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
|
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
|
3
|
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
|
4
|
Tendler M, Savino W, Almeida MSDS. Developing anti-helminth vaccines for people and cattle: (For)seeing the one health approach in action. One Health 2023; 17:100616. [PMID: 37664170 PMCID: PMC10468795 DOI: 10.1016/j.onehlt.2023.100616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/09/2023] [Accepted: 08/16/2023] [Indexed: 09/05/2023] Open
Affiliation(s)
- Miriam Tendler
- Laboratory of Anti-Helminth Vaccine Research and Development, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Wilson Savino
- Laboratory on Thymus Research, Brazilian National Institute of Science and Technology on Neuroimmunomodulation, Rio de Janeiro Research Network on Neuroinflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | | |
Collapse
|
5
|
Chayé MAM, Gasan TA, Ozir-Fazalalikhan A, Scheenstra MR, Zawistowska-Deniziak A, van Hengel ORJ, Gentenaar M, Manurung MD, Harvey MR, Codée JDC, Chiodo F, Heijke AM, Kalinowska A, van Diepen A, Hensbergen PJ, Yazdanbakhsh M, Guigas B, Hokke CH, Smits HH. Schistosoma mansoni egg-derived thioredoxin and Sm14 drive the development of IL-10 producing regulatory B cells. PLoS Negl Trop Dis 2023; 17:e0011344. [PMID: 37363916 DOI: 10.1371/journal.pntd.0011344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 05/02/2023] [Indexed: 06/28/2023] Open
Abstract
During chronic schistosome infections, a complex regulatory network is induced to regulate the host immune system, in which IL-10-producing regulatory B (Breg) cells play a significant role. Schistosoma mansoni soluble egg antigens (SEA) are bound and internalized by B cells and induce both human and mouse IL-10 producing Breg cells. To identify Breg-inducing proteins in SEA, we fractionated SEA by size exclusion chromatography and found 6 fractions able to induce IL-10 production by B cells (out of 18) in the high, medium and low molecular weight (MW) range. The high MW fractions were rich in heavily glycosylated molecules, including multi-fucosylated proteins. Using SEA glycoproteins purified by affinity chromatography and synthetic glycans coupled to gold nanoparticles, we investigated the role of these glycan structures in inducing IL-10 production by B cells. Then, we performed proteomics analysis on active low MW fractions and identified a number of proteins with putative immunomodulatory properties, notably thioredoxin (SmTrx1) and the fatty acid binding protein Sm14. Subsequent splenic murine B cell stimulations and hock immunizations with recombinant SmTrx1 and Sm14 showed their ability to dose-dependently induce IL-10 production by B cells both in vitro and in vivo. Identification of unique Breg cells-inducing molecules may pave the way to innovative therapeutic strategies for inflammatory and auto-immune diseases.
Collapse
Affiliation(s)
- Mathilde A M Chayé
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Thomas A Gasan
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Maaike R Scheenstra
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anna Zawistowska-Deniziak
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Parasitology, Institute of Functional Biology and Ecology, Faculty of Biology, University of Warsaw, Warsaw, Poland
- Department of Immunology, Institute of Functional Biology and Ecology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Oscar R J van Hengel
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Max Gentenaar
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mikhael D Manurung
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Michael R Harvey
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Jeroen D C Codée
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Fabrizio Chiodo
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
- Italian National Research Council, Institute of Biomolecular Chemistry, Pozzuoli, Italy
| | - Anouk M Heijke
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Alicja Kalinowska
- Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Warsaw, Poland
- Museum and Institute of Zoology, Polish Academy of Sciences, Warsaw, Poland
| | - Angela van Diepen
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Paul J Hensbergen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Maria Yazdanbakhsh
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bruno Guigas
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Cornelis H Hokke
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hermelijn H Smits
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
6
|
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
|
7
|
Development of the Sm14/GLA-SE Schistosomiasis Vaccine Candidate: An Open, Non-Placebo-Controlled, Standardized-Dose Immunization Phase Ib Clinical Trial Targeting Healthy Young Women. Vaccines (Basel) 2022; 10:vaccines10101724. [PMID: 36298589 PMCID: PMC9607179 DOI: 10.3390/vaccines10101724] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 09/24/2022] [Accepted: 10/12/2022] [Indexed: 12/05/2022] Open
Abstract
We report the successful closure of Phase I clinical trials, comprising Phases Ia and Ib, of the vaccine candidate against human schistosomiasis: the Schistosoma mansoni 14 kDa fatty acid-binding protein (Sm14) + glucopyranosyl lipid A in squalene emulsion (GLA-SE). Shown here are the results of Phase Ib, an open, non-placebo-controlled, standardized-dose immunization trial involving 10 healthy 18-49-year-old women. Fifty micrograms of the Sm14 protein plus 10 µg GLA-SE per dose was given intramuscularly thrice at 30-day intervals. Participants were assessed clinically, biochemically, and immunologically for up to 120 days. In preambular experiments involving vaccinated pregnant female rabbits, we did not find any toxicological features in either the offspring or mothers, and the vaccine induced adaptive immunity in the animals. In women, no adverse events were observed, and vaccination induced high titers of anti-Sm14 serum IgG antibody production. Vaccination also elicited robust cytokine responses, with increased TNFα, IFNγ, and IL-2 profiles in all vaccinees on days 90 and 120. The completion of Phase I clinical trials, which were performed to the highest standards set by Good Clinical Research Practice (GCP) standards, and preclinical data in pregnant rabbits enabled the vaccine candidate to proceed to Phase II clinical trials in endemic areas.
Collapse
|
8
|
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: 25] [Impact Index Per Article: 12.5] [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
|
9
|
Barbosa MMF, Kanno AI, Barazzone GC, Rodriguez D, Pancakova V, Trentini M, Faquim-Mauro EL, Freitas AP, Khouri MI, Lobo-Silva J, Goncalves VM, Schenkman RPF, Tanizaki MM, Boraschi D, Malley R, Farias LP, Leite LCC. Robust Immune Response Induced by Schistosoma mansoni TSP-2 Antigen Coupled to Bacterial Outer Membrane Vesicles. Int J Nanomedicine 2021; 16:7153-7168. [PMID: 34712047 PMCID: PMC8548026 DOI: 10.2147/ijn.s315786] [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: 04/22/2021] [Accepted: 09/22/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose The use of adjuvants can significantly strengthen a vaccine’s efficacy. We sought to explore the immunization efficacy of bacterial outer membrane vesicles (OMVs) displaying the Schistosoma mansoni antigen, SmTSP-2, through a biotin-rhizavidin coupling approach. The rationale is to exploit the nanoparticulate structure and the adjuvant properties of OMVs to induce a robust antigen-specific immune response, in light of developing new vaccines against S. mansoni. Materials and Methods OMVs were obtained from Neisseria lactamica and conjugated with biotin. The recombinant SmTSP-2 in fusion with the biotin-binding protein rhizavidin (rRzvSmTSP-2) was produced in E. coli and coupled to biotinylated OMVs to generate an OMV complex displaying SmTSP-2 on the membrane surface (OMV:rSmTSP-2). Transmission electron microscopy (TEM) and dynamic light scattering analysis were used to determine particle charge and size. The immunogenicity of the vaccine complex was evaluated in C57BL/6 mice. Results The rRzvSmTSP-2 protein was successfully coupled to biotinylated OMVs and purified by size-exclusion chromatography. The OMV:rSmTSP-2 nanoparticles showed an average size of 200 nm, with zeta potential around – 28 mV. Mouse Bone Marrow Dendritic Cells were activated by the nanoparticles as determined by increased expression of the co-stimulatory molecules CD40 and CD86, and the proinflammatory cytokines (TNF-α, IL-6 and IL-12) or IL-10. Splenocytes of mice immunized with OMV:rSmTSP-2 nanoparticles reacted to an in vitro challenge with SmTSP-2 with an increased production of IL-6, IL-10 and IL-17 and displayed a higher number of CD4+ and CD8+ T lymphocytes expressing IFN-γ, IL-4 and IL-2, compared to mice immunized with the antigen alone. Immunization of mice with OMV:rSmTSP-2 induced a 100-fold increase in specific anti-SmTSP-2 IgG antibody titers, as compared to the group receiving the recombinant rSmTSP-2 protein alone or even co-administered with unconjugated OMV. Conclusion Our results demonstrate that the SmTSP-2 antigen coupled with OMVs is highly immunogenic in mice, supporting the potential effectiveness of this platform for improved antigen delivery in novel vaccine strategies.
Collapse
Affiliation(s)
- Mayra M F Barbosa
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil.,Programa de Pós-Graduação Interunidades em Biotecnologia, Universidade de São Paulo, São Paulo, Brazil
| | - Alex I Kanno
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil
| | - Giovana C Barazzone
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil
| | - Dunia Rodriguez
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil
| | - Violeta Pancakova
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil.,Université Claude Bernard Lyon 1 (UCBL1), Villeurbanne, 69100, France
| | - Monalisa Trentini
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil
| | | | - Amanda P Freitas
- Laboratório de Imunopatologia, Instituto Butantan, São Paulo, Brazil
| | - Mariana I Khouri
- Laboratório de Biomarcadores e Inflamação, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Jessica Lobo-Silva
- Laboratório de Biomarcadores e Inflamação, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Viviane M Goncalves
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil
| | | | - Martha M Tanizaki
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil
| | - Diana Boraschi
- Istituto di Biochimica e Biologia Cellulare, Consiglio Nazionale delle Ricerche, Napoli, Italy.,Stazione Zoologica Anton Dohrn, Napoli, Italy.,Shenzhen Institute of Advanced Technologies (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Richard Malley
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Leonardo P Farias
- Laboratório de Biomarcadores e Inflamação, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Luciana C C Leite
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil
| |
Collapse
|
10
|
Schistosoma mansoni egg-derived extracellular vesicles: A promising vaccine candidate against murine schistosomiasis. PLoS Negl Trop Dis 2021; 15:e0009866. [PMID: 34644290 PMCID: PMC8544836 DOI: 10.1371/journal.pntd.0009866] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/25/2021] [Accepted: 09/30/2021] [Indexed: 02/05/2023] Open
Abstract
Extracellular vesicles (EVs) are protein-loaded nano-scaled particles that are extracellularly released by eukaryotes and prokaryotes. Parasite's EVs manipulate the immune system, making them probable next-generation vaccines. Schistosomal EVs carry different proteins of promising immunizing potentials. For evaluating the immune-protective role of Schistosoma mansoni (S. mansoni) egg-derived EVs against murine schistosomiasis, EVs were isolated from cultured S. mansoni eggs by progressive sequential cooling ultra-centrifugation technique. Isolated EVs were structurally identified using transmission electron microscope and their protein was quantified by Lowry's technique. Experimental mice were subcutaneously immunized with three doses of 20 μg EVs (with or without alum adjuvant); every two weeks, then were challenged with S. mansoni cercariae two weeks after the last immunizing dose. Six weeks post infection, mice were sacrificed for vaccine candidate assessment. EVs protective efficacy was evaluated through parasitological, histopathological, and immunological parameters. Results showed significant reduction of tegumentally deranged adult worms, hepatic and intestinal egg counts reduction by 46.58%, 93.14% and 93.17% respectively, accompanied by remarkable amelioration of sizes, numbers and histopathology of hepatic granulomata mediated by high interferon gamma (IFN γ) and antibody level. Using sera from vaccinated mice, the molecular weight of EVs' protein components targeted by the antibody produced was recognized by western immunoblot. Results revealed two bands of ~ 14 KDa and ~ 21 KDa, proving that EVs are able to stimulate specific antibodies response. In conclusion, the present study highlighted the role of S. mansoni-egg derived EVs as a potential vaccine candidate against murine schistosomiasis mansoni.
Collapse
|
11
|
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
|
12
|
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
|
13
|
Qokoyi NK, Masamba P, Kappo AP. Proteins as Targets in Anti-Schistosomal Drug Discovery and Vaccine Development. Vaccines (Basel) 2021; 9:762. [PMID: 34358178 PMCID: PMC8310332 DOI: 10.3390/vaccines9070762] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 01/23/2023] Open
Abstract
Proteins hardly function in isolation; they form complexes with other proteins or molecules to mediate cell signaling and control cellular processes in various organisms. Protein interactions control mechanisms that lead to normal and/or disease states. The use of competitive small molecule inhibitors to disrupt disease-relevant protein-protein interactions (PPIs) holds great promise for the development of new drugs. Schistosome invasion of the human host involves a variety of cross-species protein interactions. The pathogen expresses specific proteins that not only facilitate the breach of physical and biochemical barriers present in skin, but also evade the immune system and digestion of human hemoglobin, allowing for survival in the host for years. However, only a small number of specific protein interactions between the host and parasite have been functionally characterized; thus, in-depth understanding of the molecular mechanisms of these interactions is a key component in the development of new treatment methods. Efforts are now focused on developing a schistosomiasis vaccine, as a proposed better strategy used either alone or in combination with Praziquantel to control and eliminate this disease. This review will highlight protein interactions in schistosomes that can be targeted by specific PPI inhibitors for the design of an alternative treatment to Praziquantel.
Collapse
Affiliation(s)
| | | | - Abidemi Paul Kappo
- Molecular Biophysics and Structural Biology (MBSB) Group, Department of Biochemistry, Kingsway Campus, University of Johannesburg, Auckland Park 2006, South Africa; (N.K.Q.); (P.M.)
| |
Collapse
|
14
|
Panzner U, Boissier J. Natural Intra- and Interclade Human Hybrid Schistosomes in Africa with Considerations on Prevention through Vaccination. Microorganisms 2021; 9:microorganisms9071465. [PMID: 34361901 PMCID: PMC8305539 DOI: 10.3390/microorganisms9071465] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 12/04/2022] Open
Abstract
Causal agents of schistosomiasis are dioecious, digenean schistosomes affecting mankind in 76 countries. Preventive measures are manifold but need to be complemented by vaccination for long-term protection; vaccine candidates in advanced pre-clinical/clinical stages include Sm14, Sm-TSP-2/Sm-TSP-2Al®, Smp80/SchistoShield®, and Sh28GST/Bilhvax®. Natural and anthropogenic changes impact on breaking species isolation barriers favoring introgressive hybridization, i.e., allelic exchange among gene pools of sympatric, interbreeding species leading to instant large genetic diversity. Phylogenetic distance matters, thus the less species differ phylogenetically the more likely they hybridize. PubMed and Embase databases were searched for publications limited to hybridale confirmation by mitochondrial cytochrome c oxidase (COX) and/or nuclear ribosomal internal transcribed spacer (ITS). Human schistosomal hybrids are predominantly reported from West Africa with clustering in the Senegal River Basin, and scattering to Europe, Central and Eastern Africa. Noteworthy is the dominance of Schistosoma haematobium interbreeding with human and veterinary species leading due to hybrid vigor to extinction and homogenization as seen for S. guineensis in Cameroon and S. haematobium in Niger, respectively. Heterosis seems to advantage S. haematobium/S. bovis interbreeds with dominant S. haematobium-ITS/S. bovis-COX1 profile to spread from West to East Africa and reoccur in France. S. haematobium/S. mansoni interactions seen among Senegalese and Côte d’Ivoirian children are unexpected due to their high phylogenetic distance. Detecting pure S. bovis and S. bovis/S. curassoni crosses capable of infecting humans observed in Corsica and Côte d’Ivoire, and Niger, respectively, is worrisome. Taken together, species hybridization urges control and preventive measures targeting human and veterinary sectors in line with the One-Health concept to be complemented by vaccination protecting against transmission, infection, and disease recurrence. Functional and structural diversity of naturally occurring human schistosomal hybrids may impact current vaccine candidates requiring further research including natural history studies in endemic areas targeted for clinical trials.
Collapse
Affiliation(s)
- Ursula Panzner
- Division of Infectious Diseases and Tropical Medicine, Ludwig Maximilian University of Munich, 80539 Munich, Germany
- Swiss Tropical and Public Health Institute, University of Basel, 4002 Basel, Switzerland
- Correspondence: ; Tel.: +49-176-6657-2910
| | - Jerome Boissier
- IHPE, University of Montpellier, CNRS, Ifremer, University of Perpignan, 66860 Perpignan, France;
| |
Collapse
|
15
|
Zerna G, Spithill TW, Beddoe T. Current Status for Controlling the Overlooked Caprine Fasciolosis. Animals (Basel) 2021; 11:1819. [PMID: 34207215 PMCID: PMC8235714 DOI: 10.3390/ani11061819] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/16/2021] [Accepted: 06/16/2021] [Indexed: 01/17/2023] Open
Abstract
The disease fasciolosis is caused by the liver flukes Fasciola hepatica and F. gigantica, which infect a wide range of mammals and production livestock, including goats. These flatworm parasites are globally distributed and predicted to cost the livestock industry a now conservative USD 3 billion per year in treatment and lowered on-farm productivity. Infection poses a risk to animal welfare and results in lowered fertility rates and reduced production yields of meat, milk and wool. This zoonotic disease is estimated to infect over 600 million animals and up to 2.4 million humans. Current and future control is threatened with the global emergence of flukes resistant to anthelmintics. Drug resistance calls for immediate on-farm parasite management to ensure treatments are effective and re-infection rates are kept low, while a sustainable long-term control method, such as a vaccine, is being developed. Despite the recent expansion of the goat industry, particularly in developing countries, there are limited studies on goat-focused vaccine control studies and the effectiveness of drug treatments. There is a requirement to collate caprine-specific fasciolosis knowledge. This review will present the current status of liver fluke caprine infections and potential control methods for application in goat farming.
Collapse
Affiliation(s)
| | | | - Travis Beddoe
- Department of Animal, Plant and Soil Sciences and Centre for AgriBioscience, La Trobe University, Bundoora, Victoria 3083, Australia; (G.Z.); (T.W.S.)
| |
Collapse
|
16
|
Al-Naseri A, Al-Absi S, El Ridi R, Mahana N. A comprehensive and critical overview of schistosomiasis vaccine candidates. J Parasit Dis 2021; 45:557-580. [PMID: 33935395 PMCID: PMC8068781 DOI: 10.1007/s12639-021-01387-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 03/31/2021] [Indexed: 12/11/2022] Open
Abstract
A digenetic platyhelminth Schistosoma is the causative agent of schistosomiasis, one of the neglected tropical diseases that affect humans and animals in numerous countries in the Middle East, sub-Saharan Africa, South America and China. Several control methods were used for prevention of infection or treatment of acute and chronic disease. Mass drug administration led to reduction in heavy-intensity infections and morbidity, but failed to decrease schistosomiasis prevalence and eliminate transmission, indicating the need to develop anti-schistosome vaccine to prevent infection and parasite transmission. This review summarizes the efficacy and protective capacity of available schistosomiasis vaccine candidates with some insights and future prospects.
Collapse
Affiliation(s)
- Aya Al-Naseri
- Zoology Department, Faculty of Science, Cairo Univesity, Giza, 12613 Egypt
| | - Samar Al-Absi
- Zoology Department, Faculty of Science, Cairo Univesity, Giza, 12613 Egypt
| | - Rashika El Ridi
- Zoology Department, Faculty of Science, Cairo Univesity, Giza, 12613 Egypt
| | - Noha Mahana
- Zoology Department, Faculty of Science, Cairo Univesity, Giza, 12613 Egypt
| |
Collapse
|
17
|
Driciru E, Koopman JPR, Cose S, Siddiqui AA, Yazdanbakhsh M, Elliott AM, Roestenberg M. Immunological Considerations for Schistosoma Vaccine Development: Transitioning to Endemic Settings. Front Immunol 2021; 12:635985. [PMID: 33746974 PMCID: PMC7970007 DOI: 10.3389/fimmu.2021.635985] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/11/2021] [Indexed: 12/16/2022] Open
Abstract
Despite mass drug administration programmes with praziquantel, the prevalence of schistosomiasis remains high. A vaccine is urgently needed to control transmission of this debilitating disease. As some promising schistosomiasis vaccine candidates are moving through pre-clinical and clinical testing, we review the immunological challenges that these vaccine candidates may encounter in transitioning through the clinical trial phases in endemic settings. Prior exposure of the target population to schistosomes and other infections may impact vaccine response and efficacy and therefore requires considerable attention. Schistosomes are known for their potential to induce T-reg/IL-10 mediated immune suppression in populations which are chronically infected. Moreover, endemicity of schistosomiasis is focal whereby target and trial populations may exhibit several degrees of prior exposure as well as in utero exposure which may increase heterogeneity of vaccine responses. The age dependent distribution of exposure and development of acquired immunity, and general differences in the baseline immunological profile, adds to the complexity of selecting suitable trial populations. Similarly, prior or concurrent infections with other parasitic helminths, viral and bacterial infections, may alter immunological responses. Consequently, treatment of co-infections may benefit the immunogenicity of vaccines and may be considered despite logistical challenges. On the other hand, viral infections leave a life-long immunological imprint on the human host. Screening for serostatus may be needed to facilitate interpretation of vaccine responses. Co-delivery of schistosome vaccines with PZQ is attractive from a perspective of implementation but may complicate the immunogenicity of schistosomiasis vaccines. Several studies have reported PZQ treatment to induce both transient and long-term immuno-modulatory effects as a result of tegument destruction, worm killing and subsequent exposure of worm antigens to the host immune system. These in turn may augment or antagonize vaccine immunogenicity. Understanding the complex immunological interactions between vaccine, co-infections or prior exposure is essential in early stages of clinical development to facilitate phase 3 clinical trial design and implementation policies. Besides well-designed studies in different target populations using schistosome candidate vaccines or other vaccines as models, controlled human infections could also help identify markers of immune protection in populations with different disease and immunological backgrounds.
Collapse
Affiliation(s)
- Emmanuella Driciru
- Immunomodulation and Vaccines Programme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | | | - Stephen Cose
- Immunomodulation and Vaccines Programme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Afzal A. Siddiqui
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University School of Medicine, Lubbock, TX, United States
- Department of Internal Medicine, Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Maria Yazdanbakhsh
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Alison M. Elliott
- Immunomodulation and Vaccines Programme, Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Meta Roestenberg
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
18
|
Sanches RCO, Tiwari S, Ferreira LCG, Oliveira FM, Lopes MD, Passos MJF, Maia EHB, Taranto AG, Kato R, Azevedo VAC, Lopes DO. Immunoinformatics Design of Multi-Epitope Peptide-Based Vaccine Against Schistosoma mansoni Using Transmembrane Proteins as a Target. Front Immunol 2021; 12:621706. [PMID: 33737928 PMCID: PMC7961083 DOI: 10.3389/fimmu.2021.621706] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 02/08/2021] [Indexed: 12/17/2022] Open
Abstract
Schistosomiasis remains a serious health issue nowadays for an estimated one billion people in 79 countries around the world. Great efforts have been made to identify good vaccine candidates during the last decades, but only three molecules reached clinical trials so far. The reverse vaccinology approach has become an attractive option for vaccine design, especially regarding parasites like Schistosoma spp. that present limitations for culture maintenance. This strategy also has prompted the construction of multi-epitope based vaccines, with great immunological foreseen properties as well as being less prone to contamination, autoimmunity, and allergenic responses. Therefore, in this study we applied a robust immunoinformatics approach, targeting S. mansoni transmembrane proteins, in order to construct a chimeric antigen. Initially, the search for all hypothetical transmembrane proteins in GeneDB provided a total of 584 sequences. Using the PSORT II and CCTOP servers we reduced this to 37 plasma membrane proteins, from which extracellular domains were used for epitope prediction. Nineteen common MHC-I and MHC-II binding epitopes, from eight proteins, comprised the final multi-epitope construct, along with suitable adjuvants. The final chimeric multi-epitope vaccine was predicted as prone to induce B-cell and IFN-γ based immunity, as well as presented itself as stable and non-allergenic molecule. Finally, molecular docking and molecular dynamics foresee stable interactions between the putative antigen and the immune receptor TLR 4. Our results indicate that the multi-epitope vaccine might stimulate humoral and cellular immune responses and could be a potential vaccine candidate against schistosomiasis.
Collapse
Affiliation(s)
- Rodrigo C. O. Sanches
- Laboratório de Biologia Molecular, Universidade Federal de São João del-Rei, Divinópolis, Brazil
| | - Sandeep Tiwari
- Programa de Pós-Graduação em Bioinformática, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Laís C. G. Ferreira
- Laboratório de Biologia Molecular, Universidade Federal de São João del-Rei, Divinópolis, Brazil
| | - Flávio M. Oliveira
- Laboratório de Biologia Molecular, Universidade Federal de São João del-Rei, Divinópolis, Brazil
| | - Marcelo D. Lopes
- Laboratório de Biologia Molecular, Universidade Federal de São João del-Rei, Divinópolis, Brazil
| | - Maria J. F. Passos
- Laboratório de Biologia Molecular, Universidade Federal de São João del-Rei, Divinópolis, Brazil
| | - Eduardo H. B. Maia
- Laboratório de Química Farmacêutica Medicinal, Universidade Federal de São João del-Rei, Divinópolis, Brazil
- Centro Federal de Educação Tecnológica de Minas Gerais (CEFET-MG), Divinópolis, Brazil
| | - Alex G. Taranto
- Laboratório de Química Farmacêutica Medicinal, Universidade Federal de São João del-Rei, Divinópolis, Brazil
| | - Rodrigo Kato
- Programa de Pós-Graduação em Bioinformática, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Vasco A. C. Azevedo
- Programa de Pós-Graduação em Bioinformática, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Debora O. Lopes
- Laboratório de Biologia Molecular, Universidade Federal de São João del-Rei, Divinópolis, Brazil
| |
Collapse
|
19
|
Molehin AJ. Current Understanding of Immunity Against Schistosomiasis: Impact on Vaccine and Drug Development. Res Rep Trop Med 2020; 11:119-128. [PMID: 33173371 PMCID: PMC7646453 DOI: 10.2147/rrtm.s274518] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 09/29/2020] [Indexed: 12/17/2022] Open
Abstract
Schistosomiasis is a neglected tropical disease inflicting significant morbidity in humans worldwide. The disease is caused by infections with a parasitic trematode belonging to the genus Schistosoma. Over 250 million people are currently infected globally, with an estimated disability-adjusted life-years of 1.9 million attributed to the disease. Current understanding, based on several immunological studies using experimental and human models of schistosomiasis, reveals that complex immune mechanisms play off each other in the acquisition of immune resistance to infection/reinfection. Nevertheless, the precise characteristics of these responses, the specific antigens against which they are elicited, and how these responses are intricately regulated are still being investigated. What is apparent is that immunity to schistosome infections develops slowly and over a prolonged period of time, augmented by the death of adult worms occurring naturally or by praziquantel therapy. In this review, aspects of immunity to schistosomiasis, host–parasite interactions and their impact on schistosomiasis vaccine development are discussed.
Collapse
Affiliation(s)
- Adebayo J Molehin
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.,Center for Tropical Medicine and Infectious Diseases, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
20
|
Current status and future prospects of protein vaccine candidates against Schistosoma mansoni infection. Parasite Epidemiol Control 2020; 11:e00176. [PMID: 32923703 PMCID: PMC7475110 DOI: 10.1016/j.parepi.2020.e00176] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 06/04/2020] [Accepted: 08/16/2020] [Indexed: 12/30/2022] Open
Abstract
Schistosomiasis is an acute and chronic tropical parasitic disease caused by blood dwelling worm of the genus Schistosoma. It is the most destructive disease globally and is a major cause of morbidity and mortality for developing countries. Three main species of schistosomes infect human beings from which S. mansoni is the most common and widespread. Over the last several decades, chemotherapy using praziquantel has been a commonly used strategy for the treatment and control of schistosomiasis. However, control programs focused exclusively on chemotherapy have been challenging because of the frequency and rapidity of reinfection and these programs were expensive. Thus, new schistosomiasis control strategies will be needed. Vaccination strategy would be an ideal tool for a significant and sustainable reduction in the transmission and disease burden of schistosomiasis. An effective anti schistosome vaccine would greatly contribute to decreasing schistosomiasis-associated morbidity via protective immune responses leading to reduced worm burdens and decreased egg production. Vaccine development is a long process that can take decades. There have been three candidate vaccines that have been produced by Good Manufacturing Procedure and entered human clinical trials for S. mansoni are Sm14, SmTSP-2, and Sm-p80. Other candidates that are in pre-clinical trials at various stages include paramyosin, Sm29, SmKI-1, and Sm23. Since the growth of several new technologies, including genomics, transcriptomics, microarrays, immunomic profiling, and proteomics, have helped in the identification of promising new target schistosome antigens. Therefore, this review considers the present status of protein vaccine candidates against Schistosoma mansoni and provides some insight on prospects vaccine design and discovery.
Collapse
Key Words
- AE, Asparaginyl Endopeptidase
- Ab, Antibody
- Ag, Antigen
- CB, Cathepsin B
- CD, Cathepsin D
- CL3, Cathepsin L3
- DNA, Deoxyribonucleic Acid
- FA, Fatty Acid
- FABP, Fatty Acid Binding Protein
- GLA-Alum, Glucopyranosyl Lipid A Formulated in Aluminum
- GLA-SE, Glucopyranosyl Lipid Adjuvant Stable Emulsion
- IFN-γ, Interferon Gamma
- IL, Interleukin
- Ig, Immunoglobulin
- KI, Kunitz Type Protease Inhibitor
- LcP, Lipid Core Peptide
- Pmy, Paramyosin
- Protein vaccine
- Schistosoma mansoni
- Schistosomiasis
- Sm, Schistosoma mansoni
- TSP, Tetraspanins
- Th, T-helper Cells
- Vaccine candidates
- WHO, World Health Organization
Collapse
|
21
|
Immunogenicity and protection induced by recombinant Toxocara canis proteins in a murine model of toxocariasis. Vaccine 2020; 38:4762-4772. [PMID: 32451213 DOI: 10.1016/j.vaccine.2020.04.072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 02/06/2023]
Abstract
Toxocariasis, a natural helminth infection of dogs and cats caused by Toxocara canis and T. cati, respectively, that are transmitted to mammals, including humans. Infection control is based currently on periodic antihelmintic treatment and there is a need for the development of vaccines to prevent this infection. MATERIALS AND METHODS Eight potential vaccine candidate T. canis recombinant proteins were identified by in silico (rTcGPRs, rTcCad, rTcVcan, rTcCyst) and larval proteomics (rTES26, rTES32, rMUC-3 and rCTL-4) analyses. Immunogenicity and protection against infectious challenge for seven of these antigens were determined in a murine model of toxocariasis. C57BL/6 female mice were immunized with each of or combinations of recombinant antigens prior to challenge with 500 T. canis embryonated eggs. Levels of specific antibodies (IgG, IgG1, IgG2a and IgE) in sera and cytokines (IL-5, INF-ɣ and IL-10) produced by antigens-stimulated splenocytes, were measured. Presence of specific antibodies to the molecules was measured in sera of T. canis-seropositive dogs and humans. RESULTS All seven molecules were immunogenic in immunized mice; all stimulated significantly elevated levels of specific IgG, IgG1 or IgG2a and six were associated with elevated levels of specific IgE; all induced elevated production of IFN- ɣ and IL-10 by splenocytes, but only the in silico-identified membrane-associated recombinants (rTcCad, rTcVcan, and rTcCyst) induced significantly increased IL-5 production. Vaccination with two of the latter (rTcCad and rTcVcan) reduced larval loads in the T. canis challenged mice by 54.3% and 53.9% (P < 0.0001), respectively, compared to unimmunized controls. All seven recombinants were recognized by T. canis-seropositive dog and human sera. CONCLUSION The identification of vaccine targets by in silico analysis was an effective strategy to identify immunogenic T. canis proteins capable of reducing larval burdens following challenge with the parasite. Two recombinant proteins, rTcCad and rTcVcan, were identified as promising vaccine candidates for canine toxocariasis.
Collapse
|
22
|
Sorgi S, Bonezi V, Dominguez MR, Gimenez AM, Dobrescu I, Boscardin S, Nakaya HI, Bargieri DY, Soares IS, Silveira ELV. São Paulo School of Advanced Sciences on Vaccines: an overview. J Venom Anim Toxins Incl Trop Dis 2020; 26:e20190061. [PMID: 32362926 PMCID: PMC7187638 DOI: 10.1590/1678-9199-jvatitd-2019-0061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 02/21/2020] [Indexed: 01/08/2023] Open
Abstract
Two years ago, we held an exciting event entitled the São Paulo School of Advanced Sciences on Vaccines (SPSASV). Sixty-eight Ph.D. students, postdoctoral fellows and independent researchers from 37 different countries met at the Mendes Plaza Hotel located in the city of Santos, SP - Brazil to discuss the challenges and the new frontiers of vaccinology. The SPSASV provided a critical and comprehensive view of vaccine research from basics to the current state-of-the-art techniques performed worldwide. For 10 days, we discussed all the aspects of vaccine development in 36 lectures, 53 oral presentations and 2 poster sessions. At the end of the course, participants were further encouraged to present a model of a grant proposal related to vaccine development against individual pathogens. Among the targeted pathogens were viruses (Chikungunya, HIV, RSV, and Influenza), bacteria (Mycobacterium tuberculosis and Streptococcus pyogenes), parasites (Plasmodium falciparum or Plasmodium vivax), and the worm Strongyloides stercoralis. This report highlights some of the knowledge shared at the SPSASV.
Collapse
Affiliation(s)
- Sara Sorgi
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
- Dipartimento di Biotecnologie Mediche, Universita’ degli Studi di Siena, Siena, Italia
| | - Vivian Bonezi
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Mariana R. Dominguez
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Alba Marina Gimenez
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Irina Dobrescu
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Silvia Boscardin
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Helder I. Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Daniel Y. Bargieri
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Irene S. Soares
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Eduardo L. V. Silveira
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| |
Collapse
|
23
|
Proteomic analysis of two populations of Schistosoma mansoni-derived extracellular vesicles: 15k pellet and 120k pellet vesicles. Mol Biochem Parasitol 2020; 236:111264. [DOI: 10.1016/j.molbiopara.2020.111264] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/21/2020] [Accepted: 01/29/2020] [Indexed: 12/20/2022]
|
24
|
Reamtong O, Simanon N, Thiangtrongjit T, Limpanont Y, Chusongsang P, Chusongsang Y, Anuntakarun S, Payungporn S, Phuphisut O, Adisakwattana P. Proteomic analysis of adult Schistosoma mekongi somatic and excretory-secretory proteins. Acta Trop 2020; 202:105247. [PMID: 31672487 DOI: 10.1016/j.actatropica.2019.105247] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 07/30/2019] [Accepted: 10/25/2019] [Indexed: 12/19/2022]
Abstract
Schistosoma mekongi is a causative agent of human schistosomiasis. There is limited knowledge of the molecular biology of S. mekongi and very few studies have examined drug targets, vaccine candidates and diagnostic biomarkers for S. mekongi. To explore the biology of S. mekongi, computational as well as experimental approaches were performed on S. mekongi males and females to identify excretory-secretory (ES) proteins and proteins that are differentially expressed between genders. According to bioinformatic prediction, the S. mekongi ES product was approximately 4.7% of total annotated transcriptome sequences. The classical secretory pathway was the main process to secrete proteins. Mass spectrometry-based quantification of male and female adult S. mekongi proteins was performed. We identified 174 and 156 differential expression of proteins in male and female worms, respectively. The dominant male-biased proteins were involved in actin filament-based processes, microtubule-based processes, biosynthetic processes and homeostatic processes. The major female-biased proteins were related to biosynthetic processes, organelle organization and signal transduction. An experimental approach identified 88 proteins in the S. mekongi secretome. The S. mekongi ES proteins mainly contributed to nutrient uptake, essential substance supply and host immune evasion. This research identifies proteins in the S. mekongi secretome and provides information on ES proteins that are differentially expressed between S. mekongi genders. These findings will contribute to S. mekongi drug and vaccine development. In addition, the study enhances our understanding of basic S. mekongi biology.
Collapse
|
25
|
Molehin AJ. Schistosomiasis vaccine development: update on human clinical trials. J Biomed Sci 2020; 27:28. [PMID: 31969170 PMCID: PMC6977295 DOI: 10.1186/s12929-020-0621-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 01/16/2020] [Indexed: 01/13/2023] Open
Abstract
Schistosomiasis causes significant levels of morbidity and mortality in many geographical regions of the world. The disease is caused by infections with parasitic blood flukes known as schistosomes. The control of schistosomiasis over the last several decades has been centered on the mass drug administration (MDA) of praziquantel (PZQ), which is the only drug currently available for treatment. Despite the concerted efforts of MDA programs, the prevalence and transmission of schistosomiasis has remained largely unchecked due to the fact that PZQ is ineffective against juvenile schistosomes, does not prevent re-infection and the emergence of PZQ-resistant parasites. In addition, other measures such as the water, sanitation and hygiene programs and snail intermediate hosts control have had little to no impact. These drawbacks indicate that the current control strategies are severely inadequate at interrupting transmission and therefore, implementation of other control strategies are required. Ideally, an efficient vaccine is what is needed for long term protection thereby eliminating the current efforts of repeated mass drug administration. However, the general consensus in the field is that the integration of a viable vaccine with MDA and other control measures offer the best chance of achieving the goal of schistosomiasis elimination. This review focuses on the present status of schistosomiasis vaccine candidates in different phases of human clinical trials and provide some insight into future vaccine discovery and design.
Collapse
Affiliation(s)
- Adebayo J Molehin
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX, 79430, USA. .,Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX, 79430, USA.
| |
Collapse
|
26
|
Schistosomiasis and hookworm infection in humans: Disease burden, pathobiology and anthelmintic vaccines. Parasitol Int 2020; 75:102051. [PMID: 31911156 DOI: 10.1016/j.parint.2020.102051] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 11/01/2019] [Accepted: 01/01/2020] [Indexed: 12/12/2022]
Abstract
Helminth diseases are the ancient scourges of humans and their damages are 'silent and insidious'. Of the helminth infections, schistosomiasis and hookworm infection have a great impact. This review covers information regarding vaccine candidates against schistosomiasis and hookworms that reached at least up to the phase-1 trial and literatures regarding other vaccine candidates have been excluded. For clinical manifestations, all available literatures were included, and for epidemiology and global burden of the diseases (GBD), literatures only within 2000-2019 were included. Literatures were searched surfing various databases including PubMED, Google Scholar, and Science Direct and overall over 150 literatures were identified. Globally ~250 million people are suffering from schistosomiasis, resulting 1430 thousand DALY (disability adjusted life year) per year. On the other hand, about 1.3 billion people are infected with hookworm (HW), and according to WHO, ~878 million school-age children (SAC) are at risk. HW is estimated to cause 65,000 deaths annually, accounts for 845 thousand DALYs as well as to cause 6-35.3% loss in productivity. Despite tremendous efforts, very few anthelmintic vaccine candidates such as Na-GST-1, Na-APR-1 and Na-ASP-2 against HW, and Sm28GST/Sh28GST, Sm-p80, Sm14 and Sm-TSP-1/SmTSP-2 against schistosomiasis reached up to the clinical trials. More efforts are needed to achieve the WHO targets taken against the maladies.
Collapse
|
27
|
Masoori L, Meamar AR, Bandehpour M, Hemphill A, Razmjou E, Mokhtarian K, Roozbehani M, Badirzadeh A, Jalallou N, Akhlaghi L, Falak R. Fatty acid and retinol-binding protein: A novel antigen for immunodiagnosis of human strongyloidiasis. PLoS One 2019; 14:e0218895. [PMID: 31329601 PMCID: PMC6645452 DOI: 10.1371/journal.pone.0218895] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/11/2019] [Indexed: 11/19/2022] Open
Abstract
The tenacious human parasitic helminth Strongyloides stercoralis is a significant health problem worldwide. The current lack of a definitive diagnostic laboratory test to rule out this infection necessitates designing more specific diagnostic methods. Fatty acid and retinol-binding protein (FAR) plays a crucial role in the development and reproduction of nematodes. We generated a recombinant form of this protein and determined its applicability for immunodiagnosis of S. stercoralis. The L3 form of S. stercoralis was harvested and used for RNA extraction and cDNA synthesis. The coding sequence of S. stercoralis FAR (SsFAR) was cloned into pET28a(+) vector, expressed in E. coli BL21 and purified. ELISA and immunoblotting were employed to determine the specificity and sensitivity of rSsFAR using a set of defined sera. In addition, we analyzed the phylogenetic relationship of SsFAR with different FAR sequences from other nematodes. The cloned SsFAR had an open reading frame of 447 bp encoding 147 amino acids, with a deduced molecular mass of 19 kD. The SsFAR amino acid sequence was 93% identical to FAR of S. ratti. For differential immunodiagnosis of strongyloidiasis, rSsFAR exhibited 100% sensitivity and 97% specificity. However, cross-reactivity with FAR proteins of other parasites, namely Toxocara canis and Echinococcus granulosus, was noted. Our results provide a novel approach for immunodiagnosis of S. stercoralis infections using rSsFAR with reliable sensitivity and specificity.
Collapse
Affiliation(s)
- Leila Masoori
- Department of Parasitology and Mycology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Meamar
- Department of Parasitology and Mycology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- * E-mail: (ARM); (RF)
| | - Mojgan Bandehpour
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Andrew Hemphill
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Elham Razmjou
- Department of Parasitology and Mycology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Kobra Mokhtarian
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mona Roozbehani
- Department of Parasitology and Mycology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Badirzadeh
- Department of Parasitology and Mycology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nahid Jalallou
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Lame Akhlaghi
- Department of Parasitology and Mycology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Immunology Research center, Iran University of Medical Science, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- * E-mail: (ARM); (RF)
| |
Collapse
|
28
|
de Melo TT, Mendes MM, Alves CC, Carvalho GB, Fernandes VC, Pimenta DLF, de Moraes Mourão M, Gai F, Kalli M, Coelho A, de Azambuja Ribeiro RIM, Falcone FH, Pereira RADS, Fonseca CT. The Schistosoma mansoni cyclophilin A epitope 107-121 induces a protective immune response against schistosomiasis. Mol Immunol 2019; 111:172-181. [PMID: 31063938 DOI: 10.1016/j.molimm.2019.04.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/24/2019] [Accepted: 04/25/2019] [Indexed: 10/26/2022]
Abstract
Great efforts have been made to identify promising antigens and vaccine formulations against schistosomiasis. Among the previously described Schistosoma vaccine candidates, cyclophilins comprise an interesting antigen that could be used for vaccine formulations. Cyclophilin A is the target for the cyclosporine A, a drug with schistosomicide activity, and its orthologue from Schistosoma japonicum induces a protective immune response in mice. Although Schistosoma mansoni cyclophilin A also represents a promising target for anti-schistosome vaccines, its potential to induce protection has not been evaluated. In this study, we characterized the cyclophilin A (SmCyp), initially described as Smp17.7, analyzed its allergenic potential using in vitro functional assays, and evaluated its ability to induce protection in mice when administered as an antigen using different vaccine formulations and strategies. Results indicated that SmCyp could be successfully expressed by mammalian cells and bacteria. The recombinant protein did not promote IgE-reporter system activation in vitro, demonstrating its probable safety for use in vaccine formulations. T and B-cell epitopes were predicted in the SmCyp sequence, with two of them located within the active isomerase site. The most immunogenic antigen, SmCyp (107-121), was then used for immunization protocols. Immunization with the SmCyp gene or protein failed to reduce parasite burden but induced an immune response that modulated the granuloma area. In contrast, immunization with the synthetic peptide SmCyp (107-121) significantly reduced worm burden (48-50%) in comparison to control group, but did not regulate liver pathology. Moreover, the protection observed in mice immunized with the synthetic peptide was associated with the significant production of antibodies against the SmCyp (107-121) epitope. Therefore, in this study, we identified an epitope within the SmCyp sequence that induces a protective immune response against the parasite, thus representing a promising antigen that could be used for vaccine formulation against schistosomiasis.
Collapse
Affiliation(s)
- Tatiane Teixeira de Melo
- Laboratório de Biologia e Imunologia de Doenças Infeciosas e Parasitárias, Instituto René Rachou, Fiocruz-MG, Belo Horizonte, Minas Gerais, Brazil
| | - Mariana Moreira Mendes
- Laboratório de Biologia e Imunologia de Doenças Infeciosas e Parasitárias, Instituto René Rachou, Fiocruz-MG, Belo Horizonte, Minas Gerais, Brazil
| | - Clarice Carvalho Alves
- Laboratório de Biologia e Imunologia de Doenças Infeciosas e Parasitárias, Instituto René Rachou, Fiocruz-MG, Belo Horizonte, Minas Gerais, Brazil
| | - Gardênia Braz Carvalho
- Laboratório de Biologia e Imunologia de Doenças Infeciosas e Parasitárias, Instituto René Rachou, Fiocruz-MG, Belo Horizonte, Minas Gerais, Brazil
| | - Viviane Cristina Fernandes
- Laboratório de Biologia e Imunologia de Doenças Infeciosas e Parasitárias, Instituto René Rachou, Fiocruz-MG, Belo Horizonte, Minas Gerais, Brazil
| | - Deborah Laranjeira Ferreira Pimenta
- Laboratório de Biologia e Imunologia de Doenças Infeciosas e Parasitárias, Instituto René Rachou, Fiocruz-MG, Belo Horizonte, Minas Gerais, Brazil
| | - Marina de Moraes Mourão
- Laboratório de Helmintologia e Malacologia Médica, Instituto René Rachou, Fiocruz-MG, Belo Horizonte, Minas Gerais, Brazil
| | - Fatou Gai
- The School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Marina Kalli
- The School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Aline Coelho
- Laboratório de Patologia Experimental, Universidade Federal De São João Del Rei- Campus Divinópolis, Minas Gerais, Brazil
| | | | - Franco H Falcone
- The School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Rosiane Aparecida da Silva Pereira
- Laboratório de Biologia e Imunologia de Doenças Infeciosas e Parasitárias, Instituto René Rachou, Fiocruz-MG, Belo Horizonte, Minas Gerais, Brazil
| | - Cristina Toscano Fonseca
- Laboratório de Biologia e Imunologia de Doenças Infeciosas e Parasitárias, Instituto René Rachou, Fiocruz-MG, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
29
|
Tang CL, Pan Q, Xie YP, Xiong Y, Zhang RH, Huang J. Effect of Cytotoxic T-Lymphocyte Antigen-4 on the Efficacy of the Fatty Acid-Binding Protein Vaccine Against Schistosoma japonicum. Front Immunol 2019; 10:1022. [PMID: 31134084 PMCID: PMC6514142 DOI: 10.3389/fimmu.2019.01022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 04/23/2019] [Indexed: 01/13/2023] Open
Abstract
The present study evaluated the impact of blocking cytotoxic T-lymphocyte antigen-4 (CTLA-4) activity on the protective effect elicited by the fatty acid binding protein (FABP) vaccine against Schistosoma japonicum infection. Mice were randomly divided into uninfected, infected control, anti-CTLA-4 monoclonal antibody (anti-CTLA-4 mAb), FABP, and combination (anti-CTLA-4 mAb and FABP) groups. An assessment of the S. japonicum worm and egg burden in the infected mice revealed that the worm reduction-rate induced by FABP administration was increased from 26.58 to 54.61% by co-administration of the monoclonal anti-CTLA antibody (anti-CTLA-4 mAb). Furthermore, the regulatory T cell (Treg) percentage was significantly increased in mice after administration of the anti-CTLA-4 mAb, but not the FABP vaccine, and elevated levels of the cytokines interferon (IFN)-γ, interleukin (IL)-2, IL-4, and IL-5 were observed in infected mice that were administered the anti-CTLA-4 mAb. Notably, the diameter of egg granulomas in the anti-CTLA-4 mAb and combination groups was significantly increased compared to that observed in the infected control group. Together, these results suggest that co-administering the FABP vaccine and anti-CTLA-4 treatment may have synergistically increased the immunoprotective effect of the FABP vaccine by promoting T-helper 1-type immune responses, while incurring increased tissue damage.
Collapse
Affiliation(s)
- Chun-Lian Tang
- Wuchang Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Qun Pan
- Wuchang Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Ya-Ping Xie
- Wuchang Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Ying Xiong
- Wuchang Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Rong-Hui Zhang
- Wuchang Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Jin Huang
- Department of Clinical Laboratory, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
30
|
Tang CL, Zhang RH, Liu ZM, Jin H, He L. Effect of regulatory T cells on the efficacy of the fatty acid-binding protein vaccine against Schistosoma japonicum. Parasitol Res 2019; 118:559-566. [PMID: 30607606 DOI: 10.1007/s00436-018-6186-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 12/14/2018] [Indexed: 01/21/2023]
Abstract
Schistosomiasis is one of the most devastating parasitic diseases, making it imperative to develop efficient vaccines to control the causative flatworms called schistosomes. Regulatory T cells (Tregs) and the Th1 immune response have been implicated in the effectiveness of vaccines to control schistosomiasis, but the mechanisms underlying their effects are unclear. In this study, we evaluated the role of Tregs on the efficacy of the 14 kDa FABP (fatty acid-binding protein) vaccine against Schistosoma japonicum. BALB/c female mice were randomly divided into five groups: an uninfected group, infected control group, anti-CD25 monoclonal antibody (anti-CD25 mAb) group, FABP group, and combined anti-CD25 mAb and FABP group. Compared with FABP alone, a combined treatment with FABP and anti-CD25 mAb increased the rate of S. japonicum inhibition in mice from 30.3 to 56.08% and decreased the number of eggs per gram of liver. Compared with that of the infected control group, the percentage of Tregs in the spleen decreased significantly after single or combined treatment with FABP and anti-CD25 mAb, while it increased gradually in the anti-CD25 mAb group. Further, the secretion of Th1 cytokines, IFN-γ, and IL-2 increased in splenocytes in the anti-CD25 mAb group. Our results indicate that anti-CD25 mAb partially blocks Tregs and concomitantly enhances the Th1 type immune response, thereby enhancing the protective effect of the FABP vaccine.
Collapse
Affiliation(s)
- Chun-Lian Tang
- Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan, 430063, China
| | - Rong-Hui Zhang
- Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan, 430063, China
| | - Zhi-Ming Liu
- Wuchang Hospital affiliated to Wuhan University of Science and Technology, Wuhan, 430063, China
| | - Huang Jin
- Department of Clinical Laboratory, Wuhan Fourth Hospital; Pu'ai hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430033, China.
| | - Long He
- Department of Clinical Laboratory, Shanghai East Hospital; School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
31
|
Tendler M, Almeida MS, Vilar MM, Pinto PM, Limaverde-Sousa G. Current Status of the Sm14/GLA-SE Schistosomiasis Vaccine: Overcoming Barriers and Paradigms towards the First Anti-Parasitic Human(itarian) Vaccine. Trop Med Infect Dis 2018; 3:tropicalmed3040121. [PMID: 30469320 PMCID: PMC6306874 DOI: 10.3390/tropicalmed3040121] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/12/2018] [Accepted: 11/12/2018] [Indexed: 11/17/2022] Open
Abstract
Schistosomiasis, a disease historically associated with poverty, lack of sanitation and social inequality, is a chronic, debilitating parasitic infection, affecting hundreds of millions of people in endemic countries. Although chemotherapy is capable of reducing morbidity in humans, rapid re-infection demonstrates that the impact of drug treatment on transmission control or disease elimination is marginal. In addition, despite more than two decades of well-executed control activities based on large-scale chemotherapy, the disease is expanding in many areas including Brazil. The development of the Sm14/GLA-SE schistosomiasis vaccine is an emblematic, open knowledge innovation that has successfully completed phase I and phase IIa clinical trials, with Phase II/III trials underway in the African continent, to be followed by further trials in Brazil. The discovery and experimental phases of the development of this vaccine gathered a robust collection of data that strongly supports the ongoing clinical phase. This paper reviews the development of the Sm14 vaccine, formulated with glucopyranosyl lipid A (GLA-SE), from the initial experimental developments to clinical trials including the current status of phase II studies.
Collapse
Affiliation(s)
- Miriam Tendler
- FIOCRUZ-Instituto Oswaldo Cruz, Laboratório de Esquistossomose Experimental, Av. Brasil, 4365, Manguinhos, Rio de Janeiro 21045-900, Brazil.
| | - Marília S Almeida
- FIOCRUZ-Instituto Oswaldo Cruz, Laboratório de Esquistossomose Experimental, Av. Brasil, 4365, Manguinhos, Rio de Janeiro 21045-900, Brazil.
| | - Monica M Vilar
- FIOCRUZ-Instituto Oswaldo Cruz, Laboratório de Esquistossomose Experimental, Av. Brasil, 4365, Manguinhos, Rio de Janeiro 21045-900, Brazil.
| | - Patrícia M Pinto
- FIOCRUZ-Instituto Oswaldo Cruz, Laboratório de Esquistossomose Experimental, Av. Brasil, 4365, Manguinhos, Rio de Janeiro 21045-900, Brazil.
| | - Gabriel Limaverde-Sousa
- FIOCRUZ-Instituto Oswaldo Cruz, Laboratório de Esquistossomose Experimental, Av. Brasil, 4365, Manguinhos, Rio de Janeiro 21045-900, Brazil.
| |
Collapse
|
32
|
Aly I, ELnain G, Hamad RS, Kilany M, Ghramh HA, Alshehri A, Dajem SM, Ibrahim EH. DNA vaccination using recombinant Schistosoma mansoni fatty acid binding protein (smFABP) gene. Exp Parasitol 2018; 194:53-59. [PMID: 30266572 DOI: 10.1016/j.exppara.2018.09.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 07/23/2018] [Accepted: 09/23/2018] [Indexed: 12/17/2022]
Abstract
Schistosomiasis is a fatal disease that has a negative impact on health and economics. Praziquantel (PZQ) is the drug of choice for schistosomiasis treatment, but it has no prophylactic effect; therefore, vaccination is an essential requirement in schistosomiasis control. This work was carried out to investigate the possible effect of DNA vaccination against Schistosoma mansoni infection using recombinant S. mansoni fatty acid binding protein (rsmFABP). The smFABP gene was cloned into the eukaryotic expression vector pcDNAI/Amp in order to obtain an smFABP-pcDNAI recombinant plasmid (DNA vaccine) and was used for the intramuscular DNA vaccination of out-bread Swiss albino mice prior to infection with S. mansoni cercariae. Infected groups, either DNA vaccinated or unvaccinated, were treated with PZQ at week 6 post-infection. After 8 weeks post-infection, all mouse groups were sacrificed and parasitological, immunological and histopathological parameters were studied. DNA vaccinated mice showed a high titer of anti-smFABP-IgG antibodies and acquired significant protection (74.2%, p < 0.01) against S. mansoni infection, with a reduction in ova and granuloma counts. DNA vaccinated and PZQ treated animals had higher titers of anti-smFABP-IgG antibodies and decreased (87%, P < 0.001) parenchymal granulomas compared to the DNA vaccinated PZQ untreated group. Infected mice, either non DNA vaccinated or vaccinated, had very high collagen content and fibrous granulomas (74%) compared to the PZQ treated group (10.3% fibrous granuloma) and PZQ treated + DNA vaccinated group (0% fibrous granuloma). In conclusion, DNA vaccination had protective and anti-pathological effects in naive mice and greatly improved the pathological status in PZQ-treated animals, suggesting an immunological and pathological modulating effect of PZQ treatment.
Collapse
Affiliation(s)
- Ibrahim Aly
- Parasitology Laboratory, Theodor Bilharz Research Institute, Imbaba, P.O. Box 30, Giza, Egypt
| | - Gehan ELnain
- Parasitology Laboratory, Theodor Bilharz Research Institute, Imbaba, P.O. Box 30, Giza, Egypt; Natural Science, Mathan Science Program, University College, Abu Dhabi University, United Arab Emirates
| | - Rabab S Hamad
- Parasitology Laboratory, Theodor Bilharz Research Institute, Imbaba, P.O. Box 30, Giza, Egypt; Biological Science Department, King Faisal University, Saudi Arabia
| | - Mona Kilany
- Biology Department, Faculty of Sciences and Arts, King Khalid University, Dhahran Al Janoub, Saudi Arabia; Department of Microbiology, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt
| | - Hamed A Ghramh
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia; Unit of Bee Research and Honey Production, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia; Biology Department, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
| | - Aly Alshehri
- Biology Department, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
| | - Saad M Dajem
- Biology Department, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
| | - Essam H Ibrahim
- Biology Department, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia; Blood Products Quality Control and Research Department, National Organization for Research and Control of Biologicals, Cairo, Egypt.
| |
Collapse
|
33
|
Eguia FAP, Ramos HR, Kraschowetz S, Omote D, Ramos CRR, Ho PL, Carvalho E, Gonçalves VM. A new vector for heterologous gene expression in Escherichia coli with increased stability in the absence of antibiotic. Plasmid 2018; 98:22-30. [PMID: 30193910 DOI: 10.1016/j.plasmid.2018.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 08/28/2018] [Accepted: 08/30/2018] [Indexed: 01/12/2023]
Abstract
Expression vectors for industrial production should be stable and allow tight control of protein synthesis. This is necessary to ensure plasmid transmission to daughter cells in order to achieve a stable population capable of synthesizing high amounts of the target protein. A high-copy-number plasmid, pAE, was previously used for laboratory-scale production of recombinant human granulocyte colony-stimulating factor (rhG-CSF) and the Schistosoma mansoni fatty acid binding protein (rSm14), but it was unstable for large-scale production. Therefore, here we evaluated a new expression vector derived from pAE, pAR-KanI, which combines two plasmid replication strategies: a high-copy plasmid pUC origin of replication as pAE, and a par locus sequence derived from pSC101, which is typical of low copy plasmids, for rhG-CSF and rSm14 production in Escherichia coli. Clones bearing these constructs were cultivated in two complex media (2YT and auto-induction) and both yielded higher-than-95% resistant colonies, before and after induction, either with or without antibiotics. In 2YT medium, we obtained 244 μg/mL of rSm14, 181 μg/mL and 392 μg/mL for rhG-CSF, with and without glucose, respectively. In auto-induction medium without antibiotics, 147 μg/mL of rSm14 and 162 μg/mL of rhG-CSF were obtained. The new vector presented high stability for the production of both recombinant proteins in complex media in Escherichia coli, even in the absence of antibiotics, making the pAR-KanI a promising vector for industrial production of recombinant proteins.
Collapse
Affiliation(s)
- Fara Amelia Primelles Eguia
- Laboratório Especial de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, SP, Brazil; Programa Interunidades em Biotecnologia, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Henrique Roman Ramos
- Laboratório de Bacteriologia 2, Instituto Butantan, São Paulo, SP, Brazil; Departamento Saúde III, Universidade Nove de Julho, São Paulo, SP, Brazil
| | - Stefanie Kraschowetz
- Laboratório Especial de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, SP, Brazil; Programa Interunidades em Biotecnologia, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Daniel Omote
- Laboratório de Bacteriologia 2, Instituto Butantan, São Paulo, SP, Brazil
| | | | - Paulo Lee Ho
- Centro de Biotecnologia, Instituto Butantan, São Paulo, SP, Brazil; Divisão de Inovação, Instituto Butantan, São Paulo, SP, Brazil
| | - Eneas Carvalho
- Laboratório de Bacteriologia 2, Instituto Butantan, São Paulo, SP, Brazil
| | | |
Collapse
|
34
|
White Bear J, Long T, Skinner D, McKerrow JH. Predictions of novel Schistosoma mansoni - human protein interactions consistent with experimental data. Sci Rep 2018; 8:13092. [PMID: 30166569 PMCID: PMC6117258 DOI: 10.1038/s41598-018-31272-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 08/14/2018] [Indexed: 12/26/2022] Open
Abstract
Infection by the human blood fluke, Schistosoma mansoni involves a variety of cross-species protein- protein interactions. The pathogen expresses a diverse arsenal of proteins that facilitate the breach of physical and biochemical barriers present in skin evasion of the immune system, and digestion of human plasma proteins including albumin and hemoglobin, allowing schistosomes to reside in the host for years. However, only a small number of specific interactions between S. mansoni and human proteins have been identified. We present and apply a protocol that generates testable predictions of S. mansoni-human protein interactions. In this study, we have preliminary predictions of novel interactions between schistosome and human proteins relevant to infection and the ability of the parasite to evade the immune system. We applied a computational whole-genome comparative approach to predict potential S. mansoni-human protein interactions based on similarity to known protein complexes. We first predict S. mansoni -human protein interactions based on similarity to known protein complexes. Putative interactions were then scored and assessed using several contextual filters, including the use of annotation automatically derived from literature using a simple natural language processing methodology. Next, in vitro experiments were carried out between schistosome and host proteins to validate several prospective predictions. Our method predicted 7 out of the 10 previously known cross-species interactions involved in pathogenesis between S. mansoni and its human host. Interestingly, two novel putative interactions involving Schistosoma proteins, the cercarial elastase SmCE, and the adult tegument surface protein Sm29, were also predicted and experimentally characterized. Preliminary data suggest that elafin, a host endogenous serine protease inhibitor, may be a novel substrate for SmCE. Additionally, CD59, an inhibitor of the membrane attack complex, could interact with Sm29. Furthermore, the application framework provides an integrated methodology for investigation of host-pathogen interactions and an extensive source of orthogonal data for experimental analysis. We have made the predictions available for community perusal.
Collapse
Affiliation(s)
- J White Bear
- Department of Bioengineering and Therapeutic Sciences, Department of Pharmaceutical Chemistry, and California Institute for Quantitative Biosciences, University of California, San Francisco, CA, 94158, USA.
- Graduate Group in Bioinformatics, University of California, San Francisco, CA, 94158, USA.
- MIT Lincoln Laboratory 244 Wood St, Lexington, MA, USA.
| | - Thavy Long
- Department of Pathology and Sandler Center for Basic Research in Parasitic Diseases, University of California at San Francisco, San Francisco, California, 94158, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego 9500 Gilman Dr, La Jolla, CA, 92093, USA
- INRA - InTheRes - UMR 1436, Equipe Transporteurs Membranaires et Résistance, 180, Chemin de Tournefeuille, Toulouse, France
| | - Danielle Skinner
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego 9500 Gilman Dr, La Jolla, CA, 92093, USA
| | - James H McKerrow
- Department of Pathology and Sandler Center for Basic Research in Parasitic Diseases, University of California at San Francisco, San Francisco, California, 94158, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego 9500 Gilman Dr, La Jolla, CA, 92093, USA
| |
Collapse
|
35
|
Haçarız O, Sayers GP. Genererating a core cluster of Fasciola hepatica virulence and immunomodulation-related genes using a comparative in silico approach. Res Vet Sci 2018; 117:271-276. [DOI: 10.1016/j.rvsc.2017.12.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 12/20/2017] [Accepted: 12/27/2017] [Indexed: 01/24/2023]
|
36
|
Stutzer C, Richards SA, Ferreira M, Baron S, Maritz-Olivier C. Metazoan Parasite Vaccines: Present Status and Future Prospects. Front Cell Infect Microbiol 2018; 8:67. [PMID: 29594064 PMCID: PMC5859119 DOI: 10.3389/fcimb.2018.00067] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/26/2018] [Indexed: 12/21/2022] Open
Abstract
Eukaryotic parasites and pathogens continue to cause some of the most detrimental and difficult to treat diseases (or disease states) in both humans and animals, while also continuously expanding into non-endemic countries. Combined with the ever growing number of reports on drug-resistance and the lack of effective treatment programs for many metazoan diseases, the impact that these organisms will have on quality of life remain a global challenge. Vaccination as an effective prophylactic treatment has been demonstrated for well over 200 years for bacterial and viral diseases. From the earliest variolation procedures to the cutting edge technologies employed today, many protective preparations have been successfully developed for use in both medical and veterinary applications. In spite of the successes of these applications in the discovery of subunit vaccines against prokaryotic pathogens, not many targets have been successfully developed into vaccines directed against metazoan parasites. With the current increase in -omics technologies and metadata for eukaryotic parasites, target discovery for vaccine development can be expedited. However, a good understanding of the host/vector/pathogen interface is needed to understand the underlying biological, biochemical and immunological components that will confer a protective response in the host animal. Therefore, systems biology is rapidly coming of age in the pursuit of effective parasite vaccines. Despite the difficulties, a number of approaches have been developed and applied to parasitic helminths and arthropods. This review will focus on key aspects of vaccine development that require attention in the battle against these metazoan parasites, as well as successes in the field of vaccine development for helminthiases and ectoparasites. Lastly, we propose future direction of applying successes in pursuit of next generation vaccines.
Collapse
Affiliation(s)
- Christian Stutzer
- Tick Vaccine Group, Department of Genetics, University of Pretoria, Pretoria, South Africa
| | | | | | | | | |
Collapse
|
37
|
Mekonnen GG, Pearson M, Loukas A, Sotillo J. Extracellular vesicles from parasitic helminths and their potential utility as vaccines. Expert Rev Vaccines 2018; 17:197-205. [PMID: 29353519 DOI: 10.1080/14760584.2018.1431125] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Helminths are multicellular parasites affecting nearly three billion people worldwide. To orchestrate a parasitic existence, helminths secrete different molecules, either in soluble form or contained within extracellular vesicles (EVs). EVs are secreted by most cell types and organisms, and have varied roles in intercellular communication, including immune modulation and pathogenesis. AREAS COVERED In this review, we describe the nucleic acid and proteomic composition of EVs from helminths, with a focus on the protein vaccine candidates present on the EV surface membrane, and discuss the potential utility of helminth EVs and their constituent proteins in the fight against helminth infections. EXPERT COMMENTARY A significant number of proteins present in helminth-secreted EVs are known vaccine candidates. The characterization of helminth EV proteomes will shed light on host-pathogen interactions, facilitate the discovery of new diagnostic biomarkers, and provide a novel approach for the development of new control measures against helminth infections.
Collapse
Affiliation(s)
- Gebeyaw Getnet Mekonnen
- a Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , Australia.,b Department of Medical Parasitology , School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar , Gondar , Ethiopia
| | - Mark Pearson
- a Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , Australia
| | - Alex Loukas
- a Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , Australia
| | - Javier Sotillo
- a Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , Australia
| |
Collapse
|
38
|
de Souza C, Lopes MD, De Oliveira FM, Passos MJF, Ferreira LCG, Faria BF, Villar JAFP, Junior MC, Taranto AG, Dos Santos LL, Fonseca CT, de Oliveira Lopes D. Rational selection of immunodominant and preserved epitope Sm043300e from Schistosoma mansoni and design of a chimeric molecule for biotechnological purposes. Mol Immunol 2017; 93:133-143. [PMID: 29175593 DOI: 10.1016/j.molimm.2017.11.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/12/2017] [Accepted: 11/19/2017] [Indexed: 12/18/2022]
Abstract
Human schistosomiasis is a neglected tropical disease of great importance in public health. A large number of people are infected with schistosomiasis, making vaccine development and effective diagnosis important control strategies. A rational epitope prediction workflow using Schistosoma mansoni hypothetical proteins was previously presented by our group, and an improvement to that approach is presented here. Briefly, immunodominant epitopes from parasite membrane proteins were predicted by reverse vaccinology strategy with additional in silico analysis. Furthermore, epitope recognition was evaluated using sera of individuals infected with S. mansoni. The epitope that stood out in both in silico and in vitro assays was used to compose a rational chimeric molecule to improve immune response activation. Out of 2185 transmembrane proteins, four epitopes with high binding affinities for human and mouse MHCII molecules were selected through computational screening. These epitopes were synthesized to evaluate their ability to induce TCD4+ lymphocyte proliferation in mice. Sm204830e and Sm043300e induced significant TCD4+ proliferation. Both epitopes were submitted to enzyme-linked immunosorbent assay to evaluate their recognition by IgG antibodies from the sera of infected individuals, and epitope Sm043300 was significantly recognized in most sera samples. Epitope Sm043300 also showed good affinity for human MHCII molecules in molecular docking, and its sequence is curiously highly conserved in four S. mansoni proteins, all of which are described as G-protein-coupled receptors. In addition, we have demonstrated the feasibility of incorporating this epitope, which showed low similarity to human sequences, into a chimeric molecule. The stability of the molecule was evaluated by molecular modeling aimed at future molecule production for use in diagnosis and vaccination trials.
Collapse
Affiliation(s)
- Cláudia de Souza
- Laboratório de Biologia Molecular, Universidade Federal de São João del-Rei, Av. Sebastião Gonçalves Coelho, 400, Divinópolis, Minas Gerais, 35501-296, Brazil; Laboratório de Síntese Orgânica e Nanoestruturas, Universidade Federal de São João del-Rei, Divinópolis, Minas Gerais, Brazil, Brazil
| | - Marcelo Donizete Lopes
- Laboratório de Biologia Molecular, Universidade Federal de São João del-Rei, Av. Sebastião Gonçalves Coelho, 400, Divinópolis, Minas Gerais, 35501-296, Brazil; Laboratório de Síntese Orgânica e Nanoestruturas, Universidade Federal de São João del-Rei, Divinópolis, Minas Gerais, Brazil, Brazil
| | - Flávio Martins De Oliveira
- Laboratório de Biologia Molecular, Universidade Federal de São João del-Rei, Av. Sebastião Gonçalves Coelho, 400, Divinópolis, Minas Gerais, 35501-296, Brazil
| | - Maria Juliana Ferreira Passos
- Laboratório de Biologia Molecular, Universidade Federal de São João del-Rei, Av. Sebastião Gonçalves Coelho, 400, Divinópolis, Minas Gerais, 35501-296, Brazil; Laboratório de Síntese Orgânica e Nanoestruturas, Universidade Federal de São João del-Rei, Divinópolis, Minas Gerais, Brazil, Brazil
| | - Laís Cunha Grossi Ferreira
- Laboratório de Biologia Molecular, Universidade Federal de São João del-Rei, Av. Sebastião Gonçalves Coelho, 400, Divinópolis, Minas Gerais, 35501-296, Brazil
| | - Bruna Franciele Faria
- Laboratório de Modelagem Molecular, Universidade Federal de São João del-Rei, Divinópolis, Minas Gerais, Brazil, Brazil
| | | | - Moacyr Comar Junior
- Laboratório de Modelagem Molecular, Universidade Federal de São João del-Rei, Divinópolis, Minas Gerais, Brazil, Brazil
| | - Alex Guterres Taranto
- Laboratório de Modelagem Molecular, Universidade Federal de São João del-Rei, Divinópolis, Minas Gerais, Brazil, Brazil
| | - Luciana Lara Dos Santos
- Laboratório de Biologia Molecular, Universidade Federal de São João del-Rei, Av. Sebastião Gonçalves Coelho, 400, Divinópolis, Minas Gerais, 35501-296, Brazil
| | - Cristina Toscano Fonseca
- Grupo de Pesquisa em Biologia e Imunologia de doenças Infeciosas e Parasitária, Centro de Pesquisas René Rachou-Fundação Oswaldo Cruz, Av. Augusto de Lima, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Débora de Oliveira Lopes
- Laboratório de Biologia Molecular, Universidade Federal de São João del-Rei, Av. Sebastião Gonçalves Coelho, 400, Divinópolis, Minas Gerais, 35501-296, Brazil.
| |
Collapse
|
39
|
Floudas A, Cluxton CD, Fahel J, Khan AR, Saunders SP, Amu S, Alcami A, Fallon PG. Composition of the Schistosoma mansoni worm secretome: Identification of immune modulatory Cyclophilin A. PLoS Negl Trop Dis 2017; 11:e0006012. [PMID: 29073139 PMCID: PMC5681295 DOI: 10.1371/journal.pntd.0006012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/10/2017] [Accepted: 10/04/2017] [Indexed: 12/16/2022] Open
Abstract
The helminth Schistosoma mansoni modulates the infected host's immune system to facilitate its own survival, by producing excretory/secretory molecules that interact with a variety of the host's cell types including those of the immune system. Herein, we characterise the S. mansoni adult male worm secretome and identify 111 proteins, including 7 vaccine candidates and several molecules with potential immunomodulatory activity. Amongst the molecules present in the secretome, a 17-19kDa protein analogous to human cyclophilin A was identified. Given the ability of cyclophilin A to modulate the immune system by regulating antigen presenting cell activity, we sought to determine whether recombinant S. mansoni Cyclophilin A (rSmCypA) is capable of modulating bone-marrow derived dendritic cell (BMDC) and T cell responses under in vitro conditions. rSmCypA was enzymatically active and able to alter the pro-inflammatory cytokine profile of LPS-activated dendritic cells. rSmCypA also modulated DC function in the induction of CD4+ T cell proliferation with a preferential expansion of Treg cells. This work demonstrates the unique protein composition of the S. mansoni male worm secretome and immunomodulatory activity of S. mansoni Cyclophilin A.
Collapse
Affiliation(s)
- Achilleas Floudas
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Christopher D. Cluxton
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Julia Fahel
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Adnan R. Khan
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Sean P. Saunders
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Sylvie Amu
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Antonio Alcami
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Padraic G. Fallon
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
- * E-mail:
| |
Collapse
|
40
|
Characterization and functional analysis of fatty acid binding protein from the carcinogenic liver fluke, Opisthorchis viverrini. Parasitol Int 2017; 66:419-425. [DOI: 10.1016/j.parint.2016.04.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 03/27/2016] [Accepted: 04/29/2016] [Indexed: 01/29/2023]
|
41
|
Ramos-Benítez MJ, Ruiz-Jiménez C, Aguayo V, Espino AM. Recombinant Fasciola hepatica fatty acid binding protein suppresses toll-like receptor stimulation in response to multiple bacterial ligands. Sci Rep 2017; 7:5455. [PMID: 28710478 PMCID: PMC5511235 DOI: 10.1038/s41598-017-05735-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 06/02/2017] [Indexed: 01/07/2023] Open
Abstract
Recently, we reported that a native Fasciola hepatica fatty acid binding protein (FABP) termed Fh12 is a powerful anti-inflammatory protein capable of suppressing the LPS-induced expression of inflammatory markers in vivo and in vitro. Because the purification of a protein in native form is, in many situations not cost-beneficial and unsuitable for industrial grade scale-up, this study accomplished the task of optimizing the expression and purification of a recombinant form of FABP (Fh15). Additionally, we ascertained whether this molecule could exhibit a similar suppressive effect on TLR-stimulation and inflammatory cytokine expression from macrophages than those previously demonstrated for the native molecule. Results demonstrated that Fh15 suppresses the expression of IL-1β and TNFα in murine macrophages and THP1 Blue CD14 cells. Additionally, Fh15 suppress the LPS-induced TLR4 stimulation. This effect was not impaired by a thermal denaturing process or blocked by the presence of anti-Fh12 antibodies. Fh15 also suppressed the stimulation of various TLRs in response to whole bacteria extracts, suggesting that Fh15 could have a broad spectrum of action. These results support the possibility of using Fh15 as an excellent alternative for an anti-inflammatory drug in preclinical studies in the near future.
Collapse
Affiliation(s)
- Marcos J Ramos-Benítez
- University of Puerto Rico, Medical Sciences Campus, Department of Microbiology, PO BOX 365067, San Juan, Puerto Rico, 00936, USA
| | - Caleb Ruiz-Jiménez
- University of Puerto Rico, Medical Sciences Campus, Department of Microbiology, PO BOX 365067, San Juan, Puerto Rico, 00936, USA
| | - Vasti Aguayo
- University of Puerto Rico, Medical Sciences Campus, Department of Microbiology, PO BOX 365067, San Juan, Puerto Rico, 00936, USA
| | - Ana M Espino
- University of Puerto Rico, Medical Sciences Campus, Department of Microbiology, PO BOX 365067, San Juan, Puerto Rico, 00936, USA.
| |
Collapse
|
42
|
Damasceno L, Ritter G, Batt CA. Process development for production and purification of the Schistosoma mansoni Sm14 antigen. Protein Expr Purif 2017; 134:72-81. [DOI: 10.1016/j.pep.2017.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 02/22/2017] [Accepted: 04/03/2017] [Indexed: 10/19/2022]
|
43
|
Sotillo J, Doolan D, Loukas A. Recent advances in proteomic applications for schistosomiasis research: potential clinical impact. Expert Rev Proteomics 2016; 14:171-183. [DOI: 10.1080/14789450.2017.1271327] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Javier Sotillo
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Denise Doolan
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Alex Loukas
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
44
|
Tebeje BM, Harvie M, You H, Loukas A, McManus DP. Schistosomiasis vaccines: where do we stand? Parasit Vectors 2016; 9:528. [PMID: 27716365 PMCID: PMC5045607 DOI: 10.1186/s13071-016-1799-4] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 09/14/2016] [Indexed: 12/20/2022] Open
Abstract
Schistosomiasis, caused mainly by S. mansoni, S. haematobium and S. japonicum, continues to be a serious tropical disease and public health problem resulting in an unacceptably high level of morbidity in countries where it is endemic. Praziquantel, the only drug currently available for treatment, is unable to kill developing schistosomes, it does not prevent re-infection and its continued extensive use may result in the future emergence of drug-resistant parasites. This scenario provides impetus for the development and deployment of anti-schistosome vaccines to be used as part of an integrated approach for the prevention, control and eventual elimination of schistosomiasis. This review considers the present status of candidate vaccines for schistosomiasis, and provides some insight on future vaccine discovery and design.
Collapse
Affiliation(s)
- Biniam Mathewos Tebeje
- QIMR Berghofer Medical Research Institute, Brisbane, Australia. .,School of Public Health, University of Queensland, Brisbane, Australia. .,Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia.
| | - Marina Harvie
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Hong You
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Alex Loukas
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | | |
Collapse
|
45
|
Abstract
Schistosomiasis is a parasitic disease caused by helminths belonging to the Schistosoma genus. Approximately 700 million people are at risk of infection and 200 million people are currently infected. Schistosomiasis is the most important helminth infection, and treatment relies solely on the drug praziquantel. Worries of praziquantel resistance as well as high disease burden are only some of the justifications which support the development of a vaccine against schistosomiasis. To date, only 2 schistosome vaccines have made it into clinical trials: Sh28GST (Bilhvax) and Sm14. However, there are several vaccine candidates, such as TSP-2, sm-p8, and Sm-Cathepsin B, which are generating promising results in pre-clinical studies. Schistosomiasis vaccine development has been an uphill battle, and there are still several hurdles to overcome in the future. Fortunately, the research groups involved in the research for vaccine development have not abandoned their work. Furthermore, in the last few years, schistosomiasis has garnered some additional attention on a global scale due to its significant impact on public health.
Collapse
Affiliation(s)
- Alessandra Ricciardi
- a National Reference Center for Parasitoloy; Research Institute of the McGill University Health Center ; Montreal , Quebec , Canada.,b Department of Microbiology & Immunology ; McGill University ; Montreal , Quebec , Canada
| | - Momar Ndao
- a National Reference Center for Parasitoloy; Research Institute of the McGill University Health Center ; Montreal , Quebec , Canada.,b Department of Microbiology & Immunology ; McGill University ; Montreal , Quebec , Canada
| |
Collapse
|
46
|
Molehin AJ, Rojo JU, Siddiqui SZ, Gray SA, Carter D, Siddiqui AA. Development of a schistosomiasis vaccine. Expert Rev Vaccines 2016; 15:619-27. [PMID: 26651503 PMCID: PMC5070536 DOI: 10.1586/14760584.2016.1131127] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Schistosomiasis is a neglected tropical disease (NTD) of public health importance. Despite decades of implementation of mass praziquantel therapy programs and other control measures, schistosomiasis has not been contained and continues to spread to new geographic areas. A schistosomiasis vaccine could play an important role as part of a multifaceted control approach. With regards to vaccine development, many biological bottlenecks still exist: the lack of reliable surrogates of protection in humans; immune interactions in co-infections with other diseases in endemic areas; the potential risk of IgE responses to antigens in endemic populations; and paucity of appropriate vaccine efficacy studies in nonhuman primate models. Research is also needed on the role of modern adjuvants targeting specific parts of the innate immune system to tailor a potent and protective immune response for lead schistosome vaccine candidates with the long-term aim to achieve curative worm reduction. This review summarizes the current status of schistosomiasis vaccine development.
Collapse
Affiliation(s)
- Adebayo J. Molehin
- Department of Internal Medicine, Texas Tech University School of Medicine, Lubbock, Texas, USA
- Center of Tropical Medicine and Infectious Diseases, Texas Tech University School of Medicine, Lubbock, Texas, USA
| | - Juan U. Rojo
- Department of Internal Medicine, Texas Tech University School of Medicine, Lubbock, Texas, USA
- Center of Tropical Medicine and Infectious Diseases, Texas Tech University School of Medicine, Lubbock, Texas, USA
| | - Sabrina Z. Siddiqui
- Department of Internal Medicine, Texas Tech University School of Medicine, Lubbock, Texas, USA
- Center of Tropical Medicine and Infectious Diseases, Texas Tech University School of Medicine, Lubbock, Texas, USA
| | | | - Darrick Carter
- PAI Life Sciences, Washington, USA
- Infectious Disease Research Institute, Seattle, Washington, USA
| | - Afzal A. Siddiqui
- Department of Internal Medicine, Texas Tech University School of Medicine, Lubbock, Texas, USA
- Center of Tropical Medicine and Infectious Diseases, Texas Tech University School of Medicine, Lubbock, Texas, USA
| |
Collapse
|
47
|
Wilson RA, Li XH, Castro-Borges W. Do schistosome vaccine trials in mice have an intrinsic flaw that generates spurious protection data? Parasit Vectors 2016; 9:89. [PMID: 26888413 PMCID: PMC4756456 DOI: 10.1186/s13071-016-1369-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 02/09/2016] [Indexed: 12/23/2022] Open
Abstract
The laboratory mouse has been widely used to test the efficacy of schistosome vaccines and a long list of candidates has emerged from this work, many of them abundant internal proteins. These antigens do not have an additive effect when co-administered, or delivered as SWAP homogenate, a quarter of which comprises multiple candidates; the observed protection has an apparent ceiling of 40-50%. We contend that the low level of maturation of penetrating cercariae (~32% for Schistosoma mansoni) is a major limitation of the model since 68/100 parasites fail to mature in naïve mice due to natural causes. The pulmonary capillary bed is the obstacle encountered by schistosomula en route to the portal system. The fragility of pulmonary capillaries and their susceptibility to a cytokine-induced vascular leak syndrome have been documented. During lung transit schistosomula burst into the alveolar spaces, and possess only a limited capacity to re-enter tissues. The acquired immunity elicited by the radiation-attenuated (RA) cercarial vaccine relies on a pulmonary inflammatory response, involving cytokines such as IFNγ and TNFα, to deflect additional parasites into the alveoli. A principal difference between antigen vaccine protocols and the RA vaccine is the short interval between the last antigen boost and cercarial challenge of mice (often two weeks). Thus, after antigen vaccination, challenge parasites will reach the lungs when both activated T cells and cytokine levels are maximal in the circulation. We propose that "protection" in this situation is the result of physiological effects on the pulmonary blood vessels, increasing the proportion of parasites that enter the alveoli. This hypothesis will explain why internal antigens, which are unlikely to interact with the immune response in a living schistosomulum, plus a variety of heterologous proteins, can reduce the level of maturation in a non-antigen-specific way. These proteins are "successful" precisely because they have not been selected for immunological silence. The same arguments apply to vaccine experiments with S. japonicum in the mouse model; this schistosome species seems a more robust parasite, even harder to eliminate by acquired immune responses. We propose a number of ways in which our conclusions may be tested.
Collapse
Affiliation(s)
- R Alan Wilson
- Centre for Immunology and Infection, Department of Biology, University of York, Heslington, York, YO10 5DD, UK.
| | - Xiao-Hong Li
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025, People's Republic of China.
| | - William Castro-Borges
- Departamento de Ciências Biológicas, Universidade Federal de Ouro Preto, Campus Morro do Cruzeiro, Ouro Preto, Minas Gerais, Brasil.
| |
Collapse
|
48
|
Santini-Oliveira M, Coler RN, Parra J, Veloso V, Jayashankar L, Pinto PM, Ciol MA, Bergquist R, Reed SG, Tendler M. Schistosomiasis vaccine candidate Sm14/GLA-SE: Phase 1 safety and immunogenicity clinical trial in healthy, male adults. Vaccine 2015; 34:586-594. [PMID: 26571311 DOI: 10.1016/j.vaccine.2015.10.027] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 10/01/2015] [Accepted: 10/08/2015] [Indexed: 01/19/2023]
Abstract
DESIGN Safety and immunogenicity of a recombinant 14kDa, fatty acid-binding protein(FABP) from Schistosoma mansoni (rSm14) were evaluated through an open, non-placebo-controlled, dose-standardized trial, performed at a single research site. The vaccine was formulated with glucopyranosyl lipid A (GLA) adjuvant in an oil-in-water emulsion (SE) and investigated in 20 male volunteers from a non-endemic area for schistosomiasis in the state of Rio de Janeiro, Brazil. Fifty microgram rSm14 with 10 μg GLA-SE (rSm14/GLA-SE)/dose were given intramuscularly three times with 30-day intervals. Participants were assessed clinically, biochemically and immunologically for up to 120 days. METHODS Participants were screened for inclusion by physical examination, haematology and blood chemistry; then followed to assess adverse events and immunogenicity. Sera were tested for IgG (total and isotypes) and IgE. T cell induction of cytokines IL-2, IL-5, IL-10, IFNγ and TNFα was assessed by Milliplex kit and flow cytometry. RESULTS The investigational product showed high tolerability; some self-limited, mild adverse events were observed during and after vaccine administration. Significant increases in Sm14-specific total IgG, IgG1 and IgG3 were observed 30 days after the first vaccination with specific IgG2 and IgG4 after 60 days. An increase in IgE antibodies was not observed at any time point. The IgG response was augmented after the second dose and 88% of all vaccinated subjects had developed high anti-Sm14 IgG titres 90 days after the first injection. From day 60 and onwards, there was an increase in CD4(+) T cells producing single cytokines, particularly TNFα and IL-2, with no significant increase of multi-functional TH1 cells. CONCLUSION Clinical trial data on tolerability and specific immune responses after vaccination of adult, male volunteers in a non-endemic area for schistosomiasis with rSm14/GLA-SE, support this product as a safe, strongly immunogenic vaccine against schistosomiasis paving the way for follow-up Phase 2 trials. Study registration ID: NCT01154049 at http://www.clinicaltrials.gov.
Collapse
Affiliation(s)
- Marilia Santini-Oliveira
- Instituto Nacional de Infectologia Evandro Chagas (INI), Fiocruz, Av. Brasil, No. 4365, Manguinhos, Rio de Janeiro, 21045-900, Brazil
| | - Rhea N Coler
- Infectious Disease Research Institute (IDRI), 1616, Eastlake Ave E, Suite 400, Seattle, WA 98102, USA
| | - Juçara Parra
- Fiocruz/MS, Rua Gabriel Abrão s/n, Jardim das Nações, Campo Grande, Mato Grosso do Sul, 79.081-746, Brazil
| | - Valdilea Veloso
- Instituto Nacional de Infectologia Evandro Chagas (INI), Fiocruz, Av. Brasil, No. 4365, Manguinhos, Rio de Janeiro, 21045-900, Brazil
| | - Lakshmi Jayashankar
- Infectious Disease Research Institute (IDRI), 1616, Eastlake Ave E, Suite 400, Seattle, WA 98102, USA
| | - Patricia M Pinto
- Laboratório de Esquistossomose Experimental, Instituto Oswaldo Cruz, Fiocruz, Av. Brasil, No. 4365, Manguinhos, 21045-900 Rio de Janeiro, Brazil
| | - Marcia A Ciol
- Department of Rehabilitation Medicine, School of Medicine, University of Washington, 1959 NE Pacific St, UW Box 356490, Seattle, WA 98195-6490, USA
| | | | - Steven G Reed
- Infectious Disease Research Institute (IDRI), 1616, Eastlake Ave E, Suite 400, Seattle, WA 98102, USA
| | - Miriam Tendler
- Laboratório de Esquistossomose Experimental, Instituto Oswaldo Cruz, Fiocruz, Av. Brasil, No. 4365, Manguinhos, 21045-900 Rio de Janeiro, Brazil.
| |
Collapse
|
49
|
Cao X, Fu Z, Zhang M, Han Y, Han Q, Lu K, Li H, Zhu C, Hong Y, Lin J. Excretory/secretory proteome of 14-day schistosomula, Schistosoma japonicum. J Proteomics 2015; 130:221-30. [PMID: 26453986 DOI: 10.1016/j.jprot.2015.10.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 09/21/2015] [Accepted: 10/04/2015] [Indexed: 01/09/2023]
Abstract
Schistosomiasis remains a serious public health problem, with 200 million people infected and 779 million people at risk worldwide. The schistosomulum is the early stage of the complex lifecycle of Schistosoma japonicum in their vertebrate hosts, and is the main target of vaccine-induced protective immunity. Excretory/secretory (ES) proteins play a major role in host-parasite interactions and ES protein compositions of schistosomula of S. japonicum have not been characterized to date. In the present study, the proteome of ES proteins from 14 day schistosomula of S. japonicum was analyzed by liquid chromatography/tandem mass spectrometry and 713 unique proteins were finally identified. Gene ontology and pathway analysis revealed that identified proteins were mainly involved in carbohydrate metabolism, degradation, response to stimulus, oxidation-reduction, biological regulation and binding. Flow cytometry analysis demonstrated that thioredoxin peroxidase identified in this study had the effect on inhibiting MHCII and CD86 expression on LPS-activated macrophages. The present study provides insight into the growth and development of the schistosome in the final host and valuable information for screening vaccine candidates for schistosomiasis.
Collapse
Affiliation(s)
- Xiaodan Cao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, China
| | - Zhiqiang Fu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, China
| | - Min Zhang
- College of Animal Science and Technology, Henran University of Science and Technology, Luoyang, China
| | - Yanhui Han
- College of Animal Science, Henan Institute of Science and Technology, Xinxiang, China
| | - Qian Han
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, China
| | - Ke Lu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, China
| | - Hao Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, China
| | - Chuangang Zhu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, China
| | - Yang Hong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, China.
| | - Jiaojiao Lin
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture of China, Shanghai, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.
| |
Collapse
|
50
|
Neves LX, Sanson AL, Wilson RA, Castro-Borges W. What's in SWAP? Abundance of the principal constituents in a soluble extract of Schistosoma mansoni revealed by shotgun proteomics. Parasit Vectors 2015; 8:337. [PMID: 26088647 PMCID: PMC4484702 DOI: 10.1186/s13071-015-0943-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 06/08/2015] [Indexed: 12/27/2022] Open
Abstract
Background The soluble antigen preparation of adult schistosomes (SWAP) has often been used to probe host responses against these blood-dwelling parasites. Despite its long-established use there is limited knowledge about its composition. The information we provide here on the molecular composition of SWAP may contribute as a guide for a rational selection of antigenic targets. Methods Label-free quantitative shotgun proteomics was employed to determine the composition and abundance of SWAP constituents. Briefly, paired adult Schistosoma mansoni worms were sonicated in PBS pH 7.2 and ultracentrifuged for recovery of the soluble supernatant. An aliquot was subjected to trypsin digestion. Resulting peptides were separated under ultra-high performance liquid chromatography and analysed using an orbitrap mass spectrometer. Spectral data were interrogated using SequestHT against an in-house S. mansoni database. Proteins were quantified by recording the mean area under curve obtained for up to three most intense detected peptides. Proteins within the 90th percentile of the total SWAP mass were categorized according to their sub-cellular/tissue location. Results In this work we expanded significantly the list of known SWAP constituents. Through application of stringent criteria, a total of 633 proteins were quantitatively identified. Only 18 proteins account to 50 % of the total SWAP mass as revealed by their cumulative abundance. Among them, none is predicted as a secreted molecule reinforcing the point that SWAP is dominated by cytosolic and cytoskeletal proteins. In contrast, only 3 % of the mass comprised proteins proposed to function at the host-parasite interfaces (tegument and gut), which could conceivably represent vulnerable targets of a protective immune response. Paradoxically, at least 11 SWAP proteins, comprising ~25 % of its mass, have been tested as vaccine candidates. Conclusions Our data suggest that use of SWAP to probe host responses has greatest value for diagnostic purposes or assessing intensity of infection. However, the preparation is of limited utility as an antigen source for investigating host responses to proteins expressed at or secreted from worm-host interfaces. The data also pose the question as to why vaccination with SWAP, containing so many proposed vaccine candidates, has no additive or even synergistic effects on the induction of protection. Electronic supplementary material The online version of this article (doi:10.1186/s13071-015-0943-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Leandro Xavier Neves
- Laboratório de Enzimologia e Proteômica, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil.
| | - Ananda Lima Sanson
- Laboratório de Enzimologia e Proteômica, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil.
| | - R Alan Wilson
- Centre for Immunology & Infection, Department of Biology, University of York, PO Box 373, York, YO10 5YW, UK.
| | - William Castro-Borges
- Laboratório de Enzimologia e Proteômica, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil.
| |
Collapse
|