1
|
Guo Y, Taylor LN, Mishra R, Dolezal AG, Bonning BC. Gut-binding peptides as potential tools to reduce virus binding to honey bee gut surface proteins. Appl Environ Microbiol 2025; 91:e0241824. [PMID: 40019274 PMCID: PMC11921348 DOI: 10.1128/aem.02418-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 01/25/2025] [Indexed: 03/01/2025] Open
Abstract
Colonies of the western honey bee, Apis mellifera, are severely impacted by a wide range of stressors, with Varroa mites and associated viruses being among the most serious threats to honey bee health. Viral load plays an important role in colony demise, with the iflavirus Deformed wing virus (DWV) and the dicistrovirus Israeli acute paralysis virus (IAPV) being of particular concern. By feeding adult honey bees on a phage display library to identify gut-binding peptides (R. Mishra, Y. Guo, P. Kumar, P. E. Cantón, C. S. Tavares, R. Banerjee, S. Kuwar, and B. C. Bonning, Curr Res Insect Sci, 1:100012, https://doi.org/10.1016/j.cris.2021.100012), we identified Bee midgut-Binding Peptide (BBP2.1), which shares 75% and 85% identity with regions on the DWV capsid protein and IAPV ORFx protein, respectively. These viral protein domains are likely to be instrumental in virus interaction with the honey bee gut. Pull-down assays with honey bee gut brush border membrane vesicles were used to confirm peptide-mCherry binding to the gut for BBP2.1 and the two similar virus-derived sequences, peptides BBP2.1DWV and BBP2.1IAPV. In vitro competition assays showed that all three peptides compete with both IAPV and DWV virions for binding to honey bee gut-derived brush border membrane vesicles, suggesting that the three peptides and the two viruses bind to the same proteins. Ingestion of BBP2.1 reduced the movement of DWV, but not IAPV from the honey bee gut into the body and did not rescue IAPV-associated mortality. These results are discussed in relation to the biological function of IAPV ORFx and the potential utility of virus-blocking peptides for suppression of virus infection to reduce virus load and virus-associated honey bee mortality.IMPORTANCEEach year, approximately 40% of managed honey bee hives in the United States are lost due to a variety of environmental stressors. Although increases in virus infection are among the most important factors resulting in colony loss, there are currently no effective tools for the management of virus infection in honey bees. In this study, we identified a peptide that binds to the gut of the honey bee and competes with two of the most important honey bee viruses, Israeli acute paralysis virus of bees (IAPV) and Deformed wing virus (DWV), for binding to gut proteins. In vivo competition between this peptide and DWV demonstrates the potential utility of gut-binding peptides for the protection of honey bees from virus infection for reduced virus-associated honey bee mortality.
Collapse
Affiliation(s)
- Ya Guo
- Department of Entomology and Nematology, University of Florida, Gainesville, Florida, USA
| | - Lincoln N Taylor
- Department of Entomology, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA
| | - Ruchir Mishra
- Department of Entomology and Nematology, University of Florida, Gainesville, Florida, USA
| | - Adam G Dolezal
- Department of Entomology, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA
| | - Bryony C Bonning
- Department of Entomology and Nematology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
2
|
Bonning BC. Pathogen Binding and Entry: Molecular Interactions with the Insect Gut. ANNUAL REVIEW OF ENTOMOLOGY 2025; 70:165-184. [PMID: 39874144 DOI: 10.1146/annurev-ento-030624-014608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
The point of entry for the majority of arthropod pathogens and arthropod-vectored pathogens of plant, animal, and human health importance is the arthropod midgut. Pathogen interaction with the midgut therefore represents a primary target for intervention to prevent pathogen infection and transmission. Despite this key role in pathogen invasion, relatively little is known of the specific molecular interactions between pathogens and the surface of the arthropod gut epithelium, with few pathogen receptors having been definitively identified. This article provides an overview of pathogen molecular interactions in the arthropod midgut, with a focus on gut surface proteins that mediate pathogen entry, and highlights recent methodological advances that facilitate the identification of pathogen receptor proteins.
Collapse
Affiliation(s)
- Bryony C Bonning
- Department of Entomology and Nematology, University of Florida, Gainesville, Florida, USA;
| |
Collapse
|
3
|
Jacobs-Lorena M, Cha SJ. Unbiased phage display screening identifies hidden malaria vaccine targets. Emerg Microbes Infect 2024; 13:2429617. [PMID: 39529575 PMCID: PMC11587725 DOI: 10.1080/22221751.2024.2429617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/18/2024] [Accepted: 11/10/2024] [Indexed: 11/16/2024]
Abstract
Malaria is among the deadliest infectious diseases. Over 200 million annual clinical malaria cases are reported and more than half a million people, mostly children, die every year. The most advanced RTS,S/AS01 vaccine based on the P. falciparum circumsporozoite protein (CSP), targets sporozoite liver infection but achieved modest efficacy. To reduce malaria death, novel malaria vaccine development is a high priority. Most malaria vaccine candidates target three infection steps: sporozoite liver infection, merozoite red blood cell (RBC) infection, and mosquito midgut infection. However, only few malaria vaccine candidates target specific parasite-host cell interactions. Our group has implemented the phage peptide-display approach to discover new parasite ligands and host cell receptors. Here we summarize our findings and discuss their potential for the development of novel vaccines.
Collapse
Affiliation(s)
- Marcelo Jacobs-Lorena
- Department of Molecular Microbiology and Immunology and Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sung-Jae Cha
- Department of Medical Sciences, Mercer University School of Medicine, Macon, GA, USA
| |
Collapse
|
4
|
Kefi M, Cardoso-Jaime V, Saab SA, Dimopoulos G. Curing mosquitoes with genetic approaches for malaria control. Trends Parasitol 2024; 40:487-499. [PMID: 38760256 DOI: 10.1016/j.pt.2024.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 05/19/2024]
Abstract
Malaria remains a persistent global public health challenge because of the limitations of current prevention tools. The use of transgenic mosquitoes incapable of transmitting malaria, in conjunction with existing methods, holds promise for achieving elimination of malaria and preventing its reintroduction. In this context, population modification involves the spread of engineered genetic elements through mosquito populations that render them incapable of malaria transmission. Significant progress has been made in this field over the past decade in revealing promising targets, optimizing genetic tools, and facilitating the transition from the laboratory to successful field deployments, which are subject to regulatory scrutiny. This review summarizes recent advances and ongoing challenges in 'curing' Anopheles vectors of the malaria parasite.
Collapse
Affiliation(s)
- Mary Kefi
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Victor Cardoso-Jaime
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Sally A Saab
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - George Dimopoulos
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
5
|
Khan S, Patel MP, Patni AD, Cha SJ. Targeting Plasmodium Life Cycle with Novel Parasite Ligands as Vaccine Antigens. Vaccines (Basel) 2024; 12:484. [PMID: 38793735 PMCID: PMC11125637 DOI: 10.3390/vaccines12050484] [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: 03/28/2024] [Revised: 04/28/2024] [Accepted: 04/30/2024] [Indexed: 05/26/2024] Open
Abstract
The WHO reported an estimated 249 million malaria cases and 608,000 malaria deaths in 85 countries in 2022. A total of 94% of malaria deaths occurred in Africa, 80% of which were children under 5. In other words, one child dies every minute from malaria. The RTS,S/AS01 malaria vaccine, which uses the Plasmodium falciparum circumsporozoite protein (CSP) to target sporozoite infection of the liver, achieved modest efficacy. The Malaria Vaccine Implementation Program (MVIP), coordinated by the WHO and completed at the end of 2023, found that immunization reduced mortality by only 13%. To further reduce malaria death, the development of a more effective malaria vaccine is a high priority. Three malaria vaccine targets being considered are the sporozoite liver infection (pre-erythrocytic stage), the merozoite red blood cell infection (asexual erythrocytic stage), and the gamete/zygote mosquito infection (sexual/transmission stage). These targets involve specific ligand-receptor interactions. However, most current malaria vaccine candidates that target two major parasite population bottlenecks, liver infection, and mosquito midgut infection, do not focus on such parasite ligands. Here, we evaluate the potential of newly identified parasite ligands with a phage peptide-display technique as novel malaria vaccine antigens.
Collapse
Affiliation(s)
| | | | | | - Sung-Jae Cha
- Department of Medical Sciences, Mercer University School of Medicine, 1501 Mercer University Drive, Macon, GA 31207, USA; (S.K.); (M.P.P.); (A.D.P.)
| |
Collapse
|
6
|
Hernandez-Caballero I, Hellgren O, Garcia-Longoria Batanete L. Genomic advances in the study of the mosquito vector during avian malaria infection. Parasitology 2023; 150:1330-1339. [PMID: 37614176 PMCID: PMC10941221 DOI: 10.1017/s0031182023000756] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/25/2023]
Abstract
Invertebrate host–parasite associations are one of the keystones in order to understand vector-borne diseases. The study of these specific interactions provides information not only about how the vector is affected by the parasite at the gene-expression level, but might also reveal mosquito strategies for blocking the transmission of the parasites. A very well-known vector for human malaria is Anopheles gambiae. This mosquito species has been the main focus for genomics studies determining essential key genes and pathways over the course of a malaria infection. However, to-date there is an important knowledge gap concerning other non-mammophilic mosquito species, for example some species from the Culex genera which may transmit avian malaria but also zoonotic pathogens such as West Nile virus. From an evolutionary perspective, these 2 mosquito genera diverged 170 million years ago, hence allowing studies in both species determining evolutionary conserved genes essential during malaria infections, which in turn might help to find key genes for blocking malaria cycle inside the mosquito. Here, we extensively review the current knowledge on key genes and pathways expressed in Anopheles over the course of malaria infections and highlight the importance of conducting genomic investigations for detecting pathways in Culex mosquitoes linked to infection of avian malaria. By pooling this information, we underline the need to increase genomic studies in mosquito–parasite associations, such as the one in Culex–Plasmodium, that can provide a better understanding of the infection dynamics in wildlife and reduce the negative impact on ecosystems.
Collapse
Affiliation(s)
- Irene Hernandez-Caballero
- Department of Anatomy, Cellular Biology and Zoology, University of Extremadura, E-06071 Badajoz, Spain
| | - Olof Hellgren
- Molecular Ecology and Evolution Lab, Department of Biology, Lund University, Sölvegatan 37, SE-22362, Sweden
| | | |
Collapse
|
7
|
Kianifard L, Rafiqi AM, Akcakir O, Aly ASI, Billingsley PF, Uysal S. A recombinant Aspergillus oryzae fungus transmitted from larvae to adults of Anopheles stephensi mosquitoes inhibits malaria parasite oocyst development. Sci Rep 2023; 13:12177. [PMID: 37500682 PMCID: PMC10374630 DOI: 10.1038/s41598-023-38654-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 07/12/2023] [Indexed: 07/29/2023] Open
Abstract
The control of malaria parasite transmission from mosquitoes to humans is hampered by decreasing efficacies of insecticides, development of drug resistance against the last-resort antimalarials, and the absence of effective vaccines. Herein, the anti-plasmodial transmission blocking activity of a recombinant Aspergillus oryzae (A. oryzae-R) fungus strain, which is used in human food industry, was investigated in laboratory-reared Anopheles stephensi mosquitoes. The recombinant fungus strain was genetically modified to secrete two anti-plasmodial effector peptides, MP2 (midgut peptide 2) and EPIP (enolase-plasminogen interaction peptide) peptides. The transstadial transmission of the fungus from larvae to adult mosquitoes was confirmed following inoculation of A. oryzae-R in the water trays used for larval rearing. Secretion of the anti-plasmodial effector peptides inside the mosquito midguts inhibited oocyst formation of P. berghei parasites. These results indicate that A. oryzae can be used as a paratransgenesis model carrying effector proteins to inhibit malaria parasite development in An. stephensi. Further studies are needed to determine if this recombinant fungus can be adapted under natural conditions, with a minimal or no impact on the environment, to target mosquito-borne infectious disease agents inside their vectors.
Collapse
Affiliation(s)
- Leila Kianifard
- Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, 34820, Istanbul, Turkey
| | - Ab Matteen Rafiqi
- Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, 34820, Istanbul, Turkey
| | - Osman Akcakir
- Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, 34820, Istanbul, Turkey
| | - Ahmed S I Aly
- Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, 34820, Istanbul, Turkey
- School of Science and Engineering, Al Akhawayn University, Ifrane, 53000, Morocco
| | - Peter F Billingsley
- Sanaria Inc., 9800 Medical Center Dr., Suite A209, Rockville, MD, 20850, USA
| | - Serdar Uysal
- Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, 34820, Istanbul, Turkey.
| |
Collapse
|
8
|
Dong X, Wu W, Pan P, Zhang XZ. Engineered Living Materials for Advanced Diseases Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2304963. [PMID: 37436776 DOI: 10.1002/adma.202304963] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/04/2023] [Accepted: 07/11/2023] [Indexed: 07/13/2023]
Abstract
Natural living materials serving as biotherapeutics exhibit great potential for treating various diseases owing to their immunoactivity, tissue targeting, and other biological activities. In this review, the recent developments in engineered living materials, including mammalian cells, bacteria, viruses, fungi, microalgae, plants, and their active derivatives that are used for treating various diseases are summarized. Further, the future perspectives and challenges of such engineered living material-based biotherapeutics are discussed to provide considerations for future advances in biomedical applications.
Collapse
Affiliation(s)
- Xue Dong
- Institute for Advanced Studies, Wuhan University, Wuhan, 430072, P. R. China
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, P. R. China
| | - Wei Wu
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, P. R. China
| | - Pei Pan
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Xian-Zheng Zhang
- Institute for Advanced Studies, Wuhan University, Wuhan, 430072, P. R. China
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| |
Collapse
|
9
|
Klug D, Gautier A, Calvo E, Marois E, Blandin SA. The salivary protein Saglin facilitates efficient midgut colonization of Anopheles mosquitoes by malaria parasites. PLoS Pathog 2023; 19:e1010538. [PMID: 36862755 PMCID: PMC10013899 DOI: 10.1371/journal.ppat.1010538] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 03/14/2023] [Accepted: 02/08/2023] [Indexed: 03/03/2023] Open
Abstract
Malaria is caused by the unicellular parasite Plasmodium which is transmitted to humans through the bite of infected female Anopheles mosquitoes. To initiate sexual reproduction and to infect the midgut of the mosquito, Plasmodium gametocytes are able to recognize the intestinal environment after being ingested during blood feeding. A shift in temperature, pH change and the presence of the insect-specific compound xanthurenic acid have been shown to be important stimuli perceived by gametocytes to become activated and proceed to sexual reproduction. Here we report that the salivary protein Saglin, previously proposed to be a receptor for the recognition of salivary glands by sporozoites, facilitates Plasmodium colonization of the mosquito midgut, but does not contribute to salivary gland invasion. In mosquito mutants lacking Saglin, Plasmodium infection of Anopheles females is reduced, resulting in impaired transmission of sporozoites at low infection densities. Interestingly, Saglin can be detected in high amounts in the midgut of mosquitoes after blood ingestion, possibly indicating a previously unknown host-pathogen interaction between Saglin and midgut stages of Plasmodium. Furthermore, we were able to show that saglin deletion has no fitness cost in laboratory conditions, suggesting this gene would be an interesting target for gene drive approaches.
Collapse
Affiliation(s)
- Dennis Klug
- Université de Strasbourg, CNRS UPR9022, INSERM U1257, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
- * E-mail:
| | - Amandine Gautier
- Université de Strasbourg, CNRS UPR9022, INSERM U1257, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Eric Marois
- Université de Strasbourg, CNRS UPR9022, INSERM U1257, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Stéphanie A. Blandin
- Université de Strasbourg, CNRS UPR9022, INSERM U1257, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| |
Collapse
|
10
|
Charoenkwan P, Schaduangrat N, Lio P, Moni MA, Chumnanpuen P, Shoombuatong W. iAMAP-SCM: A Novel Computational Tool for Large-Scale Identification of Antimalarial Peptides Using Estimated Propensity Scores of Dipeptides. ACS OMEGA 2022; 7:41082-41095. [PMID: 36406571 PMCID: PMC9670693 DOI: 10.1021/acsomega.2c04465] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/20/2022] [Indexed: 06/16/2023]
Abstract
Antimalarial peptides (AMAPs) varying in length, amino acid composition, charge, conformational structure, hydrophobicity, and amphipathicity reflect their diversity in antimalarial mechanisms. Due to the worldwide major health problem concerning antimicrobial resistance, these peptides possess great therapeutic value owing to their low incidences of drug resistance as compared to conventional antibiotics. Although well-known experimental methods are able to precisely determine the antimalarial activity of peptides, these methods are still time-consuming and costly. Thus, machine learning (ML)-based methods that are capable of identifying AMAPs rapidly by using only sequence information would be beneficial for the high-throughput identification of AMAPs. In this study, we propose the first computational model (termed iAMAP-SCM) for the large-scale identification and characterization of peptides with antimalarial activity by using only sequence information. Specifically, we employed an interpretable scoring card method (SCM) to develop iAMAP-SCM and estimate propensities of 20 amino acids and 400 dipeptides to be AMAPs in a supervised manner. Experimental results showed that iAMAP-SCM could achieve a maximum accuracy and Matthew's coefficient correlation of 0.957 and 0.834, respectively, on the independent test dataset. In addition, SCM-derived propensities of 20 amino acids and selected physicochemical properties were used to provide an understanding of the functional mechanisms of AMAPs. Finally, a user-friendly online computational platform of iAMAP-SCM is publicly available at http://pmlabstack.pythonanywhere.com/iAMAP-SCM. The iAMAP-SCM predictor is anticipated to assist experimental scientists in the high-throughput identification of potential AMAP candidates for the treatment of malaria and other clinical applications.
Collapse
Affiliation(s)
- Phasit Charoenkwan
- Modern
Management and Information Technology, College of Arts, Media and
Technology, Chiang Mai University, Chiang Mai50200, Thailand
| | - Nalini Schaduangrat
- Center
of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok10700, Thailand
| | - Pietro Lio
- Department
of Computer Science and Technology, University
of Cambridge, CambridgeshireCB3 0FD, U.K.
| | - Mohammad Ali Moni
- Artificial
Intelligence & Digital Health, School of Health and Rehabilitation
Sciences, Faculty of Health and Behavioural Sciences, The University of Queensland, St LuciaQLD 4072, Australia
| | - Pramote Chumnanpuen
- Department
of Zoology, Faculty of Science, Kasetsart
University, Bangkok10900, Thailand
- Omics Center
for Agriculture, Bioresources, Food, and Health, Kasetsart University (OmiKU), Bangkok10900, Thailand
| | - Watshara Shoombuatong
- Center
of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok10700, Thailand
| |
Collapse
|
11
|
Hu X, Zhang X, Liu Y, Gao M, Lin M, Xie Y, Zhu Q, Xu C, Liu X, Vosloo D, Pooe OJ. Generation of Human Domain Antibody Fragments as Potential Insecticidal Agents against Helicoverpa armigera by Cadherin-Based Screening. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:11510-11519. [PMID: 35944165 DOI: 10.1021/acs.jafc.2c02000] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
New insecticidal genes and approaches for pest control are a hot research area. In the present study, we explored a novel strategy for the generation of insecticidal proteins. The midgut cadherin of Helicoverpa armigera (H. armigera) was used as a target to screen materials that have insecticidal activity. After three rounds of panning, the phage-displayed human domain antibody B1F6, which not only binds to the H. armigera cadherin CR9-CR11 but also significantly inhibits Cry1Ac toxins from binding to CR9-CR11, was obtained from a phage-displayed human domain antibody (DAb) library. To better analyze the relevant activity of B1F6, soluble B1F6 protein was expressed by Escherichia coli BL21 (DE3). The cytotoxicity assays demonstrated that soluble B1F6 induced Sf9 cell death when expressing H. armigera cadherin on the cell membrane. The insect bioassay results showed that soluble B1F6 protein (90 μg/cm2) caused 49.5 ± 3.3% H. armigera larvae mortality. The midgut histological results showed that soluble B1F6 caused damage to the midgut epithelium of H. armigera larvae. The present study explored a new strategy and provided a basic material for the generation of new insecticidal materials.
Collapse
Affiliation(s)
- Xiaodan Hu
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality (Ministry of Agriculture), Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Private Bag X54001, Durban 4000, South Africa
| | - Xiao Zhang
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality (Ministry of Agriculture), Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Yuan Liu
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality (Ministry of Agriculture), Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Meijing Gao
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality (Ministry of Agriculture), Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Manman Lin
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality (Ministry of Agriculture), Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Private Bag X54001, Durban 4000, South Africa
| | - Yajing Xie
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality (Ministry of Agriculture), Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Private Bag X54001, Durban 4000, South Africa
| | - Qing Zhu
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality (Ministry of Agriculture), Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Chongxin Xu
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality (Ministry of Agriculture), Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Xianjin Liu
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Key Laboratory of Control Technology and Standard for Agro-product Safety and Quality (Ministry of Agriculture), Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Dalene Vosloo
- School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Private Bag X54001, Durban 4000, South Africa
| | - Ofentse Jacob Pooe
- School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Private Bag X54001, Durban 4000, South Africa
| |
Collapse
|
12
|
Santos FA, Cruz GS, Vieira FA, Queiroz BR, Freitas CD, Mesquita FP, Souza PF. Systematic Review of Antiprotozoal Potential of Antimicrobial Peptides. Acta Trop 2022; 236:106675. [DOI: 10.1016/j.actatropica.2022.106675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/01/2022]
|
13
|
Banerjee R, Flores‐Escobar B, Chougule NP, Cantón PE, Dumitru R, Bonning BC. Peptide mediated, enhanced toxicity of a bacterial pesticidal protein against southern green stink bug. Microb Biotechnol 2022; 15:2071-2082. [PMID: 35315236 PMCID: PMC9249324 DOI: 10.1111/1751-7915.14030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 01/23/2023] Open
Abstract
The damage caused by stink bugs that feed on agricultural crops accounts for such significant losses that transgenic plant resistance to stink bugs would be highly desirable. As the level of toxicity of the Bacillus thuringiensis-derived, ETX/Mtx2 pesticidal protein Mpp83Aa1 is insufficient for practical use against the southern green stink bug Nezara viridula, we employed two disparate approaches to isolate peptides NvBP1 and ABP5 that bind to specific proteins (alpha amylase and aminopeptidase N respectively) on the surface of the N. viridula gut. Incorporation of these peptides into Mpp83Aa1 provided artificial anchors resulting in increased gut binding, and enhanced toxicity. These peptide-modified pesticidal proteins with increased toxicity provide a key advance for potential future use against N. viridula when delivered by transgenic plants to mitigate economic loss associated with this important pest.
Collapse
Affiliation(s)
- Rahul Banerjee
- Department of Entomology and NematologyUniversity of FloridaPO Box 110620GainesvilleFL32611USA
| | - Biviana Flores‐Escobar
- Department of Entomology and NematologyUniversity of FloridaPO Box 110620GainesvilleFL32611USA
| | | | - Pablo Emiliano Cantón
- Department of Entomology and NematologyUniversity of FloridaPO Box 110620GainesvilleFL32611USA
| | - Razvan Dumitru
- Innovation CenterBASF Corporation3500 Paramount ParkwayMorrisvilleNC27560USA
| | - Bryony C. Bonning
- Department of Entomology and NematologyUniversity of FloridaPO Box 110620GainesvilleFL32611USA
| |
Collapse
|
14
|
Fofana A, Yerbanga RS, Bilgo E, Ouedraogo GA, Gendrin M, Ouedraogo JB. The Strategy of Paratransgenesis for the Control of Malaria Transmission. FRONTIERS IN TROPICAL DISEASES 2022. [DOI: 10.3389/fitd.2022.867104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Insect-borne diseases are responsible for important burdens on health worldwide particularly in Africa. Malaria alone causes close to half a million deaths every year, mostly in developing, tropical and subtropical countries, with 94% of the global deaths in 2019 occurring in the WHO African region. With several decades, vector control measures have been fundamental to fight against malaria. Considering the spread of resistance to insecticides in mosquitoes and to drugs in parasites, the need for novel strategies to inhibit the transmission of the disease is pressing. In recent years, several studies have focused on the interaction of malaria parasites, bacteria and their insect vectors. Their findings suggested that the microbiota of mosquitoes could be used to block Plasmodium transmission. A strategy, termed paratransgenesis, aims to interfere with the development of malaria parasites within their vectors through genetically-modified microbes, which produce antimalarial effectors inside the insect host. Here we review the progress of the paratransgenesis approach. We provide a historical perspective and then focus on the choice of microbial strains and on genetic engineering strategies. We finally describe the different steps from laboratory design to field implementation to fight against malaria.
Collapse
|
15
|
Huang W, Cha S, Jacobs‐Lorena M. New weapons to fight malaria transmission: A historical view. ENTOMOLOGICAL RESEARCH 2022; 52:235-240. [PMID: 35846163 PMCID: PMC9272416 DOI: 10.1111/1748-5967.12585] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 05/31/2023]
Abstract
The stagnation of our fight against malaria in recent years, mainly due to the development of mosquito insecticide resistance, argues for the urgent development of new weapons. The dramatic evolution of molecular tools in the last few decades led to a better understanding of parasite-mosquito interactions and coalesced in the development of novel tools namely, mosquito transgenesis and paratransgenesis. Here we provide a historical view of the development of these new tools and point to some remaining challenges for their implementation in the field.
Collapse
Affiliation(s)
- Wei Huang
- Bloomberg School of Public HealthJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Sung‐Jae Cha
- Bloomberg School of Public HealthJohns Hopkins UniversityBaltimoreMarylandUSA
| | | |
Collapse
|
16
|
Viana TA, Barbosa WF, Jojoa LLB, Bernardes RC, da Silva JS, Jacobs-Lorena M, Martins GF. A Genetically Modified Anti-Plasmodium Bacterium Is Harmless to the Foragers of the Stingless Bee Partamona helleri. MICROBIAL ECOLOGY 2022; 83:766-775. [PMID: 34231036 PMCID: PMC9840896 DOI: 10.1007/s00248-021-01805-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 06/21/2021] [Indexed: 06/13/2023]
Abstract
Paratransgenesis consists of genetically engineering an insect symbiont to control vector-borne diseases. Biosafety assessments are a prerequisite for the use of genetically modified organisms (GMOs). Assessments rely on the measurement of the possible impacts of GMOs on different organisms, including beneficial organisms, such as pollinators. The bacterium Serratia AS1 has been genetically modified to express anti-Plasmodium effector proteins and does not impose a fitness cost on mosquitoes that carry it. In the present study, we assessed the impact of this bacterium on the native bee Partamona helleri (Meliponini), an ecologically important species in Brazil. Serratia eGFP AS1 (recombinant strain) or a wild strain of Serratia marcescens were suspended in a sucrose solution and fed to foragers, followed by measurements of survival, feeding rate, and behavior (walking and flying). These bacteria did not change any of the variables measured at 24, 72, and 144 h after the onset of the experiment. Recombinant and wild bacteria were detected in the homogenates of digestive tract during the 144 h period analyzed, but their numbers decreased with time. The recombinant strain was detected in the midgut at 24 h and in the hindgut at 72 h and 144 h after the onset of the experiment under the fluorescent microscope. As reported for mosquitoes, Serratia eGFP AS1 did not compromise the foragers of P. helleri, an ecologically relevant bee.
Collapse
Affiliation(s)
- Thaís Andrade Viana
- Departamento de Entomologia, Universidade Federal de Viçosa, Viçosa, MG, 36570-900, Brazil
| | - Wagner Faria Barbosa
- Departamento de Entomologia, Universidade Federal de Viçosa, Viçosa, MG, 36570-900, Brazil
- Departamento de Estatística, Universidade Federal de Viçosa, Viçosa, MG, 36570-900, Brazil
| | | | | | - Juliana Soares da Silva
- Departamento de Microbiologia, Universidade Federal de Viçosa, Viçosa, MG, 36570-900, Brazil
| | - Marcelo Jacobs-Lorena
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | | |
Collapse
|
17
|
Sturm A, Vos MW, Henderson R, Eldering M, Koolen KMJ, Sheshachalam A, Favia G, Samby K, Herreros E, Dechering KJ. Barcoded Asaia bacteria enable mosquito in vivo screens and identify novel systemic insecticides and inhibitors of malaria transmission. PLoS Biol 2021; 19:e3001426. [PMID: 34928952 PMCID: PMC8726507 DOI: 10.1371/journal.pbio.3001426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 01/04/2022] [Accepted: 12/03/2021] [Indexed: 11/18/2022] Open
Abstract
This work addresses the need for new chemical matter in product development for control of pest insects and vector-borne diseases. We present a barcoding strategy that enables phenotypic screens of blood-feeding insects against small molecules in microtiter plate-based arrays and apply this to discovery of novel systemic insecticides and compounds that block malaria parasite development in the mosquito vector. Encoding of the blood meals was achieved through recombinant DNA-tagged Asaia bacteria that successfully colonised Aedes and Anopheles mosquitoes. An arrayed screen of a collection of pesticides showed that chemical classes of avermectins, phenylpyrazoles, and neonicotinoids were enriched for compounds with systemic adulticide activity against Anopheles. Using a luminescent Plasmodium falciparum reporter strain, barcoded screens identified 48 drug-like transmission-blocking compounds from a 400-compound antimicrobial library. The approach significantly increases the throughput in phenotypic screening campaigns using adult insects and identifies novel candidate small molecules for disease control. This study presents a barcoding strategy that enables high-throughput phenotypic screens of blood-feeding insects against small molecules in microtiter plate-based arrays and applies this to the discovery of novel systemic insecticides and compounds that block malaria parasite development in the mosquito vector.
Collapse
|
18
|
Choi MY, Vander Meer RK. GPCR-Based Bioactive Peptide Screening Using Phage-Displayed Peptides and an Insect Cell System for Insecticide Discovery. Biomolecules 2021; 11:biom11040583. [PMID: 33923387 PMCID: PMC8071521 DOI: 10.3390/biom11040583] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 01/12/2023] Open
Abstract
The discovery of new insecticides improves integrated pest management (IPM), but is usually a long high-risk process with a low probability of success. For over two decades, insect neuropeptides (NPs) and their G-protein coupled receptors (GPCRs) have been considered as biological targets for insect pest control, because they are involved in almost all physiological processes associated with insect life stages. A key roadblock to success has been the question of how large volume chemical libraries can be efficiently screened for active compounds. New genomic and proteomic tools have advanced and facilitated the development of new approaches to insecticide discovery. In this study, we report a novel GPCR-based screening technology that uses millions of short peptides randomly generated by bacteriophages, and a method using an insect Sf9 cell expression system. The fire ant is a good model system, since bioactive peptides have been identified for a specific GPCR. The novel small peptides could interfere with the target GPCR-ligand functions. Therefore, we refer to this new mechanism as “receptor interference” (RECEPTORi). The GPCR-based bioactive peptide screening method offers multiple advantages. Libraries of phage-displayed peptides (~109 peptides) are inexpensive. An insect cell-based screening system rapidly leads to target specific GPCR agonists or antagonists in weeks. Delivery of bioactive peptides to target pests can be flexible, such as topical, ingestion, and plant-incorporated protectants. A variety of GPCR targets are available, thus minimizing the development of potential insecticide resistance. This report provides the first proof-of-concept for the development of novel arthropod pest management strategies using neuropeptides, and GPCRs.
Collapse
Affiliation(s)
- Man-Yeon Choi
- USDA-ARS, Horticultural Crops Research Laboratory, Corvallis, OR 97330, USA
- Correspondence:
| | - Robert K. Vander Meer
- USDA-ARS, Center for Medical, Agricultural, and Veterinary Entomology, Gainesville, FL 32608, USA;
| |
Collapse
|
19
|
Grogan C, Bennett M, Moore S, Lampe D. Novel Asaia bogorensis Signal Sequences for Plasmodium Inhibition in Anopheles stephensi. Front Microbiol 2021; 12:633667. [PMID: 33664722 PMCID: PMC7921796 DOI: 10.3389/fmicb.2021.633667] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/27/2021] [Indexed: 12/11/2022] Open
Abstract
Mosquitoes vector many pathogens that cause human disease, such as malaria that is caused by parasites in the genus Plasmodium. Current strategies to control vector-transmitted diseases are hindered by mosquito and pathogen resistance, so research has turned to altering the microbiota of the vectors. In this strategy, called paratransgenesis, symbiotic bacteria are genetically modified to affect the mosquito's phenotype by engineering them to deliver antiplasmodial effector molecules into the midgut to kill parasites. One paratransgenesis candidate is Asaia bogorensis, a Gram-negative, rod-shaped bacterium colonizing the midgut, ovaries, and salivary glands of Anopheles sp. mosquitoes. However, common secretion signals from E. coli and closely related species do not function in Asaia. Here, we report evaluation of 20 native Asaia N-terminal signal sequences predicted from bioinformatics for their ability to mediate increased levels of antiplasmodial effector molecules directed to the periplasm and ultimately outside the cell. We tested the hypothesis that by increasing the amount of antiplasmodials released from the cell we would also increase parasite killing power. We scanned the Asaia bogorensis SF2.1 genome to identify signal sequences from extra-cytoplasmic proteins and fused these to the reporter protein alkaline phosphatase. Six signals resulted in significant levels of protein released from the Asaia bacterium. Three signals were successfully used to drive the release of the antimicrobial peptide, scorpine. Further testing in mosquitoes demonstrated that these three Asaia strains were able to suppress the number of oocysts formed after a blood meal containing P. berghei to a significantly greater degree than wild-type Asaia, although prevalence was not decreased beyond levels obtained with a previously isolated siderophore receptor signal sequence. We interpret these results to indicate that there is a maximum level of suppression that can be achieved when the effectors are constitutively driven due to stress on the symbionts. This suggests that simply increasing the amount of antiplasmodial effector molecules in the midgut is insufficient to create superior paratransgenic bacterial strains and that symbiont fitness must be considered as well.
Collapse
Affiliation(s)
- Christina Grogan
- Department of Biological Sciences, Bayer School of Natural and Environmental Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Marissa Bennett
- Department of Biological Sciences, Bayer School of Natural and Environmental Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Shannon Moore
- Department of Biological Sciences, Bayer School of Natural and Environmental Sciences, Duquesne University, Pittsburgh, PA, United States
| | - David Lampe
- Department of Biological Sciences, Bayer School of Natural and Environmental Sciences, Duquesne University, Pittsburgh, PA, United States
| |
Collapse
|
20
|
Mishra R, Guo Y, Kumar P, Cantón PE, Tavares CS, Banerjee R, Kuwar S, Bonning BC. Streamlined phage display library protocols for identification of insect gut binding peptides highlight peptide specificity. CURRENT RESEARCH IN INSECT SCIENCE 2021; 1:100012. [PMID: 36003592 PMCID: PMC9387513 DOI: 10.1016/j.cris.2021.100012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 02/09/2021] [Accepted: 02/14/2021] [Indexed: 05/10/2023]
Abstract
Phage display libraries have been used to isolate insect gut binding peptides for use as pathogen transmission blocking agents, and to provide artificial anchors for increased toxicity of bacteria-derived pesticidal proteins. Previously, phage clones displaying enriched peptides were sequenced by Sanger sequencing. Here we present a streamlined protocol for identification of insect gut binding peptides, using insect-appropriate feeding strategies, with next generation sequencing and tailored bioinformatics analyses. The bioinformatics pipeline is designed to eliminate poorly enriched and false positive peptides, and to identify peptides predicted to be stable and hydrophilic. In addition to developing streamlined protocols, we also sought to address whether candidate gut binding peptides can bind to insects from more than one order, which is an important consideration for safe, practical use of peptide-modified pesticidal proteins. To this end, we screened phage display libraries for peptides that bind to the gut epithelia of two pest insects, the Asian citrus psyllid, Diaphorina citri (Hemiptera) and beet armyworm, Spodoptera exigua (Lepidoptera), and one beneficial insect, the western honey bee, Apis mellifera (Hymenoptera). While unique peptide sequences totaling 13,427 for D. citri, 89,561 for S. exigua and 69,053 for A. mellifera were identified from phage eluted from the surface of the insect guts, final candidate pools were comprised of 53, 107 and 1423 peptides respectively. The benefits of multiple rounds of biopanning, along with peptide binding properties in relation to practical use of peptide-modified pesticidal proteins for insect pest control are discussed.
Collapse
|
21
|
Nateghi Rostami M. CRISPR/Cas9 gene drive technology to control transmission of vector‐borne parasitic infections. Parasite Immunol 2020; 42:e12762. [DOI: 10.1111/pim.12762] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/20/2020] [Accepted: 05/26/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Mahmoud Nateghi Rostami
- Laboratory of Biology of Host‐Parasite Interactions Department of Parasitology Pasteur Institute of Iran Tehran Iran
| |
Collapse
|
22
|
Sáenz-Garcia JL, Yamanaka IB, Pacheco-Lugo LA, Miranda JS, Córneo ES, Machado-de-Ávila RA, De Moura JF, DaRocha WD. Targeting epimastigotes of Trypanosoma cruzi with a peptide isolated from a phage display random library. Exp Parasitol 2020; 210:107830. [PMID: 31917970 DOI: 10.1016/j.exppara.2020.107830] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/06/2019] [Accepted: 01/04/2020] [Indexed: 12/01/2022]
Abstract
Chagas disease, also known as American trypanosomiasis, is a potentially life-threatening illness caused by the protozoan parasite Trypanosoma cruzi, which is transmitted by insects of the family Reduviidae. Since conventional treatments with nitroheterocyclic drugs show serious adverse reactions and have questionable efficiency, different research groups have investigated polypeptide-based approaches to interfere with the parasite cell cycle in other Trypanosomatids. These strategies are supported by the fact that surface players are candidates to develop surface ligands that impair function since they may act as virulence factors. In this study, we used a phage display approach to identify peptides from one library-LX8CX8 (17 aa) (where X corresponds to any amino acid). After testing different biopanning conditions using live or fixed epimastigotes, 10 clones were sequenced that encoded the same peptide, named here as EPI18. The bacteriophage expressing EPI18 binds to epimastigotes from distinct strains of T. cruzi. To confirm these results, this peptide was synthetized, biotinylated, and assayed using flow cytometry and confocal microscopy analyses. These assays confirmed the specificity of the binding capacity of EPI18 toward epimastigote surfaces. Our findings suggest that EPI18 may have potential biotechnological applications that include peptide-based strategies to control parasite transmission.
Collapse
Affiliation(s)
- José L Sáenz-Garcia
- Laboratório de Genômica Funcional de Parasitos, Departamento de Bioquímica e Biologia Molecular, Universidade Federal Do Paraná, Curitiba, Brazil; Departamento de Ciencias Fisiológicas, Facultad de Ciencias Médicas, UNAN-Managua, Managua, Nicaragua
| | - Isabel B Yamanaka
- Laboratório de Imunoquímica, Departamento de Patologia Básica, Universidade Federal Do Paraná, Curitiba, Brazil
| | - Lisandro A Pacheco-Lugo
- Laboratório de Genômica Funcional de Parasitos, Departamento de Bioquímica e Biologia Molecular, Universidade Federal Do Paraná, Curitiba, Brazil; Universidad Simón Bolívar. Barranquilla, Colombia
| | - Juliana S Miranda
- Laboratório de Imunoquímica, Departamento de Patologia Básica, Universidade Federal Do Paraná, Curitiba, Brazil
| | - Emily S Córneo
- Laboratório de Fisiopatologia Experimental, Programa de Pós-Graduação Em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade Do Extremo Sul Catarinense, CEP, 88806-000. Criciúma, Brazil
| | - Ricardo A Machado-de-Ávila
- Laboratório de Fisiopatologia Experimental, Programa de Pós-Graduação Em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade Do Extremo Sul Catarinense, CEP, 88806-000. Criciúma, Brazil
| | - Juliana F De Moura
- Laboratório de Imunoquímica, Departamento de Patologia Básica, Universidade Federal Do Paraná, Curitiba, Brazil.
| | - Wanderson D DaRocha
- Laboratório de Genômica Funcional de Parasitos, Departamento de Bioquímica e Biologia Molecular, Universidade Federal Do Paraná, Curitiba, Brazil.
| |
Collapse
|
23
|
Kojin BB, Adelman ZN. The Sporozoite's Journey Through the Mosquito: A Critical Examination of Host and Parasite Factors Required for Salivary Gland Invasion. Front Ecol Evol 2019. [DOI: 10.3389/fevo.2019.00284] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
24
|
Liu K, Wang L, Guo Z. An extensive review of studies on mycobacterium cell wall polysaccharide-related oligosaccharides – part III: synthetic studies and biological applications of arabinofuranosyl oligosaccharides and their analogs, derivatives and conjugates. J Carbohydr Chem 2019. [DOI: 10.1080/07328303.2019.1630841] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Ji′nan, Shandong, China
| | - Lizhen Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Ji′nan, Shandong, China
| | - Zhongwu Guo
- Department of Chemistry, University of Florida, Gainesville, FL, USA
| |
Collapse
|
25
|
Zambrano-Mila MS, Sánchez Blacio KE, Santiago Vispo N. Peptide Phage Display: Molecular Principles and Biomedical Applications. Ther Innov Regul Sci 2019. [DOI: 10.1177/2168479019837624] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Marlon S. Zambrano-Mila
- School of Biological Sciences and Engineering, Yachay Tech University, San Miguel de Urcuquí, Ecuador
| | | | - Nelson Santiago Vispo
- School of Biological Sciences and Engineering, Yachay Tech University, San Miguel de Urcuquí, Ecuador
| |
Collapse
|
26
|
Li J, Han M, Yu J. Simple paratransgenic mosquitoes models and their dynamics. Math Biosci 2018; 306:20-31. [PMID: 30343053 DOI: 10.1016/j.mbs.2018.10.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 05/05/2018] [Accepted: 10/13/2018] [Indexed: 12/16/2022]
Abstract
To study the interactive dynamics of wild mosquitoes and mosquitoes carrying genetically-modified bacteria, we formulate continuous-time homogeneous and stage-structured models in this study. With appropriate transformations, complete results of the existence and stability of all boundary and positive equilibria for the homogeneous model are established and complete results of the existence and local stability of all boundary and positive equilibria for the stage-structured model are obtained as well. The outcomes from the homogeneous and the stage-structured models are similar. Based on the homogeneous model, we particularly investigate how the horizontal transmission of the transgenic bacteria, via the uptake rate of the transgenic bacteria, affects the interactive dynamics.
Collapse
Affiliation(s)
- Jia Li
- Department of Mathematics, Shanghai Normal University, Shanghai, 200234, CHINA; Department of Mathematical Sciences, University of Alabama in Huntsville, Huntsville, AL 35899, U.S.A.
| | - Maoan Han
- Department of Mathematics, Zhejiang Normal University, Jinhua, Zhejiang, 321004, CHINA; Department of Mathematics, Shanghai Normal University, Shanghai, 200234, CHINA.
| | - Jianshe Yu
- Center for Applied Mathematics, Guangzhou University, Guangzhou, 510006, CHINA.
| |
Collapse
|
27
|
Simões ML, Caragata EP, Dimopoulos G. Diverse Host and Restriction Factors Regulate Mosquito-Pathogen Interactions. Trends Parasitol 2018; 34:603-616. [PMID: 29793806 DOI: 10.1016/j.pt.2018.04.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 04/25/2018] [Accepted: 04/26/2018] [Indexed: 12/12/2022]
Abstract
Mosquitoes transmit diseases that seriously impact global human health. Despite extensive knowledge of the life cycles of mosquito-borne parasites and viruses within their hosts, control strategies have proven insufficient to halt their spread. An understanding of the relationships established between such pathogens and the host tissues they inhabit is therefore paramount for the development of new strategies that specifically target these interactions, to prevent the pathogens' maturation and transmission. Here we present an updated account of the antagonists and host factors that affect the development of Plasmodium, the parasite causing malaria, and mosquito-borne viruses, such as dengue virus and Zika virus, within their mosquito vectors, and we discuss the similarities and differences between Plasmodium and viral systems, looking toward the elucidation of new targets for disease control.
Collapse
Affiliation(s)
- Maria L Simões
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; These authors contributed equally
| | - Eric P Caragata
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; These authors contributed equally
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
28
|
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: 47] [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
|
29
|
Abstract
Novel affinity agents with high specificity are needed to make progress in disease diagnosis and therapy. Over the last several years, peptides have been considered to have fundamental benefits over other affinity agents, such as antibodies, due to their fast blood clearance, low immunogenicity, rapid tissue penetration, and reproducible chemical synthesis. These features make peptides ideal affinity agents for applications in disease diagnostics and therapeutics for a wide variety of afflictions. Virus-derived peptide techniques provide a rapid, robust, and high-throughput way to identify organism-targeting peptides with high affinity and selectivity. Here, we will review viral peptide display techniques, how these techniques have been utilized to select new organism-targeting peptides, and their numerous biomedical applications with an emphasis on targeted imaging, diagnosis, and therapeutic techniques. In the future, these virus-derived peptides may be used as common diagnosis and therapeutics tools in local clinics.
Collapse
Affiliation(s)
- Mingying Yang
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
| | - Kegan Sunderland
- Department of Chemistry & Biochemistry, Stephenson Life Science Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, Oklahoma 73019, United States
| | - Chuanbin Mao
- Department of Chemistry & Biochemistry, Stephenson Life Science Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, Oklahoma 73019, United States
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| |
Collapse
|
30
|
Jadhav PD, Shim YY, Reaney MJT. Synthesis and Characterization of Site-Selective Orbitide-BSA Conjugate to Produce Antibodies. Bioconjug Chem 2016; 27:2346-2358. [PMID: 27626291 DOI: 10.1021/acs.bioconjchem.6b00357] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Bioactive flax cyclic peptides (orbitides and linusorbs) were site-specifically ligated through methionine with bovine serum albumin (BSA) to produce immunogenic compounds. In this study, modified flaxseed immunosuppressant orbitides (linusorbs or LOs) containing hydroxyl (OH) groups were synthesized for use as haptens. These compounds were extensively characterized by 1H nuclear magnetic resonance (NMR), 13C NMR, high-performance liquid chromatography-tandem mass spectrometry, and Fourier transform infrared spectroscopy. The haptens were conjugated to BSA, and the extent of hapten incorporation was determined by matrix-assisted laser desorption and ionization, liquid chromatography-electrospray ionization-mass spectrometry, and sodium dodecyl sulfate polyacrylamide gel electrophoresis. The BSA hapten complexes were used to elicit polyclonal antibody (pAbs) production in rabbits. A competitive indirect enzyme-linked immunosorbent assay (CI-ELISA) was developed that used orbitide-specific pAbs and horseradish peroxidase (HRP) conjugates. The LO assay detection limit was approximately 0.01 μg/mL (ppm), and thus, ELISA can be used for the detection of LOs in tissue and plant samples. The pAbs can be used to detect and quantify LOs in flax and flaxseed samples, to verify the presence of LOs in flaxseed containing foods, and for the detection of LOs in tissue samples, wastes, and body fluids of animals fed flaxseed.
Collapse
Affiliation(s)
- Pramodkumar D Jadhav
- Department of Plant Sciences, University of Saskatchewan , 51 Campus Drive, Saskatoon, Saskatchewan S7N 5A8, Canada
| | - Youn Young Shim
- Department of Plant Sciences, University of Saskatchewan , 51 Campus Drive, Saskatoon, Saskatchewan S7N 5A8, Canada.,Prairie Tide Chemicals Inc. , 102 Melville Street, Saskatoon, Saskatchewan S7J 0R1, Canada
| | - Martin J T Reaney
- Department of Plant Sciences, University of Saskatchewan , 51 Campus Drive, Saskatoon, Saskatchewan S7N 5A8, Canada.,Prairie Tide Chemicals Inc. , 102 Melville Street, Saskatoon, Saskatchewan S7J 0R1, Canada.,Guangdong Saskatchewan Oilseed Joint Laboratory, Department of Food Science and Engineering, Jinan University , 601 Huangpu Avenue West, Guangzhou, Guangdong 510632, China
| |
Collapse
|
31
|
Zamanian M, Andersen EC. Prospects and challenges of CRISPR/Cas genome editing for the study and control of neglected vector-borne nematode diseases. FEBS J 2016; 283:3204-21. [PMID: 27300487 PMCID: PMC5053252 DOI: 10.1111/febs.13781] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 06/05/2016] [Accepted: 06/13/2016] [Indexed: 01/19/2023]
Abstract
Neglected tropical diseases caused by parasitic nematodes inflict an immense health and socioeconomic burden throughout much of the developing world. Current estimates indicate that more than two billion people are infected with nematodes, resulting in the loss of 14 million disability-adjusted life years per annum. Although these parasites cause significant mortality, they primarily cause chronic morbidity through a wide range of severe clinical ailments. Treatment options for nematode infections are restricted to a small number of anthelmintic drugs, and the rapid expansion of anthelmintic mass drug administration raises concerns of drug resistance. Preservation of existing drugs is necessary, as well as the development of new treatment options and methods of control. We focus this review on how the democratization of CRISPR/Cas9 genome editing technology can be enlisted to improve our understanding of the biology of nematode parasites and our ability to treat the infections they cause. We will first explore how this robust method of genome manipulation can be used to newly exploit the powerful model nematode Caenorhabditis elegans for parasitology research. We will then discuss potential avenues to develop CRISPR/Cas9 editing protocols in filarial nematodes. Lastly, we will propose potential ways in which CRISPR/Cas9 can be used to engineer gene drives that target the transmission of mosquito-borne filarial nematodes.
Collapse
Affiliation(s)
- Mostafa Zamanian
- Department of Molecular BiosciencesNorthwestern UniversityEvanstonILUSA
| | - Erik C. Andersen
- Department of Molecular BiosciencesNorthwestern UniversityEvanstonILUSA
| |
Collapse
|
32
|
Mukherjee D, Mishra P, Joshi M, Thakur PK, Hosur RV, Jarori GK. EWGWS insert in Plasmodium falciparum ookinete surface enolase is involved in binding of PWWP containing peptides: Implications to mosquito midgut invasion by the parasite. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2016; 68:13-22. [PMID: 26592350 DOI: 10.1016/j.ibmb.2015.11.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/12/2015] [Accepted: 11/16/2015] [Indexed: 06/05/2023]
Abstract
There are multiple stages in the life cycle of Plasmodium that invade host cells. Molecular machinery involved is such host-pathogen interactions constitute excellent drug targets and/or vaccine candidates. A screen using a phage display library has previously demonstrated presence of enolase on the surface of the Plasmodium ookinete. Phage-displayed peptides that bound to the ookinete contained a conserved motif (PWWP) in their sequence. Here, direct binding of these peptides with recombinant Plasmodium falciparum enolase (rPfeno) was investigated. These peptides showed specific binding to rPfeno, but failed to bind to other enolases. Plasmodium spp enolases are distinct in having an insert of five amino acids ((104)EWGWS(108)) that is not found in host enolases. The possibility of this insert being the recognition motif for the PWWP containing peptides was examined, (i) by comparing the binding of the peptides with rPfeno and a deletion variant Δ-rPfeno lacking (104)EWGWS(108), (ii) by measuring the changes in proton chemical shifts of PWWP peptides on binding to different enolases and (iii) by inter-molecular docking experiment to locate the peptide binding site. Results from these studies showed that the pentapeptide insert of Pfeno indeed constitutes the binding site for the PWWP domain containing peptide ligands. Search for sequences homologous to phage displayed peptides among peritrophic matrix proteins resulted in identification of perlecan, laminin, peritrophin and spacran. The possibility of these PWWP domain-containing proteins in the peritrophic matrix of insect gut to interact with ookinete cell surface enolase and facilitate the invasion of mosquito midgut epithelium is discussed.
Collapse
Affiliation(s)
- Debanjan Mukherjee
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Pushpa Mishra
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Mamata Joshi
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Prasoon Kumar Thakur
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisbon, 1649- 028 Lisbon, Portugal
| | - R V Hosur
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Gotam K Jarori
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India.
| |
Collapse
|
33
|
Inhibition of Plasmodium berghei Development in Mosquitoes by Effector Proteins Secreted from Asaia sp. Bacteria Using a Novel Native Secretion Signal. PLoS One 2015; 10:e0143541. [PMID: 26636338 PMCID: PMC4670117 DOI: 10.1371/journal.pone.0143541] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 11/05/2015] [Indexed: 11/25/2022] Open
Abstract
Novel interventions are needed to prevent the transmission of the Plasmodium parasites that cause malaria. One possible method is to supply mosquitoes with antiplasmodial effector proteins from bacteria by paratransgenesis. Mosquitoes have a diverse complement of midgut microbiota including the Gram-negative bacteria Asaia bogorensis. This study presents the first use of Asaia sp. bacteria for paratransgenesis against P. berghei. We identified putative secreted proteins from A. bogorensis by a genetic screen using alkaline phosphatase gene fusions. Two were secreted efficiently: a siderophore receptor protein and a YVTN beta-propeller repeat protein. The siderophore receptor gene was fused with antiplasmodial effector genes including the scorpine antimicrobial peptide and an anti-Pbs21 scFv-Shiva1 immunotoxin. Asaia SF2.1 secreting these fusion proteins were fed to mosquitoes and challenged with Plasmodium berghei-infected blood. With each of these effector constructs, significant inhibition of parasite development was observed. These results provide a novel and promising intervention against malaria transmission.
Collapse
|
34
|
Smith RC, Vega-Rodríguez J, Jacobs-Lorena M. The Plasmodium bottleneck: malaria parasite losses in the mosquito vector. Mem Inst Oswaldo Cruz 2015. [PMID: 25185005 PMCID: PMC4156458 DOI: 10.1590/0074-0276130597] [Citation(s) in RCA: 156] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nearly one million people are killed every year by the malaria parasite Plasmodium. Although the disease-causing forms of the parasite exist only in the human blood, mosquitoes of the genus Anopheles are the obligate vector for transmission. Here, we review the parasite life cycle in the vector and highlight the human and mosquito contributions that limit malaria parasite development in the mosquito host. We address parasite killing in its mosquito host and bottlenecks in parasite numbers that might guide intervention strategies to prevent transmission.
Collapse
Affiliation(s)
- Ryan C Smith
- Department of Molecular Microbiology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health and Immunology, Baltimore, MD, USA
| | - Joel Vega-Rodríguez
- Department of Molecular Microbiology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health and Immunology, Baltimore, MD, USA
| | - Marcelo Jacobs-Lorena
- Department of Molecular Microbiology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health and Immunology, Baltimore, MD, USA
| |
Collapse
|
35
|
Ruiz A, Pérez D, Muñoz MC, Molina JM, Taubert A, Jacobs-Lorena M, Vega-Rodríguez J, López AM, Hermosilla C. Targeting essential Eimeria ninakohlyakimovae sporozoite ligands for caprine host endothelial cell invasion with a phage display peptide library. Parasitol Res 2015; 114:4327-31. [PMID: 26341796 DOI: 10.1007/s00436-015-4666-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 08/06/2015] [Indexed: 01/14/2023]
Abstract
Eimeria ninakohlyakimovae is an important coccidian parasite of goats which causes severe diarrhoea in young animals. Specific molecules that mediate E. ninakohlyakimovae host interactions and molecular mechanisms involved in the pathogenesis are still unknown. Although strong circumstantial evidence indicates that E. ninakohlyakimovae sporozoite interactions with caprine endothelial host cells (ECs) are specific, hardly any information is available about the interacting molecules that confer host cell specificity. In this study, we describe a novel method to identify surface proteins of caprine umbilical vein endothelial cells (CUVEC) using a phage display library. After several panning rounds, we identified a number of peptides that specifically bind to the surface of CUVEC. Importantly, caprine endothelial cell peptide 2 (PCEC2) and PCEC5 selectively reduced the infection rate by E. ninakohlyakimovae sporozoites. These preliminary data give new insight for the molecular identification of ligands involved in the interaction between E. ninakohlyakimovae sporozoites and host ECs. Further studies using this phage approach might be useful to identify new potential target molecules for the development of anti-coccidial drugs or even new vaccine strategies.
Collapse
Affiliation(s)
- A Ruiz
- Parasitology Unit, Department of Animal Pathology, Faculty of Veterinary Medicine, University of Las Palmas de Gran Canaria, 35416, Arucas, Las Palmas, Spain.
| | - D Pérez
- Parasitology Unit, Department of Animal Pathology, Faculty of Veterinary Medicine, University of Las Palmas de Gran Canaria, 35416, Arucas, Las Palmas, Spain
| | - M C Muñoz
- Parasitology Unit, Department of Animal Pathology, Faculty of Veterinary Medicine, University of Las Palmas de Gran Canaria, 35416, Arucas, Las Palmas, Spain
| | - J M Molina
- Parasitology Unit, Department of Animal Pathology, Faculty of Veterinary Medicine, University of Las Palmas de Gran Canaria, 35416, Arucas, Las Palmas, Spain
| | - A Taubert
- Institute of Parasitology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - M Jacobs-Lorena
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - J Vega-Rodríguez
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - A M López
- Parasitology Unit, Department of Animal Pathology, Faculty of Veterinary Medicine, University of Las Palmas de Gran Canaria, 35416, Arucas, Las Palmas, Spain
| | - C Hermosilla
- Institute of Parasitology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
36
|
Cha SJ, Park K, Srinivasan P, Schindler CW, van Rooijen N, Stins M, Jacobs-Lorena M. CD68 acts as a major gateway for malaria sporozoite liver infection. ACTA ACUST UNITED AC 2015. [PMID: 26216124 PMCID: PMC4548058 DOI: 10.1084/jem.20110575] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Cha et al. use a phage display library screen to identify a peptide, P39, that binds to CD68 on the surface of Kupffer cells to inhibit malaria sporozoite cell entry. Thus, P39 may represent a therapeutic strategy for malaria by limiting hepatic infection. After being delivered by the bite from an infected mosquito, Plasmodium sporozoites enter the blood circulation and infect the liver. Previous evidence suggests that Kupffer cells, a macrophage-like component of the liver blood vessel lining, are traversed by sporozoites to initiate liver invasion. However, the molecular determinants of sporozoite–Kupffer cell interactions are unknown. Understanding the molecular basis for this specific recognition may lead to novel therapeutic strategies to control malaria. Using a phage display library screen, we identified a peptide, P39, that strongly binds to the Kupffer cell surface and, importantly, inhibits sporozoite Kupffer cell entry. Furthermore, we determined that P39 binds to CD68, a putative receptor for sporozoite invasion of Kupffer cells that acts as a gateway for malaria infection of the liver.
Collapse
Affiliation(s)
- Sung-Jae Cha
- W. Harry Feinstone Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health; and Department of Neurology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21205 W. Harry Feinstone Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health; and Department of Neurology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21205
| | - Kiwon Park
- W. Harry Feinstone Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health; and Department of Neurology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21205 W. Harry Feinstone Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health; and Department of Neurology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21205
| | - Prakash Srinivasan
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852
| | - Christian W Schindler
- Department of Microbiology and Immunology and Department of Medicine, Columbia University, New York, NY 10032 Department of Microbiology and Immunology and Department of Medicine, Columbia University, New York, NY 10032
| | - Nico van Rooijen
- Department of Molecular Cell Biology and Immunology, VUmc, 1081 BT Amsterdam, Netherlands
| | - Monique Stins
- W. Harry Feinstone Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health; and Department of Neurology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21205
| | - Marcelo Jacobs-Lorena
- W. Harry Feinstone Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health; and Department of Neurology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21205 W. Harry Feinstone Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health; and Department of Neurology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD 21205
| |
Collapse
|
37
|
Vega-Rodriguez J, Perez-Barreto D, Ruiz-Reyes A, Jacobs-Lorena M. Targeting molecular interactions essential for Plasmodium sexual reproduction. Cell Microbiol 2015; 17:1594-604. [PMID: 25944054 DOI: 10.1111/cmi.12458] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 04/30/2015] [Indexed: 01/15/2023]
Abstract
Malaria remains one of the most devastating infectious diseases, killing up to a million people every year. Whereas much progress has been made in understanding the life cycle of the parasite in the human host and in the mosquito vector, significant gaps of knowledge remain. Fertilization of malaria parasites, a process that takes place in the lumen of the mosquito midgut, is poorly understood and the molecular interactions (receptor-ligand) required for Plasmodium fertilization remain elusive. By use of a phage display library, we identified FG1 (Female Gamete peptide 1), a peptide that binds specifically to the surface of female Plasmodium berghei gametes. Importantly, FG1 but not a scrambled version of the peptide, strongly reduces P. berghei oocyst formation by interfering with fertilization. In addition, FG1 also inhibits P. falciparum oocyst formation suggesting that the peptide binds to a molecule on the surface of the female gamete whose structure is conserved. Identification of the molecular interactions disrupted by the FG1 peptide may lead to the development of novel malaria transmission-blocking strategies.
Collapse
Affiliation(s)
- Joel Vega-Rodriguez
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Davinia Perez-Barreto
- Departamento de Patología Animal, Producción Animal, Bromatología y Tecnología de los Alimentos Campus Universitario de Arucas - Facultad de Veterinaria, Universidad de Las Palmas de Gran Canarias, Gran Canarias, Spain
| | - Antonio Ruiz-Reyes
- Departamento de Patología Animal, Producción Animal, Bromatología y Tecnología de los Alimentos Campus Universitario de Arucas - Facultad de Veterinaria, Universidad de Las Palmas de Gran Canarias, Gran Canarias, Spain
| | - Marcelo Jacobs-Lorena
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
38
|
Lezaun J, Porter N. Containment and competition: transgenic animals in the One Health agenda. Soc Sci Med 2014; 129:96-105. [PMID: 24961736 DOI: 10.1016/j.socscimed.2014.06.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 05/01/2014] [Accepted: 06/12/2014] [Indexed: 10/25/2022]
Abstract
The development of the One World, One Health agenda coincides in time with the appearance of a different model for the management of human-animal relations: the genetic manipulation of animal species in order to curtail their ability as carriers of human pathogens. In this paper we examine two examples of this emergent transgenic approach to disease control: the development of transgenic chickens incapable of shedding avian flu viruses, and the creation of transgenic mosquitoes refractory to dengue or malaria infection. Our analysis elaborates three distinctions between the One World, One Health agenda and its transgenic counterpoint. The first concerns the conceptualization of outbreaks and the forms of surveillance that support disease control efforts. The second addresses the nature of the interspecies interface, and the relative role of humans and animals in preventing pathogen transmission. The third axis of comparison considers the proprietary dimensions of transgenic animals and their implications for the assumed public health ethos of One Health programs. We argue that the fundamental difference between these two approaches to infectious disease control can be summarized as one between strategies of containment and strategies of competition. While One World, One Health programs seek to establish an equilibrium in the human-animal interface in order to contain the circulation of pathogens across species, transgenic strategies deliberately trigger a new ecological dynamic by introducing novel animal varieties designed to out-compete pathogen-carrying hosts and vectors. In other words, while One World, One Health policies focus on introducing measures of inter-species containment, transgenic approaches derive their prophylactic benefit from provoking new cycles of intra-species competition between GM animals and their wild-type counterparts. The coexistence of these divergent health protection strategies, we suggest, helps to elucidate enduring tensions and concerns about how humans should relate to, appraise, and intervene on animals and their habitats.
Collapse
Affiliation(s)
| | - Natalie Porter
- University of Oxford, United Kingdom; University of New Hampshire, United States.
| |
Collapse
|
39
|
Suzuki Y, Niu G, Hughes GL, Rasgon JL. A viral over-expression system for the major malaria mosquito Anopheles gambiae. Sci Rep 2014; 4:5127. [PMID: 24875042 PMCID: PMC4038844 DOI: 10.1038/srep05127] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 05/14/2014] [Indexed: 11/08/2022] Open
Abstract
Understanding pathogen/mosquito interactions is essential for developing novel strategies to control mosquito-borne diseases. Technical advances in reverse-genetics, such as RNA interference (RNAi), have facilitated elucidation of components of the mosquito immune system that are antagonistic to pathogen development, and host proteins essential for parasite development. Forward genetic approaches, however, are limited to generation of transgenic insects, and while powerful, mosquito transgenesis is a resource- and time-intensive technique that is not broadly available to most laboratories. The ability to easily "over-express" genes would enhance molecular studies in vector biology and expedite elucidation of pathogen-refractory genes without the need to make transgenic insects. We developed and characterized an efficient Anopheles gambiae densovirus (AgDNV) over-expression system for the major malaria vector Anopheles gambiae. High-levels of gene expression were detected at 3 days post-infection and increased over time, suggesting this is an effective system for gene induction. Strong expression was observed in the fat body and ovaries. We validated multiple short promoters for gene induction studies. Finally, we developed a polycistronic system to simultaneously express multiple genes of interest. This AgDNV-based toolset allows for consistent transduction of genes of interest and will be a powerful molecular tool for research in Anopheles gambiae mosquitoes.
Collapse
Affiliation(s)
- Yasutsugu Suzuki
- Department of Entomology, Center for Infectious Disease Dynamics and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, 16802, United States of America
| | - Guodong Niu
- Department of Entomology, Center for Infectious Disease Dynamics and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, 16802, United States of America
- Current address: Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, 73019, United States of America
| | - Grant L. Hughes
- Department of Entomology, Center for Infectious Disease Dynamics and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, 16802, United States of America
| | - Jason L. Rasgon
- Department of Entomology, Center for Infectious Disease Dynamics and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, 16802, United States of America
| |
Collapse
|
40
|
YANG CUIHONG, LI JIA. DYNAMICS OF INTERACTIVE MOSQUITO POPULATIONS WITH TWO TRANSGENES. J BIOL SYST 2014. [DOI: 10.1142/s0218339013400032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We formulate a continuous-time 3D model system for the interactive mosquito populations with two different transgenes. We assume that the transgenes are dominant over the wild gene and that one of the transgenes is dominant over the other transgene. Using transformations, we translate the model system to its topologically equivalent 2D system. Based on the investigations of the reduced two-dimensional system, we obtain conditions for the existence and stability of boundary and positive equilibria. We provide numerical examples to demonstrate the rich and complex dynamical features of the model system.
Collapse
Affiliation(s)
- CUIHONG YANG
- School of Mathematics and Statistics, Central China Normal University, Wuhan, P. R. China
| | - JIA LI
- Department of Mathematical Sciences, University of Alabama in Huntsville, Huntsville, AL 35899, USA
| |
Collapse
|
41
|
Multiple pathways for Plasmodium ookinete invasion of the mosquito midgut. Proc Natl Acad Sci U S A 2014; 111:E492-500. [PMID: 24474798 DOI: 10.1073/pnas.1315517111] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Plasmodium ookinete invasion of the mosquito midgut is a crucial step of the parasite life cycle but little is known about the molecular mechanisms involved. Previously, a phage display peptide library screen identified SM1, a peptide that binds to the mosquito midgut epithelium and inhibits ookinete invasion. SM1 was characterized as a mimotope of an ookinete surface enolase and SM1 presumably competes with enolase, the presumed ligand, for binding to a putative midgut receptor. Here we identify a mosquito midgut receptor that binds both SM1 and ookinete surface enolase, termed "enolase-binding protein" (EBP). Moreover, we determined that Plasmodium berghei parasites are heterogeneous for midgut invasion, as some parasite clones are strongly inhibited by SM1 whereas others are not. The SM1-sensitive parasites required the mosquito EBP receptor for midgut invasion whereas the SM1-resistant parasites invaded the mosquito midgut independently of EBP. These experiments provide evidence that Plasmodium ookinetes can invade the mosquito midgut by alternate pathways. Furthermore, another peptide from the original phage display screen, midgut peptide 2 (MP2), strongly inhibited midgut invasion by P. berghei (SM1-sensitive and SM1-resistant) and Plasmodium falciparum ookinetes, suggesting that MP2 binds to a separate, universal receptor for midgut invasion.
Collapse
|
42
|
Basseri HR, Mohamadzadeh Hajipirloo H, Mohammadi Bavani M, Whitten MMA. Comparative susceptibility of different biological forms of Anopheles stephensi to Plasmodium berghei ANKA strain. PLoS One 2013; 8:e75413. [PMID: 24086525 PMCID: PMC3781038 DOI: 10.1371/journal.pone.0075413] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2012] [Accepted: 08/16/2013] [Indexed: 11/22/2022] Open
Abstract
Background There are varying degrees of compatibility between malaria parasite-mosquito species, and understanding this compatibility may be crucial for developing effective transmission-blocking vaccines. This study investigates the compatibility of different biological forms of a malaria vector, Anopheles stephensi, to Plasmodium berghei ANKA strain. Methods Several biologically different and allopatric forms of A. stephensi were studied. Three forms were isolated from different regions of southern Iran: the variety mysorensis, the intermediate form and the native type form, and an additional type form originated from India (Beech strain).The mosquitoes were experimentally infected with P. berghei to compare their susceptibility to parasitism. Anti-mosquito midgut antiserum was then raised in BALB/cs mice immunized against gut antigens from the most susceptible form of A. stephensi (Beech strain), and the efficacy of the antiserum was assessed in transmission-blocking assays conducted on the least susceptible mosquito biological form. Results The susceptibility of different biological forms of A. stephensi mosquito to P. berghei was specifically inter-type varied. The Beech strain and the intermediate form were both highly susceptible to infection, with higher oocyst and sporozoite infection rates than intermediate and mysorensis forms. The oocyst infection, and particularly sporozite infection, was lowest in the mysorensis strain. Antiserum raised against midgut proteins of the Indian Beech type form blocked infection in this mosquito population, but it was ineffective at blocking both oocyst and sporozoite development in the permissive but geographically distant intermediate form mosquitoes. This suggests that a strong degree of incompatibility exists between the mosquito strains in terms of midgut protein(s) acting as putative ookinete receptors. Conclusions The incompatibility in the midgut protein profiles between two biological forms of A. stephensi demonstrates a well-differentiated population structure according to geographical origin. Therefore, the design of potential transmission-blocking strategies should incorporate a more thorough understanding of intra-species variations in host-parasite interactions.
Collapse
Affiliation(s)
- Hamid R. Basseri
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Habib Mohamadzadeh Hajipirloo
- Department of Parasitology and Mycology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
- * E-mail:
| | - Mulood Mohammadi Bavani
- Department of Parasitology and Mycology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Miranda M. A. Whitten
- Institute of Life Science, College of Medicine, Swansea University, Singleton Park, Swansea, United Kingdom
| |
Collapse
|
43
|
Wang S, Jacobs-Lorena M. Genetic approaches to interfere with malaria transmission by vector mosquitoes. Trends Biotechnol 2013; 31:185-93. [PMID: 23395485 PMCID: PMC3593784 DOI: 10.1016/j.tibtech.2013.01.001] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 01/03/2013] [Accepted: 01/03/2013] [Indexed: 11/20/2022]
Abstract
Malaria remains one of the most devastating diseases worldwide, causing over 1 million deaths every year. The most vulnerable stages of Plasmodium development in the vector mosquito occur in the midgut lumen, making the midgut a prime target for intervention. Mosquito transgenesis and paratransgenesis are two novel strategies that aim at rendering the vector incapable of sustaining Plasmodium development. Mosquito transgenesis involves direct genetic engineering of the mosquito itself for delivery of anti-Plasmodium effector molecules. Conversely, paratransgenesis involves the genetic modification of mosquito symbionts for expression of anti-pathogen effector molecules. Here we consider both genetic manipulation strategies for rendering mosquitoes refractory to Plasmodium infection, and discuss challenges for the translation of laboratory findings to field applications.
Collapse
Affiliation(s)
- Sibao Wang
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China.
| | | |
Collapse
|
44
|
Eradication of malaria through genetic engineering: the current situation. ASIAN PAC J TROP MED 2013; 6:85-94. [DOI: 10.1016/s1995-7645(13)60001-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Revised: 12/15/2012] [Accepted: 01/15/2013] [Indexed: 01/03/2023] Open
|
45
|
Crawford JE, Rottschaefer SM, Coulibaly B, Sacko M, Niaré O, Riehle MM, Traore SF, Vernick KD, Lazzaro BP. No evidence for positive selection at two potential targets for malaria transmission-blocking vaccines in Anopheles gambiae s.s. INFECTION GENETICS AND EVOLUTION 2013; 16:87-92. [PMID: 23357581 DOI: 10.1016/j.meegid.2013.01.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 01/04/2013] [Accepted: 01/05/2013] [Indexed: 12/27/2022]
Abstract
Human malaria causes nearly a million deaths in sub-Saharan Africa each year. The evolution of drug-resistance in the parasite and insecticide resistance in the mosquito vector has complicated control measures and made the need for new control strategies more urgent. Anopheles gambiae s.s. is one of the primary vectors of human malaria in Africa, and parasite-transmission-blocking vaccines targeting Anopheles proteins have been proposed as a possible strategy to control the spread of the disease. However, the success of these hypothetical technologies would depend on the successful ability to broadly target mosquito populations that may be genetically heterogeneous. Understanding the evolutionary pressures shaping genetic variation among candidate target molecules offers a first step towards evaluating the prospects of successfully deploying such technologies. We studied the population genetics of genes encoding two candidate target proteins, the salivary gland protein saglin and the basal lamina structural protein laminin, in wild populations of the M and S molecular forms of A. gambiae in Mali. Through analysis of intraspecific genetic variation and interspecific comparisons, we found no evidence of positive natural selection at the genes encoding these proteins. On the contrary, we found evidence for particularly strong purifying selection at the laminin gene. These results provide insight into the patterns of genetic diversity of saglin and laminin, and we discuss these findings in relation to the potential development of these molecules as vaccine targets.
Collapse
|
46
|
Tonelli RR, Colli W, Alves MJM. Selection of binding targets in parasites using phage-display and aptamer libraries in vivo and in vitro. Front Immunol 2013; 3:419. [PMID: 23316203 PMCID: PMC3540409 DOI: 10.3389/fimmu.2012.00419] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 12/20/2012] [Indexed: 11/13/2022] Open
Abstract
Parasite infections are largely dependent on interactions between pathogen and different host cell populations to guarantee a successful infectious process. This is particularly true for obligatory intracellular parasites as Plasmodium, Toxoplasma, and Leishmania, to name a few. Adhesion to and entry into the cell are essential steps requiring specific parasite and host cell molecules. The large amount of possible involved molecules poses additional difficulties for their identification by the classical biochemical approaches. In this respect, the search for alternative techniques should be pursued. Among them two powerful methodologies can be employed, both relying upon the construction of highly diverse combinatorial libraries of peptides or oligonucleotides that randomly bind with high affinity to targets on the cell surface and are selectively displaced by putative ligands. These are, respectively, the peptide-based phage display and the oligonucleotide-based aptamer techniques. The phage display technique has been extensively employed for the identification of novel ligands in vitro and in vivo in different areas such as cancer, vaccine development, and epitope mapping. Particularly, phage display has been employed in the investigation of pathogen–host interactions. Although this methodology has been used for some parasites with encouraging results, in trypanosomatids its use is, as yet, scanty. RNA and DNA aptamers, developed by the SELEX process (Systematic Evolution of Ligands by Exponential Enrichment), were described over two decades ago and since then contributed to a large number of structured nucleic acids for diagnostic or therapeutic purposes or for the understanding of the cell biology. Similarly to the phage display technique scarce use of the SELEX process has been used in the probing of parasite–host interaction. In this review, an overall survey on the use of both phage display and aptamer technologies in different pathogenic organisms will be discussed. Using these techniques, recent results on the interaction of Trypanosoma cruzi with the host will be highlighted focusing on members of the 85 kDa protein family, a subset of the gp85/TS superfamily.
Collapse
Affiliation(s)
- R R Tonelli
- Departamento de Ciências Biológicas, Universidade Federal de São Paulo São Paulo, Brazil
| | | | | |
Collapse
|
47
|
Bazan J, Całkosiński I, Gamian A. Phage display--a powerful technique for immunotherapy: 2. Vaccine delivery. Hum Vaccin Immunother 2012; 8:1829-35. [PMID: 22906938 DOI: 10.4161/hv.21704] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Phage display is a powerful technique in medical and health biotechnology. This technology has led to formation of antibody libraries and has provided techniques for fast and efficient search of these libraries. The phage display technique has been used in studying the protein-protein or protein-ligand interactions, constructing of the antibody and antibody fragments and improving the affinity of proteins to receptors. Recently phage display has been widely used to study immunization process, develop novel vaccines and investigate allergen-antibody interactions. This technology can provide new tools for protection against viral, fungal and bacterial infections. It may become a valuable tool in cancer therapies, abuse and allergies treatment. This review presents the recent advancements in diagnostic and therapeutic applications of phage display. In particular the applicability of this technology to study the immunization process, construction of new vaccines and development of safer and more efficient delivery strategies has been described.
Collapse
Affiliation(s)
- Justyna Bazan
- Department of Medical Biochemistry; Wroclaw Medical University; Wroclaw, Poland
| | | | | |
Collapse
|
48
|
Wang S, Ghosh AK, Bongio N, Stebbings KA, Lampe DJ, Jacobs-Lorena M. Fighting malaria with engineered symbiotic bacteria from vector mosquitoes. Proc Natl Acad Sci U S A 2012; 109:12734-9. [PMID: 22802646 PMCID: PMC3412027 DOI: 10.1073/pnas.1204158109] [Citation(s) in RCA: 198] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The most vulnerable stages of Plasmodium development occur in the lumen of the mosquito midgut, a compartment shared with symbiotic bacteria. Here, we describe a strategy that uses symbiotic bacteria to deliver antimalaria effector molecules to the midgut lumen, thus rendering host mosquitoes refractory to malaria infection. The Escherichia coli hemolysin A secretion system was used to promote the secretion of a variety of anti-Plasmodium effector proteins by Pantoea agglomerans, a common mosquito symbiotic bacterium. These engineered P. agglomerans strains inhibited development of the human malaria parasite Plasmodium falciparum and rodent malaria parasite Plasmodium berghei by up to 98%. Significantly, the proportion of mosquitoes carrying parasites (prevalence) decreased by up to 84% for two of the effector molecules, scorpine, a potent antiplasmodial peptide and (EPIP)(4), four copies of Plasmodium enolase-plasminogen interaction peptide that prevents plasminogen binding to the ookinete surface. We demonstrate the use of an engineered symbiotic bacterium to interfere with the development of P. falciparum in the mosquito. These findings provide the foundation for the use of genetically modified symbiotic bacteria as a powerful tool to combat malaria.
Collapse
Affiliation(s)
- Sibao Wang
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205; and
| | - Anil K. Ghosh
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205; and
| | - Nicholas Bongio
- Department of Biological Sciences, Duquesne University, Pittsburgh, PA 15282
| | - Kevin A. Stebbings
- Department of Biological Sciences, Duquesne University, Pittsburgh, PA 15282
| | - David J. Lampe
- Department of Biological Sciences, Duquesne University, Pittsburgh, PA 15282
| | - Marcelo Jacobs-Lorena
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205; and
| |
Collapse
|
49
|
Discrete-time models with mosquitoes carrying genetically-modified bacteria. Math Biosci 2012; 240:35-44. [PMID: 22771952 DOI: 10.1016/j.mbs.2012.05.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2012] [Revised: 05/21/2012] [Accepted: 05/29/2012] [Indexed: 11/22/2022]
Abstract
We formulate a homogeneous model and a stage-structured model for the interactive wild mosquitoes and mosquitoes carrying genetically-modified bacteria. We establish conditions for the existence and stability of fixed points for both models. We show that a unique positive fixed point exists and is asymptotically stable if the two boundary fixed points are both unstable. The unique positive fixed point exists and is unstable if the two boundary fixed points are both locally asymptotically stable. Using numerical examples, we demonstrate the models undergoing a period-doubling bifurcation.
Collapse
|
50
|
Development of anti-infectives using phage display: biological agents against bacteria, viruses, and parasites. Antimicrob Agents Chemother 2012; 56:4569-82. [PMID: 22664969 DOI: 10.1128/aac.00567-12] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The vast majority of anti-infective therapeutics on the market or in development are small molecules; however, there is now a nascent pipeline of biological agents in development. Until recently, phage display technologies were used mainly to produce monoclonal antibodies (MAbs) targeted against cancer or inflammatory disease targets. Patent disputes impeded broad use of these methods and contributed to the dearth of candidates in the clinic during the 1990s. Today, however, phage display is recognized as a powerful tool for selecting novel peptides and antibodies that can bind to a wide range of antigens, ranging from whole cells to proteins and lipid targets. In this review, we highlight research that exploits phage display technology as a means of discovering novel therapeutics against infectious diseases, with a focus on antimicrobial peptides and antibodies in clinical or preclinical development. We discuss the different strategies and methods used to derive, select, and develop anti-infectives from phage display libraries and then highlight case studies of drug candidates in the process of development and commercialization. Advances in screening, manufacturing, and humanization technologies now mean that phage display can make a significant contribution in the fight against clinically important pathogens.
Collapse
|