Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Extracell Vesicles ; 11(1): e12172, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34981901

RESUMEN

Intestinal commensal bacteria contribute to maintaining gut homeostasis. Disruptions to the commensal flora are linked to the development and persistence of disease. The importance of these organisms is further demonstrated by the widespread ability of enteric viruses to exploit commensal bacteria to enhance viral infection. These viruses interact directly with commensal bacteria, and while the impact of this interaction on viral infection is well described for several viruses, the impact on the commensal bacteria has yet to be explored. In this article, we demonstrate, for the first time, that enteric viruses alter the gene expression and phenotype of individual commensal bacteria. Human and murine norovirus interaction with bacteria resulted in genome-wide differential gene expression and marked changes in the surface architecture of the bacterial cells. Furthermore, the interaction of the virus with bacteria led to increased production of smaller outer membrane vesicles (OMVs). Enhanced production of smaller vesicles was also observed when noroviruses were incubated with other commensal bacteria, indicating a potentially broad impact of norovirus interaction. The vesicle production observed in the in vivo model followed a similar trend where an increased quantity of smaller bacterial vesicles was observed in stool collected from virus-infected mice compared to mock-infected mice. Furthermore, changes in vesicle size were linked to changes in protein content and abundance, indicating that viral binding induced a shift in the mechanism of the OMV biogenesis. Collectively, these data demonstrate that enteric viruses induce specific changes in bacterial gene expression, leading to changes in bacterial extracellular vesicle production that can potentially impact host responses to infection.


Asunto(s)
Membrana Externa Bacteriana/metabolismo , Vesículas Extracelulares/metabolismo , Gastroenteritis/microbiología , Microbioma Gastrointestinal , Norovirus/fisiología , Animales , Membrana Externa Bacteriana/ultraestructura , Enterobacter cloacae/genética , Enterobacter cloacae/metabolismo , Gastroenteritis/metabolismo , Gastroenteritis/virología , Humanos , Ratones , Interacciones Microbianas
2.
Elife ; 102021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34468314

RESUMEN

The ability to produce outer membrane projections in the form of tubular membrane extensions (MEs) and membrane vesicles (MVs) is a widespread phenomenon among diderm bacteria. Despite this, our knowledge of the ultrastructure of these extensions and their associated protein complexes remains limited. Here, we surveyed the ultrastructure and formation of MEs and MVs, and their associated protein complexes, in tens of thousands of electron cryo-tomograms of ~90 bacterial species that we have collected for various projects over the past 15 years (Jensen lab database), in addition to data generated in the Briegel lab. We identified outer MEs and MVs in 13 diderm bacterial species and classified several major ultrastructures: (1) tubes with a uniform diameter (with or without an internal scaffold), (2) tubes with irregular diameter, (3) tubes with a vesicular dilation at their tip, (4) pearling tubes, (5) connected chains of vesicles (with or without neck-like connectors), (6) budding vesicles and nanopods. We also identified several protein complexes associated with these MEs and MVs which were distributed either randomly or exclusively at the tip. These complexes include a secretin-like structure and a novel crown-shaped structure observed primarily in vesicles from lysed cells. In total, this work helps to characterize the diversity of bacterial membrane projections and lays the groundwork for future research in this field.


Asunto(s)
Bacterias/ultraestructura , Proteínas de la Membrana Bacteriana Externa/ultraestructura , Membrana Externa Bacteriana/ultraestructura , Extensiones de la Superficie Celular/ultraestructura , Microscopía por Crioelectrón , Tomografía con Microscopio Electrónico , Bacterias/clasificación , Complejos Multiproteicos
3.
Res Microbiol ; 172(6): 103865, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34284091

RESUMEN

An extensive morphological analysis of the Neisseria meningitidis cell envelope, including serogroup B capsule and outer membrane, based on atomic force microscopy (AFM) together with mechanical characterization by force spectroscopic measurements, has been carried out. Three meningococcal strains were used: the encapsulated serogroup B strain B1940, and the isogenic mutants B1940 siaD(+C) (lacking capsule), and B1940 cps (lacking both capsule and lipooligosaccharide outer core). AFM experiments with the encapsulated strain B1940 provided unprecedented images of the meningococcal capsule, which seems to be characterized by protrusions ("bumps") with the lateral dimensions of about 30 nm. Measurement of the Young's modulus provided quantitative assessment of the property of the capsule to confer resistance to mechanical stress. Moreover, Raman spectroscopy gave a fingerprint by which it was possible to identify the specific molecular species of the three strains analyzed, and to highlight major differences between them.


Asunto(s)
Cápsulas Bacterianas/ultraestructura , Membrana Externa Bacteriana/ultraestructura , Neisseria meningitidis Serogrupo B/ultraestructura , Cápsulas Bacterianas/química , Cápsulas Bacterianas/fisiología , Membrana Externa Bacteriana/química , Membrana Externa Bacteriana/fisiología , Módulo de Elasticidad , Microscopía de Fuerza Atómica , Neisseria meningitidis Serogrupo B/química , Neisseria meningitidis Serogrupo B/genética , Polisacáridos Bacterianos/química , Espectrometría Raman , Estrés Mecánico , Propiedades de Superficie
4.
Biochim Biophys Acta Gen Subj ; 1865(4): 129763, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33065252

RESUMEN

Extracellular vesicles (EVs) are nanoparticles which are released by cells from all three domains of life: Archaea, Bacteria and Eukarya. They can mediate cell-cell communication by transferring cargoes such as proteins and nucleic acids between cells. EVs receive great interest in both academia and industry as they have the potential to be natural drug carriers or vaccine candidates. However, limitations to their clinical translation exist as efficient isolation, loading, labelling and surface-engineering methods are lacking. In this article, we investigate a 'post-insertion' approach, which is commonly used in the functionalization of liposomes in the pharmaceutical field, on two different EV types: mammalian cell-derived EVs and bacteria-derived EVs. We aimed to find an easy and flexible approach to functionalize EVs, thereby improving the labelling, isolation, and surface-engineering.


Asunto(s)
Bacterias/química , Membrana Externa Bacteriana/química , Vesículas Extracelulares/química , Inmunohistoquímica/métodos , Animales , Membrana Externa Bacteriana/ultraestructura , Western Blotting/métodos , Técnicas de Cultivo de Célula/métodos , Línea Celular Tumoral , Electroforesis en Gel de Poliacrilamida/métodos , Vesículas Extracelulares/ultraestructura , Citometría de Flujo/métodos , Células HEK293 , Humanos , Ratones , Microscopía Electrónica de Transmisión/métodos , Propiedades de Superficie
5.
Nat Commun ; 11(1): 6312, 2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-33298927

RESUMEN

The increase in speed of the high-speed atomic force microscopy (HS-AFM) compared to that of the conventional AFM made possible the first-ever visualisation at the molecular-level of the activity of an antimicrobial peptide on a membrane. We investigated the medically prescribed but poorly understood lipopeptide Daptomycin under infection-like conditions (37 °C, bacterial lipid composition and antibiotic concentrations). We confirmed so far hypothetical models: Dap oligomerization and the existence of half pores. Moreover, we detected unknown molecular mechanisms: new mechanisms to form toroidal pores or to resist Dap action, and to unprecedently quantify the energy profile of interacting oligomers. Finally, the biological and medical relevance of the findings was ensured by a multi-scale multi-nativeness-from the molecule to the cell-correlation of molecular-level information from living bacteria (Bacillus subtilis strains) to liquid-suspended vesicles and supported-membranes using electron and optical microscopies and the lipid tension probe FliptR, where we found that the cells with a healthier state of their cell wall show smaller membrane deformations.


Asunto(s)
Antibacterianos/farmacología , Bacillus subtilis/efectos de los fármacos , Infecciones Bacterianas/tratamiento farmacológico , Daptomicina/farmacología , Microscopía de Fuerza Atómica , Antibacterianos/uso terapéutico , Bacillus subtilis/citología , Bacillus subtilis/ultraestructura , Membrana Externa Bacteriana/efectos de los fármacos , Membrana Externa Bacteriana/ultraestructura , Pared Celular/efectos de los fármacos , Pared Celular/ultraestructura , Daptomicina/uso terapéutico , Farmacorresistencia Bacteriana , Humanos , Membrana Dobles de Lípidos , Pruebas de Sensibilidad Microbiana , Microscopía Electrónica de Transmisión , Modelos Biológicos
6.
Sci Rep ; 10(1): 21289, 2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-33277560

RESUMEN

Outer membrane vesicles (OMVs) produced by Gram-negative bacteria have roles in cell-to-cell signaling, biofilm formation, and stress responses. Here, the effects of abiotic stressors on OMV contents and composition from biofilm cells of the plant health-promoting bacterium Pseudomonas chlororaphis O6 (PcO6) are examined. Two stressors relevant to this root-colonizing bacterium were examined: CuO nanoparticles (NPs)-a potential fertilizer and fungicide- and H2O2-released from roots during plant stress responses. Atomic force microscopy revealed 40-300 nm diameter OMVs from control and stressed biofilm cells. Raman spectroscopy with linear discriminant analysis (LDA) was used to identify changes in chemical profiles of PcO6 cells and resultant OMVs according to the cellular stressor with 84.7% and 83.3% accuracies, respectively. All OMVs had higher relative concentrations of proteins, lipids, and nucleic acids than PcO6 cells. The nucleic acid concentration in OMVs exhibited a cellular stressor-dependent increase: CuO NP-induced OMVs > H2O2-induced OMVs > control OMVs. Biochemical assays confirmed the presence of lipopolysaccharides, nucleic acids, and protein in OMVs; however, these assays did not discriminate OMV composition according to the cellular stressor. These results demonstrate the sensitivity of Raman spectroscopy using LDA to characterize and distinguish cellular stress effects on OMVs composition and contents.


Asunto(s)
Membrana Externa Bacteriana/metabolismo , Vesículas Extracelulares/metabolismo , Pseudomonas chlororaphis/metabolismo , Estrés Fisiológico , Membrana Externa Bacteriana/química , Membrana Externa Bacteriana/ultraestructura , Vesículas Extracelulares/química , Vesículas Extracelulares/ultraestructura , Pseudomonas chlororaphis/química , Pseudomonas chlororaphis/ultraestructura , Espectrometría Raman
7.
Elife ; 92020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33236984

RESUMEN

In double-membraned bacteria, phospholipid transport across the cell envelope is critical to maintain the outer membrane barrier, which plays a key role in virulence and antibiotic resistance. An MCE transport system called Mla has been implicated in phospholipid trafficking and outer membrane integrity, and includes an ABC transporter, MlaFEDB. The transmembrane subunit, MlaE, has minimal sequence similarity to other transporters, and the structure of the entire inner-membrane MlaFEDB complex remains unknown. Here, we report the cryo-EM structure of MlaFEDB at 3.05 Å resolution, revealing distant relationships to the LPS and MacAB transporters, as well as the eukaryotic ABCA/ABCG families. A continuous transport pathway extends from the MlaE substrate-binding site, through the channel of MlaD, and into the periplasm. Unexpectedly, two phospholipids are bound to MlaFEDB, suggesting that multiple lipid substrates may be transported each cycle. Our structure provides mechanistic insight into substrate recognition and transport by MlaFEDB.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/química , Transportadoras de Casetes de Unión a ATP/ultraestructura , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/ultraestructura , Transportadoras de Casetes de Unión a ATP/metabolismo , Membrana Externa Bacteriana/química , Membrana Externa Bacteriana/metabolismo , Membrana Externa Bacteriana/ultraestructura , Transporte Biológico Activo/fisiología , Microscopía por Crioelectrón , Escherichia coli , Proteínas de Escherichia coli/metabolismo , Conformación Proteica
8.
NPJ Biofilms Microbiomes ; 6(1): 35, 2020 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-33037198

RESUMEN

Methods for bacterial detection are needed to advance the infection research and diagnostics. Based on conformation-sensitive fluorescent tracer molecules, optotracing was recently established for dynamic detection and visualization of structural amyloids and polysaccharides in the biofilm matrix of gram-negative bacteria. Here, we extend the use of optotracing for detection of gram-positive bacteria, focussing on the clinically relevant opportunistic human pathogen Staphylococcus aureus. We identify a donor-acceptor-donor-type optotracer, whose binding-induced fluorescence enables real-time detection, quantification, and visualization of S. aureus in monoculture and when mixed with gram-negative Salmonella Enteritidis. An algorithm-based automated high-throughput screen of 1920 S. aureus transposon mutants recognized the cell envelope as the binding target, which was corroborated by super-resolution microscopy of bacterial cells and spectroscopic analysis of purified cell wall components. The binding event was essentially governed by hydrophobic interactions, which permitted custom-designed tuning of the binding selectivity towards S. aureus versus Enterococcus faecalis by appropriate selection of buffer conditions. Collectively this work demonstrates optotracing as an enabling technology relevant for any field of basic and applied research, where visualization and detection of S. aureus is needed.


Asunto(s)
Técnicas Bacteriológicas/métodos , Mutación , Salmonella enteritidis/crecimiento & desarrollo , Staphylococcus aureus/aislamiento & purificación , Tiofenos/química , Algoritmos , Membrana Externa Bacteriana/química , Membrana Externa Bacteriana/ultraestructura , Elementos Transponibles de ADN , Fluorescencia , Ensayos Analíticos de Alto Rendimiento , Humanos , Microscopía Fluorescente , Polisacáridos Bacterianos/metabolismo , Espectrometría de Fluorescencia , Staphylococcus aureus/genética , Staphylococcus aureus/crecimiento & desarrollo
9.
Front Immunol ; 11: 1069, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32655550

RESUMEN

Acinetobacter baumannii (A. baumannii) is becoming a common global concern due to the emergence of multi-drug or pan-drug resistant strains. Confronting the issue of antimicrobial resistance by developing vaccines against the resistant pathogen is becoming a common strategy. In this study, different methods for preparing A. baumannii outer membrane vesicles (AbOMVs) vaccines were developed. sOMV (spontaneously released AbOMV) was extracted from the culture supernatant, while SuOMV (sucrose-extracted AbOMV) and nOMV (native AbOMV) were prepared from the bacterial cells. Three AbOMVs exhibited significant differences in yield, particle size, protein composition, and LPS/DNA content. To compare the protective efficacy of the three AbOMVs, groups of mice were immunized either intramuscularly or intranasally with each AbOMV. Vaccination via both routes conferred significant protection against lethal and sub-lethal A. baumannii challenge. Moreover, intranasal vaccination provided more robust protection, which may be attributed to the induction of significant sIgA response in mucosal sites. Among the three AbOMVs, SuOMV elicited the highest level of protective immunity against A. baumannii infection, whether intramuscular or intranasal immunization, which was characterized by the expression of the most profound specific serum IgG or mucosal sIgA. Taken together, the preparation method had a significant effect on the yield, morphology, and composition of AbOMVs, that further influenced the protective effect against A. baumannii infection.


Asunto(s)
Acinetobacter baumannii/inmunología , Vacunas Bacterianas/aislamiento & purificación , Infecciones por Acinetobacter/inmunología , Infecciones por Acinetobacter/microbiología , Infecciones por Acinetobacter/prevención & control , Acinetobacter baumannii/patogenicidad , Acinetobacter baumannii/ultraestructura , Administración Intranasal , Animales , Anticuerpos Antibacterianos/biosíntesis , Anticuerpos Antibacterianos/sangre , Especificidad de Anticuerpos , Membrana Externa Bacteriana/inmunología , Membrana Externa Bacteriana/ultraestructura , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas de la Membrana Bacteriana Externa/aislamiento & purificación , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/inmunología , Citocinas/metabolismo , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunidad Mucosa , Inmunoglobulina A Secretora/biosíntesis , Inmunoglobulina G/sangre , Inmunoglobulina G/clasificación , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión
10.
Proc Natl Acad Sci U S A ; 117(16): 8941-8947, 2020 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-32241888

RESUMEN

The bacterial flagellum is an amazing nanomachine. Understanding how such complex structures arose is crucial to our understanding of cellular evolution. We and others recently reported that in several Gammaproteobacterial species, a relic subcomplex comprising the decorated P and L rings persists in the outer membrane after flagellum disassembly. Imaging nine additional species with cryo-electron tomography, here, we show that this subcomplex persists after flagellum disassembly in other phyla as well. Bioinformatic analyses fail to show evidence of any recent horizontal transfers of the P- and L-ring genes, suggesting that this subcomplex and its persistence is an ancient and conserved feature of the flagellar motor. We hypothesize that one function of the P and L rings is to seal the outer membrane after motor disassembly.


Asunto(s)
Bacterias/genética , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas Bacterianas/genética , Flagelos/genética , Especiación Genética , Bacterias/citología , Bacterias/metabolismo , Membrana Externa Bacteriana/metabolismo , Membrana Externa Bacteriana/ultraestructura , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/metabolismo , Biología Computacional , Microscopía por Crioelectrón , Tomografía con Microscopio Electrónico , Flagelos/metabolismo , Genes Bacterianos , Filogenia
11.
Int J Nanomedicine ; 15: 1929-1938, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32256069

RESUMEN

BACKGROUND: Nanoscale surface roughness has been suggested to have antibacterial and antifouling properties. Several existing models have attempted to explain the antibacterial mechanism of nanoscale rough surfaces without direct observation. Here, conventional and liquid-cell TEM are implemented to observe nanoscale bacteria/surface roughness interaction. The visualization of such interactions enables the inference of possible antibacterial mechanisms. METHODS AND RESULTS: Nanotextures are synthesized on biocompatible polymer microparticles (MPs) via plasma etching. Both conventional and liquid-phase transmission electron microscopy observations suggest that these MPs may cause cell lysis via bacterial binding to a single protrusion of the nanotexture. The bacterium/protrusion interaction locally compromises the cell wall, thus causing bacterial death. This study suggests that local mechanical damage and leakage of the cytosol kill the bacteria first, with subsequent degradation of the cell envelope. CONCLUSION: Nanoscale surface roughness may act via a penetrative bactericidal mechanism. This insight suggests that future research may focus on optimizing bacterial binding to individual nanoscale projections in addition to stretching bacteria between nanopillars. Further, antibacterial nanotextures may find use in novel applications employing particles in addition to nanotextures on fibers or films.


Asunto(s)
Antibacterianos/química , Antibacterianos/farmacología , Membrana Externa Bacteriana/efectos de los fármacos , Portadores de Fármacos/química , Membrana Externa Bacteriana/ultraestructura , Portadores de Fármacos/farmacología , Escherichia coli/efectos de los fármacos , Microplásticos/química , Microplásticos/farmacología , Microscopía Electrónica de Transmisión , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Propiedades de Superficie
12.
Biomolecules ; 10(3)2020 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-32120823

RESUMEN

A number of Gram-negative bacteria have a membrane surrounding their flagella, referred to as the flagellar sheath, which is continuous with the outer membrane. The flagellar sheath was initially described in Vibrio metschnikovii in the early 1950s as an extension of the outer cell wall layer that completely surrounded the flagellar filament. Subsequent studies identified other bacteria that possess flagellar sheaths, most of which are restricted to a few genera of the phylum Proteobacteria. Biochemical analysis of the flagellar sheaths from a few bacterial species revealed the presence of lipopolysaccharide, phospholipids, and outer membrane proteins in the sheath. Some proteins localize preferentially to the flagellar sheath, indicating mechanisms exist for protein partitioning to the sheath. Recent cryo-electron tomography studies have yielded high resolution images of the flagellar sheath and other structures closely associated with the sheath, which has generated insights and new hypotheses for how the flagellar sheath is synthesized. Various functions have been proposed for the flagellar sheath, including preventing disassociation of the flagellin subunits in the presence of gastric acid, avoiding activation of the host innate immune response by flagellin, activating the host immune response, adherence to host cells, and protecting the bacterium from bacteriophages.


Asunto(s)
Flagelos/genética , Bacterias Gramnegativas/genética , Infecciones por Bacterias Gramnegativas/microbiología , Filogenia , Membrana Externa Bacteriana/metabolismo , Membrana Externa Bacteriana/ultraestructura , Flagelos/metabolismo , Flagelos/ultraestructura , Bacterias Gramnegativas/metabolismo , Bacterias Gramnegativas/ultraestructura , Humanos , Vibrio/genética , Vibrio/metabolismo , Vibrio/ultraestructura
13.
J Gen Appl Microbiol ; 66(2): 121-128, 2020 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-32173680

RESUMEN

In photosynthetic microorganisms, cell cycle progression depends on day and night cycles; however, how cell division is regulated in response to these environmental changes is poorly understood. RpaA has been implicated in the signal output from both circadian clocks and light/dark conditions in the unicellular spherical-celled cyanobacterium Synechocystis sp. PCC 6803. In the present study, we investigated the involvement of a two-component response regulator RpaA in cell division regulation. Firstly, we examined the effects of rpaA overexpression on cell morphology and the expression levels of cell division genes. We observed an increase in the volume of non-dividing cells and a high proportion of dividing cells in rpaA-overexpressing strains by light microscopy. The expression levels of selected cell division-related genes were higher in the rpaA-overexpressing strain than in the wild type, including minD of the Min system; cdv3 and zipN, which encode two divisome components; and murB, murC, and pbp2, which are involved in peptidoglycan (PG) synthesis. Moreover, in the rpaA-overexpressing strain, the outer membrane and cell wall PG layer were not smooth, and the outer membrane was not clearly visible by transmission electron microscopy. These results demonstrated that rpaA overexpression causes an impaired cell division, which is accompanied by transcriptional activation of cell division genes and morphological changes in the PG layer and outer membrane.


Asunto(s)
Membrana Externa Bacteriana/ultraestructura , Proteínas de Ciclo Celular/genética , División Celular , Relojes Circadianos/genética , Synechocystis/genética , Synechocystis/ultraestructura , Proteínas de Ciclo Celular/metabolismo , Regulación Bacteriana de la Expresión Génica , Microscopía , Microscopía Electrónica de Transmisión , Peptidoglicano/biosíntesis , Reacción en Cadena en Tiempo Real de la Polimerasa , Synechocystis/citología
14.
Artículo en Inglés | MEDLINE | ID: mdl-32211344

RESUMEN

Outer membrane vesicles (OMVs) are nanoscale proteoliposomes secreted from the cell envelope of all Gram-negative bacteria. Originally considered as an artifact of the cell wall, OMVs are now recognized as a general secretion system, which serves to improve the fitness of bacteria and facilitate bacterial interactions in polymicrobial communities as well as interactions between the microbe and the host. In general, OMVs are released in increased amounts from pathogenic bacteria and have been found to harbor much of the contents of the parental bacterium. They mainly encompass components of the outer membrane and the periplasm including various virulence factors such as toxins, adhesins, and immunomodulatory molecules. Numerous studies have clearly shown that the delivery of toxins and other virulence factors via OMVs essentially influences their interactions with host cells. Here, we review the OMV-mediated intracellular deployment of toxins and other virulence factors with a special focus on intestinal pathogenic Escherichia coli. Especially, OMVs ubiquitously produced and secreted by enterohemorrhagic E. coli (EHEC) appear as a highly advanced mechanism for secretion and simultaneous, coordinated and direct delivery of bacterial virulence factors into host cells. OMV-associated virulence factors are not only stabilized by the association with OMVs, but can also often target previously unknown target structures and perform novel activities. The toxins are released by OMVs in their active forms and are transported via cell sorting processes to their specific cell compartments, where they can develop their detrimental effects. OMVs can be considered as bacterial "long distance weapons" that attack host tissues and help bacterial pathogens to establish the colonization of their biological niche(s), impair host cell function, and modulate the defense of the host. Thus, OMVs contribute significantly to the virulence of the pathogenic bacteria.


Asunto(s)
Sistemas de Secreción Bacterianos/metabolismo , Escherichia coli Enterohemorrágica/patogenicidad , Escherichia coli Enterotoxigénica/patogenicidad , Infecciones por Escherichia coli/microbiología , Intestinos/microbiología , Proteolípidos/metabolismo , Factores de Virulencia/metabolismo , Animales , Membrana Externa Bacteriana/metabolismo , Membrana Externa Bacteriana/ultraestructura , Toxinas Bacterianas/metabolismo , Escherichia coli Enterohemorrágica/metabolismo , Escherichia coli Enterotoxigénica/metabolismo , Enterotoxinas/metabolismo , Humanos , Transporte de Proteínas , Proteolípidos/ultraestructura , Estrés Fisiológico , Virulencia
15.
Eur J Pharm Biopharm ; 146: 55-63, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31805356

RESUMEN

In recent years, extracellular vesicles (EVs) and outer membrane vesicles (OMVs) have become an extensive and diverse field of research. They hold potential as diagnostic markers, therapeutics and for fundamental biological understanding. Despite ongoing studies, numerous information regarding function, content and stability of EVs remains unclear. If EVs and OMVs ought to be used as therapeutics and in clinical environments, their stability is one of the most important factors to be considered. Especially for formulation development, EVs and OMVs need to be stable at higher temperatures. To the best of our knowledge, very little work has been published regarding heat stability of neither EVs nor OMVs. In the present study, we investigated B lymphoblastoid cell-derived EVs and OMVs derived from myxobacterial species Sorangiineae as model vesicles. We exposed the vesicles to 37 °C, 50 °C, 70 °C and 100 °C for 1 h, 6 h and 24 h, and also autoclaved them. Interestingly, physico-chemical analyses such as size, particle concentration and protein concentration showed minor alterations, particularly at 37 °C. Flow cytometry analysis emphasised these results suggesting that after heat impact, EVs and OMVs were still able to be taken up by macrophage-like dTHP-1 cells. These data indicate that both mammalian and bacterial vesicles show intrinsic stability at physiological temperature. Our findings are important to consider for vesicle formulation and for advanced bioengineering approaches.


Asunto(s)
Membrana Externa Bacteriana/química , Bioingeniería , Vesículas Extracelulares/química , Calor/efectos adversos , Membrana Externa Bacteriana/ultraestructura , Línea Celular , Microscopía por Crioelectrón , Vesículas Extracelulares/ultraestructura , Humanos , Linfocitos/citología , Myxococcales/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA