Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Microb Cell Fact ; 23(1): 61, 2024 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-38402145

RESUMEN

BACKGROUND: Helicobacter pylori (H. pylori) causes chronic gastric disease. An efficient oral vaccine would be mucosa-targeted and offer defense against colonization of invasive infection in the digestive system. Proteolytic enzymes and acidic environment in the gastrointestinal tract (GT) can, however, reduce the effectiveness of oral vaccinations. For the creation of an edible vaccine, L. lactis has been proposed as a means of delivering vaccine antigens. RESULTS: We developed a plSAM (pNZ8148-SAM) that expresses a multiepitope vaccine antigen SAM-WAE containing Urease, HpaA, HSP60, and NAP extracellularly (named LL-plSAM-WAE) to increase the efficacy of oral vaccinations. We then investigated the immunogenicity of LL-plSAM-WAE in Balb/c mice. Mice that received LL-plSAM-WAE or SAM-WAE with adjuvant showed increased levels of antibodies against H. pylori, including IgG and sIgA, and resulted in significant reductions in H. pylori colonization. Furthermore, we show that SAM-WAE and LL-plSAM-WAE improved the capacity to target the vaccine to M cells. CONCLUSIONS: These findings suggest that recombinant L. lactis could be a promising oral mucosa vaccination for preventing H. pylori infection.


Asunto(s)
Helicobacter pylori , Animales , Ratones , Inmunidad Mucosa , Factores de Virulencia , Vacunas Bacterianas , Ureasa , Vacunas Sintéticas , Ratones Endogámicos BALB C , Administración Oral
2.
Molecules ; 27(20)2022 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-36296526

RESUMEN

Cancer vaccines have been considered promising therapeutic strategies and are often constructed from whole cells, attenuated pathogens, carbohydrates, peptides, nucleic acids, etc. However, the use of whole organisms or pathogens can elicit unwanted immune responses arising from unforeseen reactions to the vaccine components. On the other hand, synthetic vaccines, which contain antigens that are conjugated, often with carrier proteins, can overcome these issues. Therefore, in this review we have highlighted the synthetic approaches and discussed several bioconjugation strategies for developing antigen-based cancer vaccines. In addition, the major synthetic biology approaches that were used to develop genetically modified cancer vaccines and their progress in clinical research are summarized here. Furthermore, to boost the immune responses of any vaccines, the addition of suitable adjuvants and a proper delivery system are essential. Hence, this review also mentions the synthesis of adjuvants and utilization of biomaterial scaffolds, which may facilitate the design of future cancer vaccines.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Ácidos Nucleicos , Humanos , Biología Sintética , Desarrollo de Vacunas , Vacunas Sintéticas , Neoplasias/prevención & control , Adyuvantes Inmunológicos/farmacología , Antígenos , Péptidos/química , Carbohidratos , Proteínas Portadoras , Materiales Biocompatibles
3.
Mol Pharm ; 17(12): 4603-4615, 2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-33175556

RESUMEN

Cancer nanovaccines have been widely explored to enhance immunotherapy efficiency, in which the significant irritation of antigen-specific cytotoxic T cells (CTLs) is the critical point. In this study, we developed a pH and reduction dual-sensitive nanovaccine (PMSN@OVA-MPN) composed of two parts. The inner part was made up of polyethyleneimine (PEI)-modified mesoporous silica nanoparticles (MSNs) loaded with model antigen ovalbumin (OVA) and the outer part was made up of disulfide bond-involved metal-phenolic networks (MPNs) as a protective corona. In vitro release experiments proved that PMSN@OVA-MPN could intelligently release OVA in the presence of reductive glutathione, but not in neutral phosphate-buffered saline (PBS). Moreover, in vitro cell assays indicated that the nanovaccine promoted not only the OVA uptake efficiency by DC2.4 cells but also antigen lysosome escape due to the proton sponge effect of PEI. Furthermore, in vivo animal experiments indicated that PMSN@OVA-MPN induced a large tumor-specific cellular immune response so as to effectively inhibit the growth of an existing tumor. Finally, the immune memory effect caused by the nanovaccine afforded conspicuous prophylaxis efficacy in neonatal tumors. Hence, the multifunctional vaccine delivery system prepared in this work exhibits a great application potential in cancer immunotherapy and offers a platform for the development of nanovaccines.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Portadores de Fármacos/química , Inmunoterapia Activa/métodos , Nanosferas/química , Neoplasias/terapia , Animales , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/farmacocinética , Línea Celular Tumoral , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Composición de Medicamentos/métodos , Liberación de Fármacos , Femenino , Humanos , Concentración de Iones de Hidrógeno , Inmunogenicidad Vacunal , Memoria Inmunológica , Estructuras Metalorgánicas/química , Ratones , Neoplasias/inmunología , Polietileneimina/química , Dióxido de Silicio/química
4.
Mol Pharm ; 15(3): 975-984, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-29359945

RESUMEN

In immunotherapy, induction of potent cellular immunity by vaccination is essential to treat intracellular infectious diseases and tumors. In this work, we designed a new synthetic peptide carrier, Cys-Trp-Trp-Arg8-Cys-Arg8-Cys-Arg8-Cys, for vaccine delivery by integrating a redox-responsive disulfide bond cross-linking and cell-penetrating peptide arginine octamer. The carrier peptide bound to the antigen protein ovalbumin (OVA) via electrostatic self-assembly to form peptide/OVA nanocomposites. Then, the spontaneous oxidization of the thiols of the cysteine residues induced interpeptide disulfide bond cross-linking to construct denser peptide/OVA condensates. The cell-penetrating peptides incorporated in the carrier peptide could increase antigen uptake by antigen presenting cells. After being internalized by antigen presenting cells, the antigen could be rapidly released in cytoplasm along with degradation of the disulfide bonds by intracellular glutathione, which could promote potent CD8+ T cell immunity. The cross-linked peptide/OVA condensates were used for subcutaneous vaccination. The results showed that the peptide carrier mediated potent antigen-specific immune response by significantly increasing IgG titer; splenocyte proliferation; the secretion level of cytokines INF-γ, IL-12, IL-4, and IL-10; immune memory function, and the activation and maturation of dendritic cells. From the results, the low-molecular weight vaccine-condensing peptide with definite chemical composition could be developed as a novel class of vaccine delivery systems.


Asunto(s)
Péptidos de Penetración Celular/química , Sistemas de Liberación de Medicamentos/métodos , Ovalbúmina/administración & dosificación , Vacunación/métodos , Vacunas de Subunidad/administración & dosificación , Animales , Antígenos/administración & dosificación , Antígenos/inmunología , Línea Celular , Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Femenino , Inmunidad Celular/efectos de los fármacos , Memoria Inmunológica/efectos de los fármacos , Inyecciones Subcutáneas , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Modelos Animales , Nanopartículas/química , Ovalbúmina/inmunología , Ovalbúmina/farmacocinética , Oxidación-Reducción , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/farmacocinética
5.
AAPS PharmSciTech ; 18(7): 2618-2625, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28243887

RESUMEN

Specific and effective delivery of DNA vaccines into dendritic cells (DCs) to express antigens is a precondition for induction of immune responses. Construction of a new DNA vaccine delivery system with the ability of programmed gene transfection may achieve this objective. In this study, we successfully integrated dendritic lipopeptide, charge-reversible polymer, and APC-targeted material into DNA vaccine delivery system through layer-by-layer (LBL) assembly. By the means of adjusting the weight ratios and concentration ratios of components, stable complexes were formulated with a particle size of 256.8 ± 10.7 nm and zeta potential of 25.1 ± 2.3 mV. Moreover, this DNA vaccine delivery system could achieve specific delivery into DCs, high transfection efficiency and low cytotoxicity, holding great promise for immunotherapy.


Asunto(s)
Células Dendríticas/inmunología , Técnicas de Transferencia de Gen , Concentración de Iones de Hidrógeno , Vacunas de ADN/administración & dosificación , Células HeLa , Humanos , Nanoestructuras , Transfección
6.
J Control Release ; 374: 39-49, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39111597

RESUMEN

Immunological adjuvants are vaccine components that enhance long-lasting adaptive immune responses to weakly immunogenic antigens. Monophosphoryl lipid A (MPLA) is a potent and safe vaccine adjuvant that initiates an early innate immune response by binding to the Toll-like receptor 4 (TLR4). Importantly, the binding and recognition process is highly dependent on the monomeric state of MPLA. However, current vaccine delivery systems often prioritize improving the loading efficiency of MPLA, while neglecting the need to maintain its monomeric form for optimal immune activation. Here, we introduce a Pickering emulsion-guided MPLA monomeric delivery system (PMMS), which embed MPLA into the oil-water interface to achieve the monomeric loading of MPLA. During interactions with antigen-presenting cells, PMMS functions as a chaperone for MPLA, facilitating efficient recognition by TLR4 regardless of the presence of lipopolysaccharide-binding proteins. At the injection site, PMMS efficiently elicited local immune responses, subsequently promoting the migration of antigen-internalized dendritic cells to the lymph nodes. Within the draining lymph nodes, PMMS enhanced antigen presentation and maturation of dendritic cells. In C57BL/6 mice models, PMMS vaccination provoked potent antigen-specific CD8+ T cell-based immune responses. Additionally, PMMS demonstrated strong anti-tumor effects against E.G7-OVA lymphoma. These data indicate that PMMS provides a straightforward and efficient strategy for delivering monomeric MPLA to achieve robust cellular immune responses and effective cancer immunotherapy.


Asunto(s)
Adyuvantes Inmunológicos , Células Dendríticas , Emulsiones , Lípido A , Ratones Endogámicos C57BL , Receptor Toll-Like 4 , Animales , Lípido A/análogos & derivados , Lípido A/administración & dosificación , Lípido A/química , Células Dendríticas/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/química , Vacunación/métodos , Femenino , Ratones , Sistemas de Liberación de Medicamentos , Adyuvantes de Vacunas/administración & dosificación , Adyuvantes de Vacunas/química , Presentación de Antígeno , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología
7.
Front Immunol ; 14: 1132129, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36845130

RESUMEN

Objective: Mucosal immunization was an effective defender against pathogens. Nasal vaccines could activate both systemic and mucosal immunity to trigger protective immune responses. However, due to the weak immunogenicity of nasal vaccines and the lack of appropriate antigen carriers, very few nasal vaccines have been clinically approved for human use, which was a major barrier to the development of nasal vaccines. Plant-derived adjuvants are promising candidates for vaccine delivery systems due to their relatively safe immunogenic properties. In particular, the distinctive structure of pollen was beneficial to the stability and retention of antigen in the nasal mucosa. Methods: Herein, a novel wild-type chrysanthemum sporopollenin vaccine delivery system loaded with a w/o/w emulsion containing squalane and protein antigen was fabricated. The unique internal cavities and the rigid external walls within the sporopollenin skeleton construction could preserve and stabilize the inner proteins. The external morphological characteristics were suitable for nasal mucosal administration with high adhesion and retention. Results: Secretory IgA antibodies in the nasal mucosa can be induced by the w/o/w emulsion with the chrysanthemum sporopollenin vaccine delivery system. Moreover, the nasal adjuvants produce a stronger humoral response (IgA and IgG) compared to squalene emulsion adjuvant. Mucosal adjuvant benefited primarily from prolongation of antigens in the nasal cavity, improvement of antigen penetration in the submucosa and promotion of CD8+ T cells in spleen. Disccusion: Based on effective delivering both the adjuvant and the antigen, the increase of protein antigen stability and the realization of mucosal retention, the chrysanthemum sporopollenin vaccine delivery system has the potential to be a promising adjuvant platform. This work provide a novel idea for the fabrication of protein-mucosal delivery vaccine.


Asunto(s)
Inmunidad Mucosa , Vacunas , Humanos , Emulsiones/farmacología , Mucosa Nasal , Adyuvantes Inmunológicos/farmacología , Antígenos
8.
Vaccine X ; 14: 100312, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37234593

RESUMEN

Drones (uncrewed aerial vehicles or UAVs) introduce new opportunities to improve vaccine distribution systems, particularly in regions with limited transportation infrastructure where maintaining the cold chain is challenging. This paper addresses the use of drones to deliver vaccines to hard-to-reach populations using a novel optimization model to strategically design a multimodal vaccine distribution network. The model is illustrated in a case study for distributing routine childhood vaccines in Vanuatu, a South Pacific island nation with limited transportation infrastructure. Our research incorporates multiple drone types, recharging of drones, a cold chain travel time limit, transshipment delays for switching transport modes, and practical limits on the vaccine paths and drone trips. The goal is to locate facilities (distribution centers, drone bases, and relay stations) and design vaccine paths to minimize transportation costs, including the fixed costs for facilities and transportation links and variable costs for transportation through the network. Results show large potential cost savings and improved service quality provided by incorporating drones in a multimodal vaccine distribution system. Results also show the impact of introducing drones on the usage of other more expensive or slower transport modes.

9.
J Control Release ; 346: 380-391, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35483639

RESUMEN

Effective antigen delivery and immune stimulation in nasal mucosa determine the success of mucosal immunity. Here, an oil-in-ionic liquid (o/IL) nanoemulsion formulated with choline and niacin IL ([Cho][Nic]), squalene, and Tween 80 surfactant is explored as a vaccine delivery system for intranasal mucosal immunization. Compared to the o/w emulsion counterpart without the ILs, the o/IL manoemulsion showed a reduced and more uniform size of approximately 168 nm and significantly improved stability. Studies in mice model showed that when was used as an intranasal vaccine delivery system for influenza split-virus antigens, the antigens in the o/IL nanoemulsion induced strong mucosal immune responses with secretory IgA titers 25- and 5.8-fold higher than those of naked and commercial MF59-adjuvanted antigens, respectively. The o/IL nanoemulsion system also induced stronger systemic humoral responses. The excellent mucosal adjuvant effects of the o/IL nanoemulsion mainly benefited from the prolonged retention of antigens in the nasal cavity, enhanced antigen permeation into the submucosa, and the consequently promoted proliferation of CD11b cells and CD4+ T cells in nasal mucosa-associated lymphoid tissue. Moreover, when used as an injection adjuvant, the o/IL nanoemulsion also induced stronger humoral immune responses than MF59. Thus, the [Cho][Nic]-based o/IL nanoemulsion vaccine delivery system can serve as a promising adjuvant platform.


Asunto(s)
Vacunas contra la Influenza , Gripe Humana , Líquidos Iónicos , Adyuvantes Inmunológicos , Administración Intranasal , Animales , Anticuerpos Antivirales , Antígenos Virales , Humanos , Inmunidad Mucosa , Ratones , Ratones Endogámicos BALB C
10.
Biomater Adv ; 136: 212752, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35929287

RESUMEN

The ideal vaccine delivery systems can not only deliver antigens in intelligent manners but also act as adjuvants. Recently found that Mn2+ can effectively stimulate anti-tumor immune responses, and Ca2+ can regulate autophagy to promote the cross-presentation of antigens. Thus, we constructed such a manganese-containing multimode vaccine delivery system by using calcium-doped manganese carbonate microspheres (Ca@MnCO3) and perforin-listeria hemolysin (LLO), as termed as Ca@MnCO3/LLO. The two components Ca@MnCO3 and LLO, not only act as vaccine adjuvants by themselves, but also contribute to achieve cellular immunity. Among them, Ca@MnCO3 microspheres as an excellent Mn2+ and Ca2+ reservoir, can continuously release adjuvants Mn2+ and Ca2+ to enhance immune response in dendritic cells, while LLO can contribute to induce lysosomal escape. Particularly, Ca2+ was added firstly to MnCO3 microspheres to improve the stability and load capacity of the microspheres. Along with the degradation of intracellular Ca@MnCO3 microspheres, and the lysosomal membrane-lytic effects of perforin LLO, the Mn2+, Ca2+ and OVA were released to the cytoplasm. These outcomes cooperatively promote antigen cross-presentation, elicit CD8+ T cell proliferation, and finally achieve prominent anti-tumor effects. The results indicate that the manganese-containing vaccine delivery system Ca@MnCO3/LLO provides a promising platform for the construction of tumor vaccines.


Asunto(s)
Toxinas Bacterianas , Vacunas contra el Cáncer , Listeria monocytogenes , Adyuvantes Inmunológicos , Calcio , Carbonatos , Proteínas de Choque Térmico , Proteínas Hemolisinas , Inmunoterapia , Manganeso , Perforina
11.
Methods Mol Biol ; 2412: 339-354, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34918254

RESUMEN

Increasing emergence of infectious diseases is driving demand for new vaccine technologies capable of improving antigen delivery and protective efficacy. Nanoparticle technology is a modern approach to antigen delivery, capable of stabilizing and increasing the amount of antigen delivered to immune cells. Protein-based nanoparticles are a biodegradable alternative to existing nanomaterials, offering a versatile and biocompatible approach to nanoparticle vaccine delivery. In this chapter, the methods for the synthesis and characterization of protein-based nanocapsule vaccines are discussed. Initially, the requirements for a suitable nanoparticle vaccine are outlined, and finally, methods for the design and synthesis of protein-based nanocapsule vaccines are explained.


Asunto(s)
Nanocápsulas , Vacunas , Antígenos , Sistemas de Liberación de Medicamentos , Nanopartículas , Proteínas
12.
Biomaterials ; 280: 121313, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34894583

RESUMEN

While research on cancer vaccines has made great strides in the field of immunotherapy, the targeted delivery of multiple effective components (rational-tailored antigens and adjuvants) remains a challenge. Here, we utilized the unique hierarchical structures of Pickering emulsions (particles, oil core, and water-oil interface) to develop mannosylated (M) Pickering emulsions (PE) that target antigen presenting cells and synergistically deliver antigenic peptides and the TLR9 agonist CpG (C) as an enhanced cancer vaccine (MPE-C). We chemically linked mannose residues to PLGA/PLAG-PEG nanoparticles and produced a dense array of mannose on the nanopatterned surface of Pickering emulsions, allowing for increased cellular targeting. Together with the inherent deformability of the oily core, MPE-C increased the droplet-cellular contact area and provoked the cellular recognition of mannose and CpG for enhanced immune activation. We found that MPE-C attracted a large number of APCs to the local site of administration, evidently increasing cellular uptake and activation. Additionally, we observed increased antigen-specific cellular immune responses, with potent anti-tumor effects against both E.G7-OVA and B16-MUCI tumors. Furthermore, MPE-C combined with PD-1 antibodies produced a significant tumor regression, resulting in synergistic increases in anti-tumor effects. Thus, through the strategic loading of mannose, antigens, and CpG, Pickering emulsions could serve as a targeted delivery platform for enhanced multicomponent cancer vaccines.


Asunto(s)
Vacunas contra el Cáncer , Nanopartículas , Adyuvantes Inmunológicos/farmacología , Emulsiones/química , Inmunidad Celular , Nanopartículas/química
13.
Adv Drug Deliv Rev ; 186: 114325, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35550392

RESUMEN

With the pandemic of severe acute respiratory syndrome coronavirus 2, vaccine delivery systems emerged as a core technology for global public health. Given that antigen processing takes place inside the cell, the intracellular delivery and trafficking of a vaccine antigen will contribute to vaccine efficiency. Investigations focusing on the in vivo behavior and intracellular transport of vaccines have improved our understanding of the mechanisms relevant to vaccine delivery systems and facilitated the design of novel potent vaccine platforms. In this review, we cover the intracellular trafficking and in vivo fate of vaccines administered via various routes and delivery systems. To improve immune responses, researchers have used various strategies to modulate vaccine platforms and intracellular trafficking. In addition to progress in vaccine trafficking studies, the challenges and future perspectives for designing next-generation vaccines are discussed.


Asunto(s)
COVID-19 , Vacunas , Antígenos , COVID-19/prevención & control , Sistemas de Liberación de Medicamentos , Humanos
14.
Int J Biol Macromol ; 220: 183-192, 2022 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-35981671

RESUMEN

The immunogenicity and toxicity of N-2-Hydroxypropyl trimethyl ammonium chloride chitosan/N, O-carboxymethyl chitosan nanoparticles (N-2-HACC/CMCS NPs) as a universal vaccine adjuvant/delivery system remains unclear. The present study indicated that the positively charged N-2-HACC/CMCS NPs showed a regular spherical morphology, with a particle size of 219 ± 13.72 nm, zeta potential of 37.28 ± 4.58 mV, had hemocompatibility and biodegradation. Acute toxicity, repeated dose toxicity, abnormal toxicity, muscle stimulation, whole body allergic reaction evaluation in vitro, and cytotoxicity in vivo confirmed N-2-HACC/CMCS NPs is safe and non-toxic. N-2-HACC/OVA/CMCS NPs were prepared to evaluate the immunogenicity, which showed a particle size of 248.1 ± 15.53 nm, zeta potential of 17.24 ± 1.28 mV, encapsulation efficiency of 92.43 ± 0.96 %, and loading capacity of 42.97 ± 0.07 %. Oral or intramuscular route with the N-2-HACC/OVA/CMCS NPs in mice not only induced higher IgG, IgG1, IgG2a, and sIgA antibody titers, but also significantly produced higher levels of IL-6, IL-4, IFN-γ, and TNF-α, demonstrating that the N-2-HACC/OVA/CMCS NPs enhance humoral, cellular, and mucosal immune responses. Our results not only support the N-2-HACC/CMCS NPs to be a safe and potential universal nano adjuvant/delivery system in vaccine development, especially mucosal vaccines, but also rich the database knowledge of adjuvant/delivery systems, and provide new direction to introduce more licensed adjuvants.


Asunto(s)
Quitosano , Nanopartículas , Vacunas Virales , Adyuvantes Inmunológicos/farmacología , Cloruro de Amonio , Animales , Pollos , Quitosano/farmacología , Derivados de la Hipromelosa , Inmunoglobulina A Secretora , Inmunoglobulina G , Interleucina-4 , Interleucina-6 , Ratones , Factor de Necrosis Tumoral alfa
15.
Vaccines (Basel) ; 10(8)2022 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-35893829

RESUMEN

Generally, the injection method is recommended as the best efficient method for vaccine applications in fish. However, labor-intensive and difficult injection for certain fish sizes is always considered as a limitation to aquatic animals. To demonstrate the effectiveness of a novel oral delivery system for the piscine vaccine with nano-delivery made from nano clay, halloysite nanotubes (HNTs) and their modified forms were loaded with killed vaccines, and we determined the ability of the system in releasing vaccines in a mimic digestive system. The efficaciousness of the oral piscine vaccine nano-delivery system was evaluated for its level of antibody production and for the level of disease prevention in tilapia. Herein, unmodified HNTs (H) and modified HNTs [HNT-Chitosan (HC), HNT-APTES (HA) and HNT-APTES-Chitosan (HAC)] successfully harbored streptococcal bivalent vaccine with inactivated S. agalactiae, designated as HF, HAF, HCF and HACF. The releasing of the loading antigens in the mimic digestive tract demonstrated a diverse pattern of protein releasing depending on the types of HNTs. Remarkably, HCF could properly release loading antigens with relevance to the increasing pH buffer. The oral vaccines revealed the greatest elevation of specific antibodies to S. agalactiae serotype Ia in HCF orally administered fish and to some extent in serotype III. The efficacy of streptococcal disease protection was determined by continually feeding with HF-, HAF-, HCF- and HACF-coated feed pellets for 7 days in the 1st and 3rd week. HCF showed significant RPS (75.00 ± 10.83%) among the other tested groups. Interestingly, the HCF-treated group exhibited noticeable efficacy similar to the bivalent-vaccine-injected group (RPS 81.25 ± 0.00%). This novel nano-delivery system for the fish vaccine was successfully developed and exhibited appropriated immune stimulation and promised disease prevention through oral administration. This delivery system can greatly support animals' immune stimulation, which conquers the limitation in vaccine applications in aquaculture systems. Moreover, this delivery system can be applied to carrying diverse types of biologics, including DNA, RNA and subunit protein vaccines.

16.
Biomed Pharmacother ; 146: 112249, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34972632

RESUMEN

The emergence of many new viruses in recent times has resulted in a significant scientific challenge for discovering drugs and vaccines that effectively treat and prevent viral diseases. Nanotechnology has opened doors to prevent the spread of several diseases, including those caused by viruses. Polymer-hybrid nanodevices are a class of nanotechnology platforms for biomedical applications that present synergistic properties among their components, with improved performance compared to conventional forms of therapy. Considering the growing interest in this emerging field and the promising technological advantages of polymer-hybrid nanodevices, this work presents the current status of these systems in the context of prevention and treatment of viral diseases. A brief description of the different types of polymer-hybrid nanodevices highlighting some peculiar characteristics such as their composition, biodistribution, delivery of antigens, and overall immune responses in systemic tissues are discussed. Finally, the work presents the future trends for new nanotechnological hybrid materials based on polymers and perspectives for clinical use.


Asunto(s)
Antivirales/administración & dosificación , Nanopartículas/administración & dosificación , Nanotecnología/tendencias , Polímeros/administración & dosificación , Virosis/prevención & control , Animales , Antivirales/metabolismo , Sistemas de Liberación de Medicamentos , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/fisiología , Nanopartículas/metabolismo , Polímeros/metabolismo , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología , Virosis/metabolismo
17.
J Control Release ; 333: 107-128, 2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33774119

RESUMEN

An appropriate delivery system can improve the immune effects of antigens against various infections or tumors. Antigen-presenting cells (APCs) are specialized to capture and process antigens in vivo, which link the innate and adaptive immune responses. Functionalization of vaccine delivery systems with targeting moieties to APCs is a promising strategy for provoking potent immune responses. Additionally, the internalization and intracellular distribution of antigens are closely related to the initiation of downstream immune responses. With a deeper understanding of the intracellular microenvironment and the mechanisms of antigen presentation, vehicles designed to respond to endogenous and external stimuli can modulate antigen processing and presentation pathways, which are critical to the types of immune response. Here, an overview of extracellular targeting delivery of antigens to APCs and intracellular stimulus-responsiveness strategies is provided, which might be helpful for the rational design of vaccine delivery systems.


Asunto(s)
Nanopartículas , Vacunas , Presentación de Antígeno , Células Presentadoras de Antígenos , Antígenos , Células Dendríticas
18.
ACS Infect Dis ; 6(9): 2502-2512, 2020 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-32786276

RESUMEN

Group A Streptococcus (GAS) and GAS-related infections are a worldwide challenge, with no commercial GAS vaccine available. Polyethylenimine (PEI) attaches to the cells' surface and delivers cargo into endosomal and cytosolic compartments. We hypothesized that this will confer mucosal adjuvant properties for peptide antigens against group A Streptococcus (GAS). In this study, we successfully demonstrated the development of PEI incorporated liposomes for the delivery of a lipopeptide-based vaccine (LCP-1) against GAS. Outbred mice were administrated with the vaccine formulations intranasally, and immunological investigation showed that the PEI liposomes elicited significant mucosal and systemic immunity with the production of IgA and IgG antibodies. Antibodies were shown to effectively opsonize multiple isolates of clinically isolated GAS. This proof-of-concept study showed the capability for PEI liposomes to act as a safe vehicle for the delivery of GAS peptide antigens to elicit immune responses against GAS infection, making PEI a promising addition to liposomal mucosal vaccines.


Asunto(s)
Vacunas Estreptocócicas , Animales , Liposomas , Ratones , Polietileneimina , Streptococcus pyogenes , Vacunas de Subunidad
19.
Vaccine ; 37(42): 6232-6239, 2019 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-31466706

RESUMEN

Clostridium perfringens is a major cause of food poisoning worldwide, with its enterotoxin (CPE) being the major virulence factor. The C-terminus of CPE (C-CPE) is non-toxic and is the part of the toxin that binds to epithelial cells via the claudins in tight junctions; however, C-CPE has low antigenicity. To address this issue, we have used protein engineering technology to augment the antigenicity of C-CPE and have developed a C-CPE-based vaccine against C. perfringens-mediated food poisoning. Moreover, C-CPE has properties that make it potentially useful for the development of vaccines against other bacterial toxins that cause food poisoning. For example, we hypothesized that the ability of C-CPE to bind to claudins could be harnessed to deliver vaccine antigens directly to mucosa-associated lymphoid tissues, and we successfully developed a nasally administered C-CPE-based vaccine delivery system that promotes antigen-specific mucosal and systemic immune responses. In addition, our group has revealed the roles that the nasal mucus plays in lowering the efficacy of C-CPE-based nasal vaccines. Here, we review recent advances in the development of C-CPE-based vaccines against the major bacterial toxins that cause food poisoning and discuss our C-CPE-based nasal vaccine delivery system.


Asunto(s)
Vacunas Bacterianas/inmunología , Clostridium perfringens/inmunología , Enterotoxinas/inmunología , Enfermedades Transmitidas por los Alimentos/prevención & control , Claudinas/metabolismo , Enterotoxinas/genética , Células Epiteliales/inmunología , Enfermedades Transmitidas por los Alimentos/microbiología , Humanos , Inmunogenicidad Vacunal/inmunología , Membrana Mucosa/inmunología , Ingeniería de Proteínas/métodos , Vacunación
20.
J Ind Eng Chem ; 77: 12-24, 2019 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-32288512

RESUMEN

Most traditional vaccines are composed either of a whole pathogen or its parts; these vaccines, however, are not always effective and can even be harmful. As such, additional agents known as adjuvants are necessary to increase vaccine safety and efficacy. This review summarizes the potential of biodegradable materials, including synthetic and natural polymers, for vaccine delivery. These materials are highly biocompatible and have minimal toxicity, and most biomaterial-based vaccines delivering antigens or adjuvants have been shown to improve immune response, compared to formulations consisting of the antigen alone. Therefore, these materials can be applied in modern vaccine development.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda