
28
|
Pedersen LC, Yi M, Pedersen LG, Kaminski AM. From Steroid and Drug Metabolism to Glycobiology, Using Sulfotransferase Structures to Understand and Tailor Function. Drug Metab Dispos 2022; 50:1027-1041. [PMID: 35197313 PMCID: PMC10753775 DOI: 10.1124/dmd.121.000478] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 12/06/2021] [Indexed: 11/22/2022] Open
Abstract
Sulfotransferases are ubiquitous enzymes that transfer a sulfo group from the universal cofactor donor 3'-phosphoadenosine 5'-phosphosulfate to a broad range of acceptor substrates. In humans, the cytosolic sulfotransferases are involved in the sulfation of endogenous compounds such as steroids, neurotransmitters, hormones, and bile acids as well as xenobiotics including drugs, toxins, and environmental chemicals. The Golgi associated membrane-bound sulfotransferases are involved in post-translational modification of macromolecules from glycosaminoglycans to proteins. The sulfation of small molecules can have profound biologic effects on the functionality of the acceptor, including activation, deactivation, or enhanced metabolism and elimination. Sulfation of macromolecules has been shown to regulate a number of physiologic and pathophysiological pathways by enhancing binding affinity to regulatory proteins or binding partners. Over the last 25 years, crystal structures of these enzymes have provided a wealth of information on the mechanisms of this process and the specificity of these enzymes. This review will focus on the general commonalities of the sulfotransferases, from enzyme structure to catalytic mechanism as well as providing examples into how structural information is being used to either design drugs that inhibit sulfotransferases or to modify the enzymes to improve drug synthesis. SIGNIFICANCE STATEMENT: This manuscript honors Dr. Masahiko Negishi's contribution to the understanding of sulfotransferase mechanism, specificity, and roles in biology by analyzing the crystal structures that have been solved over the last 25 years.
Collapse
Affiliation(s)
- Lars C Pedersen
- Genome Integrity and Structural Biology Laboratory (L.C.P., L.G.P., A.M.K.) and Reproductive and Developmental Biology Laboratory (M.Y.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina; and Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (L.G.P.)
| | - MyeongJin Yi
- Genome Integrity and Structural Biology Laboratory (L.C.P., L.G.P., A.M.K.) and Reproductive and Developmental Biology Laboratory (M.Y.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina; and Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (L.G.P.)
| | - Lee G Pedersen
- Genome Integrity and Structural Biology Laboratory (L.C.P., L.G.P., A.M.K.) and Reproductive and Developmental Biology Laboratory (M.Y.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina; and Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (L.G.P.)
| | - Andrea M Kaminski
- Genome Integrity and Structural Biology Laboratory (L.C.P., L.G.P., A.M.K.) and Reproductive and Developmental Biology Laboratory (M.Y.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina; and Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (L.G.P.)
| |
Collapse
|
29
|
Ezebuo FC, Uzochukwu IC. Drug repurposing for schistosomiasis: molecular docking and dynamics investigations. J Biomol Struct Dyn 2022; 40:995-1009. [DOI: 10.1080/07391102.2020.1820382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Fortunatus C. Ezebuo
- Department of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka, Anambra State, Nigeria
| | - Ikemefuna C. Uzochukwu
- Department of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka, Anambra State, Nigeria
| |
Collapse
|
30
|
de Menezes RPB, Viana JDO, Muratov E, Scotti L, Scotti MT. Computer-Assisted Discovery of Alkaloids with Schistosomicidal Activity. Curr Issues Mol Biol 2022; 44:383-408. [PMID: 35723407 PMCID: PMC8929062 DOI: 10.3390/cimb44010028] [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: 12/16/2021] [Revised: 01/08/2022] [Accepted: 01/09/2022] [Indexed: 11/28/2022] Open
Abstract
Schistosomiasis is a chronic parasitic disease caused by trematodes of the genus Schistosoma; it is commonly caused by Schistosoma mansoni, which is transmitted by Bioamphalaria snails. Studies show that more than 200 million people are infected and that more than 90% of them live in Africa. Treatment with praziquantel has the best cost–benefit result on the market. However, hypersensitivity, allergy, and drug resistance are frequently presented after administration. From this perspective, ligand-based and structure-based virtual screening (VS) techniques were combined to select potentially active alkaloids against S. mansoni from an internal dataset (SistematX). A set of molecules with known activity against S. mansoni was selected from the ChEMBL database to create two different models with accuracy greater than 84%, enabling ligand-based VS of the alkaloid bank. Subsequently, structure-based VS was performed through molecular docking using four targets of the parasite. Finally, five consensus hits (i.e., five alkaloids with schistosomicidal potential), were selected. In addition, in silico evaluations of the metabolism, toxicity, and drug-like profile of these five selected alkaloids were carried out. Two of them, namely, 11,12-methylethylenedioxypropoxy and methyl-3-oxo-12-methoxy-n(1)-decarbomethoxy-14,15-didehydrochanofruticosinate, had plausible toxicity, metabolomics, and toxicity profiles. These two alkaloids could serve as starting points for the development of new schistosomicidal compounds based on natural products.
Collapse
Affiliation(s)
- Renata Priscila Barros de Menezes
- Post-Graduate Program in Natural Synthetic Bioactive Products, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil; (R.P.B.d.M.); (J.d.O.V.); (L.S.)
| | - Jéssika de Oliveira Viana
- Post-Graduate Program in Natural Synthetic Bioactive Products, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil; (R.P.B.d.M.); (J.d.O.V.); (L.S.)
| | - Eugene Muratov
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA;
| | - Luciana Scotti
- Post-Graduate Program in Natural Synthetic Bioactive Products, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil; (R.P.B.d.M.); (J.d.O.V.); (L.S.)
| | - Marcus Tullius Scotti
- Post-Graduate Program in Natural Synthetic Bioactive Products, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil; (R.P.B.d.M.); (J.d.O.V.); (L.S.)
- Correspondence: ; Tel.: +55-83-998690415
| |
Collapse
|
31
|
Comparative characterization of microRNAs of Schistosoma japonicum from SCID mice and BALB/c mice: Clues to the regulation of parasite growth and development. Acta Trop 2022; 225:106200. [PMID: 34740636 DOI: 10.1016/j.actatropica.2021.106200] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 10/07/2021] [Accepted: 10/13/2021] [Indexed: 12/17/2022]
Abstract
Schistosomiasis, caused by a parasite with a wide range of mammalian hosts, remains one of the most prevailing parasitic diseases in the world. While numerous studies have reported that the growth and reproduction of schistosomes in immunodeficient mice was significantly retarded, the underlying molecular mechanisms have yet to be revealed. In this study, we comparatively analyzed the microRNA expression of Schistosoma japonicum derived from SCID and BALB/c mice on the 35th day post-infection by high-throughput RNA sequencing as prominent morphological abnormalities had been observed in schistosomes from SCID mice when compared with those from BALB/c mice. The results revealed that more than 72% and 61% of clean reads in the small RNA libraries of female and male schistosomes, respectively, could be mapped to the selected miRs in the miRBase or the sequences of species-specific genomes. Further analysis identified 122 miRNAs using TPM >0.01 as the threshold value, including 75 known and 47 novel miRNAs, 96 of which were commonly expressed across all the four tested schistosome libraries. Comparative analysis of the libraries of schistosomes from SCID and BALB/c mice identified 15 differentially expressed miRNAs (5 up-regulated and 10 down-regulated) among females and 16 among males (9 up-regulated and 7 down-regulated). Integrated analysis of the two sets of differentially expressed miRNAs of female and male worms identified 2 miRNAs (sja-miR-3488 and sja-miR-novel_29) that overlapped between female and male datasets. Prediction of miRNA targets and Gene Ontology (GO) term enrichment analysis of the predicted target genes revealed that these genes were involved in some important biological processes, such as nucleic acid metabolic process, macromolecule modification, and cellular aromatic compound metabolic process. The predicted target genes were further matched to the differentially expressed genes in male and female schistosomes from the above two hosts, obtaining 7 genes that may be responsible for regulating the growth, development and sex maturation of schistosomes. Taken together, this study provides the first identification of differentially expressed miRNAs in schistosomes from SCID and BALB/c mice. These miRNAs and their predicted target mRNAs are probably involved in the regulation of development, growth, and maturation of schistosomes. Therefore, this study expands our understanding of schistosome development regulation and host-parasite relationship, and also provides a valuable set of potential anti-schistosomal targets for prevention and control of schistosomiasis.
Collapse
|
32
|
Morphological and genomic characterisation of the Schistosoma hybrid infecting humans in Europe reveals admixture between Schistosoma haematobium and Schistosoma bovis. PLoS Negl Trop Dis 2021; 15:e0010062. [PMID: 34941866 PMCID: PMC8741037 DOI: 10.1371/journal.pntd.0010062] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 01/07/2022] [Accepted: 12/06/2021] [Indexed: 11/19/2022] Open
Abstract
Schistosomes cause schistosomiasis, the world's second most important parasitic disease after malaria in terms of public health and social-economic impacts. A peculiar feature of these dioecious parasites is their ability to produce viable and fertile hybrid offspring. Originally only present in the tropics, schistosomiasis is now also endemic in southern Europe. Based on the analysis of two genetic markers the European schistosomes had previously been identified as hybrids between the livestock- and the human-infective species Schistosoma bovis and Schistosoma haematobium, respectively. Here, using PacBio long-read sequencing technology we performed genome assembly improvement and annotation of S. bovis, one of the parental species for which no satisfactory genome assembly was available. We then describe the whole genome introgression levels of the hybrid schistosomes, their morphometric parameters (eggs and adult worms) and their compatibility with two European snail strains used as vectors (Bulinus truncatus and Planorbarius metidjensis). Schistosome-snail compatibility is a key parameter for the parasites life cycle progression, and thus the capability of the parasite to establish in a given area. Our results show that this Schistosoma hybrid is strongly introgressed genetically, composed of 77% S. haematobium and 23% S. bovis origin. This genomic admixture suggests an ancient hybridization event and subsequent backcrosses with the human-specific species, S. haematobium, before its introduction in Corsica. We also show that egg morphology (commonly used as a species diagnostic) does not allow for accurate hybrid identification while genetic tests do.
Collapse
|
33
|
Le Clec'h W, Chevalier FD, Mattos ACA, Strickland A, Diaz R, McDew-White M, Rohr CM, Kinung'hi S, Allan F, Webster BL, Webster JP, Emery AM, Rollinson D, Djirmay AG, Al Mashikhi KM, Al Yafae S, Idris MA, Moné H, Mouahid G, LoVerde P, Marchant JS, Anderson TJC. Genetic analysis of praziquantel response in schistosome parasites implicates a transient receptor potential channel. Sci Transl Med 2021; 13:eabj9114. [PMID: 34936381 DOI: 10.1126/scitranslmed.abj9114] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Winka Le Clec'h
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | | | - Ana Carolina A Mattos
- University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | | | - Robbie Diaz
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | | | - Claudia M Rohr
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Safari Kinung'hi
- National Institute for Medical Research, Mwanza, United Republic of Tanzania
| | - Fiona Allan
- London Centre for Neglected Tropical Disease Research (LCNDTR), Imperial College, London, UK.,Wolfson Wellcome Biomedical Laboratories, Natural History Museum, London, UK
| | - Bonnie L Webster
- London Centre for Neglected Tropical Disease Research (LCNDTR), Imperial College, London, UK.,Wolfson Wellcome Biomedical Laboratories, Natural History Museum, London, UK
| | - Joanne P Webster
- London Centre for Neglected Tropical Disease Research (LCNDTR), Imperial College, London, UK.,Centre for Emerging, Endemic and Exotic Diseases (CEEED), Royal Veterinary College, University of London, London, UK
| | - Aidan M Emery
- London Centre for Neglected Tropical Disease Research (LCNDTR), Imperial College, London, UK.,Wolfson Wellcome Biomedical Laboratories, Natural History Museum, London, UK
| | - David Rollinson
- London Centre for Neglected Tropical Disease Research (LCNDTR), Imperial College, London, UK.,Wolfson Wellcome Biomedical Laboratories, Natural History Museum, London, UK
| | - Amadou Garba Djirmay
- Réseau International Schistosomiases Environnemental Aménagement et Lutte (RISEAL), Niamey, Niger.,World Health Organization, Geneva, Switzerland
| | - Khalid M Al Mashikhi
- Directorate General of Health Services, Dhofar Governorate, Salalah, Sultanate of Oman
| | - Salem Al Yafae
- Directorate General of Health Services, Dhofar Governorate, Salalah, Sultanate of Oman
| | | | - Hélène Moné
- Host-Pathogen-Environment Interactions Laboratory, University of Perpignan, Perpignan, France
| | - Gabriel Mouahid
- Host-Pathogen-Environment Interactions Laboratory, University of Perpignan, Perpignan, France
| | - Philip LoVerde
- University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Jonathan S Marchant
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | |
Collapse
|
34
|
Le Clec’h W, Chevalier FD, McDew-White M, Menon V, Arya GA, Anderson TJ. Genetic architecture of transmission stage production and virulence in schistosome parasites. Virulence 2021; 12:1508-1526. [PMID: 34167443 PMCID: PMC8237990 DOI: 10.1080/21505594.2021.1932183] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 05/06/2021] [Accepted: 05/14/2021] [Indexed: 12/30/2022] Open
Abstract
Both theory and experimental data from pathogens suggest that the production of transmission stages should be strongly associated with virulence, but the genetic bases of parasite transmission/virulence traits are poorly understood. The blood fluke Schistosoma mansoni shows extensive variation in numbers of cercariae larvae shed and in their virulence to infected snail hosts, consistent with expected trade-offs between parasite transmission and virulence. We crossed schistosomes from two populations that differ 8-fold in cercarial shedding and in their virulence to Biomphalaria glabrata snail hosts, and determined four-week cercarial shedding profiles in F0 parents, F1 parents and 376 F2 progeny from two independent crosses in inbred snails. Sequencing and linkage analysis revealed that cercarial production is polygenic and controlled by five QTLs (i.e. Quantitative Trait Loci). These QTLs act additively, explaining 28.56% of the phenotypic variation. These results demonstrate that the genetic architecture of key traits relevant to schistosome ecology can be dissected using classical linkage mapping approaches.
Collapse
Affiliation(s)
- Winka Le Clec’h
- Texas Biomedical Research Institute, San Antonio, Texas, USA
| | | | | | - Vinay Menon
- Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Grace-Ann Arya
- Texas Biomedical Research Institute, San Antonio, Texas, USA
| | | |
Collapse
|
35
|
Lombardo FC, Ravaynia PS, Modena MM, Hierlemann A, Keiser J. Evaluation of Human Liver Microtissues for Drug Screening on Schistosoma mansoni Schistosomula. ACS Infect Dis 2021; 7:1894-1900. [PMID: 33105989 DOI: 10.1021/acsinfecdis.0c00614] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Schistosomiasis is a major neglected tropical disease with more than 200 million infections annually. Despite only one drug, praziquantel, being available, the drug pipeline against schistosomiasis is empty, and drug screening tools have limitations. We evaluated the potential of human liver microtissues (hLiMTs) in antischistosomal drug discovery. Because hLiMTs express all human P450 enzymes, they are an excellent tool to evaluate compounds' bioinactivation, bioactivation, and toxicity. To validate the metabolic conversion capacity of hLiMTs, we first quantified (R)- and (S)-praziquantel and the main metabolite trans-OH-praziquantel following incubation with 0.032-50 μM (0.01-15.62 μg/mL) praziquantel for up to 72 h by a validated LC-MS/MS method. We cocultured hLiMTs with newly transformed schistosomula (NTS) and evaluated the antischistosomal activity and cytotoxicity of three prodrugs terfenadine, tamoxifen citrate, and flutamide. HLiMTs converted 300-350 ng (R)-praziquantel within 24 h into trans-OH-praziquantel. We observed changes in the IC50 values for terfenadine, flutamide, and tamoxifen citrate in comparison to the standard NTS assay in vitro. Cytotoxicity was observed at high concentrations of flutamide and tamoxifen citrate. An in vitro platform containing hLiMTs could serve as an advanced drug screening tool for Schistosoma mansoni, providing information on reduced or increased activity and toxicity.
Collapse
Affiliation(s)
- Flavio C. Lombardo
- Swiss Tropical and Public Health Institute, Socinstrasse 57, CH-4002 Basel, Switzerland
- Universität Basel, Petersplatz 1, CH-4001 Basel, Switzerland
| | - Paolo S. Ravaynia
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Mario M. Modena
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Andreas Hierlemann
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Jennifer Keiser
- Swiss Tropical and Public Health Institute, Socinstrasse 57, CH-4002 Basel, Switzerland
- Universität Basel, Petersplatz 1, CH-4001 Basel, Switzerland
| |
Collapse
|
36
|
Button-Simons KA, Kumar S, Carmago N, Haile MT, Jett C, Checkley LA, Kennedy SY, Pinapati RS, Shoue DA, McDew-White M, Li X, Nosten FH, Kappe SH, Anderson TJC, Romero-Severson J, Ferdig MT, Emrich SJ, Vaughan AM, Cheeseman IH. The power and promise of genetic mapping from Plasmodium falciparum crosses utilizing human liver-chimeric mice. Commun Biol 2021; 4:734. [PMID: 34127785 PMCID: PMC8203791 DOI: 10.1038/s42003-021-02210-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 04/30/2021] [Indexed: 12/30/2022] Open
Abstract
Genetic crosses are most powerful for linkage analysis when progeny numbers are high, parental alleles segregate evenly and numbers of inbred progeny are minimized. We previously developed a novel genetic crossing platform for the human malaria parasite Plasmodium falciparum, an obligately sexual, hermaphroditic protozoan, using mice carrying human hepatocytes (the human liver-chimeric FRG NOD huHep mouse) as the vertebrate host. We report on two genetic crosses-(1) an allopatric cross between a laboratory-adapted parasite (NF54) of African origin and a recently patient-derived Asian parasite, and (2) a sympatric cross between two recently patient-derived Asian parasites. We generated 144 unique recombinant clones from the two crosses, doubling the number of unique recombinant progeny generated in the previous 30 years. The allopatric African/Asian cross has minimal levels of inbreeding and extreme segregation distortion, while in the sympatric Asian cross, inbred progeny predominate and parental alleles segregate evenly. Using simulations, we demonstrate that these progeny provide the power to map small-effect mutations and epistatic interactions. The segregation distortion in the allopatric cross slightly erodes power to detect linkage in several genome regions. We greatly increase the power and the precision to map biomedically important traits with these new large progeny panels.
Collapse
Affiliation(s)
- Katrina A Button-Simons
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA.
| | - Sudhir Kumar
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nelly Carmago
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Meseret T Haile
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Catherine Jett
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Lisa A Checkley
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Spencer Y Kennedy
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | | | - Douglas A Shoue
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Marina McDew-White
- Disease Intervention and Prevention Program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Xue Li
- Disease Intervention and Prevention Program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - François H Nosten
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Mae Sot, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine Research Building, University of Oxford Old Road Campus, Oxford, UK
| | - Stefan H Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Timothy J C Anderson
- Disease Intervention and Prevention Program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | - Michael T Ferdig
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | | | - Ashley M Vaughan
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Ian H Cheeseman
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX, USA.
| |
Collapse
|
37
|
LoVerde PT, Alwan SN, Taylor AB, Rhodes J, Chevalier FD, Anderson TJ, McHardy SF. Rational approach to drug discovery for human schistosomiasis. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2021; 16:140-147. [PMID: 34111649 PMCID: PMC8193065 DOI: 10.1016/j.ijpddr.2021.05.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 05/10/2021] [Accepted: 05/13/2021] [Indexed: 11/19/2022]
Abstract
Human schistosomiasis is a debilitating, life-threatening disease affecting more than 229 million people in as many as 78 countries. There is only one drug of choice effective against all three major species of Schistosoma, praziquantel (PZQ). However, as with many monotherapies, evidence for resistance is emerging in the field and can be selected for in the laboratory. Previously used therapies include oxamniquine (OXA), but shortcomings such as drug resistance and affordability resulted in discontinuation. Employing a genetic, biochemical and molecular approach, a sulfotransferase (SULT-OR) was identified as responsible for OXA drug resistance. By crystallizing SmSULT- OR with OXA, the mode of action of OXA was determined. This information allowed a rational approach to novel drug design. Our team approach with schistosome biologists, medicinal chemists, structural biologists and geneticists has enabled us to develop and test novel drug derivatives of OXA to treat this disease. Using an iterative process for drug development, we have successfully identified derivatives that are effective against all three species of the parasite. One derivative CIDD-0149830 kills 100% of all three human schistosome species within 5 days. The goal is to generate a second therapeutic with a different mode of action that can be used in conjunction with praziquantel to overcome the ever-growing threat of resistance and improve efficacy. The ability and need to design, screen, and develop future, affordable therapeutics to treat human schistosomiasis is critical for successful control program outcomes.
Collapse
Affiliation(s)
- Philip T LoVerde
- Departments of Biochemistry and Structural Biology, The University of Texas Health Science Center, San Antonio, TX, USA; Pathology and Laboratory Medicine, The University of Texas Health Science Center, San Antonio, TX, USA.
| | - Sevan N Alwan
- Departments of Biochemistry and Structural Biology, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Alexander B Taylor
- Departments of Biochemistry and Structural Biology, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Jayce Rhodes
- Center for Innovative Drug Discovery, Department of Chemistry, University of Texas at San Antonio, San Antonio, TX, USA
| | - Frédéric D Chevalier
- Program in Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Timothy Jc Anderson
- Program in Disease Intervention and Prevention, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Stanton F McHardy
- Center for Innovative Drug Discovery, Department of Chemistry, University of Texas at San Antonio, San Antonio, TX, USA
| |
Collapse
|
38
|
High-content approaches to anthelmintic drug screening. Trends Parasitol 2021; 37:780-789. [PMID: 34092518 DOI: 10.1016/j.pt.2021.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/03/2021] [Accepted: 05/11/2021] [Indexed: 11/23/2022]
Abstract
Most anthelmintics were discovered through in vivo screens using animal models of infection. Developing in vitro assays for parasitic worms presents several challenges. The lack of in vitro life cycle culture protocols requires harvesting worms from vertebrate hosts or vectors, limiting assay throughput. Once worms are removed from the host environment, established anthelmintics often show no obvious phenotype - raising concerns about the predictive value of many in vitro assays. However, with recent progress in understanding how anthelmintics subvert host-parasite interactions, and breakthroughs in high-content imaging and machine learning, in vitro assays have the potential to discern subtle cryptic parasite phenotypes. These may prove better endpoints than conventional in vitro viability assays.
Collapse
|
39
|
Kannigadu C, N'Da DD. Recent Advances in the Synthesis and Development of Nitroaromatics as Anti-Infective Drugs. Curr Pharm Des 2021; 26:4658-4674. [PMID: 32228417 DOI: 10.2174/1381612826666200331091853] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/28/2020] [Indexed: 12/24/2022]
Abstract
Infectious diseases commonly occur in tropical and sub-tropical countries. The pathogens of such diseases are able to multiply in human hosts, warranting their continual survival. Infections that are commonplace include malaria, chagas, trypanosomiasis, giardiasis, amoebiasis, toxoplasmosis and leishmaniasis. Malaria is known to cause symptoms, such as high fever, chills, nausea and vomiting, whereas chagas disease causes enlarged lymph glands, muscle pain, swelling and chest pain. People suffering from African trypanosomiasis may experience severe headaches, irritability, extreme fatigue and swollen lymph nodes. As an infectious disease progresses, the human host may also experience personality changes and neurologic problems. If left untreated, most of these diseases can lead to death. Parasites, microbes and bacteria are increasingly adapting and generating strains that are resistant to current clinical drugs. Drug resistance creates an urgency for the development of new drugs to treat these infections. Nitro containing drugs, such as chloramphenicol, metronidazole, tinidazole and secnidazole had been banned for use as antiparasitic agents due to their toxicity. However, recent discoveries of nitrocontaining anti-tuberculosis drugs, i.e. delamanid and pretonamid, and the repurposing of flexinidazole for use in combination with eflornithine for the treatment of human trypanosomiasis, have ignited interest in nitroaromatic scaffolds as viable sources of potential anti-infective agents. This review highlights the differences between old and new nitration methodologies. It furthermore offers insights into recent advances in the development of nitroaromatics as anti-infective drugs.
Collapse
Affiliation(s)
- Christina Kannigadu
- Centre of Excellence for Pharmaceutical Sciences (PharmacenTM), North-West University, Potchefstroom, South Africa
| | - David D N'Da
- Centre of Excellence for Pharmaceutical Sciences (PharmacenTM), North-West University, Potchefstroom, South Africa
| |
Collapse
|
40
|
Douglas B, Oyesola O, Cooper MM, Posey A, Tait Wojno E, Giacomin PR, Herbert DR. Immune System Investigation Using Parasitic Helminths. Annu Rev Immunol 2021; 39:639-665. [PMID: 33646858 DOI: 10.1146/annurev-immunol-093019-122827] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Coevolutionary adaptation between humans and helminths has developed a finely tuned balance between host immunity and chronic parasitism due to immunoregulation. Given that these reciprocal forces drive selection, experimental models of helminth infection are ideally suited for discovering how host protective immune responses adapt to the unique tissue niches inhabited by these large metazoan parasites. This review highlights the key discoveries in the immunology of helminth infection made over the last decade, from innate lymphoid cells to the emerging importance of neuroimmune connections. A particular emphasis is placed on the emerging areas within helminth immunology where the most growth is possible, including the advent of genetic manipulation of parasites to study immunology and the use of engineered T cells for therapeutic options. Lastly,we cover the status of human challenge trials with helminths as treatment for autoimmune disease, which taken together, stand to keep the study of parasitic worms at the forefront of immunology for years to come.
Collapse
Affiliation(s)
- Bonnie Douglas
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA; ,
| | - Oyebola Oyesola
- Department of Immunology, University of Washington, Seattle, Washington 98109, USA; ,
| | - Martha M Cooper
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia; ,
| | - Avery Posey
- Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA; .,Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.,Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania 19104, USA
| | - Elia Tait Wojno
- Department of Immunology, University of Washington, Seattle, Washington 98109, USA; ,
| | - Paul R Giacomin
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia; ,
| | - De'Broski R Herbert
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA; ,
| |
Collapse
|
41
|
Sankaranarayanan G, Coghlan A, Driguez P, Lotkowska ME, Sanders M, Holroyd N, Tracey A, Berriman M, Rinaldi G. Large CRISPR-Cas-induced deletions in the oxamniquine resistance locus of the human parasite Schistosoma mansoni. Wellcome Open Res 2021; 5:178. [PMID: 32789192 PMCID: PMC7405262 DOI: 10.12688/wellcomeopenres.16031.2] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2020] [Indexed: 12/18/2022] Open
Abstract
Background. At least 250 million people worldwide suffer from schistosomiasis, caused by Schistosoma worms. Genome sequences for several Schistosoma species are available, including a high-quality annotated reference for Schistosoma mansoni. There is a pressing need to develop a reliable functional toolkit to translate these data into new biological insights and targets for intervention. CRISPR-Cas9 was recently demonstrated for the first time in S. mansoni, to produce somatic mutations in the omega-1 ( ω1) gene. Methods. We employed CRISPR-Cas9 to introduce somatic mutations in a second gene, SULT-OR, a sulfotransferase expressed in the parasitic stages of S. mansoni, in which mutations confer resistance to the drug oxamniquine. A 262-bp PCR product spanning the region targeted by the gRNA against SULT-OR was amplified, and mutations identified in it by high-throughput sequencing. Results. We found that 0.3-2.0% of aligned reads from CRISPR-Cas9-treated adult worms showed deletions spanning the predicted Cas9 cut site, compared to 0.1-0.2% for sporocysts, while deletions were extremely rare in eggs. The most common deletion observed in adults and sporocysts was a 34 bp-deletion directly upstream of the predicted cut site, but rarer deletions reaching as far as 102 bp upstream of the cut site were also detected. The CRISPR-Cas9-induced deletions, if homozygous, are predicted to cause resistance to oxamniquine by producing frameshifts, ablating SULT-OR transcription, or leading to mRNA degradation via the nonsense-mediated mRNA decay pathway. However, no SULT-OR knock down at the mRNA level was observed, presumably because the cells in which CRISPR-Cas9 did induce mutations represented a small fraction of all cells expressing SULT-OR. Conclusions. Further optimisation of CRISPR-Cas protocols for different developmental stages and particular cell types, including germline cells, will contribute to the generation of a homozygous knock-out in any gene of interest, and in particular the SULT-OR gene to derive an oxamniquine-resistant stable transgenic line.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Alan Tracey
- Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
| | | | | |
Collapse
|
42
|
Nikolakis ZL, Hales NR, Perry BW, Schield DR, Timm LE, Liu Y, Zhong B, Kechris KJ, Carlton EJ, Pollock DD, Castoe TA. Patterns of relatedness and genetic diversity inferred from whole genome sequencing of archival blood fluke miracidia (Schistosoma japonicum). PLoS Negl Trop Dis 2021; 15:e0009020. [PMID: 33406094 PMCID: PMC7815185 DOI: 10.1371/journal.pntd.0009020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 01/19/2021] [Accepted: 11/30/2020] [Indexed: 02/05/2023] Open
Abstract
Genomic approaches hold great promise for resolving unanswered questions about transmission patterns and responses to control efforts for schistosomiasis and other neglected tropical diseases. However, the cost of generating genomic data and the challenges associated with obtaining sufficient DNA from individual schistosome larvae (miracidia) from mammalian hosts have limited the application of genomic data for studying schistosomes and other complex macroparasites. Here, we demonstrate the feasibility of utilizing whole genome amplification and sequencing (WGS) to analyze individual archival miracidia. As an example, we sequenced whole genomes of 22 miracidia from 11 human hosts representing two villages in rural Sichuan, China, and used these data to evaluate patterns of relatedness and genetic diversity. We also down-sampled our dataset to test how lower coverage sequencing could increase the cost effectiveness of WGS while maintaining power to accurately infer relatedness. Collectively, our results illustrate that population-level WGS datasets are attainable for individual miracidia and represent a powerful tool for ultimately providing insight into overall genetic diversity, parasite relatedness, and transmission patterns for better design and evaluation of disease control efforts.
Collapse
Affiliation(s)
- Zachary L. Nikolakis
- Department of Biology, University of Texas at Arlington, Arlington, Texas, United States of America
| | - Nicole R. Hales
- Department of Biology, University of Texas at Arlington, Arlington, Texas, United States of America
| | - Blair W. Perry
- Department of Biology, University of Texas at Arlington, Arlington, Texas, United States of America
| | - Drew R. Schield
- Department of Biology, University of Texas at Arlington, Arlington, Texas, United States of America
| | - Laura E. Timm
- Department of Biochemistry & Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Yang Liu
- Institute of Parasitic Disease, Sichuan Center for Disease Control and Prevention, Chengdu, The People’s Republic of China
| | - Bo Zhong
- Institute of Parasitic Disease, Sichuan Center for Disease Control and Prevention, Chengdu, The People’s Republic of China
| | - Katerina J. Kechris
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado, Anschutz, Aurora, Colorado, United States of America
| | - Elizabeth J. Carlton
- Department of Environmental and Occupational Health, Colorado School of Public Health, University of Colorado, Anschutz, Aurora, Colorado, United States of America
| | - David D. Pollock
- Department of Biochemistry & Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Todd A. Castoe
- Department of Biology, University of Texas at Arlington, Arlington, Texas, United States of America
| |
Collapse
|
43
|
Buchter V, Ong YC, Mouvet F, Ladaycia A, Lepeltier E, Rothlisberger U, Keiser J, Gasser G. Multidisciplinary Preclinical Investigations on Three Oxamniquine Analogues as New Drug Candidates for Schistosomiasis**. Chemistry 2020; 26:15232-15241. [DOI: 10.1002/chem.202002856] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/24/2020] [Indexed: 12/29/2022]
Affiliation(s)
- Valentin Buchter
- Swiss Tropical and Public Health Institute Socinstrasse 57, P.O. box 4002 Basel Switzerland
- University of Basel Petersplatz 1, P.O. Box 4001 Basel Switzerland
| | - Yih Ching Ong
- Chimie ParisTech PSL University CNRS, Institute of Chemistry for Life and Health Sciences Laboratory of Inorganic Chemical Biology 75005 Paris France
| | - François Mouvet
- Laboratory of Computational Chemistry and Biochemistry EPFL 1015 Lausanne Switzerland
| | - Abdallah Ladaycia
- MINT, UNIV Angers, INSERM 1066, CNRS 6021 Université Bretagne Loire 4 rue Larrey 49933 Angers Cedex 9 France
| | - Elise Lepeltier
- MINT, UNIV Angers, INSERM 1066, CNRS 6021 Université Bretagne Loire 4 rue Larrey 49933 Angers Cedex 9 France
| | - Ursula Rothlisberger
- Laboratory of Computational Chemistry and Biochemistry EPFL 1015 Lausanne Switzerland
| | - Jennifer Keiser
- Swiss Tropical and Public Health Institute Socinstrasse 57, P.O. box 4002 Basel Switzerland
- University of Basel Petersplatz 1, P.O. Box 4001 Basel Switzerland
| | - Gilles Gasser
- Chimie ParisTech PSL University CNRS, Institute of Chemistry for Life and Health Sciences Laboratory of Inorganic Chemical Biology 75005 Paris France
| |
Collapse
|
44
|
Ezebuo FC, Uzochukwu IC. Schistosomal Sulfotransferase Interaction with Oxamniquine Involves Hybrid Mechanism of Induced-fit and Conformational Selection. Curr Comput Aided Drug Des 2020; 16:451-459. [DOI: 10.2174/1573409915666190708103132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/25/2019] [Accepted: 05/06/2019] [Indexed: 11/22/2022]
Abstract
Background:
Sulfotransferase family comprises key enzymes involved in drug metabolism. Oxamniquine
is a pro-drug converted into its active form by schistosomal sulfotransferase. The conformational
dynamics of side-chain amino acid residues at the binding site of schistosomal sulfotransferase towards
activation of oxamniquine has not received attention.
Objective:
The study investigated the conformational dynamics of binding site residues in free and oxamniquine
bound schistosomal sulfotransferase systems and their contribution to the mechanism of oxamniquine
activation by schistosomal sulfotransferase using molecular dynamics simulations and binding
energy calculations.
Methods:
Schistosomal sulfotransferase was obtained from Protein Data Bank and both the free and oxamniquine
bound forms were subjected to molecular dynamics simulations using GROMACS-4.5.5 after
modeling it’s missing amino acid residues with SWISS-MODEL. Amino acid residues at its binding site
for oxamniquine was determined and used for Principal Component Analysis and calculations of side-chain
dihedrals. In addition, binding energy of the oxamniquine bound system was calculated using g_MMPBSA.
Results:
The results showed that binding site amino acid residues in free and oxamniquine bound sulfotransferase
sampled different conformational space involving several rotameric states. Importantly,
Phe45, Ile145 and Leu241 generated newly induced conformations, whereas Phe41 exhibited shift in equilibrium
of its conformational distribution. In addition, the result showed binding energy of -130.091 ±
8.800 KJ/mol and Phe45 contributed -9.8576 KJ/mol.
Conclusion:
The results showed that schistosomal sulfotransferase binds oxamniquine by relying on hybrid
mechanism of induced fit and conformational selection models. The findings offer new insight into sulfotransferase
engineering and design of new drugs that target sulfotransferase.
Collapse
Affiliation(s)
- Fortunatus C. Ezebuo
- Drug Design and Informatics Group, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, PMB 5025 Awka 420281, Anambra State, Nigeria
| | - Ikemefuna C. Uzochukwu
- Drug Design and Informatics Group, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, PMB 5025 Awka 420281, Anambra State, Nigeria
| |
Collapse
|
45
|
Guzman MA, Rugel AR, Tarpley RS, Alwan SN, Chevalier FD, Kovalskyy DP, Cao X, Holloway SP, Anderson TJC, Taylor AB, McHardy SF, LoVerde PT. An iterative process produces oxamniquine derivatives that kill the major species of schistosomes infecting humans. PLoS Negl Trop Dis 2020; 14:e0008517. [PMID: 32810153 PMCID: PMC7454593 DOI: 10.1371/journal.pntd.0008517] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 08/28/2020] [Accepted: 06/24/2020] [Indexed: 12/15/2022] Open
Abstract
Currently there is only one method of treatment for human schistosomiasis, the drug praziquantel. Strong selective pressure has caused a serious concern for a rise in resistance to praziquantel leading to the necessity for additional pharmaceuticals, with a distinctly different mechanism of action, to be used in combination therapy with praziquantel. Previous treatment of Schistosoma mansoni included the use of oxamniquine (OXA), a prodrug that is enzymatically activated in S. mansoni but is ineffective against S. haematobium and S. japonicum. The oxamniquine activating enzyme was identified as a S. mansoni sulfotransferase (SmSULT-OR). Structural data have allowed for directed drug development in reengineering oxamniquine to be effective against S. haematobium and S. japonicum. Guided by data from X-ray crystallographic studies and Schistosoma worm killing assays on oxamniquine, our structure-based drug design approach produced a robust SAR program that tested over 300 derivatives and identified several new lead compounds with effective worm killing in vitro. Previous studies resulted in the discovery of compound CIDD-0066790, which demonstrated broad-species activity in killing of schistosome species. As these compounds are racemic mixtures, we tested and demonstrate that the R enantiomer CIDD-007229 kills S. mansoni, S. haematobium and S. japonicum better than the parent drug (CIDD-0066790). The search for derivatives that kill better than CIDD-0066790 has resulted in a derivative (CIDD- 149830) that kills 100% of S. mansoni, S. haematobium and S. japonicum adult worms within 7 days. We hypothesize that the difference in activation and thus killing by the derivatives is due to the ability of the derivative to fit in the binding pocket of each sulfotransferase (SmSULT-OR, ShSULT-OR, SjSULT-OR) and to be efficiently sulfated. The purpose of this research is to develop a second drug to be used in conjunction with praziquantel to treat the major human species of Schistosoma. Collectively, our findings show that CIDD-00149830 and CIDD-0072229 are promising novel drugs for the treatment of human schistosomiasis and strongly support further development and in vivo testing.
Collapse
Affiliation(s)
- Meghan A. Guzman
- Departments of Biochemistry and Structural Biology, the University of Texas Health Science Center, San Antonio, Texas, United States of America
- Pathology and Laboratory Medicine, the University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Anastasia R. Rugel
- Departments of Biochemistry and Structural Biology, the University of Texas Health Science Center, San Antonio, Texas, United States of America
- Pathology and Laboratory Medicine, the University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Reid S. Tarpley
- Center for Innovative Drug Discovery, Department of Chemistry, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Sevan N. Alwan
- Departments of Biochemistry and Structural Biology, the University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Frédéric D. Chevalier
- Program in Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Dmytro P. Kovalskyy
- Departments of Biochemistry and Structural Biology, the University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Xiaohang Cao
- Departments of Biochemistry and Structural Biology, the University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Stephen P. Holloway
- Departments of Biochemistry and Structural Biology, the University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Timothy J. C. Anderson
- Program in Disease Intervention and Prevention, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Alexander B. Taylor
- Departments of Biochemistry and Structural Biology, the University of Texas Health Science Center, San Antonio, Texas, United States of America
- X-ray Crystallography Core Laboratory, Institutional Research Cores, University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Stanton F. McHardy
- Center for Innovative Drug Discovery, Department of Chemistry, University of Texas at San Antonio, San Antonio, Texas, United States of America
- * E-mail: (SFM); (PTL)
| | - Philip T. LoVerde
- Departments of Biochemistry and Structural Biology, the University of Texas Health Science Center, San Antonio, Texas, United States of America
- Pathology and Laboratory Medicine, the University of Texas Health Science Center, San Antonio, Texas, United States of America
- * E-mail: (SFM); (PTL)
| |
Collapse
|
46
|
Rugel AR, Guzman MA, Taylor AB, Chevalier FD, Tarpley RS, McHardy SF, Cao X, Holloway SP, Anderson TJC, Hart PJ, LoVerde PT. Why does oxamniquine kill Schistosoma mansoni and not S. haematobium and S. japonicum? Int J Parasitol Drugs Drug Resist 2020; 13:8-15. [PMID: 32315953 PMCID: PMC7167500 DOI: 10.1016/j.ijpddr.2020.04.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/01/2020] [Accepted: 04/01/2020] [Indexed: 12/03/2022]
Abstract
Human schistosomiasis is a disease which globally affects over 229 million people. Three major species affecting humans are Schistosoma mansoni, S. haematobium and S. japonicum. Previous treatment of S. mansoni includes the use of oxamniquine (OXA), a prodrug that is enzymatically activated in S. mansoni but is ineffective against S. haematobium and S. japonicum. The OXA activating enzyme was identified and crystallized, as being a S. mansoni sulfotransferase (SmSULT). S. haematobium and S. japonicum possess homologs of SmSULT (ShSULT and SjSULT) begging the question; why does oxamniquine fail to kill S. haematobium and S. japonicum adult worms? Investigation of the molecular structures of the sulfotransferases indicates that structural differences, specifically in OXA contact residues, do not abrogate OXA binding in the active sites as previously hypothesized. Data presented argue that the ability of SULTs to sulfate and thus activate OXA and its derivatives is linked to the ability of OXA to fit in the binding pocket to allow the transfer of a sulfur group.
Collapse
Affiliation(s)
- Anastasia R Rugel
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA; Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Meghan A Guzman
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA; Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Alexander B Taylor
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA; X-ray Crystallography Core Laboratory, Institutional Research Cores, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Frédéric D Chevalier
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX, 78245, USA
| | - Reid S Tarpley
- Center for Innovative Drug Discovery, Department of Chemistry, University of Texas at San Antonio, San Antonio, TX, 78249, USA
| | - Stanton F McHardy
- Center for Innovative Drug Discovery, Department of Chemistry, University of Texas at San Antonio, San Antonio, TX, 78249, USA
| | - Xiaohang Cao
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Stephen P Holloway
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Timothy J C Anderson
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX, 78245, USA
| | - P John Hart
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA; X-ray Crystallography Core Laboratory, Institutional Research Cores, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA; Department of Veterans Affairs, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
| | - Philip T LoVerde
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA; Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
47
|
Sankaranarayanan G, Coghlan A, Driguez P, Lotkowska ME, Sanders M, Holroyd N, Tracey A, Berriman M, Rinaldi G. Large CRISPR-Cas-induced deletions in the oxamniquine resistance locus of the human parasite Schistosoma mansoni. Wellcome Open Res 2020; 5:178. [PMID: 32789192 PMCID: PMC7405262 DOI: 10.12688/wellcomeopenres.16031.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2020] [Indexed: 12/29/2022] Open
Abstract
Background. At least 250 million people worldwide suffer from schistosomiasis, caused by Schistosoma worms. Genome sequences for several Schistosoma species are available, including a high-quality annotated reference for Schistosoma mansoni. There is a pressing need to develop a reliable functional toolkit to translate these data into new biological insights and targets for intervention. CRISPR-Cas9 was recently demonstrated for the first time in S. mansoni, to produce somatic mutations in the omega-1 ( ω1) gene. Methods. We employed CRISPR-Cas9 to introduce somatic mutations in a second gene, SULT-OR, a sulfotransferase expressed in the parasitic stages of S. mansoni, in which mutations confer resistance to the drug oxamniquine. A 262-bp PCR product spanning the region targeted by the gRNA against SULT-OR was amplified, and mutations identified in it by high-throughput sequencing. Results. We found that 0.3-2.0% of aligned reads from CRISPR-Cas9-treated adult worms showed deletions spanning the predicted Cas9 cut site, compared to 0.1-0.2% for sporocysts, while deletions were extremely rare in eggs. The most common deletion observed in adults and sporocysts was a 34 bp-deletion directly upstream of the predicted cut site, but rarer deletions reaching as far as 102 bp upstream of the cut site were also detected. The CRISPR-Cas9-induced deletions, if homozygous, are predicted to cause resistance to oxamniquine by producing frameshifts, ablating SULT-OR transcription, or leading to mRNA degradation via the nonsense-mediated mRNA decay pathway. However, no SULT-OR knock down at the mRNA level was observed, presumably because the cells in which CRISPR-Cas9 did induce mutations represented a small fraction of all cells expressing SULT-OR. Conclusions. Further optimisation of CRISPR-Cas protocols for different developmental stages and particular cell types, including germline cells, will contribute to the generation of a homozygous knock-out in any gene of interest, and in particular the SULT-OR gene to derive an oxamniquine-resistant stable transgenic line.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Alan Tracey
- Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
| | | | | |
Collapse
|
48
|
Roh YH, Seo J, Kim JY, Kim HU, Mun SJ, Seo JH, Bong KW. Phosphorylcholine-based encoded hydrogel microparticles with enhanced fouling resistance for multiplex immunoassays. Analyst 2020; 145:5482-5490. [PMID: 32588844 DOI: 10.1039/d0an00808g] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Due to the growing interest in multiplex protein detection, encoded hydrogel microparticles have received attention as a possible path to high performance multiplex immunoassays through a combination of high multiplexing capability and enhanced binding kinetics. However, their practical operation in real complex samples is still limited because polyethylene glycol, which is the main component of hydrogel particles, suffers from oxidative damage and relatively high fouling properties in biochemical solutions. Here, we introduce poly(2-methacryloyloxyethyl phosphorylcholine) (PMPC)-based encoded hydrogel microparticles to perform fouling-resistant multiplex immunoassays, where the anti-fouling characteristics are attributed to the zwitterionic PMPC. By applying a newly developed molding lithography technique, viscous PMPCs with low reactivity were successfully incorporated into the hydrogel network while maintaining uniformity and rigidity for use in multiplex immunoassays. Non-specific protein adsorption on the PMPC particles was reduced by about 37.5% compared to that of conventional PEG particles, which leads to better assay sensitivity. We also validate the multiplex capability of the PMPC particles by performing multiplex detection of two target proteins. Furthermore, we verify that the PMPC particles have a 70% enhancement in anti-fouling characteristics compared to PEG particles in human platelet-rich plasma, potentiating a practical immunoassay platform for clinical diagnosis.
Collapse
Affiliation(s)
- Yoon Ho Roh
- Department of Chemical and Biological Engineering, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea.
| | | | | | | | | | | | | |
Collapse
|
49
|
Duguet TB, Glebov A, Hussain A, Kulkarni S, Mochalkin I, Geary TG, Rashid M, Spangenberg T, Ribeiro P. Identification of annotated bioactive molecules that impair motility of the blood fluke Schistosoma mansoni. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2020; 13:73-88. [PMID: 32531750 PMCID: PMC7284125 DOI: 10.1016/j.ijpddr.2020.05.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 01/21/2023]
Abstract
Neglected tropical diseases are of growing worldwide concern and schistosomiasis, caused by parasitic flatworms, continues to be a major threat with more than 200 million people requiring preventive treatment. As praziquantel (PZQ) remains the treatment of choice, an urgent need for alternative treatments motivates research to identify new lead compounds that would complement PZQ by filling the therapeutic gaps associated with this treatment. Because impairing parasite neurotransmission remains a core strategy for control of parasitic helminths, we screened a library of 708 compounds with validated biological activity in humans on the blood fluke Schistosoma mansoni, measuring their effect on the motility on schistosomulae and adult worms. The primary phenotypic screen performed on schistosomulae identified 70 compounds that induced changes in viability and/or motility. Screening different concentrations and incubation times identified molecules with fast onset of activity on both life stages at low concentration (1 μM). To complement this study, similar assays were performed with chemical analogs of the cholinomimetic drug arecoline and the calcilytic molecule NPS-2143, two compounds that rapidly inhibited schistosome motility; 17 arecoline and 302 NPS-2143 analogs were tested to enlarge the pool of schistosomicidal molecules. Finally, validated hit compounds were tested on three functionally-validated neuroregulatory S. mansoni G-protein coupled receptors (GPCRs): Sm5HTR (serotonin-sensitive), SmGPR2 (histamine) and SmD2 (dopamine), revealing NPS-2143 and analogs as potent inhibitors of dopamine/epinine responses on both human and S. mansoni GPCRs. This study highlights the potential for repurposing known human therapeutic agents for potential schistosomicidal effects and expands the list of hits for further progression.
Collapse
Affiliation(s)
- Thomas B Duguet
- Institute of Parasitology, McGill University, Sainte-Anne-de-Bellevue, Quebec, Canada.
| | - Anastasia Glebov
- Institute of Parasitology, McGill University, Sainte-Anne-de-Bellevue, Quebec, Canada
| | - Asimah Hussain
- Institute of Parasitology, McGill University, Sainte-Anne-de-Bellevue, Quebec, Canada
| | | | - Igor Mochalkin
- EMD Serono Research and Development Institute, Billerica, MA, USA
| | - Timothy G Geary
- Institute of Parasitology, McGill University, Sainte-Anne-de-Bellevue, Quebec, Canada
| | - Mohammed Rashid
- Institute of Parasitology, McGill University, Sainte-Anne-de-Bellevue, Quebec, Canada
| | - Thomas Spangenberg
- Global Health Institute of Merck, Ares Trading S.A., a subsidiary of Merck KGaA (Darmstadt, Germany), Eysins, Switzerland.
| | - Paula Ribeiro
- Institute of Parasitology, McGill University, Sainte-Anne-de-Bellevue, Quebec, Canada
| |
Collapse
|
50
|
Dziwornu GA, Attram HD, Gachuhi S, Chibale K. Chemotherapy for human schistosomiasis: how far have we come? What's new? Where do we go from here? RSC Med Chem 2020; 11:455-490. [PMID: 33479649 PMCID: PMC7593896 DOI: 10.1039/d0md00062k] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 03/22/2020] [Indexed: 01/11/2023] Open
Abstract
Globally, schistosomiasis threatens more than 700 million lives, mostly children, in poor localities of tropical and sub-tropical areas with morbidity due to acute and chronic pathological manifestations of the disease. After a century since the first antimonial-based drugs were introduced to treat the disease, anti-schistosomiasis drug development is again at a bottleneck with only one drug, praziquantel, available for treatment purposes. This review focuses on promising chemotypes as potential starting points in a drug discovery effort to meet the urgent need for new schistosomicides.
Collapse
Affiliation(s)
- Godwin Akpeko Dziwornu
- Department of Chemistry , University of Cape Town , Rondebosch 7701 , South Africa . ; Tel: +27 21 6502553
| | - Henrietta Dede Attram
- Department of Chemistry , University of Cape Town , Rondebosch 7701 , South Africa . ; Tel: +27 21 6502553
| | - Samuel Gachuhi
- Department of Chemistry , University of Cape Town , Rondebosch 7701 , South Africa . ; Tel: +27 21 6502553
| | - Kelly Chibale
- Department of Chemistry , University of Cape Town , Rondebosch 7701 , South Africa . ; Tel: +27 21 6502553
- Drug Discovery and Development Centre (H3D) , University of Cape Town , Rondebosch 7701 , South Africa
- Institute of Infectious Disease and Molecular Medicine , University of Cape Town , Rondebosch 7701 , South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit , University of Cape Town , Rondebosch 7701 , South Africa
| |
Collapse
|