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
|
Mustafa S, Abbas RZ, Saeed Z, Baazaoui N, Khan AMA. Use of Metallic Nanoparticles Against Eimeria-the Coccidiosis-Causing Agents: A Comprehensive Review. Biol Trace Elem Res 2024:10.1007/s12011-024-04399-8. [PMID: 39354182 DOI: 10.1007/s12011-024-04399-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/25/2024] [Indexed: 10/03/2024]
Abstract
Coccidiosis is a protozoan disease caused by Eimeria species and is a major threat to the poultry industry. Different anti-coccidial drugs (diclazuril, amprolium, halofuginone, ionophores, sulphaquinoxaline, clopidol, and ethopabate) and vaccines have been used for their control. Still, due to the development of resistance, their efficacy has been limited. It is continuously damaging the economy of the poultry industry because under its control, almost $14 billion is spent, globally. Recent research has been introducing better and more effective control of coccidiosis by using metallic and metallic oxide nanoparticles. Zinc, zinc oxide, copper, copper oxide, silver, iron, and iron oxide are commonly used because of their drug delivery mechanism. These nanoparticles combined with other drugs enhance the effect of these drugs and give their better results. Moreover, by using nanotechnology, the resistance issue is also solved because by using several mechanisms at a time, protozoa cannot evolve and thus resistance cannot develop. Green nanotechnology has been giving better results due to its less toxic effects. Utilization of metallic and metallic oxide nanoparticles may present a new, profitable, and economical method of controlling chicken coccidiosis, thus by changing established treatment approaches and improving the health and production of chickens. Thus, the objective of this review is to discuss about economic burden of avian coccidiosis, zinc, zinc oxide, iron, iron oxide, copper, copper oxide, silver nanoparticles use in the treatment of coccidiosis, their benefits, and toxicity.
Collapse
Affiliation(s)
- Sahar Mustafa
- Department of Clinical Medicine and Surgery, University of Agriculture, Faisalabad, 38040, Pakistan.
| | - Rao Zahid Abbas
- Department of Parasitology, University of Agriculture, Faisalabad, 38040, Pakistan
| | - Zohaib Saeed
- Department of Parasitology, University of Agriculture, Faisalabad, 38040, Pakistan
| | - Narjes Baazaoui
- Applied College Muhayil Assir, King Khalid University, 61421, Abha, Saudi Arabia
| | | |
Collapse
|
3
|
Feng L, Han N, Han YB, Shang MW, Liang TW, Liu ZH, Li SK, Zhai JX, Yin J. Structural analysis of a soluble polysaccharide GSPA-0.3 from the root of Panax ginseng C. A. Meyer and its adjuvant activity with mechanism investigation. Carbohydr Polym 2024; 326:121591. [PMID: 38142068 DOI: 10.1016/j.carbpol.2023.121591] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 12/25/2023]
Abstract
A novel polysaccharide (GSPA-0.3) was isolated and purified from the root of cultivated Panax ginseng C. A. Meyer, and its structure, adjuvant activities, and mechanisms for inducing the maturation of mouse dendritic 2.4 cells (DC2.4) were extensively studied. Fraction GSPA-0.3, mainly composed by the galacturonic acid, galactose, arabinose, glucose, rhamnose, mannose, and xylose, had a molecular weight of 62,722 Da. The main chain of GSPA-0.3 was composed of →3)-α-L-Rhap-(1→, →4)-α-D-GalpA-(1→, and →3, 4)-α-D-GalpA-(1→. Branched chains comprised α-L-Araf-(1→3, 5)-α-L-Araf-(1→5)-α-L-Araf-(1→, α-D-Glcp-(1→6)-α-D-Glcp-(1→6)-α-D-Glcp-(1→, β-D-Galp-(1→4)-β-D-Galp-(1→4)-β-D-Galp-(1→, and α-D-GalpA-(1→ units connected to the C3 position of →3, 4)-α-D-GalpA-(1→. In vivo, GSPA-0.3 was found to stimulate the production of IgG, IgG1, and IgG2a; increase the splenocyte proliferation index; and promote the expression of GATA-3, T-bet, IFN-γ, and IL-4 in H1N1 vaccine-immunized mice. Moreover, GSPA-0.3 significantly increased the levels of neutralizing antibodies in the mice, and its adjuvant activity was found to be superior to aluminum adjuvant (Alum adjuvant). Mechanistic investigations showed that GSPA-0.3 activated the TLR4-dependent pathway by upregulating the expressions of TLR4, MyD88, TRAF-6, and NF-κB proteins and gens. The results presented herein suggested that GSPA-0.3 could significantly promote the efficacy of the H1N1 vaccine by modulating Th1/Th2 response via the TLR4-MyD88-NF-κB signaling pathway.
Collapse
Affiliation(s)
- Lei Feng
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, China; Department of Pharmacognosy and Utilization Key Laboratory of Northeast Plant Materials, Shenyang 110016, China
| | - Na Han
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, China; Department of Pharmacognosy and Utilization Key Laboratory of Northeast Plant Materials, Shenyang 110016, China
| | - Yu-Bo Han
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, China; Department of Pharmacognosy and Utilization Key Laboratory of Northeast Plant Materials, Shenyang 110016, China
| | - Meng-Wen Shang
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, China; Department of Pharmacognosy and Utilization Key Laboratory of Northeast Plant Materials, Shenyang 110016, China
| | - Teng-Wei Liang
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, China; Department of Pharmacognosy and Utilization Key Laboratory of Northeast Plant Materials, Shenyang 110016, China
| | - Zhi-Hui Liu
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, China; Department of Pharmacognosy and Utilization Key Laboratory of Northeast Plant Materials, Shenyang 110016, China
| | - Si-Kai Li
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, China; Department of Pharmacognosy and Utilization Key Laboratory of Northeast Plant Materials, Shenyang 110016, China
| | - Jian-Xiu Zhai
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, China; Department of Pharmacognosy and Utilization Key Laboratory of Northeast Plant Materials, Shenyang 110016, China.
| | - Jun Yin
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, China; Department of Pharmacognosy and Utilization Key Laboratory of Northeast Plant Materials, Shenyang 110016, China.
| |
Collapse
|
4
|
Xiong Y, Lou P, Xu C, Han B, Liu J, Gao J. Emerging role of extracellular vesicles in veterinary practice: novel opportunities and potential challenges. Front Vet Sci 2024; 11:1335107. [PMID: 38332755 PMCID: PMC10850357 DOI: 10.3389/fvets.2024.1335107] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/12/2024] [Indexed: 02/10/2024] Open
Abstract
Extracellular vesicles are nanoscale vesicles that transport signals between cells, mediating both physiological and pathological processes. EVs facilitate conserved intercellular communication. By transferring bioactive molecules between cells, EVs coordinate systemic responses, regulating homeostasis, immunity, and disease progression. Given their biological importance and involvement in pathogenesis, EVs show promise as biomarkers for veterinary diagnosis, and candidates for vaccine production, and treatment agents. Additionally, different treatment or engineering methods could be used to boost the capability of extracellular vesicles. Despite the emerging veterinary interest, EV research has been predominantly human-based. Critical knowledge gaps remain regarding isolation protocols, cargo loading mechanisms, in vivo biodistribution, and species-specific functions. Standardized methods for veterinary EV characterization and validation are lacking. Regulatory uncertainties impede veterinary clinical translation. Advances in fundamental EV biology and technology are needed to propel the veterinary field forward. This review introduces EVs from a veterinary perspective by introducing the latest studies, highlighting their potential while analyzing challenges to motivate expanded veterinary investigation and translation.
Collapse
Affiliation(s)
- Yindi Xiong
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Peng Lou
- NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Chuang Xu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Bo Han
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jingping Liu
- NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jian Gao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
5
|
Tan F, Zhang L, Yin L, Wang L, Zhang H, Zheng L, Cui X, Lv X, Bai R, Zheng M. Immune synergistic mechanism of recombinant plasmid adjuvant containing chicken IL-4 and IL-2 fusion genes on chicken coccidia live vaccine. Poult Sci 2024; 103:103204. [PMID: 37939587 PMCID: PMC10665987 DOI: 10.1016/j.psj.2023.103204] [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: 08/03/2023] [Revised: 10/12/2023] [Accepted: 10/12/2023] [Indexed: 11/10/2023] Open
Abstract
The recombinant plasmid pCI-IL-4-IL-2-EGFP containing fusion genes of chicken IL-4 and IL-2 can be used as an adjuvant to enhance the anticoccidiosis effect of the chicken coccidia live vaccine. The chickens were divided into 3 groups: blank control group, vaccine + pCI-IL-4-IL-2-EGFP adjuvant coimmunization group, and vaccine-only group to investigate the immune synergy mechanism of recombinant plasmid adjuvant pCI-IL-4-IL-2-EGFP. The expressions of IL-2, IL-4, TNF-α, and IFN-γ in chicken sera and tissues were detected by ELISA and RT-qPCR, and the proliferation of T and B lymphocytes and antigen presenting cells (APC) in chicken immune organs and intestines were detected by acid alpha-naphthalase (ANAE) staining, methyl green pyronine (MGP) staining, and immunofluorescence (IF) staining, respectively. Results showed that the mRNA expression of IL-2, IL-4, IFN-γ and the number of activated T and B lymphocytes were significantly upregulated in the spleen and cecum tonsils of chickens in vaccine + pCI-IL-4-IL-2-EGFP group compared with the vaccine-only group on 7 d after vaccination (P < 0.05). Protein contents of IL-2, IL-4 and TNF-α in vaccine + pCI-IL-4-IL-2-EGFP group were significantly increased compared to vaccine-only group on 28 d of inoculation (P < 0.05). The number of T and B lymphocytes and APC in chickens of the vaccine+ pCI-IL-4-IL-2-EGFP group was significantly higher than that of the vaccine-only group in cecum tonsils, thymus and spleen after 14 and 28 d of inoculation (P < 0.05). All results revealed that pCI-IL-4-IL-2-EGFP adjuvant enhanced the immune response of chicken coccidia live vaccine by upregulating the expression of IL-2, IL-4, TNF-α, and IFN-γ and promoting the proliferation of T, B lymphocytes and APCs in chicken intestines and immune organ sites. Moreover, our study provides a theoretical basis for the clinical application of cytogenic plasmids as adjuvants.
Collapse
Affiliation(s)
- Fan Tan
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
| | - Li Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
| | - Liyang Yin
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
| | - Liming Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
| | - Honghui Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
| | - Longlong Zheng
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
| | - Xiaozhen Cui
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
| | - Xiaoling Lv
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
| | - Rui Bai
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China
| | - Mingxue Zheng
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong 030801, China.
| |
Collapse
|
6
|
Khosravi M, Mohammad Rahimi H, Nazari A, Baghaei K, Asadzadeh Aghdaei H, Shahrokh S, Sharifdini M, Torrecilhas AC, Mehryab F, Mirjalali H, Shekari F, Zali MR. Characterisation of extracellular vesicles isolated from hydatid cyst fluid and evaluation of immunomodulatory effects on human monocytes. J Cell Mol Med 2023; 27:2614-2625. [PMID: 37530547 PMCID: PMC10468670 DOI: 10.1111/jcmm.17894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/03/2023] Open
Abstract
Hydatidosis is a disease caused by the larval stage of Echinococcus granulosus, which involves several organs of intermediate hosts. Evidence suggests a communication between hydatid cyst (HC) and hosts via extracellular vesicles. However, a little is known about the communication between EVs derived from HC fluid (HCF) and host cells. In the current study, EVs were isolated using differential centrifugation from sheep HCF and characterized by western blot, electron microscope and size distribution analysis. The uptake of EVs by human monocyte cell line (THP-1) was evaluated. The effects of EVs on the expression levels of pro- and anti-inflammatory cytokines were investigated using quantitative real-time PCR (RT-PCR), 3 and 24 h after incubation. Moreover, the cytokine level of IL-10 was evaluated in supernatant of THP-1 cell line at 3 and 24 h. EVs were successfully isolated and showed spherical shape with size distribution at 130.6 nm. After 3 h, the expression levels of pro-inflammatory cytokine genes (IL1Β, IL15 and IL8) were upregulated, while after 24 h, the expression levels of pro-inflammatory cytokines were decreased and IL13 gene expression showed upregulation. A statistically significant increase was seen in the levels of IL-10 after 24 h. The main mechanism of the communication between EVs derived from HCF and their host remains unclear; however, time-dependent anti-inflammatory effects in our study suggest that HC may modulate the immune responses via EVs.
Collapse
Affiliation(s)
- Mojdeh Khosravi
- Department of Pharmacy and Pharmaceutical Technology and ParasitologyUniversity of ValenciaValenciaSpain
| | - Hanieh Mohammad Rahimi
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Abdoreza Nazari
- Department of Molecular Systems Biology at Cell Science Research CenterRoyan Institute for Stem Cell Biology and TechnologyTehranIran
| | - Kaveh Baghaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Shabnam Shahrokh
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Meysam Sharifdini
- Department of Medical Parasitology and Mycology, School of MedicineGuilan University of Medical SciencesRashtIran
| | - Ana Claudia Torrecilhas
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências FarmacêuticasUniversidade Federal de São Paulo (UNIFESP)DiademaBrazil
| | - Fatemeh Mehryab
- Department of Molecular Systems Biology at Cell Science Research CenterRoyan Institute for Stem Cell Biology and TechnologyTehranIran
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of PharmacyShahid Beheshti University of Medical SciencesTehranIran
| | - Hamed Mirjalali
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Faezeh Shekari
- Department of Molecular Systems Biology at Cell Science Research CenterRoyan Institute for Stem Cell Biology and TechnologyTehranIran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
7
|
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
|
8
|
Pordanjani PM, Bolhassani A, Milani A, Pouriayevali MH. Extracellular vesicles in vaccine development and therapeutic approaches for viral diseases. Process Biochem 2023. [DOI: 10.1016/j.procbio.2023.02.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
|
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: 0] [Impact Index Per Article: 0] [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
|
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: 26] [Impact Index Per Article: 8.7] [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
|
11
|
Amrollahi P, Zheng W, Monk C, Li CZ, Hu TY. Nanoplasmonic Sensor Approaches for Sensitive Detection of Disease-Associated Exosomes. ACS APPLIED BIO MATERIALS 2021; 4:6589-6603. [PMID: 35006963 PMCID: PMC9130051 DOI: 10.1021/acsabm.1c00113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Exosomes are abundantly secreted by most cells that carry membrane and cytosolic factors that can reflect the physiologic state of their source cells and thus have strong potential to serve as biomarkers for early diagnosis, disease staging, and treatment monitoring. However, traditional diagnostic or prognostic applications that might use exosomes are hindered by the lack of rapid and sensitive assays that can exploit their biological information. An array of assay approaches have been developed to address this deficit, including those that integrate immunoassays with nanoplasmonic sensors to measure changes in optical refractive indexes in response to the binding of low concentrations of their targeted molecules. These sensors take advantage of enhanced and tunable interactions between the electron clouds of nanoplasmonic particles and structures and incident electromagnetic radiation to enable isolation-free and ultrasensitive quantification of disease-associated exosome biomarkers present in complex biological samples. These unique advantages make nanoplasmonic sensing one of the most competitive approaches available for clinical applications and point-of-care tests that evaluate exosome-based biomarkers. This review will briefly summarize the origin and clinical utility of exosomes and the limitations of current isolation and analysis approaches before reviewing the specific advantages and limitations of nanoplasmonic sensing devices and indicating what additional developments are necessary to allow the translation of these approaches into clinical applications.
Collapse
Affiliation(s)
- Pouya Amrollahi
- Center of Cellular and Molecular Diagnosis, Tulane University, New Orleans, Louisiana 70118, United States
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85281, United States
| | - Wenshu Zheng
- Center of Cellular and Molecular Diagnosis, Tulane University, New Orleans, Louisiana 70118, United States
| | - Chandler Monk
- Center of Cellular and Molecular Diagnosis, Tulane University, New Orleans, Louisiana 70118, United States
| | - Chen-Zhong Li
- Center of Cellular and Molecular Diagnosis, Tulane University, New Orleans, Louisiana 70118, United States
| | - Tony Ye Hu
- Center of Cellular and Molecular Diagnosis, Tulane University, New Orleans, Louisiana 70118, United States
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Juárez-Estrada MA, Gayosso-Vázquez A, Tellez-Isaias G, Alonso-Morales RA. Protective Immunity Induced by an Eimeria tenella Whole Sporozoite Vaccine Elicits Specific B-Cell Antigens. ANIMALS : AN OPEN ACCESS JOURNAL FROM MDPI 2021; 11:ani11051344. [PMID: 34065041 PMCID: PMC8151427 DOI: 10.3390/ani11051344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 12/13/2022]
Abstract
Simple Summary Coccidiosis caused by Eimeria tenella is a dreadful disease with a significant economic impact to the poultry industry. The disease has been controlled by routine medication of feed with synthetic chemicals or ionophore drugs. However, the rising appearance of drug resistance and public demands for reduced drug use in poultry production have driven a dramatic change, replacing anticoccidial drugs with alternative methods, such as vaccination with either virulent or attenuated Eimeria oocysts. Based on preliminary studies, the immune protection evaluating whole-sporozoites of E. tenella vaccine was verified. After this vaccine provided successful protection, the humoral response of a heterologous species like the rabbit was compared with the natural host immune response. Several B-cells antigens from the E. tenella sporozoite suitable for a genetically engineered vaccine were identified. Vaccination with newly identified recombinant antigens offers a feasible alternative for the control of avian coccidiosis into the broiler barns favoring the gradual withdrawal of the anticoccidial drugs. Abstract This study investigated protection against Eimeria tenella following the vaccination of chicks with 5.3 × 106E. tenella whole-sporozoites emulsified in the nanoparticle adjuvant IMS 1313 N VG Montanide™ (EtSz-IMS1313). One-day-old specific pathogen-free (SPF) chicks were subcutaneously injected in the neck with EtSz-IMS1313 on the 1st and 10th days of age. Acquired immunity was assayed through a challenge with 3 × 104 homologous sporulated oocysts at 21 days of age. The anticoccidial index (ACI) calculated for every group showed the effectiveness of EtSz-IMS1313 as a vaccine with an ACI of 186; the mock-injected control showed an ACI of 18 and the unimmunized, challenged control showed an ACI of −28. In a comparison assay, antibodies from rabbits and SPF birds immunized with EtSz-IMS1313 recognized almost the same polypeptides in the blotting of E. tenella sporozoites and merozoites. However, rabbit antisera showed the clearest recognition pattern. Polypeptides of 120, 105, 94, 70, 38, and 19 kDa from both E. tenella life cycle stages were the most strongly recognized by both animal species. The E. tenella zoite-specific IgG antibodies from the rabbits demonstrated the feasibility for successful B cell antigen identification.
Collapse
Affiliation(s)
- Marco A. Juárez-Estrada
- Departamento de Medicina y Zootecnia de Aves, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Coyoacán, Cd. De México 04510, Mexico
- Departamento de Genética y Bioestadística, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Coyoacán, Cd. De México 04510, Mexico; (A.G.-V.); (R.A.A.-M.)
- Correspondence:
| | - Amanda Gayosso-Vázquez
- Departamento de Genética y Bioestadística, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Coyoacán, Cd. De México 04510, Mexico; (A.G.-V.); (R.A.A.-M.)
| | | | - Rogelio A. Alonso-Morales
- Departamento de Genética y Bioestadística, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Coyoacán, Cd. De México 04510, Mexico; (A.G.-V.); (R.A.A.-M.)
| |
Collapse
|
15
|
Li S, Zhang N, Liu S, Li J, Liu L, Wang X, Li X, Gong P, Zhang X. Protective Immunity Against Neospora caninum Infection Induced by 14-3-3 Protein in Mice. Front Vet Sci 2021; 8:638173. [PMID: 33748214 PMCID: PMC7965954 DOI: 10.3389/fvets.2021.638173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 02/03/2021] [Indexed: 11/16/2022] Open
Abstract
Neospora caninum is an apicomplexan parasite that infects many mammals and remains a threatening disease worldwide because of the lack of effective drugs and vaccines. Our previous studies demonstrated that N. caninum 14-3-3 protein (Nc14-3-3), which is included in N. caninum extracellular vesicles (NEVs), can induce effective immune responses and stimulate cytokine expression in mouse peritoneal macrophages. However, whether Nc14-3-3 has a protective effect and its mechanisms are poorly understood. Here, we evaluated the immune responses and protective effects of Nc14-3-3 against exposure to 2 × 107 Nc-1 tachyzoites. Antibody (IgG, IgGl, and IgG2a) levels and Th1-type (IFN-γ and IL-12) and Th2-type (IL-4 and IL-10) cytokines in mouse serum, survival rates, survival times, and parasite burdens were detected. In the present study, the immunostimulatory effect of Nc14-3-3 was confirmed, as it triggered Th1-type cytokine (IFN-γ and IL-12) production in mouse serum 2 weeks after the final immunization. Moreover, the immunization of C57BL/6 mice with Nc14-3-3 induced high IgG antibody levels and significant increases in CD8+ T lymphocytes in the spleens of mice, indicating that the cellular immune response was significantly stimulated. Mouse survival rates and times were significantly prolonged after immunization; the survival rates were 40% for Nc14-3-3 immunization and 60% for NEV immunization, while mice that received GST, PBS, or blank control all died at 13, 9, or 8 days, respectively, after intraperitoneal N. caninum challenge. In addition, qPCR analysis indicated that there was a reduced parasite burden and diminished pathological changes in the mice immunized with Nc14-3-3. Our data demonstrate that vaccination of mice with Nc14-3-3 elicits both cellular and humoral immune responses and provides partial protection against acute neosporosis. Thus, Nc14-3-3 could be an effective antigen candidate for vaccine development for neosporosis.
Collapse
Affiliation(s)
- Shan Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China.,Department of Social Medicine and Public Health, School of Basic Medicine, Jiujiang University, Jiujiang, China
| | - Nan Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Shaoxiong Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jianhua Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Li Liu
- College of Basic Medicine, Jilin University, Changchun, China
| | - Xiaocen Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xin Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Pengtao Gong
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xichen Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
16
|
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
|
17
|
Haseeb M, Lakho SA, Huang J, Hasan MW, Ali-Ul-Husnain Naqvi M, Zhou Z, Yan R, Xu L, Song X, Li X. In vitro effects of 5 recombinant antigens of Eimeria maxima on maturation, differentiation, and immunogenic functions of dendritic cells derived from chicken spleen. Poult Sci 2020; 99:5331-5343. [PMID: 33142449 PMCID: PMC7647736 DOI: 10.1016/j.psj.2020.07.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 06/05/2020] [Accepted: 07/22/2020] [Indexed: 12/27/2022] Open
Abstract
Eimeria maxima possesses integral families of immunogenic constituents that promote differentiation of immune cells during host-parasite interactions. Dendritic cells (DCs) have an irreplaceable role in the modulation of the host immunity. However, the selection of superlative antigen with immune stimulatory efficacies on host DCs is lacking. In this study, 5 recombinant proteins of E. maxima (Em), including Em14-3-3, rhomboid family domain containing proteins (ROM) EmROM1 and EmROM2, microneme protein 2 (EmMIC2), and Em8 were identified to stimulate chicken splenic derived DCs in vitro. The cultured populations were incubated with recombinant proteins, and typical morphologies of stimulated DCs were obtained. DC-associated markers major histocompatibility complex class II, CD86, CD11c, and CD1.1, showed upregulatory expressions by flow cytometry assay. Immunofluorescence assay revealed that recombinant proteins could bind with the surface of chicken splenic derived DCs. Moreover, quantitative real-time PCR results showed that distinct gene expressions of Toll-like receptors and Wnt signaling pathway were upregulated after the coincubation of recombinant proteins with DCs. The ELISA results indicated that the DCs produced a significant higher level of interleukin (IL)-12 and interferon-γ secretions after incubation with recombinant proteins. While transforming growth factor-β was significantly increased with rEmROM1, rEmROM2, and rEmMIC2 as compared to control groups, and IL-10 did not show significant alteration. Taken together, these results concluded that among 5 potential recombinant antigens, rEm14-3-3 could promote immunogenic functions of chicken splenic derived DCs more efficiently, which might represent an effective molecule for inducing the host Th1-mediated immune response against Eimeria infection.
Collapse
Affiliation(s)
- Muhammad Haseeb
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Shakeel Ahmed Lakho
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Jianmei Huang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Muhammad Waqqas Hasan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Muhammad Ali-Ul-Husnain Naqvi
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Zhouyang Zhou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Ruofeng Yan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Lixin Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Xiaokai Song
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Xiangrui Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, PR China.
| |
Collapse
|
18
|
Huyan T, Li H, Peng H, Chen J, Yang R, Zhang W, Li Q. Extracellular Vesicles - Advanced Nanocarriers in Cancer Therapy: Progress and Achievements. Int J Nanomedicine 2020; 15:6485-6502. [PMID: 32922012 PMCID: PMC7457829 DOI: 10.2147/ijn.s238099] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 07/08/2020] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are a class of cell-derived, lipid bilayer membrane composed vesicles, and some of them such as exosomes and ectosomes have been proven, playing remarkable roles in transmitting intercellular information, and being involved in each property of cell physiological activities. Nowadays, EVs are considered as potential nanocarriers which could partially resolve the problems of current chemotherapy because of their distinctive advantages. As endogenous membrane encompassed vesicles with nanosize, EVs are able to pass through the natural barriers with prolonged circulation time in vivo and have intrinsic cell targeting properties, they are less toxic, and less immunogenic. Recently, studies focusing on EV-based drug delivery system for cancer therapy have exploded dramatically. This review aims to outline the current applications of EVs as potential nanosized drug carriers in cancer therapy. Firstly, the characteristics and biofunctions of each EV subtype are described. Then the variety of therapeutic cargoes, the loading methods, and the targeting strategy of engineered EVs are emphatically introduced. Thereafter the pros and cons of EVs applied as therapeutic carriers, as well as the future prospects in this field, are discussed.
Collapse
Affiliation(s)
- Ting Huyan
- Key Laboratory for Space Biosciences and Biotechnology, Institute of Special Environment Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, People's Republic of China.,Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, Xi'an 710072, People's Republic of China
| | - Hongduo Li
- Xi'an Institute for Food and Drug Control, Xi'an 710054, People's Republic of China
| | - Hourong Peng
- Key Laboratory for Space Biosciences and Biotechnology, Institute of Special Environment Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, People's Republic of China
| | - Jinzhao Chen
- Shanxi Weiqidaguangming Pharmaceutical Co., Ltd, Datong, Shanxi Province 037301, People's Republic of China
| | - Ruixin Yang
- Xi'an Institute for Food and Drug Control, Xi'an 710054, People's Republic of China
| | - Wei Zhang
- Department of Anesthesiology, Henan Provincial People's Hospital (People's Hospital of Zhengzhou University), Zhengzhou 450003, People's Republic of China
| | - Qi Li
- Key Laboratory for Space Biosciences and Biotechnology, Institute of Special Environment Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, People's Republic of China
| |
Collapse
|
19
|
de Souza W, Barrias ES. Membrane-bound extracellular vesicles secreted by parasitic protozoa: cellular structures involved in the communication between cells. Parasitol Res 2020; 119:2005-2023. [PMID: 32394001 DOI: 10.1007/s00436-020-06691-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/15/2020] [Indexed: 02/06/2023]
Abstract
The focus of this review is a group of structures/organelles collectively known as extracellular vesicles (EVs) that are secreted by most, if not all, cells, varying from mammalian cells to protozoa and even bacteria. They vary in size: some are small (100-200 nm) and others are larger (> 200 nm). In protozoa, however, most of them are small or medium in size (200-400 nm). These include vesicles from different origins. We briefly review the biogenesis of this distinct group that includes (a) exosome, which originates from the multivesicular bodies, an important component of the endocytic pathway; (b) ectosome, formed from a budding process that takes place in the plasma membrane of the cells; (c) vesicles released from the cell surface following a process of patching and capping of ligand/receptor complexes; (d) other processes where tubules secreted by the parasite subsequently originate exosome-like structures. Here, special emphasis is given to EVs secreted by parasitic protozoa such as Leishmania, Trypanosoma, Plasmodium, Toxoplasma, Cryptosporidium, Trichomonas, and Giardia. Most of them have been characterized as exosomes that were isolated using several approaches and characterized by electron microscopy, proteomic analysis, and RNA sequencing. The results obtained show clearly that they present several proteins and different types of RNAs. From the functional point of view, it is now clear that the secreted exosomes can be incorporated by the parasite itself as well as by mammalian cells with which they interact. As a consequence, there is interference both with the parasite (induction of differentiation, changes in infectivity, etc.) and with the host cell. Therefore, the EVs constitute a new system of transference of signals among cells. On the other hand, there are suggestions that exosomes may constitute potential biotechnology tools and are important players of what has been designated as nanobiotechnology. They may constitute an important delivery system for gene therapy and molecular-displaying cell regulation capabilities when incorporated into other cells and even by interfering with the exosomal membrane during its biogenesis, targeting the vesicles via specific ligands to different cell types. These vesicles may reach the bloodstream, overflow through intercellular junctions, and even pass through the central nervous system blood barrier. There is evidence that it is possible to interfere with the composition of the exosomes by interfering with multivesicular body biogenesis.
Collapse
Affiliation(s)
- Wanderley de Souza
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Centro de Ciências da Saúde, Bloco G, Ilha do Fundão, Rio de Janeiro, RJ, 21941-900, Brazil. .,Instituto Nacional de Ciência e Tecnologia and Núcleo de Biologia Estrutural e Bioimagens, CENABIO, Rio de Janeiro, Brazil.
| | - Emile S Barrias
- Instituto Nacional de Ciência e Tecnologia and Núcleo de Biologia Estrutural e Bioimagens, CENABIO, Rio de Janeiro, Brazil.,Laboratorio de Metrologia Aplicada à Ciências da Vida, Diretoria de Metrologia Aplicada à Ciências da Vida - Instituto Nacional de Metrologia, Qualidade e Tecnologia (Inmetro), Rio de Janeiro, Brazil
| |
Collapse
|
20
|
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
|
21
|
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
|
22
|
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
|
23
|
Retana Moreira L, Vargas Ramírez D, Linares F, Prescilla Ledezma A, Vaglio Garro A, Osuna A, Lorenzo Morales J, Abrahams Sandí E. Isolation of Acanthamoeba T5 from Water: Characterization of Its Pathogenic Potential, Including the Production of Extracellular Vesicles. Pathogens 2020; 9:pathogens9020144. [PMID: 32098034 PMCID: PMC7168589 DOI: 10.3390/pathogens9020144] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/12/2020] [Accepted: 02/19/2020] [Indexed: 12/12/2022] Open
Abstract
Acanthamoeba is a genus of free-living amoebae widely distributed in nature, associated with the development of encephalitis and keratitis. Despite the fact that it is common to find genotype T5 in environmental samples, only a few cases have been associated with clinical cases in humans. The wide distribution of Acanthamoeba, the characteristic of being amphizoic and the severity of the disease motivate researchers to focus on the isolation of these organisms, but also in demonstrating direct and indirect factors that could indicate a possible pathogenic potential. Here, we performed the characterization of the pathogenic potential of an Acanthamoeba T5 isolate collected from a water source in a hospital. Osmo- and thermotolerance, the secretion of proteases and the effect of trophozoites over cell monolayers were analyzed by different methodologies. Additionally, we confirm the secretion of extracellular vesicles (EVs) of this isolate incubated at two different temperatures, and the presence of serine and cysteine proteases in these vesicles. Finally, using atomic force microscopy, we determined some nanomechanical properties of the secreted vesicles and found a higher value of adhesion in the EVs obtained at 37 °C, which could have implications in the parasite´s survival and damaging potential in two different biological environments.
Collapse
Affiliation(s)
- Lissette Retana Moreira
- Departamento de Parasitología, Universidad de Costa Rica, San Pedro, Montes de Oca 2060, Costa Rica;
- Centro de Investigación en Enfermedades Tropicales (CIET), Universidad de Costa Rica, San Pedro, Montes de Oca 2060, Costa Rica;
- Correspondence: (L.R.M.); (E.A.S.)
| | - Daniel Vargas Ramírez
- Departamento de Parasitología, Universidad de Costa Rica, San Pedro, Montes de Oca 2060, Costa Rica;
- Centro de Investigación en Enfermedades Tropicales (CIET), Universidad de Costa Rica, San Pedro, Montes de Oca 2060, Costa Rica;
| | - Fátima Linares
- Centro de Instrumentación Científica (CIC), Universidad de Granada, Granada 18071, Spain;
| | - Alexa Prescilla Ledezma
- Departamento de Parasitología, Grupo de Bioquímica y Parasitología Molecular (CTS 183), Instituto de Biotecnología, Campus de Fuentenueva, Universidad de Granada, Granada 18071, Spain; (A.P.L.); (A.O.)
| | - Annette Vaglio Garro
- Centro de Investigación en Enfermedades Tropicales (CIET), Universidad de Costa Rica, San Pedro, Montes de Oca 2060, Costa Rica;
| | - Antonio Osuna
- Departamento de Parasitología, Grupo de Bioquímica y Parasitología Molecular (CTS 183), Instituto de Biotecnología, Campus de Fuentenueva, Universidad de Granada, Granada 18071, Spain; (A.P.L.); (A.O.)
| | - Jacob Lorenzo Morales
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, La Laguna, Tenerife, Islas Canarias 38203, Spain;
- Departamento de Obstetricia, Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, Avda. Astrofísico Fco. Sánchez, S/N, La Laguna, Tenerife, Islas Canarias 38203, Spain
| | - Elizabeth Abrahams Sandí
- Departamento de Parasitología, Universidad de Costa Rica, San Pedro, Montes de Oca 2060, Costa Rica;
- Centro de Investigación en Enfermedades Tropicales (CIET), Universidad de Costa Rica, San Pedro, Montes de Oca 2060, Costa Rica;
- Correspondence: (L.R.M.); (E.A.S.)
| |
Collapse
|
24
|
|
25
|
Nawaz M, Malik MI, Hameed M, Zhou J. Research progress on the composition and function of parasite-derived exosomes. Acta Trop 2019; 196:30-36. [PMID: 31071298 DOI: 10.1016/j.actatropica.2019.05.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/04/2019] [Accepted: 05/04/2019] [Indexed: 12/20/2022]
Abstract
Parasites use excretory-secretory pathways to communicate with the host. Characterization of exosomes within the excretory-secretory products reveal by which parasites manipulate their hosts. Parasite derived exosomes provide a mechanistic framework for protein and miRNAs transfer. Transcriptomics and proteomics of parasite exosomes identified a large number of miRNAs and proteins being utilized by parasites in their survival, reproduction and development. Characterization of proteins and miRNAs in parasite secreted exosomes provide important information on host-parasite communication and forms the basis for future studies. In this review, we summarize recent advances in isolation and molecular characterization (protein and miRNAs) of parasite derived exosomes.
Collapse
Affiliation(s)
- Mohsin Nawaz
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
| | - Muhammad Irfan Malik
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
| | - Muddassar Hameed
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
| | - Jinlin Zhou
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China.
| |
Collapse
|
26
|
Gurunathan S, Kang MH, Jeyaraj M, Qasim M, Kim JH. Review of the Isolation, Characterization, Biological Function, and Multifarious Therapeutic Approaches of Exosomes. Cells 2019; 8:307. [PMID: 30987213 PMCID: PMC6523673 DOI: 10.3390/cells8040307] [Citation(s) in RCA: 767] [Impact Index Per Article: 127.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/22/2019] [Accepted: 04/01/2019] [Indexed: 12/18/2022] Open
Abstract
Exosomes are extracellular vesicles that contain a specific composition of proteins, lipids, RNA, and DNA. They are derived from endocytic membranes and can transfer signals to recipient cells, thus mediating a novel mechanism of cell-to-cell communication. They are also thought to be involved in cellular waste disposal. Exosomes play significant roles in various biological functions, including the transfer of biomolecules such as RNA, proteins, enzymes, and lipids and the regulation of numerous physiological and pathological processes in various diseases. Because of these properties, they are considered to be promising biomarkers for the diagnosis and prognosis of various diseases and may contribute to the development of minimally invasive diagnostics and next generation therapies. The biocompatible nature of exosomes could enhance the stability and efficacy of imaging probes and therapeutics. Due to their potential use in clinical applications, exosomes have attracted much research attention on their roles in health and disease. To explore the use of exosomes in the biomedical arena, it is essential that the basic molecular mechanisms behind the transport and function of these vesicles are well-understood. Herein, we discuss the history, biogenesis, release, isolation, characterization, and biological functions of exosomes, as well as the factors influencing their biogenesis and their technical and biological challenges. We conclude this review with a discussion on the future perspectives of exosomes.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, 1 Hwayang-Dong, Gwangin-gu, Seoul 05029, Korea.
| | - Min-Hee Kang
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, 1 Hwayang-Dong, Gwangin-gu, Seoul 05029, Korea.
| | - Muniyandi Jeyaraj
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, 1 Hwayang-Dong, Gwangin-gu, Seoul 05029, Korea.
| | - Muhammad Qasim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, 1 Hwayang-Dong, Gwangin-gu, Seoul 05029, Korea.
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, 1 Hwayang-Dong, Gwangin-gu, Seoul 05029, Korea.
| |
Collapse
|
27
|
Transgenic Eimeria tenella Expressing Profilin of Eimeria maxima Elicits Enhanced Protective Immunity and Alters Gut Microbiome of Chickens. Infect Immun 2018; 86:IAI.00888-17. [PMID: 29967093 DOI: 10.1128/iai.00888-17] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 06/22/2018] [Indexed: 01/12/2023] Open
Abstract
Coccidiosis is one of the most serious diseases of livestock and birds in the world. Vaccination with live-parasite anticoccidial vaccines with genetic manipulation improving the immunogenicity of vaccine strains would be the best means for controlling coccidiosis in breeder and layer stocks, even in fast-growing broilers. Profilin from apicomplexan parasites is the first molecularly defined ligand for Toll-like receptor 11 (TLR11) and TLR12 in mice and is a potential molecular adjuvant. Here, we constructed a transgenic Eimeria tenella line (Et-EmPro) expressing the profilin of Eimeria maxima, the most immunogenic species of chicken coccidia, and evaluated the adjuvant effects of EmPro on the immunogenicity of E. tenella We found that immunization with the transgenic Eimeria parasites, compared with the wild type, elicited greater parasite antigen-specific cell-mediated immunity, characterized by increased numbers of interferon gamma (IFN-γ)-secreting lymphocytes. The transgenic parasite also induced better protective immunity against E. tenella challenge than the wild type. In addition, the diversity of the fecal microbiome of the birds immunized with the transgenic parasite differed from that of the microbiome of the wild-type-immunized birds, indicating interactions of Eimeria with the gut microbiome of chickens. Our results showing enhanced immunogenicity of E. tenella by use of EmPro as a molecular adjuvant derived from the most immunogenic affinis species represent a large step forward in the development of the next generation of coccidiosis vaccines using Eimeria as a vaccine platform expressing molecular adjuvants and potentially other pathogen antigens against not only coccidiosis but also other infectious diseases.
Collapse
|
28
|
Lovo-Martins MI, Malvezi AD, Zanluqui NG, Lucchetti BFC, Tatakihara VLH, Mörking PA, de Oliveira AG, Goldenberg S, Wowk PF, Pinge-Filho P. Extracellular Vesicles Shed By Trypanosoma cruzi Potentiate Infection and Elicit Lipid Body Formation and PGE 2 Production in Murine Macrophages. Front Immunol 2018; 9:896. [PMID: 29755471 PMCID: PMC5934475 DOI: 10.3389/fimmu.2018.00896] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/11/2018] [Indexed: 12/16/2022] Open
Abstract
During the onset of Trypanosoma cruzi infection, an effective immune response is necessary to control parasite replication and ensure host survival. Macrophages have a central role in innate immunity, acting as an important trypanocidal cell and triggering the adaptive immune response through antigen presentation and cytokine production. However, T. cruzi displays immune evasion mechanisms that allow infection and replication in macrophages, favoring its chronic persistence. One potential mechanism is the release of T. cruzi strain Y extracellular vesicle (EV Y), which participate in intracellular communication by carrying functional molecules that signal host cells and can modulate the immune response. The present work aimed to evaluate immune modulation by EV Y in C57BL/6 mice, a prototype resistant to infection by T. cruzi strain Y, and the effects of direct EV Y stimulation of macrophages in vitro. EV Y inoculation in mice prior to T. cruzi infection resulted in increased parasitemia, elevated cardiac parasitism, decreased plasma nitric oxide (NO), reduced NO production by spleen cells, and modulation of cytokine production, with a reduction in TNF-α in plasma and decreased production of TNF-α and IL-6 by spleen cells from infected animals. In vitro assays using bone marrow-derived macrophages showed that stimulation with EV Y prior to infection by T. cruzi increased the parasite internalization rate and release of infective trypomastigotes by these cells. In this same scenario, EV Y induced lipid body formation and prostaglandin E2 (PGE2) production by macrophages even in the absence of T. cruzi. In infected macrophages, EV Y decreased production of PGE2 and cytokines TNF-α and IL-6 24 h after infection. These results suggest that EV Y modulates the host response in favor of the parasite and indicates a role for lipid bodies and PGE2 in immune modulation exerted by EVs.
Collapse
Affiliation(s)
- Maria Isabel Lovo-Martins
- Instituto Carlos Chagas, Fiocruz - Paraná, Curitiba, Brazil.,Laboratório de Imunopatologia Experimental, Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Aparecida Donizette Malvezi
- Laboratório de Imunopatologia Experimental, Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | | | - Bruno Fernando Cruz Lucchetti
- Laboratório de Imunopatologia Experimental, Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Vera Lúcia Hideko Tatakihara
- Laboratório de Imunopatologia Experimental, Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | | | - Admilton Gonçalves de Oliveira
- Laboratório de Microscopia Eletrônica e Microanálises, Central de Laboratórios de Pesquisa Multiusuários, Universidade Estadual de Londrina, Londrina, Brazil
| | | | - Pryscilla Fanini Wowk
- Instituto Carlos Chagas, Fiocruz - Paraná, Curitiba, Brazil.,Laboratório de Virologia Molecular, Instituto Carlos Chagas, Fiocruz - Paraná, Curitiba, Brazil
| | - Phileno Pinge-Filho
- Laboratório de Imunopatologia Experimental, Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| |
Collapse
|
29
|
Lindenbergh MFS, Stoorvogel W. Antigen Presentation by Extracellular Vesicles from Professional Antigen-Presenting Cells. Annu Rev Immunol 2018; 36:435-459. [PMID: 29400984 DOI: 10.1146/annurev-immunol-041015-055700] [Citation(s) in RCA: 269] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The initiation and maintenance of adaptive immunity require multifaceted modes of communication between different types of immune cells, including direct intercellular contact, secreted soluble signaling molecules, and extracellular vesicles (EVs). EVs can be formed as microvesicles directly pinched off from the plasma membrane or as exosomes secreted by multivesicular endosomes. Membrane receptors guide EVs to specific target cells, allowing directional transfer of specific and complex signaling cues. EVs are released by most, if not all, immune cells. Depending on the type and status of their originating cell, EVs may facilitate the initiation, expansion, maintenance, or silencing of adaptive immune responses. This review focusses on EVs from professional antigen-presenting cells, their demonstrated and speculated roles, and their potential for cancer immunotherapy.
Collapse
Affiliation(s)
- Marthe F S Lindenbergh
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, NL-3508 TD Utrecht, The Netherlands;
| | - Willem Stoorvogel
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, NL-3508 TD Utrecht, The Netherlands;
| |
Collapse
|
30
|
Wahlund CJE, Güclüler G, Hiltbrunner S, Veerman RE, Näslund TI, Gabrielsson S. Exosomes from antigen-pulsed dendritic cells induce stronger antigen-specific immune responses than microvesicles in vivo. Sci Rep 2017; 7:17095. [PMID: 29213052 PMCID: PMC5719080 DOI: 10.1038/s41598-017-16609-6] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 11/15/2017] [Indexed: 12/18/2022] Open
Abstract
Extracellular vesicles (EV), including exosomes and microvesicles (MV), represent a rapidly expanding field of research with diagnostic and therapeutic applications. Although many aspects of EV function remain to be revealed and broad investigations are warranted, most published findings focus on only one vesicle category or a non-separated mix of EVs. In this paper, we investigated both MVs and exosomes from Ovalbumin (OVA)-pulsed dendritic cells for their immunostimulatory potential side-by-side in vivo. Only exosomes induced antigen-specific CD8+ T-cells, and were more efficient than MVs in eliciting antigen-specific IgG production. Further, mainly exosome-primed mouse splenocytes showed significant ex vivo interferon gamma production in response to antigen restimulation. Exosomes carried high levels of OVA, while OVA in MVs was barely detectable, which could explain the more potent antigen-specific response induced by exosomes. Moreover, exosomes induced increased germinal center B cell proportions, whereas MVs had no such effect. Immunisation with both vesicle types combined showed neither inhibitory nor synergistic effects. We conclude that DC-derived MVs and exosomes differ in their capacity to incorporate antigen and induce immune responses. The results are of importance for understanding the role of EVs in vivo, and for future design of vesicle-based immunotherapies and vaccines.
Collapse
Affiliation(s)
- Casper J E Wahlund
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet KS L2:04, SE-17176, Stockholm, Sweden
| | - Gözde Güclüler
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet KS L2:04, SE-17176, Stockholm, Sweden
| | - Stefanie Hiltbrunner
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet KS L2:04, SE-17176, Stockholm, Sweden
| | - Rosanne E Veerman
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet KS L2:04, SE-17176, Stockholm, Sweden
| | - Tanja I Näslund
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet KS L2:04, SE-17176, Stockholm, Sweden
| | - Susanne Gabrielsson
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet KS L2:04, SE-17176, Stockholm, Sweden.
| |
Collapse
|
31
|
|
32
|
Armstrong JPK, Holme MN, Stevens MM. Re-Engineering Extracellular Vesicles as Smart Nanoscale Therapeutics. ACS NANO 2017; 11:69-83. [PMID: 28068069 PMCID: PMC5604727 DOI: 10.1021/acsnano.6b07607] [Citation(s) in RCA: 454] [Impact Index Per Article: 56.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
In the past decade, extracellular vesicles (EVs) have emerged as a key cell-free strategy for the treatment of a range of pathologies, including cancer, myocardial infarction, and inflammatory diseases. Indeed, the field is rapidly transitioning from promising in vitro reports toward in vivo animal models and early clinical studies. These investigations exploit the high physicochemical stability and biocompatibility of EVs as well as their innate capacity to communicate with cells via signal transduction and membrane fusion. This review focuses on methods in which EVs can be chemically or biologically modified to broaden, alter, or enhance their therapeutic capability. We examine two broad strategies, which have been used to introduce a wide range of nanoparticles, reporter systems, targeting peptides, pharmaceutics, and functional RNA molecules. First, we explore how EVs can be modified by manipulating their parent cells, either through genetic or metabolic engineering or by introducing exogenous material that is subsequently incorporated into secreted EVs. Second, we consider how EVs can be directly functionalized using strategies such as hydrophobic insertion, covalent surface chemistry, and membrane permeabilization. We discuss the historical context of each specific technology, present prominent examples, and evaluate the complexities, potential pitfalls, and opportunities presented by different re-engineering strategies.
Collapse
Affiliation(s)
- James PK Armstrong
- Department of Materials, Department of Bioengineering, and Institute for Biomedical Engineering, Imperial College, London, U.K. SW7 2AZ
| | - Margaret N Holme
- Department of Materials, Department of Bioengineering, and Institute for Biomedical Engineering, Imperial College, London, U.K. SW7 2AZ
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering, and Institute for Biomedical Engineering, Imperial College, London, U.K. SW7 2AZ
| |
Collapse
|
33
|
Ingato D, Lee JU, Sim SJ, Kwon YJ. Good things come in small packages: Overcoming challenges to harness extracellular vesicles for therapeutic delivery. J Control Release 2016; 241:174-185. [DOI: 10.1016/j.jconrel.2016.09.016] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 09/18/2016] [Accepted: 09/19/2016] [Indexed: 12/15/2022]
|
34
|
Mapping B-cell responses to Salmonella enterica serovars Typhimurium and Enteritidis in chickens for the discrimination of infected from vaccinated animals. Sci Rep 2016; 6:31186. [PMID: 27510219 PMCID: PMC4980624 DOI: 10.1038/srep31186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 07/13/2016] [Indexed: 12/17/2022] Open
Abstract
Serological surveillance and vaccination are important strategies for controlling infectious diseases of food production animals. However, the compatibility of these strategies is limited by a lack of assays capable of differentiating infected from vaccinated animals (DIVA tests) for established killed or attenuated vaccines. Here, we used next generation phage-display (NGPD) and a 2-proportion Z score analysis to identify peptides that were preferentially bound by IgY from chickens infected with Salmonella Typhimurium or S. Enteritidis compared to IgY from vaccinates, for both an attenuated and an inactivated commercial vaccine. Peptides that were highly enriched against IgY from at least 4 out of 10 infected chickens were selected: 18 and 12 peptides for the killed and attenuated vaccines, respectively. The ten most discriminatory peptides for each vaccine were identified in an ELISA using a training set of IgY samples. These peptides were then used in multi-peptide assays that, when analysing a wider set of samples from infected and vaccinated animals, diagnosed infection with 100% sensitivity and specificity. The data describes a method for the development of DIVA assays for conventional attenuated and killed vaccines.
Collapse
|
35
|
del Cacho E, Gallego M, Lillehoj HS, Quilez J, Lillehoj EP, Sánchez-Acedo C. Induction of protective immunity against experimental Eimeria tenella infection using serum exosomes. Vet Parasitol 2016; 224:1-6. [DOI: 10.1016/j.vetpar.2016.04.043] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 04/24/2016] [Accepted: 04/30/2016] [Indexed: 12/31/2022]
|
36
|
Nagy N, Bódi I, Oláh I. Avian dendritic cells: Phenotype and ontogeny in lymphoid organs. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 58:47-59. [PMID: 26751596 DOI: 10.1016/j.dci.2015.12.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 12/26/2015] [Accepted: 12/26/2015] [Indexed: 06/05/2023]
Abstract
Dendritic cells (DC) are critically important accessory cells in the innate and adaptive immune systems. Avian DCs were originally identified in primary and secondary lymphoid organs by their typical morphology, displaying long cell processes with cytoplasmic granules. Several subtypes are known. Bursal secretory dendritic cells (BSDC) are elongated cells which express vimentin intermediate filaments, MHC II molecules, macrophage colony-stimulating factor 1 receptor (CSF1R), and produce 74.3+ secretory granules. Avian follicular dendritic cells (FDC) highly resemble BSDC, express the CD83, 74.3 and CSF1R molecules, and present antigen in germinal centers. Thymic dendritic cells (TDC), which express 74.3 and CD83, are concentrated in thymic medulla while interdigitating DC are found in T cell-rich areas of secondary lymphoid organs. Avian Langerhans cells are a specialized 74.3-/MHC II+ cell population found in stratified squamous epithelium and are capable of differentiating into 74.3+ migratory DCs. During organogenesis hematopoietic precursors of DC colonize the developing lymphoid organ primordia prior to immigration of lymphoid precursor cells. This review summarizes our current understanding of the ontogeny, cytoarchitecture, and immunophenotype of avian DC, and offers an antibody panel for the in vitro and in vivo identification of these heterogeneous cell types.
Collapse
Affiliation(s)
- Nándor Nagy
- Department of Human Morphology and Developmental Biology, Faculty of Medicine, Semmelweis University, 1094 Budapest, Tuzolto str. 58, Hungary.
| | - Ildikó Bódi
- Department of Human Morphology and Developmental Biology, Faculty of Medicine, Semmelweis University, 1094 Budapest, Tuzolto str. 58, Hungary
| | - Imre Oláh
- Department of Human Morphology and Developmental Biology, Faculty of Medicine, Semmelweis University, 1094 Budapest, Tuzolto str. 58, Hungary
| |
Collapse
|
37
|
|
38
|
Extracellular Vesicles: Role in Inflammatory Responses and Potential Uses in Vaccination in Cancer and Infectious Diseases. J Immunol Res 2015; 2015:832057. [PMID: 26380326 PMCID: PMC4562184 DOI: 10.1155/2015/832057] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 02/21/2015] [Indexed: 12/18/2022] Open
Abstract
Almost all cells and organisms release membrane structures containing proteins, lipids, and nucleic acids called extracellular vesicles (EVs), which have a wide range of functions concerning intercellular communication and signaling events. Recently, the characterization and understanding of their biological role have become a main research area due to their potential role in vaccination, as biomarkers antigens, early diagnostic tools, and therapeutic applications. Here, we will overview the recent advances and studies of Evs shed by tumor cells, bacteria, parasites, and fungi, focusing on their inflammatory role and their potential use in vaccination and diagnostic of cancer and infectious diseases.
Collapse
|
39
|
Schorey JS, Cheng Y, Singh PP, Smith VL. Exosomes and other extracellular vesicles in host-pathogen interactions. EMBO Rep 2014; 16:24-43. [PMID: 25488940 DOI: 10.15252/embr.201439363] [Citation(s) in RCA: 546] [Impact Index Per Article: 49.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
An effective immune response requires the engagement of host receptors by pathogen-derived molecules and the stimulation of an appropriate cellular response. Therefore, a crucial factor in our ability to control an infection is the accessibility of our immune cells to the foreign material. Exosomes-which are extracellular vesicles that function in intercellular communication-may play a key role in the dissemination of pathogen- as well as host-derived molecules during infection. In this review, we highlight the composition and function of exosomes and other extracellular vesicles produced during viral, parasitic, fungal and bacterial infections and describe how these vesicles could function to either promote or inhibit host immunity.
Collapse
Affiliation(s)
- Jeffrey S Schorey
- Department of Biological Sciences, Eck Institute for Global Health University of Notre Dame, Notre Dame, IN, USA
| | - Yong Cheng
- Department of Biological Sciences, Eck Institute for Global Health University of Notre Dame, Notre Dame, IN, USA
| | - Prachi P Singh
- Department of Biological Sciences, Eck Institute for Global Health University of Notre Dame, Notre Dame, IN, USA
| | - Victoria L Smith
- Department of Biological Sciences, Eck Institute for Global Health University of Notre Dame, Notre Dame, IN, USA
| |
Collapse
|
40
|
Zhang B, Yin Y, Lai RC, Lim SK. Immunotherapeutic potential of extracellular vesicles. Front Immunol 2014; 5:518. [PMID: 25374570 PMCID: PMC4205852 DOI: 10.3389/fimmu.2014.00518] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 10/04/2014] [Indexed: 12/20/2022] Open
Abstract
Extracellular vesicle or EV is a term that encompasses all classes of secreted lipid membrane vesicles. Despite being scientific novelties, EVs are gaining importance as a mediator of important physiological and pathological intercellular activities possibly through the transfer of their cargo of protein and RNA between cells. In particular, exosomes, the currently best characterized EVs have been notable for their in vitro and in vivo immunomodulatory activities. Exosomes are nanometer-sized endosome-derived vesicles secreted by many cell types and their immunomodulatory potential is independent of their cell source. Besides immune cells such as dendritic cells, macrophages, and T cells, cancer and stem cells also secrete immunologically active exosomes that could influence both physiological and pathological processes. The immunological activities of exosomes affect both innate and adaptive immunity and include antigen presentation, T cell activation, T cell polarization to regulatory T cells, immune suppression, and anti-inflammation. As such, exosomes carry much immunotherapeutic potential as a therapeutic agent and a therapeutic target.
Collapse
Affiliation(s)
- Bin Zhang
- Exosome and Secreted Nano-vesicle Group, ASTAR Institute of Medical Biology , Singapore
| | - Yijun Yin
- Exosome and Secreted Nano-vesicle Group, ASTAR Institute of Medical Biology , Singapore
| | - Ruenn Chai Lai
- Exosome and Secreted Nano-vesicle Group, ASTAR Institute of Medical Biology , Singapore
| | - Sai Kiang Lim
- Exosome and Secreted Nano-vesicle Group, ASTAR Institute of Medical Biology , Singapore ; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore , Singapore
| |
Collapse
|
41
|
György B, Hung ME, Breakefield XO, Leonard JN. Therapeutic applications of extracellular vesicles: clinical promise and open questions. Annu Rev Pharmacol Toxicol 2014; 55:439-464. [PMID: 25292428 DOI: 10.1146/annurev-pharmtox-010814-124630] [Citation(s) in RCA: 404] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This review provides an updated perspective on rapidly proliferating efforts to harness extracellular vesicles (EVs) for therapeutic applications. We summarize current knowledge, emerging strategies, and open questions pertaining to clinical potential and translation. Potentially useful EVs comprise diverse products of various cell types and species. EV components may also be combined with liposomes and nanoparticles to facilitate manufacturing as well as product safety and evaluation. Potential therapeutic cargoes include RNA, proteins, and drugs. Strategic issues considered herein include choice of therapeutic agent, means of loading cargoes into EVs, promotion of EV stability, tissue targeting, and functional delivery of cargo to recipient cells. Some applications may harness natural EV properties, such as immune modulation, regeneration promotion, and pathogen suppression. These properties can be enhanced or customized to enable a wide range of therapeutic applications, including vaccination, improvement of pregnancy outcome, and treatment of autoimmune disease, cancer, and tissue injury.
Collapse
Affiliation(s)
- Bence György
- Molecular Neurogenetics Unit, Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, Massachusetts 02114.,Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts 02115
| | - Michelle E Hung
- Interdepartmental Biological Sciences Graduate Program, Northwestern University, Evanston, Illinois 60208
| | - Xandra O Breakefield
- Molecular Neurogenetics Unit, Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, Massachusetts 02114
| | - Joshua N Leonard
- Department of Chemical and Biological Engineering, Robert H. Lurie Comprehensive Cancer Center, Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208
| |
Collapse
|
42
|
Cronemberger-Andrade A, Aragão-França L, de Araujo CF, Rocha VJ, Borges-Silva MDC, Figueiras CP, Oliveira PR, de Freitas LAR, Veras PST, Pontes-de-Carvalho L. Extracellular vesicles from Leishmania-infected macrophages confer an anti-infection cytokine-production profile to naïve macrophages. PLoS Negl Trop Dis 2014; 8:e3161. [PMID: 25232947 PMCID: PMC4169249 DOI: 10.1371/journal.pntd.0003161] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 08/04/2014] [Indexed: 12/13/2022] Open
Abstract
Background Extracellular vesicles (EVs) are structures with phospholipid bilayer membranes and 100–1000 nm diameters. These vesicles are released from cells upon activation of surface receptors and/or apoptosis. The production of EVs by dendritic cells, mast cells, macrophages, and B and T lymphocytes has been extensively reported in the literature. EVs may express MHC class II and other membrane surface molecules and carry antigens. The aim of this study was to investigate the role of EVs from Leishmania-infected macrophages as immune modulatory particles. Methodology/Principal Findings In this work it was shown that BALB/c mouse bone marrow-derived macrophages, either infected in vitro with Leishmania amazonensis or left uninfected, release comparable amounts of 50–300 nm-diameter extracellular vesicles (EVs). The EVs were characterized by flow cytometry and electron microscopy. The incubation of naïve macrophages with these EVs for 48 hours led to a statistically significant increase in the production of the cytokines IL-12, IL-1β, and TNF-α. Conclusions/Significance EVs derived from macrophages infected with L. amazonensis induce other macrophages, which in vivo could be bystander cells, to produce the proinflammatory cytokines IL-12, IL-1β and TNF-α. This could contribute both to modulate the immune system in favor of a Th1 immune response and to the elimination of the Leishmania, leading, therefore, to the control the infection. Leishmaniases are a group of diseases—each one individually called leishmaniasis—that are caused by the protozoan Leishmania. They affect millions of people and thousands of dogs in tropical and mediterranean countries. Macrophages are the main cellular hosts of Leishmania in the mammalian host, where it is an obligatorily intracellular parasite. In this work, it is shown that mouse bone marrow-derived macrophages, when infected in vitro with Leishmania, release small (no larger than 300 nm) extracellular vesicles (EVs), in the same way as uninfected macrophages. The EVs from the infected macrophages, however, induce in other macrophages the production of some cell hormones, named cytokines, which are involved with protection of the macrophage against infection and with the development of a protective immune response against the parasite.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Pablo R. Oliveira
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | | | | | | |
Collapse
|
43
|
Montaner S, Galiano A, Trelis M, Martin-Jaular L, Del Portillo HA, Bernal D, Marcilla A. The Role of Extracellular Vesicles in Modulating the Host Immune Response during Parasitic Infections. Front Immunol 2014; 5:433. [PMID: 25250031 PMCID: PMC4157553 DOI: 10.3389/fimmu.2014.00433] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 08/24/2014] [Indexed: 12/21/2022] Open
Abstract
Parasites are the cause of major diseases affecting billions of people. As the inflictions caused by these parasites affect mainly developing countries, they are considered as neglected diseases. These parasitic infections are often chronic and lead to significant immunomodulation of the host immune response by the parasite, which could benefit both the parasite and the host and are the result of millions of years of co-evolution. The description of parasite extracellular vesicles (EVs) in protozoa and helminths suggests that they may play an important role in host-parasite communication. In this review, recent studies on parasitic (protozoa and helminths) EVs are presented and their potential use as novel therapeutical approaches is discussed.
Collapse
Affiliation(s)
- Sergio Montaner
- Àrea de Parasitologia, Departament de Biologia Cel.lular i Parasitologia, Universitat de València , Burjassot , Spain
| | - Alicia Galiano
- Àrea de Parasitologia, Departament de Biologia Cel.lular i Parasitologia, Universitat de València , Burjassot , Spain
| | - María Trelis
- Àrea de Parasitologia, Departament de Biologia Cel.lular i Parasitologia, Universitat de València , Burjassot , Spain
| | - Lorena Martin-Jaular
- Barcelona Centre for International Health Research (CRESIB, Hospital Clínic-Universitat de Barcelona) , Barcelona , Spain
| | - Hernando A Del Portillo
- Barcelona Centre for International Health Research (CRESIB, Hospital Clínic-Universitat de Barcelona) , Barcelona , Spain ; Institució Catalana de Recerca i Estudis Avançats , Barcelona , Spain
| | - Dolores Bernal
- Departament de Bioquímica i Biologia Molecular, Universitat de València , Burjassot , Spain
| | - Antonio Marcilla
- Àrea de Parasitologia, Departament de Biologia Cel.lular i Parasitologia, Universitat de València , Burjassot , Spain
| |
Collapse
|
44
|
Fang L, Sun L, Yang J, Gu Y, Zhan B, Huang J, Zhu X. Heat shock protein 70 from Trichinella spiralis induces protective immunity in BALB/c mice by activating dendritic cells. Vaccine 2014; 32:4412-4419. [PMID: 24962751 DOI: 10.1016/j.vaccine.2014.06.055] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 06/04/2014] [Accepted: 06/11/2014] [Indexed: 11/17/2022]
Abstract
Trichinella spiralis heat shock protein 70 (Ts-Hsp70) is a protective antigen that induces partial protective immunity against T. spiralis infection in mice. To determine whether dendritic cells are involved in the mechanism responsible for the protection induced by Ts-Hsp70, mouse bone marrow-derived dendritic cells (DCs) were incubated with recombinant Ts-Hsp70 (rTs-Hsp70), and the DC-secreted cytokines and expressed surface markers were measured. The results demonstrated that rTs-Hsp70 activated DC maturation that was characterized by the secretion of IL-1β, IL-12p70, TNF-α, and IL-6 and the increased surface expression of CD11c, MHC II, CD40, CD80, and CD86. The rTs-Hsp70-activated DCs enabled the stimulation, proliferation and secretion of Th1/2 cytokines (i.e., INF-γ, IL-2, IL-4 and IL-6) in CD4(+) T cells from T. spiralis-infected mice. The mice that received rTs-Hsp70-activated DCs exhibited a 38.4% reduction in muscle larvae upon larval challenge with T. spiralis compared to the group that received PBS-incubated DCs. This partial protection was correlated with Th1 and Th2 mixed anti-Ts-Hsp70-specific immune responses that included high titers of total IgG, IgG1 and IgG2a and increased levels of Th1/2 cytokines (i.e., IFN-γ, IL-2, IL-4, IL-6). These results indicate that the rTs-Hsp70-induced protective immunity was mediated by the activation of the DCs and that rTs-Hsp70-loaded DCs could be an alternative vaccine approach against trichinellosis.
Collapse
Affiliation(s)
- Lei Fang
- Department of Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Liang Sun
- Department of Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Jing Yang
- Department of Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Yuan Gu
- Department of Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Bin Zhan
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jingjing Huang
- Department of Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Xinping Zhu
- Department of Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China.
| |
Collapse
|
45
|
Bacciu N, Bed'Hom B, Filangi O, Romé H, Gourichon D, Répérant JM, Le Roy P, Pinard-van der Laan MH, Demeure O. QTL detection for coccidiosis (Eimeria tenella) resistance in a Fayoumi × Leghorn F₂ cross, using a medium-density SNP panel. Genet Sel Evol 2014; 46:14. [PMID: 24552175 PMCID: PMC3936936 DOI: 10.1186/1297-9686-46-14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 01/28/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Coccidiosis is a major parasitic disease that causes huge economic losses to the poultry industry. Its pathogenicity leads to depression of body weight gain, lesions and, in the most serious cases, death in affected animals. Genetic variability for resistance to coccidiosis in the chicken has been demonstrated and if this natural resistance could be exploited, it would reduce the costs of the disease. Previously, a design to characterize the genetic regulation of Eimeria tenella resistance was set up in a Fayoumi × Leghorn F2 cross. The 860 F2 animals of this design were phenotyped for weight gain, plasma coloration, hematocrit level, intestinal lesion score and body temperature. In the work reported here, the 860 animals were genotyped for a panel of 1393 (157 microsatellites and 1236 single nucleotide polymorphism (SNP) markers that cover the sequenced genome (i.e. the 28 first autosomes and the Z chromosome). In addition, with the aim of finding an index capable of explaining a large amount of the variance associated with resistance to coccidiosis, a composite factor was derived by combining the variables of all these traits in a single variable. QTL detection was performed by linkage analysis using GridQTL and QTLMap. Single and multi-QTL models were applied. RESULTS Thirty-one QTL were identified i.e. 27 with the single-QTL model and four with the multi-QTL model and the average confidence interval was 5.9 cM. Only a few QTL were common with the previous study that used the same design but focused on the 260 more extreme animals that were genotyped with the 157 microsatellites only. Major differences were also found between results obtained with QTLMap and GridQTL. CONCLUSIONS The medium-density SNP panel made it possible to genotype new regions of the chicken genome (including micro-chromosomes) that were involved in the genetic control of the traits investigated. This study also highlights the strong variations in QTL detection between different models and marker densities.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Olivier Demeure
- INRA, UMR 1348 PEGASE, Domaine de la Prise, F-35590 Saint-Gilles, France.
| |
Collapse
|
46
|
Cha JO, Talha AFSM, Lim CW, Kim B. Effects of glass bead size, vortexing speed and duration on Eimeria acervulina oocyst excystation. Exp Parasitol 2014; 138:18-24. [PMID: 24457170 DOI: 10.1016/j.exppara.2014.01.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 12/01/2013] [Accepted: 01/07/2014] [Indexed: 11/18/2022]
Abstract
Improved methods for efficient excystation of Eimeria should be developed and standardized for future Eimeria-related studies. Here, the effects of different glass bead sizes (0.5, 1, 2, and 2.5 mm), and various vortex speeds (1000, 2000, and 3000 rpm) and durations (30 s, 1, 3, and 5 min) have been examined for Eimeria (E.) acervulina oocyst excystation. At 3000 rpm, all glass beads, regardless of size, efficiently ruptured E. acervulina oocysts at 5 min. At 2000 and 3000 rpm, all four glass bead sizes increasingly ruptured oocysts in a time-dependent manner. In contrast, at 1000 rpm the excystation efficiency was not related with the glass bead size or with vortexing duration. It appeared that the 1mm glass beads are most efficient for E. acervulina DNA extraction at a 3000 rpm vortexing speed for 3 and 5 min. The 2 mm glass beads delicately released the highest number of intact sporocysts at 2000 rpm for 3 min. Therefore, our data can provide valuable information for the efficient mechanical excystation of E. acervulina.
Collapse
Affiliation(s)
- Jang-Ock Cha
- Biosafety Research Institute and Laboratory of Pathology, College of Veterinary Medicine (BK21 Plus Program), Chonbuk National University, Jeonju, Republic of Korea
| | - Abul Fatah Shah Muhammad Talha
- Biosafety Research Institute and Laboratory of Pathology, College of Veterinary Medicine (BK21 Plus Program), Chonbuk National University, Jeonju, Republic of Korea
| | - Chae Woong Lim
- Biosafety Research Institute and Laboratory of Pathology, College of Veterinary Medicine (BK21 Plus Program), Chonbuk National University, Jeonju, Republic of Korea
| | - Bumseok Kim
- Biosafety Research Institute and Laboratory of Pathology, College of Veterinary Medicine (BK21 Plus Program), Chonbuk National University, Jeonju, Republic of Korea.
| |
Collapse
|
47
|
Securing poultry production from the ever-present Eimeria challenge. Trends Parasitol 2014; 30:12-9. [DOI: 10.1016/j.pt.2013.10.003] [Citation(s) in RCA: 245] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 10/02/2013] [Accepted: 10/06/2013] [Indexed: 01/16/2023]
|
48
|
Bedi B, McNair NN, Mead JR. Dendritic cells play a role in host susceptibility to Cryptosporidium parvum infection. Immunol Lett 2013; 158:42-51. [PMID: 24295591 DOI: 10.1016/j.imlet.2013.11.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 11/07/2013] [Accepted: 11/21/2013] [Indexed: 01/08/2023]
Abstract
Our previous studies have described dendritic cells (DCs) to be important sources of Th1 cytokines such as IL-12 and IL-2 in vitro, following stimulation with Cryptosporidium parvum antigens. We further established the role of DCs during cryptosporidiosis using a diphtheria toxin promoter regulated transgenic CD11c-DTR/EGFP mouse model. In vivo depletion of CD11c(+) cells in CD11c-DTR-Tg mice significantly increased susceptibility to C. parvum infection. Adoptive transfer of unstimulated or antigen stimulated DCs into CD11c(+) depleted CD11c-DTR-Tg mice resulted in an early decrease in parasite load at 4 days post infection. However, this response was transient since parasite load increased in mice engrafted with either unstimulated DCs or DCs stimulated with solubilized antigen by 6 days post infection. In contrast, in mice engrafted with DCs stimulated with live sporozoites, parasite load remained low during the entire period, suggesting the development of a more effective and sustained response. A corresponding increase in IFN-γ expression in T cells from spleen and mesenteric lymph nodes was also noted. Consistent with the in vivo engraftment study, DCs that are pulsed with live sporozoites in vitro and co-cultured with CD4(+) and CD8(+) T cells produced higher IFN-γ levels. Our study establishes the importance of DCs in susceptibility to infection by C. parvum and as important mediators of immune responses.
Collapse
Affiliation(s)
- Brahmchetna Bedi
- Atlanta VA Medical Center, Decatur, GA 30033, United States; Department of Pediatrics, Emory University, Atlanta, GA 30322, United States
| | - Nina N McNair
- Department of Pediatrics, Emory University, Atlanta, GA 30322, United States
| | - Jan R Mead
- Atlanta VA Medical Center, Decatur, GA 30033, United States; Department of Pediatrics, Emory University, Atlanta, GA 30322, United States.
| |
Collapse
|
49
|
Min W, Kim WH, Lillehoj EP, Lillehoj HS. Recent progress in host immunity to avian coccidiosis: IL-17 family cytokines as sentinels of the intestinal mucosa. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2013; 41:418-428. [PMID: 23583525 DOI: 10.1016/j.dci.2013.04.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 04/04/2013] [Accepted: 04/04/2013] [Indexed: 06/02/2023]
Abstract
The molecular and cellular mechanisms leading to immune protection against coccidiosis are complex and include multiple aspects of innate and adaptive immunities. Innate immunity is mediated by various subpopulations of immune cells that recognize pathogen associated molecular patterns (PAMPs) through their pattern recognition receptors (PRRs) leading to the secretion of soluble factors with diverse functions. Adaptive immunity, which is important in conferring protection against subsequent reinfections, involves subtypes of T and B lymphocytes that mediate antigen-specific immune responses. Recently, global gene expression microarray analysis has been used in an attempt to dissect this complex network of immune cells and molecules during avian coccidiosis. These new studies emphasized the uniqueness of the innate immune response to Eimeria infection, and directly led to the discovery of previously uncharacterized host genes and proteins whose expression levels were modulated following parasite infection. Among these is the IL-17 family of cytokines. This review highlights recent progress in IL-17 research in the context of host immunity to avian coccidiosis.
Collapse
Affiliation(s)
- Wongi Min
- College of Veterinary Medicine and Research Institute of Life Science, Gyeongsang National University, Jinju, Korea
| | | | | | | |
Collapse
|
50
|
Cheng Y, Schorey JS. Exosomes carrying mycobacterial antigens can protect mice against Mycobacterium tuberculosis infection. Eur J Immunol 2013; 43:3279-90. [PMID: 23943377 DOI: 10.1002/eji.201343727] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 07/18/2013] [Accepted: 08/09/2013] [Indexed: 11/06/2022]
Abstract
Approximately 2 billion people are infected with Mycobacterium tuberculosis, the etiological agent of tuberculosis (TB), and an estimated 1.5 million individuals die annually from TB. Presently, Mycobacterium bovis BCG remains the only licensed TB vaccine; however, previous studies suggest its protective efficacy wanes over time and fails in preventing pulmonary TB. Therefore, a safe and effective vaccine is urgently required to replace BCG or boost BCG immunizations. Our previous studies revealed that mycobacterial proteins are released via exosomes from macrophages infected with M. tuberculosis or pulsed with M. tuberculosis culture filtrate proteins (CFP). In the present study, exosomes purified from macrophages treated with M. tuberculosis CFP were found to induce antigen-specific IFN-γ and IL-2-expressing CD4(+) and CD8(+) T cells. In exosome-vaccinated mice, there was a similar TH1 immune response but a more limited TH2 response compared to BCG-vaccinated mice. Using a low-dose M. tuberculosis mouse aerosol infection model, exosomes from CFP-treated macrophages were found to both prime a protective immune response as well as boost prior BCG immunization. The protection was equal to or superior to BCG. In conclusion, our findings suggest that exosomes might serve as a novel cell-free vaccine against an M. tuberculosis infection.
Collapse
Affiliation(s)
- Yong Cheng
- Department of Biological Sciences, Center for Rare and Neglected Diseases and Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN, USA
| | | |
Collapse
|