1
|
Obonyo CO, Were VO, Wamae P, Muok EMO. Efficacy and safety of praziquantel plus artemisinin-based combinations versus praziquantel in the treatment of Kenyan children with Schistosoma mansoni infection: open-label, randomized, head-to-head, non-inferiority trial. Antimicrob Agents Chemother 2025; 69:e0073924. [PMID: 39699212 PMCID: PMC11823657 DOI: 10.1128/aac.00739-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 11/06/2024] [Indexed: 12/20/2024] Open
Abstract
Praziquantel alone is insufficient for the control of schistosomiasis due to poor efficacy against juvenile worms and increasing concerns about the risk of drug resistance. We compared the efficacy and safety of praziquantel combined with four different artemisinin-based combinations to praziquantel alone in treating Schistosoma mansoni infection in Kenyan children. In this randomized, open-label, five-arm, head-to-head, non-inferiority trial, children (aged 9-15 years) with S. mansoni infection according to duplicate Kato Katz thick smears from a stool sample in the Mwea irrigation scheme of central Kenya, were enrolled. Participants were randomly assigned (1:1:1:1:1) via a computer-generated block randomization procedure to receive a single oral dose of praziquantel (PZQ) (40 mg/kg/day) alone or in combination with a 3-day course (4 mg/kg of artesunate) of artesunate plus sulfalene-pyrimethamine (As + SP), artesunate plus amodiaquine (As + AQ), artesunate plus mefloquine (As + MQ) or dihydroartemisinin-piperaquine (DHAP). Laboratory technicians were masked to treatment allocation, but participants, clinicians, and study nurses were not. The primary outcomes were the cure rate and frequency of adverse events, which were assessed 6 weeks after treatment in the available case population using a per-protocol analysis. The non-inferiority margin was set at -10% for the risk difference in cure rates between combination therapy and PZQ alone. Between 12 September 2018 and 11 January 2019, 540 participants were assigned to receive PZQ alone (n = 108), PZQ plus As + SP (n = 108), PZQ plus As + AQ (n = 108), PZQ plus As + MQ (n = 108), or PZQ plus DHAP (n = 108). Primary outcome data were available for 523 (96.9%) participants. The cure rate was 82.5% (85/103) in PZQ alone, 81.7% (85/104) in PZQ plus As + SP, 76.2% (80/105) in PZQ plus As + AQ, 88.7% (94/106) in PZQ plus As + MQ, and 85.7% (90/105) in PZQ plus DHAP arm. Non-inferiority was declared for PZQ plus As + MQ (difference 6.2 [95% confidence interval: -3.3 to 15.6]) and PZQ plus DHAP (3.2 [-6.7 to 13.1]) but not for PZQ plus As + SP (-0.8 [-11.2 to 9.6]) or PZQ plus As + AQ (-6.3 [-17.3 to 4.6]). Adverse events were reported by 26% (138/540) of participants, including abdominal pain, headache, and vomiting. There were no serious adverse events. Alternatives to praziquantel should include praziquantel plus artesunate-mefloquine or praziquantel plus dihydroartemisinin-piperaquine. However, further multicentre trials are needed in different epidemiological settings and population groups to confirm these findings.CLINICAL TRIALSThis study is registered with the Pan-African Clinical Trials Registry under PACTR202001919442161.
Collapse
Affiliation(s)
- Charles O. Obonyo
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Vincent O. Were
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Peter Wamae
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Erick M. O. Muok
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| |
Collapse
|
2
|
de Castro Carvalho Silva L, Cunha Dos Reis LF, Malaquias LCC, Carvalho FC, Novaes RD, Marques MJ. Impact of nanostructured formulations for schistosomiasis treatment: a systematic review of in vivo preclinical evidence. J Pharm Pharmacol 2025:rgae155. [PMID: 39820345 DOI: 10.1093/jpp/rgae155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 12/26/2024] [Indexed: 01/19/2025]
Abstract
BACKGROUND Schistosomiasis is a neglected tropical disease caused by Schistosoma sp., and praziquantel (PZQ) is the first-line treatment. However, traditional PZQ formulations have low solubility and fast metabolism, limiting its effectiveness. Thus, nanoparticles have been proposed to improve the bioavailability and efficacy of poorly soluble antischistosomal drugs. AIMS This systematic review used in vivo preclinical studies to map the available evidence and compare the efficacy of free PZQ and PZQ-based nanostructured formulations (N-PZQ) for schistosomiasis treatment. METHODS PubMed, Embase, Scopus, and Web of Science were searched, and 1186 experimental studies published between 1974 and 2024 were screened. Parasitological, histopathological, pharmacokinetic, and toxicological outcomes were evaluated. RESULTS Twelve relevant studies were identified exploring N-PZQ formulations based on liposomes, nanoliposomes, and nanocrystals. N-PZQ demonstrated better therapeutic efficacy than free PZQ, reducing parasite load, modifying oogram profiles, and down-regulating liver granuloma development (number and size). N-PZQ also exhibited improved pharmacokinetic profile, with enhanced bioavailability and longer half-life, as well as reduced toxicity (cytotoxicity, genotoxicity, and hepatotoxicity) compared to free PZQ. CONCLUSION PZQ-based nanostructured formulations represent a promising strategy to enhance schistosomiasis treatment by improving chemotherapy efficacy, optimizing antiparasitic responses, pharmacokinetics, and reducing drug toxicity.
Collapse
Affiliation(s)
- Laís de Castro Carvalho Silva
- Departamento de Parasitologia e Patologia, Universidade Federal de Alfenas, Alfenas, 37130-001, Minas Gerais, Brazil
| | - Luís Felipe Cunha Dos Reis
- Departamento de Biologia Estrutural, Universidade Federal de Alfenas, Alfenas, 37130-001, Minas Gerais, Brazil
| | - Luiz Cosme Cotta Malaquias
- Departamento de Microbiologia e Imunologia, Universidade Federal de Alfenas, Alfenas, 37130-001, Minas Gerais, Brazil
| | - Flávia Chiva Carvalho
- Departamento de Alimentos e Medicamentos Faculdade de Ciências Farmacêuticas, Universidade Federal de Alfenas, Alfenas, 37130-001, Minas Gerais, Brazil
| | - Rômulo Dias Novaes
- Departamento de Biologia Estrutural, Universidade Federal de Alfenas, Alfenas, 37130-001, Minas Gerais, Brazil
| | - Marcos José Marques
- Departamento de Parasitologia e Patologia, Universidade Federal de Alfenas, Alfenas, 37130-001, Minas Gerais, Brazil
| |
Collapse
|
3
|
Motlhatlhedi K, Pilusa NB, Ndaba T, George M, Masamba P, Kappo AP. Therapeutic and vaccinomic potential of moonlighting proteins for the discovery and design of drugs and vaccines against schistosomiasis. Am J Transl Res 2024; 16:4279-4300. [PMID: 39398578 PMCID: PMC11470331 DOI: 10.62347/bxrt7210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 07/15/2024] [Indexed: 10/15/2024]
Abstract
Despite significant and coordinated efforts to combat schistosomiasis, such as providing clean water, sanitation, hygiene, and snail control, these strategies still fall short, as regions previously thought to be disease-free have shown active schistosomiasis transmission. Therefore, it is necessary to implement integrated control methods, emphasizing vaccine development for sustainable control of schistosomiasis. Vaccination has significantly contributed to global healthcare and has been the most economically friendly method for avoiding pathogenic infections. Over the years, different vaccine candidates for schistosomiasis have been investigated with varying degrees of success in clinical trials with many not proceeding past the early clinical phase. Recently, proteins have been mentioned as targets for drug discovery and vaccine development, especially those with multiple functions in schistosomes. Moonlighting proteins are a class of proteins that can perform several functions besides their known functions. This multifunctional property is believed to have been expressed through evolution, where the polypeptide chain gained the ability to perform other tasks without undergoing any structural changes. Since proteins have gained more traction as drug targets, multifunctional proteins have thus become attractive for discovering and developing novel drugs since the drug can target more than one function. Moonlighting proteins are promising drug and vaccine candidates for diseases such as schistosomiasis, since they aid in disease promotion in the human host. This manuscript elucidates vital moonlighting proteins used by schistosomes to drive their life cycle and to ensure their survival in the human host, which can be used to develop anti-schistosomal therapeutics and vaccinomics.
Collapse
Affiliation(s)
- Kagiso Motlhatlhedi
- Molecular Biophysics and Structural Biology (MBBS) Group, Department of Biochemistry, Faculty of Science, University of Johannesburg, Auckland Park Kingsway Campus Auckland Park, Johannesburg, South Africa
| | - Naledi Beatrice Pilusa
- Molecular Biophysics and Structural Biology (MBBS) Group, Department of Biochemistry, Faculty of Science, University of Johannesburg, Auckland Park Kingsway Campus Auckland Park, Johannesburg, South Africa
| | - Tshepang Ndaba
- Molecular Biophysics and Structural Biology (MBBS) Group, Department of Biochemistry, Faculty of Science, University of Johannesburg, Auckland Park Kingsway Campus Auckland Park, Johannesburg, South Africa
| | - Mary George
- Molecular Biophysics and Structural Biology (MBBS) Group, Department of Biochemistry, Faculty of Science, University of Johannesburg, Auckland Park Kingsway Campus Auckland Park, Johannesburg, South Africa
| | - Priscilla Masamba
- Molecular Biophysics and Structural Biology (MBBS) Group, Department of Biochemistry, Faculty of Science, University of Johannesburg, Auckland Park Kingsway Campus Auckland Park, Johannesburg, South Africa
| | - Abidemi Paul Kappo
- Molecular Biophysics and Structural Biology (MBBS) Group, Department of Biochemistry, Faculty of Science, University of Johannesburg, Auckland Park Kingsway Campus Auckland Park, Johannesburg, South Africa
| |
Collapse
|
4
|
Obonyo CO, Rawago FO, Makworo NK, Muok EMO. Efficacy and safety of single-dose artesunate plus sulfalene/pyrimethamine combined with praziquantel for the treatment of children with Schistosoma mansoni or Schistosoma haematobium in western Kenya: a randomised, open-label controlled trial. Parasit Vectors 2024; 17:279. [PMID: 38943214 PMCID: PMC11212220 DOI: 10.1186/s13071-024-06359-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 06/16/2024] [Indexed: 07/01/2024] Open
Abstract
BACKGROUND Reliance on praziquantel for the treatment and control of schistosomiasis is likely to facilitate the emergence of drug resistance. Combination therapy targeting adult and juvenile schistosome worms is urgently needed to improve praziquantel efficacy and delay the potential development of drug resistance. We assessed the efficacy and safety of single-dose praziquantel combined with single-dose artesunate plus sulfalene-pyrimethamine in the treatment of Kenyan children with schistosomiasis. METHODS This was an open-label, randomised clinical trial involving 426 school-aged children (7-15 years old) diagnosed with Schistosoma mansoni (by Kato-Katz) or S. haematobium (by urine filtration). They were randomly assigned (1:1:1) to receive a single dose of praziquantel (40 mg/kg), a single dose of artesunate plus sulfalene-pyrimethamine (12 mg/kg artesunate) or combination therapy using a single dose of praziquantel (40 mg/kg) combined with a single dose of artesunate plus sulfalene-pyrimethamine (12 mg/kg artesunate). The primary outcome was cure and egg reduction rates at 6 weeks post-treatment in the available case population. Adverse events were assessed within 3 h after treatment. RESULTS Of the 426 children enrolled, 135 received praziquantel, 150 received artesunate plus sulfalene-pyrimethamine, and 141 received combination therapy. Outcome data were available for 348 (81.7%) children. For S. mansoni-infected children (n = 335), the cure rates were 75.6%, 60.7%, and 77.8%, and the egg reduction rates were 80.1%, 85.0%, and 88.4% for praziquantel, artesunate plus sulfalene-pyrimethamine, and combination therapy, respectively. For S. haematobium-infected children (n = 145), the corresponding cure rates were 81.4%, 71.1%, and 82.2%, and the egg reduction rates were 95.6%, 97.1%, and 97.7%, respectively. Seventy-one (16.7%) children reported mild-intensity adverse events. The drugs were well tolerated and no serious adverse events were reported. CONCLUSIONS A single oral dose of praziquantel combined with artesunate plus sulfalene-pyrimethamine cured a high proportion of children with S. haematobium but did not significantly improve the treatment efficacy for either urinary or intestinal schistosomiasis. Sequential administration of praziquantel and artesunate plus sulfalene-pyrimethamine may enhance the efficacy and safety outcomes.
Collapse
Affiliation(s)
- Charles O Obonyo
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya.
| | - Fredrick O Rawago
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Nicholas K Makworo
- Division of Vector-Borne and Neglected Tropical Diseases, County Department of Health, Migori, Kenya
| | - Erick M O Muok
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| |
Collapse
|
5
|
Muok EMO, Were VO, Obonyo CO. Efficacy of a Single Oral Dose of Artesunate plus Sulfalene-Pyrimethamineversus Praziquantel in the Treatment of Schistosoma mansoni in Kenyan Children: An Open-Label, Randomized, Exploratory Trial. Am J Trop Med Hyg 2024; 110:677-680. [PMID: 38460198 PMCID: PMC10993826 DOI: 10.4269/ajtmh.23-0337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 12/09/2023] [Indexed: 03/11/2024] Open
Abstract
Unlike praziquantel, artemisinin derivatives are effective against juvenile schistosome worms. We assessed the efficacy and safety of a single oral dose of artesunate plus sulfalene-pyrimethamine versus praziquantel in the treatment of Schistosoma mansoni. Seventy-three schoolchildren (aged 9-15 years) with confirmed S. mansoni infection in Rarieda, western Kenya, were randomly assigned to receive either a single oral dose of artesunate plus sulfalene-pyrimethamine (n = 39) or a single dose of praziquantel (n = 34). The cure and egg reduction rates at 4 weeks posttreatment were 69.4% (25/36) versus 80.6% (25/31) (P = 0.297) and 99.1% versus 97.5% (P = 0.607) in the artesunate plus sulfalene-pyrimethamine group versus praziquantel group, respectively. Fourteen children developed adverse events, and there were no serious adverse events. A single oral dose of artesunate plus sulfalene-pyrimethamine has efficacy comparable to that of praziquantel in the treatment of S. mansoni, but these results should be confirmed in larger randomized controlled trials.
Collapse
Affiliation(s)
- Erick M. O. Muok
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Vincent O. Were
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Charles O. Obonyo
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| |
Collapse
|
6
|
Osei-Mensah B, Boakye YD, Anyan WK, Agana TA, Aboagye EA, Bentil I, Lomotey ES, Adu F, Agyare C. In Vitro Cercaricidal Activity, Acute Toxicity, and GC/MS Analysis of Some Selected Ghanaian Medicinal Plants. J Parasitol Res 2023; 2023:4589424. [PMID: 37745984 PMCID: PMC10516696 DOI: 10.1155/2023/4589424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 07/28/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023] Open
Abstract
Schistosomiasis is a human parasitic disease caused by the Schistosoma species and is recognised in public health as second to malaria in terms of its socioeconomic impact on humans. Four local plants native to many tribes in Ghana and known for their medicinal properties against some diseases were assessed for their cercaricidal activity against Schistosoma mansoni cercariae. The plants, namely, Newbouldia laevis stem bark (NLSB), Spathodea campanulata stem bark (SCSB), Momordica charantia leaves (MCL), and Ocimum viride leaves (OVL), were extracted for their active metabolites using methanol. Preliminary phytochemical screening was carried out on all plant extracts and powdered samples. The crude extracts were tested against S. mansoni cercariae in vitro using Balanites aegyptiaca as the positive control. The percentage of mortalities for each extract was recorded. Gas chromatography/mass spectrometry (GC/MS) analysis was conducted on all the plant extracts. Phytochemical analysis revealed the presence of saponins, glycosides, triterpenoids, sterols, alkaloids, flavonoids, and tannins in almost all the extracts. GC/MS analysis showed the presence of medicinally important active volatile compounds in each extract such as thymol, n-hexadecanoic acid, phytol, and maltol. All four plants showed relatively different levels of activity against S. mansoni cercariae at different times and concentrations. The LC50 values of the plant extracts were determined at the end of the assay. At 240 min, NLSB, SCSB, MCL, and OVL extracts had LC50 values of 487.564, 429.898, 197.696, and 0.129 μg/mL, respectively. Hence, this study revealed the potency of Ocimum viride leaves, Momordica charantia leaves, Spathodea campanulata stem bark, and Newbouldia laevis stem bark against S. mansoni. These plants could therefore be exploited as possible candidates for curbing schistosomiasis.
Collapse
Affiliation(s)
- Bright Osei-Mensah
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Yaw Duah Boakye
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - William Kofi Anyan
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Theresa Appiah Agana
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | | | - Ivy Bentil
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Elvis Suatey Lomotey
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Francis Adu
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Christian Agyare
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| |
Collapse
|
7
|
Hu Y, Yang L, Lai Y. Recent findings regarding the synergistic effects of emodin and its analogs with other bioactive compounds: Insights into new mechanisms. Biomed Pharmacother 2023; 162:114585. [PMID: 36989724 DOI: 10.1016/j.biopha.2023.114585] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
CONTEXT Emodin is a natural bioactive ingredient mainly extracted from traditional Chinese herbs. Increasing lines of evidence suggest that emodin and its analogs exert notable synergistic pharmacological effects with other bioactive compounds. OBJECTIVE This review provides an overview of the pharmacological activity of emodin and its analogs in combination with other physiologically active substances, describes the related molecular mechanisms, and discusses future prospects in this field. METHODS Information from multiple scientific databases, such as PubMed, the China Knowledge Resource Integrated Database from the China National Knowledge Infrastructure (CNKI), the Web of Science, Google Scholar, and Baidu Scholar, was collected between January 2006 and August 2022. The subject terms used in the literature search were emodin, pharmaceutical activities, analogs, aloe emodin, rhein, and synergistic effects. RESULTS The comprehensive literature analysis suggested that combinations of emodin or its analogs with other bioactive compounds exert notable synergistic anticancer, anti-inflammatory, and antimicrobial effects and that such combinations improve glucose and lipid metabolism and central nervous system diseases. DISCUSSION AND CONCLUSIONS Further assessments of the dose-effect relationship and the differences in the efficacy of emodin or its analogs with other bioactive compounds among various modes of administration are needed, and a drug safety evaluation of these combinations needs to be carefully performed. Future studies should also focus on determining the optimal drug combinations for specific diseases.
Collapse
|
8
|
Biendl S, Häberli C, Chen G, Wang W, Zhong L, Saunders J, Pham T, Wang X, Wu J, Charman SA, Vennerstrom JL, Keiser J. In Vitro and In Vivo Antischistosomal Activity Profiling and Pharmacokinetics of Ozonide Carboxylic Acids. ACS Infect Dis 2023; 9:643-652. [PMID: 36794836 PMCID: PMC10858445 DOI: 10.1021/acsinfecdis.2c00581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Praziquantel, the only drug in clinical use for the treatment and control of schistosomiasis, is inactive against developing infections. Ozonides are synthetic peroxide derivatives inspired by the naturally occurring artemisinin and show particularly promising activity against juvenile schistosomes. We conducted an in-depth characterization of the in vitro and in vivo antischistosomal activity and pharmacokinetics of lead ozonide carboxylic acid OZ418 and four of its active analogs. In vitro, the ozonides featured rapid and consistent activity against schistosomula and adult schistosomes at double-digit micromolar EC50 values. Potency did not vary considerably between Schistosoma spp. The zwitterionic OZ740 and OZ772 were more active in vivo compared to their non-amphoteric carboxylic acids OZ418 and OZ748, despite their much lower systemic plasma exposure (AUC). The most active compound in vivo was ethyl ester OZ780, which was rapidly transformed to its parent zwitterion OZ740 and achieved ED50 values of 35 ± 2.4 and 29 ± 2.4 mg/kg against adult and juvenile Schistosoma mansoni, respectively. Ozonide carboxylic acids represent promising candidates for further optimization and development due to their good efficacy against both life stages together with their broad activity range against all relevant parasite species.
Collapse
Affiliation(s)
- Stefan Biendl
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Kreuzstrasse 2, CH-4123 Allschwil, Switzerland
- University of Basel, CH-4003 Basel, Switzerland
| | - Cécile Häberli
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Kreuzstrasse 2, CH-4123 Allschwil, Switzerland
- University of Basel, CH-4003 Basel, Switzerland
| | - Gong Chen
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Wen Wang
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Longjin Zhong
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Jessica Saunders
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Thao Pham
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Xiaofang Wang
- College of Pharmacy, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, Nebraska 986125, United States of America
| | - Jianbo Wu
- College of Pharmacy, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, Nebraska 986125, United States of America
| | - Susan A Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Jonathan L Vennerstrom
- College of Pharmacy, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, Nebraska 986125, United States of America
| | - Jennifer Keiser
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Kreuzstrasse 2, CH-4123 Allschwil, Switzerland
- University of Basel, CH-4003 Basel, Switzerland
| |
Collapse
|
9
|
Khalil RG, Ibrahim AM, Bakery HH. Juglone: “A novel immunomodulatory, antifibrotic, and schistosomicidal agent to ameliorate liver damage in murine schistosomiasis mansoni”. Int Immunopharmacol 2022; 113:109415. [DOI: 10.1016/j.intimp.2022.109415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/27/2022] [Accepted: 10/30/2022] [Indexed: 11/11/2022]
|
10
|
Bakery HH, Allam GA, Abuelsaad ASA, Abdel‐Latif M, Elkenawy AE, Khalil RG. Anti‐inflammatory, antioxidant, anti‐fibrotic, and schistosomicidal properties of plumbagin in murine schistosomiasis. Parasite Immunol 2022; 44:e12945. [DOI: 10.1111/pim.12945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 08/01/2022] [Accepted: 08/08/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Heba H. Bakery
- Immunology Divisions, Zoology Department Faculty of Science, Beni‐Suef University Beni‐Suef Egypt
| | - Gamal A. Allam
- Immunology Divisions, Zoology Department Faculty of Science, Beni‐Suef University Beni‐Suef Egypt
| | | | - Mahmoud Abdel‐Latif
- Immunology Divisions, Zoology Department Faculty of Science, Beni‐Suef University Beni‐Suef Egypt
| | - Ayman E. Elkenawy
- Department of Pathology College of Medicine, Taif University, P.O. Box 11099 Taif Saudi Arabia
- Department of Molecular Biology, GEBRI University of Sadat City Sadat City Egypt
| | - Rehab G. Khalil
- Immunology Divisions, Zoology Department Faculty of Science, Beni‐Suef University Beni‐Suef Egypt
| |
Collapse
|
11
|
Guzman MA, Rugel A, Alwan SN, Tarpley R, Taylor AB, Chevalier FD, Wendt GR, Collins JJ, Anderson TJC, McHardy SF, LoVerde PT. Schistosome Sulfotransferases: Mode of Action, Expression and Localization. Pharmaceutics 2022; 14:1416. [PMID: 35890311 PMCID: PMC9323829 DOI: 10.3390/pharmaceutics14071416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 11/24/2022] Open
Abstract
Oxamniquine (OXA) is a prodrug activated by a sulfotransferase (SULT) that was only active against Schistosoma mansoni. We have reengineered OXA to be effective against S. haematobium and S. japonicum. Three derivatives stand out, CIDD-0066790, CIDD-0072229, and CIDD-0149830 as they kill all three major human schistosome species. However, questions remain. Is the OXA mode of action conserved in derivatives? RNA-interference experiments demonstrate that knockdown of the SmSULT, ShSULT, and SjSULT results in resistance to CIDD-0066790. Confirming that the OXA-derivative mode of action is conserved. Next is the level of expression of the schistosome SULTs in each species, as well as changes in SULT expression throughout development in S. mansoni. Using multiple tools, our data show that SmSULT has higher expression compared to ShSULT and SjSULT. Third, is the localization of SULT in the adult, multicellular eucaryotic schistosome species. We utilized fluorescence in situ hybridization and uptake of radiolabeled OXA to determine that multiple cell types throughout the adult schistosome worm express SULT. Thus, we hypothesize the ability of many cells to express the sulfotransferase accounts for the ability of the OXA derivatives to kill adult worms. Our studies demonstrate that the OXA derivatives are able to kill all three human schistosome species and thus will be a useful complement to PZQ.
Collapse
Affiliation(s)
- Meghan A. Guzman
- Department of Microbiology and Immunology, University of Texas Health, San Antonio, TX 78229, USA; (M.A.G.); (A.R.)
- Department of Biochemistry and Structural Biology, University of Texas Health, San Antonio, TX 78229, USA; (S.N.A.); (A.B.T.)
| | - Anastasia Rugel
- Department of Microbiology and Immunology, University of Texas Health, San Antonio, TX 78229, USA; (M.A.G.); (A.R.)
- Department of Biochemistry and Structural Biology, University of Texas Health, San Antonio, TX 78229, USA; (S.N.A.); (A.B.T.)
| | - Sevan N. Alwan
- Department of Biochemistry and Structural Biology, University of Texas Health, San Antonio, TX 78229, USA; (S.N.A.); (A.B.T.)
| | - Reid Tarpley
- Center for Innovative Drug Discovery, University of Texas at San Antonio, San Antonio, TX 78249, USA; (R.T.); (S.F.M.)
| | - Alexander B. Taylor
- Department of Biochemistry and Structural Biology, University of Texas Health, San Antonio, TX 78229, USA; (S.N.A.); (A.B.T.)
| | - Frédéric D. Chevalier
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA;
| | - George R. Wendt
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA; (G.R.W.); (J.J.C.III)
| | - James J. Collins
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA; (G.R.W.); (J.J.C.III)
| | - Timothy J. C. Anderson
- Disease Intervention & Prevention, Texas Biomedical Research Institute, San Antonio, TX 78227, USA;
| | - Stanton F. McHardy
- Center for Innovative Drug Discovery, University of Texas at San Antonio, San Antonio, TX 78249, USA; (R.T.); (S.F.M.)
| | - Philip T. LoVerde
- Department of Biochemistry and Structural Biology, University of Texas Health, San Antonio, TX 78229, USA; (S.N.A.); (A.B.T.)
- Department of Pathology and Laboratory Medicine, University of Texas Health, San Antonio, TX 78229, USA
| |
Collapse
|
12
|
Ossai EC, Eze AA, Ogugofor MO. Plant-derived compounds for the treatment of schistosomiasis: Improving efficacy via nano-drug delivery. Niger J Clin Pract 2022; 25:747-764. [DOI: 10.4103/njcp.njcp_1322_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
13
|
Optimal single sampling time-point for monitoring of praziquantel exposure in children. Sci Rep 2021; 11:17955. [PMID: 34504222 PMCID: PMC8429641 DOI: 10.1038/s41598-021-97409-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/24/2021] [Indexed: 12/16/2022] Open
Abstract
Praziquantel pharmacokinetics studies in schistosomiasis infected children are scarce partly due to the challenges/complexity of intensive blood sampling in the target population. This study was aimed to investigate the optimal single sampling time-point for monitoring praziquantel exposure. This was intensive pharmacokinetic study conducted among 32 Schistosoma mansoni infected children treated with an oral standard single-dose 40 mg/kg praziquantel. Plasma samples were collected at 0, 1, 2, 4, 6 and 8 h post-praziquantel administration. Quantification of praziquantel and its enantiomers (R- and S-praziquantel) concentrations was done by Liquid chromatography-tandem mass spectrometer (LC-MS/MS). The correlation between area under the plasma concentration-time curve from 0 to 8 h (AUC8) and plasma concentrations at each specific sampling time-point was determined by Pearson's correlation coefficient (r2). The median age (range) of the study population was 12.5 years (10-17). The study participants were 17 males and 15 females. Both total praziquantel and its enantiomers (R- and S-praziquantel) displayed a wide inter-individual pharmacokinetic variability. Regression analysis indicated that, plasma concentrations collected at 4 h post-dose had a significantly highest correlation with the AUC8 for both total praziquantel (r2 = 0.81, p < 0.001) and S-praziquantel (r2 = 0.84, p < 0.001) than any other sampling time-point; while for R-praziquantel, plasma concentrations collected at 6 h sampling time-point had a significantly highest correlation with the AUC8 (r2 = 0.79, p < 0.001) than any other sampling time-point. Four hours sampling time-point post-praziquantel administration is ideal optimal single sampling time-point for therapeutic monitoring of total praziquantel exposure while 6 h sampling time-point is suitable for monitoring of a pharmacologically active R-praziquantel enantiomer.
Collapse
|
14
|
Mduluza-Jokonya TL, Vengesai A, Midzi H, Kasambala M, Jokonya L, Naicker T, Mduluza T. Algorithm for diagnosis of early Schistosoma haematobium using prodromal signs and symptoms in pre-school age children in an endemic district in Zimbabwe. PLoS Negl Trop Dis 2021; 15:e0009599. [PMID: 34339415 PMCID: PMC8360514 DOI: 10.1371/journal.pntd.0009599] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 08/12/2021] [Accepted: 06/27/2021] [Indexed: 12/28/2022] Open
Abstract
Introduction Prompt diagnosis of acute schistosomiasis benefits the individual and provides opportunities for early public health intervention. In endemic areas schistosomiasis is usually contracted during the first 5 years of life, thus it is critical to look at how the infection manifests in this age group. The aim of this study was to describe the prodromal signs and symptoms of early schistosomiasis infection, correlate these with early disease progression and risk score to develop an easy to use clinical algorithm to identify early Schistosoma haematobium infection cases in resource limited settings. Methodology Two hundred and four, preschool age children who were lifelong residence of a schistosomiasis endemic district and at high risk of acquiring schistosomiasis were followed up from July 2019 to December 2019, during high transmission season. The children received interval and standard full clinical evaluations and laboratory investigations for schistosomiasis by clinicians blinded from their schistosomiasis infection status. Diagnosis of S. haematobium was by urine filtration collected over three consecutive days. Signs and symptoms of schistosomiasis at first examination visit were compared to follow-up visits. Signs and symptoms common on the last schistosomiasis negative visit (before a subsequent positive) were assigned as early schistosomiasis infection (ESI), after possible alternative causes were ruled out. Logistic regression identified clinical predictors. A model based score was assigned to each predictor to create a risk for every child. An algorithm was created based on the predictor risk scores and validated on a separate cohort of 537 preschool age children. Results Twenty-one percent (42) of the participants were negative for S. haematobium infection at baseline but turned positive at follow-up. The ESI participants at the preceding S. haematobium negative visit had the following prodromal signs and symptoms in comparison to non-ESI participants; pruritic rash adjusted odds ratio (AOR) = 21.52 (95% CI 6.38–72.66), fever AOR = 82 (95% CI 10.98–612), abdominal pain AOR = 2.6 (95% CI 1.25–5.43), pallor AOR = 4 (95% CI 1.44–11.12) and a history of facial/body swelling within the previous month AOR = 7.31 (95% CI 3.49–15.33). Furthermore 16% of the ESI group had mild normocytic anaemia, whilst 2% had moderate normocytic anaemia. A risk score model was created using a rounded integer from the relative risks ratios. The diagnostic algorithm created had a sensitivity of 81% and a specificity of 96.9%, Positive predictive value = 87.2% and NPV was 95.2%. The area under the curve for the algorithm was 0.93 (0.90–0.97) in comparison with the urine dipstick AUC = 0.58 (0.48–0.69). There was a similar appearance in the validation cohort as in the derivative cohort. Conclusion This study demonstrates for the first time prodromal signs and symptoms associated with early S. haematobium infection in pre-school age children. These prodromal signs and symptoms pave way for early intervention and management, thus decreasing the harm of late diagnosis. Our algorithm has the potential to assist in risk-stratifying pre-school age children for early S. haematobium infection. Independent validation of the algorithm on another cohort is needed to assess the utility further. Schistosoma haematobium causes urogenital infection and in endemic areas schistosomiasis is usually contracted during the first 5 years of life, thus it is critical to look at how the infection manifests in this age group. Prompt diagnosis of acute schistosomiasis is required to benefit the individuals and provide opportunities for early treatment and public health intervention. The study examined symptoms that correlated with early disease progression and risk scored to develop an easy to use clinical algorithm to identify early S. haematobium infection cases. The children received standard full clinical evaluations by clinicians who were blinded from schistosomiasis diagnosis by parasitological examination. An algorithm was created based on the predictor risk scores and participants had the following prodromal signs and symptoms; pruritic rash, abdominal pain, pallor, abdominal pain, inguinal lymphadenopathy and a history of facial/body swelling within the previous month. A risk score model, diagnostic algorithm, was created that compared to urine dipstick and parasitology. This study demonstrates the clinical signs and symptoms associated with early S. haematobium infection in pre-school age children. These prodromal signs and symptoms pave way for early intervention and management, thus decreasing the harm of late diagnosis common in populations from endemic areas.
Collapse
Affiliation(s)
- Tariro L. Mduluza-Jokonya
- Optics & Imaging, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal; KwaZulu-Natal, Durban, South Africa
- * E-mail:
| | - Arthur Vengesai
- Optics & Imaging, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal; KwaZulu-Natal, Durban, South Africa
| | - Herald Midzi
- Optics & Imaging, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal; KwaZulu-Natal, Durban, South Africa
- Department of Biochemistry, University of Zimbabwe, Harare, Zimbabwe
| | - Maritha Kasambala
- Department of Biochemistry, University of Zimbabwe, Harare, Zimbabwe
| | - Luxwell Jokonya
- Optics & Imaging, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal; KwaZulu-Natal, Durban, South Africa
- Department of Surgery, College of Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Thajasvarie Naicker
- Optics & Imaging, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal; KwaZulu-Natal, Durban, South Africa
| | - Takafira Mduluza
- Optics & Imaging, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal; KwaZulu-Natal, Durban, South Africa
- Department of Biochemistry, University of Zimbabwe, Harare, Zimbabwe
| |
Collapse
|
15
|
Rodrigues JGM, Albuquerque PSV, Nascimento JR, Campos JAV, Godinho ASS, Araújo SJ, Brito JM, Jesus CM, Miranda GS, Rezende MC, Negrão-Corrêa DA, Rocha CQ, Silva LA, Guerra RNM, Nascimento FRF. The immunomodulatory activity of Chenopodium ambrosioides reduces the parasite burden and hepatic granulomatous inflammation in Schistosoma mansoni-infection. JOURNAL OF ETHNOPHARMACOLOGY 2021; 264:113287. [PMID: 32858197 DOI: 10.1016/j.jep.2020.113287] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Folk medicine reports have described the use of Chenopodium ambrosioides as an anti-inflammatory, analgesic, and anthelmintic herb. These effects, including its activity against intestinal worms, are already scientifically observed. However, the immunological mechanisms of this species in the treatment of Schistosoma mansoni infection are unknown. AIM OF THE STUDY To evaluate the immunological and anti-Schistosoma mansoni effects of a crude Chenopodium ambrosioides hydro-alcoholic extract (HCE). MATERIALS AND METHODS For the in vitro analysis, cercariae and adult worms were exposed to different concentrations (0 to 10,000 μg/mL) of the HCE. For the in vivo evaluation, Swiss mice were infected with 50 cercariae of S. mansoni and separated into groups according to treatment as follows: a negative control (without treatment), a positive control (treated with Praziquantel®), HCE1 Group (treated with HCE during the cutaneous phase), HCE2 Group (treated with HCE during the lung phase), HCE3 Group (treated with HCE during the young worm phase), and HCE4 Group (treated with HCE during the adult worm phase). The animals treated with HCE received daily doses of 50 mg/kg, by gavage, for seven days, corresponding to the different developmental stages of S. mansoni. For comparison, a clean control group (uninfected and untreated) was also included. All animals were euthanized 60 days post-infection to allow the following assessments to be performed: a complete blood cells count, counts of eggs in the feces and liver, the quantification of cytokines and IgE levels, histopathological evaluations of the livers, and the analysis of inflammatory mediators. RESULTS HCE treatment increased the mortality of cercariae and adult worms in vitro. The HCE treatment in vivo reduced the eggs in feces and liver. The number and area of liver granulomas, independent of the phase of treatment, were also reduced. The treatment with HCE reduced the percentage of circulating eosinophils, IgE, IFN-γ, TNF-α, and IL-4. In contrast, the treatment with the HCE, dependent on the phase, increased IL-10 levels and the number of peritoneal and bone marrow cells, mainly of T lymphocytes, B lymphocytes, and macrophages. This effect could be due to secondary compounds presents in this extract, such as kaempferol, quercetin and derivatives. CONCLUSIONS This study demonstrates that Chenopodium ambrosioides has antiparasitic and immunomodulatory activity against the different phases of schistosomiasis, reducing the granulomatous inflammatory profile caused by the infection and, consequently, improving the disease prognosis.
Collapse
Affiliation(s)
- João Gustavo Mendes Rodrigues
- Laboratory of Immunophysiology, Centre for Biological and Health Sciences, Federal University of Maranhão, CEP: 65.055-970, São Luís, MA, Brazil.
| | - Paula Sibelly Veras Albuquerque
- Laboratory of Immunophysiology, Centre for Biological and Health Sciences, Federal University of Maranhão, CEP: 65.055-970, São Luís, MA, Brazil.
| | - Johnny R Nascimento
- Laboratory of Immunophysiology, Centre for Biological and Health Sciences, Federal University of Maranhão, CEP: 65.055-970, São Luís, MA, Brazil.
| | - Jaianna Andressa Viana Campos
- Laboratory of Immunophysiology, Centre for Biological and Health Sciences, Federal University of Maranhão, CEP: 65.055-970, São Luís, MA, Brazil.
| | - Andressa S S Godinho
- Laboratory of Immunophysiology, Centre for Biological and Health Sciences, Federal University of Maranhão, CEP: 65.055-970, São Luís, MA, Brazil.
| | - Sulayne Janayna Araújo
- Laboratory of Immunophysiology, Centre for Biological and Health Sciences, Federal University of Maranhão, CEP: 65.055-970, São Luís, MA, Brazil.
| | - Jefferson Mesquita Brito
- Laboratory of Immunophysiology, Centre for Biological and Health Sciences, Federal University of Maranhão, CEP: 65.055-970, São Luís, MA, Brazil.
| | - Caroline M Jesus
- Laboratory of Immunophysiology, Centre for Biological and Health Sciences, Federal University of Maranhão, CEP: 65.055-970, São Luís, MA, Brazil.
| | - Guilherme Silva Miranda
- Laboratory of Immunohelmintology, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, CEP: 31.270-901, Belo Horizonte, MG, Brazil; Laboratory of Biology, Department of Education, Federal Institute of Education, CEP: 65.840-000, São Raimundo Das Mangabeiras, MA, Brazil.
| | - Michelle C Rezende
- Laboratory of Immunohelmintology, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, CEP: 31.270-901, Belo Horizonte, MG, Brazil.
| | - Deborah Aparecida Negrão-Corrêa
- Laboratory of Immunohelmintology, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, CEP: 31.270-901, Belo Horizonte, MG, Brazil.
| | - Cláudia Q Rocha
- Laboratory of Natural Products Chemistry, Department of Chemistry, Federal University of Maranhão, CEP: 65.055-970, São Luís, MA, Brazil.
| | - Lucilene Amorim Silva
- Laboratory of Immunophysiology, Centre for Biological and Health Sciences, Federal University of Maranhão, CEP: 65.055-970, São Luís, MA, Brazil.
| | - Rosane N M Guerra
- Laboratory of Immunophysiology, Centre for Biological and Health Sciences, Federal University of Maranhão, CEP: 65.055-970, São Luís, MA, Brazil.
| | - Flávia R F Nascimento
- Laboratory of Immunophysiology, Centre for Biological and Health Sciences, Federal University of Maranhão, CEP: 65.055-970, São Luís, MA, Brazil.
| |
Collapse
|
16
|
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: 1.0] [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
|
17
|
Mnkugwe RH, Minzi O, Kinung’hi S, Kamuhabwa A, Aklillu E. Efficacy and safety of praziquantel and dihydroartemisinin piperaquine combination for treatment and control of intestinal schistosomiasis: A randomized, non-inferiority clinical trial. PLoS Negl Trop Dis 2020; 14:e0008619. [PMID: 32966290 PMCID: PMC7510991 DOI: 10.1371/journal.pntd.0008619] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 07/20/2020] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Despite the reported success in reducing morbidity, praziquantel alone is insufficient for the control and elimination of schistosomiasis, partly due to its poor efficacy against the juvenile worms. Artemisinin derivatives are effective against juvenile worms but are less effective against adult worms. We compared the safety and efficacy of praziquantel and Dihydroartemisinin-piperaquine combination against the standard praziquantel alone for treatment of intestinal schistosomiasis. METHODS In this randomized, open-label, non-inferiority trial, 639 Schistosoma mansoni infected children were enrolled and randomized to receive either praziquantel alone or praziquantel plus Dihydroartemisinin-piperaquine combination. Two stool samples were collected on consecutive days at baseline, 3 and 8 weeks post-treatment and analyzed using thick smear Kato Katz method. Efficacy was assessed by cure and egg reduction rates at 3 and 8 weeks post-treatment. Adverse events were assessed within four hours of drugs intake. The primary outcome was cure rates at 8 weeks of post-treatment. Secondary outcomes were egg reduction rates at 8 weeks of post-treatment and treatment-associated adverse events. RESULTS At 3 weeks of post-treatment, cure rates were 88.3% (263/298, 95% CI = 84.1%- 91.4%) and 81.2% (277/341, 95% CI = 76.7%- 85.0%) for the combination therapy and praziquantel alone, respectively (p < 0.01, odds ratio (OR) = 1.74, 95% CI of OR = 1.11 to 2.69). At 8 weeks, there was a significant drop in the cure rates in praziquantel alone group to 63.9% (218/341, 95% CI = 58.7%- 68.8%) compared to 81.9% (244/298, 95% CI = 77.1%- 85.8%) in the combination therapy group (p < 0.0001, OR = 2.55, 95%CI of OR = 1.75 to 3.69). Egg reduction rates at 8 weeks post-treatment were significantly higher in the combination therapy group 93.6% (95% CI = 90.8%- 96.4%) compared to 87.9% (95% CI = 84.4%- 91.4%) in the praziquantel only group (p = 0.01). On both Univariate and Multivariate regression analysis, type of treatment received was a significant predictor of cure at week 8 post-treatment. Overall, 30.8% (95% CI = 27.2%- 34.4%) of the study participants experienced mild and transient treatment-associated adverse events, post-treatment abdominal pain (27.1%) being the most common adverse event observed. There was no significant difference in the overall occurrence of adverse events between the two treatment groups. CONCLUSION Praziquantel and Dihydroartemisinin piperaquine combination therapy is safe, and more efficacious compared to praziquantel alone for the treatment of intestinal schistosomiasis. Further studies are needed to explore if the combination therapy can be considered as an option for mass drug administration to control and eventually eliminate schistosomiasis.
Collapse
Affiliation(s)
- Rajabu Hussein Mnkugwe
- Department of Clinical Pharmacology, School of Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
- Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska University Hospital-Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Omary Minzi
- Department of Clinical Pharmacy and Pharmacology, School of Pharmacy, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Safari Kinung’hi
- National Institute for Medical Research (NIMR), Mwanza Research Centre, Mwanza, Tanzania
| | - Appolinary Kamuhabwa
- Department of Clinical Pharmacy and Pharmacology, School of Pharmacy, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Eleni Aklillu
- Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska University Hospital-Huddinge, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
18
|
Zdesenko G, Mutapi F. Drug metabolism and pharmacokinetics of praziquantel: A review of variable drug exposure during schistosomiasis treatment in human hosts and experimental models. PLoS Negl Trop Dis 2020; 14:e0008649. [PMID: 32976496 PMCID: PMC7518612 DOI: 10.1371/journal.pntd.0008649] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Schistosomiasis control is heavily reliant on the drug praziquantel (PZQ), which is used as preventive chemotherapy as part of national helminth control strategies. Given the heavy reliance on PZQ for mass drug administration, there has been considerable research on the potential of parasites developing resistance to the drug, resulting in decreased drug efficacy. However, there have been comparatively fewer studies of other factors that can potentially alter PZQ efficacy. Here, we investigate whether host PZQ metabolism contributes towards variable cure rates. We evaluate factors that can influence the metabolism of PZQ and the resultant effect on the efficacy of PZQ treatment to determine factors that potentially influence an individual's response to the drug. The literature search was directed at published studies from three online databases: Web of Science, PubMed, and EMBASE. The search terms for the review comprised of ([praziquantel OR PZQ] AND [schistosom* OR bilharzia] AND [pharmaco*]) and included studies evaluating PZQ metabolism. Publications were categorised into pharmacokinetics, drug-drug interactions, pharmacogenetics, and metabolite analysis. Forty publications describing human and experimental studies fitted the inclusion criteria and were subjected to data extraction and analysis. The analyses showed that variable exposure to PZQ was associated with alterations in the liver's capacity to metabolise PZQ and observed drug-drug interactions. Other factors influencing the efficacy of PZQ were brand, formulation, and co-administered food. Although some work has been performed on metabolite identification, there was minimal information on PZQ's metabolic pathway, and no pharmacogenetics studies were identified. The study indicated that in both human and experimental studies alterations in the liver's capacity to metabolise PZQ as well as drug-drug interactions affected systemic levels of PZQ that could result in variable cure rates. The study confirmed previous findings of higher antischistosomal activity of (R)-PZQ enantiomer when administered alone compared to the racemate at the same dose as well as improved efficacy when the drug is administered with food. The study also highlighted the need for more comprehensive studies of the PZQ metabolic pathway and PZQ pharmacogenetic studies in humans.
Collapse
Affiliation(s)
- Grace Zdesenko
- 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
| | - 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
|
19
|
Hasby Saad MA, El-Anwar N. Bevacizumab as a potential anti-angiogenic therapy in schistosomiasis: A double-edged, but adjustable weapon. Parasite Immunol 2020; 42:e12724. [PMID: 32338371 DOI: 10.1111/pim.12724] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 04/09/2020] [Accepted: 04/17/2020] [Indexed: 11/30/2022]
Abstract
AIM Investigating the anti-angiogenic effect of bevacizumab on chronic schistosomiasis mansoni in a trial to hinder the Schistosome-induced angiogenesis and porto-systemic shunting complications. METHODS The immunohistochemical expression of CD34, VEGF-R1, PCNA and α-SMA (angiogenesis markers) was analysed in the lung, liver and gastrointestinal junctions of chronic S mansoni infected mice after intraperitoneal injection of bevacizumab. The effect of prolonged administration of bevacizumab with praziquantel was also assessed through parasitic load, protective index, granuloma and fibrous tissue evaluation. RESULTS A regression in the vascular activity and microvascular density was observed in the infected mice after receiving bevacizumab. They had a significantly less VEGF-R1, PCNA, CD-34 and α-SMA expression in comparison to the infected untreated mice. The least tissue egg count was reported in mice received bevacizumab for 6 weeks (Mean = 27 120). However, they had persistent liver granulomas, and massively amalgamated fibrosis. Interestingly, the least faecal egg and tissue worms counts (Mean = 112, 13.4), and the highest protection index (39.26) were reported in mice received bevacizumab for 3 weeks, with marked granuloma, and fibrous tissue resolution. CONCLUSIONS Bevacizumab has a promising protective effect against the Schistosoma-induced angiogenesis. As an adjuvant to praziquantel, it is important to adjust the appropriate duration of administration that achieves the best schistosomicidal effect without impeding granuloma and fibrous tissue resolution.
Collapse
Affiliation(s)
- Marwa A Hasby Saad
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, Gharbia Governorate, Egypt
| | - Noha El-Anwar
- Department of Pathology, Tanta University, Faculty of Medicine, Tanta, Egypt
| |
Collapse
|
20
|
Lyu H, Petukhov PA, Banta PR, Jadhav A, Lea WA, Cheng Q, Arnér ESJ, Simeonov A, Thatcher GRJ, Angelucci F, Williams DL. Characterization of Lead Compounds Targeting the Selenoprotein Thioredoxin Glutathione Reductase for Treatment of Schistosomiasis. ACS Infect Dis 2020; 6:393-405. [PMID: 31939288 DOI: 10.1021/acsinfecdis.9b00354] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Schistosomiasis is a widespread human parasitic disease currently affecting over 200 million people. Chemotherapy for schistosomiasis relies exclusively on praziquantel. Although significant advances have been made in recent years to reduce the incidence and intensity of schistosome infections, these gains will be at risk should drug-resistant parasites evolve. Thioredoxin glutathione reductase (TGR) is a selenoprotein of the parasite essential for the survival of schistosomes in the mammalian host. Several high-throughput screening campaigns have identified inhibitors of Schistosoma mansoni TGR. Follow up analyses of select active compounds form the basis of the present study. We identified eight compounds effective against ex vivo worms. Compounds 1-5 are active against all major species and development stages. The ability of these compounds to target immature worms is especially critical because praziquantel is poorly active against this stage. Compounds 1-5, 7, and 8 displayed schistosomicidal activity even after only 1 h incubation with the worms. Compounds 1-4 meet or exceed standards set by the World Health Organization for leads for schistosomiasis therapy activity. The mechanism of TGR inhibition was studied further with wild-type and mutant TGR proteins. Compounds 4-6 were found to induce an nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity in TGR, leading to the production of superoxide and hydrogen peroxide. Collectively, this effort has identified several active compound series that may serve as the basis for the development of new schistosomicidal compounds.
Collapse
Affiliation(s)
- Haining Lyu
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois 60612, United States
| | - Pavel A. Petukhov
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Paul R. Banta
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois 60612, United States
| | - Ajit Jadhav
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Wendy A. Lea
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Qing Cheng
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Elias S. J. Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Anton Simeonov
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Gregory R. J. Thatcher
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Francesco Angelucci
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - David L. Williams
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois 60612, United States
| |
Collapse
|
21
|
Santos SS, de Araújo RV, Giarolla J, Seoud OE, Ferreira EI. Searching for drugs for Chagas disease, leishmaniasis and schistosomiasis: a review. Int J Antimicrob Agents 2020; 55:105906. [PMID: 31987883 DOI: 10.1016/j.ijantimicag.2020.105906] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 01/14/2020] [Accepted: 01/18/2020] [Indexed: 12/16/2022]
Abstract
Chagas disease, leishmaniasis and schistosomiasis are neglected diseases (NDs) and are a considerable global challenge. Despite the huge number of people infected, NDs do not create interest from pharmaceutical companies because the associated revenue is generally low. Most of the research on these diseases has been conducted in academic institutions. The chemotherapeutic armamentarium for NDs is scarce and inefficient and better drugs are needed. Researchers have found some promising potential drug candidates using medicinal chemistry and computational approaches. Most of these compounds are synthetic but some are from natural sources or are semi-synthetic. Drug repurposing or repositioning has also been greatly stimulated for NDs. This review considers some potential drug candidates and provides details of their design, discovery and activity.
Collapse
Affiliation(s)
- Soraya Silva Santos
- Laboratory of Design and Synthesis of Chemotherapeutics Potentially Active in Neglected Diseases (LAPEN), Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo-USP, Avenue Professor Lineu Prestes, 580-Building 13, São Paulo SP, 05508-900, Brazil
| | - Renan Vinicius de Araújo
- Laboratory of Design and Synthesis of Chemotherapeutics Potentially Active in Neglected Diseases (LAPEN), Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo-USP, Avenue Professor Lineu Prestes, 580-Building 13, São Paulo SP, 05508-900, Brazil
| | - Jeanine Giarolla
- Laboratory of Design and Synthesis of Chemotherapeutics Potentially Active in Neglected Diseases (LAPEN), Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo-USP, Avenue Professor Lineu Prestes, 580-Building 13, São Paulo SP, 05508-900, Brazil
| | - Omar El Seoud
- Laboratory of Design and Synthesis of Chemotherapeutics Potentially Active in Neglected Diseases (LAPEN), Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo-USP, Avenue Professor Lineu Prestes, 580-Building 13, São Paulo SP, 05508-900, Brazil
| | - Elizabeth Igne Ferreira
- Laboratory of Design and Synthesis of Chemotherapeutics Potentially Active in Neglected Diseases (LAPEN), Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo-USP, Avenue Professor Lineu Prestes, 580-Building 13, São Paulo SP, 05508-900, Brazil.
| |
Collapse
|
22
|
In vitro activity of usnic acid potassium salt against different developmental stages of Schistosoma mansoni: An ultrastructural study. Acta Trop 2020; 201:105159. [PMID: 31491401 DOI: 10.1016/j.actatropica.2019.105159] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/08/2019] [Accepted: 09/02/2019] [Indexed: 12/18/2022]
Abstract
Currently, the control of schistosomiasis is based on a single drug, praziquantel, which is effective against all species of Schistosoma but only in the adult stage, presenting a schistosomicidal deficit at the other developmental stages of the parasites. Recently our research group has demonstrated that the potassium salt of usnic acid (PS-UA) presented schistosomicidal property against couples of adult worms of S. mansoni. Thus, the present study seeks to report for the first time the in vitro activity of PS-UA against different developmental stages of S. mansoni (schistosomules and young worms). As schistosomicide parameters, we evaluated motility, mortality, cell viability of the worms and tegument changes by scanning electron microscopy (SEM). After 3 h exposure, PS-UA was lethal to schistosomules at concentrations of 100 and 50 μM, whereas for concentrations 25 and 12.5 μM, 38 and 18% of mortality and 62 and 24% changes in motility, respectively, were reached. Yet for schistosomules, concentration of 25 μM caused 90 and 100% of death after 6 and 12 h, respectively. In the concentration of 12.5 μM at intervals of 12 and 24 h mortality was 68 and 100%, respectively. For young worms, after 3 h of exposure at concentrations of 200 and 100 μM caused 57 and 27% mortality, respectively. After 12 and 24 h, these concentrations caused mortality of 90 and 100% and 47 and 60% respectively. After 24 h, concentrations of 50 and 25 μM caused 80 and 30% change in motility, respectively. However, at the 12.5 μM concentration no change was observed. In addition, PS-UA reduced the cellular viability of young worms by 50.98% and 85.87% at concentrations of 100 and 200 μM, respectively. In both stages of worms and at different exposure intervals, PS-UA caused alterations such as: dorsoventral contraction, peeling, swelling, blisters, erosion, exposure of subtegumental tissue and disintegration of tegument. According to the results, changes in motility and mortality caused by PS-UA against schistosomules and young worms were concentration and time-dependents, also PS-UA even at low concentration, was able to cause profound ultrastructural changes in the integument of the worms. PS-UA is a promising candidate as prophylactic agent in the control of schistosomiasis mansoni.
Collapse
|
23
|
Khayeka-Wandabwa C, Zhou G, Magak NG, Choge JK, Kemei WK, Makwali JA, Karani LW, Kisavi MP, Ndulu JV, Anjili CO. Combined chemotherapy manifest less severe immunopathology effects in helminth-protozoa comorbidity. Exp Parasitol 2019; 204:107728. [PMID: 31348915 DOI: 10.1016/j.exppara.2019.107728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 07/15/2019] [Accepted: 07/22/2019] [Indexed: 11/24/2022]
Abstract
BACKGROUND Co-infection with Leishmania major and Schistosoma mansoni may have significant consequences for disease progression, severity and subsequent transmission dynamics. Pentavalent antimonials and Praziquantel (PZQ) are used as first line of treatment for Leishmania and Schistosoma infections respectively. However, there is limited insight on how combined therapy with the standard drugs impacts the host in comorbidity. The study aimed to determine the efficacy of combined chemotherapy using Pentostam (P) and PZQ in murine model co-infected with L. major and S. mansoni. METHODS A 3 × 4 factorial design with three parasite infection groups (Lm, Sm, Lm + Sm to represent L. major, S. mansoni and L. major + S. mansoni respectively) and four treatment regimens [P, PZQ, P + PZQ, and PBS designating Pentostam (GlaxoSmithKline UK), Praziquantel (Biltricide®, Bayer Ag. Leverkusen, Germany), Pentostam + Praziquantel and Phosphate buffered saline] as factors was applied. RESULTS Significant changes were observed in the serum Interferon gamma (IFN-γ), and Macrophage inflammatory protein-one alpha (MIP-1α) levels among various treatment groups between week 8 and week 10 (p < 0.05). There was increased IFN-γ in the L. major infected mice subjected to PZQ and PBS, and in L. major + S. mansoni infected BALB/c mice treated with P + PZQ. Subsequently, MIP-1α levels increased significantly in both the L. major infected mice under PZQ and PBS and in L. major + S. mansoni infected BALB/c mice undergoing concurrent chemotherapy with P + PZQ between 8 and 10 weeks (p < 0.05). In the comorbidity, simultaneous chemotherapy resulted in less severe histopathological effects in the liver. CONCLUSION It was evident, combined first line of treatment is a more effective strategy in managing co-infection of L. major and S. mansoni. The findings denote simultaneous chemotherapy compliments immunomodulation in the helminth-protozoa comorbidity hence, less severe pathological effects following the parasites infection. Recent cases of increased incidences of polyparasitism in vertebrates call for better ways to manage co-infections. The findings presented necessitate intrinsic biological interest on examining optimal combined chemotherapeutic agents strategies in helminth-protozoa concomitance and the related infections abatement trends vis-a-vis host-parasite relationships.
Collapse
Affiliation(s)
- Christopher Khayeka-Wandabwa
- School of Pharmaceutical Science and Technology (SPST), Health Science Platform, Tianjin University, Tianjin, 300072, China; Centre for Biotechnology Research and Development (CBRD), Kenya Medical Research Institute (KEMRI), P.O Box 54840, Nairobi, 00200, Kenya.
| | - Guan Zhou
- School of Pharmaceutical Science and Technology (SPST), Health Science Platform, Tianjin University, Tianjin, 300072, China.
| | | | - Joseph K Choge
- University of Kabianga, P.O. Box 2030, Kericho, 20200, Kenya.
| | - William Kipchirchir Kemei
- Institute of Tropical Medicine and Infectious Diseases (ITROMID), Jomo Kenyatta University of Agriculture and Technology (JKUAT), P.O. Box 62000-00200, Nairobi, Kenya.
| | - Judith Alice Makwali
- Department of Biological Science, University of Eldoret, P.O Box 1125, Eldoret, 30100, Kenya.
| | | | - Mutila Phoebe Kisavi
- School of Health Science, Machakos University, Kenya; Public Health Intervention Research Group, The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, NSW 2052 Australia.
| | - James V Ndulu
- African Population and Health Research Center (APHRC), P .O. Box 10787-00100, Nairobi, Kenya.
| | - Christopher O Anjili
- Centre for Biotechnology Research and Development (CBRD), Kenya Medical Research Institute (KEMRI), P.O Box 54840, Nairobi, 00200, Kenya.
| |
Collapse
|
24
|
Botros SS, William S, Sabra ANA, El-Lakkany NM, Seif El-Din SH, García-Rubia A, Sebastián-Pérez V, Blaazer AR, de Heuvel E, Sijm M, Zheng Y, Salado IG, Munday JC, Maes L, de Esch IJP, Sterk GJ, Augustyns K, Leurs R, Gil C, De Koning HP. Screening of a PDE-focused library identifies imidazoles with in vitro and in vivo antischistosomal activity. Int J Parasitol Drugs Drug Resist 2019; 9:35-43. [PMID: 30669086 PMCID: PMC6350229 DOI: 10.1016/j.ijpddr.2019.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/10/2019] [Accepted: 01/13/2019] [Indexed: 01/10/2023]
Abstract
We report the evaluation of 265 compounds from a PDE-focused library for their antischistosomal activity, assessed in vitro using Schistosoma mansoni. Of the tested compounds, 171 (64%) displayed selective in vitro activity, with 16 causing worm hypermotility/spastic contractions and 41 inducing various degrees of worm killing at 100 μM, with the surviving worms displaying sluggish movement, worm unpairing and complete absence of eggs. The compounds that did not affect worm viability (n = 72) induced a complete cessation of ovipositing. 82% of the compounds had an impact on male worms whereas female worms were barely affected. In vivo evaluation in S. mansoni-infected mice with the in vitro 'hit' NPD-0274 at 20 mg/kg/day orally for 5 days resulted in worm burden reductions of 29% and intestinal tissue egg load reduction of 35% at 10 days post-treatment. Combination of praziquantel (PZQ) at 10 mg/kg/day for 5 days with NPD-0274 or NPD-0298 resulted in significantly higher worm killing than PZQ alone, as well as a reduction in intestinal tissue egg load, disappearance of immature eggs and an increase in the number of dead eggs.
Collapse
Affiliation(s)
- Sanaa S Botros
- Pharmacology Department, Theodor Bilharz Research Institute, Warrak El-Hadar, Imbaba, P.O. Box 30, Giza, 12411, Egypt
| | - Samia William
- Parasitology Department, Theodor Bilharz Research Institute, Warrak El-Hadar, Imbaba, P.O. Box 30, Giza, 12411, Egypt
| | - Abdel-Nasser A Sabra
- Pharmacology Department, Theodor Bilharz Research Institute, Warrak El-Hadar, Imbaba, P.O. Box 30, Giza, 12411, Egypt
| | - Naglaa M El-Lakkany
- Pharmacology Department, Theodor Bilharz Research Institute, Warrak El-Hadar, Imbaba, P.O. Box 30, Giza, 12411, Egypt
| | - Sayed H Seif El-Din
- Pharmacology Department, Theodor Bilharz Research Institute, Warrak El-Hadar, Imbaba, P.O. Box 30, Giza, 12411, Egypt
| | | | | | - Antoni R Blaazer
- Medicinal Chemistry Vrije Universiteit Amsterdam (VUA), the Netherlands
| | - Erik de Heuvel
- Medicinal Chemistry Vrije Universiteit Amsterdam (VUA), the Netherlands
| | - Maarten Sijm
- Medicinal Chemistry Vrije Universiteit Amsterdam (VUA), the Netherlands
| | - Yang Zheng
- Medicinal Chemistry Vrije Universiteit Amsterdam (VUA), the Netherlands
| | | | - Jane C Munday
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8TA, UK
| | | | - Iwan J P de Esch
- Medicinal Chemistry Vrije Universiteit Amsterdam (VUA), the Netherlands
| | - Geert J Sterk
- Medicinal Chemistry Vrije Universiteit Amsterdam (VUA), the Netherlands
| | | | - Rob Leurs
- Medicinal Chemistry Vrije Universiteit Amsterdam (VUA), the Netherlands
| | - Carmen Gil
- Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
| | - Harry P De Koning
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8TA, UK.
| |
Collapse
|
25
|
Asian Schistosomiasis: Current Status and Prospects for Control Leading to Elimination. Trop Med Infect Dis 2019; 4:tropicalmed4010040. [PMID: 30813615 PMCID: PMC6473711 DOI: 10.3390/tropicalmed4010040] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/12/2019] [Accepted: 02/12/2019] [Indexed: 12/22/2022] Open
Abstract
Schistosomiasis is an infectious disease caused by helminth parasites of the genus Schistosoma. Worldwide, an estimated 250 million people are infected with these parasites with the majority of cases occurring in sub-Saharan Africa. Within Asia, three species of Schistosoma cause disease. Schistosoma japonicum is the most prevalent, followed by S. mekongi and S. malayensis. All three species are zoonotic, which causes concern for their control, as successful elimination not only requires management of the human definitive host, but also the animal reservoir hosts. With regard to Asian schistosomiasis, most of the published research has focused on S. japonicum with comparatively little attention paid to S. mekongi and even less focus on S. malayensis. In this review, we examine the three Asian schistosomes and their current status in their endemic countries: Cambodia, Lao People's Democratic Republic, Myanmar, and Thailand (S. mekongi); Malaysia (S. malayensis); and Indonesia, People's Republic of China, and the Philippines (S. japonicum). Prospects for control that could potentially lead to elimination are highlighted as these can inform researchers and disease control managers in other schistosomiasis-endemic areas, particularly in Africa and the Americas.
Collapse
|
26
|
Atalabi TE, Adubi TO. The epidemiology and chemotherapeutic approaches to the control of urinary schistosomiasis in school-age children (SAC): a systematic review. BMC Infect Dis 2019; 19:73. [PMID: 30658583 PMCID: PMC6339440 DOI: 10.1186/s12879-018-3647-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 12/21/2018] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Human schistosomiases are acute and chronic infectious diseases of poverty. Currently, epidemiological data of urinary schistosomiasis (US) in school-age children (SAC) and adults are often reported together making it difficult to ascertain the true status of the disease. Based on this premise, we set out to carry out this review. METHOD To achieve this aim, we carried out a computer-aided search of PubMed, Web of Science, Science Direct, African Journals OnLine (AJOL) and the database of World Health Organization. However, the information obtained from these sources was supplemented with additional literatures from Mendeley, Research Gate, and Google. RESULTS The search yielded 183 literatures of which 93 full text research, review and online articles were deemed fit for inclusion. Our key findings showed that: (1) of all World Health Organization (WHO) Regions, Africa is the most endemic zone for US, with Kenya and Senegal recording the highest prevalence and mean intensity respectively; (2) SAC within the range of 5-16 years contribute most significantly to the transmission cycle of US globally; (3) gender is a factor to watch out for, with male often recording the highest prevalence and intensity of infection; (4) contact with open, potentially infested water sources contribute significantly to transmission; (5) parental factors (occupation and education status) predispose SAC to US; (6) economic vis a vis ecological factors play a key role in infection transmission; and (7) in the last decade, a treatment coverage of 45% was never achieved globally for SAC or non-SAC treatment category for urinary schistosomiasis. CONCLUSION In view of the WHO strategic plan to eliminate schistosomiasis by 2020 and the findings from this review, it is obvious that this goal, in the face of realities, might not be achieved. It is imperative that annual control programmes be scaled up marginally, particularly in the African region of WHO. While US-based researches should be sponsored at the grass-root level to unveil hidden endemic foci, adequate facilities for Water, Sanitation, and Hygiene (WASH) should be put in place in all schools globally.
Collapse
Affiliation(s)
- Tolulope Ebenezer Atalabi
- Department of Biological Sciences, Federal University, Dutsin-Ma, Km 65, P.M.B. 5001, Dutsin-Ma, Katsina State Nigeria
| | - Taiwo Oluwakemi Adubi
- Department of Biological Sciences and Biotechnology, College of Pure and Applied Sciences, Caleb University, Imota, Lagos State Nigeria
| |
Collapse
|
27
|
Aruleba RT, Adekiya TA, Oyinloye BE, Masamba P, Mbatha LS, Pretorius A, Kappo AP. PZQ Therapy: How Close are we in the Development of Effective Alternative Anti-schistosomal Drugs? Infect Disord Drug Targets 2019; 19:337-349. [PMID: 30599112 PMCID: PMC7046992 DOI: 10.2174/1871526519666181231153139] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/24/2018] [Accepted: 12/25/2018] [Indexed: 11/22/2022]
Abstract
Today schistosomiasis, caused mainly by the three major schistosome species (S. mansoni, S. haematobium and S. japonicum), has for many decades and still continues to be on a rapid and swift rise globally, claiming thousands of lives every year and leaving 800 million people at the risk of infection. Due to the high prevalence of this disease and the steady increase in the infection rates, praziquantel (PZQ) remains the only effective drug against this acute disease although it has no effect on the juvenile schistosome parasite. However, no significant approaches have been made in recent years in the discovery of new or alternative drugs and unfortunately, resistance to this drug has been reported in some parts of the world. Therefore, it is imperative to develop a new drug for this debilitating disease. In this review, a brief history of past, present, and new promising anti-schistosomal drugs is presented.
Collapse
Affiliation(s)
- Raphael Taiwo Aruleba
- Biotechnology and Structural Biology (BSB) Group, Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
| | - Tayo Alex Adekiya
- Biotechnology and Structural Biology (BSB) Group, Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
| | - Babatunji Emmanuel Oyinloye
- Biotechnology and Structural Biology (BSB) Group, Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
- Department of Biochemistry, Afe Babalola University, PMB 5454, Ado-Ekiti 360001, Nigeria
| | - Priscilla Masamba
- Biotechnology and Structural Biology (BSB) Group, Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
| | - Londiwe Simphiwe Mbatha
- Biotechnology and Structural Biology (BSB) Group, Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
| | - Ashley Pretorius
- Bioinformatics Research Group (BRG), DST/Mintek Nanotechnology Innovation Centre, Biolabels Node, Department of Biotechnology, University of the Western Cape, Bellville 7535, South Africa
| | - Abidemi Paul Kappo
- Biotechnology and Structural Biology (BSB) Group, Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
| |
Collapse
|
28
|
Eweas AF, Allam G. Targeting thioredoxin glutathione reductase as a potential antischistosomal drug target. Mol Biochem Parasitol 2018; 225:94-102. [DOI: 10.1016/j.molbiopara.2018.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 09/09/2018] [Accepted: 09/30/2018] [Indexed: 11/30/2022]
|
29
|
White Bear J, Long T, Skinner D, McKerrow JH. Predictions of novel Schistosoma mansoni - human protein interactions consistent with experimental data. Sci Rep 2018; 8:13092. [PMID: 30166569 PMCID: PMC6117258 DOI: 10.1038/s41598-018-31272-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 08/14/2018] [Indexed: 12/26/2022] Open
Abstract
Infection by the human blood fluke, Schistosoma mansoni involves a variety of cross-species protein- protein interactions. The pathogen expresses a diverse arsenal of proteins that facilitate the breach of physical and biochemical barriers present in skin evasion of the immune system, and digestion of human plasma proteins including albumin and hemoglobin, allowing schistosomes to reside in the host for years. However, only a small number of specific interactions between S. mansoni and human proteins have been identified. We present and apply a protocol that generates testable predictions of S. mansoni-human protein interactions. In this study, we have preliminary predictions of novel interactions between schistosome and human proteins relevant to infection and the ability of the parasite to evade the immune system. We applied a computational whole-genome comparative approach to predict potential S. mansoni-human protein interactions based on similarity to known protein complexes. We first predict S. mansoni -human protein interactions based on similarity to known protein complexes. Putative interactions were then scored and assessed using several contextual filters, including the use of annotation automatically derived from literature using a simple natural language processing methodology. Next, in vitro experiments were carried out between schistosome and host proteins to validate several prospective predictions. Our method predicted 7 out of the 10 previously known cross-species interactions involved in pathogenesis between S. mansoni and its human host. Interestingly, two novel putative interactions involving Schistosoma proteins, the cercarial elastase SmCE, and the adult tegument surface protein Sm29, were also predicted and experimentally characterized. Preliminary data suggest that elafin, a host endogenous serine protease inhibitor, may be a novel substrate for SmCE. Additionally, CD59, an inhibitor of the membrane attack complex, could interact with Sm29. Furthermore, the application framework provides an integrated methodology for investigation of host-pathogen interactions and an extensive source of orthogonal data for experimental analysis. We have made the predictions available for community perusal.
Collapse
Affiliation(s)
- J White Bear
- Department of Bioengineering and Therapeutic Sciences, Department of Pharmaceutical Chemistry, and California Institute for Quantitative Biosciences, University of California, San Francisco, CA, 94158, USA.
- Graduate Group in Bioinformatics, University of California, San Francisco, CA, 94158, USA.
- MIT Lincoln Laboratory 244 Wood St, Lexington, MA, USA.
| | - Thavy Long
- Department of Pathology and Sandler Center for Basic Research in Parasitic Diseases, University of California at San Francisco, San Francisco, California, 94158, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego 9500 Gilman Dr, La Jolla, CA, 92093, USA
- INRA - InTheRes - UMR 1436, Equipe Transporteurs Membranaires et Résistance, 180, Chemin de Tournefeuille, Toulouse, France
| | - Danielle Skinner
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego 9500 Gilman Dr, La Jolla, CA, 92093, USA
| | - James H McKerrow
- Department of Pathology and Sandler Center for Basic Research in Parasitic Diseases, University of California at San Francisco, San Francisco, California, 94158, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego 9500 Gilman Dr, La Jolla, CA, 92093, USA
| |
Collapse
|
30
|
de Oliveira SA, de Oliveira Barbosa M, Filho CALM, Oliveira AR, de Sousa FA, de Farias Santiago E, de Oliveira Filho GB, de Moraes Gomes PAT, da Conceição JM, Brayner FA, Alves LC, Leite ACL. Phthalimido-thiazole as privileged scaffold: activity against immature and adult worms of Schistosoma mansoni. Parasitol Res 2018; 117:2105-2115. [DOI: 10.1007/s00436-018-5897-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/25/2018] [Indexed: 12/26/2022]
|
31
|
Gouveia MJ, Brindley PJ, Gärtner F, Costa JMCD, Vale N. Drug Repurposing for Schistosomiasis: Combinations of Drugs or Biomolecules. Pharmaceuticals (Basel) 2018; 11:E15. [PMID: 29401734 PMCID: PMC5874711 DOI: 10.3390/ph11010015] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 01/19/2018] [Accepted: 01/31/2018] [Indexed: 12/17/2022] Open
Abstract
Schistosomiasis is a major neglected tropical disease. Control of schistosomiasis currently relies on a single drug, praziquantel, and despite its efficacy against the all schistosome species that parasitize humans, it displays some problematic drawbacks and alone is ineffective in counteracting adverse pathologies associated with infection. Moreover, due to the development of the potential emergence of PZQ-resistant strains, the search for additional or alternative antischistosomal drugs have become a public health priority. The current drug discovery for schistosomiasis has been slow and uninspiring. By contrast, repurposing of existing approved drugs may offer a safe, rapid and cost-effective alternative. Combined treatment with PZQ and other drugs with different mode of action, i.e., antimalarials, shows promise results. In addition, a combination of anthelminthic drugs with antioxidant might be advantageous for modulating oxidative processes associated with schistosomiasis. Herein, we review studies dealing with combination therapies that involve PZQ and other anthelminthic drugs and/or antioxidant agents in treatment of schistosomiasis. Whereas PZQ combined with antioxidant agents might or might not interfere with anthelminthic efficacy, combinations may nonetheless ameliorate tissue damage and infection-associated complications. In fact, alone or combine with other drugs, antioxidants might be a valuable adjuvant to reduce morbidity and mortality of schistosomiasis. Therefore, attempting new combinations of anthelmintic drugs with other biomolecules such as antioxidants provides new avenues for discovery of alternatives to PZQ.
Collapse
Affiliation(s)
- Maria João Gouveia
- UCBIO/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo, 228, 4050-313 Porto, Portugal.
- Center for the Study of Animal Science, ICETA, University of Porto, Praça Gomes Teixeira, Apartado 55142, 4031-401 Porto, Portugal.
- Department of Molecular Pathology and Immunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal.
| | - Paul J Brindley
- Department of Microbiology, Immunology & Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, DC 20037, USA.
| | - Fátima Gärtner
- Department of Molecular Pathology and Immunology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal.
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal.
- Institute of Investigation and Innovation in Health (i3s), Rua Alfredo Allen, 4200-135 Porto, Portugal.
| | - José M Correia da Costa
- Center for the Study of Animal Science, ICETA, University of Porto, Praça Gomes Teixeira, Apartado 55142, 4031-401 Porto, Portugal.
- Department of Infectious Diseases, INSA-National Health Institute Dr. Ricardo Jorge, Rua Alexandre Herculano 321, 4000-055 Porto, Portugal.
| | - Nuno Vale
- UCBIO/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo, 228, 4050-313 Porto, Portugal.
| |
Collapse
|
32
|
Abstract
Praziquantel has remained the drug of choice for schistosomiasis chemotherapy for almost 40 years. The pressing need to develop a new antischistosomal drug may necessitate exploring and filtering chemotherapeutic history to search for the most promising ones. In this context, this review attempts to summarize all progress made in schistosomiasis chemotherapy from the early 20th century (mid-1910s) to 2016. We gathered almost 100 compounds providing information on therapeutic action, specifically covering at least first in vivo studies in animal model and in vitro. Pharmacokinetic and toxicity profiles of antischistosomal agents were also described. Preclinical studies indicate a handful of promising future candidates.
Collapse
|
33
|
Corstjens PLAM, Hoekstra PT, de Dood CJ, van Dam GJ. Utilizing the ultrasensitive Schistosoma up-converting phosphor lateral flow circulating anodic antigen (UCP-LF CAA) assay for sample pooling-strategies. Infect Dis Poverty 2017; 6:155. [PMID: 29089064 PMCID: PMC5664425 DOI: 10.1186/s40249-017-0368-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 10/09/2017] [Indexed: 01/08/2023] Open
Abstract
Background Methodological applications of the high sensitivity genus-specific Schistosoma CAA strip test, allowing detection of single worm active infections (ultimate sensitivity), are discussed for efficient utilization in sample pooling strategies. Besides relevant cost reduction, pooling of samples rather than individual testing can provide valuable data for large scale mapping, surveillance, and monitoring. Method The laboratory-based CAA strip test utilizes luminescent quantitative up-converting phosphor (UCP) reporter particles and a rapid user-friendly lateral flow (LF) assay format. The test includes a sample preparation step that permits virtually unlimited sample concentration with urine, reaching ultimate sensitivity (single worm detection) at 100% specificity. This facilitates testing large urine pools from many individuals with minimal loss of sensitivity and specificity. The test determines the average CAA level of the individuals in the pool thus indicating overall worm burden and prevalence. When requiring test results at the individual level, smaller pools need to be analysed with the pool-size based on expected prevalence or when unknown, on the average CAA level of a larger group; CAA negative pools do not require individual test results and thus reduce the number of tests. Results Straightforward pooling strategies indicate that at sub-population level the CAA strip test is an efficient assay for general mapping, identification of hotspots, determination of stratified infection levels, and accurate monitoring of mass drug administrations (MDA). At the individual level, the number of tests can be reduced i.e. in low endemic settings as the pool size can be increased as opposed to prevalence decrease. Conclusions At the sub-population level, average CAA concentrations determined in urine pools can be an appropriate measure indicating worm burden. Pooling strategies allowing this type of large scale testing are feasible with the various CAA strip test formats and do not affect sensitivity and specificity. It allows cost efficient stratified testing and monitoring of worm burden at the sub-population level, ideally for large-scale surveillance generating hard data for performance of MDA programs and strategic planning when moving towards transmission-stop and elimination.
Collapse
Affiliation(s)
- Paul L A M Corstjens
- Department of Molecular Cell Biology, Leiden University Medical Center, P.O. Box 9600, 2300, RC, Leiden, The Netherlands.
| | - Pytsje T Hoekstra
- Department of Parasitology, Leiden University Medical Center, P.O. Box 9600, 2300, RC, Leiden, The Netherlands
| | - Claudia J de Dood
- Department of Molecular Cell Biology, Leiden University Medical Center, P.O. Box 9600, 2300, RC, Leiden, The Netherlands
| | - Govert J van Dam
- Department of Parasitology, Leiden University Medical Center, P.O. Box 9600, 2300, RC, Leiden, The Netherlands
| |
Collapse
|
34
|
The anthelminthic drug praziquantel is a selective agonist of the sensory transient receptor potential melastatin type 8 channel. Toxicol Appl Pharmacol 2017; 336:55-65. [PMID: 29054683 DOI: 10.1016/j.taap.2017.10.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 10/10/2017] [Accepted: 10/16/2017] [Indexed: 11/23/2022]
Abstract
Praziquantel is the most effective anthelminthic drug for the treatment of schistosomiasis, an infectious disease caused by the platyhelminth Schistosoma mansoni. While praziquantel is known to trigger calcium influx into schisostomes, followed by spastic paralysis of the worms and tegumental disruption, the mechanism of action of the drug is not completely understood. Although relatively well tolerated, praziquantel has been reported to cause mild adverse effects, including nausea, abdominal pain and headaches. As a number of putative Transient Receptor Potential (TRP) channel genes have recently been predicted in S. mansoni, we sought to investigate the effect of praziquantel on three mammalian TRP channels, TRP melastatin type 8 (TRPM8), TRP vanilloid type 1 (TRPV1) and TRP ankyrin type 1 (TRPA1). Using calcium microfluorimetry and the patch clamp technique, we recorded the effect of praziquantel on HEK293T cells expressing recombinant TRPM8, TRPV1 or TRPA1, as well as on cultured dorsal root ganglion (DRG) neurons from wild type and TRPM8 null mutant mice. We discovered that praziquantel is a relatively potent and selective partial agonist of the mammalian and avian cold and menthol receptor TRPM8. The activation of cultured DRG neurons by clinically relevant concentrations of praziquantel is predominantly mediated by TRPM8. Our results may provide clues to a better understanding of praziquantel's mechanism of action and its adverse effects.
Collapse
|
35
|
Munisi DZ, Buza J, Mpolya EA, Angelo T, Kinung'hi SM. The Efficacy of Single-Dose versus Double-Dose Praziquantel Treatments on Schistosoma mansoni Infections: Its Implication on Undernutrition and Anaemia among Primary Schoolchildren in Two On-Shore Communities, Northwestern Tanzania. BIOMED RESEARCH INTERNATIONAL 2017; 2017:7035025. [PMID: 29094048 PMCID: PMC5637830 DOI: 10.1155/2017/7035025] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 08/08/2017] [Accepted: 08/21/2017] [Indexed: 12/16/2022]
Abstract
Administering more than one treatment may increase Praziquantel cure and egg reduction rates, thereby hastening achievement of schistosomiasis transmission control. A total of 431 S. mansoni-infected schoolchildren were randomized to receive either a single or repeated 40 mg/kg Praziquantel dose. Heights, weights, and haemoglobin levels were determined using a stadiometer, weighing scale, and HemoCue, respectively. At 8 weeks, cure rate was higher on repeated dose (93.10%) compared to single dose (68.68%) (p < 0.001). The egg reduction rate was higher on repeated dose (97.54%) compared to single dose (87.27%) (p = 0.0062). Geometric mean egg intensity was lower among those on repeated dose (1.30 epg) compared to single dose (3.18 epg) (p = 0.036) but not at 5 (p > 0.05) and 8 (p > 0.05) months with no difference in reinfection rate. No difference in the prevalence of stunting was observed between the two treatment regimens (p > 0.05) at 8 months, but there was an increase in the prevalence of wasting among those on repeated dose (p < 0.001). There was an increase in the mean haemoglobin levels at 8 months with no difference between the two arms (p > 0.05). To achieve reduction of transmission intensity and disease control in highly endemic areas, repeated treatments alone may not be sufficient. This trial was registered with PACTR201601001416338.
Collapse
Affiliation(s)
- David Z. Munisi
- Department of Global Health and Bio-Medical Sciences, School of Life Sciences and Bio-Engineering, Nelson Mandela African Institution of Science and Technology (NM-AIST), P.O. Box 447, Arusha, Tanzania
- Department of Bio-Medical Sciences, School of Medicine and Dentistry, College of Health Sciences, University of Dodoma, P.O. Box 259, Dodoma, Tanzania
| | - Joram Buza
- Department of Global Health and Bio-Medical Sciences, School of Life Sciences and Bio-Engineering, Nelson Mandela African Institution of Science and Technology (NM-AIST), P.O. Box 447, Arusha, Tanzania
| | - Emmanuel A. Mpolya
- Department of Global Health and Bio-Medical Sciences, School of Life Sciences and Bio-Engineering, Nelson Mandela African Institution of Science and Technology (NM-AIST), P.O. Box 447, Arusha, Tanzania
| | - Teckla Angelo
- Department of Global Health and Bio-Medical Sciences, School of Life Sciences and Bio-Engineering, Nelson Mandela African Institution of Science and Technology (NM-AIST), P.O. Box 447, Arusha, Tanzania
| | - Safari M. Kinung'hi
- National Institute for Medical Research (NIMR), Mwanza Research Centre, Isamilo Road, P.O. Box 1462, Mwanza, Tanzania
| |
Collapse
|
36
|
da Silva VBR, Campos BRKL, de Oliveira JF, Decout JL, do Carmo Alves de Lima M. Medicinal chemistry of antischistosomal drugs: Praziquantel and oxamniquine. Bioorg Med Chem 2017; 25:3259-3277. [DOI: 10.1016/j.bmc.2017.04.031] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/21/2017] [Accepted: 04/26/2017] [Indexed: 12/20/2022]
|
37
|
Campelo YDM, Mafud AC, Véras LMC, Guimarães MA, Yamaguchi LF, Lima DF, Arcanjo DDR, Kato MJ, Mendonça RZ, Pinto PLS, Mascarenhas YP, Silva MPN, de Moraes J, Eaton P, de Souza de Almeida Leite JR. Synergistic effects of in vitro combinations of piplartine, epiisopiloturine and praziquantel against Schistosoma mansoni. Biomed Pharmacother 2017; 88:488-499. [PMID: 28126674 DOI: 10.1016/j.biopha.2016.12.057] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 12/09/2016] [Accepted: 12/14/2016] [Indexed: 01/22/2023] Open
Abstract
Schistosomiasis is a world health problem, and praziquantel is the only drug currently used for the treatment. There is some evidence that extensive monotherapy of praziquantel may be leading to drug resistance in the parasite. In order to find alternative treatments, the effects of the combination of epiisopiloturine (EPI), piplartine (PPT) and praziquantel (PZQ) were evaluated. Similarity analysis of these compounds was performed using optimized molecular structures to compare the shape and the charge modeling of combinations between PZQ and EPI or PPT. Supported by this data, in vitro association of PZQ-PPT, PZQ-EPI, and EPI-PPT was carried out, and the activity of these combinations against Schistosoma mansoni was assessed. The results showed synergistic activity with a combination index (CI) of 0.42 for the treatment with PZQ-PPT. Both PZQ-EPI and EPI-PPT combinations also showed synergistic effects, with CI values of 0.86 and 0.61, respectively. Surface alterations in the tegument of adult schistosomes after the treatments were observed using laser confocal microscopy and scanning electron microscopy. Additionally, the association of EPI-PPT decreased the cytotoxicity when compared with both isolated compounds in three different lines of mammalian cells. Thus, synergistic combinations of PZQ-PPT, PZQ-EPI, and EPI-PPT create the possibility of reduced doses to be used against Schistosoma mansoni.
Collapse
Affiliation(s)
- Yuri Dias Macedo Campelo
- Biodiversity and Biotechnology Research Center, Biotec, Federal University of Piauí, UFPI, Parnaíba, PI, 64202020, Brazil; Institute of Higher Education of Vale do Parnaíba, FAHESP/IESVAP, Parnaíba, PI, Brazil
| | - Ana Carolina Mafud
- São Carlos Institute of Physics, University of São Paulo, São Carlos, SP, Brazil
| | - Leiz Maria Costa Véras
- Biodiversity and Biotechnology Research Center, Biotec, Federal University of Piauí, UFPI, Parnaíba, PI, 64202020, Brazil
| | - Maria Adelaide Guimarães
- Biodiversity and Biotechnology Research Center, Biotec, Federal University of Piauí, UFPI, Parnaíba, PI, 64202020, Brazil; Phytobios Nordeste LTDA, Centroflora Group, Parnaíba, PI, Brazil
| | - Lydia F Yamaguchi
- Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil
| | - David Fernandes Lima
- Biodiversity and Biotechnology Research Center, Biotec, Federal University of Piauí, UFPI, Parnaíba, PI, 64202020, Brazil; Federal University of Vale do São Francisco, Paulo Afonso, BA, Brazil
| | - Daniel Dias Rufino Arcanjo
- Biodiversity and Biotechnology Research Center, Biotec, Federal University of Piauí, UFPI, Parnaíba, PI, 64202020, Brazil
| | - Massuo J Kato
- Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil
| | | | | | | | - Marcos P N Silva
- Research Center for Neglected Diseases, Guarulhos University, Guarulhos, SP, Brazil
| | - Josué de Moraes
- Research Center for Neglected Diseases, Guarulhos University, Guarulhos, SP, Brazil
| | - Peter Eaton
- Biodiversity and Biotechnology Research Center, Biotec, Federal University of Piauí, UFPI, Parnaíba, PI, 64202020, Brazil; UCIBIO/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Portugal
| | - José Roberto de Souza de Almeida Leite
- Biodiversity and Biotechnology Research Center, Biotec, Federal University of Piauí, UFPI, Parnaíba, PI, 64202020, Brazil; Faculty of Medicine, University of Brasilia, UnB, Brasília, DF, Brazil.
| |
Collapse
|
38
|
Hailu GS, Robaa D, Forgione M, Sippl W, Rotili D, Mai A. Lysine Deacetylase Inhibitors in Parasites: Past, Present, and Future Perspectives. J Med Chem 2017; 60:4780-4804. [DOI: 10.1021/acs.jmedchem.6b01595] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Gebremedhin S. Hailu
- Dipartimento
di Chimica e Tecnologie del Farmaco “Sapienza” Università di Roma, 00185 Rome, Italy
| | - Dina Robaa
- Institute of Pharmacy, Martin-Luther-Universitat Halle-Wittenberg, Halle, Germany
| | - Mariantonietta Forgione
- Dipartimento
di Chimica e Tecnologie del Farmaco “Sapienza” Università di Roma, 00185 Rome, Italy
- Center
for Life Nano Science@Sapienza, Italian Institute of Technology, Viale Regina Elena 291, 00161 Rome, Italy
| | - Wolfgang Sippl
- Institute of Pharmacy, Martin-Luther-Universitat Halle-Wittenberg, Halle, Germany
| | - Dante Rotili
- Dipartimento
di Chimica e Tecnologie del Farmaco “Sapienza” Università di Roma, 00185 Rome, Italy
| | - Antonello Mai
- Dipartimento
di Chimica e Tecnologie del Farmaco “Sapienza” Università di Roma, 00185 Rome, Italy
- Istituto
Pasteur, Fondazione Cenci-Bolognetti, “Sapienza” Università di Roma, 00185 Rome, Italy
| |
Collapse
|
39
|
Abstract
The concept of neglected tropical diseases (NTDs) emerged more than a decade ago and has been recognised as a valid way to categorise diseases that affect the poorest individuals. Substantial progress in control and elimination has been achieved and policy momentum has been generated through continued bilateral, philanthropic, and non-governmental development organisation (NGDO) support, and donations of drugs from pharmaceutical companies. WHO has defined a Roadmap to reach 2020 targets, which was endorsed by member states in a World Health Assembly Resolution in 2013. NTDs have been included within the Sustainable Development Goal targets and are a crucial component of universal health coverage, conceptualised as "leaving no one behind". WHO reported that more than 1 billion people in 88 countries have benefited from preventive chemotherapy in 2014. The research agenda has defined the need for affordable products (diagnostics, drugs and insecticides). However challenges such as insecurity and weak health systems continue to prevail in the poorest countries, inhibiting progress in scaling up and also in achieving Roadmap goals.
Collapse
Affiliation(s)
- David H Molyneux
- Department of Parasitology, Liverpool School of Tropical Medicine, Liverpool, UK.
| | - Lorenzo Savioli
- Global Schistosomiasis Alliance, Chavannes de Bogis, Switzerland
| | - Dirk Engels
- Department of Control of Neglected Tropical Diseases, World Health Organization, Geneva, Switzerland
| |
Collapse
|
40
|
Kong QM, Zhu X, Tong QB, Zheng B, Shi NY, Lou D, Ding JZ, Jia JP, Chen XH, Chen R, Lu SH. Genome-wide miRNAs expression profiles of Schistosoma japonicum schistosomula in response to artesunate. Pharmacogenomics 2016; 17:2025-2037. [DOI: 10.2217/pgs.16.23] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim: miRNAs play a significant role in pharmacogenomics and are likely to be important in the molecular mechanism of atesunate (ART) effects on Schistosoma japonicum. Methods: We sequenced the RNAs using an Illumina (Solexa) DNA sequencer and compared the relative expression levels of the miRNAs in 10-day-old schistosomula from ART and the parallel control group. Results: We characterized 95 known miRNAs from S. japonicum schistosomula individuals, including 38 novel miRNA families. Among the detectable 134 miRNAs differentially expressed (>2.0-fold change, p < 0.01) after ART treatment in schistosomula, a total of seven known or novel 3p- or 5p- derived S. japonicum miRNAs were characterized. We propose that sja-miR-125b may regulate the expression of ART metabolizing enzymes, glutathione synthetase or heme-binding protein 2 to help S. japonicum resists or adapts to drug stress and also ART may significantly inhibit sexual maturation of female worms mediated by mir-71b/2 miRNA cluster. Conclusion: This was the first comprehensive miRNAs expression profile analysis of S. japonicum in response to ART, and provides an overview of the complex network of the mechanism of action of ART on S. japonicum.
Collapse
Affiliation(s)
- Qing-Ming Kong
- Department of Immunity & Biochemistry, Institute of Parasitic Disease, Zhejiang Academy of Medical Sciences, No.182, Road Tianmushan, 310013, Hangzhou, China
| | - Xiao Zhu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Scientific Research Center, Guangdong Medical University, 523808, Dongguan, China
| | - Qun-Bo Tong
- Department of Immunity & Biochemistry, Institute of Parasitic Disease, Zhejiang Academy of Medical Sciences, No.182, Road Tianmushan, 310013, Hangzhou, China
| | - Bin Zheng
- Department of Immunity & Biochemistry, Institute of Parasitic Disease, Zhejiang Academy of Medical Sciences, No.182, Road Tianmushan, 310013, Hangzhou, China
| | - Na-Yu Shi
- Department of Gynecology, Hangzhou Obstetrics & Gynecology Hospital, 310013, Hangzhou, China
| | - Di Lou
- Department of Immunity & Biochemistry, Institute of Parasitic Disease, Zhejiang Academy of Medical Sciences, No.182, Road Tianmushan, 310013, Hangzhou, China
| | - Jian-Zu Ding
- Department of Immunity & Biochemistry, Institute of Parasitic Disease, Zhejiang Academy of Medical Sciences, No.182, Road Tianmushan, 310013, Hangzhou, China
| | - Jian-Ping Jia
- Department of Immunity & Biochemistry, Institute of Parasitic Disease, Zhejiang Academy of Medical Sciences, No.182, Road Tianmushan, 310013, Hangzhou, China
| | - Xiao-Heng Chen
- Department of Immunity & Biochemistry, Institute of Parasitic Disease, Zhejiang Academy of Medical Sciences, No.182, Road Tianmushan, 310013, Hangzhou, China
| | - Rui Chen
- Department of Immunity & Biochemistry, Institute of Parasitic Disease, Zhejiang Academy of Medical Sciences, No.182, Road Tianmushan, 310013, Hangzhou, China
| | - Shao-Hong Lu
- Department of Immunity & Biochemistry, Institute of Parasitic Disease, Zhejiang Academy of Medical Sciences, No.182, Road Tianmushan, 310013, Hangzhou, China
| |
Collapse
|
41
|
Allam G, Abuelsaad AS, Alblihed MA, Alsulaimani AA. Ellagic acid reduces murine schistosomiasis mansoni immunopathology via up-regulation of IL-10 and down-modulation of pro-inflammatory cytokines production. Immunopharmacol Immunotoxicol 2016; 38:286-97. [DOI: 10.1080/08923973.2016.1189561] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
42
|
Chevalier FD, Le Clec'h W, Eng N, Rugel AR, Assis RRD, Oliveira G, Holloway SP, Cao X, Hart PJ, LoVerde PT, Anderson TJC. Independent origins of loss-of-function mutations conferring oxamniquine resistance in a Brazilian schistosome population. Int J Parasitol 2016; 46:417-24. [PMID: 27073078 DOI: 10.1016/j.ijpara.2016.03.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 03/31/2016] [Accepted: 03/31/2016] [Indexed: 11/28/2022]
Abstract
Molecular surveillance provides a powerful approach to monitoring the resistance status of parasite populations in the field and for understanding resistance evolution. Oxamniquine was used to treat Brazilian schistosomiasis patients (mid-1970s to mid-2000s) and several cases of parasite infections resistant to treatment were recorded. The gene underlying resistance (SmSULT-OR) encodes a sulfotransferase required for intracellular drug activation. Resistance has a recessive basis and occurs when both SmSULT-OR alleles encode for defective proteins. Here we examine SmSULT-OR sequence variation in a natural schistosome population in Brazil ∼40years after the first use of this drug. We sequenced SmSULT-OR from 189 individual miracidia (1-11 per patient) recovered from 49 patients, and tested proteins expressed from putative resistance alleles for their ability to activate oxamniquine. We found nine mutations (four non-synonymous single nucleotide polymorphisms, three non-coding single nucleotide polymorphisms and two indels). Both mutations (p.E142del and p.C35R) identified previously were recovered in this field population. We also found two additional mutations (a splice site variant and 1bp coding insertion) predicted to encode non-functional truncated proteins. Two additional substitutions (p.G206V, p.N215Y) tested had no impact on oxamniquine activation. Three results are of particular interest: (i) we recovered the p.E142del mutation from the field: this same deletion is responsible for resistance in an oxamniquine selected laboratory parasite population; (ii) frequencies of resistance alleles are extremely low (0.27-0.8%), perhaps due to fitness costs associated with carriage of these alleles; (iii) that four independent resistant alleles were found is consistent with the idea that multiple mutations can generate loss-of-function alleles.
Collapse
Affiliation(s)
- Frédéric D Chevalier
- Texas Biomedical Research Institute, Department of Genetics, P.O. Box 760549, San Antonio, TX 78245-0549, USA.
| | - Winka Le Clec'h
- Texas Biomedical Research Institute, Department of Genetics, P.O. Box 760549, San Antonio, TX 78245-0549, USA
| | - Nina Eng
- Texas Biomedical Research Institute, Department of Genetics, P.O. Box 760549, San Antonio, TX 78245-0549, USA
| | - Anastasia R Rugel
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA; Department of Pathology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Rafael Ramiro de Assis
- Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Av. Augusto de Lima, 1715, Belo Horizonte, Minas Gerais 30190-002, Brazil
| | - Guilherme Oliveira
- Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Av. Augusto de Lima, 1715, Belo Horizonte, Minas Gerais 30190-002, Brazil; Vale Institute of Technology, Rua Boaventura da Silva, 955, Belém, Pará 66055-090, Brazil
| | - Stephen P Holloway
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA; Department of Pathology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Xiaohang Cao
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA; Department of Pathology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - P John Hart
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA; Department of Pathology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA; Department of Veterans Affairs, South Texas Veterans Health Care System, San Antonio, TX 78229, USA
| | - Philip T LoVerde
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA; Department of Pathology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Timothy J C Anderson
- Texas Biomedical Research Institute, Department of Genetics, P.O. Box 760549, San Antonio, TX 78245-0549, USA.
| |
Collapse
|
43
|
Kong Q, Tong Q, Lou D, Ding J, Zheng B, Chen R, Zhu X, Chen X, Dong K, Lu S. Quantitative proteomic analyses of Schistosoma japonicum in response to artesunate. MOLECULAR BIOSYSTEMS 2016; 11:1400-9. [PMID: 25820832 DOI: 10.1039/c5mb00074b] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Artesunate (ART) has high prophylactic efficacy against Schistosoma japonicum infections and has been used to treat and prevent schistosomiasis in China since 1995. However, the molecular mechanism of ART's effects on S. japonicum remains unclear. Herein, we applied isobaric tagging reagents for relative and absolute quantification analyses coupled with two-dimensional liquid chromatography and tandem mass spectrometry to investigate the effect of ART on the proteome of S. japonicum in susceptible mice. 4529 proteins were quantified on the basis of 21,825 unique peptides. Comparative proteomic analyses revealed that 145, 228 and 185 proteins were significantly differentially expressed after ART treatment in schistosomula, juvenile and adult worms, respectively. Ninety proteins were differentially expressed between each two treatment groups in response to ART treatment: 67 proteins were associated with S. japonicum development/aging and 23 were specifically associated with ART treatment. Quantitative real-time PCR of selected genes verified the proteomic data. Gene ontology annotation and Kyoto encyclopedia of genes and genomes pathway mapping analysis showed that the majority of differentially expressed proteins were involved in stress/defense/detoxification, signal transduction, carbohydrate metabolism, amino acid metabolism, transcription/translation, and protein synthesis/assembly/degradation. Thirty-four of the proteins differentially expressed under ART treatment encoded hypothetical, uncharacterized proteins with unknown functions. This study obtained the first comprehensive protein expression profile of S. japonicum in response to ART, and provides a basis for a better understanding of the molecular mechanisms of ART effects on S. japonicum.
Collapse
Affiliation(s)
- QingMing Kong
- Department of Immunity and Biochemistry, Institute of Parasitic Disease, Zhejiang Academy of Medical Sciences, Hangzhou, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
El-Emam MAW, Mahmoud SS, Bayaumy FELZA. Potential role of mefloquine (antimalarial drug) and methanol extract of Chenopodium ambrosioides and Sesbania sesban in mice infected with Schistosoma mansoni. ASIAN PACIFIC JOURNAL OF TROPICAL DISEASE 2015. [DOI: 10.1016/s2222-1808(15)60898-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
45
|
Abdel-Monaem G, Farid A, Rabia I, El-Amir A. Evaluation of Echinostoma liei worm, metacercaria and redia antigens for schistosomiasis control. Exp Parasitol 2015; 157:23-9. [PMID: 26115938 DOI: 10.1016/j.exppara.2015.06.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 05/12/2015] [Accepted: 06/15/2015] [Indexed: 11/25/2022]
Abstract
While chemotherepeutic drugs, such as praziquantel, oxamniquine and metrifonate, are currently considered safe and effective drugs for schistosomiasis treatment, reinfection occurs frequently after drug treatment. Thus, a vaccine is sought to provide long-term treatment. Antigens from worm, metacercaria and redia of Echinostoma liei (E. liei) were purified using CNBr-activated Sepharose column, then used for immunization of mice prior to infection with Schistosomiasis mansoni. Worm burden, hepatic and intestinal eggs and oogram count was significantly reduced and that was reflected in normalization of liver architecture. This referred to a significant increase in the tested immunoglobulin level (IgM, IgG1 and IgG2).
Collapse
Affiliation(s)
- G Abdel-Monaem
- Zoology Dept., Faculty of Science, Cairo University, Giza, Egypt
| | - A Farid
- Zoology Dept., Faculty of Science, Cairo University, Giza, Egypt.
| | - I Rabia
- Parasitology Dept., Theodore Bilharz Research Institute, Giza, Egypt
| | - A El-Amir
- Zoology Dept., Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
46
|
Drug-induced exposure of Schistosoma mansoni antigens SmCD59a and SmKK7. PLoS Negl Trop Dis 2015; 9:e0003593. [PMID: 25774883 PMCID: PMC4361651 DOI: 10.1371/journal.pntd.0003593] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 02/05/2015] [Indexed: 11/22/2022] Open
Abstract
Background Schistosomiasis is a serious health problem especially in developing countries and affects more than 243 million people. Only few anthelmintic drugs are available up to now. A major obstacle for drug treatment is the different developmental stages and the varying host compartments during worm development. Anthelmintic drugs have been tested mainly on adult schistosomes or freshly transformed cercariae. Knowledge concerning the larval stages is lacking. Methodology/Principal Findings In this study, we used in vitro-grown schistosomula (aged between 2 to 14 days) to investigate drug effects of the three anthelmintics praziquantel, artemether, and oxamniquine. Further, we analyzed the antibody accessibility of two exemplary schistosome antigens SmCD59a and SmKK7, before and after drug treatment. Our results demonstrated that praziquantel applied at a concentration of 1 μM inhibited development of all life stages. Application of 10 μM praziquantel led to dramatic morphological changes of all schistosomula. Artemether at 1 and 10 μM had differential effects depending on whether it was applied to 2-day as compared to 7- and 14-day schistosomula. While 2-day schistosomula were not killed but inhibited from further development, severe morphological damage was seen in 7- and 14-day schistosomula. Oxamniquine (1 and 10 μM) led to severe morphological impairment in all life stages. Analyzing the accessibility of the antigens SmCD59a and SmKK7 before drug treatment showed no antibody binding on living intact schistosomula. However, when schistosomula were treated with anthelmintics, both antigens became exposed on the larvae. Oxamniquine turned out to be most effective in promoting antibody binding to all schistosomula stages. Conclusion This study has revealed marked differences in anthelmintic drug effects against larvae. Drug treatment increases surface antigen presentation and renders larvae accessible to antibody attack. Schistosomiasis is one of the major parasitic diseases in developing countries and still causes 200,000 deaths per year. Mass drug administration programs with praziquantel, the drug of choice against schistosomiasis, are currently undertaken in Sub-Saharan Africa. Praziquantel, although efficient against adult worms, fails to cure early infection. The complex developmental stages of schistosomes and migration through varying host compartments with different local drug concentration are a challenge for drug treatment. After infecting their mammalian host, schistosomula traverse through skin and the vasculature of lung, liver and intestines. During their migration, they develop from larvae to paired adults in approximately 4 to 5 weeks. So far, drug effects have been analyzed on adult worms or freshly transformed schistosomula only. Information about the effects on the larval stages is lacking. We were able to transfer the larval development of the first three weeks into the culture dish. This tool can be used for the analysis of drug effects against schistosomula and for investigation of the accessibility, expression and localization of antigens. Rendering the parasite’s larvae vulnerable to the host’s immune system by increasing antigen presentation is an important aspect of drug activity. We demonstrate on in vitro-cultured Schistosoma mansoni larvae, that SmCD59a and SmKK7, as examples for hidden antigens, become accessible to antibodies following drug treatment.
Collapse
|
47
|
Guimarães MA, de Oliveira RN, Véras LMC, Lima DF, Campelo YDM, Campos SA, Kuckelhaus SAS, Pinto PLS, Eaton P, Mafud AC, Mascarenhas YP, Allegretti SM, de Moraes J, Lolić A, Verbić T, Leite JRSA. Anthelmintic activity in vivo of epiisopiloturine against juvenile and adult worms of Schistosoma mansoni. PLoS Negl Trop Dis 2015; 9:e0003656. [PMID: 25816129 PMCID: PMC4376696 DOI: 10.1371/journal.pntd.0003656] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 02/27/2015] [Indexed: 02/02/2023] Open
Abstract
Schistosomiasis is a serious disease currently estimated to affect more that 207 million people worldwide. Due to the intensive use of praziquantel, there is increasing concern about the development of drug-resistant strains. Therefore, it is necessary to search for and investigate new potential schistosomicidal compounds. This work reports the in vivo effect of the alkaloid epiisopiloturine (EPI) against adults and juvenile worms of Schistosoma mansoni. EPI was first purified its thermal behavior and theoretical solubility parameters charaterised. In the experiment, mice were treated with EPI over the 21 days post-infection with the doses of 40 and 200 mg/kg, and 45 days post-infection with single doses of 40, 100 and 300 mg/kg. The treatment with EPI at 40 mg/kg was more effective in adult worms when compared with doses of 100 and 300 mg/kg. The treatment with 40 mg/kg in adult worms reduced parasite burden significantly, lead to reduction in hepatosplenomegaly, reduced the egg burden in faeces, and decreased granuloma diameter. Scanning electron microscopy revealed morphological changes to the parasite tegument after treatment, including the loss of important features. Additionally, the in vivo treatment against juvenile with 40 mg/kg showed a reduction of the total worm burden of 50.2%. Histopathological studies were performed on liver, spleen, lung, kidney and brain and EPI was shown to have a DL50 of 8000 mg/kg. Therefore EPI shows potential to be used in schistosomiasis treatment. This is the first time that schistosomicidal in vivo activity of EPI has been reported.
Collapse
Affiliation(s)
- Maria A. Guimarães
- Biotechnology and Biodiversity Center Research, BIOTEC, Federal University of Piauí, Parnaíba, Piauí, Brazil
| | - Rosimeire N. de Oliveira
- Department of Animal Biology, Institute of Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Leiz M. C. Véras
- Biotechnology and Biodiversity Center Research, BIOTEC, Federal University of Piauí, Parnaíba, Piauí, Brazil
- Graduate Program in Biotechnology, RENORBIO, Focal Point Federal University of Piauí, Teresina, Piauí, Brazil
| | - David F. Lima
- Biotechnology and Biodiversity Center Research, BIOTEC, Federal University of Piauí, Parnaíba, Piauí, Brazil
- Graduate Program in Biotechnology, RENORBIO, Focal Point Federal University of Piauí, Teresina, Piauí, Brazil
- Collegiate Academic Medicine, Federal University of São Francisco Valley, Campus Paulo Afonso, Paulo Afonso, Bahia, Brazil
| | - Yuri D. M. Campelo
- Biotechnology and Biodiversity Center Research, BIOTEC, Federal University of Piauí, Parnaíba, Piauí, Brazil
- Graduate Program in Biotechnology, RENORBIO, Focal Point Federal University of Piauí, Teresina, Piauí, Brazil
| | - Stefano Augusto Campos
- Biotechnology and Biodiversity Center Research, BIOTEC, Federal University of Piauí, Parnaíba, Piauí, Brazil
| | - Selma A. S. Kuckelhaus
- Faculty of Medicine, University of Brasilia, UNB Campus Dacy Ribeiro, Brasília, Distrito Federal, Brazil
| | | | - Peter Eaton
- UCIBIO, REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Science, University of Porto, Portugal
| | - Ana C. Mafud
- Group of Crystallography, Institute of Physics of São Carlos, University of São Paulo, São Carlos, São Paulo, Brazil
| | - Yvonne P. Mascarenhas
- Group of Crystallography, Institute of Physics of São Carlos, University of São Paulo, São Carlos, São Paulo, Brazil
| | - Silmara M. Allegretti
- Department of Animal Biology, Institute of Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Josué de Moraes
- Research Center for Neglected Diseases (NPDN/FACIG), Guarulhos, São Paulo, Brazil
| | | | - Tatjana Verbić
- Faculty of Chemistry, University of Belgrade, Belgrade, Serbia
| | - José Roberto S. A. Leite
- Biotechnology and Biodiversity Center Research, BIOTEC, Federal University of Piauí, Parnaíba, Piauí, Brazil
| |
Collapse
|
48
|
Abstract
Schistosomiasis is a chronic disease that affects ∼200 million people. The extended health impact of the disease has been estimated to exceed that of malaria or tuberculosis and to be nearer to that of HIV/AIDS. Within endemic areas, children carry the heaviest burden of infection. Infection/disease is controlled by the treatment of infected subjects with the anthelminthic drug praziquantel. Global initiatives from Partners of Parasite Control, including the World Health Organization (WHO), advocate regular school-based deworming strategies to reduce the development of severe morbidity, promote school-child health and development, and improve the cognitive potential of children. Until recently, preschool-aged children were excluded from schistosome treatment, creating a health inequity in affected populations. In 2010, the WHO updated their recommendations for the treatment of schistosomiasis in preschool-aged children (ie, children aged ≤5 years). This change was the culmination of several decades of research on schistosome epidemiology, immunology, and pathology in this age group. The recent development of a pediatric formulation of praziquantel (soon to enter clinical trials) should advance control efforts in preschool-aged children, with the goal of including these children in preventative chemotherapy (as currently occurs for soil-transmitted helminths). This review discusses the research work supporting the WHO revision of recommendations for treating preschool-aged children, as well as current barriers and knowledge gaps in pediatric schistosomiasis control.
Collapse
Affiliation(s)
- Francisca Mutapi
- Institute of Immunology and Infection Research, Centre for Immunity, Infection and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
49
|
Barakat R, Abou El-Ela NE, Sharaf S, El Sagheer O, Selim S, Tallima H, Bruins MJ, Hadley KB, El Ridi R. Efficacy and safety of arachidonic acid for treatment of school-age children in Schistosoma mansoni high-endemicity regions. Am J Trop Med Hyg 2015; 92:797-804. [PMID: 25624403 PMCID: PMC4385776 DOI: 10.4269/ajtmh.14-0675] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Accepted: 12/19/2014] [Indexed: 02/04/2023] Open
Abstract
Arachidonic acid (ARA), an omega-6 fatty acid, is a potent schistosomicide that displayed significant and safe therapeutic effects in Schistosoma mansoni-infected schoolchildren in S. mansoni low-prevalence regions. We here report on ARA efficacy and safety in treatment of schoolchildren in S. mansoni high-endemicity areas of Kafr El Sheikh, Egypt. The study was registered with ClinicalTrials.gov (NCT02144389). In total, 268 schoolchildren with light, moderate, or heavy S. mansoni infection were assigned to three study arms of 87, 91, and 90 children and received a single dose of 40 mg/kg praziquantel (PZQ), ARA (10 mg/kg per day for 15 days), or PZQ combined with ARA, respectively. The children were examined before and after treatment for stool parasite egg counts and blood biochemical, hematological, and immunological parameters. ARA, like PZQ, induced moderate cure rates (50% and 60%, respectively) in schoolchildren with light infection and modest cure rates (21% and 20%, respectively) in schoolchildren with high infection. PZQ and ARA combined elicited 83% and 78% cure rates in children with light and heavy infection, respectively. Biochemical and immunological profiles were either unchanged or ameliorated after ARA therapy. Combination of PZQ and ARA might be useful for treatment of children with schistosomiasis in high-endemicity regions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Rashika El Ridi
- *Address correspondence to Rashika El Ridi, Zoology Department, Faculty of Science, Cairo University, Cairo 12613, Egypt. E-mail:
| |
Collapse
|
50
|
Dorosti Z, Yousefi M, Sharafi SM, Darani HY. Mutual action of anticancer and antiparasitic drugs: are there any shared targets? Future Oncol 2014; 10:2529-39. [DOI: 10.2217/fon.14.65] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
ABSTRACT Parasites and cancers have some common features. It has been shown that some parasites interfere with tumor growth. In addition, they both have common antigens such as the Tn antigen. A communal action of anticancer and antiparasitic drugs has been reported. This shared action may be related to common targets for these drugs in cancers and parasites. In this paper, mutual action of anticancer and antiparasitic drugs, with the aim of discussing shared targets of these drugs, has been reviewed.
Collapse
Affiliation(s)
- Zahra Dorosti
- Department of Parasitology & Mycology, Faculty of Medicine, Isfahan University of Medical sciences, Isfahan, Iran
| | - Morteza Yousefi
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Seyedeh Maryam Sharafi
- Infectious Diseases & Tropical medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Yousofi Darani
- Cancer Prevention Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|