Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 162
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Allergol Int ; 73(2): 302-312, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37953104

RESUMO

BACKGROUND: Lactobacillus paracasei has been known to reduce airway resistance and inflammation in asthma. However, the therapeutic effect of its extracellular vesicles (EVs) in patients with asthma remains unclear. METHODS: To validate the clinical relevance of L. paracasei-derived EVs (LpEV) in asthma, the composition of gut microbial EVs was verified by metagenomics in LPS-induced C57BL/6 mice. The components of proteins and metabolites in LpEV were identified by peptide mass fingerprinting and metabolomic analysis. The serum levels of specific IgG1 or IgG4 antibodies to LpEV were compared by ELISA between patients with eosinophilic asthma (EA, n = 10) and those with neutrophilic asthma (NA, n = 10) as well as with healthy controls (HCs, n = 10). Finally, therapeutic effects of LpEV and their metabolites in asthma were validated in vivo/in vitro. RESULTS: Significantly lower proportions of EVs derived from Lactobacillus at the genus level were noted in mice with NA than in control mice. Moreover, the serum levels of LpEV-specific IgG4, but not IgG1, were lower in patients with NA than in those with EA or in HCs and positively correlated with FEV1 (%) values. In addition, oral administration of LpEV reduced airway resistance and inflammation in mice with NA. Finally, LpEV and their 3 metabolites (dodecanoic acid, palmitoleic acid, and D-(-)-tagatose) significantly inhibited JNK phosphorylation/IL-8 production in airway epithelium in vitro. CONCLUSIONS: These findings suggest that LpEV may have a therapeutic potential targeting NA by suppressing the JNK pathway and proinflammatory cytokine production in airway epithelium.


Assuntos
Asma , Vesículas Extracelulares , Lacticaseibacillus paracasei , Animais , Humanos , Camundongos , Asma/terapia , Modelos Animais de Doenças , Epitélio , Imunoglobulina G , Inflamação , Pulmão , Sistema de Sinalização das MAP Quinases , Camundongos Endogâmicos C57BL
2.
Psychiatry Clin Neurosci ; 77(12): 646-652, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37646189

RESUMO

AIM: The present study examined the microbiome abundance and composition of drug-naive or drug-free patients with obsessive-compulsive disorder (OCD) compared with healthy controls. In addition, in the OCD group, the microbiome composition was compared between early-onset and late-onset OCD. METHODS: Serum samples were collected from 89 patients with OCD and 107 age- and sex-matched healthy controls. Bacterial DNA was isolated from bacteria-derived extracellular vesicles in serum and then amplified and quantified using primers specific to the V3-V4 hypervariable region of the 16S ribosomal RNA gene. The 16S ribosomal DNA gene amplicon sequencing was performed. RESULTS: The pooled estimate showed that α-diversity was significantly reduced in patients with OCD compared with that in healthy controls (PShannon = 0.00015). In addition, a statistically significant difference was observed in ß-diversity between patients with OCD and healthy controls at the order (P = 0.012), family (P = 0.003), genus (P < 0.001), and species (P = 0.005) levels. In the microbiome composition, Pseudomonas, Caulobacteraceae (f), Streptococcus, Novosphingobium, and Enhydrobacter at the genus level were significantly less prevalent in patients with OCD than in controls. In addition, among patients with OCD, the microbial composition in the early-onset versus late-onset types was significantly different with respect to the genera Corynebacterium and Pelomonas. CONCLUSION: The present study showed an aberrant microbiome in patients with OCD, suggesting a role of the microbiota-brain interaction in the pathophysiology of OCD. Further longitudinal studies with larger sample sizes adjusting for various confounders are warranted.


Assuntos
Microbiota , Transtorno Obsessivo-Compulsivo , Humanos , Transtorno Obsessivo-Compulsivo/genética , Microbiota/genética , RNA Ribossômico 16S/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos
3.
Biochem Biophys Res Commun ; 523(3): 602-607, 2020 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-31941602

RESUMO

Scrub typhus is an acute vector-borne disease caused by infection with the intracellular gram-negative bacterium Orientia tsutsugamushi (Ot). The rapid production of an efficient vaccine against Ot using novel strategies is required because of the global increase in mortality caused by these infections; however, no commercial vaccine is currently available. Ot induces T-cell-mediated immunogenic responses upon infection; therefore, a new rapidly producible vaccine that maximizes T-cell responses against Ot is required. In this study, we sought to develop a model vaccine platform for T-cell-mediated Ot infection using T-cell-immunity associated Salmonella-derived extracellular vesicles (EVs). For this purpose, we optimized DNA sequences encoding the full-length Ot proteins, TSA56, ScaA, ScaC, ScaD, and ScaE, and their expression in Salmonella. The sequences were incorporated into a new platform vector, pKST, which ectopically and concurrently produces Ot proteins and EVs. Expression analysis using pKST-antigen plasmids showed that TSA56 and ScaC produced antigen-associated EVs and showed strong T-cell immunogenic responses. We found that mice vaccinated with EVs derived from TSA56-expressing cells were protected from Salmonella-induced mortality. Therefore, our findings showed that Salmonella EV-associated antigen is a model platform for T-cell immune response infections. Our system could help prepare EV-antigen vaccines against scrub typhus in an easy and rapid manner.


Assuntos
Antígenos de Bactérias/uso terapêutico , Vacinas Bacterianas/uso terapêutico , Vesículas Extracelulares/imunologia , Tifo por Ácaros/prevenção & controle , Animais , Antígenos de Bactérias/imunologia , Vacinas Bacterianas/imunologia , Humanos , Camundongos , Orientia tsutsugamushi/imunologia , Salmonella/imunologia , Infecções por Salmonella/imunologia , Infecções por Salmonella/prevenção & controle , Tifo por Ácaros/imunologia , Linfócitos T/imunologia
4.
Clin Exp Allergy ; 49(2): 227-238, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30312497

RESUMO

BACKGROUND: House dust mite (HDM) is the major source of indoor allergens that cause airway disease. Recent evidence suggests that Gram-negative/positive bacteria produce nano-sized extracellular vesicles (EVs) containing diverse components, including various immunostimulatory molecules. However, the association between bacteria-derived EVs and development of airway disease is unclear. OBJECTIVE: To identify and isolate HDM-derived EVs and to evaluate their effect on the development of airway inflammation. METHODS: Extracellular vesicles were isolated from crude HDM extracts by ultra-centrifugation, and their physical and immunological characteristics and roles in airway inflammation were tested in vitro and in murine models of airway inflammation. In addition, 16s metagenome analysis of nucleic acid from EVs was performed to identify their origin. RESULTS: Round, bilayered vesicles measuring 80-100 nanometres and containing abundant amounts of LPS were isolated. These vesicles induced innate immune responses both in vitro and in vivo. Intranasal exposure of naïve mice to HDM EVs induced production of cytokines associated with development of Th2-mediated and mixed (Th1-/Th2-/Th17-mediated) airway inflammation to allergen. Metagenome analysis identified Bacteroidetes and Proteobacteria as the probable sources of HDM EVs. CONCLUSION: House dust mite EVs originating from Gram-negative bacteria may play an important role on the development of airway inflammation.


Assuntos
Asma , Bacteroidetes , Vesículas Extracelulares , Proteobactérias , Pyroglyphidae , Linfócitos T Auxiliares-Indutores , Animais , Asma/metabolismo , Asma/microbiologia , Asma/patologia , Bacteroidetes/genética , Bacteroidetes/imunologia , Modelos Animais de Doenças , Vesículas Extracelulares/química , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/microbiologia , Metagenoma , Camundongos , Camundongos Knockout , Proteobactérias/genética , Proteobactérias/imunologia , Pyroglyphidae/química , Pyroglyphidae/imunologia , Pyroglyphidae/microbiologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/patologia
5.
Int Arch Allergy Immunol ; 178(2): 150-158, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30415264

RESUMO

BACKGROUND: Microbiota and human allergic airway diseases have been proven to be interrelated. Bacteria-derived extracellular vesicle (EV)s are known to play important roles in interbacterial and human-bacteria communications, but their relationship with allergies has not been examined yet. Urine EVs were investigated to determine whether they could be used as biomarkers for monitoring allergic airway diseases in children. METHODS: Subjects were 4 groups of chronic rhinitis (CR), allergic rhinitis (AR), atopic asthma (AS) and healthy controls. Single voided urine samples were collected. Urine EVs were isolated and their DNA was extracted for 16S-rDNA pyrosequencing. RESULTS: A total of 118 children participated in this study; 27, 39, 19, and 33 were in the CR, AR, AS, and control group, respectively. The AR had a significantly high Chao-1 index than that of controls. Principal component analysis revealed dysbiosis in the CR, AR, and AS compared to the controls. One phylum and 19 families and genera were significantly enriched or depleted in the disease groups compared to the controls; the Actinobacteria phylum and the Sphingomonadaceae family were more abundant in the AS and CR, the Comamonadaceae family, the Propionibacteraceae family, Propionibacterium and Enhydrobacter were more enriched in the CR, and the Methylobacteriaceae family and Methylobacterium were more abundant in each disease group, while the Enterobacteriaceae family was depleted in each disease group. CONCLUSIONS: CR, AR, and AS had a distinct composition of urine EVs. Urine EVs could be an indicator for assessing allergic airway diseases in children.


Assuntos
Bactérias/metabolismo , Bacteriúria/metabolismo , Bacteriúria/microbiologia , Vesículas Extracelulares/metabolismo , Hipersensibilidade Respiratória/etiologia , Hipersensibilidade Respiratória/metabolismo , Fatores Etários , Bactérias/classificação , Bactérias/genética , Criança , Pré-Escolar , Feminino , Humanos , Imunoglobulina E/sangue , Imunoglobulina E/imunologia , Masculino , Filogenia , RNA Ribossômico 16S/genética , Hipersensibilidade Respiratória/diagnóstico
6.
Gastroenterology ; 152(8): 1998-2010, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28246016

RESUMO

BACKGROUND & AIMS: Obesity and metabolic syndrome have been associated with alterations to the intestinal microbiota. However, few studies examined the effects of obesity on the intestinal immune system. We investigated changes in subsets of intestinal CD4+ T-helper (TH) cells with obesity and the effects of gut-tropic TH17 cells in mice on a high-fat diet (HFD). METHODS: We isolated immune cells from small intestine and adipose tissue of C57BL/6 mice fed a normal chow diet or a HFD for 10 weeks and analyzed the cells by flow cytometry. Mice fed a vitamin A-deficient HFD were compared with mice fed a vitamin A-sufficient HFD. Obese RAG1-deficient mice were given injections of only regulatory T cells or a combination of regulatory T cells and TH17 cells (wild type or deficient in integrin ß7 subunit or interleukin 17 [IL17]). Mice were examined for weight gain, fat mass, fatty liver, glucose tolerance, and insulin resistance. Fecal samples were collected before and after T cell transfer and analyzed for microbiota composition by metagenomic DNA sequencing and quantitative polymerase chain reaction. RESULTS: Mice placed on a HFD became obese, which affected the distribution of small intestinal CD4+ TH cells. Intestinal tissues from obese mice had significant reductions in the proportion of TH17 cells but increased proportion of TH1 cells, compared with intestinal tissues from nonobese mice. Depletion of vitamin A in obese mice further reduced the proportion of TH17 cells in small intestine; this reduction correlated with more weight gain and worsening of glucose intolerance and insulin resistance. Adoptive transfer of in vitro-differentiated gut-tropic TH17 cells to obese mice reduced these metabolic defects, which required the integrin ß7 subunit and IL17. Delivery of TH17 cells to intestines of mice led to expansion of commensal microbes associated with leanness. CONCLUSIONS: In mice, intestinal TH17 cells contribute to development of a microbiota that maintains metabolic homeostasis, via IL17. Gut-homing TH17 cells might be used to reduce metabolic disorders in obese individuals.


Assuntos
Transferência Adotiva , Imunidade nas Mucosas , Resistência à Insulina , Intestino Delgado/imunologia , Síndrome Metabólica/prevenção & controle , Obesidade/prevenção & controle , Células Th17/transplante , Animais , Células Cultivadas , Dieta Hiperlipídica , Modelos Animais de Doenças , Fezes/microbiologia , Microbioma Gastrointestinal/imunologia , Genótipo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Interações Hospedeiro-Patógeno , Cadeias beta de Integrinas/genética , Cadeias beta de Integrinas/metabolismo , Interleucina-17/deficiência , Interleucina-17/genética , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Masculino , Síndrome Metabólica/genética , Síndrome Metabólica/imunologia , Síndrome Metabólica/microbiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/genética , Obesidade/imunologia , Obesidade/microbiologia , Fenótipo , Células Th17/imunologia , Células Th17/microbiologia , Fatores de Tempo , Deficiência de Vitamina A/complicações
7.
J Immunol ; 194(7): 3361-8, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25716999

RESUMO

Recent evidence indicates that Gram-negative bacteria-derived extracellular vesicles (EVs) in indoor dust can evoke neutrophilic pulmonary inflammation, which is a key pathology of chronic obstructive pulmonary disease (COPD). Escherichia coli is a ubiquitous bacterium present in indoor dust and secretes nanometer-sized vesicles into the extracellular milieu. In the current study, we evaluated the role of E. coli-derived EVs on the development of COPD, such as emphysema. E. coli EVs were prepared by sequential ultrafiltration and ultracentrifugation. COPD phenotypes and immune responses were evaluated in C57BL/6 wild-type (WT), IFN-γ-deficient, or IL-17A-deficient mice after airway exposure to E. coli EVs. The present study showed that indoor dust from a bed mattress harbors E. coli EVs. Airway exposure to E. coli EVs increased the production of proinflammatory cytokines, such as TNF-α and IL-6. In addition, the repeated inhalation of E. coli EVs for 4 wk induced neutrophilic inflammation and emphysema, which are associated with enhanced elastase activity. Emphysema and elastase activity enhanced by E. coli EVs were reversed by the absence of IFN-γ or IL-17A genes. In addition, during the early period, lung inflammation is dependent on IL-17A and TNF-α, but not on IFN-γ, and also on TLR4. Moreover, the production of IFN-γ is eliminated by the absence of IL-17A, whereas IL-17A production is not abolished by IFN-γ absence. Taken together, the present data suggest that E. coli-derived EVs induce IL-17A-dependent neutrophilic inflammation and thereby emphysema, possibly via upregulation of elastase activity.


Assuntos
Micropartículas Derivadas de Células , Escherichia coli/metabolismo , Interleucina-17/metabolismo , Neutrófilos/metabolismo , Enfisema Pulmonar/metabolismo , Enfisema Pulmonar/microbiologia , Poluição do Ar em Ambientes Fechados , Animais , Micropartículas Derivadas de Células/imunologia , Micropartículas Derivadas de Células/metabolismo , Modelos Animais de Doenças , Poeira , Escherichia coli/imunologia , Espaço Extracelular , Interferon gama/metabolismo , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/metabolismo , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Camundongos , Camundongos Knockout , Neutrófilos/imunologia , Fenótipo , Doença Pulmonar Obstrutiva Crônica/imunologia , Doença Pulmonar Obstrutiva Crônica/metabolismo , Doença Pulmonar Obstrutiva Crônica/microbiologia , Receptores Toll-Like/metabolismo
8.
J Allergy Clin Immunol ; 137(2): 507-516.e8, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26433560

RESUMO

BACKGROUND: The incidence of food allergies has increased dramatically during the last decade. Recently, probiotics have been studied for the prevention and treatment of allergic disease. OBJECTIVE: We examined whether Bifidobacterium longum KACC 91563 and Enterococcus faecalis KACC 91532 have the capacity to suppress food allergies. METHODS: B longum KACC 91563 and E faecalis KACC 91532 were administered to BALB/c wild-type mice, in which food allergy was induced by using ovalbumin and alum. Food allergy symptoms and various immune responses were assessed. RESULTS: B longum KACC 91563, but not E faecalis KACC 91532, alleviated food allergy symptoms. Extracellular vesicles of B longum KACC 91563 bound specifically to mast cells and induced apoptosis without affecting T-cell immune responses. Furthermore, injection of family 5 extracellular solute-binding protein, a main component of extracellular vesicles, into mice markedly reduced the occurrence of diarrhea in a mouse food allergy model. CONCLUSION: B longum KACC 91563 induces apoptosis of mast cells specifically and alleviates food allergy symptoms. Accordingly, B longum KACC 91563 and family 5 extracellular solute-binding protein exhibit potential as therapeutic approaches for food allergies.


Assuntos
Proteínas de Bactérias/imunologia , Bifidobacterium/imunologia , Vesículas Extracelulares/imunologia , Hipersensibilidade Alimentar/imunologia , Hipersensibilidade Alimentar/microbiologia , Imunomodulação , Mastócitos/imunologia , Animais , Apoptose/imunologia , Proteínas de Bactérias/metabolismo , Bifidobacterium/metabolismo , Contagem de Células , Citocinas/biossíntese , Modelos Animais de Doenças , Endocitose/imunologia , Vesículas Extracelulares/metabolismo , Hipersensibilidade Alimentar/metabolismo , Intestino Delgado/imunologia , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Intestino Delgado/patologia , Mastócitos/metabolismo , Mastócitos/microbiologia , Camundongos , Probióticos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
9.
Bioinformatics ; 31(6): 933-9, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25388151

RESUMO

MOTIVATION: Extracellular vesicles (EVs) are spherical bilayered proteolipids, harboring various bioactive molecules. Due to the complexity of the vesicular nomenclatures and components, online searches for EV-related publications and vesicular components are currently challenging. RESULTS: We present an improved version of EVpedia, a public database for EVs research. This community web portal contains a database of publications and vesicular components, identification of orthologous vesicular components, bioinformatic tools and a personalized function. EVpedia includes 6879 publications, 172 080 vesicular components from 263 high-throughput datasets, and has been accessed more than 65 000 times from more than 750 cities. In addition, about 350 members from 73 international research groups have participated in developing EVpedia. This free web-based database might serve as a useful resource to stimulate the emerging field of EV research. AVAILABILITY AND IMPLEMENTATION: The web site was implemented in PHP, Java, MySQL and Apache, and is freely available at http://evpedia.info.


Assuntos
Biologia Computacional , Sistemas de Gerenciamento de Base de Dados , Bases de Dados Factuais , Exossomos/metabolismo , Espaço Extracelular/metabolismo , Software , Pesquisa Biomédica , Humanos , Interface Usuário-Computador
10.
Mass Spectrom Rev ; 34(4): 474-90, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-24421117

RESUMO

Almost all bacteria, archaea, and eukaryotic cells shed extracellular vesicles either constitutively or in a regulated manner. These nanosized membrane vesicles are spherical, bilayered proteolipids that harbor specific subsets of proteins, DNAs, RNAs, and lipids. Recent research has facilitated conceptual advancements in this emerging field that indicate that extracellular vesicles act as intercellular communicasomes by transferring signals to their target cell via surface ligands and delivering receptors and functional molecules. Recent progress in mass spectrometry-based proteomic analyses of mammalian extracellular vesicles derived from diverse cell types and body fluids has resulted in the identification of several thousand vesicular proteins that provide us with essential clues to the molecular mechanisms involved in vesicle cargo sorting and biogenesis. Furthermore, cell-type- or disease-specific vesicular proteins help us to understand the pathophysiological functions of extracellular vesicles and contribute to the discovery of diagnostic and therapeutic target proteins. This review focuses on the high-throughput mass spectrometry-based proteomic analyses of mammalian extracellular vesicles (i.e., exosomes and ectosomes), EVpedia (a free web-based integrated database of high-throughput data for systematic analyses of extracellular vesicles; http://evpedia.info), and the intravesicular protein-protein interaction network analyses of mammalian extracellular vesicles. The goal of this article is to encourage further studies to construct a comprehensive proteome database for extracellular vesicles that will help us to not only decode the biogenesis and cargo-sorting mechanisms during vesicle formation but also elucidate the pathophysiological roles of these complex extracellular organelles.


Assuntos
Micropartículas Derivadas de Células/fisiologia , Exossomos/fisiologia , Animais , Evolução Biológica , Bases de Dados Factuais , Humanos , Espectrometria de Massas/métodos , Mapas de Interação de Proteínas , Proteômica/métodos
11.
Cytokine ; 79: 23-30, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26717421

RESUMO

BACKGROUND: TNF-α has been postulated to be a critical mediator contributing to airway inflammation. The purpose of this study was to evaluate the role of TNF-α in the induction of Th17 and Th2 cells related to asthma pathogenesis. OBJECTIVE: To evaluate detailed mechanisms for the modulation of IL-23 by TNF-α in sensitization period. METHODS: During sensitization period, 10µg of rat anti-mouse TNF-α mAb was intravenously administrated one hour before the application of OVA and 0.1µg of LPS. To see the relation between TNF-α and associated effectors cytokine, we replenished TNF-α KO mice with IL-23 during sensitization period. To assess cellular resources, CD11c+ cells isolated from lung tissue after sensitization were treated with anti-TNF-α Ab. RESULTS: Treatment of anti-TNF-α mAb during sensitization period significantly reduced airway eosinophilia, serum OVA-specific IgE levels and methacholine AHR compared to isotype Ab. Anti-TNF-α mAb treated mice showed significant reduction in the levels of IL-23 after sensitization in bronchoalveolar lavage fluid (BALF) as well as IL-17A, IL-4 levels in BALF after challenge compared with isotype Ab treated mice. Supplementation of IL-23 in TNF-α KO mice resulted in complete restoration of eosinophilic airway inflammation, AHR, and IL-17A and IL-4 expression in CD4+ T cells. Anti-TNF-α mAb treatment after sensitization significantly diminished the population of IL-23p19-secreting CD11c+ cells in lung. CONCLUSION: TNF-α plays an important role in the development of airway inflammation by enhancing IL-23/Th17 and Th2 immune responses.


Assuntos
Asma/imunologia , Eosinofilia/imunologia , Subunidade p19 da Interleucina-23/imunologia , Células Th17/imunologia , Células Th2/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Anticorpos Monoclonais/imunologia , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/imunologia , Antígeno CD11c/metabolismo , Linfócitos T CD4-Positivos/imunologia , Células Cultivadas , Eosinófilos/imunologia , Feminino , Imunoglobulina E/sangue , Interleucina-17/imunologia , Subunidade p19 da Interleucina-23/metabolismo , Subunidade p19 da Interleucina-23/farmacologia , Interleucina-4/imunologia , Pulmão/imunologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovalbumina/imunologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/genética
13.
Nano Lett ; 15(1): 266-74, 2015 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-25506626

RESUMO

The notion that widespread infectious diseases could be best managed by developing potent, adjuvant-free vaccines has resulted in the use of various biological immune-stimulating components as new vaccine candidates. Recently, extracellular vesicles, also known as exosomes and microvesicles in mammalian cells and outer membrane vesicles in Gram-negative bacteria, have gained attention for the next generation vaccine. However, the more invasive and effective the vaccine is in delivery, the more risk it holds for severe immune toxicity. Here, in optimizing the current vaccine delivery system, we designed bacterial protoplast-derived nanovesicles (PDNVs), depleted of toxic outer membrane components to generate a universal adjuvant-free vaccine delivery system. These PDNVs exhibited significantly higher productivity and safety than the currently used vaccine delivery vehicles and induced strong antigen-specific humoral and cellular immune responses. Moreover, immunization with PDNVs loaded with bacterial antigens conferred effective protection against bacterial sepsis in mice. These nonliving nanovesicles derived from bacterial protoplast open up a new avenue for the creation of next generation, adjuvant-free, less toxic vaccines to be used to prevent infectious diseases.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Escherichia coli , Nanopartículas/química , Protoplastos , Infecções Estafilocócicas/prevenção & controle , Vacinas Antiestafilocócicas , Staphylococcus aureus , Animais , Escherichia coli/química , Escherichia coli/genética , Escherichia coli/imunologia , Camundongos , Protoplastos/química , Protoplastos/imunologia , Infecções Estafilocócicas/imunologia , Vacinas Antiestafilocócicas/química , Vacinas Antiestafilocócicas/genética , Vacinas Antiestafilocócicas/imunologia , Staphylococcus aureus/química , Staphylococcus aureus/genética , Staphylococcus aureus/imunologia
14.
Small ; 11(4): 456-61, 2015 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-25196673

RESUMO

Evaluation of kinetic distribution and behaviors of nanoparticles in vivo provides crucial clues into their roles in living organisms. Extracellular vesicles are evolutionary conserved nanoparticles, known to play important biological functions in intercellular, inter-species, and inter-kingdom communication. In this study, the first kinetic analysis of the biodistribution of outer membrane vesicles (OMVs)-bacterial extracellular vesicles-with immune-modulatory functions is performed. OMVs, injected intraperitoneally, spread to the whole mouse body and accumulate in the liver, lung, spleen, and kidney within 3 h of administration. As an early systemic inflammation response, increased levels of TNF-α and IL-6 are observed in serum and bronchoalveolar lavage fluid. In addition, the number of leukocytes and platelets in the blood is decreased. OMVs and cytokine concentrations, as well as body temperature are gradually decreased 6 h after OMV injection, in concomitance with the formation of eye exudates, and of an increase in ICAM-1 levels in the lung. Following OMV elimination, most of the inflammatory signs are reverted, 12 h post-injection. However, leukocytes in bronchoalveolar lavage fluid are increased as a late reaction. Taken together, these results suggest that OMVs are effective mediators of long distance communication in vivo.


Assuntos
Membrana Celular/metabolismo , Escherichia coli/metabolismo , Exossomos/metabolismo , Nanopartículas/química , Tamanho da Partícula , Animais , Líquidos Corporais/metabolismo , Injeções Intraperitoneais , Cinética , Camundongos Endogâmicos C57BL , Espectroscopia de Luz Próxima ao Infravermelho , Distribuição Tecidual
15.
Plant Biotechnol J ; 13(1): 62-72, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25065685

RESUMO

Pandemics in poultry caused by the highly pathogenic avian influenza (HPAI) A virus occur too frequently globally, and there is growing concern about the HPAI A virus due to the possibility of a pandemic among humans. Thus, it is important to develop a vaccine against HPAI suitable for both humans and animals. Various approaches are underway to develop such vaccines. In particular, an edible vaccine would be a convenient way to vaccinate poultry because of the behaviour of the animals. However, an edible vaccine is still not available. In this study, we developed a strategy of effective vaccination of mice by the oral administration of transgenic Arabidopsis plants (HA-TG) expressing haemagglutinin (HA) in the endoplasmic reticulum (ER). Expression of HA in the ER resulted in its high-level accumulation, N-glycosylation, protection from proteolytic degradation and long-term stability. Oral administration of HA-TG with saponin elicited high levels of HA-specific systemic IgG and mucosal IgA responses in mice, which resulted in protection against a lethal influenza virus infection with attenuated inflammatory symptoms. Based on these results, we propose that oral administration of freeze-dried leaf powders from transgenic plants expressing HA in the ER together with saponin is an attractive strategy for vaccination against influenza A virus.


Assuntos
Adjuvantes Imunológicos/farmacologia , Retículo Endoplasmático/metabolismo , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Virus da Influenza A Subtipo H5N1/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Saponinas/imunologia , Vacinação , Administração Oral , Animais , Formação de Anticorpos/efeitos dos fármacos , Formação de Anticorpos/imunologia , Especificidade de Anticorpos/efeitos dos fármacos , Especificidade de Anticorpos/imunologia , Antígenos Virais/imunologia , Arabidopsis/genética , Relação Dose-Resposta Imunológica , Feminino , Imunidade Humoral/efeitos dos fármacos , Imunidade nas Mucosas/efeitos dos fármacos , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , Plantas Geneticamente Modificadas , Pneumonia/imunologia , Pneumonia/patologia , Pneumonia/prevenção & controle , Pneumonia/virologia , Proteínas Recombinantes de Fusão/metabolismo
16.
J Immunol ; 190(8): 4092-102, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23514742

RESUMO

Outer membrane vesicles (OMVs), secreted from Gram-negative bacteria, are spherical nanometer-sized proteolipids enriched with outer membrane proteins. OMVs, also known as extracellular vesicles, have gained interests for use as nonliving complex vaccines and have been examined for immune-stimulating effects. However, the detailed mechanism on how OMVs elicit the vaccination effect has not been studied extensively. In this study, we investigated the immunological mechanism governing the protective immune response of OMV vaccines. Immunization with Escherichia coli-derived OMVs prevented bacteria-induced lethality and OMV-induced systemic inflammatory response syndrome. As verified by adoptive transfer and gene-knockout studies, the protective effect of OMV immunization was found to be primarily by the stimulation of T cell immunity rather than B cell immunity, especially by the OMV-Ag-specific production of IFN-γ and IL-17 from T cells. By testing the bacteria-killing ability of macrophages, we also demonstrated that IFN-γ and IL-17 production is the main factor promoting bacterial clearances. Our findings reveal that E. coli-derived OMV immunization effectively protects bacteria-induced lethality and OMV-induced systemic inflammatory response syndrome primarily via Th1 and Th17 cell responses. This study therefore provides a new perspective on the immunological detail regarding OMV vaccination.


Assuntos
Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/mortalidade , Vacinas contra Escherichia coli/administração & dosagem , Vacinas contra Escherichia coli/imunologia , Exossomos/imunologia , Células Th1/imunologia , Células Th17/imunologia , Imunidade Adaptativa , Animais , Membrana Celular/imunologia , Membrana Celular/microbiologia , Células Cultivadas , Infecções por Escherichia coli/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Sepse/imunologia , Sepse/microbiologia , Sepse/prevenção & controle , Síndrome de Resposta Inflamatória Sistêmica/imunologia , Síndrome de Resposta Inflamatória Sistêmica/microbiologia , Síndrome de Resposta Inflamatória Sistêmica/patologia , Células Th1/microbiologia , Células Th1/patologia , Células Th17/microbiologia , Células Th17/patologia
17.
J Immunol ; 188(4): 1799-808, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22262660

RESUMO

Formyl peptide receptors (FPRs) are chemoattractant receptors that mediate inflammatory cell responses to infection. Recent evidence indicates that noneosinophilic asthma phenotypes can be developed by both Th1 and Th17 cell responses when exposed to LPS-containing allergens. In this study, we evaluated the effects of airway activation of FPRs by their synthetic agonist, Trp-Lys-Tyr-Met-Val-D-Met (W-peptide), on the development of Th1 and Th17 cell responses in a noneosinophilic asthma mouse model. A noneosinophilic asthma mouse model was generated by intranasal sensitization with 10 µg of LPS plus 75 µg of OVA on days 0, 1, 2, and 7. Mice were then challenged with 50 µg of OVA alone on days 14, 15, 21, and 22. W-peptide was administered during the sensitization period, and immune and inflammatory responses were evaluated after OVA challenge. Lung inflammation after OVA challenge was partly abolished by airway activation of FPRs during sensitization. Maturation of dendritic cells (DCs) and migration of DCs from the lung to lung-draining lymph nodes were inhibited by FPR activation. In addition, airway activation of FPRs inhibited allergen-specific T cell proliferation in the lymph nodes. Production of IL-12 and IL-6 (Th1- and Th17-polarizing cytokines) from lung DCs was decreased by airway activation of FPRs. This effect resulted in the inhibition of allergen-specific Th1 and Th17 cell responses. Airway activation of FPRs during sensitization effectively prevents the development of Th1 and Th17 cell responses induced by LPS-containing allergens via multiple mechanisms, such as inhibition of DC maturation and migration and the production of Th1- and Th7-polarizing cytokines.


Assuntos
Asma/imunologia , Células Dendríticas/imunologia , Pulmão/imunologia , Receptores de Formil Peptídeo/metabolismo , Células Th1/imunologia , Células Th17/imunologia , Animais , Asma/metabolismo , Asma/patologia , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Modelos Animais de Doenças , Interleucina-12/biossíntese , Interleucina-6/biossíntese , Pulmão/metabolismo , Pulmão/patologia , Linfonodos/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Oligopeptídeos/administração & dosagem , Oligopeptídeos/farmacologia , Ovalbumina/imunologia , Receptores de Formil Peptídeo/biossíntese , Células Th1/efeitos dos fármacos , Células Th17/efeitos dos fármacos
18.
Proteomics ; 13(10-11): 1554-71, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23401200

RESUMO

Mammalian cells secrete two types of extracellular vesicles either constitutively or in a regulated manner: exosomes (50-100 nm in diameter) released from the intracellular compartment and ectosomes (also called microvesicles, 100-1000 nm in diameter) shed directly from the plasma membrane. Extracellular vesicles are bilayered proteolipids enriched with proteins, mRNAs, microRNAs, and lipids. In recent years, much data have been collected regarding the specific components of extracellular vesicles from various cell types and body fluids using proteomic, transcriptomic, and lipidomic methods. These studies have revealed that extracellular vesicles harbor specific types of proteins, mRNAs, miRNAs, and lipids rather than random cellular components. These results provide valuable information on the molecular mechanisms involved in vesicular cargo-sorting and biogenesis. Furthermore, studies of these complex extracellular organelles have facilitated conceptual advancements in the field of intercellular communication under physiological and pathological conditions as well as for disease-specific biomarker discovery. This review focuses on the proteomic, transcriptomic, and lipidomic profiles of extracellular vesicles, and will briefly summarize recent advances in the biology, function, and diagnostic potential of vesicle-specific components.


Assuntos
Micropartículas Derivadas de Células/metabolismo , Exossomos/metabolismo , Metabolismo dos Lipídeos , Proteoma/metabolismo , Transcriptoma , Animais , Comunicação Celular , Perfilação da Expressão Gênica , Humanos , Mapeamento de Interação de Proteínas , Mapas de Interação de Proteínas , Proteômica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
19.
Am J Respir Cell Mol Biol ; 49(4): 637-45, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23713467

RESUMO

Pseudomonas aeruginosa is often involved in lung diseases such as cystic fibrosis. These bacteria can release outer membrane vesicles (OMVs), which are bilayered proteolipids with diameters of approximately 20 to 250 nm. In vitro, these OMVs activate macrophages and airway epithelial cells. The aim of this study was to determine whether OMVs from P. aeruginosa can induce pulmonary inflammation in vivo and to elucidate the mechanisms involved. Bacteria-free OMVs were isolated from P. aeruginosa cultures. Wild-type, Toll-like receptor (TLR)2 and TLR4 knockout mice were exposed to OMVs by the airway, and inflammation in the lung was assessed using differential counts, histology, and quantification of chemokines and cytokines. The involvement of the TLR2 and TLR4 pathways was studied in human cells using transfection. OMVs given to the mouse lung caused dose- and time-dependent pulmonary cellular inflammation. Furthermore, OMVs increased concentrations of several chemokines and cytokines in the mouse lungs and mouse alveolar macrophages. The inflammatory responses to OMVs were comparable to those of live bacteria and were only partly regulated by the TLR2 and TLR4 pathways, according to studies in knockout mice. This study shows that OMVs from P. aeruginosa cause pulmonary inflammation without live bacteria in vivo. This effect is only partly controlled by TLR2 and TLR4. The role of OMVs in clinical disease warrants further studies because targeting of OMVs in addition to live bacteria may add clinical benefit compared with treating with antibiotics alone.


Assuntos
Pneumonia/microbiologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/metabolismo , Animais , Linhagem Celular , Quimiocinas/metabolismo , Humanos , Pulmão/metabolismo , Pulmão/microbiologia , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia/metabolismo , Infecções por Pseudomonas/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Transfecção
20.
Antimicrob Agents Chemother ; 57(6): 2589-95, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23529736

RESUMO

Gram-positive bacteria naturally produce extracellular vesicles. However, little is known regarding the functions of Gram-positive bacterial extracellular vesicles, especially in the bacterial community. Here, we investigated the role of Staphylococcus aureus extracellular vesicles in interbacterial communication to cope with antibiotic stress. We found that S. aureus liberated BlaZ, a ß-lactamase protein, via extracellular vesicles. These extracellular vesicles enabled other ampicillin-susceptible Gram-negative and Gram-positive bacteria to survive in the presence of ampicillin. However, S. aureus extracellular vesicles did not mediate the survival of tetracycline-, chloramphenicol-, or kanamycin-susceptible bacteria. Moreover, S. aureus extracellular vesicles did not contain the blaZ gene. In addition, the heat-treated S. aureus extracellular vesicles did not mediate the survival of ampicillin-susceptible bacteria. The ß-lactamase activities of S. aureus soluble and extracellular vesicle-associated BlaZ were similar, but only the extracellular vesicle-associated BlaZ was resistant to protease digestion, which suggests that the enzymatic activity of BlaZ in extracellular vesicles is largely protected by the vesicle structure. Our observations provide evidence of the important role of S. aureus extracellular vesicles in antibiotic resistance, which allows the polymicrobial community to continue to evolve and prosper against antibiotics.


Assuntos
Antibacterianos/farmacologia , Farmacorresistência Bacteriana , Escherichia coli/efeitos dos fármacos , Espaço Extracelular/enzimologia , Proteolipídeos/metabolismo , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/metabolismo , beta-Lactamases/metabolismo , Ampicilina/farmacologia , Escherichia coli/metabolismo , Humanos , Testes de Sensibilidade Microbiana , Staphylococcus aureus/genética , Staphylococcus aureus/ultraestrutura , beta-Lactamases/genética , beta-Lactamases/isolamento & purificação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA