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
|
Ouyang J, Han G, Chen J, Hu J, Luo L, Zhang H, Lan C, Lu Q, Gou Y, Gu H, Hu Y, Zhang P, Xu A, Huang S. Identification and characterization of a novel ApeC-containing transmembrane protein family in parasitic flatworms. Int J Biol Macromol 2025; 309:142866. [PMID: 40210028 DOI: 10.1016/j.ijbiomac.2025.142866] [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: 11/24/2024] [Revised: 03/25/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025]
Abstract
The Apextrin C-terminal (ApeC) domain is widely distributed across various animal clades. Several ApeC domains have been identified as carbohydrate-binding domains, contributing to extracellular adhesion and mucosal immunity. However, the functions and evolutionary history of most ApeC-containing protein (ACP) families remain largely unexplored. In this study, we identified 213 ACPs in flatworms (Platyhelminthes), with each species containing one to two such proteins. All flatworm ACPs belong to a unique transmembrane protein family, characterized by a length of 1700-2200 amino acids and a distinctive domain architecture (SP-[Kringle]1-2-[ApeC]7-[EGF]1-4-TM) unlike any found in other phyla. This ACP is conserved across all major parasitic flatworm lineages, including flukes (Trematoda), tapeworms (Cestoda) and monogeneans (Monogenea), despite their diverse morphologies and habitats. Notably, it is also present in one group of free-living flatworms, the planarians (Tricladida), suggesting that this ACP originated in the free-living ancestor of parasitic flatworms. This ACP contains seven consecutive ApeC domains, an unparalleled number among known animal proteins. Five of these ApeC domains are highly divergent, necessitating the definition of a new domain model (ApeC_Pla; Pfam: PF24148) for accurate classification. Structural predictions indicate that these ApeC domains adopt a conserved three β-sheet structure. Furthermore, transcriptomic analysis revealed that flatworm ACPs, along with several important serpins and proteases, are predominantly expressed in parenchymal cells and feeding organs, suggesting that ACPs serve as novel marker genes for parenchymal tissue and may be involved in cell adhesion, oral immunity, and parasite-host interactions. Taken together, our findings indicate that this flatworm ACP represents a promising target for vaccine development and provides key insights into the physiology of the parenchyma, a unique flatworm tissue that functions as a substitute for a body cavity.
Collapse
Affiliation(s)
- Jihua Ouyang
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Guangkun Han
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Jinsong Chen
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Jiaxuan Hu
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Lei Luo
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Hao Zhang
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Chunliu Lan
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Qianyu Lu
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Yin Gou
- Institute of Tropical Bioscience and Biotechnology, Sanya Research Institute, Chinese Academy of Tropical Agricultural Sciences, Sanya 572024, China
| | - Hanjie Gu
- Institute of Tropical Bioscience and Biotechnology, Sanya Research Institute, Chinese Academy of Tropical Agricultural Sciences, Sanya 572024, China
| | - Yonghua Hu
- Institute of Tropical Bioscience and Biotechnology, Sanya Research Institute, Chinese Academy of Tropical Agricultural Sciences, Sanya 572024, China
| | - Peng Zhang
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Anlong Xu
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Shengfeng Huang
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China.
| |
Collapse
|
3
|
Caña-Bozada VH, Dawoud AAZ, Ramos-de la Cruz I, Flores-Méndez LC, Barrera-Redondo J, Briones-Mendoza J, Yañez-Guerra LA. Global analysis of ligand-gated ion channel conservation across Platyhelminthes. Gen Comp Endocrinol 2025; 366:114718. [PMID: 40157577 DOI: 10.1016/j.ygcen.2025.114718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/16/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Ligand-gated ion channels (LGICs) are critical for neurotransmission, mediating responses to neurotransmitters and hormones, and influencing diverse physiological processes. This study identifies and classifies LGICs across Platyhelminthes, with a particular focus on parasitic neodermatans, which impact human and animal health. Using bioinformatics tools, we analyzed LGICs from 41 neodermatan species and expanded our investigation to encompass vertebrates, other invertebrates, and non-bilaterians to trace LGIC evolutionary pathways across Metazoa. We identified 2,269 putative LGICs within neodermatan species, which we classified into the cys-loop, ASIC/Deg/ENaC, iGluR, and P2X families. Our phylogenetic and clustering analyses reveal lineage-specific patterns with distinct evolutionary trajectories for each LGIC family in neodermatans compared to free-living platyhelminths and other taxa. Notably, the ASIC/Deg/ENaC family displayed the greatest degree of neodermatan-specific divergence, while cys-loop and P2X families were more conserved across taxa. To provide insight into their potential physiological roles, we analyzed LGIC expression patterns in Schistosoma mansoni, revealing widespread expression across neuronal and muscle cell types. The distribution of acid-sensing ion channels (ASICs) in both neurons and muscles suggests a role in neuromuscular signalling, while the P2X receptor (Smp_333600) exhibited sex-specific expression, potentially indicating distinct functional roles in males and females. Additionally, several cys-loop acetylcholine and GABA receptors showed differential neuronal and muscle expression, highlighting their likely contributions to cholinergic and inhibitory neurotransmission. These findings underscore the relevance of LGICs in parasite physiology, particularly in neuromuscular and sensory processes, and suggest potential targets for antiparasitic interventions.
Collapse
Affiliation(s)
- Víctor Hugo Caña-Bozada
- Centro de Investigación en Alimentación y Desarrollo, A.C. Unidad Mazatlán en Acuicultura y Manejo Ambiental, Mazatlán 82112 Sinaloa, Mexico; Centro de Investigación para la Salud en América Latina (CISeAL), Pontificia Universidad Católica del Ecuador (PUCE), Quito, Ecuador.
| | - Ahmed A Z Dawoud
- School of biology. University of Southampton, University Road, SO17 1BJ Southampton, UK
| | - Ivana Ramos-de la Cruz
- Centro de Investigación en Alimentación y Desarrollo, A.C. Unidad Mazatlán en Acuicultura y Manejo Ambiental, Mazatlán 82112 Sinaloa, Mexico
| | - Lizeth C Flores-Méndez
- Universidad Autónoma de Occidente, Unidad Regional Mazatlán. Av. del Mar, Tellería, Mazatlán 82100 Sinaloa, Mexico
| | - Josué Barrera-Redondo
- Department of Algal Development and Evolution, Max Planck Institute for Biology Tübingen 72076 Tübingen, Germany
| | - Jesús Briones-Mendoza
- Carrera de Biología, Facultad de Ciencias de la Vida y Tecnologías, Universidad Laica "Eloy Alfaro" de Manabí, Ciudadela Universitaria vía San Mateo, Manta, Ecuador
| | - Luis A Yañez-Guerra
- School of biology. University of Southampton, University Road, SO17 1BJ Southampton, UK; Institute for Life Sciences. University of Southampton, University Road SO17 1BJ Southampton, UK.
| |
Collapse
|
4
|
Henthorn CR, McCusker P, Le Clec’h W, Chevalier FD, Anderson TJ, Zamanian M, Chan JD. Transcriptional phenotype of the anti-parasitic benzodiazepine meclonazepam on the blood fluke Schistosoma mansoni. PLoS Negl Trop Dis 2025; 19:e0012969. [PMID: 40198716 PMCID: PMC12058154 DOI: 10.1371/journal.pntd.0012969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 05/07/2025] [Accepted: 03/09/2025] [Indexed: 04/10/2025] Open
Abstract
There are limited control measures for the disease schistosomiasis, despite the fact that infection with parasitic blood flukes affects hundreds of millions of people worldwide. The current treatment, praziquantel, has been in use since the 1980's and there is a concern that drug resistance may emerge with continued monotherapy. Given the need for additional antischistosomal drugs, we have re-visited an old lead, meclonazepam. In comparison to praziquantel, there has been relatively little work on its antiparasitic mechanism. Recent findings indicate that praziquantel and meclonazepam act through distinct receptors, making benzodiazepines a promising chemical series for further exploration. Previous work has profiled the transcriptional changes evoked by praziquantel treatment. Here, we examine in detail schistosome phenotypes evoked by in vitro and in vivo meclonazepam treatment. These data confirm that meclonazepam causes extensive tegument damage and directly kills parasites, as measured by pro-apoptotic caspase activation. In vivo meclonazepam exposure results in differential expression of many genes that are divergent in parasitic flatworms, as well as several gene products implicated in blood feeding and regulation of hemostasis in other parasites. Many of these transcripts are also differentially expressed with praziquantel exposure, which may reflect a common schistosome response to the two drugs. However, despite these similarities in drug response, praziquantel-resistant parasites retain susceptibility to meclonazepam's schistocidal effects. These data provide new insight into the mechanism of antischistosomal benzodiazepines, resolving similarities and differences with the current frontline therapy, praziquantel.
Collapse
Affiliation(s)
- Clair R. Henthorn
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Paul McCusker
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Institute for Global Food Security, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Winka Le Clec’h
- Disease Intervention & Prevention Program, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Frédéric D. Chevalier
- Disease Intervention & Prevention Program, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Timothy J.C. Anderson
- Disease Intervention and Prevention program, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Mostafa Zamanian
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - John D. Chan
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Global Health Institute, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
5
|
Choi YJ, Rosa BA, Fernandez-Baca MV, Ore RA, Martin J, Ortiz P, Hoban C, Cabada MM, Mitreva M. Independent origins and non-parallel selection signatures of triclabendazole resistance in Fasciola hepatica. Nat Commun 2025; 16:2996. [PMID: 40148292 PMCID: PMC11950404 DOI: 10.1038/s41467-025-57796-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 03/04/2025] [Indexed: 03/29/2025] Open
Abstract
Triclabendazole (TCBZ) is the primary treatment for fascioliasis, a global foodborne zoonosis caused by Fasciola hepatica. Widespread resistance to TCBZ (TCBZ-R) in livestock and a rapid rise in resistant human infections are significant concerns. To understand the genetic basis of TCBZ-R, we sequenced the genomes of 99 TCBZ-sensitive (TCBZ-S) and 210 TCBZ-R adult flukes from 146 bovine livers in Cusco, Peru. We identify genomic regions of high differentiation (FST outliers above the 99.9th percentile) that encod genes involved in the EGFR-PI3K-mTOR-S6K pathway and microtubule function. Transcript expression differences are observed in microtubule-related genes between TCBZ-S and -R flukes, both without drug treatment and in response to treatment. Using only 30 SNPs, it is possible to differentiate between TCBZ-S and -R parasites with ≥75% accuracy. Our outlier loci are distinct from the previously reported TCBZ-R-associated QTLs in the UK, suggesting an independent evolution of resistance alleles. Effective genetics-based TCBZ-R surveillance must consider the heterogeneity of loci under selection across diverse geographical populations.
Collapse
Affiliation(s)
- Young-Jun Choi
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Bruce A Rosa
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Martha V Fernandez-Baca
- Sede Cusco, Instituto de Medicina Tropical "Alexander von Humboldt", Universidad Peruana Cayetano Heredia, Cusco, Peru
| | - Rodrigo A Ore
- Sede Cusco, Instituto de Medicina Tropical "Alexander von Humboldt", Universidad Peruana Cayetano Heredia, Cusco, Peru
| | - John Martin
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Pedro Ortiz
- Laboratorio de Inmunología, Facultad de Ciencias Veterinarias, Universidad Nacional de Cajamarca, Cajamarca, Peru
| | - Cristian Hoban
- Laboratorio de Inmunología, Facultad de Ciencias Veterinarias, Universidad Nacional de Cajamarca, Cajamarca, Peru
| | - Miguel M Cabada
- Sede Cusco, Instituto de Medicina Tropical "Alexander von Humboldt", Universidad Peruana Cayetano Heredia, Cusco, Peru.
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA.
| | - Makedonka Mitreva
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
6
|
Xie Y, Wang X, Cheng S, Liu W, Yi C, You Y, Zhang W, Wang Y, Tang E, Wang J, Hu W. RNAi screening of uncharacterized genes identifies promising druggable targets in Schistosoma japonicum. PLoS Pathog 2025; 21:e1013014. [PMID: 40153463 PMCID: PMC11977999 DOI: 10.1371/journal.ppat.1013014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 04/08/2025] [Accepted: 03/03/2025] [Indexed: 03/30/2025] Open
Abstract
Schistosomiasis affects more than 250 million people worldwide and is one of the neglected tropical diseases. Currently, the treatment of schistosomiasis relies on a single drug-praziquantel-which has led to increasing pressure from drug resistance. Therefore, there is an urgent need to find new treatments. The development of genome sequencing has provided valuable information for understanding the biology of schistosomes. In the genome of Schistosoma japonicum, approximately 11% of the protein-coding sequences are uncharacterized genes (UGs) annotated as "hypothetical protein" or "protein of unknown function." These poorly understood genes have been unjustifiably neglected, although some may be essential for the survival of the parasites and serve as potential drug targets. In this study, we systematically mined the highly expressed UGs in both genders of this parasite throughout key developmental stages in their mammalian host, using our previously published S. japonicum genome and RNA-seq data. By employing in vitro RNA interference (RNAi), we screened 126 UGs that lack homologs in Homo sapiens and identified 8 that are essential for the parasite vitality. We further investigated two UGs, Sjc_0002003 and Sjc_0009272, which resulted in the most severe phenotypes. Fluorescence in situ hybridization demonstrated that both genes were expressed throughout the body without sex bias. Silencing either Sjc_0002003 or Sjc_0009272 reduced the cell proliferation in the body. Furthermore, in vivo RNAi indicated both genes are required for the growth and survival of the parasites in the mammalian host. For Sjc_0002003, we further characterize the underlying molecular cause of the observed phenotype. Through RNA-seq analysis and functional studies, we revealed that silencing Sjc_0002003 reduces the expression of a series of intestinal genes, including Sjc_0007312 (hypothetical protein), Sjc_0008276 (vha-17), Sjc_0002942 (PLA2G15), and Sjc_0003646 (SJCHGC09134 protein), leading to gut dilation. Our work highlights the importance of UGs in schistosomes as promising targets for drug development in the treatment of the schistosomiasis.
Collapse
Affiliation(s)
- Yuxiang Xie
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Xiaoling Wang
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Shaoyun Cheng
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Wanling Liu
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Cun Yi
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Yanmin You
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Wei Zhang
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Yuepeng Wang
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Enlu Tang
- College of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Jipeng Wang
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Wei Hu
- State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
- College of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, China
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Center for Tropical Diseases, National Center for International Research on Tropical Diseases, Shanghai, China
| |
Collapse
|
7
|
Da'Dara AA, Skelly PJ. The enigmatic heptalaminate surface membrane of intravascular schistosomes. Trends Parasitol 2025; 41:177-187. [PMID: 39915200 DOI: 10.1016/j.pt.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/14/2025] [Accepted: 01/14/2025] [Indexed: 03/08/2025]
Abstract
Intravascular schistosomes have an unusual outer, heptalaminate (seven-layered) covering consisting of not one but two lipid bilayers. Here, we present an updated model of the molecular composition of these bilayers in Schistosoma mansoni that places most identified proteins in the outer, and not the inner, membrane. Here, enzymes would have access to their recently described (non-membrane-permeable) substrates. By contrast, nutrient transporter proteins must be in both membranes to facilitate uptake into the worm's inner tissues. Ectoenzyme activities displayed by living worms suggest the presence on their outer surface of several noncanonically extracellular proteins. The advantages of having a double-bilayered covering may relate to impeding host immunological attack and/or to the worm's ability to acquire selected host molecules onto their exterior.
Collapse
Affiliation(s)
- Akram A Da'Dara
- Molecular Helminthology Laboratory, Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, USA
| | - Patrick J Skelly
- Molecular Helminthology Laboratory, Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, USA.
| |
Collapse
|
8
|
Zhao L, Wendt GR, Collins III JJ. A Krüppel-like factor establishes cellular heterogeneity during schistosome tegumental maintenance. PLoS Pathog 2025; 21:e1013002. [PMID: 40153400 PMCID: PMC11978072 DOI: 10.1371/journal.ppat.1013002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 04/08/2025] [Accepted: 02/23/2025] [Indexed: 03/30/2025] Open
Abstract
Schistosomes are blood dwelling parasitic flatworms that can survive in the circulation of their human hosts for decades. These parasites possess a unique syncytial skin-like surface tissue known as the tegument that is thought to be uniquely adapted for survival in the blood by mediating evasion of host defenses. Previous studies have shown that cell bodies within the tegumental syncytium are turned over and perpetually replaced by new tegumental cells derived from a pool of somatic stem cells called neoblasts. Thus, neoblast-driven tegumental homeostasis has been suggested to be a key part of the parasite's strategy for long-term survival in the blood. However, the comprehensive set of molecular programs that control the specification of tegumental cells are not defined. To better understand these programs, we characterized a homolog of a Krüppel-like factor 4 (klf4) transcription factor that was identified in previous single-cell RNA sequencing (scRNAseq) studies to be expressed in a putative tegument related lineage (TRL) of Schistosoma mansoni. Here, using a combination of RNAi, coupled with scRNAseq and bulk RNAseq approaches, we show that klf4 is essential for the maintenance of an entire TRL. Loss of this klf4+ TRL resulted in loss of a subpopulation of molecularly unique tegument cells, without altering the total number of mature tegumental cells. Thus, klf4 is critical for regulating the balance between different cell populations within the tegumental progenitor pool and thereby influences tegumental production dynamics and the fine-tuning of the molecular identity of the mature tegument. Understanding the functions of distinct populations of cells within the tegumental syncytium is expected to provide insights into parasite defense mechanisms and new avenues for combatting the disease these worms cause.
Collapse
Affiliation(s)
- Lu Zhao
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - George R. Wendt
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - James. J. Collins III
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Howard Hughes Medical Institute, UT Southwestern Medical Center, Dallas, Texas, United States of America
| |
Collapse
|
9
|
Abou-El-Naga IF. Receptors for growth and development of Schistosoma mansoni. J Helminthol 2025; 99:e29. [PMID: 39949117 DOI: 10.1017/s0022149x24001020] [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] [Indexed: 05/09/2025]
Abstract
The growth and development of schistosomes are tightly regulated by various receptors throughout their life cycle. Each stage of the parasite inhabits a distinct habitat and responds to different factors that drive its growth and development. With two hosts involved in its life cycle (mammalian and snail), the parasite must go through additional free-living stages to transition between them. Moreover, communication between male and female worms is essential for the maturation of females. The ability of adult schistosomes to survive in human hosts for up to thirty years demonstrates their capacity to efficiently utilize host nutrients for metabolic processes and growth. In Schistosoma mansoni, receptors mediate the utilization of growth factors derived from both the parasite itself and the host. Nuclear receptors, in particular, collaborate with other proteins to regulate the expression of genes essential for various developmental functions. Receptors also play a pivotal role in RNA export, which is crucial for the parasite development. Additionally, neurotransmitter receptors are essential for the growth and development of larval stages. This review aims to elucidate the mechanisms by which these receptors regulate cell proliferation, differentiation, and maturation throughout the parasite life cycle. Understanding these processes could provide insights into the role of receptors in Schistosoma mansoni development and potentially lead to innovative therapeutic strategies to combat human schistosomiasis.
Collapse
Affiliation(s)
- Iman F Abou-El-Naga
- Medical Parasitology Department, Faculty of Medicine, Alexandria University, Egypt
| |
Collapse
|
10
|
Moescheid MF, Lu Z, Soria CD, Quack T, Puckelwaldt O, Holroyd N, Holzaepfel P, Haeberlein S, Rinaldi G, Berriman M, Grevelding CG. The retinoic acid family-like nuclear receptor SmRAR identified by single-cell transcriptomics of ovarian cells controls oocyte differentiation in Schistosoma mansoni. Nucleic Acids Res 2025; 53:gkae1228. [PMID: 39676663 PMCID: PMC11879061 DOI: 10.1093/nar/gkae1228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 10/30/2024] [Accepted: 11/28/2024] [Indexed: 12/17/2024] Open
Abstract
Studies on transcription regulation in platyhelminth development are scarce, especially for parasitic flatworms. Here, we employed single-cell transcriptomics to identify genes involved in reproductive development in the trematode model Schistosoma mansoni. This parasite causes schistosomiasis, a major neglected infectious disease affecting >240 million people worldwide. The pathology of schistosomiasis is closely associated with schistosome eggs deposited in host organs including the liver. Unlike other trematodes, schistosomes exhibit distinct sexes, with egg production reliant on the pairing-dependent maturation of female reproductive organs. Despite this significance, the molecular mechanisms underlying ovary development and oocyte differentiation remain largely unexplored. Utilizing an organ isolation approach for S. mansoni, we extracted ovaries of paired females followed by single-cell RNA sequencing (RNA-seq) with disassociated oocytes. A total of 1967 oocytes expressing 7872 genes passed quality control (QC) filtering. Unsupervised clustering revealed four distinct cell clusters: somatic, germ cells and progeny, intermediate and late germ cells. Among distinct marker genes for each cluster, we identified a hitherto uncharacterized transcription factor of the retinoic acid receptor family, SmRAR. Functional analyses of SmRAR and associated genes like Smmeiob (meiosis-specific, oligonucleotide/oligosaccharide binding motif (OB) domain-containing) demonstrated their pairing-dependent and ovary-preferential expression and their decisive roles in oocyte differentiation of S. mansoni.
Collapse
Affiliation(s)
- Max F Moescheid
- Institute of Parasitology, Justus Liebig University, Schubertstrasse 81, 35392 Giessen, Germany
| | - Zhigang Lu
- Institute of Parasitology, Justus Liebig University, Schubertstrasse 81, 35392 Giessen, Germany
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10, 1SA, UK
| | - Carmen Diaz Soria
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10, 1SA, UK
| | - Thomas Quack
- Institute of Parasitology, Justus Liebig University, Schubertstrasse 81, 35392 Giessen, Germany
| | - Oliver Puckelwaldt
- Institute of Parasitology, Justus Liebig University, Schubertstrasse 81, 35392 Giessen, Germany
| | - Nancy Holroyd
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10, 1SA, UK
| | - Pauline Holzaepfel
- Institute of Parasitology, Justus Liebig University, Schubertstrasse 81, 35392 Giessen, Germany
| | - Simone Haeberlein
- Institute of Parasitology, Justus Liebig University, Schubertstrasse 81, 35392 Giessen, Germany
| | - Gabriel Rinaldi
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10, 1SA, UK
- Department of Life Sciences, Aberystwyth University, Penglais, Aberystwyth, Ceredigion, SY23 3DA, UK
| | - Matthew Berriman
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10, 1SA, UK
- School of Infection and Immunity, College of Medicine, Veterinary and Life Sciences, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
| | - Christoph G Grevelding
- Institute of Parasitology, Justus Liebig University, Schubertstrasse 81, 35392 Giessen, Germany
| |
Collapse
|
11
|
Wu K, Huang S, Zhao Y, Umar A, Chen H, Yu Z, Huang J. Hepatocyte nuclear factor 4 located in different developmental stages of Schistosoma japonicum and involved in important metabolic pathways. Biomed J 2025; 48:100726. [PMID: 38621646 PMCID: PMC12013132 DOI: 10.1016/j.bj.2024.100726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 04/01/2024] [Accepted: 04/08/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND Nuclear receptors (NRs) are vital for regulating gene expression in organisms. Hepatocyte nuclear factor 4 (HNF4), a class of NRs, participates in blood feeding and intestinal maintenance in schistosomes. However, there are limited researches on the molecular and functional characterization of HNF4 in Schistosoma japonicum (S. japonicum). METHODS Highly specific polyclonal antibodies were generated to analyze the expression and tissue localization of S. japonicum HNF4 (SjHNF4). The potential biological functions of SjHNF4 were characterized by transcriptome and pull-down analyses. Subsequently, enrichment analysis was performed to identify the specific signaling pathways linked to SjHNF4. RESULTS The SjHNF4 protein was expressed heterologously and purified successfully. High purity and high potency polyclonal antibodies were further prepared. The expression of SjHNF4 was higher in female compared to male worms at both transcriptional and protein levels. Female worms expressed SjHNF4 in their perithecium, reproductive system, and certain parts of the intestinal tissues. SjHNF4 was also detected in the perithecium of male worms, as well as in the head, body of cercariae, and eggs. Furthermore, our findings highlighted the potential role of SjHNF4 in blood feeding and its interaction with crucial pathways such as glucose metabolism, lipid metabolism, and nucleotide metabolism. CONCLUSIONS This study shed light on the localization of SjHNF4 in different life stages of S. japonicum, particularly associated with the female schistosomes. A strong correlation was observed between SjHNF4 and essential metabolic pathways. These findings laid a solid groundwork for the research on the relationship between NRs and schistosomes.
Collapse
Affiliation(s)
- Kaijuan Wu
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; China-Africa Research Center of Infectious Diseases, Central South University, Changsha, Hunan, China
| | - Shuaiqin Huang
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; China-Africa Research Center of Infectious Diseases, Central South University, Changsha, Hunan, China
| | - Yiming Zhao
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; China-Africa Research Center of Infectious Diseases, Central South University, Changsha, Hunan, China
| | - Abdulrahim Umar
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Hao Chen
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; China-Africa Research Center of Infectious Diseases, Central South University, Changsha, Hunan, China
| | - Zheng Yu
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; China-Africa Research Center of Infectious Diseases, Central South University, Changsha, Hunan, China
| | - Jing Huang
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; China-Africa Research Center of Infectious Diseases, Central South University, Changsha, Hunan, China.
| |
Collapse
|
12
|
Armstrong R, Marks NJ, Geary TG, Harrington J, Selzer PM, Maule AG. Wnt/β-catenin signalling underpins juvenile Fasciola hepatica growth and development. PLoS Pathog 2025; 21:e1012562. [PMID: 39919127 PMCID: PMC11805424 DOI: 10.1371/journal.ppat.1012562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/15/2025] [Indexed: 02/09/2025] Open
Abstract
Infection by the liver fluke, Fasciola hepatica, places a substantial burden on the global agri-food industry and poses a significant threat to human health in endemic regions. Widespread resistance to a limited arsenal of chemotherapeutics, including the frontline flukicide triclabendazole (TCBZ), renders F. hepatica control unsustainable and accentuates the need for novel therapeutic target discovery. A key facet of F. hepatica biology is a population of specialised stem cells which drive growth and development - their dysregulation is hypothesised to represent an appealing avenue for control. The exploitation of this system as a therapeutic target is impeded by a lack of understanding of the molecular mechanisms underpinning F. hepatica growth and development. Wnt signalling pathways govern a myriad of stem cell processes during embryogenesis and drive tumorigenesis in adult tissues in animals. Here, we identify five putative Wnt ligands and five Frizzled receptors in liver fluke transcriptomic datasets and find that Wnt/β-catenin signalling is most active in juveniles, the most pathogenic life stage. FISH-mediated transcript localisation revealed partitioning of the five Wnt ligands, with each displaying a distinct expression pattern, consistent with each Wnt regulating the development of different cell/tissue types. The silencing of each individual Wnt or Frizzled gene yielded significant reductions in juvenile worm growth and, in select cases, blunted the proliferation of neoblast-like cells. Notably, silencing FhCTNNB1, the key effector of the Wnt/β-catenin signal cascade led to aberrant development of the neuromuscular system which ultimately proved lethal - the first report of a lethal RNAi-induced phenotype in F. hepatica. The absence of any discernible phenotypes following the silencing of the inhibitory Wnt/β-catenin destruction complex components is consistent with low destruction complex activity in rapidly developing juvenile worms, corroborates transcriptomic expression profiles and underscores the importance of Wnt signalling as a key molecular driver of growth and development in early-stage juvenile fluke. The putative pharmacological inhibition of Wnt/β-catenin signalling using commercially available inhibitors phenocopied RNAi results and provides impetus for drug repurposing. Taken together, these data functionally and chemically validate the targeting of Wnt signalling as a novel strategy to undermine the pathogenicity of juvenile F. hepatica.
Collapse
Affiliation(s)
- Rebecca Armstrong
- Understanding Health and Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Nikki J. Marks
- Understanding Health and Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Timothy G. Geary
- Understanding Health and Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
- Institute of Parasitology, McGill University, Sainte-Anne-de-Bellevue, Quebec, Canada
| | - John Harrington
- Parasitology, Boehringer Ingelheim Animal Health, Duluth, Georgia, United States of America
| | - Paul M. Selzer
- Parasitology, Boehringer Ingelheim Vetmedica GmbH, Ingelheim am Rhein, Germany
| | - Aaron G. Maule
- Understanding Health and Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| |
Collapse
|
13
|
Dong B, Zhong H, Zhu D, Wu L, Wang J, Li H, Jin Y. Antibody Responses and the Vaccine Efficacy of Recombinant Glycosyltransferase and Nicastrin Against Schistosoma japonicum. Pathogens 2025; 14:70. [PMID: 39861031 PMCID: PMC11768875 DOI: 10.3390/pathogens14010070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Schistosomiasis is a neglected tropical disease and the second most common parasitic disease after malaria. While praziquantel remains the primary treatment, concerns about drug resistance highlight the urgent need for new drugs and effective vaccines to achieve sustainable control. Previous proteomic studies from our group revealed that the expression of Schistosoma japonicum glycosyltransferase and nicastrin as proteins was higher in single-sex males than mated males, suggesting their critical roles in parasite reproduction and their potential as vaccine candidates. In this study, bioinformatic tools were employed to analyze the structural and functional properties of these proteins, including their signal peptide regions, transmembrane domains, tertiary structures, and protein interaction networks. Recombinant forms of glycosyltransferase and nicastrin were expressed and purified, followed by immunization experiments in BALB/c mice. Immunized mice exhibited significantly elevated specific IgG antibody levels after three immunizations compared to adjuvant and PBS controls. Furthermore, immunization with recombinant glycosyltransferase and nicastrin significantly reduced the reproductive capacity of female worms and liver egg burden, though egg hatchability and adult worm survival were unaffected. These findings demonstrate that recombinant glycosyltransferase and nicastrin are immunogenic and reduce female worm fecundity, supporting their potential as vaccine candidates against schistosomiasis.
Collapse
Affiliation(s)
- Bowen Dong
- National Reference Laboratory for Animal Schistosomiasis, Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (B.D.); (H.Z.); (D.Z.); (H.L.)
| | - Haoran Zhong
- National Reference Laboratory for Animal Schistosomiasis, Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (B.D.); (H.Z.); (D.Z.); (H.L.)
| | - Danlin Zhu
- National Reference Laboratory for Animal Schistosomiasis, Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (B.D.); (H.Z.); (D.Z.); (H.L.)
| | - Luobin Wu
- College of Life Sciences, Shanghai Normal University, Shanghai 200234, China;
| | - Jinming Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China;
- Key Laboratory of Veterinary Parasitology of Gansu Province, Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| | - Hao Li
- National Reference Laboratory for Animal Schistosomiasis, Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (B.D.); (H.Z.); (D.Z.); (H.L.)
| | - Yamei Jin
- National Reference Laboratory for Animal Schistosomiasis, Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; (B.D.); (H.Z.); (D.Z.); (H.L.)
| |
Collapse
|
14
|
Yadav P, Simbassa SB, Sloan R, Newmark PA, Lee J. Schistosome esophageal gland factor MEG-8.2 drives host cell lysis and interacts with host immune proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.15.623777. [PMID: 39605737 PMCID: PMC11601278 DOI: 10.1101/2024.11.15.623777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Schistosomes are blood flukes that ingest large amounts of host blood during their intra-mammalian stage. The ingested blood contains leukocytes that can be harmful, yet the parasites survive inside the host for decades, reflecting superb immune evasion mechanisms that remain poorly understood. Our previous work discovered that FoxA, a forkhead transcription factor, drives the production of the esophageal gland, an anterior digestive organ essential for degrading the ingested leukocytes and for in vivo survival. However, a comprehensive molecular makeup of the esophageal gland remains unclear. Importantly, which of the esophageal gland factors are responsible for degrading the ingested leukocytes, their mechanism of action, and how such a function relates to parasite survival and immune evasion remains unknown. Here, we identify additional esophageal gland genes by taking a comparative transcriptomics approach to identify transcripts altered in foxA knockdown adult schistosomes. A targeted RNAi screen coupled with biochemistry reveals that specific domains of the micro-exon gene MEG-8.2, can drive host cell lysis in a concentration-dependent manner. Using pull-down assays coupled with mass spectrometry, we discover that MEG-8.2 interacts with several host membrane and extracellular proteins that play important roles in activating innate and/or adaptive immunity. Together, our findings suggest a dual role for MEG-8.2 in effectively lysing the ingested cells in the esophageal lumen and interacting with specific host proteins to neutralize or suppress the host immunity. These findings lay an important foundation for exploiting esophageal gland factors to treat schistosomiasis.
Collapse
Affiliation(s)
- Pallavi Yadav
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Sabona B. Simbassa
- Microbiology and Infectious Diseases Program, The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030
| | - Ryan Sloan
- Microbiology and Infectious Diseases Program, The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030
| | - Phillip A. Newmark
- Howard Hughes Medical Institute, Morgridge Institute for Research, Department of Integrative Biology, University of Wisconsin–Madison, Madison, WI 53715
| | - Jayhun Lee
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
- Microbiology and Infectious Diseases Program, The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030
| |
Collapse
|
15
|
Henthorn CR, McCusker P, Clec’h WL, Chevalier FD, Anderson TJ, Zamanian M, Chan JD. Transcriptional phenotype of the anti-parasitic benzodiazepine meclonazepam on the blood fluke Schistosoma mansoni. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.29.620505. [PMID: 39554156 PMCID: PMC11565718 DOI: 10.1101/2024.10.29.620505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
There are limited control measures for the disease schistosomiasis, despite the fact that infection with parasitic blood flukes affects hundreds of millions of people worldwide. The current treatment, praziquantel, has been in use since the 1980's and there is a concern that drug resistance may emerge with continued monotherapy. Given the need for additional antischistosomal drugs, we have re-visited an old lead, meclonazepam. In comparison to praziquantel, there has been relatively little work on its antiparasitic mechanism. Recent findings indicate that praziquantel and meclonazepam act through distinct receptors, making benzodiazepines a promising chemical series for further exploration. Previous work has profiled the transcriptional changes evoked by praziquantel treatment. Here, we examine in detail schistosome phenotypes evoked by in vitro and in vivo meclonazepam treatment. These data confirm that meclonazepam causes extensive tegument damage and directly kills parasites, as measured by pro-apoptotic caspase activation. In vivo meclonazepam exposure results in differential expression of many genes that are divergent in parasitic flatworms, as well as several gene products implicated in blood feeding and regulation of hemostasis in other parasites. Many of these transcripts are also differentially expressed with praziquantel exposure, which may reflect a common schistosome response to the two drugs. However, despite these similarities in drug response, praziquantel-resistant parasites retain susceptibility to meclonazepam's schistocidal effects. These data provide new insight into the mechanism of antischistosomal benzodiazepines, resolving similarities and differences with the current frontline therapy, praziquantel.
Collapse
Affiliation(s)
- Clair R. Henthorn
- Department of Pathobiological Sciences, University of Wisconsin - Madison, Madison, WI, USA
| | - Paul McCusker
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
- Microbe and Pathogen Biology, Institute for Global Food Security, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Winka Le Clec’h
- Host-Pathogen Interactions program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Frédéric D. Chevalier
- Host-Pathogen Interactions program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Timothy J.C. Anderson
- Disease Intervention and Prevention program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Mostafa Zamanian
- Department of Pathobiological Sciences, University of Wisconsin - Madison, Madison, WI, USA
| | - John D. Chan
- Department of Pathobiological Sciences, University of Wisconsin - Madison, Madison, WI, USA
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
- Global Health Institute, University of Wisconsin - Madison, Madison, WI, USA
| |
Collapse
|
16
|
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
|
17
|
Gramberg S, Puckelwaldt O, Schmitt T, Lu Z, Haeberlein S. Spatial transcriptomics of a parasitic flatworm provides a molecular map of drug targets and drug resistance genes. Nat Commun 2024; 15:8918. [PMID: 39414795 PMCID: PMC11484910 DOI: 10.1038/s41467-024-53215-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 10/04/2024] [Indexed: 10/18/2024] Open
Abstract
The spatial organization of gene expression dictates tissue functions in multicellular parasites. Here, we present the spatial transcriptome of a parasitic flatworm, the common liver fluke Fasciola hepatica. We identify gene expression profiles and marker genes for eight distinct tissues and validate the latter by in situ hybridization. To demonstrate the power of our spatial atlas, we focus on genes with substantial medical importance, including vaccine candidates (Ly6 proteins) and drug resistance genes (glutathione S-transferases, ABC transporters). Several of these genes exhibit unique expression patterns, indicating tissue-specific biological functions. Notably, the prioritization of tegumental protein kinases identifies a PKCβ, for which small-molecule targeting causes parasite death. Our comprehensive gene expression map provides unprecedented molecular insights into the organ systems of this complex parasitic organism, serving as a valuable tool for both basic and applied research.
Collapse
Affiliation(s)
- Svenja Gramberg
- Institute of Parasitology, Justus Liebig University Giessen, Giessen, Germany
| | - Oliver Puckelwaldt
- Institute of Parasitology, Justus Liebig University Giessen, Giessen, Germany
| | - Tobias Schmitt
- Institute of Parasitology, Justus Liebig University Giessen, Giessen, Germany
| | - Zhigang Lu
- Institute of Food Science and Biotechnology, University of Hohenheim, Stuttgart, Germany
| | - Simone Haeberlein
- Institute of Parasitology, Justus Liebig University Giessen, Giessen, Germany.
| |
Collapse
|
18
|
Chai C, Gibson J, Li P, Pampari A, Patel A, Kundaje A, Wang B. Flexible use of conserved motif vocabularies constrains genome access in cell type evolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.03.611027. [PMID: 39282369 PMCID: PMC11398382 DOI: 10.1101/2024.09.03.611027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Cell types evolve into a hierarchy with related types grouped into families. How cell type diversification is constrained by the stable separation between families over vast evolutionary times remains unknown. Here, integrating single-nucleus multiomic sequencing and deep learning, we show that hundreds of sequence features (motifs) divide into distinct sets associated with accessible genomes of specific cell type families. This division is conserved across highly divergent, early-branching animals including flatworms and cnidarians. While specific interactions between motifs delineate cell type relationships within families, surprisingly, these interactions are not conserved between species. Consistently, while deep learning models trained on one species can predict accessibility of other species' sequences, their predictions frequently rely on distinct, but synonymous, motif combinations. We propose that long-term stability of cell type families is maintained through genome access specified by conserved motif sets, or 'vocabularies', whereas cell types diversify through flexible use of motifs within each set.
Collapse
Affiliation(s)
- Chew Chai
- Department of Bioengineering, Stanford University, Stanford, USA
| | - Jesse Gibson
- Department of Bioengineering, Stanford University, Stanford, USA
| | - Pengyang Li
- Department of Bioengineering, Stanford University, Stanford, USA
| | - Anusri Pampari
- Department of Computer Science, Stanford University, Stanford, USA
| | - Aman Patel
- Department of Computer Science, Stanford University, Stanford, USA
| | - Anshul Kundaje
- Department of Computer Science, Stanford University, Stanford, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, USA
| | - Bo Wang
- Department of Bioengineering, Stanford University, Stanford, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, USA
| |
Collapse
|
19
|
Attenborough T, Rawlinson KA, Diaz Soria CL, Ambridge K, Sankaranarayanan G, Graham J, Cotton JA, Doyle SR, Rinaldi G, Berriman M. A single-cell atlas of the miracidium larva of Schistosoma mansoni reveals cell types, developmental pathways, and tissue architecture. eLife 2024; 13:RP95628. [PMID: 39190022 PMCID: PMC11349301 DOI: 10.7554/elife.95628] [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: 08/28/2024] Open
Abstract
Schistosoma mansoni is a parasitic flatworm that causes the major neglected tropical disease schistosomiasis. The miracidium is the first larval stage of the life cycle. It swims and infects a freshwater snail, transforms into a mother sporocyst, where its stem cells generate daughter sporocysts that give rise to human-infective cercariae larvae. To understand the miracidium at cellular and molecular levels, we created a whole-body atlas of its ~365 cells. Single-cell RNA sequencing identified 19 transcriptionally distinct cell clusters. In situ hybridisation of tissue-specific genes revealed that 93% of the cells in the larva are somatic (57% neural, 19% muscle, 13% epidermal or tegument, 2% parenchyma, and 2% protonephridia) and 7% are stem. Whereas neurons represent the most diverse somatic cell types, trajectory analysis of the two main stem cell populations indicates that one of them is the origin of the tegument lineage and the other likely contains pluripotent cells. Furthermore, unlike the somatic cells, each of these stem populations shows sex-biased transcriptional signatures suggesting a cell-type-specific gene dosage compensation for sex chromosome-linked loci. The miracidium represents a simple developmental stage with which to gain a fundamental understanding of the molecular biology and spatial architecture of schistosome cells.
Collapse
Affiliation(s)
- Teresa Attenborough
- Wellcome Sanger Institute, Wellcome Genome CampusHinxtonUnited Kingdom
- School of Infection and Immunity, College of Medical, Veterinary & Life Sciences, University of GlasgowGlasgowUnited Kingdom
| | - Kate A Rawlinson
- Wellcome Sanger Institute, Wellcome Genome CampusHinxtonUnited Kingdom
- Josephine Bay Paul Center, Marine Biological LaboratoryWoods HoleUnited States
| | | | - Kirsty Ambridge
- Wellcome Sanger Institute, Wellcome Genome CampusHinxtonUnited Kingdom
| | | | - Jennie Graham
- Wellcome Sanger Institute, Wellcome Genome CampusHinxtonUnited Kingdom
| | - James A Cotton
- Wellcome Sanger Institute, Wellcome Genome CampusHinxtonUnited Kingdom
- School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary & Life Sciences, University of GlasgowGlasgowUnited Kingdom
| | - Stephen R Doyle
- Wellcome Sanger Institute, Wellcome Genome CampusHinxtonUnited Kingdom
| | - Gabriel Rinaldi
- Wellcome Sanger Institute, Wellcome Genome CampusHinxtonUnited Kingdom
- Department of Life Sciences, Aberystwyth UniversityAberystwythUnited Kingdom
| | - Matthew Berriman
- Wellcome Sanger Institute, Wellcome Genome CampusHinxtonUnited Kingdom
- School of Infection and Immunity, College of Medical, Veterinary & Life Sciences, University of GlasgowGlasgowUnited Kingdom
| |
Collapse
|
20
|
Zhao L, Wendt GR, Collins JJ. A Krüppel-like factor establishes cellular heterogeneity during schistosome tegumental maintenance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.12.603265. [PMID: 39026857 PMCID: PMC11257591 DOI: 10.1101/2024.07.12.603265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Schistosomes are blood-dwelling parasitic flatworms that rely on a syncytial surface coat, known as the tegument, for long-term survival and immune evasion in the blood of their human hosts. Previous studies have shown that cells within the tegumental syncytium are perpetually turned over and renewed by somatic stem cells called neoblasts. Yet, little is known about this renewal process on a molecular level. Here, we characterized a Krüppel-like factor 4 (klf4) using a combination of bulk and single cell RNAseq approaches and demonstrate that klf4 is essential for the maintenance of a specific tegumental lineage, resulting in the loss of a subpopulation of molecularly-unique tegument cells. Thus, klf4 is critical for maintaining the balance between different tegumental progenitor pools, thereby fine-tuning the molecular composition of the mature tegument. Understanding these distinct tegumental cell populations is expected to provide insights into parasite defense mechanisms and suggest new avenues for therapeutics.
Collapse
Affiliation(s)
- Lu Zhao
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX
| | - George R. Wendt
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX
| | - James. J. Collins
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
21
|
Konečný L, Peterková K. Unveiling the peptidases of parasites from the office chair - The endothelin-converting enzyme case study. ADVANCES IN PARASITOLOGY 2024; 126:1-52. [PMID: 39448189 DOI: 10.1016/bs.apar.2024.05.003] [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: 10/26/2024]
Abstract
The emergence of high-throughput methodologies such as next-generation sequencing and proteomics has necessitated significant advancements in biological databases and bioinformatic tools, therefore reshaping the landscape of research into parasitic peptidases. In this review we outline the development of these resources along the -omics technologies and their transformative impact on the field. Apart from extensive summary of general and specific databases and tools, we provide a general pipeline on how to use these resources effectively to identify candidate peptidases from these large datasets and how to gain as much information about them as possible without leaving the office chair. This pipeline is then applied in an illustrative case study on the endothelin-converting enzyme 1 homologue from Schistosoma mansoni and attempts to highlight the contemporary capabilities of bioinformatics. The case study demonstrate how such approach can aid to hypothesize enzyme functions and interactions through computational analysis alone effectively and emphasizes how such virtual investigations can guide and optimize subsequent wet lab experiments therefore potentially saving precious time and resources. Finally, by showing what can be achieved without traditional wet laboratory methods, this review provides a compelling narrative on the use of bioinformatics to bridge the gap between big data and practical research applications, highlighting the key role of these technologies in furthering our understanding of parasitic diseases.
Collapse
Affiliation(s)
- Lukáš Konečný
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czechia; Department of Ecology, Centre of Infectious Animal Diseases, Faculty of Environmental Sciences, Czech University of Life Sciences, Prague, Czechia.
| | - Kristýna Peterková
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czechia
| |
Collapse
|
22
|
Tian Y, Cheng Z, Ge D, Xu Z, Wang H, Li X, Tian H, Liu F, Luo D, Wang Y. ROS are required for the germinative cell proliferation and metacestode larval growth of Echinococcus multilocularis. Front Microbiol 2024; 15:1410504. [PMID: 38912347 PMCID: PMC11190091 DOI: 10.3389/fmicb.2024.1410504] [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: 04/01/2024] [Accepted: 05/24/2024] [Indexed: 06/25/2024] Open
Abstract
The potentially lethal zoonotic disease alveolar echinococcosis (AE) is caused by the metacestode larval stages of the tapeworm Echinococcus multilocularis. Metacestode growth and proliferation occurs within the inner organs of mammalian hosts, which is associated with complex molecular parasite-host interactions. The host has developed various ways to resist a parasitic infection, and the production of reactive oxygen species (ROS) is one of the most important strategies. Here, we found that scavenging of ROS reduced metacestode larval growth and germinative cell proliferation in in vivo models. Furthermore, using in vitro-cultured metacestode vesicles, we found that increased ROS levels enhanced metacestode growth and germinative cell proliferation, which was achieved by positively activating the ROS-EmERK-EmHIF1α axis. These results indicate that, beside its capacity to damage the parasite, ROS also play critical roles in metacestode growth and germinative cell proliferation. This study suggests that the effects of ROS on parasite may be bidirectional during AE infection, reflecting the parasite's adaptation to the oxidative stress microenvironment.
Collapse
Affiliation(s)
- Ye Tian
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zhe Cheng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Defeng Ge
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zhijian Xu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Huijuan Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xiazhen Li
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Huimin Tian
- School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Fan Liu
- School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Damin Luo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yanhai Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Parasitology Research Laboratory, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
23
|
Wendt G, Collins JJ. Horizontal gene transfer of a functional cki homolog in the human pathogen Schistosoma mansoni. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.27.596073. [PMID: 38853947 PMCID: PMC11160599 DOI: 10.1101/2024.05.27.596073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Schistosomes are parasitic flatworms responsible for the neglected tropical disease schistosomiasis, causing devastating morbidity and mortality in the developing world. The parasites are protected by a skin-like tegument, and maintenance of this tegument is controlled by a schistosome ortholog of the tumor suppressor TP53. To understand mechanistically how p53-1 controls tegument production, we identified a cyclin dependent kinase inhibitor homolog (cki) that was co-expressed with p53-1. RNA interference of cki resulted in a hyperproliferation phenotype, that, in combination with p53-1 RNA interference yielded abundant tumor-like growths, indicating that cki and p53-1 are bona fide tumor suppressors in Schistosoma mansoni. Interestingly, cki homologs are widely present throughout parasitic flatworms but evidently absent from their free-living ancestors, suggesting this cki homolog came from an ancient horizontal gene transfer event. This in turn implies that the evolution of parasitism in flatworms may have been aided by a highly unusual means of metazoan genetic inheritance.
Collapse
Affiliation(s)
- George Wendt
- Department of Pharmacology, University of Texas Southwestern Medical Center
| | - James J Collins
- Department of Pharmacology, University of Texas Southwestern Medical Center
| |
Collapse
|
24
|
Zhou A, Zhang W, Ge X, Liu Q, Luo F, Xu S, Hu W, Lu Y. Characterizing genetic variation on the Z chromosome in Schistosoma japonicum reveals host-parasite co-evolution. Parasit Vectors 2024; 17:207. [PMID: 38720339 PMCID: PMC11080191 DOI: 10.1186/s13071-024-06250-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/18/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Schistosomiasis is a neglected tropical disease that afflicts millions of people worldwide; it is caused by Schistosoma, the only dioecious flukes with ZW systems. Schistosoma japonicum is endemic to Asia; the Z chromosome of S. japonicum comprises one-quarter of the entire genome. Detection of positive selection using resequencing data to understand adaptive evolution has been applied to a variety of pathogens, including S. japonicum. However, the contribution of the Z chromosome to evolution and adaptation is often neglected. METHODS We obtained 1,077,526 high-quality SNPs on the Z chromosome in 72 S. japonicum using re-sequencing data publicly. To examine the faster Z effect, we compared the sequence divergence of S. japonicum with two closely related species, Schistosoma haematobium and S. mansoni. Genetic diversity was compared between the Z chromosome and autosomes in S. japonicum by calculating the nucleotide diversity (π) and Dxy values. Population structure was also assessed based on PCA and structure analysis. Besides, we employed multiple methods including Tajima's D, FST, iHS, XP-EHH, and CMS to detect positive selection signals on the Z chromosome. Further RNAi knockdown experiments were performed to investigate the potential biological functions of the candidate genes. RESULTS Our study found that the Z chromosome of S. japonicum showed faster evolution and more pronounced genetic divergence than autosomes, although the effect may be smaller than the variation among genes. Compared with autosomes, the Z chromosome in S. japonicum had a more pronounced genetic divergence of sub-populations. Notably, we identified a set of candidate genes associated with host-parasite co-evolution. In particular, LCAT exhibited significant selection signals within the Taiwan population. Further RNA interference experiments suggested that LCAT is necessary for S. japonicum survival and propagation in the definitive host. In addition, we identified several genes related to the specificity of the intermediate host in the C-M population, including Rab6 and VCP, which are involved in adaptive immune evasion to the host. CONCLUSIONS Our study provides valuable insights into the adaptive evolution of the Z chromosome in S. japonicum and further advances our understanding of the co-evolution of this medically important parasite and its hosts.
Collapse
Affiliation(s)
- An Zhou
- State Key Laboratory of Genetic Engineering, Center for Evolutionary Biology, School of Life Sciences, Fudan University, Shanghai, 200438, China
- Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China
| | - Wei Zhang
- State Key Laboratory of Genetic Engineering, Center for Evolutionary Biology, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Xueling Ge
- Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Qi Liu
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and Ministry of Education Key Laboratory of Contemporary Anthropology, Fudan University, Shanghai, 201203, China
| | - Fang Luo
- State Key Laboratory of Genetic Engineering, Center for Evolutionary Biology, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Shuhua Xu
- State Key Laboratory of Genetic Engineering, Center for Evolutionary Biology, School of Life Sciences, Fudan University, Shanghai, 200438, China
- Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China
- Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and Ministry of Education Key Laboratory of Contemporary Anthropology, Fudan University, Shanghai, 201203, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai, 201210, China
| | - Wei Hu
- State Key Laboratory of Genetic Engineering, Center for Evolutionary Biology, School of Life Sciences, Fudan University, Shanghai, 200438, China
- College of Life Sciences, Inner Mongolia University, Hohhot, 010070, China
| | - Yan Lu
- State Key Laboratory of Genetic Engineering, Center for Evolutionary Biology, School of Life Sciences, Fudan University, Shanghai, 200438, China.
- Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China.
| |
Collapse
|
25
|
McCusker P, Clarke NG, Gardiner E, Armstrong R, McCammick EM, McVeigh P, Robb E, Wells D, Nowak-Roddy M, Albaqami A, Mousley A, Coulter JA, Harrington J, Marks NJ, Maule AG. Neoblast-like stem cells of Fasciola hepatica. PLoS Pathog 2024; 20:e1011903. [PMID: 38805551 PMCID: PMC11161113 DOI: 10.1371/journal.ppat.1011903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 06/07/2024] [Accepted: 05/06/2024] [Indexed: 05/30/2024] Open
Abstract
The common liver fluke (Fasciola hepatica) causes the disease fasciolosis, which results in considerable losses within the global agri-food industry. There is a shortfall in the drugs that are effective against both the adult and juvenile life stages within the mammalian host, such that new drug targets are needed. Over the last decade the stem cells of parasitic flatworms have emerged as reservoirs of putative novel targets due to their role in development and homeostasis, including at host-parasite interfaces. Here, we investigate and characterise the proliferating cells that underpin development in F. hepatica. We provide evidence that these cells are capable of self-renewal, differentiation, and are sensitive to ionising radiation- all attributes of neoblasts in other flatworms. Changes in cell proliferation were also noted during the early stages of in vitro juvenile growth/development (around four to seven days post excystment), which coincided with a marked reduction in the nuclear area of proliferating cells. Furthermore, we generated transcriptomes from worms following irradiation-based ablation of neoblasts, identifying 124 significantly downregulated transcripts, including known stem cell markers such as fgfrA and plk1. Sixty-eight of these had homologues associated with neoblast-like cells in Schistosoma mansoni. Finally, RNA interference mediated knockdown of histone h2b (a marker of proliferating cells), ablated neoblast-like cells and impaired worm development in vitro. In summary, this work demonstrates that the proliferating cells of F. hepatica are equivalent to neoblasts of other flatworm species and demonstrate that they may serve as attractive targets for novel anthelmintics.
Collapse
Affiliation(s)
- Paul McCusker
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Nathan G. Clarke
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Erica Gardiner
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Rebecca Armstrong
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Erin M. McCammick
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Paul McVeigh
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Emily Robb
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Duncan Wells
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Madelyn Nowak-Roddy
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Abdullah Albaqami
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Angela Mousley
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | | | - John Harrington
- Boehringer Ingelheim Animal Health, Duluth, Georgia, United States of America
| | - Nikki J. Marks
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| | - Aaron G. Maule
- Understanding Health & Disease, School of Biological Sciences, Queen’s University Belfast, Belfast, United Kingdom
| |
Collapse
|
26
|
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
|
27
|
Morizet D, Foucher I, Alunni A, Bally-Cuif L. Reconstruction of macroglia and adult neurogenesis evolution through cross-species single-cell transcriptomic analyses. Nat Commun 2024; 15:3306. [PMID: 38632253 PMCID: PMC11024210 DOI: 10.1038/s41467-024-47484-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 03/29/2024] [Indexed: 04/19/2024] Open
Abstract
Macroglia fulfill essential functions in the adult vertebrate brain, producing and maintaining neurons and regulating neuronal communication. However, we still know little about their emergence and diversification. We used the zebrafish D. rerio as a distant vertebrate model with moderate glial diversity as anchor to reanalyze datasets covering over 600 million years of evolution. We identify core features of adult neurogenesis and innovations in the mammalian lineage with a potential link to the rarity of radial glia-like cells in adult humans. Our results also suggest that functions associated with astrocytes originated in a multifunctional cell type fulfilling both neural stem cell and astrocytic functions before these diverged. Finally, we identify conserved elements of macroglial cell identity and function and their time of emergence during evolution.
Collapse
Affiliation(s)
- David Morizet
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, Team supported by the Ligue Nationale Contre le Cancer, F-75015, Paris, France.
- Sorbonne Université, Collège doctoral, F-75005, Paris, France.
| | - Isabelle Foucher
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, Team supported by the Ligue Nationale Contre le Cancer, F-75015, Paris, France
| | - Alessandro Alunni
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, Team supported by the Ligue Nationale Contre le Cancer, F-75015, Paris, France
- Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, CNRS UMR9197, F-91190, Gif-sur-Yvette, France
| | - Laure Bally-Cuif
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, Team supported by the Ligue Nationale Contre le Cancer, F-75015, Paris, France.
| |
Collapse
|
28
|
Diaz Soria CL, Attenborough T, Lu Z, Fontenla S, Graham J, Hall C, Thompson S, Andrews TGR, Rawlinson KA, Berriman M, Rinaldi G. Single-cell transcriptomics of the human parasite Schistosoma mansoni first intra-molluscan stage reveals tentative tegumental and stem-cell regulators. Sci Rep 2024; 14:5974. [PMID: 38472267 PMCID: PMC10933418 DOI: 10.1038/s41598-024-55790-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Schistosomiasis is a major Neglected Tropical Disease, caused by the infection with blood flukes in the genus Schistosoma. To complete the life cycle, the parasite undergoes asexual and sexual reproduction within an intermediate snail host and a definitive mammalian host, respectively. The intra-molluscan phase provides a critical amplification step that ensures a successful transmission. However, the cellular and molecular mechanisms underlying the development of the intra-molluscan stages remain poorly understood. Here, single cell suspensions from S. mansoni mother sporocysts were produced and sequenced using the droplet-based 10X Genomics Chromium platform. Six cell clusters comprising two tegument, muscle, neuron, parenchyma and stem/germinal cell clusters were identified and validated by in situ hybridisation. Gene Ontology term analysis predicted key biological processes for each of the clusters, including three stem/germinal sub-clusters. Furthermore, putative transcription factors predicted for stem/germinal and tegument clusters may play key roles during parasite development and interaction with the intermediate host.
Collapse
Affiliation(s)
| | - Teresa Attenborough
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, 120 University Place, Glasgow, G12 8TA, UK
| | - Zhigang Lu
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Santiago Fontenla
- Departamento de Genética, Facultad de Medicina, Universidad de la República (UDELAR), Montevideo, Uruguay
| | - Jennie Graham
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Christopher Hall
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Sam Thompson
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | | | - Kate A Rawlinson
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
- Josephine Bay Paul Center, Marine Biological Laboratory, Woods Hole, MA, USA
| | - Matthew Berriman
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK.
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, 120 University Place, Glasgow, G12 8TA, UK.
| | - Gabriel Rinaldi
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK.
- Department of Life Sciences, Aberystwyth University, Edward Llwyd Building, Penglais Campus, Aberystwyth, SY23 3DA, UK.
| |
Collapse
|
29
|
Costábile A, Domínguez MF, Guarnaschelli I, Preza M, Koziol U, Castillo E, Tort JF. Purification and transcriptomic characterization of proliferative cells of Mesocestoides corti selectively affected by irradiation. FRONTIERS IN PARASITOLOGY 2024; 3:1362199. [PMID: 39817174 PMCID: PMC11732142 DOI: 10.3389/fpara.2024.1362199] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 02/12/2024] [Indexed: 01/18/2025]
Abstract
Flatworms depend on stem cells for continued tissue growth and renewal during their life cycles, making these cells valuable drug targets. While neoblasts are extensively characterized in the free-living planarian Schmidtea mediterranea, and similar stem cells have been characterized in the trematode Schistosoma mansoni, their identification and characterization in cestodes is just emerging. Since stem cells are generally affected by irradiation, in this work we used this experimental approach to study the stem cells of the model cestode Mesocestoides corti. We found that gamma irradiation produces a dose-dependent decrease in proliferative cells, requiring higher doses than in other flatworms to completely abolish proliferation. The treatment results in the downregulation of candidate marker genes. Transcriptomic studies reveal that several genes downregulated after irradiation are conserved with other flatworms, and are related to cell cycle, DNA replication and repair functions. Furthermore, proliferative cells were isolated by cell sorting and also characterized transcriptomically. We found that the set of genes characteristic of proliferative cells agrees well with those downregulated during irradiation, and have a significant overlap with those expressed in planarian neoblasts or S. mansoni stem cells. Our study highlights that conserved mechanisms of stem cell biology may be functional in flatworms, suggesting that these could be relevant targets to evaluate in the control of parasitic species.
Collapse
Affiliation(s)
- Alicia Costábile
- Sección Bioquímica, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - María Fernanda Domínguez
- Departamento de Genética, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Inés Guarnaschelli
- Sección Biología Celular, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Matías Preza
- Sección Biología Celular, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Uriel Koziol
- Sección Biología Celular, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Estela Castillo
- Unidad de Biología Parasitaria, Facultad de Ciencias- Instituto de Higiene, Universidad de la República, Montevideo, Uruguay
| | - José F. Tort
- Departamento de Genética, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
30
|
Brann T, Beltramini A, Chaparro C, Berriman M, Doyle SR, Protasio AV. Subtelomeric plasticity contributes to gene family expansion in the human parasitic flatworm Schistosoma mansoni. BMC Genomics 2024; 25:217. [PMID: 38413905 PMCID: PMC10900676 DOI: 10.1186/s12864-024-10032-8] [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: 10/03/2023] [Accepted: 01/19/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND The genomic region that lies between the telomere and chromosome body, termed the subtelomere, is heterochromatic, repeat-rich, and frequently undergoes rearrangement. Within this region, large-scale structural changes enable gene diversification, and, as such, large multicopy gene families are often found at the subtelomere. In some parasites, genes associated with proliferation, invasion, and survival are often found in these regions, where they benefit from the subtelomere's highly plastic, rapidly changing nature. The increasing availability of complete (or near complete) parasite genomes provides an opportunity to investigate these typically poorly defined and overlooked genomic regions and potentially reveal relevant gene families necessary for the parasite's lifestyle. RESULTS Using the latest chromosome-scale genome assembly and hallmark repeat richness observed at chromosome termini, we have identified and characterised the subtelomeres of Schistosoma mansoni, a metazoan parasitic flatworm that infects over 250 million people worldwide. Approximately 12% of the S. mansoni genome is classified as subtelomeric, and, in line with other organisms, we find these regions to be gene-poor but rich in transposable elements. We find that S. mansoni subtelomeres have undergone extensive interchromosomal recombination and that these sites disproportionately contribute to the 2.3% of the genome derived from segmental duplications. This recombination has led to the expansion of subtelomeric gene clusters containing 103 genes, including the immunomodulatory annexins and other gene families with unknown roles. The largest of these is a 49-copy plexin domain-containing protein cluster, exclusively expressed in the tegument-the tissue located at the host-parasite physical interface-of intramolluscan life stages. CONCLUSIONS We propose that subtelomeric regions act as a genomic playground for trial-and-error of gene duplication and subsequent divergence. Owing to the importance of subtelomeric genes in other parasites, gene families implicated in this subtelomeric expansion within S. mansoni warrant further characterisation for a potential role in parasitism.
Collapse
Affiliation(s)
- T Brann
- Department of Pathology, University of Cambridge, Cambridge, CB1 2PQ, UK
| | - A Beltramini
- Department of Pathology, University of Cambridge, Cambridge, CB1 2PQ, UK
| | - C Chaparro
- IHPE, CNRS, IFREMER, UPVD, University Montpellier, Perpignan, F-66860, France
| | - M Berriman
- School of Infection and Immunity, University of Glasgow, Glasgow, G12 8TA, UK
| | - S R Doyle
- Wellcome Sanger Institute, Cambridge, CB10 1SA, UK
| | - A V Protasio
- Department of Pathology, University of Cambridge, Cambridge, CB1 2PQ, UK.
- Christ's College, Cambridge, CB2 3BU, UK.
| |
Collapse
|
31
|
Vance GM, Khouri MI, Neto APDS, James S, Leite LCC, Farias LP, Wilson RA. Antigenic epitope targets of rhesus macaques self-curing from Schistosoma mansoni infection. Front Immunol 2024; 14:1269336. [PMID: 38464672 PMCID: PMC10921417 DOI: 10.3389/fimmu.2023.1269336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 09/22/2023] [Indexed: 03/12/2024] Open
Abstract
The self-cure of rhesus macaques from a schistosome infection and their subsequent strong immunity to a cercarial challenge should provide novel insights into the way these parasites can be eliminated by immunological attack. High-density arrays comprising overlapping 15-mer peptides from target proteins printed on glass slides can be used to screen sera from host species to determine antibody reactivity at the single epitope level. Careful selection of proteins, based on compositional studies, is crucial to encompass only those exposed on or secreted from the intra-mammalian stages and is intended to focus the analysis solely on targets mediating protection. We report the results of this approach using two pools of sera from hi- and lo-responder macaques undergoing self-cure, to screen arrays comprising tegument, esophageal gland, and gastrodermis proteins. We show that, overall, the target epitopes are the same in both groups, but the intensity of response is twice as strong in the high responders. In addition, apart from Sm25, tegument proteins elicit much weaker responses than those originating in the alimentary tract, as was apparent in IFNγR KO mice. We also highlight the most reactive epitopes in key proteins. Armed with this knowledge, we intend to use multi-epitope constructs in vaccination experiments, which seek to emulate the self-cure process in experimental animals and potentially in humans.
Collapse
Affiliation(s)
| | - Mariana I. Khouri
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, BA, Brazil
| | - Almiro Pires da Silva Neto
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, BA, Brazil
| | - Sally James
- Department of Biology, University of York, York, United Kingdom
| | - Luciana C. C. Leite
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, SP, Brazil
| | - Leonardo Paiva Farias
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, SP, Brazil
| | - R. Alan Wilson
- Department of Biology, University of York, York, United Kingdom
- Biomedical Research Institute, University of York, York, United Kingdom
| |
Collapse
|
32
|
Sprague DJ, Rohr CM, Marchant JS. TRP drop, TRP drop: a steady patter of anti-schistosomal target illumination. FRONTIERS IN PARASITOLOGY 2024; 3:1349623. [PMID: 39817176 PMCID: PMC11731825 DOI: 10.3389/fpara.2024.1349623] [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: 12/05/2023] [Accepted: 01/22/2024] [Indexed: 01/18/2025]
Abstract
Infections caused by parasitic flatworms impart a significant disease burden. This is well exemplified by the neglected tropical disease schistosomiasis, which afflicts millions of people worldwide. The anti-schistosomal activity of various chemotypes has been known for decades, but the parasite targets of many of these remain undefined. Until recently, this included the current clinical therapy, praziquantel (PZQ). However, the tempo of target discovery has recently gathered pace, with discoveries of schistosome targets for praziquantel (PZQ) and the anthelmintic benzodiazepine, meclonazepam (MCLZ). This steady patter of target illumination has also revealed a pattern in that both PZQ and MCLZ target members of the same ion channel subgroup-transient receptor potential ion channels of the melastatin family (TRPM channels). PZQ activates one member of this family (TRPMPZQ) and MCLZ activates a different channel (TRPMMCLZ). Here, similarities and differences between these two new targets are discussed. These data highlight the need for further study of TRPM channels in parasitic flatworms given their vulnerability to chemotherapeutic attack.
Collapse
Affiliation(s)
| | | | - Jonathan S. Marchant
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
33
|
Marchant JS. Progress interrogating TRPMPZQ as the target of praziquantel. PLoS Negl Trop Dis 2024; 18:e0011929. [PMID: 38358948 PMCID: PMC10868838 DOI: 10.1371/journal.pntd.0011929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024] Open
Abstract
The drug praziquantel (PZQ) has served as the long-standing drug therapy for treatment of infections caused by parasitic flatworms. These encompass diseases caused by parasitic blood, lung, and liver flukes, as well as various tapeworm infections. Despite a history of clinical usage spanning over 4 decades, the parasite target of PZQ has long resisted identification. However, a flatworm transient receptor potential ion channel from the melastatin subfamily (TRPMPZQ) was recently identified as a target for PZQ action. Here, recent experimental progress interrogating TRPMPZQ is evaluated, encompassing biochemical, pharmacological, genetic, and comparative phylogenetic data that highlight the properties of this ion channel. Various lines of evidence that support TRPMPZQ being the therapeutic target of PZQ are presented, together with additional priorities for further research into the mechanism of action of this important clinical drug.
Collapse
Affiliation(s)
- Jonathan S. Marchant
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| |
Collapse
|
34
|
Herz M, Zarowiecki M, Wessels L, Pätzel K, Herrmann R, Braun C, Holroyd N, Huckvale T, Bergmann M, Spiliotis M, Koziol U, Berriman M, Brehm K. Genome-wide transcriptome analysis of Echinococcus multilocularis larvae and germinative cell cultures reveals genes involved in parasite stem cell function. Front Cell Infect Microbiol 2024; 14:1335946. [PMID: 38333034 PMCID: PMC10850878 DOI: 10.3389/fcimb.2024.1335946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/12/2024] [Indexed: 02/10/2024] Open
Abstract
The lethal zoonosis alveolar echinococcosis is caused by tumour-like growth of the metacestode stage of the tapeworm Echinococcus multilocularis within host organs. We previously demonstrated that metacestode proliferation is exclusively driven by somatic stem cells (germinative cells), which are the only mitotically active parasite cells that give rise to all differentiated cell types. The Echinococcus gene repertoire required for germinative cell maintenance and differentiation has not been characterised so far. We herein carried out Illumina sequencing on cDNA from Echinococcus metacestode vesicles, from metacestode tissue depleted of germinative cells, and from Echinococcus primary cell cultures. We identified a set of ~1,180 genes associated with germinative cells, which contained numerous known stem cell markers alongside genes involved in replication, cell cycle regulation, mitosis, meiosis, epigenetic modification, and nucleotide metabolism. Interestingly, we also identified 44 stem cell associated transcription factors that are likely involved in regulating germinative cell differentiation and/or pluripotency. By in situ hybridization and pulse-chase experiments, we also found a new general Echinococcus stem cell marker, EmCIP2Ah, and we provide evidence implying the presence of a slow cycling stem cell sub-population expressing the extracellular matrix factor Emkal1. RNA-Seq analyses on primary cell cultures revealed that metacestode-derived Echinococcus stem cells display an expanded differentiation capability and do not only form differentiated cell types of the metacestode, but also cells expressing genes specific for protoscoleces, adult worms, and oncospheres, including an ortholog of the schistosome praziquantel target, EmTRPMPZQ. Finally, we show that primary cell cultures contain a cell population expressing an ortholog of the tumour necrosis factor α receptor family and that mammalian TNFα accelerates the development of metacestode vesicles from germinative cells. Taken together, our analyses provide a robust and comprehensive characterization of the Echinococcus germinative cell transcriptome, demonstrate expanded differentiation capability of metacestode derived stem cells, and underscore the potential of primary germinative cell cultures to investigate developmental processes of the parasite. These data are relevant for studies into the role of Echinococcus stem cells in parasite development and will facilitate the design of anti-parasitic drugs that specifically act on the parasite germinative cell compartment.
Collapse
Affiliation(s)
- Michaela Herz
- Consultant Laboratory for Echinococcosis, Institute of Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | | | - Leonie Wessels
- Consultant Laboratory for Echinococcosis, Institute of Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Katharina Pätzel
- Consultant Laboratory for Echinococcosis, Institute of Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Ruth Herrmann
- Consultant Laboratory for Echinococcosis, Institute of Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Christiane Braun
- Consultant Laboratory for Echinococcosis, Institute of Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Nancy Holroyd
- Parasite Genomics, Wellcome Sanger Institute, Cambridge, United Kingdom
| | - Thomas Huckvale
- Parasite Genomics, Wellcome Sanger Institute, Cambridge, United Kingdom
| | - Monika Bergmann
- Consultant Laboratory for Echinococcosis, Institute of Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Markus Spiliotis
- Consultant Laboratory for Echinococcosis, Institute of Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Uriel Koziol
- Consultant Laboratory for Echinococcosis, Institute of Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
- Sección Biología Celular, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Matthew Berriman
- Parasite Genomics, Wellcome Sanger Institute, Cambridge, United Kingdom
| | - Klaus Brehm
- Consultant Laboratory for Echinococcosis, Institute of Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
35
|
Li X, Weth O, Haimann M, Möscheid MF, Huber TS, Grevelding CG. Rhodopsin orphan GPCR20 interacts with neuropeptides and directs growth, sexual differentiation, and egg production in female Schistosoma mansoni. Microbiol Spectr 2024; 12:e0219323. [PMID: 38047698 PMCID: PMC10783048 DOI: 10.1128/spectrum.02193-23] [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: 05/24/2023] [Accepted: 10/17/2023] [Indexed: 12/05/2023] Open
Abstract
IMPORTANCE Schistosomes cause schistosomiasis, one of the neglected tropical diseases as defined by the WHO. For decades, the treatment of schistosomiasis relies on a single drug, praziquantel. Due to its wide use, there is justified fear of resistance against this drug, and a vaccine is not available. Besides its biological relevance in signal transduction processes, the class of G protein-coupled receptors (GPCRs) is also well suited for drug design. Against this background, we characterized one GPCR of Schistosoma mansoni, SmGPCR20, at the molecular and functional level. We identified two potential neuropeptides (NPPs) as ligands, SmNPP26 and SmNPP40, and unraveled their roles, in combination with SmGPCR20, in neuronal processes controlling egg production, oogenesis, and growth of S. mansoni females. Since eggs are closely associated with the pathogenesis of schistosomiasis, our results contribute to the understanding of processes leading to egg production in schistosomes, which is under the control of pairing in this exceptional parasite.
Collapse
Affiliation(s)
- Xuesong Li
- Institute for Parasitology, BFS, Justus Liebig University Giessen, Giessen, Germany
| | - Oliver Weth
- Institute for Parasitology, BFS, Justus Liebig University Giessen, Giessen, Germany
| | - Martin Haimann
- Institute for Parasitology, BFS, Justus Liebig University Giessen, Giessen, Germany
| | - Max F. Möscheid
- Institute for Parasitology, BFS, Justus Liebig University Giessen, Giessen, Germany
| | - Theresa S. Huber
- Institute for Parasitology, BFS, Justus Liebig University Giessen, Giessen, Germany
| | | |
Collapse
|
36
|
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
|
37
|
Rinaldi G, Paz Meseguer C, Cantacessi C, Cortés A. Form and Function in the Digenea, with an Emphasis on Host-Parasite and Parasite-Bacteria Interactions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1454:3-45. [PMID: 39008262 DOI: 10.1007/978-3-031-60121-7_1] [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
This review covers the general aspects of the anatomy and physiology of the major body systems in digenetic trematodes, with an emphasis on new knowledge of the area acquired since the publication of the second edition of this book in 2019. In addition to reporting on key recent advances in the morphology and physiology of tegumentary, sensory, neuromuscular, digestive, excretory, and reproductive systems, and their roles in host-parasite interactions, this edition includes a section discussing the known and putative roles of bacteria in digenean biology and physiology. Furthermore, a brief discussion of current trends in the development of novel treatment and control strategies based on a better understanding of the trematode body systems and associated bacteria is provided.
Collapse
Affiliation(s)
- Gabriel Rinaldi
- Department of Life Sciences, Edward Llwyd Building, Aberystwyth University, Aberystwyth, UK
| | - Carla Paz Meseguer
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, School of Pharmacy and Food Sciences, Universitat de València, Valencia, Spain
| | - Cinzia Cantacessi
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Alba Cortés
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, School of Pharmacy and Food Sciences, Universitat de València, Valencia, Spain.
| |
Collapse
|
38
|
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
|
39
|
Zhong H, Hou L, Qin F, Ren Y, Dong B, Zhu D, Li H, Lu K, Fu Z, Liu J, Gu S, Jin Y. Molecular and functional characterization of Schistosoma japonicum annexin A13. Vet Res 2023; 54:116. [PMID: 38049816 PMCID: PMC10696758 DOI: 10.1186/s13567-023-01244-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/23/2023] [Indexed: 12/06/2023] Open
Abstract
Schistosomiasis is a neglected tropical disease that affects humans and animals in tropical and subtropical regions worldwide. Schistosome eggs are responsible for the pathogenesis and transmission of schistosomiasis, thus reducing egg production is vital for prevention and control of schistosomiasis. However, the mechanisms underlying schistosome reproduction remain unclear. Annexin proteins (ANXs) are involved in the physiological and pathological functions of schistosomes, but the specific regulatory mechanisms and roles of ANX A13 in the development of Schistosoma japonicum and host-parasite interactions remain poorly understood. Therefore, in this study, the expression profiles of SjANX A13 at different life cycle stages of S. japonicum were assessed using quantitative PCR. In addition, the expression profiles of the homolog in S. mansoni were analyzed in reference to public datasets. The results of RNA interference showed that knockdown of SjANX A13 significantly affected the development and egg production of female worms in vivo. The results of an immune protection assay showed that recombinant SjANX A13 increased production of immunoglobulin G-specific antibodies. Finally, co-culture of S. japonicum exosomes with LX-2 cells using a transwell system demonstrated that SjANX A13 is involved in host-parasite interactions via exosomes. Collectively, these results will help to clarify the roles of SjANX A13 in the development of S. japonicum and host-parasite interactions as a potential vaccine candidate.
Collapse
Affiliation(s)
- Haoran Zhong
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Ling Hou
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Shanxi, China
| | - Fanglin Qin
- College of Life Sciences, Shanghai Normal University, Shanghai, China
| | - Yuqi Ren
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Bowen Dong
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Danlin Zhu
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Hao Li
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Ke Lu
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Zhiqiang Fu
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Jinming Liu
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Shaopeng Gu
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Shanxi, China
| | - Yamei Jin
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China.
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China.
| |
Collapse
|
40
|
Martín-Zamora FM, Davies BE, Donnellan RD, Guynes K, Martín-Durán JM. Functional genomics in Spiralia. Brief Funct Genomics 2023; 22:487-497. [PMID: 37981859 PMCID: PMC10658182 DOI: 10.1093/bfgp/elad036] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/12/2023] [Accepted: 07/25/2023] [Indexed: 11/21/2023] Open
Abstract
Our understanding of the mechanisms that modulate gene expression in animals is strongly biased by studying a handful of model species that mainly belong to three groups: Insecta, Nematoda and Vertebrata. However, over half of the animal phyla belong to Spiralia, a morphologically and ecologically diverse animal clade with many species of economic and biomedical importance. Therefore, investigating genome regulation in this group is central to uncovering ancestral and derived features in genome functioning in animals, which can also be of significant societal impact. Here, we focus on five aspects of gene expression regulation to review our current knowledge of functional genomics in Spiralia. Although some fields, such as single-cell transcriptomics, are becoming more common, the study of chromatin accessibility, DNA methylation, histone post-translational modifications and genome architecture are still in their infancy. Recent efforts to generate chromosome-scale reference genome assemblies for greater species diversity and optimise state-of-the-art approaches for emerging spiralian research systems will address the existing knowledge gaps in functional genomics in this animal group.
Collapse
Affiliation(s)
- Francisco M Martín-Zamora
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Billie E Davies
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Rory D Donnellan
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Kero Guynes
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - José M Martín-Durán
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| |
Collapse
|
41
|
Chulkov EG, Rohr CM, Marchant JS. Praziquantel activates a native cation current in Schistosoma mansoni. FRONTIERS IN PARASITOLOGY 2023; 2:1285177. [PMID: 39816816 PMCID: PMC11732042 DOI: 10.3389/fpara.2023.1285177] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/26/2023] [Indexed: 01/18/2025]
Abstract
Introduction Praziquantel (PZQ), an anthelmintic drug discovered in the 1970s, is still used to treat schistosomiasis and various other infections caused by parasitic flatworms. PZQ causes a triad of phenotypic effects on schistosome worms - rapid depolarization, muscle contraction, and damage throughout the worm tegument. The molecular target mediating these effects has been intimated as a Ca2+-permeable ion channel, but native currents evoked by PZQ have not been reported in any schistosome cell type. The properties of the endogenous PZQ activated conductance therefore remain unknown. Methods Here, invasive electrophysiology was used to probe for responses to PZQ from different locales in a living schistosome worm. Results and discussion No direct response was seen in tegument-derived vesicles, or from the sub-tegumental muscle layer despite the presence of voltage-operated currents. However, PZQ rapidly triggered a sustained, non-selective cation current in recordings from neuronal tissue, targeting both the anterior ganglion and the main longitudinal nerve cord. The biophysical signature of this PZQ-evoked current resolved at single channel resolution matched that of a transient receptor potential ion channel named TRPMPZQ, recently proposed as the molecular target of PZQ. The endogenous PZQ-evoked current was also inhibited by a validated TRPMPZQ antagonist. PZQ therefore is a neuroactive anthelmintic, causing a sustained depolarization through ion channels with the characteristics of TRPMPZQ.
Collapse
Affiliation(s)
| | | | - Jonathan S. Marchant
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
42
|
Buddenborg SK, Lu Z, Sankaranarayan G, Doyle SR, Berriman M. The stage- and sex-specific transcriptome of the human parasite Schistosoma mansoni. Sci Data 2023; 10:775. [PMID: 37935722 PMCID: PMC10630280 DOI: 10.1038/s41597-023-02674-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 10/23/2023] [Indexed: 11/09/2023] Open
Abstract
The flatworm Schistosoma mansoni is an important but neglected pathogen that causes the disease schistosomiasis in millions of people worldwide. The parasite has a complex life cycle, undergoing sexual reproduction in a mammalian host and asexual replication in a snail host. Understanding the molecular mechanisms that the parasite uses to transition between hosts and develop into dimorphic reproductively competent adults may reveal new strategies for control. We present the first comprehensive transcriptomic analysis of S. mansoni, from eggs to sexually naïve worms. Focusing on eight life stages spanning free-living water-borne and parasitic stages from both intermediate and definitive hosts, we have generated deep RNA-seq data for five replicates per group for a total of 75 data sets. The data were produced using a single approach to increase the accuracy of stage-to-stage comparisons and made accessible via a user-friendly tool to visualise and explore gene expression ( https://lifecycle.schisto.xyz/ ). These data are valuable for understanding the biology and sex-specific development of schistosomes and the interpretation of complementary genomic and functional genetics studies.
Collapse
Affiliation(s)
| | - Zhigang Lu
- Wellcome Sanger Institute, Cambridgeshire, CB10 1SA, Hinxton, UK
| | | | - Stephen R Doyle
- Wellcome Sanger Institute, Cambridgeshire, CB10 1SA, Hinxton, UK
| | - Matthew Berriman
- Wellcome Sanger Institute, Cambridgeshire, CB10 1SA, Hinxton, UK.
- School of Infection and Immunity, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK.
| |
Collapse
|
43
|
Montagne J, Preza M, Koziol U. Stem cell proliferation and differentiation during larval metamorphosis of the model tapeworm Hymenolepis microstoma. Front Cell Infect Microbiol 2023; 13:1286190. [PMID: 37908761 PMCID: PMC10614006 DOI: 10.3389/fcimb.2023.1286190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/02/2023] [Indexed: 11/02/2023] Open
Abstract
Background Tapeworm larvae cause important diseases in humans and domestic animals. During infection, the first larval stage undergoes a metamorphosis where tissues are formed de novo from a population of stem cells called germinative cells. This process is difficult to study for human pathogens, as these larvae are infectious and difficult to obtain in the laboratory. Methods In this work, we analyzed cell proliferation and differentiation during larval metamorphosis in the model tapeworm Hymenolepis microstoma, by in vivo labelling of proliferating cells with the thymidine analogue 5-ethynyl-2'-deoxyuridine (EdU), tracing their differentiation with a suite of specific molecular markers for different cell types. Results Proliferating cells are very abundant and fast-cycling during early metamorphosis: the total number of cells duplicates every ten hours, and the length of G2 is only 75 minutes. New tegumental, muscle and nerve cells differentiate from this pool of proliferating germinative cells, and these processes are very fast, as differentiation markers for neurons and muscle cells appear within 24 hours after exiting the cell cycle, and fusion of new cells to the tegumental syncytium can be detected after only 4 hours. Tegumental and muscle cells appear from early stages of metamorphosis (24 to 48 hours post-infection); in contrast, most markers for differentiating neurons appear later, and the detection of synapsin and neuropeptides correlates with scolex retraction. Finally, we identified populations of proliferating cells that express conserved genes associated with neuronal progenitors and precursors, suggesting the existence of tissue-specific lineages among germinative cells. Discussion These results provide for the first time a comprehensive view of the development of new tissues during tapeworm larval metamorphosis, providing a framework for similar studies in human and veterinary pathogens.
Collapse
Affiliation(s)
| | | | - Uriel Koziol
- Sección Biología Celular, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
44
|
Li J, Zhang Y, Li H, Jiang J, Guo C, Zhou Z, Luo Y, Zhou C, Ming Y. Single-cell RNA sequencing reveals a peripheral landscape of immune cells in Schistosomiasis japonica. Parasit Vectors 2023; 16:356. [PMID: 37817226 PMCID: PMC10563327 DOI: 10.1186/s13071-023-05975-y] [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: 06/27/2023] [Accepted: 09/20/2023] [Indexed: 10/12/2023] Open
Abstract
BACKGROUND Schistosomiasis, also known as bilharzia, is a devastating parasitic disease. This progressive and debilitating helminth disease is often associated with poverty and can lead to chronic poor health. Despite ongoing research, there is currently no effective vaccine for schistosomiasis, and praziquantel remains the only available treatment option. According to the progression of schistosomiasis, infections caused by schistosomes are classified into three distinct clinical phases: acute, chronic and advanced schistosomiasis. However, the underlying immune mechanism involved in the progression of schistosomiasis remains poorly understood. METHODS We employed single-cell RNA sequencing (scRNA-seq) to profile the immune landscape of Schistosomiasis japonica infection based on peripheral blood mononuclear cells (PBMCs) from a healthy control group (n = 4), chronic schistosomiasis group (n = 4) and advanced schistosomiasis group (n = 2). RESULTS Of 89,896 cells, 24 major cell clusters were ultimately included in our analysis. Neutrophils and NK/T cells accounted for the major proportion in the chronic group and the healthy group, and monocytes dominated in the advanced group. A preliminary study showed that NKT cells were increased in patients with schistosomiasis and that CXCR2 + NKT cells were proinflammatory cells. Plasma cells also accounted for a large proportion of B cells in the advanced group. MHC molecules in monocytes were notably lower in the advanced group than in the chronic group or the healthy control group. However, monocytes in the advanced group exhibited high expression of FOLR3 and CCR2. CONCLUSIONS Overall, this study enhances our understanding of the immune mechanisms involved in schistosomiasis. It provides a transcriptional atlas of peripheral immune cells that may contribute to elimination of the disease. This preliminary study suggests that the increased presence of CCR2 + monocyte and CXCR2 + NKT cells might participate in the progression of schistosomiasis.
Collapse
Affiliation(s)
- Junhui Li
- Transplantation Center, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Changsha, 410013, Hunan, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, Hunan, China
| | - Yu Zhang
- Transplantation Center, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Changsha, 410013, Hunan, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, Hunan, China
| | - Hao Li
- Transplantation Center, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Changsha, 410013, Hunan, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, Hunan, China
| | - Jie Jiang
- Transplantation Center, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Changsha, 410013, Hunan, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, Hunan, China
| | - Chen Guo
- Transplantation Center, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Changsha, 410013, Hunan, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, Hunan, China
| | - Zhaoqin Zhou
- Transplantation Center, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Changsha, 410013, Hunan, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, Hunan, China
| | - Yulin Luo
- Transplantation Center, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Changsha, 410013, Hunan, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, Hunan, China
| | - Chen Zhou
- Transplantation Center, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Changsha, 410013, Hunan, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, Hunan, China
| | - Yingzi Ming
- Transplantation Center, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Changsha, 410013, Hunan, China.
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, Hunan, China.
| |
Collapse
|
45
|
Shakir EMN, Rinaldi G, Kirk RS, Walker AJ. Schistosoma mansoni excretory-secretory products induce protein kinase signalling, hyperkinesia, and stem cell proliferation in the opposite sex. Commun Biol 2023; 6:985. [PMID: 37752334 PMCID: PMC10522684 DOI: 10.1038/s42003-023-05333-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 09/08/2023] [Indexed: 09/28/2023] Open
Abstract
Adult male and female schistosomes in copula dwell within human blood vessels and lay eggs that cause the major Neglected Tropical Disease human schistosomiasis. How males and females communicate to each other is poorly understood; however, male-female physical interaction is known to be important. Here, we investigate whether excretory-secretory products (ESPs), released into the external milieu by mature Schistosoma mansoni, might induce responses in the opposite sex. We demonstrate that ESPs adhere to the surface of opposite sex worms inducing the activation of extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase (p38 MAPK) pathways, particularly in the parasite tegument. Furthermore, we show that mature worms stimulated signalling in juvenile worms. Strikingly, we demonstrate that ESPs from the opposite sex promote stem cell proliferation, in an ERK- and p38 MAPK-dependent manner, in the tegument and within the testes of males, and the ovaries and vitellaria of females. Hyperkinesia also occurs following opposite sex ESP exposure. Our findings support the hypothesis that male and female schistosomes may communicate over distance to modulate key processes underlying worm development and disease progression, opening unique avenues for schistosomiasis control.
Collapse
Affiliation(s)
- Eman M N Shakir
- Molecular Parasitology Laboratory, School of Life Sciences Pharmacy and Chemistry, Kingston University, Penrhyn Road, Kingston upon Thames, KT1 2EE, UK
| | - Gabriel Rinaldi
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
- Department of Life Sciences, Edward Llwyd Building, Aberystwyth University, Aberystwyth, SY23 3DA, UK
| | - Ruth S Kirk
- Molecular Parasitology Laboratory, School of Life Sciences Pharmacy and Chemistry, Kingston University, Penrhyn Road, Kingston upon Thames, KT1 2EE, UK
| | - Anthony J Walker
- Molecular Parasitology Laboratory, School of Life Sciences Pharmacy and Chemistry, Kingston University, Penrhyn Road, Kingston upon Thames, KT1 2EE, UK.
| |
Collapse
|
46
|
Ellis JT, Reichel MP. Twitter trends in #Parasitology determined by text mining and topic modelling. CURRENT RESEARCH IN PARASITOLOGY & VECTOR-BORNE DISEASES 2023; 4:100138. [PMID: 37670843 PMCID: PMC10475476 DOI: 10.1016/j.crpvbd.2023.100138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/08/2023] [Accepted: 08/10/2023] [Indexed: 09/07/2023]
Abstract
This study investigated the emergence and use of Twitter, as of July 2023 being rebranded as X, as the main forum for social media communication in parasitology. A dataset of tweets was constructed using a keyword search of Twitter with the search terms 'malaria', 'Plasmodium', 'Leishmania', 'Trypanosoma', 'Toxoplasma' and 'Schistosoma' for the period from 2011 to 2020. Exploratory data analyses of tweet content were conducted, including language, usernames and hashtags. To identify parasitology topics of discussion, keywords and phrases were extracted using KeyBert and biterm topic modelling. The sentiment of tweets was analysed using VADER. The results show that the number of tweets including the keywords increased from 2011 (for malaria) and 2013 (for the others) to 2020, with the highest number of tweets being recorded in 2020. The maximum number of yearly tweets for Plasmodium, Leishmania, Toxoplasma, Trypanosoma and Schistosoma was recorded in 2020 (2804, 2161, 1570, 680 and 360 tweets, respectively). English was the most commonly used language for tweeting, although the percentage varied across the searches. In tweets mentioning Leishmania, only ∼37% were in English, with Spanish being more common. Across all the searches, Portuguese was another common language found. Popular tweets on Toxoplasma contained keywords relating to mental health including depression, anxiety and schizophrenia. The Trypanosoma tweets referenced drugs (benznidazole, nifurtimox) and vectors (bugs, triatomines, tsetse), while the Schistosoma tweets referenced areas of biology including pathology, eggs and snails. A wide variety of individuals and organisations were shown to be associated with Twitter activity. Many journals in the parasitology arena regularly tweet about publications from their journal, and professional societies promote activity and events that are important to them. These represent examples of trusted sources of information, often by experts in their fields. Social media activity of influencers, however, who have large numbers of followers, might have little or no training in science. The existence of such tweeters does raise cause for concern to parasitology, as one may start to question the quality of information being disseminated.
Collapse
Affiliation(s)
- John T. Ellis
- School of Life Sciences, University of Technology Sydney, Broadway, NSW, Australia
| | - Michael P. Reichel
- Department of Population Medicine & Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| |
Collapse
|
47
|
Ittiprasert W, Moescheid MF, Chaparro C, Mann VH, Quack T, Rodpai R, Miller A, Wisitpongpun P, Buakaew W, Mentink-Kane M, Schmid S, Popratiloff A, Grevelding CG, Grunau C, Brindley PJ. Targeted insertion and reporter transgene activity at a gene safe harbor of the human blood fluke, Schistosoma mansoni. CELL REPORTS METHODS 2023; 3:100535. [PMID: 37533651 PMCID: PMC10391569 DOI: 10.1016/j.crmeth.2023.100535] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/22/2023] [Accepted: 06/25/2023] [Indexed: 08/04/2023]
Abstract
The identification and characterization of genomic safe harbor sites (GSHs) can facilitate consistent transgene activity with minimal disruption to the host cell genome. We combined computational genome annotation and chromatin structure analysis to predict the location of four GSHs in the human blood fluke, Schistosoma mansoni, a major infectious pathogen of the tropics. A transgene was introduced via CRISPR-Cas-assisted homology-directed repair into one of the GSHs in the egg of the parasite. Gene editing efficiencies of 24% and transgene-encoded fluorescence of 75% of gene-edited schistosome eggs were observed. The approach advances functional genomics for schistosomes by providing a tractable path for generating transgenics using homology-directed, repair-catalyzed transgene insertion. We also suggest that this work will serve as a roadmap for the development of similar approaches in helminths more broadly.
Collapse
Affiliation(s)
- Wannaporn Ittiprasert
- Department of Microbiology, Immunology & Tropical Medicine, School of Medicine & Health Sciences, George Washington University, Washington, DC 20037, USA
| | - Max F. Moescheid
- Department of Microbiology, Immunology & Tropical Medicine, School of Medicine & Health Sciences, George Washington University, Washington, DC 20037, USA
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| | - Cristian Chaparro
- IHPE, University of Perpignan Via Domitia, CNRS, IFREMER, University Montpellier, Perpignan, France
| | - Victoria H. Mann
- Department of Microbiology, Immunology & Tropical Medicine, School of Medicine & Health Sciences, George Washington University, Washington, DC 20037, USA
| | - Thomas Quack
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| | - Rutchanee Rodpai
- Department of Microbiology, Immunology & Tropical Medicine, School of Medicine & Health Sciences, George Washington University, Washington, DC 20037, USA
- Department of Parasitology and Excellence in Medical Innovation, and Technology Research Group, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - André Miller
- Schistosomiasis Resource Center, Biomedical Research Institute, Rockville, MD 20850, USA
| | - Prapakorn Wisitpongpun
- Department of Microbiology, Immunology & Tropical Medicine, School of Medicine & Health Sciences, George Washington University, Washington, DC 20037, USA
- Faculty of Medical Technology, Rangsit University, Pathum Thani 12000, Thailand
| | - Watunyoo Buakaew
- Department of Microbiology, Immunology & Tropical Medicine, School of Medicine & Health Sciences, George Washington University, Washington, DC 20037, USA
- Department of Microbiology, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand
| | - Margaret Mentink-Kane
- Schistosomiasis Resource Center, Biomedical Research Institute, Rockville, MD 20850, USA
| | - Sarah Schmid
- Schistosomiasis Resource Center, Biomedical Research Institute, Rockville, MD 20850, USA
| | - Anastas Popratiloff
- Nanofabrication and Imaging Center, Science & Engineering Hall, George Washington University, Washington, DC 20052, USA
| | - Christoph G. Grevelding
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| | - Christoph Grunau
- IHPE, University of Perpignan Via Domitia, CNRS, IFREMER, University Montpellier, Perpignan, France
| | - Paul J. Brindley
- Department of Microbiology, Immunology & Tropical Medicine, School of Medicine & Health Sciences, George Washington University, Washington, DC 20037, USA
| |
Collapse
|
48
|
Zhu P, Wu K, Zhang C, Batool SS, Li A, Yu Z, Huang J. Advances in new target molecules against schistosomiasis: A comprehensive discussion of physiological structure and nutrient intake. PLoS Pathog 2023; 19:e1011498. [PMID: 37498810 PMCID: PMC10374103 DOI: 10.1371/journal.ppat.1011498] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023] Open
Abstract
Schistosomiasis, a severe parasitic disease, is primarily caused by Schistosoma mansoni, Schistosoma japonicum, or Schistosoma haematobium. Currently, praziquantel is the only recommended drug for human schistosome infection. However, the lack of efficacy of praziquantel against juvenile worms and concerns about the emergence of drug resistance are driving forces behind the research for an alternative medication. Schistosomes are obligatory parasites that survive on nutrients obtained from their host. The ability of nutrient uptake depends on their physiological structure. In short, the formation and maintenance of the structure and nutrient supply are mutually reinforcing and interdependent. In this review, we focus on the structural features of the tegument, esophagus, and intestine of schistosomes and their roles in nutrient acquisition. Moreover, we introduce the significance and modes of glucose, lipids, proteins, and amino acids intake in schistosomes. We linked the schistosome structure and nutrient supply, introduced the currently emerging targets, and analyzed the current bottlenecks in the research and development of drugs and vaccines, in the hope of providing new strategies for the prevention and control of schistosomiasis.
Collapse
Affiliation(s)
- Peng Zhu
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
| | - Kaijuan Wu
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China
| | - Chaobin Zhang
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
| | - Syeda Sundas Batool
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| | - Anqiao Li
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
| | - Zheng Yu
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| | - Jing Huang
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| |
Collapse
|
49
|
Chulkov EG, Palygin O, Yahya NA, Park SK, Marchant JS. Electrophysiological characterization of a schistosome transient receptor potential channel activated by praziquantel. Int J Parasitol 2023; 53:415-425. [PMID: 36610556 PMCID: PMC10258134 DOI: 10.1016/j.ijpara.2022.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/12/2022] [Accepted: 11/16/2022] [Indexed: 01/06/2023]
Abstract
Ion channels have proved to be productive targets for anthelmintic chemotherapy. One example is the recent discovery of a parasitic flatworm ion channel targeted by praziquantel (PZQ), the main clinical therapy used for treatment of schistosomiasis. The ion channel activated by PZQ - a transient receptor potential ion channel of the melastatin subfamily, named TRPMPZQ - is a Ca2+-permeable ion channel expressed in all parasitic flatworms that are PZQ-sensitive. However, little is currently known about the electrophysiological properties of this target that mediates the deleterious action of PZQ on many trematodes and cestodes. Here, we provide a detailed biophysical characterization of the properties of Schistosoma mansoni TRPMPZQ channel (Sm.TRPMPZQ) in response to PZQ. Single channel electrophysiological analysis demonstrated that Sm.TRPMPZQ when activated by PZQ is a non-selective, large conductance, voltage-insensitive cation channel that displays distinct properties from human TRPM paralogs. Sm.TRPMPZQ is Ca2+-permeable but does not require Ca2+ for channel gating in response to PZQ. TRPMPZQ from Schistosoma japonicum (Sj.TRPMPZQ) and Schistosoma haematobium (Sh.TRPMPZQ) displayed similar characteristics. Profiling Sm.TRPMPZQ responsiveness to PZQ has established a biophysical signature for this channel that will aid future investigation of endogenous TRPMPZQ activity, as well as analyses of endogenous and exogenous regulators of this novel, druggable antiparasitic target.
Collapse
Affiliation(s)
- Evgeny G Chulkov
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Oleg Palygin
- Division of Nephrology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Nawal A Yahya
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Pharmacology, University of Minnesota Medical School, 312 Church St. SE, Minneapolis, MN 55455, USA
| | - Sang-Kyu Park
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jonathan S Marchant
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
50
|
Britton C, Laing R, McNeilly TN, Perez MG, Otto TD, Hildersley KA, Maizels RM, Devaney E, Gillan V. New technologies to study helminth development and host-parasite interactions. Int J Parasitol 2023; 53:393-403. [PMID: 36931423 DOI: 10.1016/j.ijpara.2022.11.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/24/2022] [Accepted: 11/26/2022] [Indexed: 03/17/2023]
Abstract
How parasites develop and survive, and how they stimulate or modulate host immune responses are important in understanding disease pathology and for the design of new control strategies. Microarray analysis and bulk RNA sequencing have provided a wealth of data on gene expression as parasites develop through different life-cycle stages and on host cell responses to infection. These techniques have enabled gene expression in the whole organism or host tissue to be detailed, but do not take account of the heterogeneity between cells of different types or developmental stages, nor the spatial organisation of these cells. Single-cell RNA-seq (scRNA-seq) adds a new dimension to studying parasite biology and host immunity by enabling gene profiling at the individual cell level. Here we review the application of scRNA-seq to establish gene expression cell atlases for multicellular helminths and to explore the expansion and molecular profile of individual host cell types involved in parasite immunity and tissue repair. Studying host-parasite interactions in vivo is challenging and we conclude this review by briefly discussing the applications of organoids (stem-cell derived mini-tissues) to examine host-parasite interactions at the local level, and as a potential system to study parasite development in vitro. Organoid technology and its applications have developed rapidly, and the elegant studies performed to date support the use of organoids as an alternative in vitro system for research on helminth parasites.
Collapse
Affiliation(s)
- Collette Britton
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, United Kingdom.
| | - Roz Laing
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, United Kingdom
| | - Tom N McNeilly
- Disease Control Department, Moredun Research Institute, Penicuik, United Kingdom
| | - Matias G Perez
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, United Kingdom
| | - Thomas D Otto
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, United Kingdom
| | - Katie A Hildersley
- Disease Control Department, Moredun Research Institute, Penicuik, United Kingdom
| | - Rick M Maizels
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, United Kingdom
| | - Eileen Devaney
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, United Kingdom
| | - Victoria Gillan
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|