Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Nanobiotechnology ; 21(1): 156, 2023 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-37208676

RESUMEN

The development of vaccines based on outer membrane vesicles (OMV) that naturally bud off from bacteria is an evolving field in infectious diseases. However, the inherent inflammatory nature of OMV limits their use as human vaccines. This study employed an engineered vesicle technology to develop synthetic bacterial vesicles (SyBV) that activate the immune system without the severe immunotoxicity of OMV. SyBV were generated from bacterial membranes through treatment with detergent and ionic stress. SyBV induced less inflammatory responses in macrophages and in mice compared to natural OMV. Immunization with SyBV or OMV induced comparable antigen-specific adaptive immunity. Specifically, immunization with Pseudomonas aeruginosa-derived SyBV protected mice against bacterial challenge, and this was accompanied by significant reduction in lung cell infiltration and inflammatory cytokines. Further, immunization with Escherichia coli-derived SyBV protected mice against E. coli sepsis, comparable to OMV-immunized group. The protective activity of SyBV was driven by the stimulation of B-cell and T-cell immunity. Also, SyBV were engineered to display the SARS-CoV-2 S1 protein on their surface, and these vesicles induced specific S1 protein antibody and T-cell responses. Collectively, these results demonstrate that SyBV may be a safe and efficient vaccine platform for the prevention of bacterial and viral infections.


Asunto(s)
Bacteriemia , COVID-19 , Infecciones por Escherichia coli , Vacunas , Ratones , Animales , Humanos , SARS-CoV-2 , Escherichia coli , COVID-19/prevención & control , Bacterias , Infecciones por Escherichia coli/prevención & control , Proteínas de la Membrana Bacteriana Externa , Anticuerpos Antibacterianos
2.
Respir Res ; 24(1): 3, 2023 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-36604658

RESUMEN

BACKGROUND: Allergic asthma is associated with airflow obstruction and hyper-responsiveness that arises from airway inflammation and remodeling. Cell therapy with mesenchymal stem cells (MSC) has been shown to attenuate inflammation in asthma models, and similar effects have recently been observed using extracellular vesicles (EV) obtained from these cells. Biologically functional vesicles can also be artificially generated from MSC by extruding cells through membranes to produce EV-mimetic nanovesicles (NV). In this study, we aimed to determine the effects of different MSC-derived vesicles in a murine model of allergic airway inflammation. METHODS: EV were obtained through sequential centrifugation of serum-free media conditioned by human bone marrow MSC for 24 h. NV were produced through serial extrusion of the whole cells through filters. Both types of vesicles underwent density gradient purification and were quantified through nanoparticle tracking analysis. C57BL/6 mice were sensitized to ovalbumin (OVA, 8 µg), and then randomly divided into the OVA group (intranasally exposed to 100 µg OVA for 5 days) and control group (exposed to PBS). The mice were then further divided into groups that received 2 × 109 EV or NV (intranasally or intraperitoneally) or PBS immediately following the first OVA exposure. RESULTS: Administration of EV and NV reduced cellularity and eosinophilia in bronchoalveolar lavage (BAL) fluid in OVA-sensitized and OVA-exposed mice. In addition, NV treatment resulted in decreased numbers of inflammatory cells within the lung tissue, and this was associated with lower levels of Eotaxin-2 in both BAL fluid and lung tissue. Furthermore, both intranasal and systemic administration of NV were effective in reducing inflammatory cells; however, systemic delivery resulted in a greater reduction of eosinophilia in the lung tissue. CONCLUSIONS: Taken together, our results indicate that MSC-derived NV significantly reduce OVA-induced allergic airway inflammation to a level comparable to EV. Thus, cell-derived NV may be a novel EV-mimetic therapeutic candidate for treating allergic diseases such as asthma.


Asunto(s)
Asma , Eosinofilia , Células Madre Mesenquimatosas , Humanos , Animales , Ratones , Modelos Animales de Enfermedad , Inmunoglobulina E , Ratones Endogámicos C57BL , Asma/terapia , Asma/tratamiento farmacológico , Pulmón , Líquido del Lavado Bronquioalveolar , Inflamación , Ovalbúmina/toxicidad , Ratones Endogámicos BALB C
3.
J Nanobiotechnology ; 20(1): 451, 2022 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-36243859

RESUMEN

Mesenchymal stem cells (MSC) secrete extracellular vesicles (EV) with a regenerative profile, and an increasing number of studies have focused on the utilization of MSC-EV for therapeutic drug delivery. However, EV are usually produced by cells in low quantities and are packed with numerous cytoplasmic components, which may be unfavorable for further drug loading. In this study, we developed a simple process for generating membrane vesicles directly from the cells, which we refer to as synthetic eukaryotic vesicles (SyEV). We hypothesized that MSC-derived SyEV can be efficiently loaded with an anti-inflammatory drug and the loaded vesicles can strongly suppress the systemic inflammation induced by bacterial outer membrane vesicles (OMV). SyEV were generated from MSC membranes through serial extrusion of the cells, ionic stress, and subsequent vesiculation of the membrane sheets, leading to high yield and purity of the SyEV with few cytosolic components remaining. When these SyEV were given to macrophages or mice exposed to OMV, the release of pro-inflammatory cytokines was similarly attenuated comparable to treatment with natural EV. We then loaded the SyEV with large numbers of peptides targeting Myd88 and observed enhanced therapeutic potential of the loaded vesicles in OMV-induced macrophages. Further, in vivo experiments showed that the peptide-encapsulated MSC-SyEV suppressed cytokine production synergistically. Taken together, these findings suggest that SyEV-based therapeutics is a highly interesting platform for delivering an advanced therapeutic drug for the treatment of systemic inflammation without severe side effects.


Asunto(s)
Vesículas Extracelulares , Células Madre Mesenquimatosas , Animales , Antiinflamatorios , Citocinas/metabolismo , Modelos Animales de Enfermedad , Vesículas Extracelulares/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Factor 88 de Diferenciación Mieloide/metabolismo
4.
J Extracell Vesicles ; 10(9): e12120, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34262675

RESUMEN

Bacterial outer membrane vesicles (OMV) have gained attention as a promising new cancer vaccine platform for efficiently provoking immune responses. However, OMV induce severe toxicity by activating the innate immune system. In this study, we applied a simple isolation approach to produce artificial OMV that we have named Synthetic Bacterial Vesicles (SyBV) that do not induce a severe toxic response. We also explored the potential of SyBV as an immunotherapy combined with tumour extracellular vesicles to induce anti-tumour immunity. Bacterial SyBV were produced with high yield by a protocol including lysozyme and high pH treatment, resulting in pure vesicles with very few cytosolic components and no RNA or DNA. These SyBV did not cause systemic pro-inflammatory cytokine responses in mice compared to naturally released OMV. However, SyBV and OMV were similarly effective in activation of mouse bone marrow-derived dendritic cells. Co-immunization with SyBV and melanoma extracellular vesicles elicited tumour regression in melanoma-bearing mice through Th-1 type T cell immunity and balanced antibody production. Also, the immunotherapeutic effect of SyBV was synergistically enhanced by anti-PD-1 inhibitor. Moreover, SyBV displayed significantly greater adjuvant activity than other classical adjuvants. Taken together, these results demonstrate a safe and efficient strategy for eliciting specific anti-tumour responses using immunotherapeutic bacterial SyBV.


Asunto(s)
Membrana Externa Bacteriana/inmunología , Escherichia coli/inmunología , Vesículas Extracelulares/inmunología , Inmunoterapia , Melanoma Experimental/inmunología , Adyuvantes Inmunológicos/metabolismo , Animales , Células Artificiales/inmunología , Membrana Externa Bacteriana/metabolismo , Línea Celular Tumoral , Citocinas/metabolismo , Células Dendríticas , Vesículas Extracelulares/metabolismo , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunización , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Células TH1/inmunología
5.
Int J Mol Sci ; 22(9)2021 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-33946872

RESUMEN

Studying the proteomes of tissue-derived extracellular vesicles (EVs) can lead to the identification of biomarkers of disease and can provide a better understanding of cell-to-cell communication in both healthy and diseased tissue. The aim of this study was to apply our previously established tissue-derived EV isolation protocol to mouse lungs in order to determine the changes in the proteomes of lung tissue-derived EVs during allergen-induced eosinophilic airway inflammation. A mouse model for allergic airway inflammation was used by sensitizing the mice intraperitoneal with ovalbumin (OVA), and one week after the final sensitization, the mice were challenged intranasal with OVA or PBS. The animals were sacrificed 24 h after the final challenge, and their lungs were removed and sliced into smaller pieces that were incubated in culture media with DNase I and Collagenase D for 30 min at 37 °C. Vesicles were isolated from the medium by ultracentrifugation and bottom-loaded iodixanol density cushions, and the proteomes were determined using quantitative mass spectrometry. More EVs were present in the lungs of the OVA-challenged mice compared to the PBS-challenged control mice. In total, 4510 proteins were quantified in all samples. Among them, over 1000 proteins were significantly altered (fold change >2), with 614 proteins being increased and 425 proteins being decreased in the EVs from OVA-challenged mice compared to EVs from PBS-challenged animals. The associated cellular components and biological processes were analyzed for the altered EV proteins, and the proteins enriched during allergen-induced airway inflammation were mainly associated with gene ontology (GO) terms related to immune responses. In conclusion, EVs can be isolated from mouse lung tissue, and the EVs' proteomes undergo changes in response to allergen-induced airway inflammation. This suggests that the composition of lung-derived EVs is altered in diseases associated with inflammation of the lung, which may have implications in type-2 driven eosinophilic asthma pathogenesis.


Asunto(s)
Vesículas Extracelulares/inmunología , Pulmón/inmunología , Proteoma , Eosinofilia Pulmonar/inmunología , Hipersensibilidad Respiratoria/inmunología , Alérgenos/toxicidad , Animales , Asma , Líquido del Lavado Bronquioalveolar/citología , Modelos Animales de Enfermedad , Vesículas Extracelulares/metabolismo , Ontología de Genes , Pulmón/química , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Nanopartículas , Ovalbúmina/toxicidad , Eosinofilia Pulmonar/etiología , Eosinofilia Pulmonar/metabolismo , Hipersensibilidad Respiratoria/etiología , Hipersensibilidad Respiratoria/metabolismo
6.
Stem Cell Res Ther ; 10(1): 288, 2019 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-31547882

RESUMEN

After the initial investigations into applications of mesenchymal stem cells (MSCs) for cell therapy, there was increased interest in their secreted soluble factors. Following studies of MSCs and their secreted factors, extracellular vesicles (EVs) released from MSCs have emerged as a new mode of intercellular crosstalk. MSC-derived EVs have been identified as essential signaling mediators under both physiological and pathological conditions, and they appear to be responsible for many of the therapeutic effects of MSCs. In several in vitro and in vivo models, EVs have been observed to have supportive functions in modulating the immune system, mainly mediated by EV-associated proteins and nucleic acids. Moreover, stimulation of MSCs with biophysical or biochemical cues, including EVs from other cells, has been shown to influence the contents and biological activities of subsequent MSC-derived EVs. This review provides on overview of the contents of MSC-derived EVs in terms of their supportive effects, and it provides different perspectives on the manipulation of MSCs to improve the secretion of EVs and subsequent EV-mediated activities. In this review, we discuss the possibilities for manipulating MSCs for EV-based cell therapy and for using EVs to affect the expression of elements of interest in MSCs. In this way, we provide a clear perspective on the state of the art of EVs in cell therapy focusing on MSCs, and we raise pertinent questions and suggestions for knowledge gaps to be filled.


Asunto(s)
Vesículas Extracelulares/trasplante , Trasplante de Células Madre Mesenquimatosas/métodos , Animales , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Transducción de Señal
7.
Stem Cell Res Ther ; 10(1): 231, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31370884

RESUMEN

BACKGROUND: Sepsis remains a source of high mortality in hospitalized patients despite proper antibiotic approaches. Encouragingly, mesenchymal stromal cells (MSCs) and their produced extracellular vesicles (EVs) have been shown to elicit anti-inflammatory effects in multiple inflammatory conditions including sepsis. However, EVs are generally released from mammalian cells in relatively low amounts, and high-yield isolation of EVs is still challenging due to a complicated procedure. To get over these limitations, vesicles very similar to EVs can be produced by serial extrusions of cells, after which they are called nanovesicles (NVs). We hypothesized that MSC-derived NVs can attenuate the cytokine storm induced by bacterial outer membrane vesicles (OMVs) in mice, and we aimed to elucidate the mechanism involved. METHODS: NVs were produced from MSCs by the breakdown of cells through serial extrusions and were subsequently floated in a density gradient. Morphology and the number of NVs were analyzed by transmission electron microscopy and nanoparticle tracking analysis. Mice were intraperitoneally injected with Escherichia coli-derived OMVs to establish sepsis, and then injected with 2 × 109 NVs. Innate inflammation was assessed in peritoneal fluid and blood through investigation of infiltration of cells and cytokine production. The biodistribution of NVs labeled with Cy7 dye was analyzed using near-infrared imaging. RESULTS: Electron microscopy showed that NVs have a nanometer-size spherical shape and harbor classical EV marker proteins. In mice, NVs inhibited eye exudates and hypothermia, signs of a systemic cytokine storm, induced by intraperitoneal injection of OMVs. Moreover, NVs significantly suppressed cytokine release into the systemic circulation, as well as neutrophil and monocyte infiltration in the peritoneum. The protective effect of NVs was significantly reduced by prior treatment with anti-interleukin (IL)-10 monoclonal antibody. In biodistribution study, NVs spread to the whole mouse body and localized in the lung, liver, and kidney at 6 h. CONCLUSIONS: Taken together, these data indicate that MSC-derived NVs have beneficial effects in a mouse model of sepsis by upregulating the IL-10 production, suggesting that artificial NVs may be novel EV-mimetics clinically applicable to septic patients.


Asunto(s)
Membrana Externa Bacteriana/metabolismo , Exosomas/metabolismo , Vesículas Extracelulares/metabolismo , Interleucina-10/metabolismo , Nanoestructuras/química , Sepsis/prevención & control , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Modelos Animales de Enfermedad , Endocitosis , Escherichia coli/metabolismo , Vesículas Extracelulares/química , Interleucina-10/inmunología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Monocitos/citología , Monocitos/inmunología , Infiltración Neutrófila , Peritoneo/metabolismo , Peritoneo/patología , Proteoma/análisis , Células RAW 264.7 , Sepsis/patología , Distribución Tisular
8.
Infect Immun ; 87(9)2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31262980

RESUMEN

Pneumonia due to Gram-negative bacteria is associated with high mortality. Acinetobacter baumannii is a Gram-negative bacterium that is associated with hospital-acquired and ventilator-associated pneumonia. Bacteria have been described to release outer membrane vesicles (OMVs) that are capable of mediating systemic inflammation. The mechanism by which A. baumannii OMVs mediate inflammation is not fully defined. We sought to investigate the roles that Toll-like receptors (TLRs) play in A. baumannii OMV-mediated pulmonary inflammation. We isolated OMVs from A. baumannii cultures and intranasally introduced the OMVs into mice. Intranasal introduction of A. baumannii OMVs mediated pulmonary inflammation, which is associated with neutrophil recruitment and weight loss. In addition, A. baumannii OMVs increased the release of several chemokines and cytokines in the mouse lungs. The proinflammatory responses were partially inhibited in TLR2- and TLR4-deficient mice compared to those of wild-type mice. This study highlights the important roles of TLRs in A. baumannii OMV-induced pulmonary inflammation in vivo.


Asunto(s)
Infecciones por Acinetobacter/microbiología , Acinetobacter baumannii/fisiología , Neumonía/microbiología , Vesículas Secretoras/fisiología , Receptor Toll-Like 2/fisiología , Receptor Toll-Like 4/fisiología , Infecciones por Acinetobacter/metabolismo , Animales , Proteínas de la Membrana Bacteriana Externa , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ratones
9.
Front Microbiol ; 9: 2268, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30369908

RESUMEN

Outer membrane vesicles (OMVs) are spherical, proteolipid nanostructures that are constitutively released by Gram-negative bacteria including Escherichia coli. Although it has been shown that administration of E. coli OMVs stimulates a strong pulmonary inflammatory response with infiltration of neutrophils into the lungs in vivo, the mechanism of E. coli OMV-mediated neutrophil recruitment is poorly characterized. In this study, we observed significant infiltration of neutrophils into the mouse lung tissues in vivo, with increased expression of the neutrophil chemoattractant CXCL1, a murine functional homolog of human IL-8, on intraperitoneal administration of E. coli OMVs. In addition, OMVs and CD31-positive endothelial cells colocalized in the mouse lungs. Moreover, in vitro results showed that E. coli OMVs significantly increased IL-8 release from human microvascular endothelial cells and toll-like receptor (TLR)4 was found to be the main component for recognizing E. coli OMVs among human endothelial cell-associated TLRs. Furthermore, the transmigration of neutrophils was suppressed in the lung tissues obtained from TLR4 knockout mice treated with E. coli OMVs. Taken together, our data demonstrated that E. coli OMVs potently recruit neutrophils into the lung via the release of IL-8/CXCL1 from endothelial cells in TLR4- and NF-κB-dependent manners.

10.
Front Microbiol ; 9: 1735, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30131776

RESUMEN

Nano-sized extracellular vesicles (EVs), including exosomes, microvesicles, and other types of vesicles, are released by most mammalian cells and bacteria. We here ask whether feces contain EVs of mammalian and/or bacterial origin, and whether these EVs induce systemic inflammation. Fecal extracellular vesicles (fEVs) were isolated from mice and humans. The presence of EVs from Gram-negative and Gram-positive bacteria was detected by enzyme-linked immunosorbent assay using anti-lipid A and anti-lipoteichoic acid antibodies, whereas Western blot using anti-beta-actin antibody was employed to detect host-derived EVs in the fEVs. Further, fEVs were administered into mice by intraperitoneal injection, and inflammatory responses were investigated in the peritoneum, blood, and lungs. The role of TLR2 and TLR4 were studied using knockout mice. Significant quantities of EVs were present in feces from mice as well as humans, and derived from Gram-negative and Gram-positive bacteria, as well as the host. Bacteria-free fEVs introduced into the peritoneum induced local and systemic inflammation (including in the lungs), but fEVs from germ-free animals had weaker effects. This pronounced local and systemic inflammatory responses seemed to be induced by EVs from both Gram-negative and Gram-positive bacteria, and was attenuated in mice lacking TLR2 or TLR4. Our findings show that fEVs cause sepsis-like systemic inflammation, when introduced intraperitoneally, a process regulated by TLR2 and TLR4.

11.
Adv Healthc Mater ; 7(13): e1701476, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29683274

RESUMEN

Sepsis is characterized by systemic inflammatory response syndrome (SIRS) accompanied with infection. Gram-negative bacteria can evoke sepsis by activating the host immune system, such as the release of IL-6 and TNF-α, through their virulence factors. Outer membrane vesicles (OMVs), nanosized bilayered proteolipids derived from Gram-negative bacteria, harbor various virulence factors and are shown to induce SIRS. Here, drugs are repositioned to alleviate SIRS caused by Gram-negative bacterial OMVs. Using novel OMV-based drug screening systems, a total of 178 commercially available drugs are primarily screened, and a total of 18 repositioned drug candidates are found to effectively block IL-6 and TNF-α production from OMV-stimulated macrophages. After excluding the compounds which are previously known to intervene sepsis or which show cytotoxicity to macrophages, the compounds which show dose-dependency in inhibiting the release of IL-6 and TNF-α by the OMV-stimulated macrophages in vitro and which reduce OMV-induced SIRS in vivo are selected. Salbutamol, a ß2 adrenergic receptor agonist, is selected as a novel candidate to alleviate OMV-induced SIRS. This study sheds light on using Gram-negative bacterial OMVs in exploring novel candidate compounds to alleviate inflammatory diseases including sepsis.


Asunto(s)
Albuterol/farmacología , Reposicionamiento de Medicamentos , Bacterias Gramnegativas/patogenicidad , Síndrome de Respuesta Inflamatoria Sistémica/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Vesículas Extracelulares , Interleucina-6/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Nortriptilina/farmacología , Células RAW 264.7 , Síndrome de Respuesta Inflamatoria Sistémica/microbiología , Factor de Necrosis Tumoral alfa/genética
12.
Sci Rep ; 7(1): 17434, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29234030

RESUMEN

Sepsis induced cardiac dysfunction (SIC) is a severe complication to sepsis which significantly worsens patient outcomes. It is known that bacteria have the capacity to release outer membrane vesicles (OMVs), which are nano-sized bilayered vesicles composed of lipids and proteins, that can induce a fatal inflammatory response. The aim of this study was to determine whether OMVs from a uropathogenic Escherichia coli strain can induce cardiac dysfunction, and to elucidate any mechanisms involved. OMVs induced irregular Ca2+ oscillations with a decreased frequency in cardiomyocytes through recordings of intracellular Ca2+ dynamics. Mice were intraperitoneally injected with bacteria-free OMVs, which resulted in increased concentration of pro-inflammatory cytokine levels in blood. Cytokines were increased in heart lysates, and OMVs could be detected in the heart after OMVs injection. Troponin T was significantly increased in blood, and echocardiography showed increased heart wall thickness as well as increased heart rate. This study shows that E. coli OMVs induce cardiac injury in vitro and in vivo, in the absence of bacteria, and may be a causative microbial signal in SIC. The role of OMVs in clinical disease warrant further studies, as bacterial OMVs in addition to live bacteria may be good therapeutic targets to control sepsis.


Asunto(s)
Micropartículas Derivadas de Células/inmunología , Infecciones por Escherichia coli/inmunología , Escherichia coli/inmunología , Cardiopatías/inmunología , Sepsis/inmunología , Animales , Antibacterianos/farmacología , Proteínas de la Membrana Bacteriana Externa/metabolismo , Calcio/metabolismo , Línea Celular , Supervivencia Celular/fisiología , Micropartículas Derivadas de Células/efectos de los fármacos , Citocinas/metabolismo , Infecciones por Escherichia coli/diagnóstico por imagen , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/patología , Cardiopatías/diagnóstico por imagen , Cardiopatías/tratamiento farmacológico , Cardiopatías/patología , Lipopolisacáridos , Ratones Endogámicos C57BL , Miocardio/inmunología , Miocardio/patología , Polimixina B/farmacología , Distribución Aleatoria , Sepsis/diagnóstico por imagen , Sepsis/tratamiento farmacológico , Sepsis/patología , Troponina T/sangre
13.
Small ; 11(4): 456-61, 2015 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-25196673

RESUMEN

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.


Asunto(s)
Membrana Celular/metabolismo , Escherichia coli/metabolismo , Exosomas/metabolismo , Nanopartículas/química , Tamaño de la Partícula , Animales , Líquidos Corporales/metabolismo , Inyecciones Intraperitoneales , Cinética , Ratones Endogámicos C57BL , Espectroscopía Infrarroja Corta , Distribución Tisular
14.
Bioinformatics ; 31(6): 933-9, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25388151

RESUMEN

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.


Asunto(s)
Biología Computacional , Sistemas de Administración de Bases de Datos , Bases de Datos Factuales , Exosomas/metabolismo , Espacio Extracelular/metabolismo , Programas Informáticos , Investigación Biomédica , Humanos , Interfaz Usuario-Computador
15.
Nano Lett ; 15(1): 266-74, 2015 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-25506626

RESUMEN

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.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Escherichia coli , Nanopartículas/química , Protoplastos , Infecciones Estafilocócicas/prevención & control , Vacunas Estafilocócicas , Staphylococcus aureus , Animales , Escherichia coli/química , Escherichia coli/genética , Escherichia coli/inmunología , Ratones , Protoplastos/química , Protoplastos/inmunología , Infecciones Estafilocócicas/inmunología , Vacunas Estafilocócicas/química , Vacunas Estafilocócicas/genética , Vacunas Estafilocócicas/inmunología , Staphylococcus aureus/química , Staphylococcus aureus/genética , Staphylococcus aureus/inmunología
16.
Artículo en Inglés | MEDLINE | ID: mdl-24009897

RESUMEN

Secretion of extracellular vesicles is a general cellular activity that spans the range from simple unicellular organisms (e.g. archaea; Gram-positive and Gram-negative bacteria) to complex multicellular ones, suggesting that this extracellular vesicle-mediated communication is evolutionarily conserved. Extracellular vesicles are spherical bilayered proteolipids with a mean diameter of 20-1,000 nm, which are known to contain various bioactive molecules including proteins, lipids, and nucleic acids. Here, we present EVpedia, which is an integrated database of high-throughput datasets from prokaryotic and eukaryotic extracellular vesicles. EVpedia provides high-throughput datasets of vesicular components (proteins, mRNAs, miRNAs, and lipids) present on prokaryotic, non-mammalian eukaryotic, and mammalian extracellular vesicles. In addition, EVpedia also provides an array of tools, such as the search and browse of vesicular components, Gene Ontology enrichment analysis, network analysis of vesicular proteins and mRNAs, and a comparison of vesicular datasets by ortholog identification. Moreover, publications on extracellular vesicle studies are listed in the database. This free web-based database of EVpedia (http://evpedia.info) might serve as a fundamental repository to stimulate the advancement of extracellular vesicle studies and to elucidate the novel functions of these complex extracellular organelles.

17.
Am J Respir Cell Mol Biol ; 49(4): 637-45, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23713467

RESUMEN

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.


Asunto(s)
Neumonía/microbiología , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/metabolismo , Animales , Línea Celular , Quimiocinas/metabolismo , Humanos , Pulmón/metabolismo , Pulmón/microbiología , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Neumonía/metabolismo , Infecciones por Pseudomonas/metabolismo , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Transfección
18.
Antimicrob Agents Chemother ; 57(6): 2589-95, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23529736

RESUMEN

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.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana , Escherichia coli/efectos de los fármacos , Espacio Extracelular/enzimología , Proteolípidos/metabolismo , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/metabolismo , beta-Lactamasas/metabolismo , Ampicilina/farmacología , Escherichia coli/metabolismo , Humanos , Pruebas de Sensibilidad Microbiana , Staphylococcus aureus/genética , Staphylococcus aureus/ultraestructura , beta-Lactamasas/genética , beta-Lactamasas/aislamiento & purificación
19.
PLoS One ; 8(3): e59276, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23516621

RESUMEN

Escherichia coli, as one of the gut microbiota, can evoke severe inflammatory diseases including peritonitis and sepsis. Gram-negative bacteria including E. coli constitutively release nano-sized outer membrane vesicles (OMVs). Although E. coli OMVs can induce the inflammatory responses without live bacteria, the effect of E. coli OMVs in vivo on endothelial cell function has not been previously elucidated. In this study, we show that bacteria-free OMVs increased the expression of endothelial intercellular adhesion molecule-1 (ICAM-1), E-selectin and vascular cell adhesion molecule-1, and enhanced the leukocyte binding on human microvascular endothelial cells in vitro. Inhibition of NF-κB and TLR4 reduced the expression of cell adhesion molecules in vitro. OMVs given intraperitoneally to the mice induced ICAM-1 expression and neutrophil sequestration in the lung endothelium, and the effects were reduced in ICAM-1(-/-) and TLR4(-/-) mice. When compared to free lipopolysaccharide, OMVs were more potent in inducing both ICAM-1 expression as well as leukocyte adhesion in vitro, and ICAM-1 expression and neutrophil sequestration in the lungs in vivo. This study shows that OMVs potently up-regulate functional cell adhesion molecules via NF-κB- and TLR4-dependent pathways, and that OMVs are more potent than free lipopolysaccharide.


Asunto(s)
Escherichia coli/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Animales , Selectina E/genética , Selectina E/metabolismo , Escherichia coli/efectos de los fármacos , Molécula 1 de Adhesión Intercelular/genética , Lipopolisacáridos/farmacología , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
20.
J Immunol ; 190(8): 4092-102, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23514742

RESUMEN

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.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/mortalidad , Vacunas contra Escherichia coli/administración & dosificación , Vacunas contra Escherichia coli/inmunología , Exosomas/inmunología , Células TH1/inmunología , Células Th17/inmunología , Inmunidad Adaptativa , Animales , Membrana Celular/inmunología , Membrana Celular/microbiología , Células Cultivadas , Infecciones por Escherichia coli/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Sepsis/inmunología , Sepsis/microbiología , Sepsis/prevención & control , Síndrome de Respuesta Inflamatoria Sistémica/inmunología , Síndrome de Respuesta Inflamatoria Sistémica/microbiología , Síndrome de Respuesta Inflamatoria Sistémica/patología , Células TH1/microbiología , Células TH1/patología , Células Th17/microbiología , Células Th17/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA