1
|
Abida, Alhuthali HM, Alshehri JM, Alkathiri A, Almaghrabi ROM, Alsaeed SS, Albebi SAH, Almethn RM, Alfuraydi BA, Alharbi SB, Kamal M, Imran M. Exosomes in infectious diseases: insights into leishmaniasis pathogenesis, immune modulation, and therapeutic potential. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4913-4931. [PMID: 39702600 DOI: 10.1007/s00210-024-03702-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024]
Abstract
Leishmaniasis continues to be a critical international health issue due to the scarcity of efficient treatment and the development of drug tolerance. New developments in the research of extracellular vesicles (EVs), especially exosomes, have revealed novel disease management approaches. Exosomes are small vesicles that transport lipids, nucleic acids, and proteins in cell signalling. Its biogenesis depends on several cellular processes, and their functions in immune response, encompassing innate and adaptive immunity, underline their function in the pathogen-host interface. Exosomes play a significant role in the pathogenesis of some parasitic infections, especially Leishmaniasis, by helping parasites escape host immunity and promote disease progression. This article explains that in the framework of parasitic diseases, exosomes can act as master regulators that define the pathogenesis of the disease, as illustrated by the engagement of exosomes in the Leishmaniasis parasite and immune escape processes. Based on many published articles on Leishmaniasis, this review aims to summarize the biogenesis of exosomes, the properties of the cargo in exosomes, and the modulation of immune responses. We delve deeper into the prospect of using exosomes for the therapy of Leishmaniasis based on the possibility of using these extracellular vesicles for drug delivery and as diagnostic and prognostic biomarkers. Lastly, we focus on the recent research perspectives and future developments, underlining the necessity to continue the investigation of exosome-mediated approaches in Leishmaniasis treatment. Thus, this review intends to draw attention to exosomes as a bright new perspective in the battle against this disabling affliction.
Collapse
Affiliation(s)
- Abida
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia
| | - Hayaa M Alhuthali
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Jawaher Mohammad Alshehri
- Optometry Department, Faculty of Applied Medical Sciences, Albaha University, 65431, Albaha, Saudi Arabia
| | - Afnan Alkathiri
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Albaha University, 65431, Albaha, Saudi Arabia
| | - Ruba Omar M Almaghrabi
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Albaha University, 65431, Albaha, Saudi Arabia
| | | | | | | | | | | | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al-Kharj, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia.
- Center for Health Research, Northern Border University, Arar, Saudi Arabia.
| |
Collapse
|
2
|
Yu R, Song Z, Jin L, Jiao L, Liu H, Zhang S, Hu Y, Sun Y, Li E, Zhao G, Liu Z, Cai T. Polyethyleneimine-modified Laminarin nanoparticles as a novel vaccine adjuvant for ovalbumin to enhance the immune responses. Int J Biol Macromol 2025; 292:139157. [PMID: 39725115 DOI: 10.1016/j.ijbiomac.2024.139157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
Functional modification of drugs can significantly improve their efficacy and safety, thus enabling targeted therapy. Functional modifications based on polysaccharides can alter their molecular structure, and effectively enhance their functional properties and biological activities. Herein, we designed and synthesized cationic Laminarin (CLam) modified with polyethyleneimine (PEI) and explored its application as a vaccine adjuvant. The PEI modification resulted in a positively charged surface of CLam, which was mixed with model antigen (Ovalbumin, OVA) to form CLam/OVA nanoparticles with an optimal particle size of about 380.07 nm, a uniform distribution of the particle size and a stable system. In vitro experiments showed that the positive charge on the surface of CLam/OVA enabled it to be effectively internalized by bone marrow dendritic cells (BMDCs), promoted cell maturation, lysosomal escape, and the efficiency of antigen cross-presentation. Mechanically, CLam/OVA induces BMDC function via toll-like receptors, cytokine receptors, and chemokine-mediated signaling pathways. CLam/OVA induced stronger humoral and cellular immunity compared to the aluminum adjuvant. CLam/OVA induces higher levels of OVA-specific antibodies, generates cytotoxic T lymphocyte immune responses, and stimulates IFN-γ secretion. Overall, this study demonstrates that functionalization is critical for the rational design of polysaccharides to boost antigen-specific immune responses for more effective and long-lasting vaccination.
Collapse
Affiliation(s)
- Ruihong Yu
- Ningbo No. 2 Hospital, Ningbo 315010, Zhejiang, China; Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo 315000, Zhejiang, China; College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Zuchen Song
- Ningbo No. 2 Hospital, Ningbo 315010, Zhejiang, China; College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Lan Jin
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Lina Jiao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Huina Liu
- Ningbo No. 2 Hospital, Ningbo 315010, Zhejiang, China; Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo 315000, Zhejiang, China
| | - Shun Zhang
- Ningbo No. 2 Hospital, Ningbo 315010, Zhejiang, China; Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo 315000, Zhejiang, China
| | - Yaoren Hu
- Ningbo No. 2 Hospital, Ningbo 315010, Zhejiang, China; Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo 315000, Zhejiang, China
| | - Yuechao Sun
- Ningbo No. 2 Hospital, Ningbo 315010, Zhejiang, China; Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo 315000, Zhejiang, China
| | - Entao Li
- Ningbo No. 2 Hospital, Ningbo 315010, Zhejiang, China; Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo 315000, Zhejiang, China
| | - Guofang Zhao
- Ningbo No. 2 Hospital, Ningbo 315010, Zhejiang, China; Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo 315000, Zhejiang, China.
| | - Zhenguang Liu
- Ningbo No. 2 Hospital, Ningbo 315010, Zhejiang, China; Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo 315000, Zhejiang, China; College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China.
| | - Ting Cai
- Ningbo No. 2 Hospital, Ningbo 315010, Zhejiang, China; Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo 315000, Zhejiang, China.
| |
Collapse
|
3
|
Jayathilaka EHTT, Edirisinghe SL, De Zoysa M, Nikapitiya C. Exosomes derived from olive flounders infected with Streptococcus parauberis: Proteomic analysis, immunomodulation, and disease resistance capacity. FISH & SHELLFISH IMMUNOLOGY 2024; 148:109478. [PMID: 38452957 DOI: 10.1016/j.fsi.2024.109478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/02/2024] [Accepted: 03/04/2024] [Indexed: 03/09/2024]
Abstract
Multidrug-resistant Streptococcus parauberis causes high fish mortality in aquaculture, necessitating an urgent need for innovative control strategies. This study aimed to develop an immunizing agent against S. parauberis using exosomes isolated from the plasma of olive flounders infected experimentally with S. parauberis (Sp-Exo). Initially, we tested the in vitro immunomodulatory effect of Sp-Exo in murine macrophage RAW264.7 cells and compared it to that of exosomes isolated from naïve fish (PBS-Exo-treated). Notably, Sp-Exo treatment significantly (p < 0.05) upregulated pro-and anti-inflammatory cytokines (Il1β, Tnfα, and Il10), antimicrobial peptide, defensin isoforms (Def-rs2 and Def-ps1), and antiviral (Ifnβ1 and Isg15) genes. In vivo studies in larval and adult zebrafish revealed similar patterns of immunomodulation. Furthermore, larval and adult zebrafish exhibited significantly (p < 0.05) enhanced resistance to S. parauberis infection following treatment with Sp-Exo compared to that with PBS-Exo. Proteomic analysis using isobaric tags for relative and absolute quantitation (iTRAQ) approach revealed the presence of 77 upregulated and 94 downregulated differentially expressed proteins (DEPs) in Sp-Exo, with 22 and 37 significantly (p < 0.05) upregulated and downregulated DEPs, respectively. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and Search Tool for the Retrieval of Interacting Genes/Proteins analyses revealed that these genes are associated with key pathways, such as innate immune responses, complement system, acute phase responses, phospholipid efflux, and chylomicron remodeling. In conclusion, Sp-Exo demonstrated superior immunomodulatory activity and significant resistance against S. parauberis infection relative to that on treatment with PBS-Exo. Proteomic analysis further verified that most DEPs in Sp-Exo were associated with immune induction or modulation. These findings highlight the potential of Sp-Exo as a promising vaccine candidate against S. parauberis and other bacterial infections in olive flounder.
Collapse
Affiliation(s)
- E H T Thulshan Jayathilaka
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Shan Lakmal Edirisinghe
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Mahanama De Zoysa
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Yuseong-gu, Daejeon 34134, Republic of Korea.
| | - Chamilani Nikapitiya
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Yuseong-gu, Daejeon 34134, Republic of Korea.
| |
Collapse
|
4
|
Medina AC, Acevedo Ospina H, Descoteaux A. Immunomodulatory properties of Leishmania tarentolae extracellular vesicles containing the Spike protein of SARS-CoV-2. FRONTIERS IN PARASITOLOGY 2024; 3:1306478. [PMID: 39817166 PMCID: PMC11731608 DOI: 10.3389/fpara.2024.1306478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 03/22/2024] [Indexed: 01/18/2025]
Abstract
Extracellular vesicles released by the protozoan parasite Leishmania display immunomodulatory properties towards mammalian immune cells. In this study, we have evaluated the potential of extracellular vesicles derived from the non-pathogenic protozoan Leishmania tarentolae towards the development of a vaccine adjuvant. As a proof of concept, we expressed in L. tarentolae a codon-optimized SARS-CoV-2 Spike protein fused to the L. mexicana secreted acid phosphatase signal peptide in the N-terminal and to a 6×-His stretch in the C-terminal. Extracellular vesicles released by the engineered L. tarentolae were isolated by ultracentrifugation and fast protein liquid chromatography and were characterized via nanoparticle tracking analysis and transmission electron microscopy. The recombinant S protein was present in extracellular vesicles released by L. tarentolae, as determined by Western blot analyses and immunoelectron microscopy. Next, we evaluated the immunomodulatory potential of extracellular vesicles containing the S protein towards bone-marrow-derived macrophages and bone-marrow-derived dendritic cells. Our data show that in bone-marrow-derived dendritic cells, extracellular vesicles containing the S protein induced an increased expression of proinflammatory genes compared to plain extracellular vesicles whereas the opposite was observed in bone-marrow-derived macrophages. These findings reveal the immunomodulatory potential of L. tarentolae extracellular vesicles and provide a proof of concept that they can be used as adjuvant in the context of dendritic cell stimulation.
Collapse
Affiliation(s)
- Ana Catalina Medina
- INRS- Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, QC, Canada
- Infectiopôle INRS, Laval, QC, Canada
| | - Hamlet Acevedo Ospina
- INRS- Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, QC, Canada
- Infectiopôle INRS, Laval, QC, Canada
| | - Albert Descoteaux
- INRS- Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, QC, Canada
- Infectiopôle INRS, Laval, QC, Canada
| |
Collapse
|
5
|
Chatterjee S, Kordbacheh R, Sin J. Extracellular Vesicles: A Novel Mode of Viral Propagation Exploited by Enveloped and Non-Enveloped Viruses. Microorganisms 2024; 12:274. [PMID: 38399678 PMCID: PMC10892846 DOI: 10.3390/microorganisms12020274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
Extracellular vesicles (EVs) are small membrane-enclosed structures that have gained much attention from researchers across varying scientific fields in the past few decades. Cells secrete diverse types of EVs into the extracellular milieu which include exosomes, microvesicles, and apoptotic bodies. These EVs play a crucial role in facilitating intracellular communication via the transport of proteins, lipids, DNA, rRNA, and miRNAs. It is well known that a number of viruses hijack several cellular pathways involved in EV biogenesis to aid in their replication, assembly, and egress. On the other hand, EVs can also trigger host antiviral immune responses by carrying immunomodulatory molecules and viral antigens on their surface. Owing to this intricate relationship between EVs and viruses, intriguing studies have identified various EV-mediated viral infections and interrogated how EVs can alter overall viral spread and longevity. This review provides a comprehensive overview on the EV-virus relationship, and details various modes of EV-mediated viral spread in the context of clinically relevant enveloped and non-enveloped viruses.
Collapse
Affiliation(s)
| | | | - Jon Sin
- Department of Biological Sciences, University of Alabama, 1325 Hackberry Lane, Tuscaloosa, AL 35401, USA; (S.C.); (R.K.)
| |
Collapse
|
6
|
Gómez-Chávez F, Murrieta-Coxca JM, Caballero-Ortega H, Morales-Prieto DM, Markert UR. Host-pathogen interactions mediated by extracellular vesicles in Toxoplasma gondii infection during pregnancy. J Reprod Immunol 2023; 158:103957. [PMID: 37253287 DOI: 10.1016/j.jri.2023.103957] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/15/2023] [Accepted: 05/17/2023] [Indexed: 06/01/2023]
Abstract
Molecular communication between a pathogen and its host is crucial for a successful interplay. Extracellular vesicles (EVs) act as mediators for the delivery of molecular signals among pathogens or between pathogens and the host. Toxoplasma gondii (T. gondii), an intracellular parasite with a worldwide presence, produces EVs itself, or induces the secretion of EVs from infected host cells potentially having capacities to modulate the host immune response. T. gondii infection is particularly important during pregnancy. Depending on the gestational age at the time of infection, the parasite can be transmitted through the placenta to the fetus, causing clinical complications such as jaundice, hepatosplenomegaly, chorioretinitis, cranioencephalic abnormalities, or even death. T. gondii infection is related to a pro-inflammatory immune response in both mother and fetus, which may enhance parasite transmission, but the implication of EV signaling in this process remains unclear. In this review, we summarize the current knowledge on EV release from T. gondii and its human host cells in regard to the immunological consequences and the passage through the placenta.
Collapse
Affiliation(s)
- Fernando Gómez-Chávez
- Sección de Estudios de Posgrado e Investigación, Escuela Nacional de Medicina y Homeopatía-Instituto Politécnico Nacional, Mexico City, Mexico; Programa de Posgrado en Ciencia y Tecnología de Vacunas y Bioterapéuticos, Instituto Politécnico Nacional, Mexico City, Mexico
| | | | - Heriberto Caballero-Ortega
- Secretaría de Salud, Laboratorio de Inmunología Experimental, Instituto Nacional de Pediatría, Mexico City, Mexico
| | | | - Udo R Markert
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany.
| |
Collapse
|
7
|
Yang F, Zhang M, Jin YG, Chen JC, Duan MH, Liu Y, Li ZE, Li XP, Yang F. Development and Application of a Physiologically Based Pharmacokinetic Model for Diclazuril in Broiler Chickens. Animals (Basel) 2023; 13:1512. [PMID: 37174549 PMCID: PMC10177140 DOI: 10.3390/ani13091512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Withdrawal periods for diclazuril in broilers have traditionally been determined through regression analysis. However, over the last two decades, the physiologically based pharmacokinetic (PBPK) model has gained prominence as a predictive tool for veterinary drug residues, which offers an alternative method for establishing appropriate withdrawal periods for veterinary drugs. In this current study, a flow-limited PBPK model was developed to predict diclazuril concentrations in broilers following long-duration administration via medicated feed and water. This model consists of nine compartments, including arterial and venous plasma, lung, muscle, skin + fat, kidney, liver, intestine contents, and the rest of the body compartment. Physiological parameters such as tissue weights (Vcxx) and blood flow (Qcxx) were gathered from published studies, and tissue/plasma partition coefficients (Pxx) were calculated through the area method or parameter optimization. Published diclazuril concentrations were compared to the predicted values, indicating the accuracy and validity of the model. The sensitivity analysis showed that parameters associated with cardiac output, drug absorption, and elimination significantly affected diclazuril concentrations in the muscle. Finally, a Monte Carlo analysis, consisting of 1000 iterations, was conducted to calculate the withdrawal period. Based on the Chinese MRL values, we calculated a withdrawal period of 0 days for both recommended dosing regimens (through mediated water and feed at concentrations of 0.5-1 mg/L and 1 mg/kg, respectively). However, based on the European MRLs, longer periods were determined for the mediated feed dosing route. Our model provides a foundation for scaling other coccidiostats and poultry species.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Fan Yang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China (Y.-G.J.)
| |
Collapse
|
8
|
Lu M, Lee Y, Lillehoj HS. Evolution of developmental and comparative immunology in poultry: The regulators and the regulated. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 138:104525. [PMID: 36058383 DOI: 10.1016/j.dci.2022.104525] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/25/2022] [Accepted: 08/28/2022] [Indexed: 06/15/2023]
Abstract
Avian has a unique immune system that evolved in response to environmental pressures in all aspects of innate and adaptive immune responses, including localized and circulating lymphocytes, diversity of immunoglobulin repertoire, and various cytokines and chemokines. All of these attributes make birds an indispensable vertebrate model for studying the fundamental immunological concepts and comparative immunology. However, research on the immune system in birds lags far behind that of humans, mice, and other agricultural animal species, and limited immune tools have hindered the adequate application of birds as disease models for mammalian systems. An in-depth understanding of the avian immune system relies on the detailed studies of various regulated and regulatory mediators, such as cell surface antigens, cytokines, and chemokines. Here, we review current knowledge centered on the roles of avian cell surface antigens, cytokines, chemokines, and beyond. Moreover, we provide an update on recent progress in this rapidly developing field of study with respect to the availability of immune reagents that will facilitate the study of regulatory and regulated components of poultry immunity. The new information on avian immunity and available immune tools will benefit avian researchers and evolutionary biologists in conducting fundamental and applied research.
Collapse
Affiliation(s)
- Mingmin Lu
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, U.S. Department of Agriculture-Agricultural Research Service, Beltsville, MD, 20705, USA.
| | - Youngsub Lee
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, U.S. Department of Agriculture-Agricultural Research Service, Beltsville, MD, 20705, USA.
| | - Hyun S Lillehoj
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, U.S. Department of Agriculture-Agricultural Research Service, Beltsville, MD, 20705, USA.
| |
Collapse
|
9
|
Eimeria tenella 14-kDa phosphohistidine phosphatase stimulates maturation of chicken dendritic cells and mediates DC-induced T cell priming in a Th1 cytokine interface. Res Vet Sci 2022; 152:61-71. [DOI: 10.1016/j.rvsc.2022.07.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 07/18/2022] [Accepted: 07/24/2022] [Indexed: 11/15/2022]
|
10
|
Extracellular Vesicles in Veterinary Medicine. Animals (Basel) 2022; 12:ani12192716. [PMID: 36230457 PMCID: PMC9559303 DOI: 10.3390/ani12192716] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/23/2022] [Accepted: 10/03/2022] [Indexed: 11/16/2022] Open
Abstract
Extracellular vesicles (EVs) are cell-derived membrane-bound vesicles involved in many physiological and pathological processes not only in humans but also in all the organisms of the eukaryotic and prokaryotic kingdoms. EV shedding constitutes a fundamental universal mechanism of intra-kingdom and inter-kingdom intercellular communication. A tremendous increase of interest in EVs has therefore grown in the last decades, mainly in humans, but progressively also in animals, parasites, and bacteria. With the present review, we aim to summarize the current status of the EV research on domestic and wild animals, analyzing the content of scientific literature, including approximately 220 papers published between 1984 and 2021. Critical aspects evidenced through the veterinarian EV literature are discussed. Then, specific subsections describe details regarding EVs in physiology and pathophysiology, as biomarkers, and in therapy and vaccines. Further, the wide area of research related to animal milk-derived EVs is also presented in brief. The numerous studies on EVs related to parasites and parasitic diseases are excluded, deserving further specific attention. The literature shows that EVs are becoming increasingly addressed in veterinary studies and standardization in protocols and procedures is mandatory, as in human research, to maximize the knowledge and the possibility to exploit these naturally produced nanoparticles.
Collapse
|
11
|
Li H, Sun L, Jiang Y, Wang B, Wu Z, Sun J, Zhang X, Li H, Zhao X. Identification and characterization of Eimeria tenella EtTrx1 protein. Vet Parasitol 2022; 310:109785. [PMID: 35994916 DOI: 10.1016/j.vetpar.2022.109785] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 12/01/2022]
Abstract
Thioredoxin (Trx) is a widespread protein regulator of redox reactions in all organisms. It operates together with NADPH and thioredoxin reductase as a general protein disulfide catalytic system. Recently, Trx has been found to be related to the process by which apicomplexan protozoa invade host cells. In this study, Eimeria tenella thioredoxin (EtTrx1) was identified and its gene structural features, expression levels at different developmental stages, localization in sporozoites, roles in adhesion and invasion, and immunogenicity were investigated. Sequence analysis indicated that EtTrx1 contains a Trx domain with a WCGPC motif in 29-33 aa and a typical Trx fold, and belongs to thioredoxin family. EtTrx1 was detected on the surface of sporozoites using anti-EtTrx1 polyclonal antibodies under non-permeabilized conditions by indirect immunofluorescence assay (IFA) and also in a secretion form. EtTrx1 protein was highly transcribed and expressed in merozoites and sporozoites by quantitative PCR and western blot. The attachment assay showed that the adherence rates of yeast cells expressing EtTrx1 on the surface to host cells were 3.1-fold higher than those of the blank control. Specific anti-EtTrx1 antibodies inhibited the invasion of sporozoites into DF-1 cells. The highest inhibition rate was up to 36.75% compared to the control group. Immunization with recombinant EtTrx1 peptides also showed significant protection against lethal infections in chickens. It could offer moderate protective efficacy (Anticoccidial Index [ACI]: 163.70), induce humoral responses, and be an effective candidate for the development of new vaccines.
Collapse
Affiliation(s)
- Huihui Li
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China
| | - Lingyu Sun
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China
| | - Yingying Jiang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China
| | - Bingxiang Wang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China
| | - Zhiyuan Wu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China
| | - Jinkun Sun
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China
| | - Xiao Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China
| | - Hongmei Li
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China.
| | - Xiaomin Zhao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China.
| |
Collapse
|
12
|
Zaheer T, Abbas RZ, Imran M, Abbas A, Butt A, Aslam S, Ahmad J. Vaccines against chicken coccidiosis with particular reference to previous decade: progress, challenges, and opportunities. Parasitol Res 2022; 121:2749-2763. [PMID: 35925452 PMCID: PMC9362588 DOI: 10.1007/s00436-022-07612-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 07/24/2022] [Indexed: 11/29/2022]
Abstract
Chicken coccidiosis is an economically significant disease of commercial chicken industry accounting for losses of more than £10.4 billion (according to 2016 prices). Additionally, the costs incurred in prophylaxis and therapeutics against chicken coccidiosis in developing countries (for instance Pakistan according to 2018 prices) reached US $45,000.00 while production losses for various categories of chicken ranges 104.74 to US $2,750,779.00. The infection has been reported from all types of commercial chickens (broiler, layer, breeder) having a range of reported prevalence of 7-90%. The concern of resistance towards major anticoccidials has provided a way forward to vaccine research and development. For prophylaxis of chicken coccidiosis, live virulent, attenuated, ionophore tolerant strains and recombinant vaccines have been extensively trialed and commercialized. Eimeria antigens and novel vaccine adjuvants have elicited the protective efficacy against coccidial challenge. The cost of production and achieving robust immune responses in birds are major challenges for commercial vaccine production. In the future, research should be focused on the development of multivalent anticoccidial vaccines for commercial poultry. Efforts should also be made on the discovery of novel antigens for incorporation into vaccine designs which might be more effective against multiple Eimeria species. This review presents a recap to the overall progress against chicken Eimeria with particular reference to previous decade. The article presents critical analysis of potential areas for future research in chicken Eimeria vaccine development.
Collapse
Affiliation(s)
- Tean Zaheer
- Department of Parasitology, University of Agriculture, Faisalabad, Pakistan
| | - Rao Zahid Abbas
- Department of Parasitology, University of Agriculture, Faisalabad, Pakistan.
| | - Muhammad Imran
- Department of Parasitology, University of Agriculture, Faisalabad, Pakistan
| | - Asghar Abbas
- Faculty of Veterinary Science, Muhammad Nawaz Shareef University of Agriculture Multan, Multan, Pakistan
| | - Ali Butt
- Department of Parasitology, University of Agriculture, Faisalabad, Pakistan
| | - Sarfraz Aslam
- Institute of Physiology, Pharmacology and Pharmaceutics, University of Agriculture, Faisalabad, Pakistan
| | - Jameel Ahmad
- Institute of Physiology, Pharmacology and Pharmaceutics, University of Agriculture, Faisalabad, Pakistan
| |
Collapse
|
13
|
Hong Y, Truong AD, Vu TH, Lee S, Heo J, Kang S, Lillehoj HS, Hong YH. Exosomes from H5N1 avian influenza virus-infected chickens regulate antiviral immune responses of chicken immune cells. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 130:104368. [PMID: 35104460 DOI: 10.1016/j.dci.2022.104368] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 06/14/2023]
Abstract
Exosomes (membrane-derived vesicles) enable intracellular communication by delivering lipids, proteins, DNA, and RNA from one cell to another. Highly pathogenic avian influenza virus (HPAIV) H5N1 causes considerable economic loss in the poultry industry and poses a public health concern. The host innate immune system defends against H5N1 infection by activating antiviral immune responses. This study aimed to demonstrated that immunomodulatory effects of exosomes from HPAIV H5N1-infected White Leghorn chickens on chicken macrophages, fibroblasts, T cell, and B cell lines. The expression of type I interferons (IFN-α and -β) were highly upregulated in immune-related cell lines after treatment with exosomes derived from H5N1-infected chickens. Levels of pro-inflammatory cytokines, such as IFN-γ, IL-1β, and CXCL8, were also elevated by the exosomes. The mitogen-activated protein kinase (MAPK) signaling pathway was stimulated in immune-related cells by such exosomes via phosphorylation of extracellular regulated kinases 1/2 and p38 signaling molecules. Furthermore, the H5N1 viral proteins, nucleoprotein (NP) and non-structural protein (NS1), were packaged in exosomes and successfully transferred to non-infected immune-related cells. Therefore, exosomes from H5N1-infected chickens induced pro-inflammatory cytokine expression and stimulated the MAPK signaling pathway by delivering key viral proteins. These findings would aid better understanding of the mechanism underlying the modulation of antiviral immune responses of host immune-related cells by viral-protein-carrying exosomes and support their further application as a novel exosome-based H5N1 AIV vaccine platform.
Collapse
Affiliation(s)
- Yeojin Hong
- Department of Animal Science and Technology, Chung-Ang University, Anseong, 17546, Republic of Korea
| | - Anh Duc Truong
- Department of Biochemistry and Immunology, National Institute of Veterinary Research, 86 Truong Chinh, Dong Da, Hanoi, 100000, Viet Nam
| | - Thi Hao Vu
- Department of Animal Science and Technology, Chung-Ang University, Anseong, 17546, Republic of Korea
| | - Sooyeon Lee
- Department of Animal Science and Technology, Chung-Ang University, Anseong, 17546, Republic of Korea
| | - Jubi Heo
- Department of Animal Science and Technology, Chung-Ang University, Anseong, 17546, Republic of Korea
| | - Suyeon Kang
- Department of Animal Science and Technology, Chung-Ang University, Anseong, 17546, Republic of Korea
| | - Hyun S Lillehoj
- Animal Biosciences and Biotechnology Laboratory, Agricultural Research Services, United States Department of Agriculture, Beltsville, MD, 20705, USA
| | - Yeong Ho Hong
- Department of Animal Science and Technology, Chung-Ang University, Anseong, 17546, Republic of Korea.
| |
Collapse
|
14
|
Shi C, Zhou X, Yang W, Wu J, Bai M, Zhang Y, Zhao W, Yang H, Nagai A, Yin M, Gao X, Ding S, Zhao J. Proteomic Analysis of Plasma-Derived Extracellular Vesicles From Mice With Echinococcus granulosus at Different Infection Stages and Their Immunomodulatory Functions. Front Cell Infect Microbiol 2022; 12:805010. [PMID: 35360110 PMCID: PMC8960237 DOI: 10.3389/fcimb.2022.805010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/14/2022] [Indexed: 01/15/2023] Open
Abstract
The globally distributed cystic echinococcosis (CE) is caused by the larval stage of Echinococcus granulosus (E. granulosus), a cosmopolitan and zoonotic disease with potentially life-threatening complications in humans. The emerging roles for extracellular vesicles (EVs) in parasitic infection include transferring proteins and modifying host cell gene expression to modulate host immune responses. Few studies focused on the host-derived EVs and its protein profiles. We focused on the EVs from mouse infected with E. granulosus at different stages. ExoQuick kit was used for isolating EVs from mouse plasma and ExoEasy Maxi kit was used for isolating protoscolex culture supernatant (PCS) and hydatid cyst fluid (HCF). Firstly, EVs were characterized by transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA) and immunoblot. Secondly, the proteins of plasma EVs were identified using liquid chromatography-tandem mass spectrometry (LC–MS/MS). The resulting LC–MS/MS data were processed using Maxquant search engine (v 1.5.2.8). Tandem mass spectra were researched against the mice and E. granulosus proteins database in the NCBI. The differentially expressed proteins are performed by proteomic label-free quantitative analysis and bioinformatics. Thirdly, in vitro experiment, the results of co-culture of plasma EVs and spleen mononuclear cells showed that 7W-EVs can increase the relative abundance of regulatory T (Treg) cells and IL-10. We further verified that EVs can be internalized by CD4+ and CD8+ T cells, B cells, and myeloid-derived suppressor cells (MDSC). These results implied host-derived EVs are multidirectional immune modulators. The findings can contribute to a better understanding of the role of host-derived EVs which are the optimal vehicle to transfer important cargo into host immune system. In addition, we have found several important proteins associated with E. granulosus and identified in infected mouse plasma at different stages. Furthermore, our study further highlighted the proteomics and immunological function of EVs from mouse infected with E. granulosus protoscoleces at different infection stages. We have laid a solid foundation for the role of EVs in cystic echinococcosis in the future research and supplemented a unique dataset for this E. granulosus.
Collapse
Affiliation(s)
- Chunli Shi
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China
- Department of Molecular Biology, Shanghai Centre for Clinical Laboratory, Shanghai, China
| | - Xiaojing Zhou
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- Department of Neurology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Wenjuan Yang
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Jianwen Wu
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China
| | - Min Bai
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China
| | - Ying Zhang
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China
| | - Wei Zhao
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China
- Research Center for Medical Science and Technology, Ningxia Medical University, Yinchuan, China
- Ningxia Institute of Medical Science, Yinchuan, China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, Yinchuan, China
| | - Hui Yang
- Research Center for Medical Science and Technology, Ningxia Medical University, Yinchuan, China
- Ningxia Institute of Medical Science, Yinchuan, China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, Yinchuan, China
| | - Atsushi Nagai
- Department of Neurology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Mei Yin
- Department of Respiratory Medicine, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Xiaoping Gao
- Department of Otolaryngology Head and Neck Surgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Shuqin Ding
- Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- *Correspondence: Jiaqing Zhao, ; Shuqin Ding,
| | - Jiaqing Zhao
- School of Basic Medicine, Ningxia Medical University, Yinchuan, China
- Research Center for Medical Science and Technology, Ningxia Medical University, Yinchuan, China
- Ningxia Institute of Medical Science, Yinchuan, China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, Yinchuan, China
- *Correspondence: Jiaqing Zhao, ; Shuqin Ding,
| |
Collapse
|
15
|
Ozturk EA, Caner A. Liquid Biopsy for Promising Non-invasive Diagnostic Biomarkers in Parasitic Infections. Acta Parasitol 2022; 67:1-17. [PMID: 34176040 DOI: 10.1007/s11686-021-00444-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Liquid biopsy refers to the sampling and molecular analysis of body fluids such as blood, saliva, and urine in contrast to conventional tissue biopsies. Liquid biopsy approach can offer powerful non-invasive biomarkers (circulating markers) for diagnosis and monitoring treatment response of a variety of diseases, including parasitic infections. METHODS In this review, we concentrate on cell-free DNA (cfDNA), microRNA (miRNA), and exosomes in the published literature. RESULTS Considering the high prevalence and severity of parasitic infections worldwide, circulating biomarkers can provide a new insight into the diagnosis and prognosis of parasites in the near future. Moreover, identifying and characterizing parasite- or host-derived circulating markers are important for a better understanding of the pathogenesis of parasite infection and host-parasite relationship at the molecular level. Profiling of biomarkers for parasitic diseases is a promising potential field, though further studies and optimization strategies are required, both in vitro and in vivo. CONCLUSION In this review, we discuss three approaches in the liquid biopsy including circulating cfDNA, miRNAs, and exosomes for diagnosis and evaluation of parasites and summarize circulating biomarkers in non-invasive samples during parasitic infections.
Collapse
Affiliation(s)
- Eylem Akdur Ozturk
- Department of Parasitology, Çukurova University Faculty of Medicine, Adana, Turkey
| | - Ayse Caner
- Department of Parasitology, Ege University Faculty of Medicine, 35100, Izmir, Turkey.
- Cancer Research Center, Ege University, Izmir, Turkey.
| |
Collapse
|
16
|
Liu Z, Geng X, Zhao Q, Zhu S, Han H, Yu Y, Huang W, Yao Y, Huang B, Dong H. Effects of host vimentin on Eimeria tenella sporozoite invasion. Parasit Vectors 2022; 15:8. [PMID: 34983604 PMCID: PMC8729122 DOI: 10.1186/s13071-021-05107-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 11/19/2021] [Indexed: 12/23/2022] Open
Abstract
Background Chicken coccidiosis is a parasitic disease caused by Eimeria of Apicomplexa, which has caused great economic loss to the poultry breeding industry. Host vimentin is a key protein in the process of infection of many pathogens. In an earlier phosphorylation proteomics study, we found that the phosphorylation level of host vimentin was significantly regulated after Eimeria tenella sporozoite infection. Therefore, we explored the role of host vimentin in the invasion of host cells by sporozoites. Methods Chicken vimentin protein was cloned and expressed. We used qPCR, western blotting, and indirect immunofluorescence to detect levels of mRNA transcription, translation, and phosphorylation, and changes in the distribution of vimentin after E. tenella sporozoite infection. The sporozoite invasion rate in DF-1 cells treated with vimentin polyclonal antibody or with small interfering RNA (siRNA), which downregulated vimentin expression, was assessed by an in vitro invasion test. Results The results showed that vimentin transcription and translation levels increased continually at 6–72 h after E. tenella sporozoite infection, and the total phosphorylation levels of vimentin also changed. About 24 h after sporozoite infection, vimentin accumulated around sporozoites in DF-1 cells. Treating DF-1 cells with vimentin polyclonal antibody or downregulating vimentin expression by siRNA significantly improved the invasion efficiency of sporozoites. Conclusion In this study, we showed that vimentin played an inhibitory role during the invasion of sporozoites. These data provided a foundation for clarifying the relationship between Eimeria and the host. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13071-021-05107-4.
Collapse
Affiliation(s)
- Zhan Liu
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Minhang, 200241, Shanghai, People's Republic of China
| | - Xiangfei Geng
- Beijing YuanDa Spark Medicine Technology Co., Ltd, Beijing, 100088, People's Republic of China
| | - Qiping Zhao
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Minhang, 200241, Shanghai, People's Republic of China
| | - Shunhai Zhu
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Minhang, 200241, Shanghai, People's Republic of China
| | - Hongyu Han
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Minhang, 200241, Shanghai, People's Republic of China
| | - Yu Yu
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Minhang, 200241, Shanghai, People's Republic of China
| | - Wenhao Huang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Minhang, 200241, Shanghai, People's Republic of China
| | - Yawen Yao
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Minhang, 200241, Shanghai, People's Republic of China
| | - Bing Huang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Minhang, 200241, Shanghai, People's Republic of China
| | - Hui Dong
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Minhang, 200241, Shanghai, People's Republic of China.
| |
Collapse
|
17
|
O’Dowd K, Sánchez L, Ben Salem J, Beaudry F, Barjesteh N. Characterization of the Role of Extracellular Vesicles Released from Chicken Tracheal Cells in the Antiviral Responses against Avian Influenza Virus. MEMBRANES 2021; 12:membranes12010053. [PMID: 35054579 PMCID: PMC8780788 DOI: 10.3390/membranes12010053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/24/2021] [Accepted: 12/26/2021] [Indexed: 11/16/2022]
Abstract
During viral respiratory infections, the innate antiviral response engages a complex network of cells and coordinates the secretion of key antiviral factors, such as cytokines, which requires high levels of regulation and communication. Extracellular vesicles (EVs) are particles released from cells that contain an array of biomolecules, including lipids, proteins, and RNAs. The contents of EVs can be influenced by viral infections and may play a role in the regulation of antiviral responses. We hypothesized that the contents of EVs released from chicken tracheal cells are influenced by viral infection and that these EVs regulate the function of other immune cells, such as macrophages. To this end, we characterized the protein profile of EVs during avian influenza virus (AIV) infection and evaluated the impact of EV stimulation on chicken macrophage functions. A total of 140 differentially expressed proteins were identified upon stimulation with various stimuli. These proteins were shown to be involved in immune responses and cell signaling pathways. In addition, we demonstrated that EVs can activate macrophages. These results suggest that EVs play a role in the induction and modulation of antiviral responses during viral respiratory infections in chickens.
Collapse
Affiliation(s)
- Kelsey O’Dowd
- Research Group on Infectious Diseases in Production Animals (GREMIP), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada;
- Swine and Poultry Infectious Disease Research Center (CRIPA), Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada;
| | - Laura Sánchez
- Swine and Poultry Infectious Disease Research Center (CRIPA), Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada;
| | - Jennifer Ben Salem
- Animal Pharmacology Research Group of Quebec (GREPAQ), Department of Veterinary Medicine, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada; (J.B.S.); (F.B.)
- Centre de Recherche sur le Cerveau et L’apprentissage (CIRCA), Université de Montréal, Montreal, QC J2S 2M2, Canada
| | - Francis Beaudry
- Animal Pharmacology Research Group of Quebec (GREPAQ), Department of Veterinary Medicine, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada; (J.B.S.); (F.B.)
- Centre de Recherche sur le Cerveau et L’apprentissage (CIRCA), Université de Montréal, Montreal, QC J2S 2M2, Canada
| | - Neda Barjesteh
- Research Group on Infectious Diseases in Production Animals (GREMIP), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada;
- Swine and Poultry Infectious Disease Research Center (CRIPA), Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada;
- Correspondence:
| |
Collapse
|
18
|
Evaluation of immunoprotective effects of recombinant proteins and DNA vaccines derived from Eimeria tenella surface antigen 6 and 15 in vivo. Parasitol Res 2021; 121:235-243. [PMID: 34816300 DOI: 10.1007/s00436-021-07364-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 10/26/2021] [Indexed: 10/19/2022]
Abstract
Coccidiosis is an intestinal parasitic disease that causes huge economic losses to the poultry industry globally. Eimeria tenella belonging to protozoon is the causative agent of cecal coccidiosis in chicken, and it causes enormous damage to poultry industry. The surface antigens (SAGs) of apicomplexan parasites have functions of attachment and invasion in host-parasite interaction. As a result of parasitic invasion, host immune response is triggered. However, the immunogenicity and potency of E. tenella surface antigen 6 and 15 (EtSAG 6 and 15), as vaccinal candidate antigen, remain largely unknown. Therefore, gene fragments of E. tenella EtSAG 6 and 15 were amplified and transformed to pET28a prokaryotic vector for recombinant protein expression. The pEGFP-N1 eukaryotic vectors with EtSAG 6 and 15 amplification fragments (pEGFP-N1-EtSAG 5 and 16) were transformed into 293 T cell line. The results of reverse transcription-polymerase chain reaction (RT-PCR) and western blot analysis revealed successful expressions of EtSAG 6 and 15 in Escherichia coli and 293 T cells. Subsequently, animal experiments of 49 cobb broilers were performed to evaluate immunoprotection of recombinant proteins and DNA vaccines derived from E. tenella EtSAG 5 and 16 with an immunizing dose of 100 μg, respectively. Chickens vaccinated with rEtSAG 6 protein, rEtSAG 15 protein, pEGFP-N1-EtSAG 6 plasmid, or pEGFP-N1-EtSAG 15 plasmid showed no significant increase in IFN-γor interleukin-4 (IL-4) level compared with control groups. Chickens vaccinated with protein rEtSAG 6, protein rEtSAG 15, pEGFP-N1-EtSAG 6 plasmid, or pEGFP-N1-EtSAG 15 exhibited higher weight gains, lower oocyst output, and lower mean lesion scores, compared with infection control group. Among the four immunized groups, plasmid EGFP-N1-EtSAG 6 (100 μg) group exhibited the highest anticoccidial index (ACI) value (150.20). Overall, plasmids EGFP-N1-EtSAG 6 and 15, as DNA vaccines, provided a more effective immunoprotection for chickens against E. tenella than protein rEtSAG 6 and protein rEtSAG 15 as subunit vaccines. EtSAG 6 and 15 are promising candidate antigen genes for developing coccidiosis vaccine.
Collapse
|
19
|
Hong Y, Lee J, Vu TH, Lee S, Lillehoj HS, Hong YH. Immunomodulatory effects of poly(I:C)-stimulated exosomes derived from chicken macrophages. Poult Sci 2021; 100:101247. [PMID: 34174563 PMCID: PMC8242060 DOI: 10.1016/j.psj.2021.101247] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/14/2021] [Accepted: 05/02/2021] [Indexed: 11/13/2022] Open
Abstract
Exosomes are small membrane vesicles that contain proteins and nucleic acids derived from secretory cells and mediate intracellular communication. Immune cell-derived exosomes regulate immune responses and gene expression of recipient cells. Macrophages recognize viral dsRNA via Toll-like receptor 3, thereby inducing the activation of transcription factors such as interferon regulatory factor 3 and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). In this study, we aimed to identify the immunomodulatory functions of exosomes derived from chicken macrophages (HD11) stimulated with polyinosinic-polycytidylic acid (poly[I:C]); exosomes were then delivered into HD11 cells and CU91 chicken T cells. Exosomes purified from poly(I:C)-activated macrophages stimulated the expression of type I interferons, proinflammatory cytokines, anti-inflammatory cytokines, and chemokines in HD11 and CU91 cells. Moreover, poly(I:C)-stimulated exosomes induced the NF-κB signaling pathway by phosphorylating TAK1 and NF-κB1. Therefore, we suggest that after the activation of Toll-like receptor 3 ligands following infection with dsRNA virus, chicken macrophages regulate the immune response of naive macrophages and T cells through the NF-κB signaling pathway. Furthermore, poly(I:C)-activated exosomes can be potentially utilized as immunostimulators.
Collapse
Affiliation(s)
- Yeojin Hong
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Jiae Lee
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Thi Hao Vu
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Sooyeon Lee
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Hyun S Lillehoj
- Animal Biosciences and Biotechnology Laboratory, Agricultural Research Services, United States Department of Agriculture, Beltsville, MD 20705, USA
| | - Yeong Ho Hong
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea.
| |
Collapse
|
20
|
Exosome-Based Vaccines: Pros and Cons in the World of Animal Health. Viruses 2021; 13:v13081499. [PMID: 34452364 PMCID: PMC8402771 DOI: 10.3390/v13081499] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/15/2022] Open
Abstract
Due to the emergence of antibiotic resistance and new and more complex diseases that affect livestock animal health and food security, the control of epidemics has become a top priority worldwide. Vaccination represents the most important and cost-effective measure to control infectious diseases in animal health, but it represents only 23% of the total global animal health market, highlighting the need to develop new vaccines. A recent strategy in animal health vaccination is the use of extracellular vesicles (EVs), lipid bilayer nanovesicles produced by almost all living cells, including both prokaryotes and eukaryotes. EVs have been evaluated as a prominent source of viral antigens to elicit specific immune responses and to develop new vaccination platforms as viruses and EVs share biogenesis pathways. Preliminary trials with lymphocytic choriomeningitis virus infection (LCMV), porcine reproductive and respiratory syndrome virus (PRRSV), and Marek's disease virus (MDV) have demonstrated that EVs have a role in the activation of cellular and antibody immune responses. Moreover, in parasitic diseases such as Eimeria (chickens) and Plasmodium yoelii (mice) protection has been achieved. Research into EVs is therefore opening an opportunity for new strategies to overcome old problems affecting food security, animal health, and emerging diseases. Here, we review different conventional approaches for vaccine design and compare them with examples of EV-based vaccines that have already been tested in relation to animal health.
Collapse
|
21
|
Zhao P, Wang C, Ding J, Zhao C, Xia Y, Hu Y, Zhang L, Zhou Y, Zhao J, Fang R. Evaluation of immunoprotective effects of recombinant protein and DNA vaccine based on Eimeria tenella surface antigen 16 and 22 in vivo. Parasitol Res 2021; 120:1861-1871. [PMID: 33689009 PMCID: PMC7943400 DOI: 10.1007/s00436-021-07105-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/25/2021] [Indexed: 12/05/2022]
Abstract
Coccidiosis triggered by Eimeria tenella is accompanied by haemorrhagic caecum and high morbidity. Vaccines are preferable choices to replace chemical drugs against coccidiosis. Surface antigens of apicomplexan parasites can adhere to host cells during the infection process. Therefore, truncated fragments coding E. tenella surface antigen 16 (EtSAG16) and 22 (EtSAG22) were cloned into pET-28a prokaryotic vector to express recombinant protein 16 (rEtSAG16) and 22 (rEtSAG22), respectively. Likewise, pEGFP-N1-EtSAG16 and pEGFP-N1-EtSAG22 plasmids were constructed using pEGFP-N1 eukaryotic vector. Further, pEGFP-N1-EtSAG4-16-22 multiple gene plasmid carrying EtSAG4, 16 and 22 were designed as cocktail vaccines to study integral immunoprotective effects. Western blot and RT-PCR (reverse transcription) assay were performed to verify expressions of EtSAG16 and 22 genes. Immunoprotective effects of recombinant protein or DNA vaccine were evaluated using different doses (50 or 100 μg) in vivo. All chickens in the vaccination group showed higher cytokine concentration (IFN-γ and IL-17), raised IgY antibody level, increased weight gain, lower caecum lesion score and reduced oocyst shedding compared with infection control groups (p < 0.05). The highest anticoccidial index (ACI) value 173.11 was from the pEGFP-N1-EtSAG4-16-22 plasmid (50 μg) group. In conclusion, EtSAG16 and 22 might be alternative candidate genes for generating vaccines against E. tenella infection.
Collapse
Affiliation(s)
- Pengfei Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070 Hubei China
| | - Chaofei Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070 Hubei China
| | - Jun Ding
- Animal Disease Prevention and Control Center, Jingshan, 431800 Hubei China
| | - Chengfeng Zhao
- Animal Disease Prevention and Control Center, Anlu, 432600 Hubei China
| | - Yingjun Xia
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070 Hubei China
| | - Yanli Hu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070 Hubei China
| | - Li Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070 Hubei China
| | - Yanqin Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070 Hubei China
| | - Junlong Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070 Hubei China
| | - Rui Fang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070 Hubei China
| |
Collapse
|
22
|
Hong Y, Lee J, Vu TH, Lee S, Lillehoj HS, Hong YH. Exosomes of lipopolysaccharide-stimulated chicken macrophages modulate immune response through the MyD88/NF-κB signaling pathway. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 115:103908. [PMID: 33115603 DOI: 10.1016/j.dci.2020.103908] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/22/2020] [Accepted: 10/22/2020] [Indexed: 06/11/2023]
Abstract
Exosomes are small membrane-extracellular vesicles produced from multivesicular bodies and play a role in cell-to-cell signaling. Exosomes from immune cells can regulate immune responses of recipient cells by releasing their contents. In the immune system, macrophages recognize lipopolysaccharides (LPSs) of gram-negative bacteria by toll-like receptor 4 (TLR4) and intracellular pathways, such as NF-κB pathway, are activated, inducing proinflammatory cytokine expression. However, no studies have investigated the functions of exosomes in chicken macrophages. The purpose of this study was to demonstrate the immunoregulatory functions of LPS-activated exosomes in chicken immune systems. Therefore, chicken macrophages cells (HD11) were activated with LPS, and exosomes were purified. The LPS-activated exosomes enhanced the gene expression of cytokines and chemokines, including IL-1β, IFN-γ, IFN-α, IL-4, CCL4, CCL17, and CCL19, in naive chicken macrophages. Furthermore, LPS-activated exosomes induced the MyD88/NF-κB signaling pathway. Therefore, as an immune response against gram-negative bacterial infection, LPS-activated chicken macrophages can release exosomes that are delivered to inactivated macrophages by regulating the expression of immune-related genes and the MyD88/NF-κB signaling pathway. In the future, LPS-stimulated exosomes may be utilized as an immune stimulator.
Collapse
Affiliation(s)
- Yeojin Hong
- Department of Animal Science and Technology, Chung-Ang University, Anseong, 17546, Republic of Korea
| | - Jiae Lee
- Department of Animal Science and Technology, Chung-Ang University, Anseong, 17546, Republic of Korea
| | - Thi Hao Vu
- Department of Animal Science and Technology, Chung-Ang University, Anseong, 17546, Republic of Korea
| | - Sooyeon Lee
- Department of Animal Science and Technology, Chung-Ang University, Anseong, 17546, Republic of Korea
| | - Hyun S Lillehoj
- Animal Biosciences and Biotechnology Laboratory, Agricultural Research Services, United States Department of Agriculture, Beltsville, MD, 20705, USA
| | - Yeong Ho Hong
- Department of Animal Science and Technology, Chung-Ang University, Anseong, 17546, Republic of Korea.
| |
Collapse
|
23
|
Xu Z, Zheng M, Liu Y, Zhang L, Zhang X, Bai R. Roles of TNF receptor-associated and Fas-associated death domain proteins in the apoptosis of Eimeria tenella host cells. Vet Parasitol 2021; 290:109351. [PMID: 33476903 DOI: 10.1016/j.vetpar.2021.109351] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 12/30/2020] [Accepted: 01/02/2021] [Indexed: 12/22/2022]
Abstract
The present study aimed to investigate the effects of death receptor adapter proteins, namely, TNF receptor-associated death domain (TRADD) and Fas-associated death domain (FADD) proteins, on Eimeria tenella-induced host cell apoptosis. Gene silencing, culture technique for primary chick embryo cecal epithelial cells, enzyme-linked immunosorbent assay, Hoechst-Annexin V/PI apoptosis staining, fluorescence quantitative PCR, and flow cytometry were used to detect the E. tenella host cell apoptotic rate, RIP1 and FADD protein expression levels, and caspase-8 activity of the TRADD siRNA-treated and FADD siRNA-treated groups. Results showed that the apoptotic rate in the TRADD siRNA group was significantly higher than that in the NC siRNA group at 4 h post-infection with E. tenella (P < 0.05). The RIP1 protein expression level in the TRADD siRNA group was significantly lower than that in the NC siRNA group at 4-24 h (P < 0.05). The FADD expression and apoptotic rates in the TRADD siRNA group were significantly lower than those in the NC siRNA group at 24-120 h (P < 0.05). The caspase-8 activity and apoptotic rates in the FADD siRNA group were significantly lower than those in the NC siRNA group (P < 0.05) at 24-120 h. These findings indicated that E. tenella inhibited the host cell apoptosis through the TRADD-RIP1 pathway at the early developmental stage and promoted host cell apoptosis via the TRADD-FADD-caspase-8 apoptotic pathway at the middle and late developmental stages.
Collapse
Affiliation(s)
- ZhiYong Xu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, China; College of Animal Science and Technology, Henan Institute of Science and Technology, Xinxiang, 453003, China
| | - MingXue Zheng
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, China.
| | - Yan Liu
- Army Eighty-Three Army Hospital, Xinxiang, 453000, China
| | - Li Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, China
| | - XueSong Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, China
| | - Rui Bai
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, China
| |
Collapse
|
24
|
Mtshali SA, Adeleke MA. A review of adaptive immune responses to Eimeria tenella and Eimeria maxima challenge in chickens. WORLD POULTRY SCI J 2020. [DOI: 10.1080/00439339.2020.1833693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- S. A. Mtshali
- Discipline of Genetics, School of Life Sciences, University of Kwa-Zulu Natal, Durban, South Africa
| | - M. A. Adeleke
- Discipline of Genetics, School of Life Sciences, University of Kwa-Zulu Natal, Durban, South Africa
| |
Collapse
|
25
|
Recent Advances in Extracellular Vesicles as Drug Delivery Systems and Their Potential in Precision Medicine. Pharmaceutics 2020; 12:pharmaceutics12111006. [PMID: 33105857 PMCID: PMC7690579 DOI: 10.3390/pharmaceutics12111006] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) are membrane-bilayered nanoparticles released by most cell types. Recently, an enormous number of studies have been published on the potential of EVs as carriers of therapeutic agents. In contrast to systems such as liposomes, EVs exhibit less immunogenicity and higher engineering potential. Here, we review the most relevant publications addressing the potential and use of EVs as a drug delivery system (DDS). The information is divided based on the key steps for designing an EV-mediated delivery strategy. We discuss possible sources and isolation methods of EVs. We address the administration routes that have been tested in vivo and the tissue distribution observed. We describe the current knowledge on EV clearance, a significant challenge towards enhancing bioavailability. Also, EV-engineering approaches are described as alternatives to improve tissue and cell-specificity. Finally, a summary of the ongoing clinical trials is performed. Although the application of EVs in the clinical practice is still at an early stage, a high number of studies in animals support their potential as DDS. Thus, better treatment options could be designed to precisely increase target specificity and therapeutic efficacy while reducing off-target effects and toxicity according to the individual requirements of each patient.
Collapse
|
26
|
Zhang M, Qiu J, Shu X, Tang X, Sha X, Wu L, Fan J, Zeng D, He R, Zhang W, Zeng Z, Liu R, He L. Pharmacokinetics, Activity, and Residue Elimination of R- and S-Diclazuril in Broiler Chickens. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:8987-8995. [PMID: 32692165 DOI: 10.1021/acs.jafc.0c03091] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Diclazuril (DIC) is widely used as a racemic mixture to prevent and treat coccidiosis in farm animals, while the pharmacokinetics, bioactivity, and toxicity of DIC enantiomers are not known at all. This study first established a simple, sensitive, and reliable liquid chromatography tandem mass spectrometry method for separation of R-DIC and S-DIC and their analyses. Then, it was applied to investigate the stereoselective pharmacokinetics and residual elimination of individual enantiomers, and their anticoccidial activity was also evaluated in broiler chickens. The results indicated that the area under the concentration-time curve (AUC) and elimination half-life (t1/2β) were significantly different (p < 0.05) for two enantiomers in chicken plasma. The AUC and t1/2β of S-DIC were approximately 2 and 1.4 times those of R-DIC, respectively. The residual elimination of DIC enantiomers in chicken tissues was also stereoselective. The concentrations of S-DIC in chicken muscle and liver were greater than those of R-DIC, and it is the opposite in the kidney. There was no significant difference (p > 0.05) in the anticoccidial activity of racemate and enantiomers when a single enantiomer in feed was added above 0.5 mg kg-1. However, the anticoccidial activity of R-DIC (0.25 mg kg-1) was significantly higher (p < 0.05) than that of S-DIC (0.25 mg kg-1) in the diet. It should be mentioned that in chicken small intestine and cecum, the enantiomerization rate of each enantiomer in the infection group was faster than that in the uninfected group.
Collapse
Affiliation(s)
- Meiyu Zhang
- National Reference Laboratory of Veterinary Drug Residues, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Jingli Qiu
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Xiaogui Shu
- National Reference Laboratory of Veterinary Drug Residues, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Xiaoling Tang
- National Reference Laboratory of Veterinary Drug Residues, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Xuefang Sha
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Li Wu
- Guangdong Huanong Hi-Tech Bio-Pharmaceutical Co. Limited, Guangzhou 510642, China
| | - Jun Fan
- School of Chemistry, South China Normal University, Guangzhou 510006, China
| | - Dongping Zeng
- National Reference Laboratory of Veterinary Drug Residues, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Rujian He
- School of Chemistry, South China Normal University, Guangzhou 510006, China
| | - Weiguang Zhang
- School of Chemistry, South China Normal University, Guangzhou 510006, China
| | - Zhenling Zeng
- National Reference Laboratory of Veterinary Drug Residues, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Rong Liu
- National Reference Laboratory of Veterinary Drug Residues, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Limin He
- National Reference Laboratory of Veterinary Drug Residues, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| |
Collapse
|
27
|
Recombinant invasive Lactobacillus plantarum expressing the Eimeria tenella fusion gene TA4 and AMA1 induces protection against coccidiosis in chickens. Vet Parasitol 2020; 283:109161. [PMID: 32526607 DOI: 10.1016/j.vetpar.2020.109161] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 05/29/2020] [Accepted: 05/30/2020] [Indexed: 01/29/2023]
Abstract
Coccidiosis is an intestinal parasitic disease that is caused by Eimeria tenella and other species, and it seriously restricts the economic development of the broiler breeding industry. In this study, a recombinant Lactobacillus plantarum with an invasive effect was constructed, and it expressed the TA4-AMA1 protein of E. tenella. After oral immunization with recombinant L. plantarum, specific humoral and mucosal immune levels were measured by indirect ELISA, and the differentiation of T cells was analysed by flow cytometry. After challenge with sporulated oocysts, the body weight, oocyst shedding and cecum lesions of the chicken were evaluated. The results indicated that chickens immunized with recombinant invasive L. plantarum produced higher levels of specific antibodies in the serum than did the non-immunized controls, and the secretory IgA (sIgA) levels were increased in the intestinal washes compared to those of the controls (P < 0.05). Flow cytometry showed that recombinant invasive L. plantarum significantly stimulated T cell differentiation compared to the PBS group (P < 0.01, P < 0.001), and a higher proportion of CD4+ and CD8+ T cells were detected in peripheral blood. Moreover, the lesion scores and histopathological caecum sections showed that immunizing chickens with recombinant invasive L. plantarum can significantly relieve pathological damage in the cecum (P < 0.01), and the relative body weight gain was 89.64 %, which was higher than the 79.83 % gain in the chickens immunized with non-invasive L. plantarum. After the challenge, faeces from ten chickens in each group were collected between 4 and 7 days, and the oocysts per gram (OPG) was determined by the McMaster technique. The data indicated that oocysts in the faeces of chickens immunized with the recombinant invasive L. plantarum were significantly lower than those of the controls (P < 0.01). The results suggest that recombinant invasive L. plantarum effectively activated immune responses against E. tenella infection and can be used as a candidate vaccine against E. tenella infection.
Collapse
|
28
|
Lu M, Huang Y. Bioinspired exosome-like therapeutics and delivery nanoplatforms. Biomaterials 2020; 242:119925. [PMID: 32151860 DOI: 10.1016/j.biomaterials.2020.119925] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/09/2020] [Accepted: 02/26/2020] [Indexed: 02/08/2023]
Abstract
Exosomes have emerged as appealing candidate therapeutic agents and delivery nanoplatforms due to their endogenous features and unique biological properties. However, obstacles such as low isolation yield, considerable complexity and potential safety concerns, and inefficient drug payload substantially hamper their therapeutic applicability. To this end, developing bioinspired exosome-like nanoparticles has become a promising area to overcome certain limitations of their natural counterparts. Synthetically fabrication of exosome-like nanoparticles that harbor only crucial components of exosomes through controllable protocols strongly increases the pharmaceutical acceptability of these vesicles. Assembly of exosome-like nanovesicles derived from producer cells allows for a promising strategy for scale-up production. To improve the loading capability and delivery efficiency of exosomes, hybrid exosome-like nanovesicles and membrane-camouflaged nanoparticles towards better bridging synthetic nanocarriers with natural exosomes could be designed. Building off these observations, herein, efforts are made to give an overview of bioinspired exosome-like therapeutics and delivery nanoplatforms. We briefly recapitulate the recent advance in exosome biology with focus on tailoring exosomes as therapeutics and delivery vehicles. Furthermore, we elaborately discuss the biomimicry methodologies for preparation of exosome-like nanoparticles with special emphasis on offering insights into strategies for rational design of exosome-like biomaterials as effective and safe therapeutics and delivery nanoplatforms.
Collapse
Affiliation(s)
- Mei Lu
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Institute of Engineering Medicine, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, 100081, PR China
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Institute of Engineering Medicine, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, 100081, PR China.
| |
Collapse
|
29
|
Khosravi M, Mirsamadi ES, Mirjalali H, Zali MR. Isolation and Functions of Extracellular Vesicles Derived from Parasites: The Promise of a New Era in Immunotherapy, Vaccination, and Diagnosis. Int J Nanomedicine 2020; 15:2957-2969. [PMID: 32425527 PMCID: PMC7196212 DOI: 10.2147/ijn.s250993] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 04/10/2020] [Indexed: 12/13/2022] Open
Abstract
Experimental and epidemiological evidence shows that parasites, particularly helminths, play a central role in balancing the host immunity. It was demonstrated that parasites can modulate immune responses via their excretory/secretory (ES) and some specific proteins. Extracellular vesicles (EVs) are nano-scale particles that are released from eukaryotic and prokaryotic cells. EVs in parasitological studies have been mostly employed for immunotherapy of autoimmune diseases, vaccination, and diagnosis. EVs can carry virulence factors and play a central role in the development of parasites in host cells. These molecules can manipulate the immune responses through transcriptional changes. Moreover, EVs derived from helminths modulate the immune system via provoking anti-inflammatory cytokines. On the other hand, EVs from parasite protozoa can induce efficient immunity, that makes them useful for probable next-generation vaccines. In addition, it seems that EVs from parasites may provide new diagnostic approaches for parasitic infections. In the current study, we reviewed isolation methods, functions, and applications of parasite's EVs in immunotherapy, vaccination, and diagnosis.
Collapse
Affiliation(s)
- Mojdeh Khosravi
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Valencia, Spain
| | - Elnaz Sadat Mirsamadi
- Department of Microbiology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hamed Mirjalali
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Panebra A, Lillehoj HS. Eimeria tenella Elongation Factor-1α (EF-1α) Coadministered with Chicken IL-7 (chIL-7) DNA Vaccine Emulsified in Montanide Gel 01 Adjuvant Enhanced the Immune Response to E. acervulina Infection in Broiler Chickens. Avian Dis 2020; 63:342-350. [PMID: 31251536 DOI: 10.1637/11976-092418-reg.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 02/07/2019] [Indexed: 11/05/2022]
Abstract
The current study was undertaken to assess the vaccine efficacy of Eimeria tenella EF-1α/chicken IL-7 (chIL-7) DNA vaccine when administered with Montanide Gel 01 adjuvant against live Eimeria acervulina challenge in commercial broiler chickens. The criteria used for the evaluation of vaccine efficacy were weight gain, duodenal lesion scores, oocyst counts, humoral antibody response, and duodenal proinflammatory cytokine gene expression. Chickens vaccinated with EF-1α (100 µg)/chIL-7 (20 µg) in Gel 01 PR adjuvant showed body weight gain similar to the uninfected control and higher oocyst shedding, a lower gut lesion score, and higher proinflammatory cytokine gene expression than did the infected controls. Moreover, chickens vaccinated with chIL-7 (20 µg) in Gel 01 PR adjuvant shed fewer oocysts with reduced gut lesion scores and produced higher levels of anti-EF-1α serum antibody than did the infected control. Chickens vaccinated with EF-1α (50 µg)/chIL-7 (20 µg) in Gel 01 PR adjuvant showed higher weight gains than did the infected control and shed significantly fewer oocysts than the infected control. Furthermore, chickens vaccinated with EF-1α (100 µg) in Gel 01 PR adjuvant demonstrated the lowest anti-EF-1α serum antibody levels. This study demonstrated the beneficial effects of using EF-1α and/or host cytokine chIL-7 DNA vaccine together with Gel 01 PR adjuvant to improve T-cell-mediated effector function in broiler chickens challenged with live E. acervulina.
Collapse
Affiliation(s)
- Alfredo Panebra
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Service, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD 20705
| | - Hyun S Lillehoj
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Service, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD 20705,
| |
Collapse
|
31
|
Zmrhal V, Slama P. Current knowledge about interactions between avian dendritic cells and poultry pathogens. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 104:103565. [PMID: 31830703 DOI: 10.1016/j.dci.2019.103565] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/03/2019] [Accepted: 12/03/2019] [Indexed: 06/10/2023]
Abstract
In poultry production conditions today, birds are surrounded by viral, bacterial, and parasitic agents. DCs are the main antigen-presenting cells located in tissues of the body, and their role involves recognizing antigen structures, engulfing and processing them, and subsequently presenting antigen peptides on their surface by major histocompatibility complex, where T cells and B cells are stimulated and can begin appropriate cellular and antibody immune response. This unique function indicates that these cells can be used in producing vaccines, but first it is necessary to culture DCs in vitro to identify the principles of their interactions with pathogens. The following review summarizes our current knowledge about avian dendritic cells and their interactions with pathogens. It provides a basis for future studies of these unique cells and their use in vaccine development.
Collapse
Affiliation(s)
- Vladimir Zmrhal
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Zemedelska 1, 613 00, Brno, Czech Republic
| | - Petr Slama
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Zemedelska 1, 613 00, Brno, Czech Republic.
| |
Collapse
|
32
|
Zhao N, Lv J, Lu Y, Jiang Y, Li H, Liu Y, Zhang X, Zhao X. Prolonging and enhancing the protective efficacy of the EtMIC3-C-MAR against eimeria tenella through delivered by attenuated salmonella typhimurium. Vet Parasitol 2020; 279:109061. [PMID: 32143014 DOI: 10.1016/j.vetpar.2020.109061] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 02/20/2020] [Accepted: 02/23/2020] [Indexed: 01/09/2023]
Abstract
The microneme adhesive repeats (MAR) of Eimeria tenella microneme protein 3 (EtMIC3) are associated with binding to and invasion of host cells. Adhesion and invasion-related proteins or domains are often strongly immunogenic, immune responses mounted against these factors that play a key role in blocking invasion. In the present study, an oral live vaccine consisting of attenuated Salmonella typhimurium X4550 carrying two MAR domains fragment (St-X4550-MAR) was constructed and its protective efficacies were evaluated. The results showed that St-X4550-MAR was more immunogenic and conferred a higher degree of protection than recombinant MAR polypeptide as reflected by increased body weight, decreased oocyst shedding and lesion scores, increased serum IgG and cecal sIgA antibody production, and increasing levels of interferon-γ and interleukin-10. Thus, MAR domains are highly immunogenic and St-X4550-MAR had moderate activity against E. tenella infection by stimulating humoral, mucosal and cellular immunity. Chickens immunized with our constructed live vaccine provided considerable protections as early as at 10 d post-immunization (ACI: 155.17), and maintained higher protection levels at 20 d post-immunization (ACI: 173.66), and at 30 d post-immunization (ACI: 162.4). While the protective efficacy of chickens immunized with the recombinant MAR peptides showed a decreased trend as the post immunization time prolonging. Thus, using live-attenuated S. typhimurium X4550 as a vaccine expression and delivery system can significantly improve the protective efficacy and duration of protective immunity of MAR of EtMIC3.
Collapse
Affiliation(s)
- Ningning Zhao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian City, Shandong Province, China
| | - Junfeng Lv
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian City, Shandong Province, China
| | - Yaru Lu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian City, Shandong Province, China
| | - Yingying Jiang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian City, Shandong Province, China
| | - Hongmei Li
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian City, Shandong Province, China
| | - Yumin Liu
- Shandong Huamutianyuan Agriculture and Animal Husbandry Co., Ltd., 1 Gangxing 3 Road, Jinan, Shandong Province, 250101, China
| | - Xiao Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian City, Shandong Province, China.
| | - Xiaomin Zhao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian City, Shandong Province, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian City, Shandong Province, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Taian City, Shandong Province, 271018, China.
| |
Collapse
|
33
|
Aguanta BN, Fuller AL, Milfort MC, Williams SM, Rekaya R, Aggrey SE. Histologic Effects of Concurrent Heat Stress and Coccidial Infection on the Lymphoid Tissues of Broiler Chickens. Avian Dis 2019; 62:345-350. [PMID: 31119917 DOI: 10.1637/11907-052818-reg.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/20/2018] [Indexed: 11/05/2022]
Abstract
We characterized the histologic effects of two stressors (heat and coccidial infection) alone or in combination on bursa of Fabricius, thymus, and spleen in broiler chickens. Four hundred and eighty Cobb500 male chicks at 14 days of age were randomly assigned to two treatments in a 2×2 factorial design, with 15 replicates per treatment and eight birds per replicate. The treatment factors were temperature (25 and 35 C) and a mixed culture of 2.5 × 105 sporulated Eimeria acervulina and Eimeria maxima oocysts (infection or no infection). Histologic lesion severity was scored in these tissues at different ages. At 21 and 28 days of age, bursal and thymic tissues from birds raised at 35 C exhibited significant increases in lymphoid depletion severity compared with those raised at 25 C. No significant differences were detected in the lymphoid depletion severity of birds infected with Eimeria when compared with uninfected birds. These results indicate that continuous exposure to heat stress-inducing temperatures results in lymphoid depletion of the bursa and thymus in broiler chickens, a potential histologic marker for the immunologic changes known to arise as a result of heat stress. Bursal and thymic atrophy are thought to contribute to immunologic changes that underlie the negative effects of heat stress on poultry production characteristics.
Collapse
Affiliation(s)
- Bryan N Aguanta
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602
| | - Alberta L Fuller
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602
| | - Marie C Milfort
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602
| | - Susan M Williams
- Department of Population Health, University of Georgia, Athens, GA 30602
| | - Romdhane Rekaya
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602
| | - Samuel E Aggrey
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602,
| |
Collapse
|
34
|
Wei Z, Zhao Y, Zhang N, Han Z, Liu X, Jiang A, Zhang Y, Wang C, Gong P, Li J, Zhang X, Yang Z. Eimeria tenella induces the release of chicken heterophil extracellular traps. Vet Parasitol 2019; 275:108931. [PMID: 31605936 DOI: 10.1016/j.vetpar.2019.108931] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 09/23/2019] [Accepted: 09/23/2019] [Indexed: 01/04/2023]
Abstract
Avian coccidiosis makes a great threat and economic loss to the poultry industry, and fully understanding the innate immune response of chicken against E. tenella infection will play a significant role in avian coccidiosis prevention and treatment. Extracellular traps have been reported as a novel defense mechanism of host against pathogens infection. However, the interaction between chicken heterophil extracellular traps and E. tenella has remained not well known. Thus, this study aims to investigate the effects of E. tenella on chicken heterophil extracellular traps (ETs), and try to clarify the regulatory mechanisms in this process. E. tenella-triggered chicken heterophil ETs structures were analyzed by using scanning electron microscopy (SEM) and scanning confocal microscope. Inhibitors and Pico Green® were used to quantify E. tenella - triggered chicken heterophil ETs release. The results showed that E. tenella sporozoites significantly induced chicken heterophil ETs-like structures release, and histone and elastin co-existed with DNA in these structures of chicken heterophil ETs. Furthermore, it was also demonstrated that NADPH, p38 or Rac1 signaling pathways participated in E. tenella sporozoites-induced chicken heterophil ETs release, but more key molecules or signaling pathways involved in this process still needed to be further investigated. Taken together, this study reports that E. tenella sporozoites could induce chicken heterophil ETs formation via NADPH, p38 and Rac1 signaling pathways, which further suggests the critical role of heterophil ETs in the process of chicken against E. tenella infection.
Collapse
Affiliation(s)
- Zhengkai Wei
- College of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong Province, China; Key Laboratory of Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin province, China
| | - Yingchi Zhao
- Key Laboratory of Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin province, China
| | - Nan Zhang
- Key Laboratory of Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin province, China
| | - Zhen Han
- Key Laboratory of Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin province, China
| | - Xiao Liu
- Key Laboratory of Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin province, China
| | - Aimin Jiang
- Key Laboratory of Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin province, China
| | - Yong Zhang
- Key Laboratory of Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin province, China
| | - Chaoqun Wang
- Key Laboratory of Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin province, China
| | - Pengtao Gong
- Key Laboratory of Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin province, China
| | - Jianhua Li
- Key Laboratory of Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin province, China
| | - Xichen Zhang
- Key Laboratory of Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin province, China.
| | - Zhengtao Yang
- College of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong Province, China; Key Laboratory of Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin province, China.
| |
Collapse
|
35
|
|
36
|
Montaner-Tarbes S, Pujol M, Jabbar T, Hawes P, Chapman D, Portillo HD, Fraile L, Sánchez-Cordón PJ, Dixon L, Montoya M. Serum-Derived Extracellular Vesicles from African Swine Fever Virus-Infected Pigs Selectively Recruit Viral and Porcine Proteins. Viruses 2019; 11:v11100882. [PMID: 31547130 PMCID: PMC6832119 DOI: 10.3390/v11100882] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/13/2019] [Accepted: 09/16/2019] [Indexed: 12/14/2022] Open
Abstract
: African swine fever is a devastating hemorrhagic infectious disease, which affects domestic and wild swines (Susscrofa) of all breeds and ages, with a high lethality of up to 90-100% in naïve animals. The causative agent, African swine fever virus (ASFV), is a large and complex double-stranded DNA arbovirus which is currently spreading worldwide, with serious socioeconomic consequences. There is no treatment or effective vaccine commercially available, and most of the current research is focused on attenuated viral models, with limited success so far. Thus, new strategies are under investigation. Extracellular vesicles (EVs) have proven to be a promising new vaccination platform for veterinary diseases in situations in which conventional approaches have not been completely successful. Here, serum extracellular vesicles from infected pigs using two different ASFV viruses (OURT 88/3 and Benin ΔMGF), corresponding to a naturally attenuated virus and a deletion mutant, respectively, were characterized in order to determine possible differences in the content of swine and viral proteins in EV-enriched fractions. Firstly, EVs were characterized by their CD5, CD63, CD81 and CD163 surface expression. Secondly, ASFV proteins were detected on the surface of EVs from ASFV-infected pig serum. Finally, proteomic analysis revealed few specific proteins from ASFV in the EVs, but 942 swine proteins were detected in all EV preparations (negative controls, and OURT 88/3 and Benin ΔMGF-infected preparations). However, in samples from OURT 88/3-infected animals, only a small number of proteins were differentially identified compared to control uninfected animals. Fifty-six swine proteins (Group Benin) and seven proteins (Group OURT 88/3) were differentially detected on EVs when compared to the EV control group. Most of these were related to coagulation cascades. The results presented here could contribute to a better understanding of ASFV pathogenesis and immune/protective responses in the host.
Collapse
Affiliation(s)
- Sergio Montaner-Tarbes
- Innovex Therapeutics S.L., 08916 Badalona, Barcelona, Spain.
- Departamento de Ciència Animal, Escola Tècnica Superior d'Enginyeria Agrària, Avenida Alcalde Rovira Roure, 191, 25198 Lleida, Spain.
| | - Myriam Pujol
- Faculty of Medicine, Universidad de Chile, Santiago 7591538, Chile.
| | - Tamara Jabbar
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey GU24 0NF, UK.
| | - Philippa Hawes
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey GU24 0NF, UK.
| | - Dave Chapman
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey GU24 0NF, UK.
| | | | - Lorenzo Fraile
- Innovex Therapeutics S.L., 08916 Badalona, Barcelona, Spain.
- Departamento de Ciència Animal, Escola Tècnica Superior d'Enginyeria Agrària, Avenida Alcalde Rovira Roure, 191, 25198 Lleida, Spain.
| | | | - Linda Dixon
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey GU24 0NF, UK.
| | - Maria Montoya
- Innovex Therapeutics S.L., 08916 Badalona, Barcelona, Spain.
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey GU24 0NF, UK.
- Centro de Investigaciones Biológicas (CIB-CSIC), Universidad Complutense de Madrid, Ramiro de Maeztu 9, Madrid 28040, Spain.
| |
Collapse
|
37
|
Gao K, Jin J, Huang C, Li J, Luo H, Li L, Huang Y, Jiang Y. Exosomes Derived From Septic Mouse Serum Modulate Immune Responses via Exosome-Associated Cytokines. Front Immunol 2019; 10:1560. [PMID: 31354717 PMCID: PMC6640201 DOI: 10.3389/fimmu.2019.01560] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 06/24/2019] [Indexed: 12/12/2022] Open
Abstract
Sepsis is a life-threatening condition caused by an immune response triggered by infection, and highly elevated cytokine/chemokine levels in the blood play crucial roles in the progression of sepsis. Serum exosomes are nanovesicles that have multiple biological functions, playing roles in antigen presentation, intercellular signal communication, inflammatory response and immune surveillance. However, the biological functions and related molecular bases remain to be elucidated. In this study, we investigated the profiles of cytokines/chemokines harbored in the exosomes of septic mice and explored the mechanisms of immunomodulation on T cells treated with exosomes harvested from septic mice. Blood cytokines/chemokines existed in both the soluble form and in the insoluble exosomal form; the profiles of the cytokines/chemokines in these two forms displayed different dynamics in the blood of mice challenged with LPS. Exosomes from septic mice induced the differentiation of Th1/Th2 cells, which was blocked by specific antibodies targeting IL-12 and IL-4. In addition, these exosomes significantly augmented the proliferation and migration of T lymphocytes. Furthermore, preadministration of exosomes by intravenous injection restrained the inflammatory response, attenuated lung and liver tissue damage, and prolonged the survival of cecal ligation and puncture (CLP) mice. Our results indicate that exosomes enriched with cytokines/chemokines play critical roles in T cell differentiation, proliferation and chemotaxis during the sepsis process and have a protective effect on cecal ligation and puncture (CLP) mice. Thus, these findings not only strengthen our understanding of the role of sepsis via exosomes but also provide potential targets for therapeutic applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yong Jiang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
38
|
Tang X, Wang C, Liang L, Hu D, Zhang S, Duan C, Suo J, Liu X, Suo X, Cui S. Co-immunization with two recombinant Eimeria tenella lines expressing immunoprotective antigens of E. maxima elicits enhanced protection against E. maxima infection. Parasit Vectors 2019; 12:347. [PMID: 31300007 PMCID: PMC6626336 DOI: 10.1186/s13071-019-3605-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 07/06/2019] [Indexed: 01/23/2023] Open
Abstract
Background Live anticoccidial vaccines have been a tremendous success for disease prevention. The establishment of the reverse genetic manipulation platform has enabled the development of Eimeria parasites, the live anticoccidial vaccine strains, as vaccine vectors. In our previous study, recombinant E. tenella expressing a single immunodominant antigen of E. maxima (Et-EmIMP1) was able to protect chickens against challenge infection with E. maxima. This promising result encouraged us to further explore strategies to improve the protection efficacy of recombinant Eimeria and develop it as a vaccine vector. Results We constructed a novel recombinant Eimeria line expressing apical membrane antigen 1 of E. maxima (Et-EmAMA1) and then immunized chickens with Et-EmAMA1 and/or Et-EmIMP1. We found that the E. maxima soluble antigen-specific cell-mediated immunity was much stronger in the birds that were co-immunized with Et-EmAMA1 and Et-EmIMP1 than in those that were immunized with Et-EmAMA1 or Et-EmIMP1 alone. The oocyst production after E. maxima infection was significantly reduced in the recombinant Eimeria-immunized birds compared with the wild-type-immunized and naïve birds. The oocyst production in the birds co-immunized with Et-EmAMA1 and Et-EmIMP1 was consistently the lowest among the treatment groups after E. maxima infection. Conclusions These results demonstrated that Eimeria is an effective vaccine vector that can carry and deliver heterologous Eimeria antigens to the host immune system and trigger specific immune responses. Our results also suggested that increasing the number of recombinant Eimeria lines is an effective approach to enhance protective immunity against infections with heterologous pathogens.
Collapse
Affiliation(s)
- Xinming Tang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Chaoyue Wang
- Key Laboratory of Zoonosis of Ministry of Agriculture & National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Lin Liang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China.,Beijing Scientific Observation and Experimental Station of Veterinary Drugs and Diagnostic Technology, Ministry of Agriculture, Beijing, China
| | - Dandan Hu
- Key Laboratory of Zoonosis of Ministry of Agriculture & National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Sixin Zhang
- Key Laboratory of Zoonosis of Ministry of Agriculture & National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Chunhui Duan
- Key Laboratory of Zoonosis of Ministry of Agriculture & National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jingxia Suo
- Key Laboratory of Zoonosis of Ministry of Agriculture & National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xianyong Liu
- Key Laboratory of Zoonosis of Ministry of Agriculture & National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xun Suo
- Key Laboratory of Zoonosis of Ministry of Agriculture & National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Shangjin Cui
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China. .,Beijing Scientific Observation and Experimental Station of Veterinary Drugs and Diagnostic Technology, Ministry of Agriculture, Beijing, China.
| |
Collapse
|
39
|
Montaner-Tarbes S, Del Portillo HA, Montoya M, Fraile L. Key Gaps in the Knowledge of the Porcine Respiratory Reproductive Syndrome Virus (PRRSV). Front Vet Sci 2019; 6:38. [PMID: 30842948 PMCID: PMC6391865 DOI: 10.3389/fvets.2019.00038] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 01/30/2019] [Indexed: 12/11/2022] Open
Abstract
The porcine reproductive and respiratory syndrome virus (PRRSV) is one of the most important swine diseases in the world. It is causing an enormous economic burden due to reproductive failure in sows and a complex respiratory syndrome in pigs of all ages, with mortality varying from 2 to 100% in the most extreme cases of emergent highly pathogenic strains. PRRSV displays complex interactions with the immune system and a high mutation rate, making the development, and implementation of control strategies a major challenge. In this review, the biology of the virus will be addressed focusing on newly discovered functions of non-structural proteins and novel dissemination mechanisms. Secondly, the role of different cell types and viral proteins will be reviewed in natural and vaccine-induced immune response together with the role of different immune evasion mechanisms focusing on those gaps of knowledge that are critical to generate more efficacious vaccines. Finally, novel strategies for antigen discovery and vaccine development will be discussed, in particular the use of exosomes (extracellular vesicles of endocytic origin). As nanocarriers of lipids, proteins and nucleic acids, exosomes have potential effects on cell activation, modulation of immune responses and antigen presentation. Thus, representing a novel vaccination approach against this devastating disease.
Collapse
Affiliation(s)
- Sergio Montaner-Tarbes
- Innovex Therapeutics S.L, Badalona, Spain.,Departamento de Ciencia Animal, Escuela Técnica Superior de Ingenieria Agraria (ETSEA), Universidad de Lleida, Lleida, Spain
| | - Hernando A Del Portillo
- Innovex Therapeutics S.L, Badalona, Spain.,Germans Trias i Pujol Health Science Research Institute, Badalona, Spain.,ISGlobal, Hospital Clínic-Universitat de Barcelona, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - María Montoya
- Innovex Therapeutics S.L, Badalona, Spain.,Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Cientificas, Madrid, Spain
| | - Lorenzo Fraile
- Innovex Therapeutics S.L, Badalona, Spain.,Departamento de Ciencia Animal, Escuela Técnica Superior de Ingenieria Agraria (ETSEA), Universidad de Lleida, Lleida, Spain
| |
Collapse
|
40
|
Montaner-Tarbes S, Novell E, Tarancón V, Borrás FE, Montoya M, Fraile L, Del Portillo HA. Targeted-pig trial on safety and immunogenicity of serum-derived extracellular vesicles enriched fractions obtained from Porcine Respiratory and Reproductive virus infections. Sci Rep 2018; 8:17487. [PMID: 30504834 PMCID: PMC6269534 DOI: 10.1038/s41598-018-36141-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 11/01/2018] [Indexed: 11/09/2022] Open
Abstract
The Porcine Reproductive and Respiratory Syndrome Virus (PRRSV) is the etiological agent of one of the most important swine diseases with a significant economic burden worldwide. Unfortunately, available vaccines are partially effective highlighting the need of novel approaches. Previously, antigenic viral proteins were described in serum-derived extracellular vesicles (EVs) from pigs previously infected with PRRSV. Here, a targeted-pig trial was designed to determine the safety and immunogenicity of such extracellular vesicles enriched fractions. Our results showed that immunizations with EV-enriched fractions from convalescence animals in combination with montanide is safe and free of virus as immunizations with up-to two milligrams of EV-enriched fractions did not induce clinical symptoms, adverse effects and detectable viral replication. In addition, this vaccine formulation was able to elicit specific humoral IgG immune response in vaccinated animals, albeit variably. Noticeably, sera from vaccinated animals was diagnosed negative when tested for PRRSV using a commercial ELISA test; thus, indicating that this new approach differentiates vaccinated from infected animals. Lastly, after priming animals with EV-enriched fractions from sera of convalescence animals and boosting them with synthetic viral peptides identified by mass spectrometry, a distinctive high and specific IFN-γ response was elicited. Altogether, our data strongly suggest the use of serum EV-enriched fractions as a novel vaccine strategy against PRRSV.
Collapse
Affiliation(s)
- Sergio Montaner-Tarbes
- Innovex Therapeutics S.L, Badalona, Spain
- Departamento de Ciència Animal, ETSEA, Avenida Alcalde Rovira Roure, 191, Universidad de Lleida, Lleida, Spain
| | | | | | - Francesc E Borrás
- Innovex Therapeutics S.L, Badalona, Spain
- Germans Trias i Pujol Health Science Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Spain
| | | | - Lorenzo Fraile
- Innovex Therapeutics S.L, Badalona, Spain.
- Departamento de Ciència Animal, ETSEA, Avenida Alcalde Rovira Roure, 191, Universidad de Lleida, Lleida, Spain.
| | - Hernando A Del Portillo
- Innovex Therapeutics S.L, Badalona, Spain.
- Germans Trias i Pujol Health Science Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Spain.
- ISGlobal, Hospital Clínic - Universitat de Barcelona, C/ Roselló 153, 08036, Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Passeig Luis Companys 23, 08010, Barcelona, Spain.
| |
Collapse
|
41
|
Li S, Gong P, Tai L, Li X, Wang X, Zhao C, Zhang X, Yang Z, Yang J, Li J, Zhang X. Extracellular Vesicles Secreted by Neospora caninum Are Recognized by Toll-Like Receptor 2 and Modulate Host Cell Innate Immunity Through the MAPK Signaling Pathway. Front Immunol 2018; 9:1633. [PMID: 30087675 PMCID: PMC6066505 DOI: 10.3389/fimmu.2018.01633] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 07/02/2018] [Indexed: 12/23/2022] Open
Abstract
Neospora caninum is an obligate intracellular parasite, which causes significant economic losses in the cattle industry. However, the immune mechanism of the parasite–host interaction is not yet fully understood. Extracellular vesicles (EVs) have emerged as a ubiquitous mechanism by which almost all cells, especially immune and tumor cells, participate in intercellular communications. Although studies have indicated that EVs secreted by Toxoplasma gondii or Trypanosoma brucei promote exchanges of biological molecules important for the host–parasite interplay, however, EVs and their biological activities in N. caninum is not clear. Here, we used multiple methods, including electron microscopy, nanoparticle tracking analysis, RT-PCR, immunofluorescence, western blot, proteomics, and cytokine analyses, to examine the properties of N. caninum EVs. We found that N. caninum produced EVs that are similar to mammalian exosomes, which generally range from 30 to 150 nm in diameter. It was shown that N. caninum EVs could remarkably increase the production of pro-inflammatory cytokines IL-12p40, TNF-α, IL-1β, IL-6, and IFN-γ by wild-type (WT) mouse bone marrow-derived macrophages (BMDMs) whereas the secretion of IL-12p40, TNF-α, and IFN-γ was very strongly downregulated in TLR2−/− mouse BMDMs. The levels of IL-6 were not affected, but the secretion of IL-10 was upregulated. We found that the phosphorylation levels of P38, ERK, and JNK were significantly reduced in the TLR2−/− cells compared with those in WT mouse BMDMs and that treatment with chemical inhibiters of P38, ERK, and JNK resulted in upregulation of IL-6, IL-12p40, and IL-10 production. Together, these results demonstrated that N. caninum EVs could be rapidly internalized to deliver proteins to the host cells and modulate the host cell immune responses through MAPK signaling pathway in a TLR2-dependent manner. Our study is the first to reveal potential roles for N. caninum EVs in host communication and immune response in parasite–host interactions.
Collapse
Affiliation(s)
- Shan Li
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Pengtao Gong
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Lixin Tai
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xin Li
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiaocen Wang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Chunyan Zhao
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xu Zhang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zhengtao Yang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ju Yang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jianhua Li
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xichen Zhang
- College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
42
|
|
43
|
Zheng Y, Guo X, Su M, Guo A, Ding J, Yang J, Xiang H, Cao X, Zhang S, Ayaz M, Luo X. Regulatory effects of Echinococcus multilocularis extracellular vesicles on RAW264.7 macrophages. Vet Parasitol 2017; 235:29-36. [PMID: 28215864 DOI: 10.1016/j.vetpar.2017.01.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 01/11/2017] [Accepted: 01/13/2017] [Indexed: 12/21/2022]
Abstract
Extracellular vesicles (EVs) play a role in intercellular communications via exchanging biological molecules, being involved in host-parasite interplay. Little is to date known about E. multilocularis EVs and their biological activities. Here spherical EVs secreted by E. multilocularis metacestodes were shown to range predominately from 34nm to 95nm in diameter. A total of 433 proteins were identified in the EVs, and the proteins involved in binding (42%) and catalytic activity (41%) were most frequently represented. Moreover, the proteins associated with EV biogenesis and trafficking, including annexin, 14-3-3, tetraspanin and heat shock protein 70kDa, were highly enriched. It was shown that the EVs remarkably suppressed NO produced by activated RAW macrophages via downregulation of inducible nitric oxide synthase expression (p <0.01). Suppression of pro-inflammatory cytokines, especially IL-1α and IL-1β, was also observed post treatment with the EVs. Conversely, increased expression of the majority (10/11) of key components involved in the LPS/TLR4 pathway was induced by the EVs. These results demonstrate a regulatory effect of E. multilocularis EVs on macrophages, suggesting a role in parasite-host interactions.
Collapse
Affiliation(s)
- Yadong Zheng
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou 730046, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| | - Xiaola Guo
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou 730046, China
| | - Meng Su
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou 730046, China
| | - Aijiang Guo
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou 730046, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Juntao Ding
- College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Jing Yang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou 730046, China
| | - Haitao Xiang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Xiaoan Cao
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou 730046, China
| | - Shaohua Zhang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou 730046, China
| | - Mazhar Ayaz
- Department of Pathobiology, Faculty of Veterinary Sciences, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Xuenong Luo
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou 730046, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| |
Collapse
|
44
|
Song X, Zhao X, Xu L, Yan R, Li X. Immune protection duration and efficacy stability of DNA vaccine encoding Eimeria tenella TA4 and chicken IL-2 against coccidiosis. Res Vet Sci 2016; 111:31-35. [PMID: 27914219 DOI: 10.1016/j.rvsc.2016.11.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/01/2016] [Accepted: 11/18/2016] [Indexed: 12/12/2022]
Abstract
In our previous study, an effective DNA vaccine encoding Eimeria tenella TA4 and chicken IL-2 was constructed. In the present study, the immunization dose of the DNA vaccine pVAX1.0-TA4-IL-2 was further optimized. With the optimized dose, the dynamics of antibodies induced by the DNA vaccine was determined using indirect ELISA. To evaluate the immune protection duration of the DNA vaccine, two-week-old chickens were intramuscularly immunized twice and the induced efficacy was evaluated by challenging with E. tenella at 5, 9, 13, 17 and 21weeks post the last immunization (PLI) separately. To evaluate the efficacy stability of the DNA vaccine, two-week-old chickens were immunized with 3 batches of the DNA vaccine, and the induced efficacy was evaluated by challenging with E. tenella. The results showed that the optimal dose was 25μg. The induced antibody level persisted until 10weeks PPI. For the challenge time of 5 and 9weeks PLI, the immunization resulted in ACIs of 182.28 and 162.23 beyond 160, showing effective protection. However, for the challenge time of 13, 17 and 21weeks PLI, the immunization resulted in ACIs below 160 which means poor protection. Therefore, the immune protection duration of the DNA vaccination was at least 9weeks PLI. DNA immunization with three batches DNA vaccine resulted in ACIs of 187.52, 191.57 and 185.22, which demonstrated that efficacies of the three batches DNA vaccine were effective and stable. Overall, our results indicate that DNA vaccine pVAX1.0-TA4-IL-2 has the potential to be developed as effective vaccine against coccidiosis.
Collapse
Affiliation(s)
- Xiaokai Song
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Xiaofang Zhao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Lixin Xu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Ruofeng Yan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Xiangrui Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China.
| |
Collapse
|