Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 950
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Nature ; 628(8008): 612-619, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38509366

RESUMO

There is increasing interest in how immune cells in the meninges-the membranes that surround the brain and spinal cord-contribute to homeostasis and disease in the central nervous system1,2. The outer layer of the meninges, the dura mater, has recently been described to contain both innate and adaptive immune cells, and functions as a site for B cell development3-6. Here we identify organized lymphoid structures that protect fenestrated vasculature in the dura mater. The most elaborate of these dural-associated lymphoid tissues (DALT) surrounded the rostral-rhinal confluence of the sinuses and included lymphatic vessels. We termed this structure, which interfaces with the skull bone marrow and a comparable venous plexus at the skull base, the rostral-rhinal venolymphatic hub. Immune aggregates were present in DALT during homeostasis and expanded with age or after challenge with systemic or nasal antigens. DALT contain germinal centre B cells and support the generation of somatically mutated, antibody-producing cells in response to a nasal pathogen challenge. Inhibition of lymphocyte entry into the rostral-rhinal hub at the time of nasal viral challenge abrogated the generation of germinal centre B cells and class-switched plasma cells, as did perturbation of B-T cell interactions. These data demonstrate a lymphoid structure around vasculature in the dura mater that can sample antigens and rapidly support humoral immune responses after local pathogen challenge.


Assuntos
Dura-Máter , Imunidade Humoral , Tecido Linfoide , Veias , Administração Intranasal , Antígenos/administração & dosagem , Antígenos/imunologia , Medula Óssea/imunologia , Sistema Nervoso Central/irrigação sanguínea , Sistema Nervoso Central/imunologia , Dura-Máter/irrigação sanguínea , Dura-Máter/imunologia , Centro Germinativo/citologia , Centro Germinativo/imunologia , Vasos Linfáticos/imunologia , Tecido Linfoide/irrigação sanguínea , Tecido Linfoide/imunologia , Plasmócitos/imunologia , Crânio/irrigação sanguínea , Linfócitos T/imunologia , Veias/fisiologia , Humanos , Masculino , Feminino , Adulto , Pessoa de Meia-Idade , Animais , Camundongos , Idoso de 80 Anos ou mais
2.
Int Immunopharmacol ; 104: 108522, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35032825

RESUMO

Induction of tumor-specific CD8 + T cell responses is known as a major challenge for cancer vaccine development; here we presented a strategy to improve peptide nanofibers-mounted antitumor immune responses. To this end, peptide nanofibers bearing class I (Kb)-restricted epitope (Epi-Nano) were formulated with polyethylene imine backbone (Epi-Nano-PEI), and characterized using morphological and physicochemicalcharacterizationtechniques. Nanofibers were studied in terms of their uptake by antigen-presenting cells (APCs), antigen cross-presentation capacity, and cytotoxic activity. Furthermore, nanofibers were assessed by their potency to induce NLRP3 inflammasome-related cytokines and factors. Finally, the ability of nanofibers to induce tumor-specific CD8 T cells and tumor protection were investigated in tumor-bearing mice. The formulation of Epi-Nano with PEI led to the formation of short strand nanofibers with a positive surface charge, a low critical aggregation concentration (CAC), and an increased resistancetoproteolytic degradation. Epi-Nano-PEI was significantly taken up more efficiently by antigen-presenting cells (APCs), and was more potent in cross-presentation when compared to Epi-Nano. Moreover, Epi-Nano-PEI, in comparison to Epi-Nano, efficiently up-regulated the expression of NLRP3, caspase-1, IL-1b, IL18 and IL-6. Cell viability analysis showed that formulation of PEI with Epi-Nano not only abolished its cytotoxic activity, but surprisingly induced macrophage proliferation. Furthermore, it demonstrated that Epi-Nano-PEI triggered robust antigen-specific CD8+ T cell responses, and induced maximum antitumor response (tumor growth inhibition and prolonged survival) in tumor-bearing mice that were significantly higher compared to Epi-Nano. Taken together, the formulation of Epi-Nano with PEI is suggested as a promising strategy to improve nanofibers-mounted antitumor immune response.


Assuntos
Antígenos/administração & dosagem , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Epitopos/administração & dosagem , Nanofibras/administração & dosagem , Neoplasias/imunologia , Ovalbumina/administração & dosagem , Peptídeos/administração & dosagem , Polietilenoimina/administração & dosagem , Animais , Células Apresentadoras de Antígenos/imunologia , Linhagem Celular Tumoral , Feminino , Camundongos Endogâmicos C57BL
3.
Adv Sci (Weinh) ; 8(23): e2100118, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34693665

RESUMO

Recently, viral infectious diseases, including COVID-19 and Influenza, are the subjects of major concerns worldwide. One strategy for addressing these concerns focuses on nasal vaccines, which have great potential for achieving successful immunization via safe, easy, and affordable approaches. However, conventional nasal vaccines have major limitations resulting from fast removal when pass through nasal mucosa and mucociliary clearance hindering their effectiveness. Herein a nanoparticulate vaccine (NanoVac) exhibiting photochemical immunomodulation and constituting a new self-assembled immunization system of a photoactivatable polymeric adjuvant with influenza virus hemagglutinin for efficient nasal delivery and antigen-specific immunity against pathogenic influenza viruses is described. NanoVac increases the residence period of antigens and further enhances by spatiotemporal photochemical modulation in the nasal cavity. As a consequence, photochemical immunomodulation of NanoVacs successfully induces humoral and cellular immune responses followed by stimulation of mature dendritic cells, plasma cells, memory B cells, and CD4+ and CD8+ T cells, resulting in secretion of antigen-specific immunoglobulins, cytokines, and CD8+ T cells. Notably, challenge with influenza virus after nasal immunization with NanoVacs demonstrates robust prevention of viral infection. Thus, this newly designed vaccine system can serve as a promising strategy for developing vaccines that are active against current hazardous pathogen outbreaks and pandemics.


Assuntos
Hemaglutininas/química , Vacinas contra Influenza/administração & dosagem , Luz , Nanopartículas/química , Infecções por Orthomyxoviridae/prevenção & controle , Adjuvantes Imunológicos/administração & dosagem , Administração por Inalação , Animais , Antígenos/administração & dosagem , Antígenos/química , Antígenos/imunologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Hemaglutininas/administração & dosagem , Hemaglutininas/imunologia , Humanos , Imunidade Celular , Imunidade Humoral , Vacinas contra Influenza/química , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Influenza Humana/virologia , Interferon gama/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , Fármacos Fotossensibilizantes/química , Polímeros/química
4.
Goiânia; SES-GO; 31 ago 2021. 1-9 p. ilus.
Não convencional em Português | SES-GO, ColecionaSUS, CONASS, LILACS | ID: biblio-1290834

RESUMO

Em janeiro de 2020, os testes para detectar o SARS-CoV-2 em amostras coletadas de pacientes foram desenvolvidos logo após o sequenciamento e divulgação do genoma do vírus (CORMAN et al., 2020) e, desde então, diferentes metodologias de testagem , têm sido empregadas em contextos distintos. Embora sejam o padrão de referência para diagnóstico da infecção aguda pelo SARS-CoV-2, os testes moleculares de reação em cadeia da polimerase (RT-PCR) não podem ser dimensionados para atender às demandas extensas da saúde pública (MINA & ANDERSEN, 2021). O processo é limitado para algumas regiões devido à necessidade de equipamentos sofisticados, operadores extremamente qualificados ao tempo que pode decorrer. Contudo, os testes de antígeno, permitem limitar de forma eficaz a disseminação da COVID-19 e responder a surtos da pandemia. Foram levantadas no estudo as recomendações quanto aos testes de antígeno emitidos pela Organização Mundial da Saúde (OMS), pela Europa, pelo Reino Unido, Estados Unidos, Israel e Brasil.


In January 2020, tests to detect SARS-CoV-2 in samples collected from patients were developed soon after the sequencing and dissemination of the virus genome (CORMAN et al., 2020) and, since then, different testing methodologies, have been used in different contexts. Although they are the reference standard for diagnosing acute SARS-CoV-2 infection, molecular polymerase chain reaction (RT-PCR) tests cannot be scaled to meet the extensive demands of public health (MINA & ANDERSEN, 2021 ). The process is limited to some regions due to the need for sophisticated equipment, extremely qualified operators and the time that may elapse. However, antigen tests effectively limit the spread of COVID-19 and respond to pandemic outbreaks. Recommendations for antigen tests issued by the World Health Organization (WHO), Europe, the United Kingdom, the United States, Israel and Brazil were raised in the study.


Assuntos
Humanos , Masculino , Feminino , Gravidez , Recém-Nascido , Lactente , Pré-Escolar , Criança , Adolescente , Adulto , Pessoa de Meia-Idade , Idoso , Idoso de 80 Anos ou mais , Adulto Jovem , COVID-19/prevenção & controle , Antígenos/administração & dosagem
5.
Mol Pharm ; 18(8): 2867-2888, 2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-34264684

RESUMO

Despite the many advances that have occurred in the field of vaccine adjuvants, there are still unmet needs that may enable the development of vaccines suitable for more challenging pathogens (e.g., HIV and tuberculosis) and for cancer vaccines. Liposomes have already been shown to be highly effective as adjuvant/delivery systems due to their versatility and likely will find further uses in this space. The broad potential of lipid-based delivery systems is highlighted by the recent approval of COVID-19 vaccines comprising lipid nanoparticles with encapsulated mRNA. This review provides an overview of the different approaches that can be evaluated for the design of lipid-based vaccine adjuvant/delivery systems for protein, carbohydrate, and nucleic acid-based antigens and how these strategies might be combined to develop multicomponent vaccines.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Antígenos/administração & dosagem , Sistemas de Liberação de Medicamentos , Lipídeos/química , Nanopartículas/química , Vacinas/administração & dosagem , Vacinas contra COVID-19/administração & dosagem , Humanos , Lipossomos , SARS-CoV-2/imunologia , Vacinas/química
6.
Immunobiology ; 226(4): 152108, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34247017

RESUMO

In Algeria, Androctonus australis hector scorpion envenomation remains a major problem of public health because of non-efficient therapy. The development of safe vaccine against scorpion venom could be one key strategy for the envenomation prevention. The irradiation of venom by γ-rays develops suitable immunogens which produced effective antivenom and safe vaccine. In this study, we investigated the ability of the irradiated toxic fraction (γ-FtoxG50) to induce long-term memory humoral response in immunized animals (mice and rabbits), by involving the long-lived plasma cells to prevent efficiently the lethality of scorpion envenomation. For this purpose, an appropriate immunization schedule was established in mice and rabbits using three (3) similar doses of γ-FtoxG50 associated with Alum adjuvant. Obtained results indicate that the long-term immunogenicity of γ-FtoxG50 is able to induce the long-term memory humoral response with a high level of specific antibodies. The long-term persistence of antibody levels could depend on bone marrow memory plasma cells. These cells produce continuously antibodies without antigen stimulus. Furthermore, an enhanced memory response was obtained post-repeated envenomation with toxic native venom that leads to improved protection of animals. Together, pre-existing protective antibodies and the activation of memory B-cells could induce a rapid neutralization of scorpion toxins and long-term protection against scorpion envenomation.


Assuntos
Antígenos/administração & dosagem , Imunoglobulina G/imunologia , Neurotoxinas/administração & dosagem , Plasmócitos/imunologia , Venenos de Escorpião/administração & dosagem , Vacinas/administração & dosagem , Adjuvantes de Vacinas/administração & dosagem , Compostos de Alúmen/administração & dosagem , Animais , Antígenos/efeitos da radiação , Medula Óssea/imunologia , Feminino , Raios gama , Memória Imunológica , Camundongos , Neurotoxinas/efeitos da radiação , Coelhos , Venenos de Escorpião/efeitos da radiação , Baço/imunologia
7.
Ann Clin Lab Sci ; 51(3): 359-367, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-34162566

RESUMO

OBJECTIVE: The aim of this investigation was to evaluate the property of bovine lactoferrin (LF) in the generation of delayed type hypersensitivity (DTH) as an oral adjuvant during immunization with ovalbumin (OVA) and BCG. METHODS: LF admixed with OVA or BCG was used for immunization of CBA or C57BL/6 mice when given via oral or subcutaneous routes. Elicited DTH response was measured post immunization. Inhibition studies using mannose or galactose were accomplished by gavage prior to oral administration of antigens. LF was also examined for effects on BCG uptake by bone marrow derived macrophages (BMM). RESULTS: LF at doses of 1.0 mg and 10.0 mg, admixed with OVA (10.0 mg), significantly enhanced the antigen-specific DTH reaction. The stimulatory effects of LF were inhibited by the oral pretreatment of mice with 50.0 mg of mannose but not galactose. LF also enhanced the DTH reaction to orally administered BCG. LF enhanced uptake of BCG by BMM in a dose-dependent manner. CONCLUSION: LF was able to augment development of DTH when orally administered with OVA or BCG antigens. Inhibition studies suggest the involvement of the receptor with an affinity to mannose in mediation of the adjuvant effect. LF augmentation of the DTH response was partially effective when given in advance of oral delivery of the antigen; this effect could also be saturated by mannose. BCG studies provide preliminary evidence for LF in the potential augmentation of oral vaccination to prevent mycobacterial infection. In vitro experiments provide evidence that LF plays a role in modulation of antigen presenting cell activation.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Antígenos/administração & dosagem , Hipersensibilidade Tardia/patologia , Lactoferrina/administração & dosagem , Macrófagos/imunologia , Mycobacterium bovis/imunologia , Ovalbumina/administração & dosagem , Administração Oral , Animais , Antígenos/imunologia , Hipersensibilidade Tardia/etiologia , Lactoferrina/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Ovalbumina/imunologia
8.
Front Immunol ; 12: 736936, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35003059

RESUMO

Palladium (Pd) is a widely used metal and extremely important biomaterial for the reconstruction of occlusions during dental restorations. However, metallic biomaterials can cause serious allergic reactions, such as Pd-related oral mucositis seen in dentistry. Metal allergy is categorized as a type IV allergy and we demonstrated that CD8 T cells play an important role in Pd allergy previously. As TCR of CD8 T cells recognizes MHC class I/peptide complex, the antigen specificity to this complex seems to be generated during Pd allergy. However, it remains unknown if Pd affects the MHC class I/peptide complex. In this study, we investigated the behavior of the MHC class I/peptide complex in response to Pd treatment. We found that PdCl2 treatment altered peptide presentation on MHC class I and that co-culture with Pd-treated DC2.4 cells induced activation of Pd-responsive TCR-expressing T cell line. Furthermore, PdCl2 treatment induced temporal MHC class I internalization and inhibition of membrane movement suppressed Pd-induced T cell-mediated antigenicity. These data suggest that Pd-induced MHC class I internalization is critical for generation of antigenicity through a mechanism including differential peptide loading on MHC class I, which results in Pd allergy.


Assuntos
Antígenos/efeitos adversos , Linfócitos T CD8-Positivos/imunologia , Hipersensibilidade a Drogas/etiologia , Hipersensibilidade a Drogas/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Paládio/efeitos adversos , Animais , Antígenos/administração & dosagem , Linhagem Celular , Membrana Celular/metabolismo , Células Dendríticas/imunologia , Feminino , Humanos , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/efeitos adversos , Camundongos , Camundongos Endogâmicos C57BL , Paládio/administração & dosagem , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo
9.
Nat Nanotechnol ; 15(12): 1053-1064, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33106640

RESUMO

Cancer vaccines hold great promise for improved cancer treatment. However, endosomal trapping and low immunogenicity of tumour antigens usually limit the efficiency of vaccination strategies. Here, we present a proton-driven nanotransformer-based vaccine, comprising a polymer-peptide conjugate-based nanotransformer and loaded antigenic peptide. The nanotransformer-based vaccine induces a strong immune response without substantial systemic toxicity. In the acidic endosomal environment, the nanotransformer-based vaccine undergoes a dramatic morphological change from nanospheres (about 100 nanometres in diameter) into nanosheets (several micrometres in length or width), which mechanically disrupts the endosomal membrane and directly delivers the antigenic peptide into the cytoplasm. The re-assembled nanosheets also boost tumour immunity via activation of specific inflammation pathways. The nanotransformer-based vaccine effectively inhibits tumour growth in the B16F10-OVA and human papilloma virus-E6/E7 tumour models in mice. Moreover, combining the nanotransformer-based vaccine with anti-PD-L1 antibodies results in over 83 days of survival and in about half of the mice produces complete tumour regression in the B16F10 model. This proton-driven transformable nanovaccine offers a robust and safe strategy for cancer immunotherapy.


Assuntos
Antígenos/administração & dosagem , Vacinas Anticâncer/administração & dosagem , Preparações de Ação Retardada/química , Nanosferas/química , Neoplasias/prevenção & controle , Animais , Antígenos/uso terapêutico , Vacinas Anticâncer/uso terapêutico , Linhagem Celular Tumoral , Feminino , Humanos , Concentração de Íons de Hidrogênio , Imunoterapia , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/patologia , Polímeros/química , Prótons
10.
Biotechnol J ; 15(12): e2000100, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32679620

RESUMO

Biotinylated peptide amphiphile (Biotin-PA) nanofibers, are designed as a noncovalent binding location for antigens, which are adjuvants to enhance, accelerate, and prolong the immune response triggered by antigens. Presenting antigens on synthetic Biotin-PA nanofibers generated a higher immune response than the free antigens delivered with a cytosine-phosphate-guanine oligodeoxynucleotides (CpG ODN) (TLR9 agonist) adjuvant. Antigen attached Biotin-PA nanofibers trigger splenocytes to produce high levels of cytokines (IFN-γ, IL-12, TNF-α, and IL-6) and to exhibit a superior cross-presentation of the antigen. Both Biotin-PA nanofibers and CpG ODN induce a Th-1-biased IgG subclass response; however, delivering the antigen with Biotin-PA nanofibers induce significantly greater production of total IgG and subclasses of IgG compared to delivering the antigen with CpG ODN. Contrary to CpG ODN, Biotin-PA nanofibers also enhance antigen-specific splenocyte proliferation and increase the proportion of the antigen-specific CD8(+) T cells. Given their biodegradability and biocompatibility, Biotin-PA nanofibers have a significant potential in immunoengineering applications as a biomaterial for the delivery of a diverse set of antigens derived from intracellular pathogens, emerging viral diseases such as COVID-19, or cancer cells to induce humoral and cellular immune responses against the antigens.


Assuntos
Adjuvantes Imunológicos/química , Nanofibras/química , Peptídeos/química , Peptídeos/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Apresentação de Antígeno , Células Apresentadoras de Antígenos/citologia , Células Apresentadoras de Antígenos/imunologia , Antígenos/administração & dosagem , Antígenos/química , Materiais Biocompatíveis/química , Biotecnologia , Biotina/análogos & derivados , Citocinas/metabolismo , Desenho de Fármacos , Imunidade Celular , Imunidade Humoral , Técnicas In Vitro , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Nanofibras/administração & dosagem , Nanofibras/ultraestrutura , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Peptídeos/administração & dosagem , Engenharia de Proteínas
11.
Int J Nanomedicine ; 15: 2685-2697, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32368049

RESUMO

BACKGROUND: Nanocarriers could deliver significantly higher amounts of antigen to antigen-presenting cells (APCs), which have great potential to stimulate humoral and cellular response in cancer immunotherapy. Thereafter, silica solid nanosphere (SiO2) was prepared, and a model antigen (ovalbumin, OVA) was covalently conjugated on the surface of SiO2 to form nanovaccine (OVA@SiO2). And the application of OVA@SiO2 for cancer immunotherapy was evaluated. MATERIALS AND METHODS: SiO2 solid nanosphere was prepared by the Stöber method, then successively aminated by aminopropyltriethoxysilane and activated with glutaraldehyde. OVA was covalently conjugated on the surface of activated SiO2 to obtain nanovaccine (OVA@SiO2). Dynamic light scattering, scanning electron microscope, and transmission electron microscope were conducted to identify the size distribution, zeta potential and morphology of OVA@SiO2. The OVA loading capacity was investigated by varying glutaraldehyde concentration. The biocompatibility of OVA@SiO2 to DC2.4 and RAW246.7 cells was evaluated by a Cell Counting Kit-8 assay. The uptake of OVA@SiO2 by DC2.4 and its internalization pathway were evaluated in the absence or presence of different inhibitors. The activation and maturation of bone marrow-derived DC cells by OVA@SiO2 were also investigated. Finally, the in vivo transport of OVA@SiO2 and its toxicity to organs were appraised. RESULTS: All results indicated the successful covalent conjugation of OVA on the surface of SiO2. The as-prepared OVA@SiO2 possessed high antigen loading capacity, which had good biocompatibility to APCs and major organs. Besides, OVA@SiO2 facilitated antigen uptake by DC2.4 cells and its cytosolic release. Noteworthily, OVA@SiO2 significantly promoted the maturation of dendritic cells and up-regulation of cytokine secretion by co-administration of adjuvant CpG-ODN. CONCLUSION: The as-prepared SiO2 shows promising potential for use as an antigen delivery carrier.


Assuntos
Antígenos/metabolismo , Vacinas Anticâncer/farmacologia , Imunoterapia/métodos , Nanosferas/química , Ovalbumina/química , Adjuvantes Imunológicos/administração & dosagem , Animais , Apresentação de Antígeno , Antígenos/administração & dosagem , Antígenos/química , Antígenos/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/química , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Nanosferas/administração & dosagem , Oligodesoxirribonucleotídeos/administração & dosagem , Ovalbumina/imunologia , Ovalbumina/farmacocinética , Células RAW 264.7 , Dióxido de Silício/química
12.
PLoS One ; 15(2): e0229042, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32097442

RESUMO

T cell anergy is known to be a crucial mechanism for various types of immune tolerance, including oral tolerance. The expression of several anergy-specific genes was reportedly up-regulated in anergic T cells, and played important roles in the cells. However, how the genes were up-regulated has not been understood. In this study, we comprehensively analyzed the altered gene expression and DNA methylation status in T cells tolerized by oral antigen in vivo. Our results showed that many genes were significantly up-regulated in the orally tolerized T cells, and most of the genes found in this study have not been reported previously as anergy related genes; for example, ribosomal protein L41 (FC = 3.54E06, p = 3.70E-09: Fisher's exact test; the same applies hereinafter) and CD52 (FC = 2.18E05, p = 3.44E-06). Furthermore, we showed that the DNA methylation statuses of many genes; for example, enoyl-coenzyme A delta isomerase 3 (FC = 3.62E-01, p = 3.01E-02) and leucine zipper protein 1 (FC = 4.80E-01, p = 3.25E-02), including the ones distinctly expressed in tolerized T cells; for example, latexin (FC = 3.85E03, p = 4.06E-02 for expression; FC = 7.75E-01, p = 4.13E-01 for DNA methylation) and small nuclear ribonucleoprotein polypeptide F (FC = 3.12E04, p = 4.46E-04 for expression; FC = 8.56E-01, p = 5.15E-01 for DNA methylation), changed during tolerization, suggesting that the distinct expression of some genes was epigenetically regulated in the tolerized T cells. This study would contribute to providing a novel clue to the fine understanding of the mechanism for T cell anergy and oral tolerance.


Assuntos
Metilação de DNA , Regulação da Expressão Gênica , Linfócitos T/metabolismo , Administração Oral , Animais , Antígenos/administração & dosagem , Antígenos/imunologia , Biomarcadores , Ilhas de CpG , Citosina , Epigênese Genética , Perfilação da Expressão Gênica , Tolerância Imunológica , Ativação Linfocitária/imunologia , Camundongos , Transdução de Sinais , Linfócitos T/imunologia
13.
Biomater Sci ; 8(4): 1020-1044, 2020 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-31621709

RESUMO

Oral drug delivery remains the most preferred approach due to its multiple advantages. Recently there has been increasing interest in the development of advanced vehicles for oral delivery of different therapeutics. Among them, biomimetic and bioinspired strategies are emerging as novel approaches that are promising for addressing biological barriers encountered by traditional drug delivery systems. Herein we provide a state-of-the-art review on the current progress of biomimetic particulate oral delivery systems. Different biomimetic nanoparticles used for oral drug delivery are first discussed, mainly including ligand/antibody-functionalized nanoparticles, transporter-mediated nanoplatforms, and nanoscale extracellular vesicles. Then we describe bacteria-derived biomimetic systems, with respect to oral delivery of therapeutic proteins or antigens. Subsequently, yeast-derived oral delivery systems, based on either chemical engineering or bioengineering approaches are discussed, with emphasis on the treatment of inflammatory diseases and cancer as well as oral vaccination. Finally, bioengineered plant cells are introduced for oral delivery of biological agents. A future perspective is also provided to highlight the existing challenges and possible resolution toward clinical translation of currently developed biomimetic oral therapies.


Assuntos
Antígenos/administração & dosagem , Biomimética/métodos , Proteínas/administração & dosagem , Administração Oral , Antígenos/química , Bioengenharia , Sistemas de Liberação de Medicamentos , Humanos , Nanopartículas/química , Proteínas/química
14.
Int J Nanomedicine ; 14: 8235-8249, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31802864

RESUMO

BACKGROUND: The effective induction of an antigen-specific T cell immune response through dendritic cell activation is one of the key goals of tumor immunotherapy. METHODS: In this study, efficient antigen-delivery carriers using silica-coated magnetic nanoparticles were designed and, their antigen-specific T cell immune response through dendritic cell activation investigated. RESULTS: The results showed that the silica-coated magnetic nanoparticles with conjugated ovalbumin enhanced the production of cytokines and antigen uptake in bone marrow-derived dendritic cells. Also, this induced an antigen-specific cytotoxic T lymphocyte (CTL) immune response and activated antigen-specific Th1 cell responses, including IL-2 and IFN-γ production and proliferation. We proved that the immune-stimulatory effects of silica-coated magnetic nanoparticles with conjugated ovalbumin were efficient in inhibiting of tumor growth in EG7-OVA (mouse lymphoma-expressing ovalbumin tumor-bearing mice model). CONCLUSION: Therefore, the silica-coated magnetic nanoparticles with conjugated ovalbumin are expected to be useful as efficient anti-cancer immunotherapy agents.


Assuntos
Imunoterapia , Nanopartículas de Magnetita/química , Neoplasias/imunologia , Neoplasias/terapia , Ovalbumina/química , Dióxido de Silício/química , Animais , Antígenos/administração & dosagem , Galinhas , Citocinas/biossíntese , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Feminino , Imunidade , Nanopartículas de Magnetita/toxicidade , Camundongos Endogâmicos C57BL
15.
Int J Pharm ; 572: 118777, 2019 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-31678377

RESUMO

En masse vaccination is a promising strategy for combatting infectious diseases. Intranasal vaccination is a viable route of mass vaccination, and it could be performed easily via needle-free administration. However, it is not widely used because it tends not to evoke sufficient immunity. The aim of the present study was to improve the performance of intranasal vaccination by extending the amount of time that administered antigens remain in the nasal cavity, and enhancing immune responses via a nanocarrier-based adjuvant. A simple and safe solid-in-oil (S/O) system was investigated as a nanocarrier in intranasal vaccination. S/O nanodispersions are oil-based dispersions of antigens coated with surfactants. Because of the mucoadhesive capacities of surfactant and oil they have high potential to extend the amount of time that administered antigens remain in the nasal cavity, and can induce strong immune responses due to a nanocarrier-based adjuvant effect. In nasal absorption experiments antigens administered intranasally via S/O nanodispersions remained in the nasal cavity longer and induced strong mucosal and systemic immune responses. Histopathology analysis indicated that S/O nanodispersions did not modify the nasal epithelium or cilia, suggesting non-toxicity of the carrier. These results indicate the potential of intranasal vaccination using S/O nanodispersions for future vaccination.


Assuntos
Adjuvantes Imunológicos/química , Imunidade nas Mucosas/imunologia , Nanopartículas/química , Mucosa Nasal/imunologia , Óleos/química , Tensoativos/química , Adjuvantes Imunológicos/administração & dosagem , Administração Intranasal/métodos , Animais , Antígenos/administração & dosagem , Antígenos/química , Antígenos/imunologia , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Vacinação/métodos
16.
Biomaterials ; 225: 119518, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31586864

RESUMO

Tumor-cell derived small extracellular vesicle (sEV) combined with immunostimulatory adjuvants may serve as a promising tumor vaccine through the induction of the cytotoxic T cell response. To achieve an efficient immune response, the prolonged tissue residence after intradermal injection followed by the sustained and efficient delivery of tumor-cell derived sEV combined with adjuvants to antigen-presenting cells (APCs) is a promising strategy. In the present study, we constructed a DNA-anchored sEV superstructure in which tumor-cell derived sEVs were assembled with each other to achieve prolonged tissue residence and the ability to encourage selective uptake by dendritic cells. We prepared sEVs modified with immunostimulatory CpG-DNA containing an additional "sticky end" (CpG-sEV). CpG-sEVs were mixed with an oligonucleotide duplex containing the sequence complementary to the "sticky end" of the CpG-DNA, resulting in the self-assembly of CpG-sEV into a micrometer-sized superstructure. The CpG-DNA anchored sEV assembly (CpG-sEV assembly) was selectively taken up by APCs, compared to tumor cells or fibroblast cells, and it efficiently activated dendritic cells in vitro. Moreover, CpG-sEV assembly formation significantly prolonged tissue residence and increased the immune responses of immunostimulatory CpG-DNA intradermally injected into mice. These results indicate that CpG-sEV assembly is an effective system which may be useful for tumor immunotherapy.


Assuntos
Células Apresentadoras de Antígenos/metabolismo , Antígenos/administração & dosagem , DNA/metabolismo , Vesículas Extracelulares/metabolismo , Animais , Citocinas/metabolismo , Endocitose/efeitos dos fármacos , Imunomodulação/efeitos dos fármacos , Injeções Intradérmicas , Masculino , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos Endogâmicos C57BL , Oligodesoxirribonucleotídeos/farmacologia
17.
Biomater Sci ; 7(10): 4022-4026, 2019 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-31506651

RESUMO

Since current subunit vaccines are limited by a short half-life in vivo and weak immune responses when used alone without adjuvants, there is an unmet need for combing carriers with complement activation signals to interrupt outbreaks in real-time. Amino-functionalized zirconium-based MOFs (UiO-AM) could activate the complement system, which plays an important role in innate and adaptive immunity. Our data provide design principles for studies on complement activation as a safe vaccine carrier that can effectively enhance immune responses against antigens in vivo.


Assuntos
Antígenos/administração & dosagem , Estruturas Metalorgânicas/administração & dosagem , Nanopartículas/administração & dosagem , Ovalbumina/administração & dosagem , Vacinas/administração & dosagem , Zircônio/administração & dosagem , Animais , Sobrevivência Celular/efeitos dos fármacos , Ativação do Complemento/efeitos dos fármacos , Citocinas/metabolismo , Células HeLa , Células Hep G2 , Humanos , Camundongos , Células RAW 264.7
18.
J Control Release ; 311-312: 50-64, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31465827

RESUMO

The current conventional injectable vaccines face several drawbacks such as inconvenience and ineffectiveness in mucosal immunization. Therefore, the current development of effective oral vaccines is vital to enable the generation of dual systemic and mucosal immunity. In the present study, we examine the potential of pH-responsive bacterial nanocellulose/polyacrylic acid (BNC/PAA) hydrogel microparticles (MPs) as an oral vaccine carrier. In-vitro entrapment efficiency and release study of Ovalbumin (Ova) demonstrated that as high as 72% of Ova were entrapped in the hydrogel, and the release of loaded Ova was pH-dependent. The released Ova remained structurally conserved as evident by Western blot and circular dichroism. Hydrogel MPs reduced the TEER measurement of HT29MTX/Caco2/Raji B triple co-culture monolayer by reversibly opening the tight junctions (TJs) as shown in the TEM images. The ligated ileal loop assay revealed that hydrogel MPs could facilitate the penetration of FITC-Ova into the Peyer's patches in small intestine. Ova and cholera toxin B (CTB) were utilized in in-vivo oral immunization as model antigen and mucosal adjuvant. The in-vivo immunization revealed mice orally administered with Ova and CTB-loaded hydrogel MPs generated significantly higher level of serum anti-Ova IgG and mucosal anti-Ova IgA in the intestinal washes, compared to intramuscular administrated Ova. These results conclude that BNC/PAA hydrogel MPs is a potential oral vaccine carrier for effective oral immunization.


Assuntos
Resinas Acrílicas/administração & dosagem , Antígenos/administração & dosagem , Celulose/administração & dosagem , Portadores de Fármacos/administração & dosagem , Hidrogéis/administração & dosagem , Imunização/métodos , Ovalbumina/administração & dosagem , Administração Oral , Animais , Antígenos/química , Linhagem Celular Tumoral , Portadores de Fármacos/química , Liberação Controlada de Fármacos , Feminino , Humanos , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Camundongos Endogâmicos BALB C , Ovalbumina/química , Junções Íntimas/metabolismo
19.
PLoS One ; 14(8): e0220382, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31386690

RESUMO

The skin is a very suitable organ for the induction of immune responses to vaccine antigens. Antigen delivery systems to the skin by needle and syringe directly deposit the antigen into the epidermal-dermal compartment, one of the most immunocompetent sites due to the presence of professional antigen-presenting cells aimed at the induction of antigen-specific T cells. In this study, we analyzed the amount of ovalbumin as an antigen delivered to the skin by a microneedle. When ovalbumin protein as an antigen was delivered to the skin of mice using a dissolving microneedle, it induced an immune response through the enhanced proliferation and cytokines production by the splenocytes and lymph nodes. Also, it effectively increased the ovalbumin-specific CD8+ T cell and CD4+ T cell population and induced an ovalbumin-specific CTL response against the graft of ovalbumin-expressing EG7 tumor cells in the immunized mice. Also, we identified the inhibition of tumor growth and prevention of tumor formation in the context of the therapeutic and prophylactic vaccine, respectively through EG-7 tumor mouse model. Finally, these data show the potential of patches as attractive antigen delivery vehicles.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Imunoterapia/métodos , Agulhas , Ovalbumina/administração & dosagem , Adesivo Transdérmico , Administração Cutânea , Animais , Antígenos/administração & dosagem , Antígenos/farmacologia , Antígenos/uso terapêutico , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD8-Positivos/citologia , Proliferação de Células , Sistemas de Liberação de Medicamentos/normas , Imunidade , Camundongos , Neoplasias/terapia , Ovalbumina/uso terapêutico , Linfócitos T Citotóxicos/citologia , Adesivo Transdérmico/normas , Resultado do Tratamento
20.
Int J Nanomedicine ; 14: 4867-4880, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31308663

RESUMO

Background: The demand for an effective vaccine delivery system that drives a suitable immune response is increasing. The oxidized carbon nanosphere (OCN), a negatively charged carbon nanoparticle, has the potential to fulfill this requirement because it can efficiently deliver macromolecules into cells and allows endosomal leakage. However, fundamental insights into how OCNs are taken up by antigen-presenting cells, and the intracellular behavior of delivered molecules is lacking. Furthermore, how immune responses are stimulated by OCN-mediated delivery has not been investigated. Purpose: In this study, the model protein antigen ovalbumin (OVA) was used to investigate the uptake mechanism and intracellular fate of OCN-mediated delivery of protein in macrophages. Moreover, the immune response triggered by OVA delivered by OCNs was characterized. Methods: Bone-marrow-derived macrophages (BMDMs) from mice were used to study antigen uptake and intracellular trafficking. Mice were immunized using OCN-OVA combined with known adjuvants, and the specific immune response was measured. Results: OCNs showed no cytotoxicity against BMDMs. OCN-mediated delivery of OVA into BMDMs was partially temperature independent process. Using specific inhibitors, it was revealed that intracellular delivery of OCN-OVA does not rely on phagocytosis or the clathrin- and lipid raft/caveolae-mediated pathways. Delivered OVA was found to colocalize with compartments containing MHC class I, but not with early endosomes, lysosomes, and autophagosomes. Immunization of OVA using OCNs in combination with the known adjuvant monophosphoryl lipid A specifically enhanced interferon gamma (IFNγ)- and granzyme B-producing cytotoxic T cells (CTLs). Conclusion: OCNs effectively delivered protein antigens into macrophages that localized with compartments containing MHC class I partially by the temperature independent, but not clathrin- and lipid raft/caveolae-mediated pathways. Increased CD8+ T-cell activity was induced by OCN-delivered antigens, suggesting antigen processing toward antigen presentation for CTLs. Taken together, OCNs are a potential protein antigen delivery system that stimulates the cell-mediated immune response.


Assuntos
Antígenos/administração & dosagem , Carbono/química , Sistemas de Liberação de Medicamentos , Imunidade Celular , Nanopartículas/química , Adjuvantes Imunológicos/farmacologia , Animais , Antígenos/imunologia , Autofagossomos/efeitos dos fármacos , Autofagossomos/metabolismo , Linhagem Celular , Endocitose/efeitos dos fármacos , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Feminino , Imunidade Celular/efeitos dos fármacos , Cinética , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos Endogâmicos BALB C , Nanopartículas/toxicidade , Oxirredução , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA