51
|
Yamasaki K, Taguchi K, Nishi K, Otagiri M, Seo H. Enhanced dissolution and oral bioavailability of praziquantel by emulsification with human serum albumin followed by spray drying. Eur J Pharm Sci 2019; 139:105064. [PMID: 31491499 DOI: 10.1016/j.ejps.2019.105064] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/08/2019] [Accepted: 09/02/2019] [Indexed: 10/26/2022]
Abstract
The goal of this study was to enhance the oral bioavailability of praziquantel through its conjugation with human serum albumin (HSA). Praziquantel-HSA particles were produced by spray drying an emulsion of an aqueous solution of HSA and a solution of praziquantel in oil. The particles were agglomerates of multiple smooth corrugated particles containing amorphous praziquantel nearly equivalent to the theoretical doses. The solubility of praziquantel in an aqueous medium was enhanced in both the produced particles and the physical mixture. In addition, the dissolution rate in an aqueous medium was enhanced in the case of particles, but not in a physical mixture. Thus, the inclusion of HSA by emulsification followed by spray drying appeared to contribute to the enhanced dissolution rate. In a pharmacokinetic study, the maximum plasma concentration (Cmax) and the area under the concentration-time curve (AUC) for the produced particles (HSA/praziquantel = 1/1 w/w) were approximately two times higher than the corresponding values for raw praziquantel. This increased oral bioavailability of the particles was considered to be due to the enhanced dissolution rate. This process for producing praziquantel-HSA particles could be useful in terms of improving the oral bioavailability of the other hydrophobic drugs.
Collapse
Affiliation(s)
- Keishi Yamasaki
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto, Japan; DDS Research Institute, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto, Japan.
| | - Kazuaki Taguchi
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto, Japan; Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-ku, Tokyo, Japan
| | - Koji Nishi
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto, Japan; DDS Research Institute, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto, Japan
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto, Japan; DDS Research Institute, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto, Japan
| | - Hakaru Seo
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto, Japan; DDS Research Institute, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto, Japan
| |
Collapse
|
52
|
Hernández-Goenaga J, López-Abán J, Protasio AV, Vicente Santiago B, del Olmo E, Vanegas M, Fernández-Soto P, Patarroyo MA, Muro A. Peptides Derived of Kunitz-Type Serine Protease Inhibitor as Potential Vaccine Against Experimental Schistosomiasis. Front Immunol 2019; 10:2498. [PMID: 31736947 PMCID: PMC6838133 DOI: 10.3389/fimmu.2019.02498] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 10/07/2019] [Indexed: 12/27/2022] Open
Abstract
Schistosomiasis is a significant public health problem in sub-Saharan Africa, China, Southeast Asia, and regions of South and Central America affecting about 189 million people. Kunitz-type serine protease inhibitors have been identified as important players in the interaction of other flatworm parasites with their mammalian hosts. They are involved in host blood coagulation, fibrinolysis, inflammation, and ion channel blocking, all of them critical biological processes, which make them interesting targets to develop a vaccine. Here, we evaluate the protective efficacy of chemically synthesized T- and B-cell peptide epitopes derived from a kunitz protein from Schistosoma mansoni. Putative kunitz-type protease inhibitor proteins were identified in the S. mansoni genome, and their expression was analyzed by RNA-seq. Gene expression analyses showed that the kunitz protein Smp_147730 (Syn. Smp_311670) was dramatically and significantly up-regulated in schistosomula and adult worms when compared to the invading cercariae. T- and B-cell epitopes were predicted using bioinformatics tools, chemically synthesized, and formulated in the Adjuvant Adaptation (ADAD) vaccination system. BALB/c mice were vaccinated and challenged with S. mansoni cercariae. Kunitz peptides were highly protective in vaccinated BALB/c mice showing significant reductions in recovery of adult females (89-91%) and in the numbers of eggs trapped in the livers (77-81%) and guts (57-77%) of mice. Moreover, liver lesions were significantly reduced in vaccinated mice (64-65%) compared to infected control mice. The vaccination regime was well-tolerated with both peptides. We propose the use of these peptides, alone or in combination, as reliable candidates for vaccination against schistosomiasis.
Collapse
Affiliation(s)
- Juan Hernández-Goenaga
- Infectious and Tropical Diseases Group (e-INTRO), IBSAL-CIETUS (Biomedical Research Institute of Salamanca-Research Centre for Tropical Diseases at the University of Salamanca), Faculty of Pharmacy, University of Salamanca, Salamanca, Spain
| | - Julio López-Abán
- Infectious and Tropical Diseases Group (e-INTRO), IBSAL-CIETUS (Biomedical Research Institute of Salamanca-Research Centre for Tropical Diseases at the University of Salamanca), Faculty of Pharmacy, University of Salamanca, Salamanca, Spain
| | - Anna V. Protasio
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Belén Vicente Santiago
- Infectious and Tropical Diseases Group (e-INTRO), IBSAL-CIETUS (Biomedical Research Institute of Salamanca-Research Centre for Tropical Diseases at the University of Salamanca), Faculty of Pharmacy, University of Salamanca, Salamanca, Spain
| | - Esther del Olmo
- Department of Pharmaceutical Chemistry, IBSAL-CIETUS, Faculty of Pharmacy, University of Salamanca, Salamanca, Spain
| | - Magnolia Vanegas
- Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
| | - Pedro Fernández-Soto
- Infectious and Tropical Diseases Group (e-INTRO), IBSAL-CIETUS (Biomedical Research Institute of Salamanca-Research Centre for Tropical Diseases at the University of Salamanca), Faculty of Pharmacy, University of Salamanca, Salamanca, Spain
| | - Manuel Alfonso Patarroyo
- Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
- School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Antonio Muro
- Infectious and Tropical Diseases Group (e-INTRO), IBSAL-CIETUS (Biomedical Research Institute of Salamanca-Research Centre for Tropical Diseases at the University of Salamanca), Faculty of Pharmacy, University of Salamanca, Salamanca, Spain
| |
Collapse
|
53
|
Transjugular Intrahepatic Portosystemic Shunt Placement in Patients with Schistosomiasis-Induced Liver Fibrosis. Cardiovasc Intervent Radiol 2019; 42:1760-1770. [PMID: 31363898 PMCID: PMC6823317 DOI: 10.1007/s00270-019-02295-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 07/20/2019] [Indexed: 12/15/2022]
Abstract
Purpose Evaluate the efficacy and safety of transjugular intrahepatic portosystemic shunt (TIPS) insertion on patients with schistosomiasis-induced liver fibrosis, and compare with that of patients with HBV-induced cirrhosis. Materials and Methods This was a retrospective study from November 2015 to December 2018 including 82 patients diagnosed with portal hypertension, one group of which is induced by schistosomiasis (n = 20), the other by hepatitis B virus (HBV) (n = 62). Both groups of subjects underwent TIPS placement for the management of portal hypertension complications. Results TIPS was inserted successfully in all patients (technical success 100%). After a median follow-up of 14 months following TIPS insertion, portal pressure gradient (PPG) value in both schistosomiasis-induced group and HBV-induced group underwent a significant decrease with no major difference between the two groups. There exists no significant difference demonstrated by Kaplan–Meier curves between two groups concerning cumulative rate of hepatic encephalopathy (HE) (log-rank p = 0.681), variceal rebleeding (log-rank p = 0.837) and survival (log-rank p = 0.429), and no statistically difference was found in terms of alleviation of portal vein thrombosis (PVT). In addition, splenectomy (HR 19, 95% CI 4–90, p < 0.001) was identified as independent predictor of PVT. Conclusions TIPS placement is well-founded to be considered as a safe and effective treatment in patients with schistosomiasis-induced portal hypertension and relevant severe complications. We also found the risk of PVT is 19 times higher in patients who underwent splenectomy than in untreated patients. Level of Evidence Historically controlled studies, level 4.
Collapse
|
54
|
Zaqout A, Abid FB, Murshed K, Al-Bozom I, Al-Rumaihi G, Al Soub H, Al Maslamani M, Al Khal A. Cerebral schistosomiasis: Case series from Qatar. Int J Infect Dis 2019; 86:167-170. [PMID: 31295550 DOI: 10.1016/j.ijid.2019.07.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/30/2019] [Accepted: 07/02/2019] [Indexed: 01/12/2023] Open
Abstract
Schistosomiasis is the third most widespread devastating parasitic disease worldwide and has a high mortality burden. Neuroschistosomiasis is one of the rare and most severe clinical presentations of the disease. It is caused by granuloma formation around eggs that lodge in the central nervous system, with Schistosoma japonicum usually causing most reported cerebral disease. Three unusual presentations of schistosomiasis in Qatar are described herein. The three patients were young males who presented with seizures and tumor-like lesions on brain imaging. The diagnosis was confirmed by biopsy, which showed necrotizing granulomas containing Schistosoma eggs. These cases raise awareness of neuroschistosomiasis as a potential cause of tumor-like brain lesions in migrants and returning travelers from endemic areas.
Collapse
Affiliation(s)
- Ahmed Zaqout
- Department of Medicine, Division of Infectious Diseases, Hamad Medical Corporation, Doha, Qatar.
| | - Fatma Ben Abid
- Department of Medicine, Division of Infectious Diseases, Hamad Medical Corporation, Doha, Qatar.
| | - Khaled Murshed
- Histopathology Department, Hamad Medical Corporation, Doha, Qatar.
| | - Issam Al-Bozom
- Histopathology Department, Hamad Medical Corporation, Doha, Qatar; Weill Cornell Medical College, Doha, Qatar.
| | - Ghaya Al-Rumaihi
- Department of Neurosurgery, Hamad Medical Corporation, Doha, Qatar.
| | - Hussam Al Soub
- Department of Medicine, Division of Infectious Diseases, Hamad Medical Corporation, Doha, Qatar; Weill Cornell Medical College, Doha, Qatar.
| | - Muna Al Maslamani
- Department of Medicine, Division of Infectious Diseases, Hamad Medical Corporation, Doha, Qatar; Weill Cornell Medical College, Doha, Qatar.
| | - Abdullatif Al Khal
- Department of Medicine, Division of Infectious Diseases, Hamad Medical Corporation, Doha, Qatar; Weill Cornell Medical College, Doha, Qatar.
| |
Collapse
|
55
|
Yegorov S, Joag V, Galiwango RM, Good SV, Mpendo J, Tannich E, Boggild AK, Kiwanuka N, Bagaya BS, Kaul R. Schistosoma mansoni treatment reduces HIV entry into cervical CD4+ T cells and induces IFN-I pathways. Nat Commun 2019; 10:2296. [PMID: 31127086 PMCID: PMC6534541 DOI: 10.1038/s41467-019-09900-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 04/02/2019] [Indexed: 01/21/2023] Open
Abstract
Schistosoma mansoni (Sm) infection has been linked with an increased risk of HIV acquisition in women. Therefore, defining the mechanism(s) by which Sm alters HIV susceptibility might lead to new HIV prevention strategies. Here, we analyze the impact of standard Sm therapy in HIV-uninfected Sm+ Ugandan adult women on genital HIV susceptibility and mucosal and systemic immunology. Schistosomiasis treatment induces a profound reduction of HIV entry into cervical and blood CD4+ T cells that is sustained for up to two months, despite transient systemic and mucosal immune activation and elevated genital IL-1α levels. Genital IFN-α2a levels are also elevated post-treatment, and IFN-α2a blocks HIV entry into primary CD4+ T cells ex vivo. Transcriptomic analysis of blood mononuclear cells post-Sm treatment shows IFN-I pathway up-regulation and partial reversal of Sm-dysregulated interferon signaling. These findings indicate that Sm therapy may reduce HIV susceptibility for women with Sm infection, potentially through de-repression of IFN-I pathways.
Collapse
Affiliation(s)
- Sergey Yegorov
- Departments of Immunology and Medicine, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada. .,Department of Science Education, Faculty of Education and Humanities, Suleyman Demirel University, 1/1 Abylai Khan Street, Kaskelen, Almaty, 040900, Kazakhstan.
| | - Vineet Joag
- Departments of Immunology and Medicine, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.,Department of Microbiology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Ronald M Galiwango
- Departments of Immunology and Medicine, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Sara V Good
- Genetics & Genome Biology, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay St., Toronto, ON, M5G 0A4, Canada.,Community Health Sciences, University of Manitoba, 750 Bannatyne Ave, Winnipeg, MB, R3E 0W2, Canada
| | - Juliet Mpendo
- Uganda Virus Research Institute -International AIDS Vaccine Initiative HIV Vaccine Program, 51/59 Nakiwogo Rd, P.O.Box 49, Entebbe, Uganda
| | - Egbert Tannich
- Bernhard Nocht Institute for Tropical Medicine, National Reference Centre for Tropical Pathogens, Bernhard-Nocht-Str. 74, 20359, Hamburg, Germany
| | - Andrea K Boggild
- Department of Medicine, University Health Network, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada.,Public Health Ontario Laboratories, 661 University Ave, Toronto, ON, M5G 1M1, Canada
| | - Noah Kiwanuka
- Uganda Virus Research Institute -International AIDS Vaccine Initiative HIV Vaccine Program, 51/59 Nakiwogo Rd, P.O.Box 49, Entebbe, Uganda.,Department of Epidemiology and Biostatistics, School of Public Health, College of Health Sciences, Makerere University, P.O. Box 7072, Kampala, Uganda
| | - Bernard S Bagaya
- Uganda Virus Research Institute -International AIDS Vaccine Initiative HIV Vaccine Program, 51/59 Nakiwogo Rd, P.O.Box 49, Entebbe, Uganda.,Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P.O. Box 7072, Kampala, Uganda
| | - Rupert Kaul
- Departments of Immunology and Medicine, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.,Department of Medicine, University Health Network, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada
| |
Collapse
|
56
|
Wang X, Yu D, Li C, Zhan T, Zhang T, Ma H, Xu J, Xia C. In vitro and in vivo activities of DW-3-15, a commercial praziquantel derivative, against Schistosoma japonicum. Parasit Vectors 2019; 12:199. [PMID: 31053083 PMCID: PMC6500042 DOI: 10.1186/s13071-019-3442-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 04/12/2019] [Indexed: 12/15/2022] Open
Abstract
Background Schistosomiasis is a debilitating neglected tropical disease that affects approximately 190 million people around the world. Praziquantel (PZQ) is the only drug available for use against all Schistosoma species. Although PZQ has a high efficacy, recognized concerns have prompted the development of new, alternative drugs for repeated use in endemic areas where PZQ efficacy against strains of Schistosoma is reduced. A hybrid drug containing different pharmacophores within a single molecule is a promising strategy. Our earlier in vivo studies showed the significant antiparasitic activity of a praziquantel derivative, DW-3-15, against Schistosoma japonicum. In the present study, DW-3-15 was synthesized in large amounts by a pharmaceutical company and its schistosomicidal efficacy and stability were further confirmed. Parameters such as parasite viability, pairing and oviposition were evaluated in vitro. An in vivo study was conducted to assess the effect of commercial DW-3-15 on worm burden, egg production and diameter of granulomas. Additionally, to gain insight into the mechanism of action for DW-3-15, morphological changes in the tegument of S. japonicum were also examined. Results The in vitro study showed the antiparasitic activity of DW-3-15 against S. japonicum, with significant reductions in viability of adult and juvenile worms, worm pairings and egg output. Compared to PZQ, DW-3-15 induced similar ultrastructural changes and evident destruction of the tegument surface in male worms. In vivo, the oral administration of DW-3-15 at a dose of 400 mg/kg per day for five consecutive days in mice significantly reduced the total worm burden and number of eggs in the liver. Histological analysis of the livers showed a marked reduction in the average diameter of the egg granuloma. Conclusions Our findings suggest that DW-3-15, a PZQ derivative with the prospect of commercial production, can be developed as a potential promising schistosomicide. Electronic supplementary material The online version of this article (10.1186/s13071-019-3442-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaoli Wang
- Department of Parasitology, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, China.,Department of Microbiology and Parasitology, Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, 2600 Donghai Road, Bengbu, 233030, China
| | - Dan Yu
- Department of Parasitology, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, China
| | - Chunxiang Li
- Department of Parasitology, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, China
| | - Tingzheng Zhan
- Department of Parasitology, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, China.,Department of Parasitology, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China
| | - Tingting Zhang
- Department of Parasitology, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, China
| | - Huihui Ma
- Department of Parasitology, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, China
| | - Jing Xu
- Department of Parasitology, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, China
| | - Chaoming Xia
- Department of Parasitology, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, China.
| |
Collapse
|
57
|
Zhao Y, Yang S, Li B, Li W, Wang J, Chen Z, Yang J, Tan H, Li J. Alterations of the Mice Gut Microbiome via Schistosoma japonicum Ova-Induced Granuloma. Front Microbiol 2019; 10:352. [PMID: 30891012 PMCID: PMC6411663 DOI: 10.3389/fmicb.2019.00352] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 02/11/2019] [Indexed: 12/17/2022] Open
Abstract
Schistosomiasis, also called bilharziasis, is a neglected tropical disease induced by Schistosoma spp. that causes hundreds of millions of infections. Although Schistosoma ova-induced granulomas commonly cause inflammation, hyperplasia, ulceration, micro abscess formation, and polyposis, the role of the egg granuloma on the gut microbiome remains unclear. To explore the role, gut microbial communities in mice infected with Schistosoma japonicum were surveyed. Female C57BL/6 and BALB/c mice were exposed to cercariae of S. japonicum for 45 and 65 days and then sacrificed. Intestinal contents and feces were collected, DNA was extracted, and high-throughput 16S rRNA gene-based pyrosequencing was used to provide a comparative analysis of gut microbial diversity. The intestinal mucosal tissues were also examined. Histopathologic analysis demonstrated that the basic structure of the colonic mucosa was damaged by ova-induced granuloma. Regarding the gut microbiome, 2,578,303 good-quality sequences were studied and assigned to 25,278 Operational Taxonomic Units (OTUs) at a threshold of 97% similarity. The average number of OTUs for C57BL/6 and BALB/c were 545 and 530, respectively. At the phylum level, intestinal microbial communities were dominated by Firmicutes, Bacteroidetes, Proteobacteria, and Verrucomicrobia. Infection with S. japonicum modified bacterial richness in the fecal associated microbiota. Exposure significantly modified bacterial community composition among different groups. At the phylogenetic levels, LEfSe analysis revealed that several bacterial taxa were significantly associated with the S. japonicum-infected mice. The present results suggest that egg granulomas in the intestine influence differentiation of the gut microbial community under pathophysiological conditions. This result suggests that intestinal microbiome-based strategies should be considered for early diagnosis, clinical treatment, and prognosis evaluation of schistosomiasis.
Collapse
Affiliation(s)
- Yanqing Zhao
- Department of Human Parasitology, School of Basic Medical Science, Shiyan, China.,Department of Infectious Diseases, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Shuguo Yang
- Department of Human Parasitology, School of Basic Medical Science, Shiyan, China.,Department of Infectious Diseases, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Bei Li
- Department of Human Parasitology, School of Basic Medical Science, Shiyan, China.,Department of Infectious Diseases, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Wei Li
- Department of Prevention and Control of Schistosomiasis, Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Jue Wang
- Department of Human Parasitology, School of Basic Medical Science, Shiyan, China.,Department of Infectious Diseases, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Zongyun Chen
- Department of Human Parasitology, School of Basic Medical Science, Shiyan, China.,Department of Infectious Diseases, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Jing Yang
- Department of Human Parasitology, School of Basic Medical Science, Shiyan, China.,Department of Infectious Diseases, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Huabing Tan
- Department of Human Parasitology, School of Basic Medical Science, Shiyan, China.,Department of Infectious Diseases, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Jian Li
- Department of Human Parasitology, School of Basic Medical Science, Shiyan, China.,Department of Infectious Diseases, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
58
|
In Vitro and In Vivo Studies of Spironolactone as an Antischistosomal Drug Capable of Clinical Repurposing. Antimicrob Agents Chemother 2019; 63:AAC.01722-18. [PMID: 30559137 DOI: 10.1128/aac.01722-18] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 12/10/2018] [Indexed: 12/21/2022] Open
Abstract
Schistosomiasis is a parasitic flatworm disease that infects over 200 million people worldwide, especially in poor communities. Treatment and control of the disease rely on just one drug, praziquantel. Since funding for drug development for poverty-associated diseases is very limited, drug repurposing is a promising strategy. In this study, from a screening of 13 marketed diuretics, we identified that spironolactone, a potassium-sparing diuretic, had potent antischistosomal effects on Schistosoma mansoni in vitro and in vivo in a murine model of schistosomiasis. In vitro, spironolactone at low concentrations (<10 µM) is able to alter worm motor activity and the morphology of adult schistosomes, leading to parasitic death. In vivo, oral treatment with spironolactone at a single dose (400 mg/kg) or daily for five consecutive days (100 mg/kg/day) in mice harboring either patent or prepatent infections significantly reduced worm burden, egg production, and hepato- and splenomegaly (P < 0.05 to P < 0.001). Taken together, with the safety profile of spironolactone, supported by its potential to affect schistosomes, these results indicate that spironolactone could be a potential treatment for schistosomiasis and make it promising for repurposing.
Collapse
|
59
|
|
60
|
Gemma S, Federico S, Brogi S, Brindisi M, Butini S, Campiani G. Dealing with schistosomiasis: Current drug discovery strategies. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2019. [DOI: 10.1016/bs.armc.2019.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
61
|
Fukushige M, Mutapi F, Woolhouse ME. Population level changes in schistosome-specific antibody levels following chemotherapy. Parasite Immunol 2019; 41:e12604. [PMID: 30467873 PMCID: PMC6492179 DOI: 10.1111/pim.12604] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 11/13/2018] [Indexed: 11/28/2022]
Abstract
AIMS Previous studies have reported that chemotherapy of schistosomiasis by praziquantel in humans boosts protective antibody responses against S mansoni and S haematobium. A number of studies have reported schistosome-specific antibody levels before and after chemotherapy. Using these reports, a meta-analysis was conducted to identify predictors of population level change in schistosome-specific antibody levels after chemotherapy. METHODS AND RESULTS Following a systematic review, 92 observations from 26 articles published between 1988 and 2013 were included in this study. Observations were grouped by antigen type and antibody isotypes for the classification and regression tree (CART) analysis. The study showed that the change in antibody levels was variable: (a) between different human populations and (b) according to the parasite antigen and antibody isotypes. Thus, while anti-worm responses predominantly increased after chemotherapy, anti-egg responses decreased or did not show a significant trend. The change in antibody levels depended on a combination of age and infection intensity for anti-egg IgA, IgM, IgG1, IgG2 and anti-worm IgM and IgG. CONCLUSION The study results are consistent with praziquantel treatment boosting anti-worm antibody responses. However, there is considerable heterogeneity in post-treatment changes in specific antibody levels that is related to host age and pre-treatment infection intensity.
Collapse
Affiliation(s)
- Mizuho Fukushige
- Present address:
Faculty of MedicineUniversity of TsukubaTsukubaJapan
- Centre for ImmunityInfection & EvolutionCollege of Medicine and Veterinary MedicineUniversity of EdinburghEdinburghUK
| | - Francisca Mutapi
- Institute of Immunology and Infection ResearchCentre for ImmunityInfection & EvolutionSchool of Biological SciencesNIHR Global Health Research Unit Tackling Infections to Benefit Africa (TIBA)University of EdinburghEdinburghUK
| | - Mark E.J. Woolhouse
- Centre for ImmunityInfection & Evolution, and Usher Institute of Population Health Sciences & InformaticsCollege of Medicine and Veterinary MedicineUniversity of EdinburghEdinburghUK
| |
Collapse
|
62
|
Parreira RLT, Costa ES, Heleno VCG, Magalhães LG, Souza JM, Pauletti PM, Cunha WR, Januário AH, Símaro GV, Bastos JK, Laurentiz RS, Kar T, Caramori GF, Kawano DF, Andrade E Silva ML. Evaluation of Lignans from Piper cubeba against Schistosoma mansoni Adult Worms: A Combined Experimental and Theoretical Study. Chem Biodivers 2018; 16:e1800305. [PMID: 30335227 DOI: 10.1002/cbdv.201800305] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 10/17/2018] [Indexed: 01/01/2023]
Abstract
Six dibenzylbutyrolactonic lignans ((-)-hinokinin (1), (-)-cubebin (2), (-)-yatein (3), (-)-5-methoxyyatein (4), dihydrocubebin (5) and dihydroclusin (6)) were isolated from Piper cubeba seed extract and evaluated against Schistosoma mansoni. All lignans, except 5, were able to separate the adult worm pairs and reduce the egg numbers during 24 h of incubation. Lignans 1, 3 and 4 (containing a lactone ring) were the most efficient concerning antiparasitary activity. Comparing structures 3 and 4, the presence of the methoxy group at position 5 appears to be important for this activity. Considering 1 and 3, it is possible to see that the substitution pattern change (methylenedioxy or methoxy groups) in positions 3' and 4' alter the biological response, with 1 being the second most active compound. Computational calculations suggest that the activity of compound 4 can be correlated with the largest lipophilicity value.
Collapse
Affiliation(s)
- Renato L T Parreira
- Núcleo de Pesquisas em Ciências Exatas e Tecnológicas, Universidade de Franca, Av. Dr. Armando Salles Oliveira 201, 14404-600 Franca, São Paulo, Brazil
| | - Eveline S Costa
- Núcleo de Pesquisas em Ciências Exatas e Tecnológicas, Universidade de Franca, Av. Dr. Armando Salles Oliveira 201, 14404-600 Franca, São Paulo, Brazil
| | - Vladimir C G Heleno
- Núcleo de Pesquisas em Ciências Exatas e Tecnológicas, Universidade de Franca, Av. Dr. Armando Salles Oliveira 201, 14404-600 Franca, São Paulo, Brazil
| | - Lizandra G Magalhães
- Núcleo de Pesquisas em Ciências Exatas e Tecnológicas, Universidade de Franca, Av. Dr. Armando Salles Oliveira 201, 14404-600 Franca, São Paulo, Brazil
| | - Julia M Souza
- Núcleo de Pesquisas em Ciências Exatas e Tecnológicas, Universidade de Franca, Av. Dr. Armando Salles Oliveira 201, 14404-600 Franca, São Paulo, Brazil
| | - Patrícia M Pauletti
- Núcleo de Pesquisas em Ciências Exatas e Tecnológicas, Universidade de Franca, Av. Dr. Armando Salles Oliveira 201, 14404-600 Franca, São Paulo, Brazil
| | - Wilson R Cunha
- Núcleo de Pesquisas em Ciências Exatas e Tecnológicas, Universidade de Franca, Av. Dr. Armando Salles Oliveira 201, 14404-600 Franca, São Paulo, Brazil
| | - Ana H Januário
- Núcleo de Pesquisas em Ciências Exatas e Tecnológicas, Universidade de Franca, Av. Dr. Armando Salles Oliveira 201, 14404-600 Franca, São Paulo, Brazil
| | - Guilherme V Símaro
- Núcleo de Pesquisas em Ciências Exatas e Tecnológicas, Universidade de Franca, Av. Dr. Armando Salles Oliveira 201, 14404-600 Franca, São Paulo, Brazil
| | - Jairo K Bastos
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Av. do Café s/n, 14040-903 Ribeirão Preto, São Paulo, Brazil
| | - Rosangela S Laurentiz
- Faculdade de Engenharia de Ilha Solteira, Universidade Estadual Paulista Júlio de Mesquita Filho, Avenida Brasil 56, 15385-000 Ilha Solteira, São Paulo, Brazil
| | - Tapas Kar
- Department of Chemistry and Biochemistry, Utah State University, Logan, Utah, 84322-0300, USA
| | - Giovanni F Caramori
- Departamento de Química, Universidade Federal de Santa Catarina, Campus Universitário Trindade, 88040-900, CP 476 Florianópolis, Santa Catarina, Brazil
| | - Daniel Fábio Kawano
- Universidade de Campinas, Faculdade de Ciências Farmacêuticas, Rua Cândido Portinari 200, 13083-871 Campinas, São Paulo, Brazil
| | - Márcio L Andrade E Silva
- Núcleo de Pesquisas em Ciências Exatas e Tecnológicas, Universidade de Franca, Av. Dr. Armando Salles Oliveira 201, 14404-600 Franca, São Paulo, Brazil
| |
Collapse
|
63
|
Abstract
Schistosomiasis is a serious but neglected tropical infectious disease, afflicting more than 240 million people in 78 countries. Lack of an effective vaccine and obscuring disease mechanism could be the main hurdles to effectively control and eradicate this disease. A better understanding of the host-schistosome interaction is the key to clearing these hurdles. Recently, accumulating evidence shows that alarmin cytokines and microRNAs (miRNAs) are crucial regulators in the host-schistosome interaction. Alarmin cytokines are proven to be potent mechanisms driving type 2 immunity, which is the central disease mechanism of schistosomiasis. MiRNA deregulation is a hallmark of a variety of human diseases, including schistosomiasis. In this review, we summarize the research advances on the role of alarmin cytokines and miRNAs in the host-schistosome interaction.
Collapse
Affiliation(s)
- Xing He
- Department of Tropical diseases, Second Military Medical University, Shanghai, 200433, China
| | - Weiqing Pan
- Department of Tropical diseases, Second Military Medical University, Shanghai, 200433, China
| |
Collapse
|
64
|
Viana AG, Coelho ALG, Gazzinelli-Guimarães PH, Phillips AE, Bartholomeu DC, Bueno LL, Fujiwara RT. Development of the PraziCalc mobile device-app to calculate praziquantel dosage in the treatment of schistosomiasis. Rev Inst Med Trop Sao Paulo 2018; 60:e42. [PMID: 30133602 PMCID: PMC6103324 DOI: 10.1590/s1678-9946201860042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 07/11/2018] [Indexed: 11/08/2023] Open
Affiliation(s)
- Agostinho Gonçalves Viana
- Universidade Federal de Minas Gerais, Instituto de Ciências
Biológicas, Departamento de Parasitologia, Laboratório de Imunologia e Genômica de
Parasitos, Belo Horizonte, Minas Gerais, Brazil
| | - André Luiz Gomes Coelho
- Universidade Federal de Minas Gerais, Instituto de Ciências
Biológicas, Departamento de Parasitologia, Laboratório de Imunologia e Genômica de
Parasitos, Belo Horizonte, Minas Gerais, Brazil
| | | | | | - Daniella Castanheira Bartholomeu
- Universidade Federal de Minas Gerais, Instituto de Ciências
Biológicas, Departamento de Parasitologia, Laboratório de Imunologia e Genômica de
Parasitos, Belo Horizonte, Minas Gerais, Brazil
| | - Lilian Lacerda Bueno
- Universidade Federal de Minas Gerais, Instituto de Ciências
Biológicas, Departamento de Parasitologia, Laboratório de Imunologia e Genômica de
Parasitos, Belo Horizonte, Minas Gerais, Brazil
| | - Ricardo Toshio Fujiwara
- Universidade Federal de Minas Gerais, Instituto de Ciências
Biológicas, Departamento de Parasitologia, Laboratório de Imunologia e Genômica de
Parasitos, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
65
|
Abstract
Schistosomiasis (bilharzia) is a neglected tropical disease caused by parasitic flatworms (blood flukes) of the genus Schistosoma, with considerable morbidity in parts of the Middle East, South America, Southeast Asia and, particularly, in sub-Saharan Africa. Infective larvae grow in an intermediate host (fresh-water snails) before penetrating the skin of the definitive human host. Mature adult worms reside in the mesenteric (Schistosoma mansoni and Schistosoma japonicum) or pelvic (Schistosoma haematobium) veins, where female worms lay eggs, which are secreted in stool or urine. Eggs trapped in the surrounding tissues and organs, such as the liver and bladder, cause inflammatory immune responses (including granulomas) that result in intestinal, hepato-splenic or urogenital disease. Diagnosis requires the detection of eggs in excreta or worm antigens in the serum, and sensitive, rapid, point-of-care tests for populations living in endemic areas are needed. The anti-schistosomal drug praziquantel is safe and efficacious against adult worms of all the six Schistosoma spp. infecting humans; however, it does not prevent reinfection and the emergence of drug resistance is a concern. Schistosomiasis elimination will require a multifaceted approach, including: treatment; snail control; information, education and communication; improved water, sanitation and hygiene; accurate diagnostics; and surveillance-response systems that are readily tailored to social-ecological settings.
Collapse
Affiliation(s)
- Donald P McManus
- Immunology Department, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia.
| | - David W Dunne
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Moussa Sacko
- Department of Diagnostic and Biomedical Research, Institut National de Recherche en Santé Publique, Bamako, Mali
| | - Jürg Utzinger
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Birgitte J Vennervald
- Department of Veterinary and Animal Science, University of Copenhagen, Copenhagen, Denmark
| | - Xiao-Nong Zhou
- National Institute of Parasitic Diseases, Shanghai, People's Republic of China
| |
Collapse
|
66
|
Macrophage Activation and Functions during Helminth Infection: Recent Advances from the Laboratory Mouse. J Immunol Res 2018; 2018:2790627. [PMID: 30057915 PMCID: PMC6051086 DOI: 10.1155/2018/2790627] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 05/23/2018] [Indexed: 12/20/2022] Open
Abstract
Macrophages are highly plastic innate immune cells that adopt an important diversity of phenotypes in response to environmental cues. Helminth infections induce strong type 2 cell-mediated immune responses, characterized among other things by production of high levels of interleukin- (IL-) 4 and IL-13. Alternative activation of macrophages by IL-4 in vitro was described as an opposite phenotype of classically activated macrophages, but the in vivo reality is much more complex. Their exact activation state as well as the role of these cells and associated molecules in type 2 immune responses remains to be fully understood. We can take advantage of a variety of helminth models available, each of which have their own feature including life cycle, site of infection, or pathological mechanisms influencing macrophage biology. Here, we reviewed the recent advances from the laboratory mouse about macrophage origin, polarization, activation, and effector functions during parasitic helminth infection.
Collapse
|
67
|
Pacheco PAF, Dantas LP, Ferreira LGB, Faria RX. Purinergic receptors and neglected tropical diseases: why ignore purinergic signaling in the search for new molecular targets? J Bioenerg Biomembr 2018; 50:307-313. [PMID: 29882206 DOI: 10.1007/s10863-018-9761-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 05/07/2018] [Indexed: 12/20/2022]
Abstract
Purinergic receptors are widespread in the human organism and are involved in several physiological functions like neurotransmission, nociception, platelet aggregation, etc. In the immune system, they may regulate the expression and release of pro-inflammatory factors as well as the activation and death of several cell types. It is already described the participation of some purinergic receptors in the inflammation and pathological processes, such as a few neglected tropical diseases (NTDs) which affect more than 1 billion people in the world. Although the high social influence those diseases represent endemic countries, most of them do not have an efficient, safe or affordable drug treatment. In that way, this review aims to discuss the current literature involving purinergic receptor and immune response to NTDs pathogens, which may contribute in the search for new therapeutic possibilities.
Collapse
Affiliation(s)
- P A F Pacheco
- Department of Chemistry, Chemistry Institute, Fluminense Federal University, Niterói, Brazil
| | - L P Dantas
- Laboratory of Molecular Virology, Biology Institute, Fluminense Federal University, Niterói, Brazil
| | - L G B Ferreira
- Laboratory of Inflammation, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Robson Xavier Faria
- Laboratory of Toxoplasmosis and other Protozoans, FIOCRUZ, Oswaldo Cruz Institute, Rio de Janeiro, Brazil. .,Fundação Oswaldo Cruz, Laboratório de Toxoplasmose e outras Protozooses, Instituto Oswaldo Cruz, Avenida Brasil 4365, sala 32; Manguinhos, Rio de Janeiro, RJ, CEP 21045-900, Brazil.
| |
Collapse
|
68
|
Standley CJ, Graeden E, Kerr J, Sorrell EM, Katz R. Decision support for evidence-based integration of disease control: A proof of concept for malaria and schistosomiasis. PLoS Negl Trop Dis 2018; 12:e0006328. [PMID: 29649260 PMCID: PMC5896906 DOI: 10.1371/journal.pntd.0006328] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 02/19/2018] [Indexed: 11/19/2022] Open
Abstract
AUTHOR SUMMARY Designing and implementing effective programs for infectious disease control requires complex decision-making, informed by an understanding of the diseases, the types of disease interventions and control measures available, and the disease-relevant characteristics of the local community. Though disease modeling frameworks have been developed to address these questions and support decision-making, the complexity of current models presents a significant barrier to on-the-ground end users. The picture is further complicated when considering approaches for integration of different disease control programs, where co-infection dynamics, treatment interactions, and other variables must also be taken into account. Here, we describe the development of an application available on the internet with a simple user interface, to support on-the-ground decision-making for integrating disease control, given local conditions and practical constraints. The model upon which the tool is built provides predictive analysis for the effectiveness of integration of schistosomiasis and malaria control, two diseases with extensive geographical and epidemiological overlap. This proof-of-concept method and tool demonstrate significant progress in effectively translating the best available scientific models to support pragmatic decision-making on the ground, with the potential to significantly increase the impact and cost-effectiveness of disease control.
Collapse
Affiliation(s)
- Claire J. Standley
- Center for Global Health Science and Security, Georgetown University, Washington, DC, United States of America
- * E-mail:
| | - Ellie Graeden
- Talus Analytics, LLC, Lyons, CO, United States of America
| | - Justin Kerr
- Talus Analytics, LLC, Lyons, CO, United States of America
| | - Erin M. Sorrell
- Center for Global Health Science and Security, Georgetown University, Washington, DC, United States of America
| | - Rebecca Katz
- Center for Global Health Science and Security, Georgetown University, Washington, DC, United States of America
| |
Collapse
|
69
|
Blay EA, Kumagai T, Yamabe M, Hino A, Shimogawara R, Kim HS, Sato A, Ichimura K, Ayi I, Iwanaga S, Ohta N. Insights into the mode of action of 1,2,6,7-tetraoxaspiro [7.11] nonadecane (N-89) against adult Schistosoma mansoni worms. Parasitol Int 2018; 67:403-412. [PMID: 29617630 DOI: 10.1016/j.parint.2018.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 03/14/2018] [Accepted: 03/28/2018] [Indexed: 01/16/2023]
Abstract
Control of morbidity associated with schistosomiasis via chemotherapy largely relies on the drug praziquantel. Repeated therapy with praziquantel has created concerns about the possible selection of resistant worms and necessitated the search for novel drugs to treat schistosomiasis. Here, a murine model was infected with Schistosoma mansoni and treated with oral 1,2,6,7-tetraoxaspiro [7.11] nonadecane (N-89), which caused a significant reduction in fecundity and egg burden and reduced morbidity when administered at 5-weeks post-infection. The analysis showed that the mode of action occurred through the ingestion of activated N-89 by the worms, and that there was no direct external effect on the S. mansoni worms. Ultrastructural analysis of the treated worms showed disruptions in the gut lumen and the presence of large volumes of material, suggestive of undigested blood meals or red blood cells. In addition, there were reduced vitelline cells in female worms and damage to sub-tegmental musculature in male worms. Eggs recovered from the treated mice showed both damage to the eggs and the production of immature eggs. Expression of mRNA responsible for gut and digestive function and egg production was also significantly affected by N-89 treatment, whereas control genes for musculature showed no significant changes. Thus, N-89 drastically affected the total digestive function and egg production of S. mansoni worms. Physiological processes requiring heme uptake such as egg production and eggshell formation were subsequently affected, suggesting that the compound could be a possible therapeutic drug candidate for schistosomiasis control.
Collapse
Affiliation(s)
- Emmanuel Awusah Blay
- Department of Environmental Parasitology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Takashi Kumagai
- Department of Environmental Parasitology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Masafumi Yamabe
- Department of Environmental Parasitology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Akina Hino
- Department of Environmental Parasitology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Rieko Shimogawara
- Department of Environmental Parasitology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Hye-Sook Kim
- Division of International Infectious Disease Control, Faculty of Pharmaceutical Sciences, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| | - Akira Sato
- Division of International Infectious Disease Control, Faculty of Pharmaceutical Sciences, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| | - Koichiro Ichimura
- Department of Anatomy and Life Structure, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Irene Ayi
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, P. O. Box LG581, Legon, Accra, Ghana
| | - Shiroh Iwanaga
- Department of Environmental Parasitology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Nobuo Ohta
- Department of Environmental Parasitology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan.
| |
Collapse
|
70
|
Schistosomiasis and Strongyloidiasis Recommendations for Solid-Organ Transplant Recipients and Donors. Transplantation 2018; 102:S27-S34. [DOI: 10.1097/tp.0000000000002016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
71
|
Schistosoma mansoni and endocarditis: from egg to free DNA detection in Egyptian patients and infected BALB/c mice. J Helminthol 2018; 93:139-148. [PMID: 29352830 DOI: 10.1017/s0022149x17001183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
With the growing incidence of closed schistosomiasis and uncommon presentations, there is a risk of the infection rate being underestimated. A study in Japan reported an unexplained case of endocarditis that was finally diagnosed as a complex Schistosoma japonicum infection; in the absence of advanced techniques, the diagnosis was delayed. We therefore set out to explore the incidence of Schistosoma mansoni in endocarditis patients coming from areas of Egypt where S. mansoni is endemic. We also investigated histopathological changes in the cardiac valves and the presence of cell-free parasite DNA (CFPD) in cardiac tissues of laboratory mice infected with S. mansoni. The study included 186 patients with the manifestations of infective endocarditis. Eggs were detected in the stool samples of 5.91% of patients. Seropositivity was reported in 23.66% of patients and antigen was detected in the urine samples of 10.21%. Using real-time polymerase chain reaction (PCR), CFPD was detected in the blood of 6.98% of the endocarditis patients and 95% of the infected mice, while the cardiac samples of 45% of the mice tested positive for CFPD (means ± SD = 1390.2 ± 283.65, 2158.72 ± 1103.1 and 5.71 ± 2.91, respectively). Histopathological examination revealed abnormal collagen deposition, inflammatory cells and haemorrhagic pigmentation in the heart sections. Despite the low incidence of S. mansoni infection in the studied cohort, the presence of CFPD in the cardiac tissue of infected mice makes it necessary to: (1) investigate the hazards of CFPD deposition in endothelium-rich organs; and (2) test the potential of CFPD to trigger tissue inflammation, abnormal proliferation or genome integration.
Collapse
|
72
|
Friedman JF, Olveda RM, Mirochnick MH, Bustinduy AL, Elliott AM. Praziquantel for the treatment of schistosomiasis during human pregnancy. Bull World Health Organ 2017; 96:59-65. [PMID: 29403101 PMCID: PMC5791873 DOI: 10.2471/blt.17.198879] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 10/31/2017] [Accepted: 11/15/2017] [Indexed: 02/08/2023] Open
Abstract
In 2014, an estimated 40 million women of reproductive age were infected with Schistosoma haematobium, S. japonicum and/or S. mansoni. In both 2003 and 2006, the World Health Organization (WHO) recommended that all schistosome-infected pregnant and breastfeeding women be offered treatment, with praziquantel, either individually or during treatment campaigns. In 2006, WHO also stated the need for randomized controlled trials to assess the safety and efficacy of such treatment. Some countries have yet to follow the recommendation on treatment and many programme managers and pregnant women in other countries remain reluctant to follow the recommended approach. Since 2006, two randomized controlled trials on the use of praziquantel during pregnancy have been conducted: one against S. mansoni in Uganda and the other against S. japonicum in the Philippines. In these trials, praziquantel treatment of pregnant women had no significant effect on birth weight, appeared safe and caused minimal side-effects that were similar to those seen in treated non-pregnant subjects. Having summarized the encouraging data, on efficacy, pharmacokinetics and safety, from these two trials and reviewed the safety data from non-interventional human studies, we recommend that all countries include pregnant women in praziquantel treatment campaigns. We identify the barriers to the treatment of pregnant women, in countries that already include such women in individual treatments and mass drug administration campaigns, and discuss ways to address these barriers.
Collapse
Affiliation(s)
- Jennifer F Friedman
- Center for International Health Research at Rhode Island Hospital, 55 Claverick Street, Suite 101, Providence, RI 02903, United States of America (USA)
| | - Remigio M Olveda
- Department of Immunology, Research Institute for Tropical Medicine, Manila, Philippines
| | - Mark H Mirochnick
- Division of Neonatology, Department of Pediatrics, Boston University School of Medicine, Boston, USA
| | - Amaya L Bustinduy
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, England
| | - Alison M Elliott
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, England
| |
Collapse
|
73
|
The Interdependence between Schistosome Transmission and Protective Immunity. Trop Med Infect Dis 2017; 2:tropicalmed2030042. [PMID: 30270899 PMCID: PMC6082113 DOI: 10.3390/tropicalmed2030042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 08/02/2017] [Accepted: 08/08/2017] [Indexed: 01/14/2023] Open
Abstract
Mass drug administration (MDA) for control of schistosomiasis is likely to affect transmission dynamics through a combination of passive vaccination and reduction of local transmission intensity. This is indicated in phenomenological models of immunity and the impact of MDA, yet immunity parameters in these models are not validated by empirical data that reflects protective immunity to reinfection. There is significant empirical evidence supporting the role of IgE in acquired protective immunity. This is proposed to be a form of delayed concomitant immunity, driven at least in part by protective IgE responses to the tegument allergen-like (TAL) family of proteins. Specific questions have arisen from modeling studies regarding the strength and duration of the protective immune response. At present, field studies have not been specifically designed to address these questions. There is therefore a need for field studies that are explicitly designed to capture epidemiological effects of acquired immunity to elucidate these immunological interactions. In doing so, it is important to address the discourse between theoretical modelers and immuno-epidemiologists and develop mechanistic models that empirically define immunity parameters. This is of increasing significance in a climate of potential changing transmission dynamics following long-term implementation of MDA.
Collapse
|
74
|
Sun LP, Wang W, Zuo YP, Hong QB, Du GL, Ma YC, Wang J, Yang GJ, Zhu DJ, Liang YS. A multidisciplinary, integrated approach for the elimination of schistosomiasis: a longitudinal study in a historically hyper-endemic region in the lower reaches of the Yangtze River, China from 2005 to 2014. Infect Dis Poverty 2017; 6:56. [PMID: 28288689 PMCID: PMC5348877 DOI: 10.1186/s40249-017-0270-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 02/27/2017] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Although great success has been achieved, schistosomiasis remains a major public health concern in China, and the remaining core endemic regions are concentrated along the middle and lower reaches of the Yangtze River. In this longitudinal study, we evaluated the effectiveness of a multidisciplinary, integrated approach for schistosomiasis elimination in a historically hyper-endemic region in the lower reaches of the Yangtze River, China over the 10-year period from 2005 through 2014. METHODS A three-step roadmap for schistosomiasis elimination was designed in the study site, and multidisciplinary, integrated interventions were implemented by the health, agriculture, water resources development, land and resources, and forestry sectors from 2005 to 2014, including chemotherapy for infected individuals, health education, management of the source of Schistosoma japonicum infection, and intermediate host snail control. The annual number of schistosomiasis patients, S. japonicum infection in humans, bovines and Oncomelania hupensis snails, and water infectivity were observed to assess the effectiveness of the multidisciplinary, integrated approach for the elimination of schistosomiasis. RESULTS There was a tendency towards a gradual decline in both the number of schistosomiasis cases and the prevalence of S. japonicum human infection across the study period from 2005 through 2014. No S. japonicum human infection was detected since 2012, and no acute infection was seen since 2006. During the study period, no infection was found in bovines, and a 0.03% overall infection rate was observed in O. hupensis snails. Since 2009, no infected snails were identified, and the area of both snail habitats and infected snail habitats appeared a reduction over the study period. Following the 3-year multidisciplinary, integrated control, infection control was achieved, and transmission control was achieved after 6-year implementation, with all infected snails and water infectivity eliminated; in addition, the 10-year implementation resulted in interruption of schistosomiasis transmission in the study site in 2014. CONCLUSIONS The results of the present 10-year longitudinal study demonstrate that the multidisciplinary, integrated approach is effective for the elimination of schistosomiasis as a public health problem in the lower reaches of the Yangtze River, China.
Collapse
Affiliation(s)
- Le-Ping Sun
- Key Laboratory of National Health and Family Planning Commission on Parasitic Disease Control and Prevention, No. 117 Yangxiang, Meiyuan, Wuxi City, Jiangsu Province 214064 China
- Jiangsu Provincial Key Laboratory on Parasites and Vector Control Technology, No. 117 Yangxiang, Meiyuan, Wuxi City, Jiangsu Province 214064 China
- Jiangsu Institute of Parasitic Diseases, No. 117 Yangxiang, Meiyuan, Wuxi City, Jiangsu Province 214064 China
| | - Wei Wang
- Key Laboratory of National Health and Family Planning Commission on Parasitic Disease Control and Prevention, No. 117 Yangxiang, Meiyuan, Wuxi City, Jiangsu Province 214064 China
- Jiangsu Provincial Key Laboratory on Parasites and Vector Control Technology, No. 117 Yangxiang, Meiyuan, Wuxi City, Jiangsu Province 214064 China
- Jiangsu Institute of Parasitic Diseases, No. 117 Yangxiang, Meiyuan, Wuxi City, Jiangsu Province 214064 China
| | - Yin-Ping Zuo
- Yangzhou Municipal Center for Disease Control and Prevention, No. 36 Yanfu East Road, Yangzhou City, Jiangsu Province 225000 China
| | - Qing-Biao Hong
- Key Laboratory of National Health and Family Planning Commission on Parasitic Disease Control and Prevention, No. 117 Yangxiang, Meiyuan, Wuxi City, Jiangsu Province 214064 China
- Jiangsu Provincial Key Laboratory on Parasites and Vector Control Technology, No. 117 Yangxiang, Meiyuan, Wuxi City, Jiangsu Province 214064 China
- Jiangsu Institute of Parasitic Diseases, No. 117 Yangxiang, Meiyuan, Wuxi City, Jiangsu Province 214064 China
| | - Guang-Lin Du
- Yangzhou Municipal Center for Disease Control and Prevention, No. 36 Yanfu East Road, Yangzhou City, Jiangsu Province 225000 China
| | - Yu-Cai Ma
- Hanjiang District Center for Disease Control and Prevention, Wenhui West Road, Yangzhou City, Jiangsu Province 225000 China
| | - Jian Wang
- Yangzhou Municipal Center for Disease Control and Prevention, No. 36 Yanfu East Road, Yangzhou City, Jiangsu Province 225000 China
| | - Guo-Jing Yang
- Key Laboratory of National Health and Family Planning Commission on Parasitic Disease Control and Prevention, No. 117 Yangxiang, Meiyuan, Wuxi City, Jiangsu Province 214064 China
- Jiangsu Provincial Key Laboratory on Parasites and Vector Control Technology, No. 117 Yangxiang, Meiyuan, Wuxi City, Jiangsu Province 214064 China
- Jiangsu Institute of Parasitic Diseases, No. 117 Yangxiang, Meiyuan, Wuxi City, Jiangsu Province 214064 China
| | - Dao-Jian Zhu
- Yangzhou Municipal Center for Disease Control and Prevention, No. 36 Yanfu East Road, Yangzhou City, Jiangsu Province 225000 China
| | - You-Sheng Liang
- Key Laboratory of National Health and Family Planning Commission on Parasitic Disease Control and Prevention, No. 117 Yangxiang, Meiyuan, Wuxi City, Jiangsu Province 214064 China
- Jiangsu Provincial Key Laboratory on Parasites and Vector Control Technology, No. 117 Yangxiang, Meiyuan, Wuxi City, Jiangsu Province 214064 China
- Jiangsu Institute of Parasitic Diseases, No. 117 Yangxiang, Meiyuan, Wuxi City, Jiangsu Province 214064 China
| |
Collapse
|