1
|
Chatterji T, Khanna N, Alghamdi S, Bhagat T, Gupta N, Alkurbi MO, Sen M, Alghamdi SM, Bamagous GA, Sahoo DK, Patel A, Kumar P, Yadav VK. A Recent Advance in the Diagnosis, Treatment, and Vaccine Development for Human Schistosomiasis. Trop Med Infect Dis 2024; 9:243. [PMID: 39453270 PMCID: PMC11511416 DOI: 10.3390/tropicalmed9100243] [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: 08/28/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Schistosomiasis, which affects a large number of people worldwide, is among the most overlooked parasitic diseases. The disease is mainly prevalent in sub-Saharan Africa, southeast Asian countries, and South America due to the lack of adequate sanitation. The disease is mainly associated with poor hygiene, sanitation, and contaminated water, so it is also known as a disease of poverty. Three Schistosoma species (S. mansoni, S. japonicum, and S. haematobium) cause significant human infections. Co-infections with Schistosoma and other parasites are widely common. All these parasites may cause intestinal or urogenital schistosomiasis, where the disease may be categorized into the acute, sensitized, and chronic phases. The disease is more prevalent among school children, which may cause anemia and reduce development. Chronic infections frequently cause significant liver, intestinal, and bladder damage. Women exposed to contaminated water while performing normal duties like washing clothes might acquire urogenital schistosomiasis (UGS), which can cause tissue damage and raise the risk of blood-borne disease transmission, including human immunodeficiency virus (HIV) transmission. Praziquantel (PZQ) is the World Health Organization (WHO)-prescribed treatment for individuals who are known to be infected, but it does not prevent further re-infections with larval worms. Vaccine development and new molecular-based diagnosis techniques have promised to be a reliable approach to the diagnosis and prevention of schistosomiasis. The current review emphasizes the recent advancement in the diagnosis of schistosomiasis by molecular techniques and the treatment of schistosomiasis by combined and alternative regimes of drugs. Moreover, this review has also focused on the recent outbreak of schistosomiasis, the development of vaccines, and their clinical trials.
Collapse
Affiliation(s)
- Tanushri Chatterji
- Department of Biosciences, Institute of Management Studies Ghaziabad (University Courses Campus), Adhyatmik Nagar, NH-09, Ghaziabad 201015, Uttar Pradesh, India;
| | - Namrata Khanna
- Department of Biochemistry, M A Rangoonwala College of Dental Sciences and Research Centre, 2390-B, K.B. Hidayatullah Road, Azam Campus, Camp, Pune 411001, Maharashtra, India;
| | - Saad Alghamdi
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (S.A.); (M.O.A.)
| | - Tanya Bhagat
- Department of Biosciences, Institute of Management Studies Ghaziabad (University Courses Campus), Adhyatmik Nagar, NH-09, Ghaziabad 201015, Uttar Pradesh, India;
| | - Nishant Gupta
- Engineering Department, River Engineering Pvt Ltd., Toy City, Ecotech–III, Greater Noida 201306, Uttar Pradesh, India;
| | - Mohammad Othman Alkurbi
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (S.A.); (M.O.A.)
| | - Manodeep Sen
- Department of Microbiology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Vibhuti Khand, Gomti Nagar, Lucknow 226010, Uttar Pradesh, India;
| | - Saeed Mardy Alghamdi
- Respiratory Care Program, Clinical Technology Department, Faculty of Applied Medical Science, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Ghazi A. Bamagous
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Dipak Kumar Sahoo
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA;
| | - Ashish Patel
- Department of Life Sciences, Hemchandracharya North Gujarat University, Patan 384265, Gujarat, India;
| | - Pankaj Kumar
- Department of Environmental Science, Parul Institute of Applied Sciences, Parul University, Vadodara 391760, Gujarat, India;
| | - Virendra Kumar Yadav
- Marwadi University Research Center, Department of Microbiology, Faculty of Sciences, Marwadi University, Rajkot 360003, Gujarat, India
| |
Collapse
|
2
|
Chen H, Huang S, Yao S, Wang J, Huang J, Yu Z. Multi-omics analyses of Bacillus amyloliquefaciens treated mice infected with Schistosoma japonicum reveal dynamics change of intestinal microbiome and its associations with host metabolism. PLoS Negl Trop Dis 2024; 18:e0012583. [PMID: 39466852 PMCID: PMC11515987 DOI: 10.1371/journal.pntd.0012583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Schistosomiasis japonica is a serious threat to human health. It causes damage to the intestine and liver. Probiotic therapy has been shown to be effective in alleviating intestinal diseases and improving host health. Previous studies have found that Bacillus amyloliquefaciens could alleviate the pathological symptoms of schistosomiasis japonica, but the regulatory mechanism of alleviating schistosomiasis japonica is still unknown. PRINCIPAL FINDINGS This study analyzed the dynamic changes of intestinal microbiome in mice infected with Schistosoma japonicum after the intervention of B. amyloliquefaciens and its connection to host metabolism by multi-omics sequencing technology. B. amyloliquefaciens was found to significantly regulate the homeostasis of intestinal microbiota by promoting the growth of beneficial bacteria and inhibiting potential pathogenic bacteria and protect the number of core microbes. Meanwhile, the genes related to the metabolism of glycerophospholipids and amino acid from intestinal microbiome changed significantly, and were shown to be significantly positively correlated with the associated metabolites of microbial origin. Moreover, host metabolism (lipid metabolism and steroid hormone biosynthesis) was also found to be significantly regulated. CONCLUSIONS The recovery of intestinal microbial homeostasis and the regulation of host metabolism revealed the potential probiotic properties of B. amyloliquefaciens, which also provided new ideas for the prevention and adjuvant treatment of schistosomiasis japonica.
Collapse
Affiliation(s)
- Hao Chen
- Human Microbiome and Health Group, Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Shuaiqin Huang
- Human Microbiome and Health Group, Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Siqi Yao
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Jingyan Wang
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Jing Huang
- Human Microbiome and Health Group, Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Zheng Yu
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| |
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
|
Kim J, Davis J, Lee J, Cho SN, Yang K, Yang J, Bae S, Son J, Kim B, Whittington D, Siddiqui AA, Carter D, Gray SA. An assessment of a GMP schistosomiasis vaccine (SchistoShield ®). FRONTIERS IN TROPICAL DISEASES 2024; 5:1404943. [PMID: 39483645 PMCID: PMC11525685 DOI: 10.3389/fitd.2024.1404943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024] Open
Abstract
Introduction Schistosomiasis is a neglected tropical disease that puts over 200 million people at risk, and prevention options are sparse with no approved vaccine. Our vaccine candidate, SchistoShield®, is based on an approximately 87 kDa large subunit of calcium activated neutral protease - termed Sm-p80 - combined with a potent TLR4 agonist-based adjuvant. SchistoShield® has been shown to prevent disease throughout the parasitic life cycle - including egg, juvenile, and adult worm stages - in numerous animal models up to and including baboons. SchistoShield® has been shown safe in both preclinical toxicology studies in rabbits and in a Phase 1 clinical trial in the USA. A Phase 1b trial was initiated in 2023 in endemic regions of Africa, and to date no serious safety signals have been reported. Methods In preparation for large-scale Phase 2 clinical trials and eventual vaccine deployment, the Sm-p80 antigen production process has been transferred to a manufacturing organization, Quratis Corporation in South Korea, which specializes in preparation of vaccines for large-scale European and African trials. The process of scaling from our current production level of ~2000 vaccine doses, to a process that will generate more than 100 million doses has required multiple improvement steps in the process including fermentation, downstream purification of the protein antigen, lyophilization, and fill and finish. Results In this study, we detail the large-scale production process of the SchistoShield® protein product by Quratis. In addition, an effort was made to analyze and compare the Quratis-made lot of Sm-p80, referred to as QTP-105, to the cGMP lot of Sm-p80 which is in use in human trials in the USA and Africa, referred to as Sm-p80 DP (made in USA). We show that QTP-105 demonstrates excellent potency, purity, identity, and endotoxin levels compared to our Phase 1 Sm-p80 DP and is suitable for use in Phase 2 studies and beyond.
Collapse
Affiliation(s)
- Jiho Kim
- PAI Life Sciences, Seattle, WA, United States
| | - Jenn Davis
- PAI Life Sciences, Seattle, WA, United States
| | - Jinhee Lee
- Quratis Corp, Cheongju, Republic of Korea
| | - Sang-Nae Cho
- Quratis Corp, Cheongju, Republic of Korea
- Department of Microbiology, Yonsei University, Seoul, Republic of Korea
| | | | | | | | - Joohee Son
- Quratis Corp, Cheongju, Republic of Korea
| | | | - Dale Whittington
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, United States
| | - Afzal A. Siddiqui
- Department of Immunology & Molecular Microbiology, Center for Tropical Medicine & Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Darrick Carter
- PAI Life Sciences, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Sean A. Gray
- PAI Life Sciences, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
| |
Collapse
|
5
|
Ndiour CN, Senghor B, Thiam O, Niang S, Wotodjo AN, Faye BT, Ndiaye NA, Sow O, Sylla K, Ndiaye M, Gaye O, Faye B, Sokhna C, Doucouré S, Sow D. Prevalence and associated factors of schistosomiasis among pregnant women in northern Senegal. BMC Infect Dis 2024; 24:682. [PMID: 38982383 PMCID: PMC11232235 DOI: 10.1186/s12879-024-09443-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 05/28/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Schistosomiasis remains a public health concern worldwide. It is responsible for more than 240 million cases in 78 countries, 40 million of whom are women of childbearing age. In the Senegal River basin, both Schistosoma haematobium and Schistosoma mansoni are very prevalent in school-age children. However, there is a lack of information on the burden of schistosomiasis in pregnant women, which can cause complications in the pregnancy outcome. This study aimed to determine the prevalence and associated factors of schistosomiasis in pregnant women. METHODS We conducted a prospective cross-sectional study of pregnant women attending antenatal clinics at the health center of the Senegalese Sugar Company and at the hospital of Richard Toll between August and December 2021. The urine and stool samples collected were examined using microscopy techniques and quantitative polymerase chain reaction (qPCR) to detect the presence of S. haematobium and S. mansoni. The urines were previously tested using urine reagent strips to detect hematuria and proteinuria. Socio-demographical, clinical, and diagnostically data were recorded by the midwife and the gynaecologist. The data were analyzed using a logistic regression model. RESULTS Among the 298 women examined for the infection by microscopic, 65 (21.81%) were infected with urogenital schistosomiasis, 10 (3.36%) with intestinal schistosomiasis, and 4 (1.34%) were co-infected with both types of schistosomiasis. Out of the 288 samples tested by qPCR, 146 (48.99%) were positive for S. haematobium, 49 (35.51%) for S. mansoni and 22 (15.94%) for both species (co-infection). Pregnant women having microscopic haematuria and proteinuria were significantly more infected (p < 0.05). CONCLUSION This study has revealed a high prevalence of schistosomiasis in pregnant women in Senegal. The qPCR allowed us to detect more cases compared to the microscopy. There is a need to conduct more studies to understand the real burden of the disease and to set up a surveillance system to prevent pregnancy-related complications.
Collapse
Affiliation(s)
- Coumba Nar Ndiour
- Service de Parasitologie-Mycologie, UFR Sciences de la Santé, Université Gaston Berger, de Saint-Louis, Senegal
| | - Bruno Senghor
- EMR MINES: Maladies Infectieuses, Négligées et Émergentes au Sud, Institut de Recherche pour le Développement, Campus International Institut de Recherche pour le Développement-Université-Cheikh Anta Diop of Hann, BP 1386, Dakar, Sénégal
| | - Ousmane Thiam
- Service de Gynécologie-Obstétrique, UFR Sciences de la Santé, Université Gaston Berger, de Saint-Louis, Senegal
| | - Souleymane Niang
- Centre de Santé, Compagnie Sucrière Sénégalaise, Richard Toll, Richard Toll, Senegal
| | - Amélé Nyedzie Wotodjo
- EMR MINES: Maladies Infectieuses, Négligées et Émergentes au Sud, Institut de Recherche pour le Développement, Campus International Institut de Recherche pour le Développement-Université-Cheikh Anta Diop of Hann, BP 1386, Dakar, Sénégal
| | - Babacar Thiendella Faye
- Service de Parasitologie-Mycologie, UFR Sciences de la Santé, Université Gaston Berger, de Saint-Louis, Senegal
| | - Ndeye Amy Ndiaye
- Direction de la Santé de la Mère et de l'Enfant, Ministère de la Santé et de l'Action Sociale, Dakar, Senegal
| | - Omar Sow
- Service de Parasitologie-Mycologie, UFR Sciences de la Santé, Université Gaston Berger, de Saint-Louis, Senegal
| | - Khadime Sylla
- Service de Parasitologie-Mycologie, FMPO, Université Cheikh Anta Diop, de Dakar, Senegal
| | - Magatte Ndiaye
- Service de Parasitologie-Mycologie, FMPO, Université Cheikh Anta Diop, de Dakar, Senegal
| | - Oumar Gaye
- Service de Parasitologie-Mycologie, FMPO, Université Cheikh Anta Diop, de Dakar, Senegal
| | - Babacar Faye
- Service de Parasitologie-Mycologie, FMPO, Université Cheikh Anta Diop, de Dakar, Senegal
| | - Cheikh Sokhna
- EMR MINES: Maladies Infectieuses, Négligées et Émergentes au Sud, Institut de Recherche pour le Développement, Campus International Institut de Recherche pour le Développement-Université-Cheikh Anta Diop of Hann, BP 1386, Dakar, Sénégal
| | - Souleymane Doucouré
- EMR MINES: Maladies Infectieuses, Négligées et Émergentes au Sud, Institut de Recherche pour le Développement, Campus International Institut de Recherche pour le Développement-Université-Cheikh Anta Diop of Hann, BP 1386, Dakar, Sénégal
| | - Doudou Sow
- Service de Parasitologie-Mycologie, UFR Sciences de la Santé, Université Gaston Berger, de Saint-Louis, Senegal.
- EMR MINES: Maladies Infectieuses, Négligées et Émergentes au Sud, Institut de Recherche pour le Développement, Campus International Institut de Recherche pour le Développement-Université-Cheikh Anta Diop of Hann, BP 1386, Dakar, Sénégal.
| |
Collapse
|
6
|
Almeida Júnior ASD, Freitas Viana Leal MM, Marques DSC, Silva ALD, Souza Bezerra RD, Siqueira de Souza YF, Mendonça Silveira ME, Santos FA, Alves LC, de Lima Aires A, Cruz Filho IJD, do Carmo Alves de Lima M. Therapeutic potential of hydantoin and thiohydantoin compounds against Schistosoma mansoni: An integrated in vitro, DNA, ultrastructural, and ADMET in silico approach. Mol Biochem Parasitol 2024; 260:111646. [PMID: 38950658 DOI: 10.1016/j.molbiopara.2024.111646] [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: 02/10/2024] [Revised: 06/01/2024] [Accepted: 06/18/2024] [Indexed: 07/03/2024]
Abstract
The study aimed to conduct in vitro biological assessments of hydantoin and thiohydantoin compounds against mature Schistosoma mansoni worms, evaluate their cytotoxic effects and predict their pharmacokinetic parameters using computational methods. The compounds showed low in vitro cytotoxicity and were not considered hemolytic. Antiparasitic activity against adult S. mansoni worms was tested with all compounds at concentrations ranging from 200 to 6.25 μM. Compounds SC01, SC02, and SC03 exhibited low activity. Compounds SC04, SC05, SC06 and SC07 caused 100 % mortality within 24 h of incubation at a concentration of 100 and 200 μM. Thiohydantoin SC04 exhibited the highest activity, resulting in 100 % mortality after 24 h of incubation at a concentration of 50 μM and IC50 of 28 µM. In the ultrastructural analysis (SEM), the compound SC04 (200 µM) induced integumentary changes, formation of integumentary blisters, and destruction of tubercles and spicules. Therefore, the SC04 compound shows promise as an antiparasitic against S. mansoni.
Collapse
Affiliation(s)
- Antônio Sérgio de Almeida Júnior
- Department of Antibiotics, Biosciences Center, Federal University of Pernambuco (UFPE), Avenida Prof. Moraes Rego, s/n, Cidade Universitária, Recife, PE 50740-600, Brazil
| | - Mayse Manuele Freitas Viana Leal
- Department of Antibiotics, Biosciences Center, Federal University of Pernambuco (UFPE), Avenida Prof. Moraes Rego, s/n, Cidade Universitária, Recife, PE 50740-600, Brazil
| | - Diego Santa Clara Marques
- Department of Antibiotics, Biosciences Center, Federal University of Pernambuco (UFPE), Avenida Prof. Moraes Rego, s/n, Cidade Universitária, Recife, PE 50740-600, Brazil.
| | - Anekécia Lauro da Silva
- Department of Medicine, Federal University of Vale do Rio São Francisco (UNIVASF), Avenida da Amizade, s/n, Sal Torrado, Paulo Afonso, BA 48605-780, Brazil
| | - Rafael de Souza Bezerra
- Department of Medicine, Federal University of Vale do Rio São Francisco (UNIVASF), Avenida da Amizade, s/n, Sal Torrado, Paulo Afonso, BA 48605-780, Brazil
| | - Yandra Flaviana Siqueira de Souza
- Department of Medicine, Federal University of Vale do Rio São Francisco (UNIVASF), Avenida da Amizade, s/n, Sal Torrado, Paulo Afonso, BA 48605-780, Brazil
| | - Maria Eduardade Mendonça Silveira
- Department of Antibiotics, Biosciences Center, Federal University of Pernambuco (UFPE), Avenida Prof. Moraes Rego, s/n, Cidade Universitária, Recife, PE 50740-600, Brazil
| | - Fábio Ab Santos
- Aggeu Magalhães Institute. Oswaldo Cruz Foundation (IAM-FIOCRUZ), Cidade Universitária, Recife, PE 50670-420, Brazil
| | - Luiz Carlos Alves
- Aggeu Magalhães Institute. Oswaldo Cruz Foundation (IAM-FIOCRUZ), Cidade Universitária, Recife, PE 50670-420, Brazil
| | - André de Lima Aires
- Department of Tropical Medicine, Health Sciences Center, Federal University of Pernambuco (UFPE), Avenida Prof. Moraes Rego, s/n, Cidade Universitária, Recife, PE 50740-600, Brazil
| | - Iranildo José da Cruz Filho
- Department of Antibiotics, Biosciences Center, Federal University of Pernambuco (UFPE), Avenida Prof. Moraes Rego, s/n, Cidade Universitária, Recife, PE 50740-600, Brazil
| | - Maria do Carmo Alves de Lima
- Department of Antibiotics, Biosciences Center, Federal University of Pernambuco (UFPE), Avenida Prof. Moraes Rego, s/n, Cidade Universitária, Recife, PE 50740-600, Brazil
| |
Collapse
|
7
|
Gopinath SCB. Response to Article "Development of a Magnetically-Assisted SERS Biosensor for Rapid Bacterial Detection" [Letter]. Int J Nanomedicine 2024; 19:5951-5952. [PMID: 38895148 PMCID: PMC11184219 DOI: 10.2147/ijn.s480177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 06/10/2024] [Indexed: 06/21/2024] Open
Affiliation(s)
- Subash C B Gopinath
- Faculty of Chemical Engineering & Technology, Universiti Malaysia Perlis (UniMAP), 02600 Arau, Perlis, Malaysia
- Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), 01000 Arau, Perlis, Malaysia
- Center for Global Health Research, Saveetha Medical College & Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai-602 105, Tamil Nadu, India
| |
Collapse
|
8
|
Lee CN, Hall BA, Sanford L, Molehin AJ. Molecular Characterization and Functional Analysis of a Schistosoma mansoni Serine Protease Inhibitor, Smserpin-p46. Microorganisms 2024; 12:1164. [PMID: 38930546 PMCID: PMC11205507 DOI: 10.3390/microorganisms12061164] [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: 05/10/2024] [Revised: 05/30/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Serine protease inhibitors are a superfamily of proteins that regulate various physiological processes including fibrinolysis, inflammation and immune responses. In parasite systems, serpins are believed to play important roles in parasite colonization, inhibition of host immune serine proteases and penetration of defensive barriers. However, serpins are less well characterized in schistosomes. In this study, a Schistosoma mansoni serpin (Smserpin-p46) containing a 1360 base pair open reading frame, was cloned, expressed and functionally characterized. Bioinformatics analysis revealed that Smserpin-p46 contains the key residues, structural domains and motifs characteristic of inhibitory serpins. Gene expression profiling demonstrated stage-specific expression of Smserpin-p46 with the highest expression in adult male worms. Recombinant Smserpin-p46 (rSmserpin-p46) inhibited both human neutrophil cathepsin G and elastase, key serine proteases involved in NETosis, a program for the formation of neutrophil extracellular traps. Using specific rabbit antiserum, Smserpin-p46 was detected in soluble worm antigen preparation and was localized to the adult worm tegument. Cumulatively, the expression of Smserpin-p46 on the parasite tegument and its ability to inhibit proteases involved in NETosis highlights the importance of this serpin in parasite-host interactions and encourages its further investigation as a candidate vaccine antigen for the control of schistosomiasis.
Collapse
Affiliation(s)
- Christine N. Lee
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA;
| | - Brooke Ashlyn Hall
- Department of Microbiology and Immunology, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA;
| | - Leah Sanford
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA;
| | - Adebayo J. Molehin
- Department of Microbiology and Immunology, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA;
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA;
| |
Collapse
|
9
|
Tallima H, Tadros MM, El Ridi R. Differential protective impact of peptide vaccine formulae targeting the lung- and liver-stage of challenge Schistosoma mansoni infection in mice. Acta Trop 2024; 254:107208. [PMID: 38621620 DOI: 10.1016/j.actatropica.2024.107208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 02/11/2024] [Accepted: 04/05/2024] [Indexed: 04/17/2024]
Abstract
The study aimed to elicit protective immune responses against murine schistosomiasis mansoni at the parasite lung- and liver stage. Two peptides showing amino acid sequence similarity to gut cysteine peptidases, which induce strong memory immune effectors in the liver, were combined with a peptide based on S. mansoni thioredoxin peroxidase (TPX), a prominent lung-stage schistosomula excretory-secretory product, and alum as adjuvant. Only one of the 2 cysteine peptidases-based peptides in a multiple antigenic peptide construct (MAP-3 and MAP-4) appeared to adjuvant protective immune responses induced by the TPX peptide in a MAP form. Production of TPX MAP-specific IgG1 serum antibodies, and increase in lung interleukin-1 (IL-1), uric acid, and reactive oxygen species (ROS) content were associated with significant (P < 0.05) 50 % reduction in recovery of lung-stage larvae. Increase in lung triglycerides and cholesterol levels appeared to provide the surviving worms with nutrients necessary for a stout double lipid bilayer barrier at the parasite-host interface. Surviving worms-released products elicited memory responses to the MAP-3 immunogen, including production of specific IgG1 antibodies and increase in liver IL-33 and ROS. Reduction in challenge worm burden recorded 45 days post infection did not exceed 48 % associated with no differences in parasite egg counts in the host liver and small intestine compared to unimmunized adjuvant control mice. Alum adjuvant assisted the second peptide, MAP-4, in production of IgG1, IgG2a, IgG2b and IgA specific antibodies and increase in liver ROS, but with no protective potential, raising doubt about the necessity of adjuvant addition. Accordingly, different vaccine formulas containing TPX MAP and 1, 2 or 3 cysteine peptidases-derived peptides with or without alum were used to immunize parallel groups of mice. Compared to unimmunized control mice, significant (P < 0.05 to < 0.005) 22 to 54 % reduction in worm burden was recorded in the different groups associated with insignificant changes in parasite egg output. The results together indicated that a schistosomiasis vaccine able to entirely prevent disease and halt its transmission still remains elusive.
Collapse
Affiliation(s)
- Hatem Tallima
- Zoology Department, Faculty of Science, Cairo University, Giza 12613, Egypt; Department of Chemistry, School of Sciences and Engineering, American University in Cairo, New Cairo 11835, Cairo, Egypt.
| | - Menerva M Tadros
- Department of Parasitology, Theodore Bilharz Research Institute, Warrak El-Hadar, Imbaba, Giza 12411, Egypt
| | - Rashika El Ridi
- Zoology Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| |
Collapse
|
10
|
Perera DJ, Koger-Pease C, Paulini K, Daoudi M, Ndao M. Beyond schistosomiasis: unraveling co-infections and altered immunity. Clin Microbiol Rev 2024; 37:e0009823. [PMID: 38319102 PMCID: PMC10938899 DOI: 10.1128/cmr.00098-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024] Open
Abstract
Schistosomiasis is a neglected tropical disease caused by the helminth Schistosoma spp. and has the second highest global impact of all parasites. Schistosoma are transmitted through contact with contaminated fresh water predominantly in Africa, Asia, the Middle East, and South America. Due to the widespread prevalence of Schistosoma, co-infection with other infectious agents is common but often poorly described. Herein, we review recent literature describing the impact of Schistosoma co-infection between species and Schistosoma co-infection with blood-borne protozoa, soil-transmitted helminths, various intestinal protozoa, Mycobacterium, Salmonella, various urinary tract infection-causing agents, and viral pathogens. In each case, disease severity and, of particular interest, the immune landscape, are altered as a consequence of co-infection. Understanding the impact of schistosomiasis co-infections will be important when considering treatment strategies and vaccine development moving forward.
Collapse
Affiliation(s)
- Dilhan J. Perera
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Cal Koger-Pease
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Kayla Paulini
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
| | - Mohamed Daoudi
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
| | - Momar Ndao
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
- National Reference Centre for Parasitology, Research Institute of the McGill University Health Centre, Montreal, Canada
| |
Collapse
|
11
|
Cheng S, Tu Z, Zheng S, Khan A, Yang P, Shen H, Gu B. Development of a Magnetically-Assisted SERS Biosensor for Rapid Bacterial Detection. Int J Nanomedicine 2024; 19:389-401. [PMID: 38250194 PMCID: PMC10799629 DOI: 10.2147/ijn.s433316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/31/2023] [Indexed: 01/23/2024] Open
Abstract
Introduction Ultrasensitive bacterial detection methods are crucial to ensuring accurate diagnosis and effective clinical monitoring, given the significant threat bacterial infections pose to human health. The aim of this study is to develop a biosensor with capabilities for broad-spectrum bacterial detection, rapid processing, and cost-effectiveness. Methods A magnetically-assisted SERS biosensor was designed, employing wheat germ agglutinin (WGA) for broad-spectrum recognition and antibodies for specific capture. Gold nanostars (AuNSs) were sequentially modified with the Raman reporter molecules and WGA, creating a versatile SERS tag with high affinity for a diverse range of bacteria. Staphylococcus aureus (S. aureus) and Pseudomonas aeruginosa (P. aeruginosa) antibody-modified Fe3O4 magnetic gold nanoparticles (MGNPs) served as the capture probes. Target bacteria were captured by MGNPs and combined with SERS tags, forming a "sandwich" composite structure for bacterial detection. Results AuNSs, with a core size of 65 nm, exhibited excellent storage stability (RSD=5.6%) and demonstrated superior SERS enhancement compared to colloidal gold nanoparticles. Efficient binding of S. aureus and P. aeruginosa to MGNPs resulted in capture efficiencies of 89.13% and 85.31%, respectively. Under optimized conditions, the developed assay achieved a limit of detection (LOD) of 7 CFU/mL for S. aureus and 5 CFU/mL for P. aeruginosa. The bacterial concentration (10-106 CFU/mL) showed a strong linear correlation with the SERS intensity at 1331 cm-1. Additionally, high recoveries (84.8% - 118.0%) and low RSD (6.21% - 11.42%) were observed in spiked human urine samples. Conclusion This study introduces a simple and innovative magnetically-assisted SERS biosensor for the sensitive and quantitative detection of S. aureus or P. aeruginosa, utilizing WGA and antibodies. The developed biosensor enhances the capabilities of the "sandwich" type SERS biosensor, offering a novel and effective platform for accurate and timely clinical diagnosis of bacterial infections.
Collapse
Affiliation(s)
- Siyun Cheng
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
- Medical Technology School of Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Zhijie Tu
- Medical Technology School of Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Shuai Zheng
- Department of Clinical Laboratory Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People’s Republic of China
| | - Adeel Khan
- Department of Biotechnology, University of Science and Technology, Bannu, KP, Pakistan
| | - Ping Yang
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Han Shen
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Bing Gu
- Medical Technology School of Xuzhou Medical University, Xuzhou, People’s Republic of China
- Department of Clinical Laboratory Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People’s Republic of China
| |
Collapse
|
12
|
Woellner-Santos D, Tahira AC, Malvezzi JVM, Mesel V, Morales-Vicente DA, Trentini MM, Marques-Neto LM, Matos IA, Kanno AI, Pereira ASA, Teixeira AAR, Giordano RJ, Leite LCC, Pereira CAB, DeMarco R, Amaral MS, Verjovski-Almeida S. Schistosoma mansoni vaccine candidates identified by unbiased phage display screening in self-cured rhesus macaques. NPJ Vaccines 2024; 9:5. [PMID: 38177171 PMCID: PMC10767053 DOI: 10.1038/s41541-023-00803-x] [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: 08/03/2023] [Accepted: 11/30/2023] [Indexed: 01/06/2024] Open
Abstract
Schistosomiasis, a challenging neglected tropical disease, affects millions of people worldwide. Developing a prophylactic vaccine against Schistosoma mansoni has been hindered by the parasite's biological complexity. In this study, we utilized the innovative phage-display immunoprecipitation followed by a sequencing approach (PhIP-Seq) to screen the immune response of 10 infected rhesus macaques during self-cure and challenge-resistant phases, identifying vaccine candidates. Our high-throughput S. mansoni synthetic DNA phage-display library encoded 99.6% of 119,747 58-mer peptides, providing comprehensive coverage of the parasite's proteome. Library screening with rhesus macaques' antibodies, from the early phase of establishment of parasite infection, identified significantly enriched epitopes of parasite extracellular proteins known to be expressed in the digestive tract, shifting towards intracellular proteins during the late phase of parasite clearance. Immunization of mice with a selected pool of PhIP-Seq-enriched phage-displayed peptides from MEG proteins, cathepsins B, and asparaginyl endopeptidase significantly reduced worm burden in a vaccination assay. These findings enhance our understanding of parasite-host immune responses and provide promising prospects for developing an effective schistosomiasis vaccine.
Collapse
Affiliation(s)
- Daisy Woellner-Santos
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, SP, Brazil
- Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Ana C Tahira
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, SP, Brazil
| | - João V M Malvezzi
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, SP, Brazil
- Instituto de Matemática e Estatística, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Vinicius Mesel
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, SP, Brazil
| | - David A Morales-Vicente
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, SP, Brazil
- Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Monalisa M Trentini
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, SP, Brazil
| | - Lázaro M Marques-Neto
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, SP, Brazil
| | - Isaac A Matos
- Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Alex I Kanno
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, SP, Brazil
| | - Adriana S A Pereira
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, SP, Brazil
- Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - André A R Teixeira
- Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
- Institute for Protein Innovation, Boston, MA, USA
| | | | - Luciana C C Leite
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, SP, Brazil
| | - Carlos A B Pereira
- Instituto de Matemática e Estatística, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Ricardo DeMarco
- Instituto de Física de São Carlos, Universidade de São Paulo, São Carlos, SP, Brazil
| | - Murilo S Amaral
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, SP, Brazil
| | - Sergio Verjovski-Almeida
- Laboratório de Ciclo Celular, Instituto Butantan, São Paulo, SP, Brazil.
- Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
13
|
LoVerde PT. Schistosomiasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1454:75-105. [PMID: 39008264 DOI: 10.1007/978-3-031-60121-7_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Schistosomiasis is a major cause of morbidity in the world and almost 800 million people worldwide are at risk for schistosomiasis; it is second only to malaria as a major infectious disease. Globally, it is estimated that the disease affects more than 250 million people in 78 countries of the world and is responsible for some 280,000-500,000 deaths each year. The three major schistosomes infecting humans are Schistosoma mansoni, S. japonicum, and S. haematobium. This chapter covers a wide range of aspects of schistosomiasis, including basic biology of the parasites, epidemiology, immunopathology, treatment, control, vaccines, and genomics/proteomics. In this chapter, the reader will understand the significant toll this disease takes in terms of mortality and morbidity. A description of the various life stages of schistosomes is presented, which will be informative for both those unfamiliar with the disease and experienced scientists. Clinical and public health aspects are addressed that cover acute and chronic disease, diagnosis, current treatment regimens and alternative drugs, and schistosomiasis control programs. A brief overview of genomics and proteomics is included that details recent advances in the field that will help scientists investigate the molecular biology of schistosomes. The reader will take away an appreciation for general aspects of schistosomiasis and the current research advances.
Collapse
Affiliation(s)
- Philip T LoVerde
- Department of Biochemistry and Structural Biology, University of Texas Health, San Antonio, TX, USA.
| |
Collapse
|
14
|
Tadele T, Astatkie A, Tadesse BT, Makonnen E, Aklillu E, Abay SM. Efficacy and safety of praziquantel treatment against Schistosoma mansoni infection among pre-school age children in southern Ethiopia. Trop Med Health 2023; 51:72. [PMID: 38124206 PMCID: PMC10731898 DOI: 10.1186/s41182-023-00562-4] [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: 09/24/2023] [Accepted: 12/09/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Preventive chemotherapy with a single dose of praziquantel given to an all-at-risk population through mass drug administration is the cornerstone intervention to control and eliminate schistosomiasis as a public health problem. This intervention mainly targets school age children, and pre-school age children (pre-SAC) are excluded from receiving preventive chemotherapy, partly due to scarcity of data on praziquantel treatment outcomes. METHODS We conducted active efficacy and safety surveillance of praziquantel treatment among 240 Schistosoma mansoni-infected pre-SAC who received a single dose of praziquantel (40 mg/kg) in southern Ethiopia. The study outcomes were egg reduction rates (ERR) and cure rates (CRs) four weeks after treatment using the Kato-Katz technique and treatment-associated adverse events (AEs) that occurred within 8 days post-treatment. RESULTS The overall ERR was 93.3% (WHO reference threshold ≥ 90%), while the CR was 85.2% (95% CI = 80.0-89.5%). Baseline S. mansoni infection intensity was significantly associated with CRs, 100% among light infected than moderate (83.4%) or heavy (29.4%) infected children. An increase of 100 in baseline S. mansoni egg count per gram of stool resulted in a 26% (95% CI: 17%, 34%) reduction in the odds of cure. The incidence of experiencing at least one type of AE was 23.1% (95% CI: 18.0%, 29.0%). Stomachache, diarrhea, and nausea were the most common AEs. AEs were mild-to-moderate grade and transient. Pre-treatment moderate (ARR = 3.2, 95% CI: 1.69, 6.14) or heavy infection intensity (ARR = 6.5, 95% CI: 3.62, 11.52) was a significant predictor of AEs (p < 0.001). Sex, age, or soil-transmitted helminth coinfections were not significant predictors of CR or AEs. CONCLUSIONS Single-dose praziquantel is tolerable and effective against S. mansoni infection among pre-SAC, and associated AEs are mostly mild-to-moderate and transient. However, the reduced CR in heavily infected and AEs in one-fourth of S. mansoni-infected pre-SAC underscores the need for safety and efficacy monitoring, especially in moderate-to-high infection settings. Integrating pre-SACs in the national deworming programs is recommended to accelerate the elimination of schistosomiasis as a public health problem.
Collapse
Affiliation(s)
- Tafese Tadele
- School of Public Health, College of Medicine and Health Sciences, Hawassa University, P.O. Box 1560, Hawassa, Ethiopia
| | - Ayalew Astatkie
- School of Public Health, College of Medicine and Health Sciences, Hawassa University, P.O. Box 1560, Hawassa, Ethiopia
| | - Birkneh Tilahun Tadesse
- Department of Pediatrics, College of Medicine and Health Sciences, Hawassa University, P.O. Box 1560, Hawassa, Ethiopia
| | - Eyasu Makonnen
- Center for Innovative Drug Development and Therapeutic Trials for Africa, College of Health Sciences, Addis Ababa University, P.O. Box 9086, Addis Ababa, Ethiopia
- Department of Pharmacology and Clinical Pharmacy, College of Health Sciences, Addis Ababa University, P.O. Box 9086, Addis Ababa, Ethiopia
| | - Eleni Aklillu
- Department of Global Public Health, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | - Solomon Mequanente Abay
- Department of Pharmacology and Clinical Pharmacy, College of Health Sciences, Addis Ababa University, P.O. Box 9086, Addis Ababa, Ethiopia
| |
Collapse
|
15
|
Li H, Chen Y, Zhu Y, Feng Y, Qian Y, Ye X, Xu J, Yang H, Yu J, Chen J, Chen K. Exploring the immune interactions between Oncomelania hupensis and Schistosoma japonicum, with a cross-comparison of immunological research progress in other intermediate host snails. Parasit Vectors 2023; 16:453. [PMID: 38093363 PMCID: PMC10717515 DOI: 10.1186/s13071-023-06011-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 10/11/2023] [Indexed: 12/17/2023] Open
Abstract
Schistosomiasis, the second largest parasitic disease in the world after malaria, poses a significant threat to human health and causes public health issues. The disease primarily affects populations in economically underdeveloped tropical regions, earning it the title of "neglected tropical disease". Schistosomiasis is difficult to eradicate globally if medication alone is used. One of the essential elements of thorough schistosomiasis prevention and control is the management and disruption of the life cycle of intermediate host snails. The key approach to controlling the transmission of schistosomiasis is to control the intermediate hosts of the schistosome to disrupt its life cycle. We believe that approaching it from the perspective of the intermediate host's immunity could be an environmentally friendly and potentially effective method. Currently, globally significant intermediate host snails for schistosomes include Oncomelania hupensis, Biomphalaria glabrata, and Bulinus truncatus. The immune interaction research between B. glabrata and Schistosoma mansoni has a history of several decades, and the complete genome sequencing of both B. glabrata and B. truncatus has been accomplished. We have summarized the immune-related factors and research progress primarily studied in B. glabrata and B. truncatus and compared them with several humoral immune factors that O. hupensis research focuses on: macrophage migration inhibitory factor (MIF), Toll-like receptors (TLRs), and thioredoxin (Trx). We believe that continued exploration of the immune interactions between O. hupensis and Schistosoma japonicum is valuable. This comparative analysis can provide some direction and clues for further in-depth research. Comparative immunological studies between them not only expand our understanding of the immune defense responses of snails that act as intermediaries for schistosomes but also facilitate the development of more comprehensive and integrated strategies for schistosomiasis prevention and control. Furthermore, it offers an excellent opportunity to study the immune system of gastropods and their co-evolution with pathogenic organisms.
Collapse
Affiliation(s)
- Hongyu Li
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China.
- Ocean College, Beibu Gulf University, Qinzhou, China.
| | - Yihan Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yunhuan Zhu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yilu Feng
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yuncheng Qian
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Xiaoyu Ye
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Jiatong Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Hanyu Yang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Jiawei Yu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Jingyu Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Keda Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China.
| |
Collapse
|
16
|
Jain S, Rana M, Choubey P, Kumar S. Schistosoma japonicum Associated Colorectal Cancer and Its Management. Acta Parasitol 2023; 68:723-734. [PMID: 37594685 DOI: 10.1007/s11686-023-00707-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 07/31/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND An association between Schistosoma japonicum and colorectal cancer in humans has been known since a long time; however, this association remains understudied and lacks comprehensive experimentation support. OBJECTIVE Various epidemiological and pathological studies have established the role of chronic inflammation as a major factor behind the induction of colorectal cancer. The aim of this review is to present the current knowledge on the association of Schistosoma japonicum with colorectal cancer. RESULT Mechanisms which lead to induction and progression of colorectal cancer are highlighted along with diagnosis and treatment for the same. Further, various methodologies, including mass drug administration, use of new drugs and vaccines, role of apoptosis, and histone-modifying enzymes, have been described which can either prevent the schistosomal infection itself or can check it from reaching an advanced stage. CONCLUSIONS Epidemiological, clinical, pathological and surgical studies suggest that Schistosoma japonicum is responsible for induction of colorectal cancer. However, thorough clinical studies are required to support and globally accept this notion. Further, methodologies highlighted in this work can be employed in order to take care of schistosomal infection or address the cancer induction and progression.
Collapse
Affiliation(s)
- Sidhant Jain
- Institute for Globally Distributed Open Research and Education (IGDORE), Rewari, Haryana, India.
| | - Meenakshi Rana
- Dyal Singh College, University of Delhi, Lodhi Road, Pragati Vihaar, New Delhi, 110003, India
| | - Pooja Choubey
- Department of Zoology, Gate No. 3, Chaatra Marg, University of Delhi, North Campus, Delhi, 110007, India
| | - Sahil Kumar
- Department of Pharmacology, ESIC Dental College and Hospital, Rohini, Sector-15, Delhi, 110089, India
| |
Collapse
|
17
|
Zhao S, Zhang Q, Wang X, Li W, Juma S, Berquist R, Zhang J, Yang K. Development and performance of recombinase-aided amplification (RAA) assay for detecting Schistosoma haematobium DNA in urine samples. Heliyon 2023; 9:e23031. [PMID: 38144328 PMCID: PMC10746445 DOI: 10.1016/j.heliyon.2023.e23031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 11/17/2023] [Accepted: 11/24/2023] [Indexed: 12/26/2023] Open
Abstract
Rapid diagnosis of urogenital schistosomiasis caused by Schistosoma haematobium requires an accurate and timely assay, especially for low-intensity S. haematobium infection cases and in non-endemic areas. The mitochondrial cytochrome c oxidase 1 (cox1) gene fragment of S. haematobium was selected as detection target as this short fragment, which can be rapidly sequenced and yet possess good diagnostic resolution. A pair of primers and a fluorescent probe were designed according to the principle of recombinase-aided amplification (RAA), which was subsequently optimized and applied as an S. haematobium-specific RAA assay. Its diagnostic performance was validated for sensitivity and specificity in comparison to microscopy-based egg counting after urine filtration. The RAA assay could detect as little as 10 copies/μL of S. haematobium recombinant plasmid, and no cross-reactions were observed with S. mansoni, S. japonicum, Ancylostoma duodenale, Clonorchis sinensis, Echinococcus granulosus, or Ascaris lumbricoides. This test can be conducted at 39 °C and the whole RAA reaction can be completed within 20 min. The validation of the RAA assay showed that it had 100 % consistency with urine-egg microscopy, as it does not require an elaborate reading tool, is simple to use, and should be useful for field diagnostics and point-of-care applications.
Collapse
Affiliation(s)
- Song Zhao
- Jiangsu Institute of Parasitic Diseases, Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health, Jiangsu Provincial Key Laboratory on the Molecular Biology of Parasites, Wuxi, Jiangsu, China
- Jiangnan University, Wuxi, Jiangsu, China
| | - Qiaoqiao Zhang
- Jiangsu Institute of Parasitic Diseases, Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health, Jiangsu Provincial Key Laboratory on the Molecular Biology of Parasites, Wuxi, Jiangsu, China
- Department of Clinical Laboratory, The 904th Hospital of Joint Logistic Support Force of PLA, Wuxi, Jiangsu, 214044, China
| | - Xinyao Wang
- Jiangsu Institute of Parasitic Diseases, Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health, Jiangsu Provincial Key Laboratory on the Molecular Biology of Parasites, Wuxi, Jiangsu, China
| | - Wei Li
- Jiangsu Institute of Parasitic Diseases, Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health, Jiangsu Provincial Key Laboratory on the Molecular Biology of Parasites, Wuxi, Jiangsu, China
| | - Saleh Juma
- Ministry of Health of Zanzibar, P.O. Box 236, Zanzibar, United Republic of Tanzania
| | - Robert Berquist
- Ingerod, Brastad, Sweden (formerly with the UNICEF/UNDP/World Bank/WHO Special Programme for Research and Training in Tropical Diseases, World Health Organization, Geneva, Switzerland
| | - Jianfeng Zhang
- Jiangsu Institute of Parasitic Diseases, Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health, Jiangsu Provincial Key Laboratory on the Molecular Biology of Parasites, Wuxi, Jiangsu, China
| | - Kun Yang
- Jiangsu Institute of Parasitic Diseases, Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health, Jiangsu Provincial Key Laboratory on the Molecular Biology of Parasites, Wuxi, Jiangsu, China
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
- Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
18
|
Zhu J, Zhang L, Xue Z, Li Z, Wang C, Chen F, Li Y, Dai Y, Zhou Y, Zhou S, Chen X, Okumura-Noji K, Lu R, Yokoyama S, Su C. Vaccination against the HDL receptor of S. japonicum inhibits egg embryonation and prevents fatal hepatic complication in rabbit model. PLoS Negl Trop Dis 2023; 17:e0011749. [PMID: 38019787 PMCID: PMC10686426 DOI: 10.1371/journal.pntd.0011749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Schistosomiasis is one of the most important neglected tropical infectious diseases to overcome and the primary cause of its pathogenesis is ectopic maturation of the parasite eggs. Uptake of cholesteryl ester from the host high-density lipoprotein (HDL) is a key in this process in Schistosoma japonicum and CD36-related protein (CD36RP) has been identified as the receptor for this reaction. Antibody against the extracellular domain of CD36RP (Ex160) efficiently blocked the HDL cholesteryl ester uptake and the egg embryonation in vitro. However, whether Ex160 immunization could efficiently raise proper antibody responses to sufficiently block HDL cholesteryl ester uptake and the egg embryonation to protect host in vivo is very interesting but unknown. METHODOLOGY/PRINCIPAL FINDINGS In this study, rabbits were immunized with the recombinant Ex160 peptide (rEx160) to evaluate its anti-pathogenic vaccine potential. Immunization with rEx160 induced consistent anti-Ex160 IgG antibody and significant reduction in development of the liver granulomatosis lesions associated with suppressed intrahepatic maturation of the schistosome eggs. The immunization with rEx160 rescued reduction of serum HDL by the infection without changing its size distribution, being consistent with interference of the HDL lipid uptake by the parasites or their eggs by antibody against Ex160 in in vitro culture. CONCLUSIONS/SIGNIFICANCE The results demonstrated that vaccination strategy against nutritional supply pathway of the parasite is effective for reducing its pathogenesis.
Collapse
Affiliation(s)
- Jifeng Zhu
- National Vaccine Innovation Platform, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Pathogen Biology and Immunology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lina Zhang
- Department of Blood Transfusion, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
- Department of Blood Transfusion of Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zechao Xue
- National Vaccine Innovation Platform, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Pathogen Biology and Immunology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zilüe Li
- National Vaccine Innovation Platform, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Pathogen Biology and Immunology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chun Wang
- National Vaccine Innovation Platform, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Pathogen Biology and Immunology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fanyan Chen
- National Vaccine Innovation Platform, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Pathogen Biology and Immunology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yalin Li
- National Vaccine Innovation Platform, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Pathogen Biology and Immunology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yang Dai
- Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu, China
| | - Yonghua Zhou
- Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu, China
| | - Sha Zhou
- National Vaccine Innovation Platform, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Pathogen Biology and Immunology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaojun Chen
- National Vaccine Innovation Platform, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Pathogen Biology and Immunology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | | | - Rui Lu
- Food and Nutritional Sciences, Chubu University, Kasugai, Japan
| | - Shinji Yokoyama
- Food and Nutritional Sciences, Chubu University, Kasugai, Japan
| | - Chuan Su
- National Vaccine Innovation Platform, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Pathogen Biology and Immunology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
19
|
Díaz AV, Walker M, Webster JP. Reaching the World Health Organization elimination targets for schistosomiasis: the importance of a One Health perspective. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220274. [PMID: 37598697 PMCID: PMC10440173 DOI: 10.1098/rstb.2022.0274] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 07/19/2023] [Indexed: 08/22/2023] Open
Abstract
The past three years has seen the launch of a new World Health Organization (WHO) neglected tropical diseases (NTDs) roadmap, together with revised control and elimination guidelines. Across all, there is now a clear emphasis on the need to incorporate a One Health approach, recognizing the critical links between human and animal health and the environment. Schistosomiasis, caused by Schistosoma spp. trematodes, is a NTD of global medical and veterinary importance, with over 220 million people and untold millions of livestock currently infected. Its burden remains extremely high in certain regions, particularly within sub-Saharan Africa, despite over two decades of mass preventive chemotherapy (mass drug administration), predominantly to school-aged children. In Africa, in contrast to Asia, any zoonotic component of schistosomiasis transmission and its implications for disease control has, until recently, been largely ignored. Here, we review recent epidemiological, clinical, molecular, and modelling work across both Asia and Africa. We outline the evolutionary history and transmission dynamics of Schistosoma species, and emphasize the emerging risk raised by both wildlife reservoirs and viable hybridization between human and animal schistosomes. To achieve the 2030 WHO roadmap elimination targets, a truly multi-disciplinary One Health perspective must be implemented. This article is part of the theme issue 'Challenges and opportunities in the fight against neglected tropical diseases: a decade from the London Declaration on NTDs'.
Collapse
Affiliation(s)
- Adriana V. Díaz
- Department of Pathobiology and Population Sciences, Royal Veterinary College, Hatfield AL9 7TA, UK
| | - Martin Walker
- Department of Pathobiology and Population Sciences, Royal Veterinary College, Hatfield AL9 7TA, UK
- Department of Infectious Disease Epidemiology, London Centre for Neglected Tropical Disease Research, Faculty of Medicine, Imperial College, London W2 1PG, UK
| | - Joanne P. Webster
- Department of Pathobiology and Population Sciences, Royal Veterinary College, Hatfield AL9 7TA, UK
- Department of Infectious Disease Epidemiology, London Centre for Neglected Tropical Disease Research, Faculty of Medicine, Imperial College, London W2 1PG, UK
| |
Collapse
|
20
|
Zhu P, Wu K, Zhang C, Batool SS, Li A, Yu Z, Huang J. Advances in new target molecules against schistosomiasis: A comprehensive discussion of physiological structure and nutrient intake. PLoS Pathog 2023; 19:e1011498. [PMID: 37498810 PMCID: PMC10374103 DOI: 10.1371/journal.ppat.1011498] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023] Open
Abstract
Schistosomiasis, a severe parasitic disease, is primarily caused by Schistosoma mansoni, Schistosoma japonicum, or Schistosoma haematobium. Currently, praziquantel is the only recommended drug for human schistosome infection. However, the lack of efficacy of praziquantel against juvenile worms and concerns about the emergence of drug resistance are driving forces behind the research for an alternative medication. Schistosomes are obligatory parasites that survive on nutrients obtained from their host. The ability of nutrient uptake depends on their physiological structure. In short, the formation and maintenance of the structure and nutrient supply are mutually reinforcing and interdependent. In this review, we focus on the structural features of the tegument, esophagus, and intestine of schistosomes and their roles in nutrient acquisition. Moreover, we introduce the significance and modes of glucose, lipids, proteins, and amino acids intake in schistosomes. We linked the schistosome structure and nutrient supply, introduced the currently emerging targets, and analyzed the current bottlenecks in the research and development of drugs and vaccines, in the hope of providing new strategies for the prevention and control of schistosomiasis.
Collapse
Affiliation(s)
- Peng Zhu
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
| | - Kaijuan Wu
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China
| | - Chaobin Zhang
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
| | - Syeda Sundas Batool
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| | - Anqiao Li
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
| | - Zheng Yu
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| | - Jing Huang
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| |
Collapse
|
21
|
Amer AS, Othman AA, Dawood LM, El-Nouby KA, Gobert GN, Abou Rayia DM. The interaction of Schistosoma mansoni infection with diabetes mellitus and obesity in mice. Sci Rep 2023; 13:9417. [PMID: 37296126 PMCID: PMC10256771 DOI: 10.1038/s41598-023-36112-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Human schistosomiasis is one of the most prevalent parasitic diseases worldwide. Various host factors can affect the host-parasite interactions. Therefore, the aim of the present work was to determine the parasitological, histopathological, biochemical, and immunological status of Schistosoma mansoni-infected hosts with metabolic disorders to identify the underlying possible mechanisms of these comorbidities. The study animals were divided into four groups. Group I represented the control groups, namely, the normal control group, the S. mansoni-infected control group, and the noninfected type 1 diabetes (T1DM), type 2 diabetes (T2DM), and obesity groups. The mice of the other three groups underwent induction of T1DM (Group II), T2DM (Group III) and obesity (Group IV) before being infected with S. mansoni. All mice were subjected to body weight measurement, blood glucose and insulin assessment, parasitological evaluation of adult worm count, tissue egg count and intestinal oogram. Histopathological and immunohistochemical study using anti-glial fibrillary acidic protein (GFAP) in hepatic stellate cells (HSCs) and image analysis of Masson's trichrome-stained liver sections using ImageJ (Fiji) software were carried out. Additionally, immunological analysis of tumour necrosis factor (TNF) beta, interleukin-5 (IL-5), IL-10, Forkhead box P3 (FOXP3) and pentraxin 3 (PTX3) levels besides biochemical study of total lipid profile were evaluated. The present study revealed a significant increase in the adult worm count and tissue egg output in the obesity group compared to the infected control group. The oogram of counted eggs showed prevalence of immature eggs in T1DM group, while T2DM and obese groups showed prevalence of mature eggs. The fibrosis area percentage showed significant increase in T2DM and obese groups while it was decreased in T1DM group in comparison to infected control group. Our data also showed significant increase in the levels of TNF-β, IL-5, PTX3 in T1DM, T2DM and obesity groups in comparison to infected control group, whilst the levels of FOXP3 and IL-10 were increased in the infected groups in comparison to their noninfected controls. Moreover, infected T1DM, T2DM and obesity groups showed higher blood glucose and lipid profile in comparison to the infected control group. However, these parameters were improved in comparison to their noninfected controls. In sum, induction of T2DM and obesity increased tissue egg counts, mature egg percentage, and fibrosis density, while schistosome infection induced changes in the lipid profile and blood glucose levels in infected diabetic and obese groups and impacted favorably insulin levels in obese mice. By better understanding the complexities of host-parasite interactions, efforts to reduce the burden of these debilitating diseases can be improved.
Collapse
Affiliation(s)
- Alaa S Amer
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, 31527, Egypt.
| | - Ahmad A Othman
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, 31527, Egypt
| | - Lamees M Dawood
- Biochemistry Department, Faculty of Medicine, Tanta University, Tanta, 31527, Egypt
| | - Kholoud A El-Nouby
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, 31527, Egypt
| | - Geoffrey N Gobert
- School of Biological Science, Institute for Global Food Security, Queen's University Belfast, Belfast, BT9 5DL, UK
| | - Dina M Abou Rayia
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
22
|
Alzain AA, Mukhtar RM, Abdelmoniem N, Elbadwi FA, Hussien A, Garelnabi EAE, Osman W, Sherif AE, Khedr AIM, Ghazawi KF, Samman WA, Ibrahim SRM, Mohamed GA, Ashour A. Computational Insights into Natural Antischistosomal Metabolites as SmHDAC8 Inhibitors: Molecular Docking, ADMET Profiling, and Molecular Dynamics Simulation. Metabolites 2023; 13:metabo13050658. [PMID: 37233699 DOI: 10.3390/metabo13050658] [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: 04/16/2023] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 05/27/2023] Open
Abstract
Schistosomiasis is a neglected tropical disease with a significant socioeconomic impact. It is caused by several species of blood trematodes from the genus Schistosoma, with S. mansoni being the most prevalent. Praziquantel (PZQ) is the only drug available for treatment, but it is vulnerable to drug resistance and ineffective in the juvenile stage. Therefore, identifying new treatments is crucial. SmHDAC8 is a promising therapeutic target, and a new allosteric site was discovered, providing the opportunity for the identification of a new class of inhibitors. In this study, molecular docking was used to screen 13,257 phytochemicals from 80 Saudi medicinal plants for inhibitory activity on the SmHDAC8 allosteric site. Nine compounds with better docking scores than the reference were identified, and four of them (LTS0233470, LTS0020703, LTS0033093, and LTS0028823) exhibited promising results in ADMET analysis and molecular dynamics simulation. These compounds should be further explored experimentally as potential allosteric inhibitors of SmHDAC8.
Collapse
Affiliation(s)
- Abdulrahim A Alzain
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Wad Madani 21111, Sudan
| | - Rua M Mukhtar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Wad Madani 21111, Sudan
| | - Nihal Abdelmoniem
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Wad Madani 21111, Sudan
| | - Fatima A Elbadwi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Gezira, Wad Madani 21111, Sudan
| | - Amira Hussien
- Department of Pharmacology, Faculty of Pharmacy, University of Gezira, Wad Madani 21111, Sudan
| | - Elrashied A E Garelnabi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Khartoum, Al-Qasr Ave, Khartoum 11111, Sudan
| | - Wadah Osman
- Department of Pharmacognosy, Faculty of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, University of Khartoum, Al-Qasr Ave, Khartoum 11111, Sudan
| | - Asmaa E Sherif
- Department of Pharmacognosy, Faculty of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Amgad I M Khedr
- Department of Pharmacognosy, Faculty of Pharmacy, Port Said University, Port Said 42526, Egypt
| | - Kholoud F Ghazawi
- Clinical Pharmacy Department, College of Pharmacy, Umm Al-Qura University, Makkah 24382, Saudi Arabia
| | - Waad A Samman
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Al-Madinah Al-Munawwarah 30078, Saudi Arabia
| | - Sabrin R M Ibrahim
- Preparatory Year Program, Department of Chemistry, Batterjee Medical College, Jeddah 21442, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Gamal A Mohamed
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ahmed Ashour
- Department of Pharmacognosy, Faculty of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
23
|
Siddiqui AJ, Bhardwaj J, Saxena J, Jahan S, Snoussi M, Bardakci F, Badraoui R, Adnan M. A Critical Review on Human Malaria and Schistosomiasis Vaccines: Current State, Recent Advancements, and Developments. Vaccines (Basel) 2023; 11:vaccines11040792. [PMID: 37112704 PMCID: PMC10146311 DOI: 10.3390/vaccines11040792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/07/2023] Open
Abstract
Malaria and schistosomiasis are two major parasitic diseases that remain leading causes of morbidity and mortality worldwide. Co-infections of these two parasites are common in the tropics, where both diseases are endemic. The clinical consequences of schistosomiasis and malaria are determined by a variety of host, parasitic, and environmental variables. Chronic schistosomiasis causes malnutrition and cognitive impairments in children, while malaria can cause fatal acute infections. There are effective drugs available to treat malaria and schistosomiasis. However, the occurrence of allelic polymorphisms and the rapid selection of parasites with genetic mutations can confer reduced susceptibility and lead to the emergence of drug resistance. Moreover, the successful elimination and complete management of these parasites are difficult due to the lack of effective vaccines against Plasmodium and Schistosoma infections. Therefore, it is important to highlight all current vaccine candidates undergoing clinical trials, such as pre-erythrocytic and erythrocytic stage malaria, as well as a next-generation RTS,S-like vaccine, the R21/Matrix-M vaccine, that conferred 77% protection against clinical malaria in a Phase 2b trial. Moreover, this review also discusses the progress and development of schistosomiasis vaccines. Furthermore, significant information is provided through this review on the effectiveness and progress of schistosomiasis vaccines currently under clinical trials, such as Sh28GST, Sm-14, and Sm-p80. Overall, this review provides insights into recent progress in malarial and schistosomiasis vaccines and their developmental approaches.
Collapse
Affiliation(s)
- Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia
| | - Jyoti Bhardwaj
- Division of Infectious Diseases, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Juhi Saxena
- Department of Biotechnology, University Institute of Biotechnology, Chandigarh University, Gharuan, NH-95, Ludhiana—Chandigarh State Hwy, Mohali 140413, India
| | - Sadaf Jahan
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia
| | - Mejdi Snoussi
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia
- Laboratory of Genetics, Biodiversity and Valorization of Bio-Resources (LR11ES41), Higher Institute of Biotechnology of Monastir, University of Monastir, Avenue TaharHaddas BP74, Monastir 5000, Tunisia
| | - Fevzi Bardakci
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia
| | - Riadh Badraoui
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia
- Section of Histology-Cytology, Medicine Faculty of Tunis, University of Tunis El Manar, Tunis 1017, Tunisia
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia
| |
Collapse
|
24
|
Diemert DJ, Correa-Oliveira R, Fraga CG, Talles F, Silva MR, Patel SM, Galbiati S, Kennedy JK, Lundeen JS, Gazzinelli MF, Li G, Hoeweler L, Deye GA, Bottazzi ME, Hotez PJ, El Sahly HM, Keitel WA, Bethony J, Atmar RL. A randomized, controlled Phase 1b trial of the Sm-TSP-2 Vaccine for intestinal schistosomiasis in healthy Brazilian adults living in an endemic area. PLoS Negl Trop Dis 2023; 17:e0011236. [PMID: 36996185 PMCID: PMC10089325 DOI: 10.1371/journal.pntd.0011236] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 04/11/2023] [Accepted: 03/12/2023] [Indexed: 04/01/2023] Open
Abstract
BACKGROUND Recombinant Schistosoma mansoni Tetraspanin-2 formulated on Alhydrogel (Sm-TSP-2/Alhydrogel) is being developed to prevent intestinal and hepatic disease caused by S. mansoni. The tegumentary Sm-TSP-2 antigen was selected based on its unique recognition by cytophilic antibodies in putatively immune individuals living in areas of ongoing S. mansoni transmission in Brazil, and preclinical studies in which vaccination with Sm-TSP-2 protected mice following infection challenge. METHODS A randomized, observer-blind, controlled, Phase 1b clinical trial was conducted in 60 healthy adults living in a region of Brazil with ongoing S. mansoni transmission. In each cohort of 20 participants, 16 were randomized to receive one of two formulations of Sm-TSP-2 vaccine (adjuvanted with Alhydrogel only, or with Alhydrogel plus the Toll-like receptor-4 agonist, AP 10-701), and 4 to receive Euvax B hepatitis B vaccine. Successively higher doses of antigen (10 μg, 30 μg, and 100 μg) were administered in a dose-escalation fashion, with progression to the next dose cohort being dependent upon evaluation of 7-day safety data after all participants in the preceding cohort had received their first dose of vaccine. Each participant received 3 intramuscular injections of study product at intervals of 2 months and was followed for 12 months after the third vaccination. IgG and IgG subclass antibody responses to Sm-TSP-2 were measured by qualified indirect ELISAs at pre- and post-vaccination time points through the final study visit. RESULTS Sm-TSP-2/Alhydrogel administered with or without AP-10-701 was well-tolerated in this population. The most common solicited adverse events were mild injection site tenderness and pain, and mild headache. No vaccine-related serious adverse events or adverse events of special interest were observed. Groups administered Sm-TSP-2/Alhydrogel with AP 10-701 had higher post-vaccination levels of antigen-specific IgG antibody. A significant dose-response relationship was seen in those administered Sm-TSP-2/Alhydrogel with AP 10-701. Peak anti-Sm-TSP-2 IgG levels were observed approximately 2 weeks following the third dose, regardless of Sm-TSP-2 formulation. IgG levels fell to low levels by Day 478 in all groups except the 100 μg with AP 10-701 group, in which 50% of subjects (4 of 8) still had IgG levels that were ≥4-fold higher than baseline. IgG subclass levels mirrored those of total IgG, with IgG1 being the predominant subclass response. CONCLUSIONS Vaccination of adults with Sm-TSP-2/Alhydrogel in an area of ongoing S. mansoni transmission was safe, minimally reactogenic, and elicited significant IgG and IgG subclass responses against the vaccine antigen. These promising results have led to initiation of a Phase 2 clinical trial of this vaccine in an endemic region of Uganda. TRIAL REGISTRATION NCT03110757.
Collapse
Affiliation(s)
- David J Diemert
- Department of Medicine, School of Medicine and Health Sciences, The George Washington University, Washington DC, United States of America
- Department of Microbiology, Immunology and Tropical Medicine, School of Medicine and Health Sciences, The George Washington University, Washington DC, United States of America
| | - Rodrigo Correa-Oliveira
- Instituto René Rachou, Fundação Oswaldo Cruz em Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Carlo Geraldo Fraga
- Instituto René Rachou, Fundação Oswaldo Cruz em Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Frederico Talles
- Instituto René Rachou, Fundação Oswaldo Cruz em Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marcella Rezende Silva
- Instituto René Rachou, Fundação Oswaldo Cruz em Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Shital M Patel
- Departments of Molecular Virology & Microbiology and Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Shirley Galbiati
- The Emmes Company, LLC, Frederick, Maryland, United States of America
| | - Jessie K Kennedy
- The Emmes Company, LLC, Frederick, Maryland, United States of America
| | - Jordan S Lundeen
- The Emmes Company, LLC, Frederick, Maryland, United States of America
| | - Maria Flavia Gazzinelli
- Instituto René Rachou, Fundação Oswaldo Cruz em Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Guangzhao Li
- Department of Microbiology, Immunology and Tropical Medicine, School of Medicine and Health Sciences, The George Washington University, Washington DC, United States of America
| | - Lara Hoeweler
- Department of Microbiology, Immunology and Tropical Medicine, School of Medicine and Health Sciences, The George Washington University, Washington DC, United States of America
| | - Gregory A Deye
- Division of Microbiology and Infectious Diseases (DMID), National Institutes of Allergy and Infectious, Diseases (NIAID), National Institutes of Health (NIH), United States of America
| | - Maria Elena Bottazzi
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Peter J Hotez
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Hana M El Sahly
- Departments of Molecular Virology & Microbiology and Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Wendy A Keitel
- Departments of Molecular Virology & Microbiology and Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jeffrey Bethony
- Department of Microbiology, Immunology and Tropical Medicine, School of Medicine and Health Sciences, The George Washington University, Washington DC, United States of America
| | - Robert L Atmar
- Departments of Molecular Virology & Microbiology and Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
25
|
Ford A, Hwang A, Mo AX, Baqar S, Touchette N, Deal C, King D, Earle K, Giersing B, Dull P, Hall BF. Meeting Summary: Global Vaccine and Immunization Research Forum, 2021. Vaccine 2023; 41:1799-1807. [PMID: 36803897 PMCID: PMC9938725 DOI: 10.1016/j.vaccine.2023.02.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 01/17/2023] [Accepted: 02/09/2023] [Indexed: 02/21/2023]
Abstract
The 2021 Global Vaccine and Immunization Research Forum highlighted the considerable advances and recent progress in research and development for vaccines and immunization, critically reviewed lessons learned from COVID-19 vaccine programs, and looked ahead to opportunities for this decade. For COVID-19, decades of investments in basic and translational research, new technology platforms, and vaccines targeting prototype pathogens enabled a rapid, global response. Unprecedented global coordination and partnership have played an essential role in creating and delivering COVID-19 vaccines. More improvement is needed in product attributes such as deliverability, and in equitable access to vaccines. Developments in other priority areas included: the halting of two human immunodeficiency virus vaccine trials due to lack of efficacy in preventing infection; promising efficacy results in Phase 2 trials of two tuberculosis vaccines; pilot implementation of the most advanced malaria vaccine candidate in three countries; trials of human papillomavirus vaccines given in single-dose regimens; and emergency use listing of a novel, oral poliomyelitis type 2 vaccine. More systematic, proactive approaches are being developed for fostering vaccine uptake and demand, aligning on priorities for investment by the public and private sectors, and accelerating policy making. Participants emphasized that addressing endemic disease is intertwined with emergency preparedness and pandemic response, so that advances in one area create opportunities in the other. In this decade, advances made in response to the COVID-19 pandemic should accelerate availability of vaccines for other diseases, contribute to preparedness for future pandemics, and help to achieve impact and equity under Immunization Agenda 2030.
Collapse
Key Words
- bcg, bacille calmette-guérin
- bnab, broadly neutralizing antibody
- cepi, coalition for epidemic preparedness innovations
- chim, controlled human infection model
- ecvp, evidence considerations for vaccine policy
- eua, emergency use authorization
- eul, emergency use listing
- gvap, global vaccine action plan
- gvirf, global vaccine and immunization research forum
- hiv, human immunodeficiency virus
- hpv, human papillomavirus
- ia2030, immunization agenda 2030
- mers, middle east respiratory syndrome
- nopv-2, novel oral poliomyelitis type 2 vaccine
- ppp, public–private partnership
- r&d, research and development
- sars, severe acute respiratory syndrome
- vips, vaccine innovation prioritisation strategy
Collapse
Affiliation(s)
- Andrew Ford
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, MSC 9825, Bethesda, MD 20892-9825, USA.
| | - Angela Hwang
- Angela Hwang Consulting, PO Box 6601, Albany, CA 94706, USA.
| | - Annie X. Mo
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, MSC 9825, Bethesda, MD 20892-9825, USA
| | - Shahida Baqar
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, MSC 9825, Bethesda, MD 20892-9825, USA.
| | - Nancy Touchette
- Office of Global Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Carolyn Deal
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, MSC 9825, Bethesda, MD 20892-9825, USA.
| | - Deborah King
- Infectious Disease Health Challenge - Prevention, Wellcome Trust, London NW1 2BE, United Kingdom.
| | - Kristen Earle
- Vaccine Development & Surveillance, Bill & Melinda Gates Foundation, PO Box 23350, Seattle, Washington 98102, USA.
| | - Birgitte Giersing
- Department of Immunization, Vaccines, and Biologicals, World Health Organization, Geneva, Switzerland.
| | - Peter Dull
- Vaccine Development & Surveillance, Bill & Melinda Gates Foundation, PO Box 23350, Seattle, Washington 98102, USA.
| | - B. Fenton Hall
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, MSC 9825, Bethesda, MD 20892-9825, USA
| |
Collapse
|
26
|
Salmonella Typhimurium expressing chromosomally integrated Schistosoma mansoni Cathepsin B protects against schistosomiasis in mice. NPJ Vaccines 2023; 8:27. [PMID: 36849453 PMCID: PMC9969381 DOI: 10.1038/s41541-023-00599-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 01/13/2023] [Indexed: 03/01/2023] Open
Abstract
Schistosomiasis threatens hundreds of millions of people worldwide. The larval stage of Schistosoma mansoni migrates through the lung and adult worms reside adjacent to the colonic mucosa. Several candidate vaccines are in preclinical development, but none is designed to elicit both systemic and mucosal responses. We have repurposed an attenuated Salmonella enterica Typhimurium strain (YS1646) to express Cathepsin B (CatB), a digestive enzyme important for the juvenile and adult stages of the S. mansoni life cycle. Previous studies have demonstrated the prophylactic and therapeutic efficacy of our plasmid-based vaccine. Here, we have generated chromosomally integrated (CI) YS1646 strains that express CatB to produce a viable candidate vaccine for eventual human use (stability, no antibiotic resistance). 6-8-week-old C57BL/6 mice were vaccinated in a multimodal oral (PO) and intramuscular (IM) regimen, and then sacrificed 3 weeks later. The PO + IM group had significantly higher anti-CatB IgG titers with greater avidity and mounted significant intestinal anti-CatB IgA responses compared to PBS control mice (all P < 0.0001). Multimodal vaccination generated balanced TH1/TH2 humoral and cellular immune responses. Production of IFNγ by both CD4+ and CD8+ T cells was confirmed by flow cytometry (P < 0.0001 & P < 0.01). Multimodal vaccination reduced worm burden by 80.4%, hepatic egg counts by 75.2%, and intestinal egg burden by 78.4% (all P < 0.0001). A stable and safe vaccine that has both prophylactic and therapeutic activity would be ideal for use in conjunction with praziquantel mass treatment campaigns.
Collapse
|
27
|
Senghor B, Webster B, Pennance T, Sène M, Doucouré S, Sow D, Sokhna C. Molecular characterization of schistosome cercariae and their Bulinus snail hosts from Niakhar, a seasonal transmission focus in central Senegal. CURRENT RESEARCH IN PARASITOLOGY & VECTOR-BORNE DISEASES 2023; 3:100114. [PMID: 36824299 PMCID: PMC9941053 DOI: 10.1016/j.crpvbd.2023.100114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/11/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023]
Abstract
Bulinus senegalensis and Bulinus umbilicatus, two sympatric freshwater snails found in temporal ponds in Senegal, were thought to be involved in the transmission of Schistosoma haematobium and/or Schistosoma curassoni. To better understand the role of these Bulinus species in the transmission of human and animal Schistosoma species, B. senegalensis and B. umbilicatus were collected in 2015, during a malacological survey, from a temporal pond in Niakhar, central Senegal. Snails were induced to shed cercariae on two consecutive days. Individual cercariae from each snail were collected and preserved for molecular identification. Infected snails were identified by analysis of a partial region of the cytochrome c oxidase subunit 1 (cox1) gene. Six individual cercariae shed from each infected snail were identified by analyses of the cox1, nuclear ITS and partial 18S rDNA regions. Of the 98 snails collected, one B. senegalensis had a mixed infection shedding S. haematobium, S. bovis and S. haematobium-S. bovis hybrid cercariae and one B. umbilicatus was found to be shedding only S. haematobium. These data provide molecular confirmation for B. senegalensis transmitting S. bovis and S. haematobium-S. bovis hybrids in Senegal. The multiple Bulinus species involved in the human urogenital schistosomiasis in Senegal provides a high force of transmission warranting detailed mapping, surveillance and regular treatment of at-risk populations.
Collapse
Affiliation(s)
- Bruno Senghor
- VITROME, Campus International IRD-UCAD de Hann, 1386, Dakar, Senegal
| | - Bonnie Webster
- Natural History Museum, Wolfson Wellcome Biomedical Laboratories, Department of Science, Cromwell Road, London, SW7 5BD, UK
- London Centre for Neglected Tropical Disease Research, Imperial College London, School of Public Health, Norfolk Pl, Paddington, London, W21PG, UK
| | - Tom Pennance
- Natural History Museum, Wolfson Wellcome Biomedical Laboratories, Department of Science, Cromwell Road, London, SW7 5BD, UK
- London Centre for Neglected Tropical Disease Research, Imperial College London, School of Public Health, Norfolk Pl, Paddington, London, W21PG, UK
- Department of Basic Medical Sciences, Western University of Health Sciences, Lebanon, OR, 97355, USA
| | - Mariama Sène
- Laboratoire des Sciences Biologiques, Agronomiques et de Modélisation des Systems Complexes, UFRS2ATA, Université Gaston Berger de Saint-Louis, Saint-Louis, Senegal
| | | | - Doudou Sow
- Department of Parasitology-Mycology, UFR Sciences de la Santé, Université Gaston Berger, 234, Saint-Louis, Senegal
| | - Cheikh Sokhna
- VITROME, Campus International IRD-UCAD de Hann, 1386, Dakar, Senegal
- VITROME, Aix-Marseille Univ, IRD, SSA, AP-HM, IHU-Mediterranean Infection, Marseille, France
| |
Collapse
|
28
|
Fattori ACM, Montija EDA, Fragelli BDDL, Correia RDO, de Castro CA, Romanello L, Nogueira CT, Allegretti SM, Soares EG, Pereira HD, Anibal FDF. Effects of Immunization with Recombinant Schistosoma mansoni Enzymes AK and HGPRT: Murine Infection Control. Pathogens 2023; 12:pathogens12010069. [PMID: 36678417 PMCID: PMC9866087 DOI: 10.3390/pathogens12010069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/16/2022] [Accepted: 12/23/2022] [Indexed: 01/03/2023] Open
Abstract
Schistosomiasis is one of the most important human helminthiases worldwide. Praziquantel is the current treatment, and no vaccine is available until the present. Thus, the presented study aimed to evaluate the immunization effects with recombinant Schistosoma mansoni enzymes: Adenosine Kinase (AK) and Hypoxanthine-Guanine Phosphoribosyltransferase (HGPRT), as well as a MIX of the two enzymes. Female Balb/c mice were immunized in three doses, and 15 days after the last immunization, animals were infected with S. mansoni. Our results showed that the group MIX presented a reduction in the eggs in feces by 30.74% and 29%, respectively, in the adult worms. The groups AK, HGPRT and MIX could produce IgG1 antibodies, and the groups AK and MIX produced IgE antibodies anti-enzymes and anti-S. mansoni total proteins. The groups AK, HGPRT and MIX induced a reduction in the eosinophils in the peritoneal cavity. Besides, the group AK showed a decrease in the number of hepatic granulomas (41.81%) and the eggs present in the liver (42.30%). Therefore, it suggests that immunization with these enzymes can contribute to schistosomiasis control, as well as help to modulate experimental infection inducing a reduction of physiopathology in the disease.
Collapse
Affiliation(s)
- Ana Carolina Maragno Fattori
- Laboratório de Inflamação e Doenças Infecciosas, Departamento de Morfologia e Patologia, Universidade Federal de São Carlos, São Carlos 13565-905, Brazil
- Correspondence: (A.C.M.F.); (F.d.F.A.)
| | - Elisandra de A. Montija
- Laboratório de Inflamação e Doenças Infecciosas, Departamento de Morfologia e Patologia, Universidade Federal de São Carlos, São Carlos 13565-905, Brazil
| | - Bruna D. de L. Fragelli
- Laboratório de Inflamação e Doenças Infecciosas, Departamento de Morfologia e Patologia, Universidade Federal de São Carlos, São Carlos 13565-905, Brazil
| | - Ricardo de O. Correia
- Laboratório de Inflamação e Doenças Infecciosas, Departamento de Morfologia e Patologia, Universidade Federal de São Carlos, São Carlos 13565-905, Brazil
| | - Cynthia Aparecida de Castro
- Laboratório de Inflamação e Doenças Infecciosas, Departamento de Morfologia e Patologia, Universidade Federal de São Carlos, São Carlos 13565-905, Brazil
| | - Larissa Romanello
- Departamento de Saúde e Psicologia, Universidade do Estado de Minas Gerais, Ituiutaba 38302-192, Brazil
| | - Camila T. Nogueira
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04039-032, Brazil
| | - Silmara M. Allegretti
- Instituto de Biologia, Departamento de Biologia Animal, Universidade Estadual de Campinas, Campinas 13083-970, Brazil
| | - Edson G. Soares
- Laboratório de Citopatologia, Departamento de Patologia e Medicina Legal, Universidade de São Paulo, Ribeirão Preto 14040-900, Brazil
| | - Humberto D. Pereira
- Instituto de Física de São Carlos, Universidade de São Paulo, São Carlos 13566-590, Brazil
| | - Fernanda de F. Anibal
- Laboratório de Inflamação e Doenças Infecciosas, Departamento de Morfologia e Patologia, Universidade Federal de São Carlos, São Carlos 13565-905, Brazil
- Correspondence: (A.C.M.F.); (F.d.F.A.)
| |
Collapse
|
29
|
Chanhanga N, Mindu T, Mogaka J, Chimbari M. The Impact of Targeted Treatment and Mass Drug Administration Delivery Strategies on the Prevalence and Intensity of Schistosomiasis in School Aged Children in Africa: A Systematic Review. Infect Drug Resist 2023; 16:2453-2466. [PMID: 37138838 PMCID: PMC10150034 DOI: 10.2147/idr.s395382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/16/2023] [Indexed: 05/05/2023] Open
Abstract
Schistosomiasis is a public health problem in more than 78 countries in the world. The disease is most prevalent among children than adults due to their high exposure to infectious water sources. Various interventions such as mass drug administration (MDA), snail control, safe water provision and health education have been implemented independently or jointly to control, reduce and ultimately eliminate Schistosomiasis. This scoping review focused on studies reporting the impact of different delivery strategies of targeted treatment and MDA on the prevalence and intensity of schistosomiasis infection in school aged children in Africa. The review focused on Schistosoma haematobium and Schistosoma mansoni species. A systematic search for eligible literature from peer-reviewed articles was done from Google Scholar, Medline, PubMed and EBSCO host databases. The search yielded twenty-seven peer-reviewed articles. All articles found reported a decrease in the prevalence of schistosomiasis infection. Five studies (18.5%) reported a prevalence change below 40%, eighteen studies (66.7%) reported a change between 40% and 80%, and four studies (14.8%) reported a change above 80%. The infection intensity post-treatment was varied: twenty-four studies reported a decrease, while two studies reported an increase. The review showed that the impact of targeted treatment on the prevalence and intensity of schistosomiasis depended on the frequency at which it was offered, complementary interventions, and its uptake by the target population. Targeted treatment can significantly control the infection burden, but cannot eliminate the disease. Constant MDA programs coupled with preventative and health promotional programs are required to reach the elimination stage.
Collapse
Affiliation(s)
- Nathan Chanhanga
- School of Nursing and Public Health, University of KwaZulu Natal, Durban, South Africa
| | - Tafadzwa Mindu
- School of Nursing and Public Health, University of KwaZulu Natal, Durban, South Africa
- Correspondence: Tafadzwa Mindu, Email
| | - John Mogaka
- School of Nursing and Public Health, University of KwaZulu Natal, Durban, South Africa
| | - Moses Chimbari
- School of Nursing and Public Health, University of KwaZulu Natal, Durban, South Africa
- Research and Innovation, Great Zimbabwe University, Masvingo, Zimbabwe
| |
Collapse
|
30
|
Girod V, Houssier R, Sahmer K, Ghoris MJ, Caby S, Melnyk O, Dissous C, Senez V, Vicogne J. A self-purifying microfluidic system for identifying drugs acting against adult schistosomes. ROYAL SOCIETY OPEN SCIENCE 2022; 9:220648. [PMID: 36465675 PMCID: PMC9709518 DOI: 10.1098/rsos.220648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 11/09/2022] [Indexed: 06/17/2023]
Abstract
The discovery of novel antihelmintic molecules to combat the development and spread of schistosomiasis, a disease caused by several Schistosoma flatworm species, mobilizes significant research efforts worldwide. With a limited number of biochemical assays for measuring the viability of adult worms, the antischistosomicidal activity of molecules is usually evaluated by a microscopic observation of worm mobility and/or integrity upon drug exposure. Even if these phenotypical assays enable multiple parameters analysis, they are often conducted during several days and need to be associated with image-based analysis to minimized subjectivity. We describe here a self-purifying microfluidic system enabling the selection of healthy adult worms and the identification of molecules acting instantly on the parasite. The worms are assayed in a dynamic environment that eliminates unhealthy worms that cannot attach firmly to the chip walls prior to being exposed to the drug. The detachment of the worms is also used as second step readout for identifying active compounds. We have validated this new fluidic screening approach using the two major antihelmintic drugs, praziquantel and artemisinin. The reported dynamic system is simple to produce and to parallelize. Importantly, it enables a quick and sensitive detection of antischistosomal compounds in no more than one hour.
Collapse
Affiliation(s)
- Vincent Girod
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 – CANTHER – Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille F-59000, France
- CNRS, University of Tokyo, IRL2820 – LIMMS, Lille F-59000, France
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017 – Center for Infection and Immunity of Lille, F-59000 Lille, France
- University of Lille, CNRS, UPHF, JUNIA, CLI, UMR 8520 – IEMN – Institut d'Electronique, de Microélectronique et de Nanotechnologie, Villeneuve d'Ascq F-59650, France
| | - Robin Houssier
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 – CANTHER – Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille F-59000, France
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017 – Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Karin Sahmer
- University of Lille, IMT Lille Douai, University of Artois, JUNIA, ULR 4515 – LGCgE, Laboratoire de Génie Civil et géo-Environnement, F-59000 Lille, France
| | - Marie-José Ghoris
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017 – Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Stéphanie Caby
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017 – Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Oleg Melnyk
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017 – Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Colette Dissous
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017 – Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Vincent Senez
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 – CANTHER – Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille F-59000, France
- CNRS, University of Tokyo, IRL2820 – LIMMS, Lille F-59000, France
| | - Jérôme Vicogne
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017 – Center for Infection and Immunity of Lille, F-59000 Lille, France
| |
Collapse
|
31
|
Dibo N, Liu X, Chang Y, Huang S, Wu X. Pattern recognition receptor signaling and innate immune responses to schistosome infection. Front Cell Infect Microbiol 2022; 12:1040270. [PMID: 36339337 PMCID: PMC9633954 DOI: 10.3389/fcimb.2022.1040270] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/03/2022] [Indexed: 08/22/2023] Open
Abstract
Schistosomiasis remains to be a significant public health problem in tropical and subtropical regions. Despite remarkable progress that has been made in the control of the disease over the past decades, its elimination remains a daunting challenge in many countries. This disease is an inflammatory response-driven, and the positive outcome after infection depends on the regulation of immune responses that efficiently clear worms and allow protective immunity to develop. The innate immune responses play a critical role in host defense against schistosome infection and pathogenesis. Initial pro-inflammatory responses are essential for clearing invading parasites by promoting appropriate cell-mediated and humoral immunity. However, elevated and prolonged inflammatory responses against the eggs trapped in the host tissues contribute to disease progression. A better understanding of the molecular mechanisms of innate immune responses is important for developing effective therapies and vaccines. Here, we update the recent advances in the definitive host innate immune response to schistosome infection, especially highlighting the critical roles of pattern recognition receptors and cytokines. The considerations for further research are also provided.
Collapse
Affiliation(s)
- Nouhoum Dibo
- Department of medical parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, China
| | - Xianshu Liu
- Department of medical parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, China
| | - Yunfeng Chang
- Department of Forensic Medicine Science, Xiangya School of Basic Medicine, Central South University, Yueyang, China
| | - Shuaiqin Huang
- Department of medical parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, China
- Hunan Provincial Key Lab of Immunology and Transmission Control on Schistosomiasis, Hunan Provincial Institute of Schistosomiasis Control, Yueyang, China
| | - Xiang Wu
- Department of medical parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, China
- Hunan Provincial Key Lab of Immunology and Transmission Control on Schistosomiasis, Hunan Provincial Institute of Schistosomiasis Control, Yueyang, China
| |
Collapse
|
32
|
Morales-Vicente DA, Zhao L, Silveira GO, Tahira AC, Amaral MS, Collins JJ, Verjovski-Almeida S. Single-cell RNA-seq analyses show that long non-coding RNAs are conspicuously expressed in Schistosoma mansoni gamete and tegument progenitor cell populations. Front Genet 2022; 13:924877. [PMID: 36204320 PMCID: PMC9531161 DOI: 10.3389/fgene.2022.924877] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
Schistosoma mansoni is a flatworm that causes schistosomiasis, a neglected tropical disease that affects over 200 million people worldwide. New therapeutic targets are needed with only one drug available for treatment and no vaccine. Long non-coding RNAs (lncRNAs) are transcripts longer than 200 nucleotides with low or no protein-coding potential. In other organisms, they have been shown as involved with reproduction, stem cell maintenance and drug resistance, and they tend to exhibit tissue-specific expression patterns. S. mansoni expresses thousands of lncRNA genes; however, the cell type expression patterns of lncRNAs in the parasite remain uncharacterized. Here, we have re-analyzed publicly available single-cell RNA-sequencing (scRNA-seq) data obtained from adult S. mansoni to identify the lncRNAs signature of adult schistosome cell types. A total of 8023 lncRNAs (79% of all lncRNAs) were detected. Analyses of the lncRNAs expression profiles in the cells using statistically stringent criteria were performed to identify 74 lncRNA gene markers of cell clusters. Male gamete and tegument progenitor lineages clusters contained most of the cluster-specific lncRNA markers. We also identified lncRNA markers of specific neural clusters. Whole-mount in situ hybridization (WISH) and double fluorescence in situ hybridization were used to validate the cluster-specific expression of 13 out of 16 selected lncRNA genes (81%) in the male and female adult parasite tissues; for one of these 16 gene loci, probes for two different lncRNA isoforms were used, which showed differential isoform expression in testis and ovary. An atlas of the expression profiles across the cell clusters of all lncRNAs detected in our analysis is available as a public website resource (http://verjolab.usp.br:8081). The results presented here give strong support to a tissue-specific expression and to a regulated expression program of lncRNAs in S. mansoni. This will be the basis for further exploration of lncRNA genes as potential therapeutic targets.
Collapse
Affiliation(s)
- David A. Morales-Vicente
- Laboratório de Parasitologia, Instituto Butantan, São Paulo, Brazil
- Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Lu Zhao
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Gilbert O. Silveira
- Laboratório de Parasitologia, Instituto Butantan, São Paulo, Brazil
- Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Ana C. Tahira
- Laboratório de Parasitologia, Instituto Butantan, São Paulo, Brazil
| | - Murilo S. Amaral
- Laboratório de Parasitologia, Instituto Butantan, São Paulo, Brazil
| | - James J. Collins
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Sergio Verjovski-Almeida
- Laboratório de Parasitologia, Instituto Butantan, São Paulo, Brazil
- Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
33
|
In Vitro Cercaricidal and Schistosomicidal Activities of the Raffia Wine and Hydroethanolic Extracts of Pedilanthus tithymaloides Linn (Poit). Stem Barks. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2672150. [PMID: 36159554 PMCID: PMC9507742 DOI: 10.1155/2022/2672150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/14/2022] [Accepted: 08/16/2022] [Indexed: 11/18/2022]
Abstract
Schistosomiasis control remains a public health concern, and there is a need to evaluate new strategies for targeting larval and adult stages of the parasite. As Pedilanthus tithymaloides is empirically used to treat schistosomiasis, it becomes essential to know its effective action scientifically. This study assessed the cercaricidal and schistosomicidal activity of P. tithymaloides stem barks raffia wine extract (RwPt) and hydroethanolic extract (HePt). Different concentrations of these extracts were tested against cercariae (31.25–1000 μg/mL) and adult worms (62.5–2000 μg/mL) of Schistosoma mansoni. Niclosamide-olamine 5% (1 μg/mL) and praziquantel (10 μg/mL) were used as pharmacological controls. Cercariae viability was determined every 30 min for 180 min, and adult worms’ motor activity and viability after 24 and 48 h incubation. In addition, cytotoxicity and phytochemical analysis were performed. HePt was lethal to cercariae and adult worms with LC50 of 73.91 μg/mL after 60 min of incubation and 731.17 μg/mL after 48 h of incubation, respectively. Furthermore, a significant reduction of 94.44% in motor activity was observed in surviving worms at the concentration of 2000 μg/mL. RwPt was less effective on S. mansoni cercariae with an LC50 of 617.86 μg/mL after 180 min and on adult worms with a mortality rate of 9.83% at 2000 μg/mL for 48 h incubation. Both extracts showed a weak cytotoxicity profile with an IC50 of 983.50 μg/mL for HePt and more than 1000 μg/mL for RwPt. The LC-MS analysis of HePt allowed the detection of two annotated diterpenoids. Based on the selectivity index, the hydroethanolic extract of P. tithymaloides stem barks disclosed an intense cercaricidal activity and a moderate schistosomicidal effect with low cytotoxicity. These findings may support the potential use of Pedilanthus tithymaloides as a natural product or a source of natural-derived compounds for interrupting schistosomiasis transmission.
Collapse
|
34
|
Skelly PJ, Da'dara AA. Schistosome secretomes. Acta Trop 2022; 236:106676. [PMID: 36113567 DOI: 10.1016/j.actatropica.2022.106676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/08/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022]
Abstract
Schistosomes are intravascular parasitic platyhelminths (blood flukes) that infect over 200 million people globally. Biomolecules secreted by the worms likely contribute to their ability to survive in the bloodstreams of immunocompetent hosts for many years. Here we review what is known about the protein composition of material released by the worms. Prominent among cercarial excretions/secretions (ES) is a ∼ 30 kDa serine protease called cercarial elastase (SmCE in Schistosoma mansoni), likely important in host invasion. Also prominent is a 117 amino acid non-glycosylated polypeptide (Sm16) that can impact several host cell-types to impinge on immunological outcomes. Similarly, components of the egg secretome (notably the 134 amino acid homodimeric glycoprotein "IL-4 inducing principle of schistosome eggs", IPSE, and the 225-amino acid monomeric T2 ribonuclease - omega-1) are capable of driving Th2-biased immune responses. A ∼36kDa chemokine binding glycoprotein SmCKBP, secreted by eggs, can negate the impact of several cytokines and can impede neutrophil migration. Of special interest is a disparate collection of classically cytosolic proteins that are surprisingly often identified in schistosome ES across life stages. These proteins, perhaps released as components of extracellular vesicles (EVs), include glycolytic enzymes, redox proteins, proteases and protease inhibitors, heat shock proteins, proteins involved in translation/turnover, histones, and others. Some such proteins may display "moonlighting" functions and, for example, impede blood clot formation around the worms. More prosaically, since several are particularly abundant soluble proteins, their appearance in the ES fraction may be indicative of worm damage ex vivo leading to protein leakage. Some bioactive schistosome ES proteins are in development as novel therapeutics against autoimmune, inflammatory, and other, non-parasitic, diseases.
Collapse
Affiliation(s)
- Patrick J Skelly
- Molecular Helminthology Laboratory, Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, USA.
| | - Akram A Da'dara
- Molecular Helminthology Laboratory, Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, USA
| |
Collapse
|
35
|
Phuphisut O, Kobpornchai P, Chusongsang P, Limpanont Y, Kanjanapruthipong T, Ampawong S, Reamtong O, Adisakwattana P. Molecular characterization and functional analysis of Schistosoma mekongi neuroglobin homolog. Acta Trop 2022; 231:106433. [PMID: 35364046 DOI: 10.1016/j.actatropica.2022.106433] [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: 09/22/2021] [Revised: 03/11/2022] [Accepted: 03/28/2022] [Indexed: 11/01/2022]
Abstract
Schistosomes are blood-dwelling parasites that are constantly exposed to high-level oxidative stress arising from parasite-intrinsic and host defense mechanisms. To survive in their hosts, schistosomes require an antioxidant system to minimize with oxidative stress. Several schistosome antioxidant enzymes have been identified and have been suggested to play indispensable antioxidant roles for the parasite. In addition to antioxidant enzymes, non-enzymatic antioxidants including small molecules, peptides, and proteins have been identified and characterized. Neuroglobin (Ngb), a nervous system-specific heme-binding protein, has been classified as a non-enzymatic antioxidant and is capable of scavenging a variety of free radical species. The antioxidant activity of Ngb has been well-studied in humans. Ngb is involved in cellular oxygen homeostasis and reactive oxygen/nitrogen scavenging in the central and peripheral nervous systems, but its functions in schistosome parasites have not yet been characterized. In this study, we aimed to characterize the molecular properties and functions of Schistosoma mekongi Ngb (SmeNgb) using bioinformatic, biochemical, and molecular biology approaches. The amino acid sequence of Ngb was highly conserved among schistosomes as well as closely related trematodes. SmeNgb was abundantly localized in the gastrodermis, vitelline, and ovary of adult female S. mekongi worms as well as in the tegument of adult male worms. Assessment of antioxidant activity demonstrated that recombinant SmeNgb had Fe2+ chelating and hydrogen peroxide scavenging activities. Intriguingly, siRNA silencing of SmeNgb gene expression resulted in tegument pathology. Understanding the properties and functions of SmNgb will help in future development of effective treatments and vaccines against S. mekongi, other schistosome parasites, and other platyhelminths.
Collapse
|
36
|
Mtemeli FL, Shoko R, Ndlovu J, Mugumbate G. In Silico Study of Cucurbita maxima Compounds as Potential Therapeutics Against Schistosomiasis. Bioinform Biol Insights 2022; 16:11779322221100741. [PMID: 35615403 PMCID: PMC9125113 DOI: 10.1177/11779322221100741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/21/2022] [Indexed: 11/15/2022] Open
Abstract
Schistosomiasis, a disease usually related to poverty and poor sanitation, affects more than 200 million people worldwide. Since the 1970s, the medical sector has depended on a single drug, praziquantel, for the treatment of the disease. The emerging evidence of resistance of the Schistosoma parasite to praziquantel and the drug’s inefficacy against juvenile stages of the parasite makes the need to find alternative drugs an urgent matter. In this study, we explored the inhibition potential of compounds from Cucurbita maxima using molecular docking studies on Schistosoma mansoni purine nucleoside phosphorylase ( SmPNP) and Schistosoma haematobium 28-kDa glutathione S-transferase ( Sh28kDaGST). Following molecular docking studies and analysis of the active sites, the primary amino acids that were observed and shown to be involved in the SmPNP-ligand interaction are CYS 33, ARG 86, HIS 88, TYR 90, ALA 118, ALA 119, PRO 200, TYR 202, GLU 203, VAL 219, MET 221, THR 244, ASN 245, PRO 257 and HIS 259. For the Sh28dKa-ligand interaction, the primary amino acids were PHE 11, ARG 16, TRP 41, LEU 53, GLU 70 and SER 71. Momordicoside I aglycone binds to SmPNP with the lowest binding affinity of -7.9 kcal/mol by pi sigma bond interactions with HIS 88. Balsaminoside B binds to Sh28kDaGST with a binding affinity of −7.6 kcal/mol by hydrogen bond interaction with TRP 41, LEU 53 and SER 71. Pharmacokinetic studies showed favourable drug-like properties for the 10 compounds that exhibited the lowest binding energies. Therefore, we propose that bioactive compounds from C. maxima be considered as potential novel drug hits in the treatment of schistosomiasis.
Collapse
Affiliation(s)
- Floryn Lynorah Mtemeli
- Department of Biology, School of Natural Sciences and Mathematics, Chinhoyi University of Technology, Chinhoyi, Zimbabwe
| | - Ryman Shoko
- Department of Biology, School of Natural Sciences and Mathematics, Chinhoyi University of Technology, Chinhoyi, Zimbabwe
| | - Joice Ndlovu
- Department of Biology, School of Natural Sciences and Mathematics, Chinhoyi University of Technology, Chinhoyi, Zimbabwe
| | - Grace Mugumbate
- Department of Chemical Technology, Midlands State University, Gweru, Zimbabwe
| |
Collapse
|
37
|
Ogongo P, Nyakundi RK, Chege GK, Ochola L. The Road to Elimination: Current State of Schistosomiasis Research and Progress Towards the End Game. Front Immunol 2022; 13:846108. [PMID: 35592327 PMCID: PMC9112563 DOI: 10.3389/fimmu.2022.846108] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/21/2022] [Indexed: 12/14/2022] Open
Abstract
The new WHO Roadmap for Neglected Tropical Diseases targets the global elimination of schistosomiasis as a public health problem. To date, control strategies have focused on effective diagnostics, mass drug administration, complementary and integrative public health interventions. Non-mammalian intermediate hosts and other vertebrates promote transmission of schistosomiasis and have been utilized as experimental model systems. Experimental animal models that recapitulate schistosomiasis immunology, disease progression, and pathology observed in humans are important in testing and validation of control interventions. We discuss the pivotal value of these models in contributing to elimination of schistosomiasis. Treatment of schistosomiasis relies heavily on mass drug administration of praziquantel whose efficacy is comprised due to re-infections and experimental systems have revealed the inability to kill juvenile schistosomes. In terms of diagnosis, nonhuman primate models have demonstrated the low sensitivity of the gold standard Kato Katz smear technique. Antibody assays are valuable tools for evaluating efficacy of candidate vaccines, and sera from graded infection experiments are useful for evaluating diagnostic sensitivity of different targets. Lastly, the presence of Schistosomes can compromise the efficacy of vaccines to other infectious diseases and its elimination will benefit control programs of the other diseases. As the focus moves towards schistosomiasis elimination, it will be critical to integrate treatment, diagnostics, novel research tools such as sequencing, improved understanding of disease pathogenesis and utilization of experimental models to assist with evaluating performance of new approaches.
Collapse
Affiliation(s)
- Paul Ogongo
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
- Department of Tropical and Infectious Diseases, Institute of Primate Research, Nairobi, Kenya
| | - Ruth K. Nyakundi
- Department of Tropical and Infectious Diseases, Institute of Primate Research, Nairobi, Kenya
| | - Gerald K. Chege
- Primate Unit & Delft Animal Centre, South African Medical Research Council, Cape Town, South Africa
- Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Lucy Ochola
- Department of Tropical and Infectious Diseases, Institute of Primate Research, Nairobi, Kenya
- Department of Environmental Health, School of Behavioural and Lifestyle Sciences, Faculty of Health Sciences, Nelson Mandela University, Gqeberha, South Africa
| |
Collapse
|
38
|
Ravaynia PS, Biendl S, Grassi F, Keiser J, Hierlemann A, Modena MM. Real-time and automated monitoring of antischistosomal drug activity profiles for screening of compound libraries. iScience 2022; 25:104087. [PMID: 35378863 PMCID: PMC8976133 DOI: 10.1016/j.isci.2022.104087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 02/02/2022] [Accepted: 03/14/2022] [Indexed: 11/26/2022] Open
Abstract
Schistosomiasis is a neglected tropical disease that affects over 200 million people annually. As the antischistosomal drug pipeline is currently empty, repurposing of compound libraries has become a source for accelerating drug development, which demands the implementation of high-throughput and efficient screening strategies. Here, we present a parallelized impedance-based platform for continuous and automated viability evaluation of Schistosoma mansoni schistosomula in 128 microwells during 72 h to identify antischistosomal hits in vitro. By initially screening 57 repurposed compounds against larvae, five drugs are identified, which reduce parasite viability by more than 70%. The activity profiles of the selected drugs are then investigated via real-time dose-response monitoring, and four compounds reveal high potency and rapid action, which renders them suitable candidates for follow-up tests against adult parasites. The study shows that our device is a reliable tool for real-time drug screening analysis of libraries to identify new promising therapeutics against schistosomiasis. Scalable, plastic microwell chip with integrated platinum electrodes Automated impedance-based recording of 128 microwell units in parallel Continuous monitoring of in vitro drug library efficacy on schistosomula for 72 h Identification of four fast-acting antischistosomal drugs for in vivo testing
Collapse
Affiliation(s)
- Paolo S Ravaynia
- Bioengineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Stefan Biendl
- Swiss Tropical and Public Health Institute, Department of Medical Parasitology and Infection Biology, University of Basel, Socinstrasse 57, 4051 Basel, Switzerland
| | - Francesco Grassi
- Centre for Microsystems Technology, Department of Electronics and Information Systems, Ghent University, Technologiepark-Zwijnaarde 126, 9052 Gent, Belgium
| | - Jennifer Keiser
- Swiss Tropical and Public Health Institute, Department of Medical Parasitology and Infection Biology, University of Basel, Socinstrasse 57, 4051 Basel, Switzerland
| | - Andreas Hierlemann
- Bioengineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Mario M Modena
- Bioengineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, 4058 Basel, Switzerland
| |
Collapse
|
39
|
Hou N, Piao X, Jiang N, Liu S, Cai P, Liu B, McManus DP, Chen Q. Novel Hepatic Schistosomula Antigens as Promising Targets for Immunodiagnosis and Immunoprotection of Schistosomiasis japonica. J Infect Dis 2022; 225:1991-2001. [PMID: 35235942 DOI: 10.1093/infdis/jiac077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/01/2022] [Indexed: 11/12/2022] Open
Abstract
Antigens of migrating schistosomula are promising candidates as schistosomiasis vaccine targets, since immune attack on hepatic schistosomula would interrupt the parasites life cycle and reduce egg burden on the host. Here, we report a collection of Schistosoma japonicum schistosomula proteins (SjScP), which are highly expressed in hepatic schistosomula. We found that a number of these SjScPs were highly antigenic and could effectively stimulate humoral immune responses in both human and other mammalian hosts. In particular, SjScP25, SjScP37, SjScP41, SjScP80, SjScP88 showed high potential as biomarkers for schistosomiasis immunodiagnosis. Furthermore, we demonstrated that immunization with several of the recombinant SjScPs were able to protect mice from S. japonicum challenge infection, with SjScP25 generating the most protective results. Our work represents a group of novel schistosome immunogens, which may be promsing schistosomiasis japonica diagonosis and vaccine candidates.
Collapse
Affiliation(s)
- Nan Hou
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Dong Dan San Tiao, Beijing, China
| | - Xianyu Piao
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Dong Dan San Tiao, Beijing, China
| | - Ning Jiang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China.,The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China
| | - Shuai Liu
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Dong Dan San Tiao, Beijing, China
| | - Pengfei Cai
- Molecular Parasitology Laboratory, Infectious Diseases Program, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Australia
| | - Bing Liu
- Institute for Protein Science and Phage Research, the First Affiliated Hospital of Xi'an Jiaotong University, Shanxi, China
| | - Donald P McManus
- Molecular Parasitology Laboratory, Infectious Diseases Program, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Australia
| | - Qijun Chen
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Dong Dan San Tiao, Beijing, China.,Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China.,The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China
| |
Collapse
|
40
|
Pereira Moreira B, Weber MHW, Haeberlein S, Mokosch AS, Spengler B, Grevelding CG, Falcone FH. Drug Repurposing and De Novo Drug Discovery of Protein Kinase Inhibitors as New Drugs against Schistosomiasis. Molecules 2022; 27:molecules27041414. [PMID: 35209202 PMCID: PMC8879451 DOI: 10.3390/molecules27041414] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/09/2022] [Accepted: 02/16/2022] [Indexed: 02/01/2023] Open
Abstract
Schistosomiasis is a neglected tropical disease affecting more than 200 million people worldwide. Chemotherapy relies on one single drug, praziquantel, which is safe but ineffective at killing larval stages of this parasite. Furthermore, concerns have been expressed about the rise in resistance against this drug. In the absence of an antischistosomal vaccine, it is, therefore, necessary to develop new drugs against the different species of schistosomes. Protein kinases are important molecules involved in key cellular processes such as signaling, growth, and differentiation. The kinome of schistosomes has been studied and the suitability of schistosomal protein kinases as targets demonstrated by RNA interference studies. Although protein kinase inhibitors are mostly used in cancer therapy, e.g., for the treatment of chronic myeloid leukemia or melanoma, they are now being increasingly explored for the treatment of non-oncological conditions, including schistosomiasis. Here, we discuss the various approaches including screening of natural and synthetic compounds, de novo drug development, and drug repurposing in the context of the search for protein kinase inhibitors against schistosomiasis. We discuss the status quo of the development of kinase inhibitors against schistosomal serine/threonine kinases such as polo-like kinases (PLKs) and mitogen-activated protein kinases (MAP kinases), as well as protein tyrosine kinases (PTKs).
Collapse
Affiliation(s)
- Bernardo Pereira Moreira
- Biomedical Research Center Seltersberg (BFS), Institute of Parasitology, Justus Liebig University Giessen, 35392 Giessen, Germany; (B.P.M.); (M.H.W.W.); (S.H.); (C.G.G.)
| | - Michael H. W. Weber
- Biomedical Research Center Seltersberg (BFS), Institute of Parasitology, Justus Liebig University Giessen, 35392 Giessen, Germany; (B.P.M.); (M.H.W.W.); (S.H.); (C.G.G.)
| | - Simone Haeberlein
- Biomedical Research Center Seltersberg (BFS), Institute of Parasitology, Justus Liebig University Giessen, 35392 Giessen, Germany; (B.P.M.); (M.H.W.W.); (S.H.); (C.G.G.)
| | - Annika S. Mokosch
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, 35392 Giessen, Germany; (A.S.M.); (B.S.)
| | - Bernhard Spengler
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, 35392 Giessen, Germany; (A.S.M.); (B.S.)
| | - Christoph G. Grevelding
- Biomedical Research Center Seltersberg (BFS), Institute of Parasitology, Justus Liebig University Giessen, 35392 Giessen, Germany; (B.P.M.); (M.H.W.W.); (S.H.); (C.G.G.)
| | - Franco H. Falcone
- Biomedical Research Center Seltersberg (BFS), Institute of Parasitology, Justus Liebig University Giessen, 35392 Giessen, Germany; (B.P.M.); (M.H.W.W.); (S.H.); (C.G.G.)
- Correspondence:
| |
Collapse
|
41
|
Vaccines for Human Schistosomiasis: Recent Progress, New Developments and Future Prospects. Int J Mol Sci 2022; 23:ijms23042255. [PMID: 35216369 PMCID: PMC8879820 DOI: 10.3390/ijms23042255] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 12/18/2022] Open
Abstract
Schistosomiasis, caused by human trematode blood flukes (schistosomes), remains one of the most prevalent and serious of the neglected tropical parasitic diseases. Currently, treatment of schistosomiasis relies solely on a single drug, the anthelmintic praziquantel, and with increased usage in mass drug administration control programs for the disease, the specter of drug resistance developing is a constant threat. Vaccination is recognized as one of the most sustainable options for the control of any pathogen, but despite the discovery and reporting of numerous potentially promising schistosome vaccine antigens, to date, no schistosomiasis vaccine for human or animal deployment is available. This is despite the fact that Science ranked such an intervention as one of the top 10 vaccines that need to be urgently developed to improve public health globally. This review summarizes current progress of schistosomiasis vaccines under clinical development and advocates the urgent need for the establishment of a revolutionary and effective anti-schistosome vaccine pipeline utilizing cutting-edge technologies (including developing mRNA vaccines and exploiting CRISPR-based technologies) to provide novel insight into future vaccine discovery, design, manufacture and deployment.
Collapse
|
42
|
Al-Naseri A, Al-Absi S, Mahana N, Tallima H, El Ridi R. Protective immune potential of multiple antigenic peptide (MAP) constructs comprising peptides that are shared by several cysteine peptidases against Schistosoma mansoni infection in mice. Mol Biochem Parasitol 2022; 248:111459. [PMID: 35041897 DOI: 10.1016/j.molbiopara.2022.111459] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/31/2021] [Accepted: 01/13/2022] [Indexed: 10/19/2022]
Abstract
In vaccine trials, Schistosoma mansoni cathepsin B1 (SmCB1), helminth cathepsins of the L family (e.g., SmCL3), and papain consistently induce highly significant reductions in challenge worm burden and egg viability, but generated no additive protective effects when used in combination. The protective capacity of the cysteine peptidases is associated with modest (SmCB1) and poor (cathepsins L) production of cytokines and antibodies, essentially of the type 2 axis, and is only marginally reduced upon use of proteolytically inactive enzymes. In this work, peptides shared by SmCB1, cathepsins of the L family, papain and other allergens were selected, synthesized as tetrabranched multiple antigen peptide constructs (MAP-1 and MAP-2), and used in two independent experiments to immunize outbred mice, in parallel with papain. The two peptides elicited significant (P < 0.05) reduction in challenge worm burden when compared to unimmunized mice, albeit lower than that achieved by papain. Protection was associated with modest serum type 2 cytokines and antibody levels in MAP-, and papain-immunized mice. Immunization with papain also elicited a reduction in parasite egg load, viability, and granuloma numbers in liver and intestine. MAP-1 and MAP-2 immunogens displayed some opposite effects- MAP-1 leading to higher egg numbers with poor vitality, whereas MAP-2 immunization yielded fewer eggs. Cysteine peptidase thus appear to carry peptides that elicit opposing outcomes, highlighting the difficulty of reaching fully fledged protection, unless a vaccine is based on carefully selected peptides and combined with an effective adjuvant.
Collapse
Affiliation(s)
- Aya Al-Naseri
- Zoology Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Samar Al-Absi
- Zoology Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Noha Mahana
- Zoology Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Hatem Tallima
- Zoology Department, Faculty of Science, Cairo University, Giza, 12613, Egypt; Department of Chemistry, School of Science and Engineering, American University in Cairo, New Cairo, 11835, Cairo, Egypt.
| | - Rashika El Ridi
- Zoology Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| |
Collapse
|
43
|
Adegboye O, Field MA, Kupz A, Pai S, Sharma D, Smout MJ, Wangchuk P, Wong Y, Loiseau C. Natural-Product-Based Solutions for Tropical Infectious Diseases. Clin Microbiol Rev 2021; 34:e0034820. [PMID: 34494873 PMCID: PMC8673330 DOI: 10.1128/cmr.00348-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
About half of the world's population and 80% of the world's biodiversity can be found in the tropics. Many diseases are specific to the tropics, with at least 41 diseases caused by endemic bacteria, viruses, parasites, and fungi. Such diseases are of increasing concern, as the geographic range of tropical diseases is expanding due to climate change, urbanization, change in agricultural practices, deforestation, and loss of biodiversity. While traditional medicines have been used for centuries in the treatment of tropical diseases, the active natural compounds within these medicines remain largely unknown. In this review, we describe infectious diseases specific to the tropics, including their causative pathogens, modes of transmission, recent major outbreaks, and geographic locations. We further review current treatments for these tropical diseases, carefully consider the biodiscovery potential of the tropical biome, and discuss a range of technologies being used for drug development from natural resources. We provide a list of natural products with antimicrobial activity, detailing the source organisms and their effectiveness as treatment. We discuss how technological advancements, such as next-generation sequencing, are driving high-throughput natural product screening pipelines to identify compounds with therapeutic properties. This review demonstrates the impact natural products from the vast tropical biome have in the treatment of tropical infectious diseases and how high-throughput technical capacity will accelerate this discovery process.
Collapse
Affiliation(s)
- Oyelola Adegboye
- Public Health and Tropical Medicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia
- World Health Organization Collaborating Center for Vector-Borne and Neglected Tropical Diseases, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Matt A. Field
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia
- Garvin Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Andreas Kupz
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia
| | - Saparna Pai
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia
| | - Dileep Sharma
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- College of Medicine & Dentistry, James Cook University, Cairns, QLD, Australia
| | - Michael J. Smout
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia
| | - Phurpa Wangchuk
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia
| | - Yide Wong
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia
| | - Claire Loiseau
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
44
|
Tissue Pathogens and Cancers: A Review of Commonly Seen Manifestations in Histo- and Cytopathology. Pathogens 2021; 10:pathogens10111410. [PMID: 34832566 PMCID: PMC8624235 DOI: 10.3390/pathogens10111410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 02/07/2023] Open
Abstract
Tissue pathogens are commonly encountered in histopathology and cytology practice, where they can present as either benign mimickers of malignancy or true malignancies. The aim of this review is to provide a timely synthesis of our understanding of these tissue pathogens, with an emphasis on pertinent diagnostic conundrums associated with the benign mimickers of malignancy that can be seen with viral infections and those which manifest as granulomas. The oncogenic pathogens, including viruses, bacteria, and parasites, are then discussed with relationship to their associated malignancies. Although not exhaustive, the epidemiology, clinical manifestations, pathogenesis, and histological findings are included, along with a short review of emerging therapies.
Collapse
|
45
|
Yang ZY, Liu ZH, Zhang YN, Li C, Liu L, Pu WJ, Xie SQ, Xu J, Xia CM. Synergistic effect of combination chemotherapy with praziquantel and DW-3-15 for Schistosoma japonicum in vitro and in vivo. Parasit Vectors 2021; 14:550. [PMID: 34702326 PMCID: PMC8549225 DOI: 10.1186/s13071-021-05065-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/12/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Schistosomiasis is a debilitating and neglected tropical disease for which praziquantel (PZQ) remains the first-choice drug for treatment and control of the disease. In our previous studies, we found that the patented compound DW-3-15 (patent no. ZL201110142538.2) displayed significant and stabilized antiparasitic activity through a mechanism that might be distinct from PZQ. Here, we investigated the antischistosomal efficacy of PZQ combined with DW-3-15 against schistosomula and adult worms of Schistosoma japonicum in vitro and in vivo, to verify whether there was a synergistic effect of the two compounds. METHODS The antischistosomal efficacy of PZQ combined with DW-3-15 in comparison with an untreated control and monotherapy group against schistosomula and adult worms was assessed both in vitro and in vivo. Parasitological studies, scanning electron microscopy, combination index, and histopathological analysis were used for the assessment. RESULTS The results showed significantly reduced viability of schistosomes, achieving 100% viability reduction for juveniles and males by combination chemotherapy using PZQ together with DW-3-15 in vitro. The combination index was 0.28, 0.27, and 0.53 at the higher concentration of PZQ combined with DW-3-15 against juveniles, males, and females, respectively, indicating that the two compounds display strong synergism. Scanning electron microscopy observations also demonstrated that the compound combination induced more severe and extensive alterations to the tegument and subtegument of S. japonicum than those with each compound alone. In vivo, compared with the single-compound-treated group, the group treated with the higher-dose combination demonstrated the best schistosomicidal efficacy, with significantly reduced worm burden, egg burden, and granuloma count and area, which was evident against schistosomula and adult worms. CONCLUSIONS Our study provides a potential novel chemotherapy for schistosomiasis caused by S. japonicum. It would improve the antischistosomal effect on schistosomula and adult worms of S. japonicum, and decrease individual dosages.
Collapse
Affiliation(s)
- Zi-Yin Yang
- Department of Parasitology, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123 China
| | - Zi-Hao Liu
- Department of Parasitology, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123 China
| | - Ya-Nan Zhang
- Department of Parasitology, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123 China
| | - Chen Li
- Department of Parasitology, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123 China
| | - Lei Liu
- Department of Parasitology, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123 China
| | - Wen-Jie Pu
- Department of Parasitology, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123 China
| | - Shi-Qi Xie
- Department of Parasitology, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123 China
| | - Jing Xu
- Department of Parasitology, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123 China
| | - Chao-Ming Xia
- Department of Parasitology, Medical College of Soochow University, 199 Renai Road, Suzhou, 215123 China
| |
Collapse
|
46
|
Heat-killed Propionibacterium acnes augment the protective effect of 28-kDa glutathione S-transferases antigen against Schistosoma mansoni infection. Acta Trop 2021; 222:106033. [PMID: 34224719 DOI: 10.1016/j.actatropica.2021.106033] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 05/13/2021] [Accepted: 06/27/2021] [Indexed: 02/06/2023]
Abstract
Sm28GST is one of the candidate antigens for Schistosoma mansoni vaccine. Already Sm28GST vaccine formulations have shown to be protective against S. mansoni infection. Currently, efforts have been put into finding an adjuvant to enhance the immunity induced by Sm28GST. In the present work, we investigated whether heat-killed Propionibacterium acnes can be served as a potential adjuvant for recombinant Sm28GST (rSm28GST) antigen. As the results showed, P. acnes successfully modulated the Th1 humoral immune response induced by rSm28GST. Stronger Th1 cytokines responses were also observed in mice immunized with P. acnes-adjuvanted rSm28GST. Immunization of mice with P. acnes-adjuvanted rSm28GST was able to reduce worm burden and hepatic egg burden by 54.20 and 73.61%. Reduced granuloma size and count, as well as improved liver histology, were seen in P. acnes-adjuvanted rSm28GST immunized mice. These data suggest that P. acnes may evoke a stronger rSm28GST-induced immune response, higher resistance to S. mansoni infection, and more profound protection against S. mansoni-induced liver damages.
Collapse
|
47
|
Lam HYP, Huang SP, Liang TR, Wu WJ, Cheng PC, Chang KC, Peng SY. Increased immunogenicity and protection of recombinant Sm14 antigens by heat-killed Cutibacterium acnes in BALB/c mice infected with Schistosoma mansoni. Parasitol Int 2021; 86:102446. [PMID: 34481947 DOI: 10.1016/j.parint.2021.102446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 12/21/2022]
Abstract
After many years of the excessive use of praziquantel against Schistosoma mansoni (S. mansoni), it has already led to the development of drug resistance. While schistosomiasis is still affecting millions of people every year, vaccination may be one realistic alternative way to control the disease. Currently, S. mansoni 14-kDa fatty acid-binding protein (Sm14) has shown promising results as a vaccine antigen. Yet, the use of an adjuvant may be necessary to further increase the effectiveness of the vaccine. Herein, we investigated the potential of using heat-killed Cutibacterium acnes (C. acnes) as an adjuvant for recombinant Sm14 (rSm14). Immunization of mice with C. acnes-adjuvanted rSm14 showed increased humoral immune responses, compared with mice immunized with rSm14 alone. Additionally, C. acnes-adjuvanted rSm14 vaccination provided higher protection to mice against S. mansoni infection and liver injuries. These results suggest that C. acnes increases the immunogenicity of rSm14, which leads to better protection against S. mansoni infection. Therefore, heat-killed C. acnes may be a promising adjuvant to use with rSm14.
Collapse
Affiliation(s)
- Ho Yin Pekkle Lam
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan; Institute of Medical Sciences, Tzu Chi University, Hualien 97004, Taiwan
| | - Shu-Ping Huang
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien 97004, Taiwan; Department of Laboratory Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City 60002, Taiwan
| | - Ting-Ruei Liang
- PhD Program in Pharmacology and Toxicology, Tzu Chi University, Hualien 97004, Taiwan
| | - Wen-Jui Wu
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Po-Ching Cheng
- Department of Molecular Parasitology and Tropical Diseases, School of Medicine, Taipei Medical University, Taipei 110, Taiwan; Research Center of International Tropical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Kai-Chih Chang
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien 97004, Taiwan.
| | - Shih-Yi Peng
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan; Institute of Medical Sciences, Tzu Chi University, Hualien 97004, Taiwan; PhD Program in Pharmacology and Toxicology, Tzu Chi University, Hualien 97004, Taiwan.
| |
Collapse
|
48
|
McManus DP. The Search for a Schistosomiasis Vaccine: Australia's Contribution. Vaccines (Basel) 2021; 9:vaccines9080872. [PMID: 34451997 PMCID: PMC8402410 DOI: 10.3390/vaccines9080872] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/05/2021] [Accepted: 08/05/2021] [Indexed: 01/10/2023] Open
Abstract
Schistosomiasis, a neglected tropical disease caused by parasitic flatworms of the genus Schistosoma, results in considerable human morbidity in sub-Saharan Africa, in particular, but also parts of the Middle East, South America, and Southeast Asia. The anti-schistosome drug praziquantel is efficacious and safe against the adult parasites of all Schistosoma species infecting humans; however, it does not prevent reinfection and the development of drug resistance is a constant concern. The need to develop an effective vaccine is of great importance if the health of many in the developing world is to be improved. Indeed, vaccination, in combination with other public health measures, can provide an invaluable tool to achieve lasting control, leading to schistosomiasis elimination. Australia has played a leading role in schistosomiasis vaccine research over many years and this review presents an overview of some of the significant contributions made by Australian scientists in this important area.
Collapse
Affiliation(s)
- Donald P McManus
- Molecular Parasitology Laboratory, Infectious Diseases Division, QIMR Berghofer Medical Research Institute, Brisbane 4006, Australia
| |
Collapse
|
49
|
Mitochondria as a potential target for the development of prophylactic and therapeutic drugs against Schistosoma mansoni infection. Antimicrob Agents Chemother 2021; 65:e0041821. [PMID: 34339272 DOI: 10.1128/aac.00418-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Emergence of parasites resistant to praziquantel, the only therapeutic agent, and its ineffectiveness as a prophylactic agent (inactive against the migratory/juvenile Schistosoma mansoni), makes the development of new antischistosomal drugs urgent. The parasite's mitochondrion is an attractive target for drug development because this organelle is essential for survival throughout the parasite's life cycle. We investigated the effects of 116 compounds against Schistosoma mansoni cercariae motility that have been reported to affect mitochondria-related processes in other organisms. Next, eight compounds plus two controls (mefloquine and praziquantel) were selected and assayed against motility of schistosomula (in vitro) and adults (ex vivo). Prophylactic and therapeutic assays were performed using infected mouse models. Inhibition of oxygen consumption rate (OCR) was assayed using Seahorse XFe24 Analyzer. All selected compounds showed excellent prophylactic activity, reducing the worm burden in the lungs to less than 15% that obtained in the vehicle control. Notably, ascofuranone showed the highest activity with a 98% reduction of the worm burden, suggesting the potential for development of ascofuranone as a prophylactic agent. The worm burden of infected mice with S. mansoni at the adult stage was reduced by more than 50% in mice treated with mefloquine, nitazoxanide, amiodarone, ascofuranone, pyrvinium pamoate, or plumbagin. Moreover, adult mitochondrial OCR was severely inhibited by ascofuranone, atovaquone, and nitazoxanide, while pyrvinium pamoate inhibited both mitochondrial and non-mitochondrial OCRs. These results demonstrate that the mitochondria of S. mansoni are feasible target for drug development.
Collapse
|
50
|
Panzner U, Excler JL, Kim JH, Marks F, Carter D, Siddiqui AA. Recent Advances and Methodological Considerations on Vaccine Candidates for Human Schistosomiasis. FRONTIERS IN TROPICAL DISEASES 2021; 2:719369. [PMID: 39280170 PMCID: PMC11392908 DOI: 10.3389/fitd.2021.719369] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/18/2024] Open
Abstract
Schistosomiasis remains a neglected tropical disease of major public health concern with high levels of morbidity in various parts of the world. Although considerable efforts in implementing mass drug administration programs utilizing praziquantel have been deployed, schistosomiasis is still not contained. A vaccine may therefore be an essential part of multifaceted prevention control efforts. In the 1990s, a joint United Nations committee promoting parasite vaccines shortlisted promising candidates including for schistosomiasis discussed below. After examining the complexity of immune responses in human hosts infected with schistosomes, we review and discuss the antigen design and preclinical and clinical development of the four leading vaccine candidates: Sm-TSP-2 in Phase 1b/2b, Sm14 in Phase 2a/2b, Sm-p80 in Phase 1 preparation, and Sh28GST in Phase 3. Our assessment of currently leading vaccine candidates revealed some methodological issues that preclude a fair comparison between candidates and the rationale to advance in clinical development. These include (1) variability in animal models - in particular non-human primate studies - and predictive values of each for protection in humans; (2) lack of consensus on the assessment of parasitological and immunological parameters; (3) absence of reliable surrogate markers of protection; (4) lack of well-designed parasitological and immunological natural history studies in the context of mass drug administration with praziquantel. The controlled human infection model - while promising and unique - requires validation against efficacy outcomes in endemic settings. Further research is also needed on the impact of advanced adjuvants targeting specific parts of the innate immune system that may induce potent, protective and durable immune responses with the ultimate goal of achieving meaningful worm reduction.
Collapse
Affiliation(s)
- Ursula Panzner
- International Vaccine Institute, Seoul, South Korea
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | | | - Jerome H Kim
- International Vaccine Institute, Seoul, South Korea
| | - Florian Marks
- International Vaccine Institute, Seoul, South Korea
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- University of Antananarivo, Antananarivo, Madagascar
| | | | - Afzal A Siddiqui
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|