451
|
Sankaranarayanan G, Coghlan A, Driguez P, Lotkowska ME, Sanders M, Holroyd N, Tracey A, Berriman M, Rinaldi G. Large CRISPR-Cas-induced deletions in the oxamniquine resistance locus of the human parasite Schistosoma mansoni. Wellcome Open Res 2021; 5:178. [PMID: 32789192 PMCID: PMC7405262 DOI: 10.12688/wellcomeopenres.16031.2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2020] [Indexed: 12/18/2022] Open
Abstract
Background. At least 250 million people worldwide suffer from schistosomiasis, caused by Schistosoma worms. Genome sequences for several Schistosoma species are available, including a high-quality annotated reference for Schistosoma mansoni. There is a pressing need to develop a reliable functional toolkit to translate these data into new biological insights and targets for intervention. CRISPR-Cas9 was recently demonstrated for the first time in S. mansoni, to produce somatic mutations in the omega-1 ( ω1) gene. Methods. We employed CRISPR-Cas9 to introduce somatic mutations in a second gene, SULT-OR, a sulfotransferase expressed in the parasitic stages of S. mansoni, in which mutations confer resistance to the drug oxamniquine. A 262-bp PCR product spanning the region targeted by the gRNA against SULT-OR was amplified, and mutations identified in it by high-throughput sequencing. Results. We found that 0.3-2.0% of aligned reads from CRISPR-Cas9-treated adult worms showed deletions spanning the predicted Cas9 cut site, compared to 0.1-0.2% for sporocysts, while deletions were extremely rare in eggs. The most common deletion observed in adults and sporocysts was a 34 bp-deletion directly upstream of the predicted cut site, but rarer deletions reaching as far as 102 bp upstream of the cut site were also detected. The CRISPR-Cas9-induced deletions, if homozygous, are predicted to cause resistance to oxamniquine by producing frameshifts, ablating SULT-OR transcription, or leading to mRNA degradation via the nonsense-mediated mRNA decay pathway. However, no SULT-OR knock down at the mRNA level was observed, presumably because the cells in which CRISPR-Cas9 did induce mutations represented a small fraction of all cells expressing SULT-OR. Conclusions. Further optimisation of CRISPR-Cas protocols for different developmental stages and particular cell types, including germline cells, will contribute to the generation of a homozygous knock-out in any gene of interest, and in particular the SULT-OR gene to derive an oxamniquine-resistant stable transgenic line.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Alan Tracey
- Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
| | | | | |
Collapse
|
452
|
Diarylureas: Repositioning from Antitumor to Antimicrobials or Multi-Target Agents against New Pandemics. Antibiotics (Basel) 2021; 10:antibiotics10010092. [PMID: 33477901 PMCID: PMC7833385 DOI: 10.3390/antibiotics10010092] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/12/2021] [Accepted: 01/18/2021] [Indexed: 12/19/2022] Open
Abstract
Antimicrobials have allowed medical advancements over several decades. However, the continuous emergence of antimicrobial resistance restricts efficacy in treating infectious diseases. In this context, the drug repositioning of already known biological active compounds to antimicrobials could represent a useful strategy. In 2002 and 2003, the SARS-CoV pandemic immobilized the Far East regions. However, the drug discovery attempts to study the virus have stopped after the crisis declined. Today’s COVID-19 pandemic could probably have been avoided if those efforts against SARS-CoV had continued. Recently, a new coronavirus variant was identified in the UK. Because of this, the search for safe and potent antimicrobials and antivirals is urgent. Apart from antiviral treatment for severe cases of COVID-19, many patients with mild disease without pneumonia or moderate disease with pneumonia have received different classes of antibiotics. Diarylureas are tyrosine kinase inhibitors well known in the art as anticancer agents, which might be useful tools for a reposition as antimicrobials. The first to come onto the market as anticancer was sorafenib, followed by some other active molecules. For this interesting class of organic compounds antimicrobial, antiviral, antithrombotic, antimalarial, and anti-inflammatory properties have been reported in the literature. These numerous properties make these compounds interesting for a new possible pandemic considering that, as well as for other viral infections also for CoVID-19, a multitarget therapeutic strategy could be favorable. This review is meant to be an overview on diarylureas, focusing on their biological activities, not dwelling on the already known antitumor activity. Quite a lot of papers present in the literature underline and highlight the importance of these molecules as versatile scaffolds for the development of new and promising antimicrobials and multitarget agents against new pandemic events.
Collapse
|
453
|
Tumor Necrosis Factor and Schistosoma mansoni egg antigen omega-1 shape distinct aspects of the early egg-induced granulomatous response. PLoS Negl Trop Dis 2021; 15:e0008814. [PMID: 33465071 PMCID: PMC7845976 DOI: 10.1371/journal.pntd.0008814] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/29/2021] [Accepted: 11/17/2020] [Indexed: 11/29/2022] Open
Abstract
Infections by schistosomes result in granulomatous lesions around parasite eggs entrapped within the host tissues. The host and parasite determinants of the Schistosoma mansoni egg-induced granulomatous response are areas of active investigation. Some studies in mice implicate Tumor Necrosis Factor (TNF) produced in response to the infection whereas others fail to find a role for it. In addition, in the mouse model, the S. mansoni secreted egg antigen omega-1 is found to induce granulomas but the underlying mechanism remains unknown. We have recently developed the zebrafish larva as a model to study macrophage recruitment and granuloma formation in response to Schistosoma mansoni eggs. Here we use this model to investigate the mechanisms by which TNF and omega-1 shape the early granulomatous response. We find that TNF, specifically signaling through TNF receptor 1, is not required for macrophage recruitment to the egg and granuloma initiation but does mediate granuloma enlargement. In contrast, omega-1 mediates initial macrophage recruitment, with this chemotactic activity being dependent on its RNase activity. Our findings further the understanding of the role of these host- and parasite-derived factors and show that they impact distinct facets of the granulomatous response to the schistosome egg. Schistosomiasis is a disease caused by parasitic flatworms which lay eggs within the veins of their human host. Upon sensing the parasite egg, macrophages, the first line defense cells, aggregate tightly around the egg to encapsulate it within an immune structure known as a granuloma. These granulomas are the key pathological structures which determine both host disease outcome and parasite transmission. Studies in mice have implicated omega-1, a secreted parasite protein. Omega-1 is an RNase, an enzyme that degrades host RNA. Mouse studies have also suggested that a host defense protein, Tumor Necrosis Factor (TNF), is required to form granulomas around the egg. We used the small and transparent zebrafish larva to examine the requirement of omega-1 and TNF for granuloma formation. We find that omega-1 induces rapid macrophage migration and that its RNase activity is required for this. In contrast, TNF is not involved in the initial recruitment of macrophages. Rather, it enlarges granulomas after they are initiated. These findings improve our understanding of the role of omega-1 and TNF, and show that they impact distinct facets of granuloma formation around Schistosoma eggs.
Collapse
|
454
|
Marín-Martínez L, Kyriakos G, Sánchez-Gutiérrez D. Pseudotumoral form of schistosomiasis mimicking neuroendocrine tumor: a case report and brief review of the differential diagnosis of retroperitoneal masses. Pan Afr Med J 2021; 37:186. [PMID: 33447341 PMCID: PMC7778187 DOI: 10.11604/pamj.2020.37.186.26344] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 10/15/2020] [Indexed: 02/02/2023] Open
Abstract
Differential diagnosis of retroperitoneal masses may become complex and requires careful anamnesis, physical examination and several complementary tests. We present the clinical case of a male patient aged 45 years who was diagnosed with a 4cm paraaortic lesion compatible with neuroendocrine tumor in the abdominal computed tomography (CT) exam. The workup performed with SPECT-CT, somatostatin receptors scintigraphy, MIBG scintigraphy, 24-hour urine total and fractionated catecholamines and 24-hour urine 5-OH indoleacetic did not confirm the first diagnostic impression. Finally, the lesion was biopsied and presence of micro-organisms was revealed. Further exams confirmed schistosomiasis as the cause of the paraaortic lesion. Histological diagnosis can be helpful with regard to the differential diagnosis of retroperitoneal masses.
Collapse
Affiliation(s)
- Luis Marín-Martínez
- Sección de Endocrinología y Nutrición, Hospital General Universitario Santa Lucía, Cartagena, Spain
| | - Georgios Kyriakos
- Sección de Endocrinología y Nutrición, Hospital General Universitario Santa Lucía, Cartagena, Spain
| | - David Sánchez-Gutiérrez
- Servicio de Anatomía Patológica, Hospital General Universitario Santa Lucía, Cartagena, Spain
| |
Collapse
|
455
|
Ouattara M, Diakité NR, Yao PK, Saric J, Coulibaly JT, Assaré RK, Bassa FK, Koné N, Guindo-Coulibaly N, Hattendorf J, Utzinger J, N’Goran EK. Effectiveness of school-based preventive chemotherapy strategies for sustaining the control of schistosomiasis in Côte d'Ivoire: Results of a 5-year cluster randomized trial. PLoS Negl Trop Dis 2021; 15:e0008845. [PMID: 33449924 PMCID: PMC7810315 DOI: 10.1371/journal.pntd.0008845] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 09/30/2020] [Indexed: 11/18/2022] Open
Abstract
Background Preventive chemotherapy using praziquantel is the mainstay for schistosomiasis control. However, there is little evidence on what is supposed to be the most effective school-based treatment strategy to sustain morbidity control. The aim of this study was to compare differences in Schistosoma mansoni prevalence and infection intensity between three different schedules of school-based preventive chemotherapy in an area with moderate prevalence of S. mansoni in Côte d’Ivoire. Methodology Seventy-five schools were randomly assigned to one of three intervention arms: (i) annual school-based preventive chemotherapy with praziquantel (40 mg/kg) over four years; (ii) praziquantel treatment only in the first two years, followed by two years whithout treatment; and (iii) praziquantel treatment in years 1 and 3 without treatment in-between. Cross-sectional parasitologic surveys were carried out prior to each round of preventive chemotherapy. The difference in S. mansoni prevalence and infection intensity was assessed by multiple Kato-Katz thick smears, among children aged 9–12 years at the time of each survey. First-grade children, aged 5–8 years who had never received praziquantel, were also tested at baseline and at the end of the study. Principal findings Overall, 7,410 children aged 9–12 years were examined at baseline and 7,223 at the final survey. The baseline prevalence of S. mansoni was 17.4%, 20.2%, and 25.2% in arms 1, 2, and 3, respectively. In the final year, we observed the lowest prevalence of 10.4% in arm 1, compared to 18.2% in arm 2 and 17.5% in arm 3. The comparison between arms 1 and 2 estimated an odds ratio (OR) of 0.52 but the difference was not statistically significant (95% confidence interval (CI) = 0.23–1.16). Likewise the difference between arms 1 and 3 lacked statistical significance (OR = 0.55, 95% CI = 0.23–1.29). There was no noteworthy difference observed between arms 2 and 3 (OR = 1.06, 95% CI = 0.64–1.75). The lowest S. mansoni fecal egg counts in the final year survey were observed in arm 1 (7.9 eggs per gram of stool (EPG)). However, compared with 11.5 EPG in arm 2 and 15.4 EPG in arm 3, the difference lacked statistical significance. There were 4,812 first-grade children examined at baseline and 4,513 in the final survey. The overall prevalence of S. mansoni in these children slightly decreased in arms 1 (from 4.5% to 3.6%) and 2 (from 4.7% to 4.3%), but increased in arm 3 (from 6.8% to 7.9%). However, there was no significant difference in prevalence and infection intensity observed between study arms. Conclusions/significance The three treatment schedules investigated led to a reduction in the prevalence and intensity of S. mansoni infection among children aged 9–12 years. Comparing intervention arms at the end of the study, no statistically significant differences were observed between annual treatement and the other two treatment schedules, neither in reduction of prevalence nor intensity of infection. It is important to combine our results with those of three sister trials conducted simultaneously in other African countries, before final recommendations can be drawn. The World Health Organization (WHO) recommends preventive chemotherapy with praziquantel as the global strategy for morbidity control of schistosomiasis. The guidelines include target groups and treatment frequencies based on prevalence in school-age children. However, these recommendations are based on expert opinion. The Schistosomiasis Consortium for Operational Research and Evaluation (SCORE) put forward a series of cluster-randomized trials in different African countries, including Côte d’Ivoire, to identify the most suitable approach to gain and sustain the control of schistosomiasis. Results from Côte d’Ivoire did not show statistically significant differences between three school-based treatment schedules (i.e., annual treatment over four years; treatment only in the first two years, followed by two years whithout treatment; and treatment every other year without treatment in-between) in reducing prevalence and intensity of Schistosoma mansoni infection among children aged 9–12 years. The results in first-grade children with an age of 5–8 years entering school who had never received deworming drugs showed no significant difference in the prevalence and intensity of S. mansoni infection between the different treatments at the study end, suggesting that the three strategies were not significantly different for reducing the disease transmission in affected communities. However, our data should be combined with other SCORE studies carried out elsewhere in Africa. A meta-analysis including the results of the sister trials could help to conclude and make more generic recommendations.
Collapse
Affiliation(s)
- Mamadou Ouattara
- Unité de Formation et de Recherche Biosciences, Université Félix Houphouët-Boigny, Abidjan, Côte d’Ivoire
- Centre Suisse de Recherches Scientifiques en Côte d’Ivoire, Abidjan, Côte d’Ivoire
- * E-mail:
| | - Nana R. Diakité
- Unité de Formation et de Recherche Biosciences, Université Félix Houphouët-Boigny, Abidjan, Côte d’Ivoire
- Centre Suisse de Recherches Scientifiques en Côte d’Ivoire, Abidjan, Côte d’Ivoire
| | - Patrick K. Yao
- Unité de Formation et de Recherche Biosciences, Université Félix Houphouët-Boigny, Abidjan, Côte d’Ivoire
| | - Jasmina Saric
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Jean T. Coulibaly
- Unité de Formation et de Recherche Biosciences, Université Félix Houphouët-Boigny, Abidjan, Côte d’Ivoire
- Centre Suisse de Recherches Scientifiques en Côte d’Ivoire, Abidjan, Côte d’Ivoire
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Rufin K. Assaré
- Unité de Formation et de Recherche Biosciences, Université Félix Houphouët-Boigny, Abidjan, Côte d’Ivoire
- Centre Suisse de Recherches Scientifiques en Côte d’Ivoire, Abidjan, Côte d’Ivoire
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Fidèle K. Bassa
- Unité de Formation et de Recherche Biosciences, Université Félix Houphouët-Boigny, Abidjan, Côte d’Ivoire
- Centre Suisse de Recherches Scientifiques en Côte d’Ivoire, Abidjan, Côte d’Ivoire
| | - Naférima Koné
- Unité de Formation et de Recherche Biosciences, Université Félix Houphouët-Boigny, Abidjan, Côte d’Ivoire
| | - Négnorogo Guindo-Coulibaly
- Unité de Formation et de Recherche Biosciences, Université Félix Houphouët-Boigny, Abidjan, Côte d’Ivoire
| | - Jan Hattendorf
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Jürg Utzinger
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Eliézer K. N’Goran
- Unité de Formation et de Recherche Biosciences, Université Félix Houphouët-Boigny, Abidjan, Côte d’Ivoire
- Centre Suisse de Recherches Scientifiques en Côte d’Ivoire, Abidjan, Côte d’Ivoire
| |
Collapse
|
456
|
Zhang P, Wang BJ, Wang JZ, Xie XM, Tong QX. Association of CX3CL1 and CX3CR1 Expression with Liver Fibrosis in a Mouse Model of Schistosomiasis. Curr Med Sci 2021; 40:1121-1127. [PMID: 33428140 DOI: 10.1007/s11596-020-2294-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 07/13/2020] [Indexed: 10/22/2022]
Abstract
Immunopathological mechanisms of schistosomiasis, a debilitating parasitic disease, are still unclear. In this study, we investigated the involvement of CX3C chemokine ligand 1 (CX3CL1) and its sole receptor CX3CR1 in the development of liver fibrosis in schistosomiasis. The animal model of schistosomiasis was established by infection of C57BL/6 mice with Schistosoma japonicum cercariae; mice injected with carbon tetrachloride (CCl4) were used as positive control of liver injury. After 4 and 8 weeks, the degree of liver lesions was assessed by hematoxylin and eosin staining, serum levels of hyaluronic acid (HA) were analyzed by a chemiluminescence immunoassay, liver fibrosis was evaluated by immunohistochemistry analysis of α-smooth muscle actin (α-SMA) expression, and CX3CL1 and CX3CR1 expression in the liver was measured by immunohistochemistry and real-time PCR. The results showed that at 8 weeks after Schistosoma infection, serum HA levels were increased and α-SMA-expressing cells appeared in the liver, indicating fibrogenesis. CX3CL1- and CX3CR1-positive cells were observed in the outer layer of granulomas formed around Schistosoma eggs in liver tissues, which was consistent with the significant upregulation of hepatic CX3CL1 and CX3CR1 mRNA expression at 4 and 8 weeks post-infection. Furthermore, correlation analysis revealed positive association between CX3CL1 and CX3CR1 expression and serum HA levels at 8 weeks post-infection, indicating a link between fibrogenesis and the CX3CL1/CX3CR1 axis in schistosomiasis. In conclusion, our data suggest the involvement of CX3CL1 and CX3CR1 in the progression of liver fibrosis caused by Schistosoma infection.
Collapse
Affiliation(s)
- Pan Zhang
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bao-Ju Wang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jun-Zhong Wang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xu-Mao Xie
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qiao-Xiao Tong
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
457
|
Silva MP, Silva TM, Mengarda AC, Salvadori MC, Teixeira FS, Alencar SM, Luz Filho GC, Bueno-Silva B, de Moraes J. Brazilian red propolis exhibits antiparasitic properties in vitro and reduces worm burden and egg production in an mouse model harboring either early or chronic Schistosoma mansoni infection. JOURNAL OF ETHNOPHARMACOLOGY 2021; 264:113387. [PMID: 32918996 DOI: 10.1016/j.jep.2020.113387] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 08/25/2020] [Accepted: 09/04/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Propolis has been used in folk medicine for thousands of years and, in the past few decades, it has attracted renewed interest. Although propolis has been traditionally used in many communities worldwide against parasitic diseases, its effect against Schistosoma mansoni infection remains unclear. AIM OF THE STUDY To demonstrate the effects of Brazilian red propolis on Schistosoma mansoni ex vivo and in an animal model of schistosomiasis. MATERIALS AND METHODS In vitro, we monitored phenotypic and tegumental changes as well as the effects of the crude extract of propolis on pairing and egg production. In a mouse infected with either immature (early infection) or adult (chronic infection) worms, propolis was administered by oral gavage and we studied the influence of this natural product on worm burden and egg production. RESULTS Propolis 25 μg/mL reduced motility and caused 100% mortality of adult parasites ex vivo. Further analysis revealed a pronounced reduction in oviposition after exposure to propolis at sub-lethal concentrations. In addition, scanning electron microscopy showed morphological alterations in the tegument of schistosomes. In the animal model, propolis markedly reduced worm burden and egg production in both early and chronic S. mansoni infection when compared to untreated control animals. CONCLUSIONS The efficacy of Brazilian red propolis in both in vitro and in vivo studies suggests its potential anthelmintic properties against S. mansoni infection.
Collapse
Affiliation(s)
- Marcos P Silva
- Núcleo de Pesquisa Em Doenças Negligenciadas, Universidade Guarulhos, Guarulhos, SP, Brazil.
| | - Thiago M Silva
- Núcleo de Pesquisa Em Doenças Negligenciadas, Universidade Guarulhos, Guarulhos, SP, Brazil.
| | - Ana C Mengarda
- Núcleo de Pesquisa Em Doenças Negligenciadas, Universidade Guarulhos, Guarulhos, SP, Brazil.
| | - Maria C Salvadori
- Instituto de Física, Universidade de São Paulo, São Paulo, SP, Brazil.
| | | | - Severino M Alencar
- Escola Superior de Agricultura "Luiz de Queiroz", Universidade de São Paulo, São Paulo, SP, Brazil.
| | | | - Bruno Bueno-Silva
- Departamento de Odontologia, Universidade Guarulhos, Guarulhos, SP, Brazil.
| | - Josué de Moraes
- Núcleo de Pesquisa Em Doenças Negligenciadas, Universidade Guarulhos, Guarulhos, SP, Brazil.
| |
Collapse
|
458
|
Nyangiri OA, Edwige SA, Koffi M, Mewamba E, Simo G, Namulondo J, Mulindwa J, Nassuuna J, Elliott A, Karume K, Mumba D, Corstjens P, Casacuberta-Partal M, van Dam G, Bucheton B, Noyes H, Matovu E. Candidate gene family-based and case-control studies of susceptibility to high Schistosoma mansoni worm burden in African children: a protocol. AAS Open Res 2021; 4:36. [PMID: 35252746 PMCID: PMC8861467 DOI: 10.12688/aasopenres.13203.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2021] [Indexed: 11/20/2022] Open
Abstract
Background: Approximately 25% of the risk of Schistosoma mansoni is associated with host genetic variation. We will test 24 candidate genes, mainly in the T h2 and T h17 pathways, for association with S. mansoni infection intensity in four African countries, using family based and case-control approaches. Methods: Children aged 5-15 years will be recruited in S. mansoni endemic areas of Ivory Coast, Cameroon, Uganda and the Democratic Republic of Congo (DRC). We will use family based (study 1) and case-control (study 2) designs. Study 1 will take place in Ivory Coast, Cameroon, Uganda and the DRC. We aim to recruit 100 high worm burden families from each country except Uganda, where a previous study recruited at least 40 families. For phenotyping, cases will be defined as the 20% of children in each community with heaviest worm burdens as measured by the circulating cathodic antigen (CCA) assay. Study 2 will take place in Uganda. We will recruit 500 children in a highly endemic community. For phenotyping, cases will be defined as the 20% of children with heaviest worm burdens as measured by the CAA assay, while controls will be the 20% of infected children with the lightest worm burdens. Deoxyribonucleic acid (DNA) will be genotyped on the Illumina H3Africa SNP (single nucleotide polymorphisms) chip and genotypes will be converted to sets of haplotypes that span the gene region for analysis. We have selected 24 genes for genotyping that are mainly in the Th2 and Th17 pathways and that have variants that have been demonstrated to be or could be associated with Schistosoma infection intensity. Analysis: In the family-based design, we will identify SNP haplotypes disproportionately transmitted to children with high worm burden. Case-control analysis will detect overrepresentation of haplotypes in extreme phenotypes with correction for relatedness by using whole genome principal components.
Collapse
Affiliation(s)
- Oscar A. Nyangiri
- College of Veterinary Medicine, Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Sokouri A. Edwige
- Université Jean Lorougnon Guédé (UJLoG) de Daloa, Daloa, Cote d'Ivoire
| | - Mathurin Koffi
- Université Jean Lorougnon Guédé (UJLoG) de Daloa, Daloa, Cote d'Ivoire
| | - Estelle Mewamba
- Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Gustave Simo
- Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Joyce Namulondo
- College of Veterinary Medicine, Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Julius Mulindwa
- College of Veterinary Medicine, Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Jacent Nassuuna
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Alison Elliott
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- London School of Hygiene and Tropical Medicine, London, WC1E, UK
| | - Kévin Karume
- Institut National de Recherche Biomedicale, Kinshasa, Democratic Republic of the Congo
| | - Dieudonne Mumba
- Institut National de Recherche Biomedicale, Kinshasa, Democratic Republic of the Congo
| | - P.L.A.M Corstjens
- Dept. of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - G.J. van Dam
- Dept. of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bruno Bucheton
- Institut de Recherche pour le Développement (IRD), IRD-CIRAD, Montpellier, France
| | - Harry Noyes
- Centre for Genomic Research, University of Liverpool, Liverpool, UK
| | - Enock Matovu
- College of Veterinary Medicine, Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | | |
Collapse
|
459
|
Nyangiri OA, Edwige SA, Koffi M, Mewamba E, Simo G, Namulondo J, Mulindwa J, Nassuuna J, Elliott A, Karume K, Mumba D, Corstjens P, Casacuberta-Partal M, van Dam G, Bucheton B, Noyes H, Matovu E. Candidate gene family-based and case-control studies of susceptibility to high Schistosoma mansoni worm burden in African children: a protocol. AAS Open Res 2021; 4:36. [PMID: 35252746 PMCID: PMC8861467 DOI: 10.12688/aasopenres.13203.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2021] [Indexed: 11/20/2022] Open
Abstract
Background: Approximately 25% of the risk of Schistosoma mansoni is associated with host genetic variation. We will test 24 candidate genes, mainly in the T h2 and T h17 pathways, for association with S. mansoni infection intensity in four African countries, using family based and case-control approaches. Methods: Children aged 5-15 years will be recruited in S. mansoni endemic areas of Ivory Coast, Cameroon, Uganda and the Democratic Republic of Congo (DRC). We will use family based (study 1) and case-control (study 2) designs. Study 1 will take place in Ivory Coast, Cameroon, Uganda and the DRC. We aim to recruit 100 high worm burden families from each country except Uganda, where a previous study recruited at least 40 families. For phenotyping, cases will be defined as the 20% of children in each community with heaviest worm burdens as measured by the circulating cathodic antigen (CCA) assay. Study 2 will take place in Uganda. We will recruit 500 children in a highly endemic community. For phenotyping, cases will be defined as the 20% of children with heaviest worm burdens as measured by the CAA assay, while controls will be the 20% of infected children with the lightest worm burdens. Deoxyribonucleic acid (DNA) will be genotyped on the Illumina H3Africa SNP (single nucleotide polymorphisms) chip and genotypes will be converted to sets of haplotypes that span the gene region for analysis. We have selected 24 genes for genotyping that are mainly in the Th2 and Th17 pathways and that have variants that have been demonstrated to be or could be associated with Schistosoma infection intensity. Analysis: In the family-based design, we will identify SNP haplotypes disproportionately transmitted to children with high worm burden. Case-control analysis will detect overrepresentation of haplotypes in extreme phenotypes with correction for relatedness by using whole genome principal components.
Collapse
Affiliation(s)
- Oscar A. Nyangiri
- College of Veterinary Medicine, Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Sokouri A. Edwige
- Université Jean Lorougnon Guédé (UJLoG) de Daloa, Daloa, Cote d'Ivoire
| | - Mathurin Koffi
- Université Jean Lorougnon Guédé (UJLoG) de Daloa, Daloa, Cote d'Ivoire
| | - Estelle Mewamba
- Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Gustave Simo
- Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Joyce Namulondo
- College of Veterinary Medicine, Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Julius Mulindwa
- College of Veterinary Medicine, Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Jacent Nassuuna
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Alison Elliott
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- London School of Hygiene and Tropical Medicine, London, WC1E, UK
| | - Kévin Karume
- Institut National de Recherche Biomedicale, Kinshasa, Democratic Republic of the Congo
| | - Dieudonne Mumba
- Institut National de Recherche Biomedicale, Kinshasa, Democratic Republic of the Congo
| | - P.L.A.M Corstjens
- Dept. of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - G.J. van Dam
- Dept. of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bruno Bucheton
- Institut de Recherche pour le Développement (IRD), IRD-CIRAD, Montpellier, France
| | - Harry Noyes
- Centre for Genomic Research, University of Liverpool, Liverpool, UK
| | - Enock Matovu
- College of Veterinary Medicine, Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | | |
Collapse
|
460
|
Sessa DP, Mengarda AC, Simplicio PE, Antar GM, Lago JHG, de Moraes J. 15β-Senecioyl-oxy- ent-kaur-16-en-19-oic Acid, a Diterpene Isolated from Baccharis lateralis, as Promising Oral Compound for the Treatment of Schistosomiasis. JOURNAL OF NATURAL PRODUCTS 2020; 83:3744-3750. [PMID: 33236902 DOI: 10.1021/acs.jnatprod.0c01050] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Praziquantel is the only available drug to treat schistosomiasis, and therefore, urgent studies must be performed to identify new anthelmintic agents. This study reports the anthelmintic evaluation of two related ent-kaurane diterpenes isolated from aerial parts of Baccharis lateralis (Asteraceae), ent-kaur-16-en-19-oic acid (1) and 15β-senecioyl-oxy-ent-kaur-16-en-19-oic acid (2) against Schistosoma mansoni in vitro and in a murine model of schistosomiasis. Both compounds exhibited in vitro activity with lethal concentration 50% (LC50) values of 26.1 μM (1) and 11.6 μM (2) as well as reduced toxicity against human cell lines, revealing a good selectivity profile, mainly with compound 2 (selectivity index > 10). Compound 2 also decreased egg production and caused morphological alterations in the parasite reproductive system. In mice infected with S. mansoni, oral treatment with compound 2 at 400 mg/kg, the standard dose used in this model of schistosomiasis, caused a significant reduction in a total worm burden of 61.9% (P < 0.01). S. mansoni egg production, a key mechanism for both transmission and pathogenesis, was also markedly reduced. In addition, compound 2 achieved a significant reduction in hepatosplenomegaly. Therefore, the diterpene 15β-senecioyl-oxy-ent-kaur-16-en-19-oic acid (2) has an acceptable cytotoxicity profile and is orally active in a murine schistosomiasis model.
Collapse
Affiliation(s)
- Deborah P Sessa
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, São Paulo 09210-180, Brazil
| | - Ana C Mengarda
- Núcleo de Pesquisa em Doenças Negligenciadas, Universidade Guarulhos, Guarulhos, São Paulo 07023-070, Brazil
| | - Paula E Simplicio
- Núcleo de Pesquisa em Doenças Negligenciadas, Universidade Guarulhos, Guarulhos, São Paulo 07023-070, Brazil
| | - Guilherme M Antar
- Departamento de Botânica, Instituto de Biociências, Universidade de São Paulo, São Paulo 05508-090, Brazil
| | - João Henrique G Lago
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, São Paulo 09210-180, Brazil
| | - Josué de Moraes
- Núcleo de Pesquisa em Doenças Negligenciadas, Universidade Guarulhos, Guarulhos, São Paulo 07023-070, Brazil
| |
Collapse
|
461
|
Lei Z, Tang R, Qi Q, Gu P, Wang J, Xu L, Wei C, Pu Y, Qi X, Chen Y, Yu B, Yu Y, Chen X, Zhu J, Li Y, Zhou S, Su C. Hepatocyte CD1d protects against liver immunopathology in mice with schistosomiasis japonica. Immunology 2020; 162:328-338. [PMID: 33283278 DOI: 10.1111/imm.13288] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/17/2020] [Accepted: 11/21/2020] [Indexed: 12/16/2022] Open
Abstract
Schistosomiasis is a neglected tropical disease with over 250 million people infected worldwide. The main clinically important species Schistosoma mansoni (S. mansoni) and Schistosoma japonicum (S. japonicum) cause inflammatory responses against tissue-trapped eggs, resulting in formation of granulomas mainly in host liver. Persistent granulomatous response results in severe fibrosis in the liver, leading to irreversible impairment of the liver and even death of the host. CD1d, a highly conserved MHC class I-like molecule, is expressed by both haematopoietic and non-haematopoietic cells. CD1d on antigen-presenting cells (APCs) of haematopoietic origin presents pathogen-derived lipid antigens to natural killer T (NKT) cells, which enables them to rapidly produce large amounts of various cytokines and facilitate CD4+ T helper (Th) cell differentiation upon invading pathogens. Noteworthy, hepatocytes of non-haematopoietic origin have recently been shown to be involved in maintaining liver NKT cell homeostasis through a CD1d-dependent manner. However, whether hepatocyte CD1d-dependent regulation of NKT cell homeostasis also modulates CD4+ Th cell responses and liver immunopathology in murine schistosomiasis remains to be addressed. Here, we show in mice that CD1d expression on hepatocytes was decreased dramatically upon S. japonicum infection, accompanied by increased NKT cells, as well as upregulated Th1 and Th2 responses. Overexpression of CD1d in hepatocytes significantly decreased local NKT numbers and cytokines (IFN-γ, IL-4, IL-13), concomitantly with downregulation of both Th1 and Th2 responses and alleviation in pathological damage in livers of S. japonicum-infected mice. These findings highlight the potential of hepatocyte CD1d-targeted therapies for liver immunopathology control in schistosomiasis.
Collapse
Affiliation(s)
- Zhigang Lei
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rui Tang
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qianqian Qi
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Pan Gu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junling Wang
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Xu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chuan Wei
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yanan Pu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xin Qi
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ying Chen
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Beibei Yu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yanxiong Yu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaojun Chen
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jifeng Zhu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yalin Li
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Sha Zhou
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chuan Su
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
462
|
Loss of natural resistance to schistosome in T cell deficient rat. PLoS Negl Trop Dis 2020; 14:e0008909. [PMID: 33347431 PMCID: PMC7785244 DOI: 10.1371/journal.pntd.0008909] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 01/05/2021] [Accepted: 10/21/2020] [Indexed: 01/08/2023] Open
Abstract
Schistosomiasis is among the major neglected tropical diseases and effective prevention by boosting the immune system is still not available. T cells are key cellular components governing adaptive immune response to various infections. While common laboratory mice, such as C57BL/6, are highly susceptible to schistosomiasis, the SD rats are extremely resistant. However, whether adaptive immunity is necessary for such natural resistance to schistosomiasis in rats remains to be determined. Therefore, it is necessary to establish genetic model deficient in T cells and adaptive immunity on the resistant SD background, and to characterize liver pathology during schistosomiasis. In this study we compared experimental schistosomiasis in highly susceptible C57BL/6 (B6) mice and in resistant SD rats, using cercariae of Schistosoma japonicum. We observed a marked T cell expansion in the spleen of infected B6 mice, but not resistant SD rats. Interestingly, CD3e−/− B6 mice in which T cells are completely absent, the infectious burden of adult worms was significantly higher than that in WT mice, suggesting an anti-parasitic role for T cells in B6 mice during schistosome infection. In further experiments, we established Lck deficient SD rats by using CRISPR/Cas9 in which T cell development was completely abolished. Strikingly, we found that such Lck deficiency in SD rats severely impaired their natural resistance to schistosome infection, and fostered parasite growth. Together with an additional genetic model deficient in T cells, the CD3e−/− SD rats, we confirmed the absence of T cell resulted in loss of natural resistance to schistosome infection, but also mitigated liver immunopathology. Our further experiments showed that regulatory T cell differentiation in infected SD rats was significantly decreased during schistosomiasis, in contrast to significant increase of regulatory T cells in infected B6 mice. These data suggest that T cell mediated immune tolerance facilitates persistent infection in mice but not in SD rats. The demonstration of an important role for T cells in natural resistance of SD rats to schistosomiasis provides experimental evidences supporting the rationale to boost T cell responses in humans to prevent and treat schistosomiasis. Schistosomiasis is among the major neglected tropical diseases and affects mainly the developing countries. Although the role of the immune system in driving immunopathology in schistosomiasis has been extensively studied, how adaptive immunity contributes to disease resistance during schistosome infection is still not completely understood. Most livestock species as well as humans are susceptible to schistosomiasis, while some mammals are extremely resistant. The common laboratory C57BL/6 mice are highly susceptible to schistosomiasis; however, the SD rats are extremely resistant. In this study, we first used T cell deficient CD3e−/− C57BL/6 mice and experimental Schistosoma japonicum infection and further established novel T cell deficient models in SD rats to assess anti-parasite roles of T cells. Strikingly, we found that the natural resistance of SD rat to schistosomiasis was abolished in the absence of T cells, despite the fact that the liver pathology was mitigated following infection. Therefore, our study presented experimental support for the rationale to boost T cell function for clearance of schistosome parasites.
Collapse
|
463
|
Maggi L, Rocha IC, Camelo GMA, Fernandes VR, Negrão-Corrêa D. The IL-33/ST2 pathway is not essential to Th2 stimulation but is key for modulation and survival during chronic infection with Schistosoma mansoni in mice. Cytokine 2020; 138:155390. [PMID: 33341001 DOI: 10.1016/j.cyto.2020.155390] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 11/11/2020] [Accepted: 11/25/2020] [Indexed: 12/30/2022]
Abstract
Morbidity during chronic schistosomiasis has been associated with the induction and modulation of type-2 granulomatous inflammatory response induced by antigens secreted by the eggs, which become trapped in capillary venules of the host tissues, especially in the liver and intestines. IL-33, an alarmin released after cell damage, binds to its ST2 (suppressor of tumorigenicity 2) receptor, expressed in an variety of immune cells, including ILC2 and macrophages, and stimulates the early production of IL-5 and IL-13, which leads to eosinophil infiltration and activation of a Th2 response. However, the role of IL-33/ST2 activation on Schistosoma-induced granuloma formation and modulation is mostly unknown. In the current work, we comparatively evaluated the immune response and granuloma formation in wild-type BALB/c (WT) and BALB/c mice genetically deficient in the IL-33 receptor (ST2-/-) experimentally infected with Schistosoma mansoni. Mice were infected with 25 or 50 S. mansoni cercariae and followed for up to 14 weeks to assess mortality. Mice from each experimental group were comparatively evaluated for parasite burden, liver immune response, and granuloma appearance during acute and chronic schistosomiasis. Our data showed that the number of circulating worms and eggs retained in the liver and eliminated in the feces was similar in WT and ST2-/- infected mice, but infected ST2-/- mice presented an enhanced rate of mortality. Interestingly, the production of type-2 cytokines by soluble egg antigens (SEA)-stimulated spleen cells, the serum concentrations of IL-5 and Immunoglobulin (Ig)-E, and the level of parasite-reactive IgG1 were similar in infected mice of both experimental groups. The concentrations of IL-4, IL-5, IL-13, and IFN-γ in liver homogenate of infected mice also did not differ between the strains at acute schistosomiasis, but there was a significant increase in IL-17 levels in ST2-/- infected mice at this phase. On the other hand, IL-4, IL-13, IL-10, IL-17, and IFN-γ concentrations were reduced and the ratios of IL-4/IFN-γ and IL-17/IFN-γ were higher in liver homogenate of chronically infected ST2-/- mice, suggesting unbalanced Th2 and Th17 responses. Moreover, liver granulomas of ST2-/- mice were larger and disorganized, showing an intense cellular infiltrate, rich in eosinophils and neutrophils. Our results suggest that the absence of the IL-33/ST2 pathway is not essential for the Schistosoma-induced Th2 response, but is necessary to prevent host mortality by modulating granuloma-mediated pathology.
Collapse
Affiliation(s)
- Laura Maggi
- Laboratório de Esquistossomose e Imunohelmintologia, Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Izabella Chrystina Rocha
- Laboratório de Esquistossomose e Imunohelmintologia, Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Curso de Enfermagem, Instituto de Ciências Biológicas e Saúde, Universidade Federal de Mato Grosso, Barra do Garça, Brazil
| | - Genil Mororó Araújo Camelo
- Laboratório de Esquistossomose e Imunohelmintologia, Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vanessa Rodrigues Fernandes
- Laboratório de Esquistossomose e Imunohelmintologia, Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Deborah Negrão-Corrêa
- Laboratório de Esquistossomose e Imunohelmintologia, Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
464
|
Osakunor DNM, Mduluza T, Osei-Hyiaman D, Burgess K, Woolhouse MEJ, Mutapi F. Schistosoma haematobium infection is associated with alterations in energy and purine-related metabolism in preschool-aged children. PLoS Negl Trop Dis 2020; 14:e0008866. [PMID: 33315875 PMCID: PMC7735607 DOI: 10.1371/journal.pntd.0008866] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/08/2020] [Indexed: 12/17/2022] Open
Abstract
Helminths are parasitic worms that infect over a billion people worldwide. The pathological consequences from infection are due in part, to parasite-induced changes in host metabolic pathways. Here, we analyse the changes in host metabolic profiles, in response to the first Schistosoma haematobium infection and treatment in Zimbabwean children. A cohort of 83 schistosome-negative children (2-5 years old) as determined by parasitological examination, guardian interviews and examination of medical records, was recruited at baseline. Children were followed up after three months for parasitological diagnosis of their first S. haematobium infection, by detection of parasite eggs excreted in urine. Children positive for infection were treated with the antihelminthic drug praziquantel, and treatment efficacy checked three months after treatment. Blood samples were taken at each time point, and capillary electrophoresis mass spectrometry in conjunction with multivariate analysis were used to compare the change in serum metabolite profiles in schistosome-infected versus uninfected children. Following baseline at the three-month follow up, 11 children had become infected with S. haematobium (incidence = 13.3%). Our results showed that infection with S. haematobium was associated with significant increases (>2-fold) in discriminatory metabolites, linked primarily with energy (G6P, 3-PG, AMP, ADP) and purine (AMP, ADP) metabolism. These observed changes were commensurate with schistosome infection intensity, and levels of the affected metabolites were reduced following treatment, albeit not significantly. This study demonstrates that early infection with S. haematobium is associated with alterations in host energy and purine metabolism. Taken together, these changes are consistent with parasite-related clinical manifestations of malnutrition, poor growth and poor physical and cognitive performance observed in schistosome-infected children.
Collapse
Affiliation(s)
- Derick N. M. Osakunor
- Institute of Immunology & Infection Research, University of Edinburgh, Ashworth Laboratories, King’s Buildings, Edinburgh, United Kingdom
- * E-mail:
| | - Takafira Mduluza
- Biochemistry Department, University of Zimbabwe, Mount Pleasant, Harare, Zimbabwe
| | - Douglas Osei-Hyiaman
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States of America
- Metabolomics Research Division, Human Metabolome Technologies Inc., Tsuruoka, Yamagata, Japan
- Department of Systems Neurophysiology, Graduate School of Medical & Dental Science, Tokyo Medical and Dental University, Bunkyo City, Tokyo, Japan
| | - Karl Burgess
- Centre for Synthetic and Systems Biology, University of Edinburgh, CH Waddington Building, King’s Buildings, Edinburgh, United Kingdom
| | - Mark E. J. Woolhouse
- Usher Institute, University of Edinburgh, Ashworth Laboratories, King’s Buildings, Edinburgh, United Kingdom
- NIHR Global Health Research Unit Tackling Infections to Benefit Africa (TIBA), University of Edinburgh, Ashworth Laboratories, King’s Buildings, Edinburgh, United Kingdom
| | - Francisca Mutapi
- Institute of Immunology & Infection Research, University of Edinburgh, Ashworth Laboratories, King’s Buildings, Edinburgh, United Kingdom
- NIHR Global Health Research Unit Tackling Infections to Benefit Africa (TIBA), University of Edinburgh, Ashworth Laboratories, King’s Buildings, Edinburgh, United Kingdom
| |
Collapse
|
465
|
Paul S, Ruiz-Manriquez LM, Serrano-Cano FI, Estrada-Meza C, Solorio-Diaz KA, Srivastava A. Human microRNAs in host-parasite interaction: a review. 3 Biotech 2020; 10:510. [PMID: 33178551 PMCID: PMC7644590 DOI: 10.1007/s13205-020-02498-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are a group of small noncoding RNA molecules with significant capacity to regulate the gene expression at the post-transcriptional level in a sequence-specific manner either through translation repression or mRNA degradation triggering a fine-tuning biological impact. They have been implicated in several processes, including cell growth and development, signal transduction, cell proliferation and differentiation, metabolism, apoptosis, inflammation, and immune response modulation. However, over the last few years, extensive studies have shown the relevance of miRNAs in human pathophysiology. Common human parasitic diseases, such as Malaria, Leishmaniasis, Amoebiasis, Chagas disease, Schistosomiasis, Toxoplasmosis, Cryptosporidiosis, Clonorchiasis, and Echinococcosis are the leading cause of death worldwide. Thus, identifying and characterizing parasite-specific miRNAs and their host targets, as well as host-related miRNAs, are important for a deeper understanding of the pathophysiology of parasite-specific diseases at the molecular level. In this review, we have demonstrated the impact of human microRNAs during host-parasite interaction as well as their potential to be used for diagnosis and prognosis purposes.
Collapse
Affiliation(s)
- Sujay Paul
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130 Querétaro, Mexico
| | - Luis M. Ruiz-Manriquez
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130 Querétaro, Mexico
| | - Francisco I. Serrano-Cano
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130 Querétaro, Mexico
| | - Carolina Estrada-Meza
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130 Querétaro, Mexico
| | - Karla A. Solorio-Diaz
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130 Querétaro, Mexico
| | - Aashish Srivastava
- Section of Bioinformatics, Clinical Laboratory, Haukeland University Hospital, 5021 Bergen, Norway
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
| |
Collapse
|
466
|
Kouadio JN, Giovanoli Evack J, Achi LY, Fritsche D, Ouattara M, Silué KD, Bonfoh B, Hattendorf J, Utzinger J, Zinsstag J, Balmer O, N'Goran EK. Prevalence and distribution of livestock schistosomiasis and fascioliasis in Côte d'Ivoire: results from a cross-sectional survey. BMC Vet Res 2020; 16:446. [PMID: 33203412 PMCID: PMC7672978 DOI: 10.1186/s12917-020-02667-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 11/04/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Schistosoma and Fasciola are zoonotic parasites of public health and veterinary importance. However, while the epidemiology of schistosomiasis in humans is well studied, little is known about fascioliasis and schistosomiasis in livestock in Côte d'Ivoire. This study aimed to determine the prevalence and the distribution of livestock schistosomiasis and fascioliasis across Côte d'Ivoire. In 2018, we conducted a cross-sectional survey in abattoirs and farms in 13 departments of Côte d'Ivoire. In abattoirs, the mesenteric veins and livers of slaughtered cattle, sheep and goats were examined for adult Schistosoma and Fasciola flukes. Faeces from live cattle, goats and sheep were collected and examined for Schistosoma and Fasciola eggs using a sedimentation technique. RESULTS A total of 386 cattle, 174 goats and 151 sheep from abattoirs and 435 cattle, 22 goats and 176 sheep from farms were sampled. The observed prevalence of schistosomiasis was higher in slaughtered animals. Fascioliasis was more prevalent in farm animals. The prevalence of schistosomiasis in slaughtered cattle varied between 5.9% (95% confidence interval (CI): 0.7-19.7%) and 53.3% (95% CI: 37.9-68.3%) with the highest prevalence observed in Ouangolodougou in the North. Cattle from farms had a relatively low prevalence of schistosomiasis, with the highest prevalence found in Ouangolodougou (2.4%, 95% CI: 0.7-6.1%). The prevalence of fascioliasis varied considerably from one department to another, ranging from nil (95% CI: 0.0-18.5%) to 50.8% (95% CI: 43.4-58.2%), with the highest prevalence found in farm cattle in Dikodougou in the North. Sheep and goats had a lower prevalence of schistosomiasis and fascioliasis than cattle. In slaughtered animals, cattle aged 4 years and older were at highest risk for schistosomiasis (odds ratio (OR): 2.4; 95% CI: 1.0-5.6) and fascioliasis (OR: 2.1; 95% CI: 1.1-3.9). In farm animals, male cattle had higher odds of being infected with Schistosoma (OR: 4.3; 95% CI: 0.7-26.9) than females. CONCLUSIONS Our study confirms that schistosomiasis and fascioliasis are endemic in livestock across Côte d'Ivoire. A strategic control programme should be considered, especially for cattle, including providing drinking water in troughs to reduce faecal contamination of water sources by cattle.
Collapse
Affiliation(s)
- Jules N Kouadio
- Unité de Formation et de Recherche Biosciences, Université Félix Houphouët-Boigny, 22 BP 770 Abidjan 22, Abidjan, Côte d'Ivoire. .,Centre Suisse de Recherches Scientifiques en Côte d'Ivoire, 01 BP 1303 Abidjan 01, Abidjan, Côte d'Ivoire. .,Swiss Tropical and Public Health Institute, P.O. Box CH-4002, Basel, Switzerland. .,University of Basel, P.O. Box CH-4001, Basel, Switzerland.
| | - Jennifer Giovanoli Evack
- Swiss Tropical and Public Health Institute, P.O. Box CH-4002, Basel, Switzerland.,University of Basel, P.O. Box CH-4001, Basel, Switzerland
| | - Louise Y Achi
- Centre Suisse de Recherches Scientifiques en Côte d'Ivoire, 01 BP 1303 Abidjan 01, Abidjan, Côte d'Ivoire.,Ecole de Spécialisation en Elevage et des Métiers de la Viande de Bingerville, BP 58 Bingerville, Abidjan, Côte d'Ivoire
| | - Dominik Fritsche
- University of Zurich, Ramistrasse 71, P.O. Box CH-8006, Zurich, Switzerland
| | - Mamadou Ouattara
- Unité de Formation et de Recherche Biosciences, Université Félix Houphouët-Boigny, 22 BP 770 Abidjan 22, Abidjan, Côte d'Ivoire.,Centre Suisse de Recherches Scientifiques en Côte d'Ivoire, 01 BP 1303 Abidjan 01, Abidjan, Côte d'Ivoire
| | - Kigbafori D Silué
- Unité de Formation et de Recherche Biosciences, Université Félix Houphouët-Boigny, 22 BP 770 Abidjan 22, Abidjan, Côte d'Ivoire.,Centre Suisse de Recherches Scientifiques en Côte d'Ivoire, 01 BP 1303 Abidjan 01, Abidjan, Côte d'Ivoire
| | - Bassirou Bonfoh
- Centre Suisse de Recherches Scientifiques en Côte d'Ivoire, 01 BP 1303 Abidjan 01, Abidjan, Côte d'Ivoire
| | - Jan Hattendorf
- Swiss Tropical and Public Health Institute, P.O. Box CH-4002, Basel, Switzerland.,University of Basel, P.O. Box CH-4001, Basel, Switzerland
| | - Jürg Utzinger
- Swiss Tropical and Public Health Institute, P.O. Box CH-4002, Basel, Switzerland.,University of Basel, P.O. Box CH-4001, Basel, Switzerland
| | - Jakob Zinsstag
- Swiss Tropical and Public Health Institute, P.O. Box CH-4002, Basel, Switzerland.,University of Basel, P.O. Box CH-4001, Basel, Switzerland
| | - Oliver Balmer
- Swiss Tropical and Public Health Institute, P.O. Box CH-4002, Basel, Switzerland.,University of Basel, P.O. Box CH-4001, Basel, Switzerland
| | - Eliézer K N'Goran
- Unité de Formation et de Recherche Biosciences, Université Félix Houphouët-Boigny, 22 BP 770 Abidjan 22, Abidjan, Côte d'Ivoire.,Centre Suisse de Recherches Scientifiques en Côte d'Ivoire, 01 BP 1303 Abidjan 01, Abidjan, Côte d'Ivoire
| |
Collapse
|
467
|
Perera DJ, Hassan AS, Jia Y, Ricciardi A, McCluskie MJ, Weeratna RD, Ndao M. Adjuvanted Schistosoma mansoni-Cathepsin B With Sulfated Lactosyl Archaeol Archaeosomes or AddaVax™ Provides Protection in a Pre-Clinical Schistosomiasis Model. Front Immunol 2020; 11:605288. [PMID: 33304354 PMCID: PMC7701121 DOI: 10.3389/fimmu.2020.605288] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 10/20/2020] [Indexed: 12/15/2022] Open
Abstract
Schistosomiasis threatens 800 million people worldwide. Chronic pathology manifests as hepatosplenomegaly, and intestinal schistosomiasis caused by Schistosoma mansoni can lead to liver fibrosis, cirrhosis, and blood in the stool. To assist the only FDA-approved drug, praziquantel, in parasite elimination, the development of a vaccine would be of high value. S. mansoni Cathepsin B (SmCB) is a well-documented vaccine target for intestinal schistosomiasis. Herein, we test the increased efficacy and immunogenicity of SmCB when combined with sulfated lactosyl archaeol (SLA) archaeosomes or AddaVax™ (a squalene based oil-in-water emulsion). Both vaccine formulations resulted in robust humoral and cell mediated immune responses. Impressively, both formulations were able to reduce parasite burden greater than 40% (WHO standard), with AddaVax™ reaching 86.8%. Additionally, SmCB with both adjuvants were able to reduce granuloma size and the amount of larval parasite hatched from feces, which would reduce transmission. Our data support SmCB as a target for S. mansoni vaccination; especially when used in an adjuvanted formulation.
Collapse
Affiliation(s)
- Dilhan J Perera
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, Canada.,Infectious Diseases and Immunity in Global Health Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Adam S Hassan
- Infectious Diseases and Immunity in Global Health Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Yimei Jia
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Alessandra Ricciardi
- Infectious Diseases and Immunity in Global Health Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Michael J McCluskie
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Risini D Weeratna
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Momar Ndao
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, Canada.,Infectious Diseases and Immunity in Global Health Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada.,National Reference Center for Parasitology, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
468
|
Zhang B, Wu X, Song Q, Ning A, Liang J, Song L, Liu J, Zhang Y, Yuan D, Sun X, Wu Z. Gut Microbiota Modulates Intestinal Pathological Injury in Schistosoma japonicum-Infected Mice. Front Med (Lausanne) 2020; 7:588928. [PMID: 33313045 PMCID: PMC7703745 DOI: 10.3389/fmed.2020.588928] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
Trapping of Schistosoma japonicum (S. japonicum) eggs in host tissue, mainly in the intestine and liver, causes severe gastrointestinal and hepatic granulomatous immune responses and irreversible fibrosis. Although the gut microbiota plays a central role in regulating pathological responses in several diseases, the effect of the gut microbiota on the pathologenesis progression of schistosomiasis remains largely unknown. In this study, we aimed to investigate the regulatory function of the gut microbiota in schistosomiasis japonica. We found that the depletion of the gut microbiota significantly ameliorated egg granulomas formation and fibrosis in the intestine of infected mice. This role of the gut microbiota in intestinal granuloma formation and fibrosis was reinforced when normal and infected mice were housed together in one cage. Notably, changes in the gut microbiota induced by S. japonicum infection were partly reversible with microbiota transfer in the cohousing experiment. Transfer of the gut microbiota from normal to infected mice attenuated the intestinal pathological responses. Depletion of the gut microbiota by antibiotics, or transfer of the gut microbiota from normal to infected mice decreased the levels of IL-4, IL-5, and IL-13 and promoted the production of cytokines and mRNA levels of IL-10 and TGF-β in infected mice. Our findings indicated a regulatory effect of the gut microbiota on intestinal pathological injury associated with schistosomiasis japonica in mice, and thus suggested a potential strategy for schistosomiasis treatment.
Collapse
Affiliation(s)
- Beibei Zhang
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Sun Yat-sen University, Guangzhou, China.,Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China.,Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, China
| | - Xiaoying Wu
- Department of Gastroenterology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qiuyue Song
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Sun Yat-sen University, Guangzhou, China
| | - An Ning
- Jiangxi Provincial Institute of Parasitic Diseases, Nanchang, China
| | - Jinyi Liang
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Sun Yat-sen University, Guangzhou, China
| | - Langui Song
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Sun Yat-sen University, Guangzhou, China
| | - Jiahua Liu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Sun Yat-sen University, Guangzhou, China
| | - Yishu Zhang
- College of Basic Medical Sciences, Guilin Medical University, Guilin, China
| | - Dongjuan Yuan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xi Sun
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Sun Yat-sen University, Guangzhou, China
| | - Zhongdao Wu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
469
|
Hillyer JF. Parasites and Parasitology in this SARS-CoV-2, COVID-19 World: An American Society of Parasitologists Presidential Address. J Parasitol 2020; 106:859-868. [PMID: 33450760 DOI: 10.1645/20-158] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The novel coronavirus disease 2019 (COVID-19) is one of the worst global health crises of this generation. The core of this pandemic is the rapid transmissibility of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus, its high morbidity and mortality, and the presence of infectious asymptomatic carriers. As a result, COVID-19 has dominated this year's headlines and commanded significant research attention. As we consider SARS-CoV-2 and the COVID-19 pandemic, it is essential that scientists, governments, the media, and the general population also come to grips with the everyday cost of parasitic diseases. Plasmodium (malaria), schistosomes, filarial worms, hookworms, Ascaris, whipworms, and other protozoan and metazoan parasites take a tremendous toll on local communities. Yet, because most of these diseases are no longer endemic to developed countries, their research and intervention are not funded at levels that are proportional to their global morbidity and mortality. The scientific and public health communities must indeed vigorously fight SARS-CoV-2 and COVID-19, but while doing so and beyond, it will be essential to demonstrate steadfast resolve toward understanding and combating the parasitic diseases that for centuries have haunted humankind.
Collapse
Affiliation(s)
- Julián F Hillyer
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee 37235
| |
Collapse
|
470
|
Du X, McManus DP, French JD, Jones MK, You H. CRISPR/Cas9: A new tool for the study and control of helminth parasites. Bioessays 2020; 43:e2000185. [PMID: 33145822 DOI: 10.1002/bies.202000185] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 12/17/2022]
Abstract
Recent reports of CRISPR/Cas9 genome editing in parasitic helminths open up new avenues for research on these dangerous pathogens. However, the complex morphology and life cycles inherent to these parasites present obstacles for the efficient application of CRISPR/Cas9-targeted mutagenesis. This is especially true with the trematode flukes where only modest levels of gene mutation efficiency have been achieved. Current major challenges in the application of CRISPR/Cas9 for study of parasitic worms thus lie in enhancing gene mutation efficiency and overcoming issues involved in host passage so that mutated parasites survive. Strategies developed for CRISPR/Cas9 studies on Caenorhabditis elegans, protozoa and mammalian cells, including novel delivery methods, the choice of selectable markers, and refining mutation precision represent novel tactics whereby these impediments can be overcome. Furthermore, employing CRISPR/Cas9-mediated gene drive to interfere with vector transmission represents a novel approach for the control of parasitic worms that is worthy of further exploration.
Collapse
Affiliation(s)
- Xiaofeng Du
- Immunology Department, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Herston, Brisbane, Queensland, Australia
| | - Donald P McManus
- Immunology Department, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Herston, Brisbane, Queensland, Australia
| | - Juliet D French
- Genetics & Computational Biology Department, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | - Malcolm K Jones
- School of Veterinary Science, The University of Queensland, Gatton, Queensland, Australia
| | - Hong You
- Immunology Department, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| |
Collapse
|
471
|
Molehin AJ. Current Understanding of Immunity Against Schistosomiasis: Impact on Vaccine and Drug Development. Res Rep Trop Med 2020; 11:119-128. [PMID: 33173371 PMCID: PMC7646453 DOI: 10.2147/rrtm.s274518] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 09/29/2020] [Indexed: 12/17/2022] Open
Abstract
Schistosomiasis is a neglected tropical disease inflicting significant morbidity in humans worldwide. The disease is caused by infections with a parasitic trematode belonging to the genus Schistosoma. Over 250 million people are currently infected globally, with an estimated disability-adjusted life-years of 1.9 million attributed to the disease. Current understanding, based on several immunological studies using experimental and human models of schistosomiasis, reveals that complex immune mechanisms play off each other in the acquisition of immune resistance to infection/reinfection. Nevertheless, the precise characteristics of these responses, the specific antigens against which they are elicited, and how these responses are intricately regulated are still being investigated. What is apparent is that immunity to schistosome infections develops slowly and over a prolonged period of time, augmented by the death of adult worms occurring naturally or by praziquantel therapy. In this review, aspects of immunity to schistosomiasis, host–parasite interactions and their impact on schistosomiasis vaccine development are discussed.
Collapse
Affiliation(s)
- Adebayo J Molehin
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.,Center for Tropical Medicine and Infectious Diseases, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
472
|
Combined transcriptomics and proteomics to identify differential proteins involved in the immune response to the parasite schistosoma japonicum in snail hosts pre-infected with exorchis sp. Acta Trop 2020; 211:105623. [PMID: 32645302 DOI: 10.1016/j.actatropica.2020.105623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 06/28/2020] [Accepted: 07/06/2020] [Indexed: 01/01/2023]
Abstract
Oncomelania hupensis is the obligate intermediate host of Schistosoma japonicum, and it also serves as the first intermediate host for Exorchis sp., which uses Parasilurus asoyus as its definitive host rather than humans. In previous studies, Tang et al. found that all S. japonicum larvae can be blocked and killed in O. hupensis pre-infected with Exorchis sp. eggs. However, the molecular and cellular mechanisms involved in this process remain unclear. Therefore, in the present study, a combined transcriptomic and proteomic analysis was performed to identify the differential proteins involved in the immune response to the parasite S. japonicum in the O. hupensis snail host pre-infected with Exorchis sp. trematodes. The results showed that a total of 46,162 unigenes were obtained with 23,535 (50.98%) unigenes annotated in relevant databases, and 3811 proteins from O. hupensis were identified. In addition, iTRAQ-based quantitative proteomic analysis demonstrated that among three groups (OhSj-1_vs_OhN-1, OhE-1_vs_OhN-1 and OhES-1_vs_OhN-1), there were 146 common differential proteins including 44 up-regulated proteins and 90 down-regulated proteins, and 195 differential proteins exclusive to only one experimental group, including 91 up-regulated proteins and 104 down-regulated proteins, which were defined as the Common group and the Only group, respectively. KEGG analysis showed that 15 and 11 differential proteins were annotated in "Infectious diseases" in the Common group and the Only group, respectively, indicating that these proteins may be involved in the snail host immune response to parasite infection. These data will be helpful for better understanding the host-parasite interaction, and could pave the way towards exploring the mechanisms involved in the biological control on S. japonicum in O. hupensis. They also provide valuable information about developing new anti-schistosomiasis strategies.
Collapse
|
473
|
Oliveira LC, Porto TS, Junior AHC, Santos MFC, Ramos HP, Braun GH, de Lima Paula LA, Bastos JK, Furtado NAJC, Parreira RLT, Veneziani RCS, Magalhães LG, Ambrósio SR. Schistosomicidal activity of kaurane, labdane and clerodane-type diterpenes obtained by fungal transformation. Process Biochem 2020. [DOI: 10.1016/j.procbio.2020.07.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
474
|
Ammar AI, Afifi AF, Essa A, Galal-Khallaf A, Mokhtar MM, Shehab-Eldeen S, Rady AA. Cucurbita pepo Seed Oil Induces Microsatellite Instability and Tegumental Damage to Schistosoma mansoni Immature and Adult Worms In vitro. Infect Drug Resist 2020; 13:3469-3484. [PMID: 33116667 PMCID: PMC7549022 DOI: 10.2147/idr.s265699] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 08/17/2020] [Indexed: 12/16/2022] Open
Abstract
Background Schistosomiasis is a tropical parasitic disease treated exclusively with praziquantel (PZQ). However, PZQ has low efficacy against schistosomula and juveniles. In addition, the emergence of PZQ resistance has prompted the search for new drugs. Methods This study investigated the effects of pumpkin (Cucurbita pepo)-seed oil (PSO) on Schistosoma mansoni adults, juveniles, and newly formed schistosomula in vitro by exposing the parasites to increasing concentrations of PSO (20, 40, 60, 80, and 100 µl/mL) with variable incubation periods (24, 48, and 72 hours). Dose-response effects of PSO on mortality rate, worm activity, and tegumental changes were studied. Also, effect on DNA were assessed with microsatellite analysis. Results All tested stages of S. mansoni were susceptible to PSO, which was more effective than PZQ on juvenile worms and schistosomula. Juveniles and schistosomula S. mansoni were more sensitive to the antischistosomal activity of PSO than adult worms. PSO showed evident changes in the integuments of adults, juveniles, and schistosomula. These changes were more evident with increased concentrations. At the genomic level, PSO induced clear qualitative and quantitative changes in the microsatellite loci R95529 and SMD57 of S. mansoni adults and schistosomula. This microsatellite instability is being reported through the current study for S. mansoni in response to PSO for the first time. Conclusion This study suggested that PSO possesses effective antischistosomal activity against various stages of S. mansoni. Further investigations are needed to figure out the mechanism of action of PSO on this parasite.
Collapse
Affiliation(s)
- Amany Ibrahim Ammar
- Department of Medical Parasitology, Faculty of Medicine, Menoufia University, Shebin El Kom, Menoufia, Egypt
| | - Amira Fathy Afifi
- Department of Medical Parasitology, Faculty of Medicine, Menoufia University, Shebin El Kom, Menoufia, Egypt
| | - Abdallah Essa
- Tropical Medicine Department, Faculty of Medicine, Menoufia University, Shebin El Kom, Menoufia, Egypt.,Internal Medicine Department, College of Medicine, King Faisal University, Al-Ahsaa, Saudi Arabia
| | - Asmaa Galal-Khallaf
- Molecular Biology and Biotechnology Laboratory, Department of Zoology, Faculty of Science, Menoufia University, Shebin El- Kom, Menoufia, Egypt
| | - Mostafa Mohamed Mokhtar
- Department of Zoology and Entomology, Faculty of Science, Al-Azhar University, Nasr City, Egypt
| | - Somaia Shehab-Eldeen
- Tropical Medicine Department, Faculty of Medicine, Menoufia University, Shebin El Kom, Menoufia, Egypt.,Internal Medicine Department, College of Medicine, King Faisal University, Al-Ahsaa, Saudi Arabia
| | - Amany Ahmed Rady
- Department of Medical Parasitology, Faculty of Medicine, Menoufia University, Shebin El Kom, Menoufia, Egypt
| |
Collapse
|
475
|
Takaki KK, Rinaldi G, Berriman M, Pagán AJ, Ramakrishnan L. Schistosoma mansoni Eggs Modulate the Timing of Granuloma Formation to Promote Transmission. Cell Host Microbe 2020; 29:58-67.e5. [PMID: 33120115 PMCID: PMC7815046 DOI: 10.1016/j.chom.2020.10.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 07/28/2020] [Accepted: 09/30/2020] [Indexed: 01/07/2023]
Abstract
Schistosome eggs provoke the formation of granulomas, organized immune aggregates, around them. For the host, the granulomatous response can be both protective and pathological. Granulomas are also postulated to facilitate egg extrusion through the gut lumen, a necessary step for parasite transmission. We used zebrafish larvae to visualize the granulomatous response to Schistosomamansoni eggs and inert egg-sized beads. Mature eggs rapidly recruit macrophages, which form granulomas within days. Beads also induce granulomas rapidly, through a foreign body response. Strikingly, immature eggs do not recruit macrophages, revealing that the eggshell is immunologically inert. Our findings suggest that the eggshell inhibits foreign body granuloma formation long enough for the miracidium to mature. Then parasite antigens secreted through the eggshell trigger granulomas that facilitate egg extrusion into the environment. In support of this model, we find that only mature S. mansoni eggs are shed into the feces of mice and humans. Foreign bodies are walled off by immune structures called granulomas Schistosoma mansoni eggshells prevent the formation of granulomas around immature parasites Secreted antigens from mature parasites induce granulomas that promote egg shedding S. mansoni modulates granuloma formation to selectively shed mature eggs into feces
Collapse
Affiliation(s)
- Kevin K Takaki
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Gabriel Rinaldi
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, UK
| | - Matthew Berriman
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, UK
| | - Antonio J Pagán
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK.
| | - Lalita Ramakrishnan
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK.
| |
Collapse
|
476
|
Zhu B, Luo F, Shen Y, Yang W, Sun C, Wang J, Li J, Mo X, Xu B, Zhang X, Li Y, Hu W. Schistosoma japonicum cathepsin B2 (SjCB2) facilitates parasite invasion through the skin. PLoS Negl Trop Dis 2020; 14:e0008810. [PMID: 33104723 PMCID: PMC7644097 DOI: 10.1371/journal.pntd.0008810] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 11/05/2020] [Accepted: 09/22/2020] [Indexed: 12/15/2022] Open
Abstract
Cercariae invasion of the human skin is the first step in schistosome infection. Proteases play key roles in this process. However, little is known about the related hydrolytic enzymes in Schistosoma japonicum. Here, we investigated the biochemical features, tissue distribution and biological roles of a cathepsin B cysteine protease, SjCB2, in the invasion process of S. japonicum cercariae. Enzyme activity analysis revealed that recombinant SjCB2 is a typical cysteine protease with optimum temperature and pH for activity at 37°C and 4.0, respectively, and can be totally inhibited by the cysteine protease inhibitor E-64. Immunoblotting showed that both the zymogen (50 kDa) and mature enzyme (30.5 kDa) forms of SjCB2 are expressed in the cercariae. It was observed that SjCB2 localized predominantly in the acetabular glands and their ducts of cercariae, suggesting that the protease could be released during the invasion process. The protease degraded collagen, elastin, keratin, fibronectin, immunoglobulin (A, G and M) and complement C3, protein components of the dermis and immune system. In addition, proteomic analysis demonstrated that SjCB2 can degrade the human epidermis. Furthermore, it was showed that anti-rSjCB2 IgG significantly reduced (22.94%) the ability of the cercariae to invade the skin. The cysteine protease, SjCB2, located in the acetabular glands and their ducts of S. japonicum cercariae. We propose that SjCB2 facilitates skin invasion by degrading the major proteins of the epidermis and dermis. However, this cysteine protease may play additional roles in host-parasite interaction by degrading immunoglobins and complement protein. Schistosomiasis is one of the most prevalent parasitic diseases in the world, with about 200 million humans infected in 74 tropical countries. The infection of schistosome is initiated when the larvae, cercariae, penetrate the human skin. Proteolytic enzymes are likely involved in the invasion process, but these have yet to be characterized for S. japonicum. Here, we have functionally expressed a recombinant form of the cathepsin B cysteine protease SjCB2 in the yeast Pichia pastoris. Our study showed that SjCB2 degraded a number of proteins associated with the skin and immune systems, and disrupted the structure of the human epidermis. The enzyme was located in the acetabular glands and their ducts in the cercariae, where it would be stored before released into the skin. Antibody-blocking studies revealed that SjCB2 had a 22.94% contribution during the cercariae invasion process. Taken together, our findings suggest that SjCB2 helped cercariae penetrating the skin barrier and evading the immune attack to allow successful infection in the mammalian host.
Collapse
Affiliation(s)
- Bingkuan Zhu
- Department of infectious diseases, Huashan Hospital, State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Science, Fudan University, Shanghai, China
| | - Fang Luo
- Department of infectious diseases, Huashan Hospital, State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Science, Fudan University, Shanghai, China
| | - Yi Shen
- Department of infectious diseases, Huashan Hospital, State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Science, Fudan University, Shanghai, China
| | - Wenbin Yang
- Department of infectious diseases, Huashan Hospital, State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Science, Fudan University, Shanghai, China
| | - Chengsong Sun
- Department of infectious diseases, Huashan Hospital, State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Science, Fudan University, Shanghai, China
| | - Jipeng Wang
- Department of infectious diseases, Huashan Hospital, State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Science, Fudan University, Shanghai, China
| | - Jian Li
- Dermatology Department, Huashan Hospital of Fudan University, Shanghai, China
| | - Xiaojin Mo
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of China Ministry of Health, WHO Collaborating Centre for Tropical Diseases, Joint Research Laboratory of Genetics and Ecology on Parasite-host Interaction, Chinese Center for Disease Control and Prevention & Fudan University, Shanghai, China
| | - Bin Xu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of China Ministry of Health, WHO Collaborating Centre for Tropical Diseases, Joint Research Laboratory of Genetics and Ecology on Parasite-host Interaction, Chinese Center for Disease Control and Prevention & Fudan University, Shanghai, China
| | - Xumin Zhang
- Department of infectious diseases, Huashan Hospital, State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Science, Fudan University, Shanghai, China
| | - Yongdong Li
- Key Laboratory of Organo-Pharmaceutical Chemistry, Gannan Normal University, Ganzhou, China
- * E-mail: (YL); (WH)
| | - Wei Hu
- Department of infectious diseases, Huashan Hospital, State Key Laboratory of Genetic Engineering, Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Science, Fudan University, Shanghai, China
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of China Ministry of Health, WHO Collaborating Centre for Tropical Diseases, Joint Research Laboratory of Genetics and Ecology on Parasite-host Interaction, Chinese Center for Disease Control and Prevention & Fudan University, Shanghai, China
- * E-mail: (YL); (WH)
| |
Collapse
|
477
|
Wang L, Wu X, Li X, Zheng X, Wang F, Qi Z, Huang M, Zou Y. Imported Schistosomiasis: A New Public Health Challenge for China. Front Med (Lausanne) 2020; 7:553487. [PMID: 33195303 PMCID: PMC7642816 DOI: 10.3389/fmed.2020.553487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 08/18/2020] [Indexed: 12/18/2022] Open
Abstract
Significantly increased imported schistosomiasis cases have been reported in China as the economy grows. The aim of this study is to review and summarize the current status, clinical features, and transmission risk of imported infections of Schistosoma mansoni and Schistosoma haematobium in China. A retrospective study was performed to review all information regarding the imported cases of schistosomiasis collected from published literature and the database of the National Notifiable Disease Report System from 1979 to 2019. The characterization of epidemiological and clinical features was analyzed. A total of 355 cases of imported schistosomiasis have been reported in 15 provinces (autonomous regions, municipalities) in China since 1979, including 78 cases of infection with S. mansoni (21.97%), 262 cases with S. haematobium (73.80%), and 15 cases with unidentified Schistosoma species. Eosinophilia was the most common sign of the infection with S. mansoni (91.03%). The parasitological findings were confirmed in 89.74% (70/78) of cases infected with S. mansoni and 32.06% (84/262) of cases infected with S. haematobium. There was no imported case of infection of Schistosoma japonicum, Schistosoma intercalatum, or Schistosoma mekongi reported in China during this period. Praziquantel is the best therapeutic drug for curing imported schistosomiasis. In addition, Biomphalaria straminea, the intermediate host of S. mansoni, has already been found in Guangdong province in south China. There is a rising risk that the existence of the intermediate host B. straminea and the imported cases of S. mansoni infection could cause the spread of the infections and make these endemic. Thus, better understanding of the clinical features and the transmission pattern of these Schistosoma infections would assist Chinese physicians in the diagnosis and treatment of these imported schistosomiasis cases.
Collapse
Affiliation(s)
- Lei Wang
- Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Tropical Medicine, Beijing, China.,Beijing Key Laboratory for Research on Prevention and Treatment of Tropical Diseases, Beijing, China
| | - Xiaoying Wu
- Department of Gastroenterology, 3rd Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoli Li
- Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Tropical Medicine, Beijing, China.,Beijing Key Laboratory for Research on Prevention and Treatment of Tropical Diseases, Beijing, China
| | - Xiaoyan Zheng
- Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Tropical Medicine, Beijing, China.,Beijing Key Laboratory for Research on Prevention and Treatment of Tropical Diseases, Beijing, China
| | - Fei Wang
- Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Tropical Medicine, Beijing, China.,Beijing Key Laboratory for Research on Prevention and Treatment of Tropical Diseases, Beijing, China
| | - Zhiqun Qi
- Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Tropical Medicine, Beijing, China.,Beijing Key Laboratory for Research on Prevention and Treatment of Tropical Diseases, Beijing, China
| | - Minjun Huang
- Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Tropical Medicine, Beijing, China.,Beijing Key Laboratory for Research on Prevention and Treatment of Tropical Diseases, Beijing, China
| | - Yang Zou
- Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Tropical Medicine, Beijing, China.,Beijing Key Laboratory for Research on Prevention and Treatment of Tropical Diseases, Beijing, China
| |
Collapse
|
478
|
Therapeutic Effect of Diminazene Aceturate on Parasitic Blood Fluke Schistosoma mansoni Infection. Antimicrob Agents Chemother 2020; 64:AAC.01372-20. [PMID: 32816737 DOI: 10.1128/aac.01372-20] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/13/2020] [Indexed: 01/30/2023] Open
Abstract
Praziquantel is currently the only drug available to treat schistosomiasis, a disease of enormous public health significance caused by a blood fluke of the genus Schistosoma Diminazene, a drug approved by the FDA, has been successfully used to treat diseases caused by blood protozoan parasites. In this study, we evaluated the antiparasitic properties of diminazene against Schistosoma mansoni ex vivo and in mice harboring either chronic or early S. mansoni infections. In vitro, we monitored phenotypic and tegumental changes as well as the effects of the drug on pairing and egg production. In mice infected with either adult (chronic infection) or immature (early infection) worms, diminazene was administered intraperitoneally (10 to 100 mg/kg of body weight) or by oral gavage (100 to 400 mg/kg), and we studied the influence of the drug on worm burden and egg production. Liver and spleen pathologies and serum aminotransferase levels were also analyzed. In vitro, 50% effective concentrations (EC50) and EC90 revealed that diminazene is able to kill both immature and adult parasites, and its effect was time and concentration dependent. In addition, confocal laser scanning microscopy showed morphological alterations in the teguments of schistosomes. In an animal model, the influence of the drug on worm burden, egg production, hepatomegaly, and splenomegaly depended on the dosing regimen applied and the route of administration. Diminazene also caused a significant reduction in aminotransferase levels. Comparatively, diminazene treatment was more effective in chronic infection than in early infection. In tandem, our study revealed that diminazene possesses anthelmintic properties and inhibits liver injury caused by Schistosoma eggs.
Collapse
|
479
|
Hu Y, Chen J, Xu Y, Zhou H, Huang P, Ma Y, Gao M, Cheng S, Zhou H, Lv Z. Alterations of Gut Microbiome and Metabolite Profiling in Mice Infected by Schistosoma japonicum. Front Immunol 2020; 11:569727. [PMID: 33162984 PMCID: PMC7580221 DOI: 10.3389/fimmu.2020.569727] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/24/2020] [Indexed: 12/12/2022] Open
Abstract
Schistosoma japonicum (S. japonicum) is one of the etiological agents of schistosomiasis, a widespread zoonotic parasitic disease. However, the mechanism of the balanced co-existence between the host immune system and S. japonicum as well as their complex interaction remains unclear. In this study, 16S rRNA gene sequencing, combined with metagenomic sequencing approach as well as ultraperformance liquid chromatography–mass spectrometry metabolic profiling, was applied to demonstrate changes in the gut microbiome community structure during schistosomiasis progression, the functional interactions between the gut bacteria and S. japonicum infection in BALB/c mice, and the dynamic metabolite changes of the host. The results showed that both gut microbiome and the metabolites were significantly altered at different time points after the infection. Decrease in richness and diversity as well as differed composition of the gut microbiota was observed in the infected status when compared with the uninfected status. At the phylum level, the gut microbial communities in all samples were dominated by Firmicutes, Bacteroidetes, Proteobacteria, and Deferribacteres, while at the genus level, Lactobacillus, Lachnospiraceae NK4A136 group, Bacteroides, Staphylococcus, and Alloprevotella were the most abundant. After exposure, Roseburia, and Ruminococcaceae UCG-014 decreased, while Staphylococcus, Alistipes, and Parabacteroides increased, which could raise the risk of infections. Furthermore, LEfSe demonstrated several bacterial taxa that could discriminate between each time point of S. japonicum infection. Besides that, metagenomic analysis illuminated that the AMP-activated protein kinase (AMPK) signaling pathway and the chemokine signaling pathway were significantly perturbed after the infection. Phosphatidylcholine and colfosceril palmitate in serum as well as xanthurenic acid, naphthalenesulfonic acid, and pimelylcarnitine in urine might be metabolic biomarkers due to their promising diagnostic potential at the early stage of the infection. Alterations of glycerophospholipid and purine metabolism were also discovered in the infection. The present study might provide further understanding of the mechanisms during schistosome infection in aspects of gut microbiome and metabolites, and facilitate the discovery of new targets for early diagnosis and prognostic purposes. Further validations of potential biomarkers in human populations are necessary, and the exploration of interactions among S. japonicum, gut microbiome, and metabolites is to be deepened in the future.
Collapse
Affiliation(s)
- Yue Hu
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.,Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, China.,Joint Program of Pathobiology, Fifth Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jiansong Chen
- Instrumental Analysis and Research Center, Sun Yat-sen University, Guangzhou, China
| | - Yiyue Xu
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.,Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, China
| | - Hongli Zhou
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.,Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, China
| | - Ping Huang
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.,Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, China
| | - Yubin Ma
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.,Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, China
| | - Minzhao Gao
- Department of Gastroenterology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Shaoyun Cheng
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.,Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, China
| | - Haiyun Zhou
- Instrumental Analysis and Research Center, Sun Yat-sen University, Guangzhou, China
| | - Zhiyue Lv
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.,Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, China.,Joint Program of Pathobiology, Fifth Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
480
|
Pinheiro MCC, Ferreira AF, Silva Filho JDD, Lima MDS, Martins-Melo FR, Bezerra FSM, Sousa MS, Ramos AN. Burden of schistosomiasis-related mortality in Brazil: epidemiological patterns and spatial-temporal distribution, 2003-2018. Trop Med Int Health 2020; 25:1395-1407. [PMID: 32881312 DOI: 10.1111/tmi.13483] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To characterise the epidemiological patterns and the spatial-temporal distribution of schistosomiasis-related mortality in Brazil from 2003 to 2018. METHODS A national population-based ecological study that used official data from the Mortality Information System. The data included all deaths recorded in Brazil from 2003 to 2018 in which schistosomiasis was mentioned in the death certificate as an underlying or associated cause of death (multiple causes). The municipalities of residence were used as units of geographic analysis, and standardised and smoothed mortality rates (per 100 000 inhabitants) were calculated using the local empirical Bayes method. Spatial autocorrelation was evaluated using global and local Moran indexes. To analyse the spatial dependence, the Getis-Ord G and Gi* statistics were used. RESULTS During the study period, 18 421 113 deaths were recorded in Brazil. Schistosomiasis was mentioned in 11 487 deaths (proportional mortality: 0.06%); for 8141 deaths (70.87%), it was listed as the underlying cause, and for 3346 deaths (29.13%), it was listed as an associated cause. The mean mortality rate was 0.38 deaths/100 000 inhabitants. Individuals ≥ 70 years of age (RR: 115.34, 95% CI: 68.56-194.03) and residents in the Northeast region (RR: 10.81, 95% CI: 5.95-19.66) presented higher risks related to schistosomiasis. Municipalities with high mortality rates were identified in all regions, and high-risk clusters were found in municipalities located in the Northeast and Southeast regions of the country. CONCLUSIONS Schistosomiasis remains an important cause of death in persistently endemic areas in Brazil, particularly in those with a high prevalence of the disease and a marked parasite load.
Collapse
Affiliation(s)
| | - Anderson Fuentes Ferreira
- Department of Clinical and Toxicological Analysis, Laboratory of Research in Molluscan Parasitology and Biology, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - José Damiao da Silva Filho
- Department of Clinical and Toxicological Analysis, Laboratory of Research in Molluscan Parasitology and Biology, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Mauricélia da Silveira Lima
- Department of Clinical and Toxicological Analysis, Laboratory of Research in Molluscan Parasitology and Biology, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | | | - Fernando Schmelzer Moraes Bezerra
- Post-Graduate Programme in Pathology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil.,Federal Institute of Education, Science and Technology of Ceará, Caucaia, Ceará, Brazil.,Post-Graduate Programme in Medical Sciences, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Mariana Silva Sousa
- Post-Graduate Programme in Medical Sciences, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Alberto Novaes Ramos
- Department of Clinical and Toxicological Analysis, Laboratory of Research in Molluscan Parasitology and Biology, Federal University of Ceará, Fortaleza, Ceará, Brazil.,Department of Community Health, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| |
Collapse
|
481
|
Domingues ALC, Barbosa CS, Agt TFA, Mota AB, Franco CMR, Lopes EP, Loyo R, Gomes ECS. Spinal neuroschistosomiasis caused by Schistoma mansoni: cases reported in two brothers. BMC Infect Dis 2020; 20:724. [PMID: 33008310 PMCID: PMC7530957 DOI: 10.1186/s12879-020-05428-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/16/2020] [Indexed: 11/23/2022] Open
Abstract
Background Spinal neuroschistosomiasis (SN) is one of the most severe clinical presentations of schistosomiasis infection and an ectopic form of the disease caused by any species of Schistosoma. In Brazil, all cases of this clinical manifestation are related to Schistosoma mansoni, the only species present in the country. Although many cases have been reported in various endemic areas in Brazil, this is the first time in the literature that SN is described in two brothers. Case presentation Two cases of SN were accidentally diagnosed during an epidemiological survey in an urban area endemic for schistosomiasis transmission. Both patients complained of low back pain and muscle weakness in the lower limbs. Sphincter dysfunction and various degrees of paresthesia were also reported. The patients’ disease was classified as hepato-intestinal stage schistosomiasis mansoni at the onset of the chronic form. A positive parasitological stool test for S. mansoni, clinical evidence of myeloradicular damage and exclusion of other causes of damage were the basic criteria for diagnosis. After treatment with praziquantel and corticosteroid, the patients presented an improvement in symptoms, although some complaints persisted. Conclusions It is important to consider SN when patients come from areas endemic for transmission of schistosomiasis mansoni. Clinical physicians and neurologists should consider this diagnostic hypothesis, because recovery from neurological injuries is directly related to early treatment. As, described here in two brothers, a genetic predisposition may be related to neurological involvement. Primary care physicians should thus try to evaluate family members and close relatives in order to arrive at prompt schistosomiasis diagnosis in asymptomatic individuals and propose treatment in an attempt to avoid progression to SN.
Collapse
Affiliation(s)
- Ana Lúcia Coutinho Domingues
- Department of Gastroenterology, Clinical Hospital, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Constança Simões Barbosa
- Department of Parasitology, Schistosomiasis Reference Laboratory, Aggeu Magalhães Institute, Fiocruz - Ministry of Health, Recife, PE, 50740-465, Brazil
| | - Thiago Frederico Andrade Agt
- Department of Neurology, Clinical Hospital, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Andréia Braga Mota
- Department of Neurology, Clinical Hospital, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Clélia Maria Ribeiro Franco
- Department of Neurology, Clinical Hospital, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Edmundo Pessoa Lopes
- Department of Gastroenterology, Clinical Hospital, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Rodrigo Loyo
- Department of Parasitology, Schistosomiasis Reference Laboratory, Aggeu Magalhães Institute, Fiocruz - Ministry of Health, Recife, PE, 50740-465, Brazil
| | - Elainne Christine Souza Gomes
- Department of Parasitology, Schistosomiasis Reference Laboratory, Aggeu Magalhães Institute, Fiocruz - Ministry of Health, Recife, PE, 50740-465, Brazil.
| |
Collapse
|
482
|
Guo JY, Xu J, Zhang LJ, Lv S, Cao CL, Li SZ, Zhou XN. Surveillance on schistosomiasis in five provincial-level administrative divisions of the People's Republic of China in the post-elimination era. Infect Dis Poverty 2020; 9:136. [PMID: 33004080 PMCID: PMC7528395 DOI: 10.1186/s40249-020-00758-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/22/2020] [Indexed: 02/15/2023] Open
Abstract
Background The People’s Republic of China (P. R. China) has made significant progress on schistosomiasis control. Among the 12 provincial-level administrative divisions (PLADs) with schistosomiasis endemic in P. R. China, Guangdong, Shanghai, Fujian, Guangxi and Zhejiang PLADs (following as five PLADs) had successively eliminated schistosomiasis during 1985–1995. However, consolidation of the schistosomiasis elimination in these five PLADs remains challenging. In the current study, we sought to understand the epidemic situation in these post-elimination areas and their surveillance capabilities on schistosomiasis. Methods Annual data reflecting the interventions and surveillance on human beings, cattle and snails based on county level from 2005 to 2016 were collected through the national schistosomiasis reporting system and the data were analyzed to understand the epidemic status of schistosomiasis in the five PLADs. A standardized score sheet was designed to assess the surveillance capacity for schistosomiasis of selected disease control agencies in five PLADs and ten counties. Assessment on surveillance capacity including schistosomiasis diagnostic skills, identification of snails’ living and infection status and knowledge about schistosomiasis and its control were made. Descriptive analysis was used to analyze the epidemic status and evaluation results on surveillance capacities. Results The assessments showed that no local cases in humans and cattle or infected snail were found in these five PLADs since 2005. However, from 2005 to 2016, a total of 221 imported cases were detected in Zhejiang, Shanghai and Fujian, and 11.98 hm2 of new snail habitats were found in Zhejiang, Shanghai and Guangxi. In addition, snail infestation reoccurred in 247.55 hm2 of former snail habitats since 2011. For the surveillance capacity assessment, the accuracy rate of IHA and MHT were 100 and 89.3%, respectively. All participants could judge the living status of snails accurately and 98.1% on the infection status of snails. The accuracy rate of the questionnaire survey was 98.0%. Conclusions Elimination of schistosomiasis was consolidated successfully in five PLADs of P. R. China due to effective and strong post-elimination surveillance. Comprehensive consolidation strategies should be focused on the elimination of residual snails and the prevention of imported infection sources to consolidate the achievements of schistosomiasis control.
Collapse
Affiliation(s)
- Jing-Yi Guo
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, Chinese Center for Tropical Disease Research, Shanghai, 200025, People's Republic of China
| | - Jing Xu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, Chinese Center for Tropical Disease Research, Shanghai, 200025, People's Republic of China
| | - Li-Juan Zhang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, Chinese Center for Tropical Disease Research, Shanghai, 200025, People's Republic of China.
| | - Shan Lv
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, Chinese Center for Tropical Disease Research, Shanghai, 200025, People's Republic of China
| | - Chun-Li Cao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, Chinese Center for Tropical Disease Research, Shanghai, 200025, People's Republic of China
| | - Shi-Zhu Li
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, Chinese Center for Tropical Disease Research, Shanghai, 200025, People's Republic of China
| | - Xiao-Nong Zhou
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, Chinese Center for Tropical Disease Research, Shanghai, 200025, People's Republic of China
| |
Collapse
|
483
|
|
484
|
Recent Progress in the Development of Liver Fluke and Blood Fluke Vaccines. Vaccines (Basel) 2020; 8:vaccines8030553. [PMID: 32971734 PMCID: PMC7564142 DOI: 10.3390/vaccines8030553] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/18/2020] [Accepted: 09/18/2020] [Indexed: 12/14/2022] Open
Abstract
Liver flukes (Fasciola spp., Opisthorchis spp., Clonorchis sinensis) and blood flukes (Schistosoma spp.) are parasitic helminths causing neglected tropical diseases that result in substantial morbidity afflicting millions globally. Affecting the world’s poorest people, fasciolosis, opisthorchiasis, clonorchiasis and schistosomiasis cause severe disability; hinder growth, productivity and cognitive development; and can end in death. Children are often disproportionately affected. F. hepatica and F. gigantica are also the most important trematode flukes parasitising ruminants and cause substantial economic losses annually. Mass drug administration (MDA) programs for the control of these liver and blood fluke infections are in place in a number of countries but treatment coverage is often low, re-infection rates are high and drug compliance and effectiveness can vary. Furthermore, the spectre of drug resistance is ever-present, so MDA is not effective or sustainable long term. Vaccination would provide an invaluable tool to achieve lasting control leading to elimination. This review summarises the status currently of vaccine development, identifies some of the major scientific targets for progression and briefly discusses future innovations that may provide effective protective immunity against these helminth parasites and the diseases they cause.
Collapse
|
485
|
Anter A, El-Ghany MA, Abou El Dahab M, Mahana N. Does Curcumin Have a Role in the Interaction between Gut Microbiota and Schistosoma mansoni in Mice? Pathogens 2020; 9:pathogens9090767. [PMID: 32961786 PMCID: PMC7558489 DOI: 10.3390/pathogens9090767] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 08/19/2020] [Accepted: 08/19/2020] [Indexed: 12/11/2022] Open
Abstract
There is strong correlation between changes in abundance of specific bacterial species and several diseases including schistosomiasis. Several studies have described therapeutic effects of curcumin (CUR) which may arise from its regulative effects on intestinal microbiota. Thus, we examined the impact of CUR on the diversity of intestinal microbiota with/without infection by Schistosoma mansoni cercariae for 56 days. Enterobacteriaceae was dominating in a naive and S. mansoni infected mice group without CUR treatment, the most predominant species was Escherichia coli with relative density (R.D%) = 80.66% and the least one was Pseudomonas sp. (0.52%). The influence of CUR on murine microbiota composition was examined one week after oral administration of high (40) and low (20 mg/kg b.w.) CUR doses were administered three times, with two day intervals. CUR induced high variation in the Enterobacteriaceae family, characterized by a significant (p < 0.001) reduction in E. coli and asignificant (p < 0.001) increase in Pseudomonas sp. in both naïve and S. mansoni-infected mice, compared to untreated mice, in a dose-dependent manner. Additionally, our study showed the effects of high CUR doses on S. mansoni infection immunological and parasitological parameters. These data support CUR’s ability to promote Pseudomonas sp. known to produce schistosomicidal toxins and offset the sequelae of murine schistosomiasis.
Collapse
Affiliation(s)
- Assmaa Anter
- Zoology Department, Faculty of Science, Cairo University, Giza 12613, Egypt;
| | - Mohamed Abd El-Ghany
- Botany and Microbiology Department, Faculty of Science, Cairo University, Giza 12613, Egypt;
| | - Marwa Abou El Dahab
- Zoology Department, Faculty of Science, Ain Shams University, Cairo 11566, Egypt;
| | - Noha Mahana
- Zoology Department, Faculty of Science, Cairo University, Giza 12613, Egypt;
- Correspondence: or ; Tel.: +20-2-3567-6708
| |
Collapse
|
486
|
Wiedemann M, Voehringer D. Immunomodulation and Immune Escape Strategies of Gastrointestinal Helminths and Schistosomes. Front Immunol 2020; 11:572865. [PMID: 33042153 PMCID: PMC7527441 DOI: 10.3389/fimmu.2020.572865] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022] Open
Abstract
Parasitic worms (helminths) developed various immunoregulatory mechanisms to counteract the immune system of their host. The increasing identification and characterization of helminth-derived factors with strong immune modulatory activity provides novel insights into immune escape strategies of helminths. Such factors might be good targets to enhance anti-helminthic immune responses. In addition, immunosuppressive helminth-derived factors could be useful to develop new therapeutic strategies for treatment of chronic inflammatory conditions. This review will take an in depth look at the effects of immunomodulatory molecules produced by different helminths with a focus on schistosomes and mouse models of hookworm infections.
Collapse
Affiliation(s)
- Marie Wiedemann
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| |
Collapse
|
487
|
Ortega MÁ, Guzmán Merino A, Fraile-Martínez O, Recio-Ruiz J, Pekarek L, G. Guijarro L, García-Honduvilla N, Álvarez-Mon M, Buján J, García-Gallego S. Dendrimers and Dendritic Materials: From Laboratory to Medical Practice in Infectious Diseases. Pharmaceutics 2020; 12:pharmaceutics12090874. [PMID: 32937793 PMCID: PMC7560085 DOI: 10.3390/pharmaceutics12090874] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/11/2020] [Accepted: 09/11/2020] [Indexed: 02/07/2023] Open
Abstract
Infectious diseases are one of the main global public health risks, predominantly caused by viruses, bacteria, fungi, and parasites. The control of infections is founded on three main pillars: prevention, treatment, and diagnosis. However, the appearance of microbial resistance has challenged traditional strategies and demands new approaches. Dendrimers are a type of polymeric nanoparticles whose nanometric size, multivalency, biocompatibility, and structural perfection offer boundless possibilities in multiple biomedical applications. This review provides the reader a general overview about the uses of dendrimers and dendritic materials in the treatment, prevention, and diagnosis of highly prevalent infectious diseases, and their advantages compared to traditional approaches. Examples of dendrimers as antimicrobial agents per se, as nanocarriers of antimicrobial drugs, as well as their uses in gene transfection, in vaccines or as contrast agents in imaging assays are presented. Despite the need to address some challenges in order to be used in the clinic, dendritic materials appear as an innovative tool with a brilliant future ahead in the clinical management of infectious diseases and many other health issues.
Collapse
Affiliation(s)
- Miguel Ángel Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.Á.O.); (A.G.M.); (O.F.-M.); (L.P.); (N.G.-H.); (M.Á.-M.); (J.B.)
- Institute Ramón y Cajal for Health Research (IRYCIS), 28034 Madrid, Spain
- Tumour Registry, Pathological Anatomy Service, University Hospital Príncipe de Asturias, 28805 Alcalá de Henares, Spain
- University Center for the Defense of Madrid (CUD-ACD), 28047 Madrid, Spain
| | - Alberto Guzmán Merino
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.Á.O.); (A.G.M.); (O.F.-M.); (L.P.); (N.G.-H.); (M.Á.-M.); (J.B.)
| | - Oscar Fraile-Martínez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.Á.O.); (A.G.M.); (O.F.-M.); (L.P.); (N.G.-H.); (M.Á.-M.); (J.B.)
| | - Judith Recio-Ruiz
- Department of Organic and Inorganic Chemistry, Faculty of Sciences, and Research Institute in Chemistry “Andrés M. del Río” (IQAR), University of Alcalá, 28801 Alcalá de Henares, Spain;
| | - Leonel Pekarek
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.Á.O.); (A.G.M.); (O.F.-M.); (L.P.); (N.G.-H.); (M.Á.-M.); (J.B.)
| | - Luis G. Guijarro
- Department of Systems Biology, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain;
- Networking Research Centre on Hepatic and Digestive Diseases (CIBER-EHD), 28029 Madrid, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.Á.O.); (A.G.M.); (O.F.-M.); (L.P.); (N.G.-H.); (M.Á.-M.); (J.B.)
- Institute Ramón y Cajal for Health Research (IRYCIS), 28034 Madrid, Spain
- University Center for the Defense of Madrid (CUD-ACD), 28047 Madrid, Spain
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.Á.O.); (A.G.M.); (O.F.-M.); (L.P.); (N.G.-H.); (M.Á.-M.); (J.B.)
- Institute Ramón y Cajal for Health Research (IRYCIS), 28034 Madrid, Spain
- University Center for the Defense of Madrid (CUD-ACD), 28047 Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology and Medicine Service, University Hospital Príncipe de Asturias, 28805 Alcalá de Henares, Madrid, Spain
| | - Julia Buján
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.Á.O.); (A.G.M.); (O.F.-M.); (L.P.); (N.G.-H.); (M.Á.-M.); (J.B.)
- Institute Ramón y Cajal for Health Research (IRYCIS), 28034 Madrid, Spain
- Tumour Registry, Pathological Anatomy Service, University Hospital Príncipe de Asturias, 28805 Alcalá de Henares, Spain
- University Center for the Defense of Madrid (CUD-ACD), 28047 Madrid, Spain
| | - Sandra García-Gallego
- Institute Ramón y Cajal for Health Research (IRYCIS), 28034 Madrid, Spain
- Department of Organic and Inorganic Chemistry, Faculty of Sciences, and Research Institute in Chemistry “Andrés M. del Río” (IQAR), University of Alcalá, 28801 Alcalá de Henares, Spain;
- Correspondence:
| |
Collapse
|
488
|
Bai X, Li M, Wang X, Chang H, Ni Y, Li C, He K, Wang H, Yang Y, Tian T, Hou M, Ji M, Xu Z. Therapeutic potential of fucoidan in the reduction of hepatic pathology in murine schistosomiasis japonica. Parasit Vectors 2020; 13:451. [PMID: 32894174 PMCID: PMC7487607 DOI: 10.1186/s13071-020-04332-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/30/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Hepatic granuloma formation and fibrosis as the consequence of tissue entrapped eggs produced by female schistosomes characterize the pathology of Schistosoma japonicum infection. It has been proposed that fucoidan, a sulfated polysaccharide existing naturally in brown seaweed Fucus vesiculosus, plays a diversified role to perform immunomodulatory activities. However, whether fucoidan functions in the host hepatic pathology is unknown and identifying the potential mechanism that is responsible for hepatic improvement is still necessary. METHODS We evaluated the hepatic pathology from S. japonicum-infected mice after treatment with fucoidan. qRT-PCR and immunofluorescence were used to detect the pro- or anti-inflammatory factors and the phosphorylated p65 in the livers. In addition, flow cytometry was also performed to investigate the T cell subsets in the S. japonicum-infected mice after treatment with fucoidan, and functional molecules relatively specific to Treg cells were detected in vitro. Furthermore, macrophages were treated with fucoidan in vitro and to detect the inflammatory cytokines. RESULTS Treatment with fucoidan significantly reduced the hepatic granuloma size and fibrosis response during S. japonicum infection. The attenuated phospho-p65 protein levels and the mRNA levels of pro-inflammatory cytokines (IL-6, IL-12 and TNF-α) were observed in the livers from fucoidan-treated S. japonicum-infected mice; however, the mRNA levels of anti-inflammatory cytokines (IL-4 and IL-13) were increased. In addition, the infiltration of Treg cells was significantly enhanced both in the livers and spleens from fucoidan-treated S. japonicum-infected mice. Consistent with this, the mRNA levels of IL-10 and TGF-β were dramatically increased in the livers from S. japonicum-infected mice after fucoidan treatment. Furthermore, in vitro stimulated splenocytes with fucoidan resulted in increasing Treg cells in splenocytes as well as the functional expression of CC chemokine receptor type 4 (CCR4) and CXC chemokine receptor type 5 (CXCR5) in Treg cells. Additionally, fucoidan promoted the mRNA levels of IL-4 and IL-13 in macrophages. CONCLUSIONS These findings suggest an important role of natural fucoidan in reducing hepatic pathology in the progress of S. japonicum infection with a stronger Treg response, which may reveal a new potential therapeutic strategy for hepatic disease caused by parasitic chronic infection.
Collapse
Affiliation(s)
- Xueqi Bai
- Department of Pathogen Biology, Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 China
| | - Maining Li
- Department of Pathogen Biology, Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 China
| | - Xinyue Wang
- Department of Pathogen Biology, Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 China
| | - Hao Chang
- Department of Pathogen Biology, Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 China
| | - Yangyue Ni
- Department of Pathogen Biology, Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 China
| | - Chen Li
- Department of Pathogen Biology, Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 China
| | - Kaiyue He
- Department of Pathogen Biology, Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 China
| | - Huiquan Wang
- Department of Pathogen Biology, Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 China
| | - Yuxuan Yang
- Department of Pathogen Biology, Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 China
| | - Tian Tian
- Department of Dermatology, The Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing, Jiangsu 211100 China
| | - Min Hou
- Department of Pathogen Biology, Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 China
| | - Minjun Ji
- Department of Pathogen Biology, Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 China
| | - Zhipeng Xu
- Department of Pathogen Biology, Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 China
| |
Collapse
|
489
|
Volkman T, Clifford V, Paxton GA. Schistosoma serology after praziquantel treatment of Schistosoma infection in refugee children resettled in Australia: A retrospective analysis. Travel Med Infect Dis 2020; 37:101680. [DOI: 10.1016/j.tmaid.2020.101680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/12/2020] [Accepted: 04/15/2020] [Indexed: 11/30/2022]
|
490
|
Sanches RCO, Souza C, Oliveira SC. Schistosoma antigens as activators of inflammasome pathway: from an unexpected stimulus to an intriguing role. Microbes Infect 2020; 22:534-539. [PMID: 32841730 DOI: 10.1016/j.micinf.2020.08.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/14/2020] [Accepted: 08/16/2020] [Indexed: 01/14/2023]
Abstract
Parasites of the genus Schistosoma are organisms capable of living for decades within the definitive host. They interfere with the immune response by interacting with host's receptors. In this review, we discuss from the first reports to the most recent discoveries regarding the ability of Schistosoma antigens in triggering intracellular receptors and inducing inflammasome activation.
Collapse
Affiliation(s)
- Rodrigo C O Sanches
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Cláudia Souza
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Sergio C Oliveira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq MCT, 31270-901, Salvador, Brazil.
| |
Collapse
|
491
|
Liu S, Piao X, Hou N, Cai P, Ma Y, Chen Q. Duplex real-time PCR for sexing Schistosoma japonicum cercariae based on W chromosome-specific genes and its applications. PLoS Negl Trop Dis 2020; 14:e0008609. [PMID: 32822351 PMCID: PMC7467314 DOI: 10.1371/journal.pntd.0008609] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 09/02/2020] [Accepted: 07/15/2020] [Indexed: 02/07/2023] Open
Abstract
As a unique feature among otherwise hermaphroditic trematodes, Schistosoma species are gonochoric parasites whose sex is genetically determined (ZZ for males and ZW for females). However, schistosome larvae are morphologically identical, and sex can only be discriminated by molecular methods. Here, we integrated published Schistosoma. japonicum transcriptome and genome data to identify W chromosome-specific genes as sex biomarkers. Three W chromosome-specific genes of S. japonicum were identified as sex biomarkers from a panel of 12 genes expressed only in females. An efficient duplex real-time PCR (qPCR) method for sexing cercariae was developed which could identify the sex of cercariae within 2 h without DNA extraction. Moreover, this method can be used to identify not only single-sex but also mixed-sex schistosome-infected snails. We observed a nearly equal proportion of single-male, single-female, and mixed-sex schistosome infections in artificially infected snails. Sex-known schistosome-infected snail models can be efficiently constructed with the aid of duplex qPCR. A field study revealed that single-sex schistosome infections were predominant among naturally infected snails. Finally, a schistosomiasis mouse model based on sex-known cercariae infection was shown to be more reliable than a model based on sex-unknown cercariae infection. The developed duplex qPCR method for sexing S. japonicum cercariae can be widely used for schistosomiasis modeling, genetic experiments, and field-based molecular epidemiological studies. Schistosoma japonicum is a major causative agent of human schistosomiasis. Unlike other parasitic worms, S. japonicum females are determined by the heterogametic sex chromosome (ZW) and males by the homogametic sex chromosome (ZZ). The life cycle of S. japonicum includes the egg, miracidium, mother sporocyst, daughter sporocyst, cercaria, schistosomulum, and adult stages. The sex of adult male and female worms can be morphologically distinguished, whereas the sex of larvae, such as cercariae, can only be discriminated by molecular methods. In this study, we established an efficient duplex real-time PCR method for sexing S. japonicum cercariae based on newly identified W chromosome-specific genes. The established duplex real-time PCR method will facilitate construction of sex-controlled schistosome-infected intermediate host or definitive host models for schistosome-host interplays and schistosomiasis studies. This method is also a powerful tool for investigating the epidemiology of single-sex and mixed-sex schistosome-infected snails in the field.
Collapse
Affiliation(s)
- Shuai Liu
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
- * E-mail: (SL); (QC)
| | - Xianyu Piao
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Nan Hou
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Pengfei Cai
- Molecular Parasitology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Yu Ma
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Qijun Chen
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agriculture University, Shenyang, P.R. China
- * E-mail: (SL); (QC)
| |
Collapse
|
492
|
Cleenewerk L, Garssen J, Hogenkamp A. Clinical Use of Schistosoma mansoni Antigens as Novel Immunotherapies for Autoimmune Disorders. Front Immunol 2020; 11:1821. [PMID: 32903582 PMCID: PMC7438586 DOI: 10.3389/fimmu.2020.01821] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/07/2020] [Indexed: 12/11/2022] Open
Abstract
The hygiene hypothesis states that improved hygiene and the resulting disappearance of once endemic diseases is at the origin of the enormous increase in immune related disorders such as autoimmune diseases seen in the industrialized world. Helminths, such as Schistosoma mansoni, are thought to provide protection against the development of autoimmune diseases by regulating the host's immune response. This modulation primarily involves induction of regulatory immune responses, such as generation of tolerogenic dendritic cells and alternatively activated macrophages. This points toward the potential of employing helminths or their products/metabolites as therapeutics for autoimmune diseases that are characterized by an excessive inflammatory state, such as multiple sclerosis (MS), type I diabetes (T1D) and inflammatory bowel disease (IBD). In this review, we examine the known mechanisms of immune modulation by S. mansoni, explore preclinical and clinical studies that investigated the use of an array helminthic products in these diseases, and propose that helminthic therapy opens opportunities in the treatment of chronic inflammatory disorders.
Collapse
Affiliation(s)
- L Cleenewerk
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Beta Sciences, Utrecht University, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Beta Sciences, Utrecht University, Utrecht, Netherlands.,Division of Immunology, Danone Nutricia Research B.V., Utrecht, Netherlands
| | - Astrid Hogenkamp
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Beta Sciences, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
493
|
Giorgio S, Gallo-Francisco PH, Roque GAS, Flóro E Silva M. Granulomas in parasitic diseases: the good and the bad. Parasitol Res 2020; 119:3165-3180. [PMID: 32789534 DOI: 10.1007/s00436-020-06841-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 08/02/2020] [Indexed: 12/12/2022]
Abstract
Parasitic diseases affect more than one billion people worldwide, and most of them are chronic conditions in which the treatment and prevention are difficult. The appearance of granulomas, defined as organized and compact structures of macrophages and other immune cells, during various parasitic diseases is frequent, since these structures will only form when individual immune cells do not control the invading agent. Th2-typering various parasitic diseases are frequent, since these structures will only form when individual immune cells do not control the invading agent. The characterization of granulomas in different parasitic diseases, as well as recent findings in this field, is discussed in this review, in order to understand the significance of the granuloma and its modulation in the host-parasite interaction and in the immune, pathological, and parasitological aspects of this interaction. The parasitic granulomatous diseases granulomatous amebic encephalitis, toxoplasmosis, leishmaniasis, neurocysticercosis, and schistosomiasis mansoni are discussed as well as the mechanistic and dynamical aspects of the infectious granulomas.
Collapse
Affiliation(s)
- Selma Giorgio
- Department of Animal Biology, Biology Institute, State University of Campinas, Campinas, 13083-865, Brazil.
| | | | | | - Marina Flóro E Silva
- Department of Animal Biology, Biology Institute, State University of Campinas, Campinas, 13083-865, Brazil
| |
Collapse
|
494
|
Souza COS, Gardinassi LG, Rodrigues V, Faccioli LH. Monocyte and Macrophage-Mediated Pathology and Protective Immunity During Schistosomiasis. Front Microbiol 2020; 11:1973. [PMID: 32922381 PMCID: PMC7456899 DOI: 10.3389/fmicb.2020.01973] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
Infection by Schistosoma parasites culminates in a chronic granulomatous disease characterized by intense tissue fibrosis. Along the course of schistosomiasis, diverse leukocytes are recruited for inflammatory foci. Innate immune cell accumulation in Th2-driven granulomas around Schistosoma eggs is associated with increased collagen deposition, while monocytes and macrophages exert critical roles during this process. Monocytes are recruited to damaged tissues from blood, produce TGF-β and differentiate into monocyte-derived macrophages (MDMs), which become alternatively activated by IL-4/IL-13 signaling via IL-4Rα (AAMs). AAMs are key players of tissue repair and wound healing in response to Schistosoma infection. Alternative activation of macrophages is characterized by the activation of STAT6 that coordinates the transcription of Arg1, Chi3l3, Relma, and Mrc1. In addition to these markers, monocyte-derived AAMs also express Raldh2 and Pdl2. AAMs produce high levels of IL-10 and TGF-β that minimizes tissue damage caused by Schistosoma egg accumulation in tissues. In this review, we provide support to previous findings about the host response to Schistosoma infection reusing public transcriptome data. Importantly, we discuss the role of monocytes and macrophages with emphasis on the mechanisms of alternative macrophage activation during schistosomiasis.
Collapse
Affiliation(s)
- Camila Oliveira Silva Souza
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Luiz Gustavo Gardinassi
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil
| | - Vanderlei Rodrigues
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Lúcia Helena Faccioli
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
495
|
Lee J, Chong T, Newmark PA. The esophageal gland mediates host immune evasion by the human parasite Schistosoma mansoni. Proc Natl Acad Sci U S A 2020; 117:19299-19309. [PMID: 32737161 PMCID: PMC7431036 DOI: 10.1073/pnas.2006553117] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Schistosomes are parasitic flatworms that cause schistosomiasis, a neglected tropical disease affecting over 200 million people. Schistosomes develop multiple body plans while navigating their complex life cycle, which involves two different hosts: a mammalian definitive host and a molluscan intermediate host. Their survival and propagation depend upon proliferation and differentiation of stem cells necessary for parasite homeostasis and reproduction. Infective larvae released from snails carry a handful of stem cells that serve as the likely source of new tissues as the parasite adapts to life inside the mammalian host; however, the role of these stem cells during this critical life cycle stage remains unclear. Here, we characterize stem cell fates during early intramammalian development. Surprisingly, we find that the esophageal gland, an accessory organ of the digestive tract, develops before the rest of the digestive system is formed and blood feeding is initiated, suggesting a role in processes beyond nutrient uptake. To explore such a role, we examine schistosomes that lack the esophageal gland due to knockdown of a forkhead-box transcription factor, Sm-foxA, which blocks development and maintenance of the esophageal gland, without affecting the development of other somatic tissues. Intriguingly, schistosomes lacking the esophageal gland die after transplantation into naive mice, but survive in immunodeficient mice lacking B cells. We show that parasites lacking the esophageal gland are unable to lyse ingested immune cells within the esophagus before passing them into the gut. These results unveil an immune-evasion mechanism mediated by the esophageal gland, which is essential for schistosome survival and pathogenesis.
Collapse
Affiliation(s)
- Jayhun Lee
- Regenerative Biology, Morgridge Institute for Research, Madison, WI 53715
| | - Tracy Chong
- Regenerative Biology, Morgridge Institute for Research, Madison, WI 53715
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53715
| | - Phillip A Newmark
- Regenerative Biology, Morgridge Institute for Research, Madison, WI 53715;
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53715
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI 53715
| |
Collapse
|
496
|
Qiu J, Li R, Zhu H, Xia J, Xiao Y, Huang D, Wang Y. The effect of ecological environmental changes and mollusciciding on snail intermediate host of Schistosoma in Qianjiang city of China from 1985 to 2015. Parasit Vectors 2020; 13:397. [PMID: 32758280 PMCID: PMC7409449 DOI: 10.1186/s13071-020-04273-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/30/2020] [Indexed: 12/31/2022] Open
Abstract
Background Schistosomiasis remains prevalent in Africa, Asia and South America with an estimated burden of 1.9 million disability-adjusted life years in 2016. Targeting snails as a key to success for schistosomiasis control has been widely approved, but the long-term quantitative effects of interventions on snail control that would inform and improve future control programmes are unclear. Over the last six decades, schistosomiasis in China had been brought largely under control, and snail control as supplementary methods or part of integrated multisectoral approaches in different historical periods has played an essential role. Methods Ecological environment factors, prevalence and control data on Oncomelania hupensis between 1985 and 2015 at 5-year intervals in Qianjiang city, China, were collected. A multilevel growth model approach was used to examine the long-term effects of ecological environmental changes and mollusciciding on snail-infested area (SIA) and living snail density (LSD) during the 30 years. Results The variation of SIA was 68.4% in spatial distribution, while the variation of LSD was 68.4% in temporal distribution. Continuous mollusciciding could result in significant LSD reduction, but may not lead to significant SIA reduction. The normalized difference vegetation index (NDVI), patch size coefficient of variation (PSCoV) and mean patch size (MPS) reduction, slightly due to eco-environmental changes decreased SIA, while mean perimeter-area ratio (MPAR) and dry farm-land proportion (DFLP) reduction might increase SIA. Only NDVI and MPAR reduction led to a lower LSD. Conclusions Mollusciciding was more effective in reducing snail density, but it is not easy to eliminate snails completely. Environmental modifications could completely change the snail breeding environment and reduce its infestation area. Due to difficulty of scaling-up the current environmental modifications in waterway network regions, more effective snail control methods are needed. The experience in China could thereby provide guidance for other schistosomiasis endemic areas with a high snail prevalence.![]()
Collapse
Affiliation(s)
- Juan Qiu
- Key Laboratory of Monitoring and Estimate for Environment and Disaster of Hubei Province, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, People's Republic of China.
| | - Rendong Li
- Key Laboratory of Monitoring and Estimate for Environment and Disaster of Hubei Province, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, People's Republic of China
| | - Hong Zhu
- Hubei Center for Disease Control and Prevention, Hubei Provincial Academy of Preventive Medicine, Wuhan, People's Republic of China
| | - Jing Xia
- Hubei Center for Disease Control and Prevention, Hubei Provincial Academy of Preventive Medicine, Wuhan, People's Republic of China
| | - Ying Xiao
- Hubei Center for Disease Control and Prevention, Hubei Provincial Academy of Preventive Medicine, Wuhan, People's Republic of China
| | - Duan Huang
- Faculty of Geomatics, East China University of Technology, Nanchang, People's Republic of China
| | - Yong Wang
- State Key Laboratory of Resources and Environmental Information Systems, Institute of Geographical Sciences and Natural Resources Research, Chinese Academy of Sciences, Beijing, People's Republic of China.
| |
Collapse
|
497
|
Use of kinase inhibitors against schistosomes to improve and broaden praziquantel efficacy. Parasitology 2020; 147:1488-1498. [PMID: 32741402 DOI: 10.1017/s0031182020001250] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Praziquantel (PZQ) is the drug of choice for schistosomiasis. The potential drug resistance necessitates the search for adjunct or alternative therapies to PZQ. Previous functional genomics has shown that RNAi inhibition of Ca2+/calmodulin-dependent protein kinase II (CaMKII) gene in Schistosoma adult worms significantly improved the effectiveness of PZQ. Here we tested the in vitro efficacy of 15 selective and non-selective CaMK inhibitors against Schistosoma mansoni and showed that PZQ efficacy was improved against refractory juvenile parasites when combined with these CaMK inhibitors. By measuring CaMK activity and the mobility of adult S. mansoni, we identified two non-selective CaMK inhibitors, Staurosporine (STSP) and 1Naphthyl PP1 (1NAPP1), as promising candidates for further study. The impact of STSP and 1NAPP1 was investigated in mice infected with S. mansoni in the presence or absence of a sub-lethal dose of PZQ against 2- and 7-day-old schistosomula and adults. Treatment with STSP/PZQ induced a significant (47-68%) liver egg burden reduction compared with mice treated with PZQ alone. The findings indicate that the combination of STSP and PZQ dosages significantly improved anti-schistosomal activity compared to PZQ alone, demonstrating the potential of selective and non-selective CaMK/kinase inhibitors as a combination therapy with PZQ in treating schistosomiasis.
Collapse
|
498
|
Kifle DW, Chaiyadet S, Waardenberg AJ, Wise I, Cooper M, Becker L, Doolan DL, Laha T, Sotillo J, Pearson MS, Loukas A. Uptake of Schistosoma mansoni extracellular vesicles by human endothelial and monocytic cell lines and impact on vascular endothelial cell gene expression. Int J Parasitol 2020; 50:685-696. [DOI: 10.1016/j.ijpara.2020.05.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/18/2020] [Accepted: 05/21/2020] [Indexed: 12/27/2022]
|
499
|
Bais S, Greenberg RM. Schistosome TRP channels: An appraisal. Int J Parasitol Drugs Drug Resist 2020; 13:1-7. [PMID: 32250774 PMCID: PMC7138929 DOI: 10.1016/j.ijpddr.2020.02.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/14/2020] [Accepted: 02/16/2020] [Indexed: 02/07/2023]
Abstract
Ion channels underlie electrical excitability in cells and are essential for a variety of functions, most notably neuromuscular and sensory activity. They are also validated targets for a preponderance of approved anthelmintic compounds. Transient receptor potential (TRP) channels constitute an ion channel superfamily whose members play important roles in sensory signaling, regulation of ion homeostasis, organellar trafficking, and other key cellular and organismal activities. Unlike most other ion channels, TRP channels are often polymodal, gated by a variety of mechanisms. Furthermore, TRP channels fall into several classes or subtypes based on sequence and structure. Until recently, there had been very little investigation of the properties and functions of TRP channels from parasitic helminths, including schistosomes, but that situation has changed in the past few years. Indeed, it is now clear that at least some schistosome TRP channels exhibit unusual pharmacological properties, and, intriguingly, both a mammalian and a schistosome TRP channel are activated by praziquantel, the current antischistosomal drug of choice. With the latest release of the Schistosoma mansoni genome database, several changes in predicted TRP channel sequences appeared, some of which were significant. This review updates and reassesses the TRP channel repertoire in S. mansoni, examines recent findings regarding these potential therapeutic targets, and provides guideposts for some of the physiological functions that may be mediated by these channels in schistosomes.
Collapse
Affiliation(s)
- Swarna Bais
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA, 19104, USA
| | - Robert M Greenberg
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA, 19104, USA.
| |
Collapse
|
500
|
Midzi N, Bärenbold O, Manangazira P, Phiri I, Mutsaka-Makuvaza MJ, Mhlanga G, Utzinger J, Vounatsou P. Accuracy of different diagnostic techniques for Schistosoma haematobium to estimate treatment needs in Zimbabwe: Application of a hierarchical Bayesian egg count model. PLoS Negl Trop Dis 2020; 14:e0008451. [PMID: 32817650 PMCID: PMC7462259 DOI: 10.1371/journal.pntd.0008451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 09/01/2020] [Accepted: 06/04/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Treatment needs for Schistosoma haematobium are commonly evaluated using urine filtration with detection of parasite eggs under a microscope. A common symptom of S. haematobium is hematuria, the passing of blood in urine. Hence, the use of hematuria-based diagnostic techniques as a proxy for the assessment of treatment needs has been considered. This study evaluates data from a national survey in Zimbabwe, where three hematuria-based diagnostic techniques, that is microhematuria, macrohematuria, and an anamnestic questionnaire pertaining to self-reported blood in urine, have been included in addition to urine filtration in 280 schools across 70 districts. METHODOLOGY We developed an egg count model, which evaluates the infection intensity-dependent sensitivity and the specificity of each diagnostic technique without relying on a 'gold' standard. Subsequently, we determined prevalence thresholds for each diagnostic technique, equivalent to a 10% urine filtration-based prevalence and compared classification of districts according to treatment strategy based on the different diagnostic methods. PRINCIPAL FINDINGS A 10% urine filtration prevalence threshold corresponded to a 17.9% and 13.3% prevalence based on questionnaire and microhematuria, respectively. Both the questionnaire and the microhematuria showed a sensitivity and specificity of more than 85% for estimating treatment needs at the above thresholds. For diagnosis at individual level, the questionnaire showed the highest sensitivity (70.0%) followed by urine filtration (53.8%) and microhematuria (52.2%). CONCLUSIONS/SIGNIFICANCE The high sensitivity and specificity of a simple questionnaire to estimate treatment needs of S. haematobium suggests that it can be used as a rapid, low-cost method to estimate district prevalence. Our modeling approach can be expanded to include setting-dependent specificity of the technique and should be assessed in relation to other diagnostic methods due to potential cross-reaction with other diseases.
Collapse
Affiliation(s)
- Nicholas Midzi
- National Institute of Health Research, Ministry of Health and Child Care, Harare, Zimbabwe
| | - Oliver Bärenbold
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- Faculty of Science, University of Basel, Basel, Switzerland
| | - Portia Manangazira
- National Institute of Health Research, Ministry of Health and Child Care, Harare, Zimbabwe
| | - Isaac Phiri
- National Institute of Health Research, Ministry of Health and Child Care, Harare, Zimbabwe
| | - Masceline J. Mutsaka-Makuvaza
- National Institute of Health Research, Ministry of Health and Child Care, Harare, Zimbabwe
- Department of Medical Microbiology, University of Zimbabwe, Harare, Zimbabwe
| | - Gibson Mhlanga
- National Institute of Health Research, Ministry of Health and Child Care, Harare, Zimbabwe
| | - Jürg Utzinger
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- Faculty of Science, University of Basel, Basel, Switzerland
| | - Penelope Vounatsou
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- Faculty of Science, University of Basel, Basel, Switzerland
| |
Collapse
|