Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ACS Appl Mater Interfaces ; 15(3): 3744-3759, 2023 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-36630299

RESUMEN

Inducing immunogenic cell death (ICD) is a critical strategy for enhancing cancer immunotherapy. However, inefficient and risky ICD inducers along with a tumor hypoxia microenvironment seriously limit the immunotherapy efficacy. Non-specific delivery is also responsible for this inefficiency. In this work, we report a drug-free bacteria-derived outer membrane vesicle (OMV)-functionalized Fe3O4-MnO2 (FMO) nanoplatform that realized neutrophil-mediated targeted delivery and photothermally enhanced cancer immunotherapy. In this system, modification of OMVs derived from Escherichia coli enhanced the accumulation of FMO NPs at the tumor tissue through neutrophil-mediated targeted delivery. The FMO NPs underwent reactive decomposition in the tumor site, generating manganese and iron ions that induced ICD and O2 that regulated the tumor hypoxia environment. Moreover, OMVs are rich in pathogen-associated pattern molecules that can overcome the tumor immunosuppressive microenvironment and effectively activate immune cells, thereby enhancing specific immune responses. Photothermal therapy (PTT) caused by MnO2 and Fe3O4 can not only indirectly stimulate systemic immunity by directly destroying tumor cells but also promote the enrichment of neutrophil-equipped nanoparticles by enhancing the inflammatory response at the tumor site. Finally, the proposed multi-modal treatment system with targeted delivery capability realized effective tumor immunotherapy to prevent tumor growth and recurrence.


Asunto(s)
Bioingeniería , Inmunoterapia , Nanopartículas Multifuncionales , Neoplasias , Humanos , Línea Celular Tumoral , Inmunoterapia/métodos , Nanopartículas Multifuncionales/uso terapéutico , Neoplasias/terapia , Microambiente Tumoral/inmunología , Vesículas Transportadoras/química , Vesículas Transportadoras/inmunología , Membrana Externa Bacteriana/química , Membrana Externa Bacteriana/inmunología , Escherichia coli
2.
Infect Immun ; 89(4)2021 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-33361202

RESUMEN

Sphingolipids (SLs) are essential structural components of mammalian cell membranes. Our group recently determined that the oral anaerobe Porphyromonas gingivalis delivers its SLs to host cells and that the ability of P. gingivalis to synthesize SLs limits the elicited host inflammatory response during cellular infection. As P. gingivalis robustly produces outer membrane vesicles (OMVs), we hypothesized that OMVs serve as a delivery vehicle for SLs, that the SL status of the OMVs may impact cargo loading to OMVs, and that SL-containing OMVs limit elicited host inflammation similar to that observed by direct bacterial challenge. Transwell cell culture experiments determined that in comparison to the parent strain W83, the SL-null mutant elicited a hyperinflammatory immune response from THP-1 macrophage-like cells with elevated tumor necrosis factor alpha (TNF-α), interleukin 1ß (IL-1ß), and IL-6. Targeted assessment of Toll-like receptors (TLRs) identified elevated expression of TLR2, unchanged TLR4, and elevated expression of the adaptor molecules MyD88 and TRIF (Toll/IL-1 receptor domain-containing adaptor-inducing beta interferon) by SL-null P. gingivalis No significant differences in gingipain activity were observed in our infection models, and both strains produced OMVs of similar sizes. Using comparative two-dimensional gel electrophoresis, we identified differences in the protein cargo of the OMVs between parent and SL-null strain. Importantly, use of purified OMVs recapitulated the cellular inflammatory response observed in the transwell system with whole bacteria. These findings provide new insights into the role of SLs in P. gingivalis OMV cargo assembly and expand our understanding of SL-OMVs as bacterial structures that modulate the host inflammatory response.


Asunto(s)
Infecciones por Bacteroidaceae/inmunología , Infecciones por Bacteroidaceae/microbiología , Macrófagos/inmunología , Porphyromonas gingivalis/inmunología , Porphyromonas gingivalis/metabolismo , Esfingolípidos/inmunología , Vesículas Transportadoras/inmunología , Infecciones por Bacteroidaceae/patología , Transporte Biológico , Interacciones Huésped-Patógeno , Inmunomodulación , Mutación , Porphyromonas gingivalis/genética , Proteómica/métodos , Esfingolípidos/metabolismo , Vesículas Transportadoras/metabolismo
3.
Clin Exp Immunol ; 196(3): 287-304, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30985006

RESUMEN

Plague caused by the Gram-negative bacterium, Yersinia pestis, is still endemic in parts of the world today. Protection against pneumonic plague is essential to prevent the development and spread of epidemics. Despite this, there are currently no licensed plague vaccines in the western world. Here we describe the means of delivering biologically active plague vaccine antigens directly to mucosal sites of plague infection using highly stable microvesicles (outer membrane vesicles; OMVs) that are naturally produced by the abundant and harmless human commensal gut bacterium Bacteroides thetaiotaomicron (Bt). Bt was engineered to express major plague protective antigens in its OMVs, specifically Fraction 1 (F1) in the outer membrane and LcrV (V antigen) in the lumen, for targeted delivery to the gastrointestinal (GI) and respiratory tracts in a non-human primate (NHP) host. Our key findings were that Bt OMVs stably expresses F1 and V plague antigens, particularly the V antigen, in the correct, immunogenic form. When delivered intranasally V-OMVs elicited substantive and specific immune and antibody responses, both in the serum [immunoglobulin (Ig)G] and in the upper and lower respiratory tract (IgA); this included the generation of serum antibodies able to kill plague bacteria. Our results also showed that Bt OMV-based vaccines had many desirable characteristics, including: biosafety and an absence of any adverse effects, pathology or gross alteration of resident microbial communities (microbiotas); high stability and thermo-tolerance; needle-free delivery; intrinsic adjuvanticity; the ability to stimulate both humoral and cell-mediated immune responses; and targeting of primary sites of plague infection.


Asunto(s)
Antígenos Bacterianos/metabolismo , Membrana Externa Bacteriana/metabolismo , Bacteroides thetaiotaomicron/metabolismo , Vacuna contra la Peste/inmunología , Peste/inmunología , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Vesículas Transportadoras/inmunología , Yersinia pestis/fisiología , Administración Intranasal , Animales , Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/genética , Bacteroides thetaiotaomicron/genética , Bioingeniería , Muerte Celular , Células Cultivadas , Microbioma Gastrointestinal/genética , Humanos , Inmunidad Celular , Inmunidad Humoral , Inmunoglobulina A/metabolismo , Inmunoglobulina G/sangre , Macaca , Peste/prevención & control , Vacuna contra la Peste/metabolismo , Proteínas Citotóxicas Formadoras de Poros/genética , Vesículas Transportadoras/metabolismo
4.
Proc Natl Acad Sci U S A ; 115(14): E3106-E3115, 2018 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-29555731

RESUMEN

Many microbial pathogens produce a ß-(1→6)-linked poly-N-acetyl-d-glucosamine (PNAG) surface capsule, including bacterial, fungal, and protozoan cells. Broadly protective immune responses to this single conserved polysaccharide antigen in animals are possible but only when a deacetylated poly-N-acetyl-d-glucosamine (dPNAG; <30% acetate) glycoform is administered as a conjugate to a carrier protein. Unfortunately, conventional methods for natural extraction or chemical synthesis of dPNAG and its subsequent conjugation to protein carriers can be technically demanding and expensive. Here, we describe an alternative strategy for creating broadly protective vaccine candidates that involved coordinating recombinant poly-N-acetyl-d-glucosamine (rPNAG) biosynthesis with outer membrane vesicle (OMV) formation in laboratory strains of Escherichia coli The glycosylated outer membrane vesicles (glycOMVs) released by these engineered bacteria were decorated with the PNAG glycopolymer and induced high titers of PNAG-specific IgG antibodies after immunization in mice. When a Staphylococcus aureus enzyme responsible for PNAG deacetylation was additionally expressed in these cells, glycOMVs were generated that elicited antibodies to both highly acetylated PNAG (∼95-100% acetate) and a chemically deacetylated dPNAG derivative (∼15% acetate). These antibodies mediated efficient in vitro killing of two distinct PNAG-positive bacterial species, namely S. aureus and Francisella tularensis subsp. holarctica, and mice immunized with PNAG-containing glycOMVs developed protective immunity against these unrelated pathogens. Collectively, our results reveal the potential of glycOMVs for targeting this conserved polysaccharide antigen and engendering protective immunity against the broad range of pathogens that produce surface PNAG.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Antígenos de Superficie/inmunología , Bacterias/inmunología , Infecciones Bacterianas/prevención & control , Vacunas Bacterianas/uso terapéutico , Inmunización/métodos , Vesículas Transportadoras/inmunología , Animales , Infecciones Bacterianas/inmunología , Vacunas Bacterianas/inmunología , Femenino , Ratones , Ratones Endogámicos BALB C , Vacunas Conjugadas/inmunología , Vacunas Conjugadas/uso terapéutico , beta-Glucanos/metabolismo
5.
Microbiol Res ; 200: 25-32, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28527761

RESUMEN

During infection, the release of nano-sized membrane vesicle is a process which is common both for bacteria and host cells. Host cell-derived membrane vesicles can be involved in innate and adaptive immunity whereas bacterial membrane vesicles can contribute to bacterial pathogenicity. To study the contribution of both membrane vesicle populations during infection is highly complicated as most vesicles fall within a similar size range of 30-300nm. Specialized techniques for purification are required and often no single technique complies on its own. Moreover, techniques for vesicle quantification are either complicated to use or do not distinguish between host cell-derived and bacterial membrane vesicle subpopulations. Here we demonstrate a bead-based platform that allows a semi-quantitatively analysis by flow-cytometry of bacterial and host-cell derived membrane vesicles. We show this method can be used to study heterogeneous and complex vesicle populations composed of bacterial and host-cell membrane vesicles. The easy accessible design of the protocol makes it also highly suitable for screening procedures to assess how intrinsic and environmental factors affect vesicle release.


Asunto(s)
Bacterias/citología , Línea Celular/citología , Citometría de Flujo/métodos , Vesículas Transportadoras/química , Anticuerpos , Línea Celular/microbiología , Membrana Celular , Recuento de Colonia Microbiana , Epítopos , Humanos , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/microbiología , Moraxella catarrhalis/clasificación , Pseudomonas aeruginosa/citología , Vesículas Transportadoras/inmunología
6.
Sci Rep ; 6: 37242, 2016 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-27849050

RESUMEN

Outer membrane vesicles (OMVs) have proven to be highly immunogenic and induced an immune response against bacterial infection in human clinics and animal models. We sought to investigate whether engineered OMVs can be a feasible antigen-delivery platform for efficiently inducing specific antibody responses. In this study, Omp22 (an outer membrane protein of A. baumannii) was displayed on E. coli DH5α-derived OMVs (Omp22-OMVs) using recombinant gene technology. The morphological features of Omp22-OMVs were similar to those of wild-type OMVs (wtOMVs). Immunization with Omp22-OMVs induced high titers of Omp22-specific antibodies. In a murine sepsis model, Omp22-OMV immunization significantly protected mice from lethal challenge with a clinically isolated A. baumannii strain, which was evidenced by the increased survival rate of the mice, the reduced bacterial burdens in the lung, spleen, liver, kidney, and blood, and the suppressed serum levels of inflammatory cytokines. In vitro opsonophagocytosis assays showed that antiserum collected from Omp22-OMV-immunized mice had bactericidal activity against clinical isolates, which was partly specific antibody-dependent. These results strongly indicated that engineered OMVs could display a whole heterologous protein (~22 kDa) on the surface and effectively induce specific antibody responses, and thus OMVs have the potential to be a feasible vaccine platform.


Asunto(s)
Infecciones por Acinetobacter/inmunología , Acinetobacter baumannii/inmunología , Antígenos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Escherichia coli/inmunología , Vesículas Transportadoras/inmunología , Infecciones por Acinetobacter/microbiología , Acinetobacter baumannii/fisiología , Secuencia de Aminoácidos , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Vacunas Bacterianas/inmunología , Secuencia de Bases , Escherichia coli/genética , Escherichia coli/metabolismo , Femenino , Inmunización/métodos , Ratones Endogámicos ICR , Sepsis/sangre , Sepsis/inmunología , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico , Vesículas Transportadoras/metabolismo
7.
Vaccine ; 34(35): 4250-4256, 2016 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-27269057

RESUMEN

Trivalent native outer membrane vesicles (nOMVs) derived from three genetically modified Neisseria meningitidis serogroup B strains have been previously evaluated immunologically in mice and rabbits. This nOMV vaccine elicited serum bactericidal activity (SBA) against multiple N. meningitidis serogroup B strains as well as strains from serogroups C, Y, W, and X. In this study, we used trivalent nOMVs isolated from the same vaccine strains and evaluated their immunogenicity in an infant Rhesus macaque (IRM) model whose immune responses to the vaccine are likely to be more predictive of the responses in human infants. IRMs were immunized with trivalent nOMV vaccines and sera were evaluated for exogenous human serum complement-dependent SBA (hSBA). Antibody responses to selected hSBA generating antigens contained within the trivalent nOMVs were also measured and we found that antibody titers against factor H binding protein variant 2 (fHbpv2) were very low in the sera from animals immunized with these original nOMV vaccines. To increase the fHbp content in the nOMVs, the vaccine strains were further genetically altered by addition of another fHbp gene copy into the porB locus. Trivalent nOMVs from the three new vaccine strains had higher fHbp antigen levels and generated higher anti-fHbp antibody responses in immunized mice and IRMs. As expected, fHbp insertion into the porB locus resulted in no PorB expression. Interestingly, higher expression of PorA, an hSBA generating antigen, was observed for all three modified vaccine strains. Compared to the trivalent nOMVs from the original strains, higher PorA levels in the improved nOMVs resulted in higher anti-PorA antibody responses in mice and IRMs. In addition, hSBA titers against other strains with PorA as the only hSBA antigen in common with the vaccine strains also increased.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Ingeniería Genética , Inmunogenicidad Vacunal , Vacunas Meningococicas/inmunología , Vesículas Transportadoras/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Formación de Anticuerpos , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Macaca mulatta , Vacunas Meningococicas/genética , Neisseria meningitidis , Neisseria meningitidis Serogrupo B , Porinas/genética
8.
Sci Rep ; 6: 23089, 2016 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-26980157

RESUMEN

Survival of Mycobacterium tuberculosis (Mtb) within the host macrophage is mediated through pathogen-dependent inhibition of phagosome-lysosome fusion, which enables bacteria to persist within the immature phagosomal compartment. By employing ultrastructural examination of different field isolates supported by biochemical analysis, we found that some of the Mtb strains were in fact poorly adapted for subsistence within endocytic vesicles of infected macrophages. Instead, through a mechanism involving activation of host cytosolic phospholipase A2, these bacteria rapidly escaped from phagosomes, and established residence in the cytoplasm of the host cell. Interestingly, by facilitating an enhanced suppression of host cellular autophagy, this translocation served as an alternate virulence acquisition mechanism. Thus, our studies reveal plasticity in the adaptation strategies employed by Mtb, for survival in the host macrophage.


Asunto(s)
Adaptación Fisiológica/inmunología , Citoplasma/inmunología , Macrófagos/inmunología , Mycobacterium tuberculosis/inmunología , Fagosomas/inmunología , Autofagia/inmunología , Línea Celular Tumoral , Células Cultivadas , Citoplasma/microbiología , Citoplasma/ultraestructura , Interacciones Huésped-Patógeno/inmunología , Humanos , Evasión Inmune/inmunología , Macrófagos/microbiología , Macrófagos/ultraestructura , Microscopía Confocal , Microscopía Electrónica de Transmisión , Mycobacterium tuberculosis/patogenicidad , Mycobacterium tuberculosis/fisiología , Fagocitosis/inmunología , Fagosomas/microbiología , Fagosomas/ultraestructura , Fosfolipasas A2 Citosólicas/inmunología , Fosfolipasas A2 Citosólicas/metabolismo , Vesículas Transportadoras/inmunología , Vesículas Transportadoras/microbiología , Vesículas Transportadoras/ultraestructura , Virulencia/inmunología
9.
Wei Sheng Wu Xue Bao ; 56(6): 911-21, 2016 Jun 04.
Artículo en Chino | MEDLINE | ID: mdl-29727152

RESUMEN

Outer membrane vesicles (OMVs) are vesicle-like structures, widely present in gram-negative bacteria and even in some gram-positive bacteria. OMVs contain biological active substances and their sizes are normally between 20 to 250 nm. Components of OMVs include lipopolysaccharide, outer membrane protein, phospholipids, DNA, as well as the periplasmic components produced during their formation. OMVs are non-viable vesicles that contain multiple antigenic proteins from the bacterial outer membrane, and are capable of activating the immune system, therefore they are considered to be potential vaccine candidates. Although outer membrane vesicles were discoverd more than 50 years ago, hardly any reports were published in China. In this review, we summarized the progress of outer membrane vesicles as a novel strategy for disease prevention and control in two aspects: the mechanism of the outer membrane vesicle-induced immune response and the advances in OMVs vaccine. This review provides some information on outer membrane vesicles as vaccine development.


Asunto(s)
Vacunas Bacterianas/inmunología , Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/prevención & control , Vesículas Transportadoras/inmunología , Vacunas de Subunidad/inmunología , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Vacunas Bacterianas/genética , Bacterias Gramnegativas/genética , Infecciones por Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/microbiología , Humanos , Vacunas de Subunidad/genética
10.
Fish Shellfish Immunol ; 42(1): 50-7, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25449706

RESUMEN

Infection of fish with the facultative intracellular bacterium Francisella noatunensis remains an unresolved problem for aquaculture industry worldwide as it is difficult to vaccinate against without using live attenuated vaccines. Outer membrane vesicles (OMVs) are biological structures shed by Gram-negative bacteria in response to various environmental stimuli. OMVs have successfully been used to vaccinate against both intracellular and extracellular pathogens, due to an ability to stimulate innate, cell-mediated and humoral immune responses. We show by using atomic force and electron microscopy that the fish pathogenic bacterium F. noatunensis subspecies noatunensis (F.n.n.) shed OMVs both in vitro into culture medium and in vivo in a zebrafish infection model. The main protein constituents of the OMV are IglC, PdpD and PdpA, all known Francisella virulence factors, in addition to the outer membrane protein FopA and the chaperonin GroEL, as analyzed by mass spectrometry. The vesicles, when used as a vaccine, reduced proliferation of the bacterium and protected zebrafish when subsequently challenged with a high dose of F.n.n. without causing adverse effects for the host. Also granulomatous responses were reduced in F.n.n.-challenged zebrafish after OMV vaccination. Taken together, the data support the possible use of OMVs as vaccines against francisellosis in fish.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/inmunología , Vacunas Bacterianas/farmacología , Francisella/inmunología , Infecciones por Bacterias Gramnegativas/prevención & control , Inmunidad Humoral/inmunología , Vesículas Transportadoras/inmunología , Vacunación/métodos , Animales , Cromatografía Liquida , Electroforesis en Gel de Poliacrilamida , Francisella/ultraestructura , Inmunidad Humoral/efectos de los fármacos , Estimación de Kaplan-Meier , Microscopía de Fuerza Atómica , Microscopía Electrónica de Transmisión , Espectrometría de Masas en Tándem , Vesículas Transportadoras/ultraestructura , Pez Cebra
11.
Sci Signal ; 7(339): ra79, 2014 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-25140054

RESUMEN

The traditional view of how intracellular effector proteins are recruited to the B cell antigen receptor (BCR) complex at the plasma membrane is based on the occurrence of direct protein-protein interactions, as exemplified by the recruitment of the tyrosine kinase Syk (spleen tyrosine kinase) to phosphorylated motifs in BCR signaling subunits. By contrast, the subcellular targeting of the cytosolic adaptor protein SLP-65 (Src homology 2 domain-containing leukocyte adaptor protein of 65 kD), which serves as a proximal Syk substrate, is unclear. We showed that SLP-65 activation required its association at vesicular compartments in resting B cells. A module of ~50 amino acid residues located at the amino terminus of SLP-65 anchored SLP-65 to the vesicles. Nuclear magnetic resonance spectroscopy showed that the SLP-65 amino terminus was structurally disordered in solution but could bind in a structured manner to noncharged lipid components of cellular membranes. Our finding that preformed vesicular signaling scaffolds are required for B cell activation indicates that vesicles may deliver preassembled signaling cargo to sites of BCR activation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Linfocitos B/metabolismo , Vesículas Transportadoras/química , Vesículas Transportadoras/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Linfocitos B/química , Membrana Celular/genética , Membrana Celular/inmunología , Membrana Celular/metabolismo , Humanos , Resonancia Magnética Nuclear Biomolecular , Estructura Terciaria de Proteína , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/inmunología , Receptores de Antígenos de Linfocitos B/metabolismo , Vesículas Transportadoras/genética , Vesículas Transportadoras/inmunología
12.
Proc Natl Acad Sci U S A ; 111(15): E1463-72, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24706780

RESUMEN

Recognition of phosphatidylserine (PS) lipids exposed on the extracellular leaflet of plasma membranes is implicated in both apoptotic cell removal and immune regulation. The PS receptor T cell immunoglobulin and mucin-domain-containing molecule 4 (Tim4) regulates T-cell immunity via phagocytosis of both apoptotic (high PS exposure) and nonapoptotic (intermediate PS exposure) activated T cells. The latter population must be removed at lower efficiency to sensitively control immune tolerance and memory cell population size, but the molecular basis for how Tim4 achieves this sensitivity is unknown. Using a combination of interfacial X-ray scattering, molecular dynamics simulations, and membrane binding assays, we demonstrate how Tim4 recognizes PS in the context of a lipid bilayer. Our data reveal that in addition to the known Ca(2+)-coordinated, single-PS binding pocket, Tim4 has four weaker sites of potential ionic interactions with PS lipids. This organization makes Tim4 sensitive to PS surface concentration in a manner capable of supporting differential recognition on the basis of PS exposure level. The structurally homologous, but functionally distinct, Tim1 and Tim3 are significantly less sensitive to PS surface density, likely reflecting the differences in immunological function between the Tim proteins. These results establish the potential for lipid membrane parameters, such as PS surface density, to play a critical role in facilitating selective recognition of PS-exposing cells. Furthermore, our multidisciplinary approach overcomes the difficulties associated with characterizing dynamic protein/membrane systems to reveal the molecular mechanisms underlying Tim4's recognition properties, and thereby provides an approach capable of providing atomic-level detail to uncover the nuances of protein/membrane interactions.


Asunto(s)
Inmunidad Celular/inmunología , Proteínas de la Membrana/inmunología , Modelos Moleculares , Fosfatidilserinas/inmunología , Conformación Proteica , Linfocitos T/inmunología , Animales , Receptor Celular 1 del Virus de la Hepatitis A , Receptor 2 Celular del Virus de la Hepatitis A , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Ratones , Modelos Inmunológicos , Simulación de Dinámica Molecular , Unión Proteica , Receptores Virales/inmunología , Dispersión de Radiación , Vesículas Transportadoras/inmunología , Triptófano/metabolismo
13.
PLoS One ; 9(2): e89121, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24586537

RESUMEN

Oral delivery of Gram positive bacteria, often derived from the genera Lactobacillus or Bifidobacterium, can modulate immune function. Although the exact mechanisms remain unclear, immunomodulatory effects may be elicited through the direct interaction of these bacteria with the intestinal epithelium or resident dendritic cell (DC) populations. We analyzed the immune activation properties of Lactobacilli and Bifidobacterium species and made the surprising observation that cellular responses in vitro were differentially influenced by the presence of serum, specifically the extracellular vesicle (EV) fraction. In contrast to the tested Lactobacilli species, tested Bifidobacterium species induce TLR2/6 activity which is inhibited by the presence of EVs. Using specific TLR ligands, EVs were found to enhance cellular TLR2/1 and TLR4 responses while TLR2/6 responses were suppressed. No effect could be observed on cellular TLR5 responses. We determined that EVs play a role in bacterial aggregation, suggesting that EVs interact with bacterial surfaces. EVs were found to slightly enhance DC phagocytosis of Bifidobacterium breve whereas phagocytosis of Lactobacillus rhamnosus was virtually absent upon serum EV depletion. DC uptake of a non-microbial substance (dextran) was not affected by the different serum fractions suggesting that EVs do not interfere with DC phagocytic capacity but rather modify the DC-microbe interaction. Depending on the microbe, combined effects of EVs on TLR activity and phagocytosis result in a differential proinflammatory DC cytokine release. Overall, these data suggest that EVs play a yet unrecognized role in host-microbe responses, not by interfering in recipient cellular responses but via attachment to, or scavenging of, microbe-associated molecular patterns. EVs can be found in any tissue or bodily fluid, therefore insights into EV-microbe interactions are important in understanding the mechanism of action of potential probiotics and gut immune homeostasis.


Asunto(s)
Bifidobacterium/fisiología , Interacciones Huésped-Patógeno , Fagocitosis , Receptor Toll-Like 2/metabolismo , Vesículas Transportadoras/metabolismo , Animales , Bifidobacterium/metabolismo , Citocinas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células HEK293 , Humanos , Ácido Láctico/biosíntesis , Ratones , Especificidad de la Especie , Receptor Toll-Like 6/metabolismo , Vesículas Transportadoras/inmunología , Vesículas Transportadoras/microbiología
14.
Nat Rev Rheumatol ; 10(6): 356-64, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24535546

RESUMEN

The discovery that submicron-sized extracellular vesicles (EVs) are generated by both prokaryotic and eukaryotic cells might have a profound effect on experimental and clinical sciences, and could pave the way for new strategies to combat various diseases. EVs are carriers of pathogen-associated and damage-associated molecular patterns, cytokines, autoantigens and tissue-degrading enzymes. In addition to a possible role in the pathogenesis of a number of inflammatory conditions, such as infections and autoimmune diseases, EVs, including microvesicles (also known as microparticles), exosomes and apoptotic vesicles, have therapeutic potential and might be used as biomarkers for inflammatory diseases. Therefore, molecular diagnostics and targeted therapy could benefit from expanding knowledge in the field. In this Review, we summarize important developments and propose that extracellular vesicles could be used as therapeutic vehicles and as targets for the treatment and prevention of inflammatory diseases.


Asunto(s)
Comunicación Celular/fisiología , Inflamación/fisiopatología , Vesículas Transportadoras/fisiología , Comunicación Celular/inmunología , Humanos , Inflamación/inmunología , Vesículas Transportadoras/inmunología
16.
Immunol Rev ; 251(1): 97-112, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23278743

RESUMEN

Accumulating evidence underscores the immune synapse (IS) of naive T cells as a site of intense vesicular trafficking. At variance with helper and cytolytic effectors, which use the IS as a secretory platform to deliver cytokines and/or lytic granules to their cellular targets, this process is exploited by naive T cells as a means to regulate the assembly and maintenance of the IS, on which productive signaling and cell activation crucially depend. We have recently identified a role of the intraflagellar transport (IFT) system, which is responsible for the assembly of the primary cilium, in the non-ciliated T-cell, where it controls IS assembly by promoting polarized T-cell receptor recycling. This unexpected finding not only provides new insight into the mechanisms of IS assembly but also strongly supports the notion that the IS and the primary cilium, which are both characterized by a specialized membrane domain highly enriched in receptors and signaling mediators, share architectural similarities and are homologous structures. Here, we review our current understanding of vesicular trafficking in the regulation of the assembly and maintenance of the naive T-cell IS and the primary cilium, with a focus on the IFT system.


Asunto(s)
Compartimento Celular/inmunología , Cilios/inmunología , Sinapsis Inmunológicas/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Vesículas Transportadoras/inmunología , Animales , Citocinas/inmunología , Citocinesis/inmunología , Citotoxicidad Inmunológica , Humanos , Transducción de Señal/inmunología
17.
J Bacteriol ; 195(6): 1120-32, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23264574

RESUMEN

Francisella spp. are highly infectious and virulent bacteria that cause the zoonotic disease tularemia. Knowledge is lacking for the virulence factors expressed by Francisella and how these factors are secreted and delivered to host cells. Gram-negative bacteria constitutively release outer membrane vesicles (OMV), which may function in the delivery of virulence factors to host cells. We identified growth conditions under which Francisella novicida produces abundant OMV. Purification of the vesicles revealed the presence of tube-shaped vesicles in addition to typical spherical OMV, and examination of whole bacteria revealed the presence of tubes extending out from the bacterial surface. Recently, both prokaryotic and eukaryotic cells have been shown to produce membrane-enclosed projections, termed nanotubes, which appear to function in cell-cell communication and the exchange of molecules. In contrast to these previously characterized structures, the F. novicida tubes are produced in liquid as well as on solid medium and are derived from the OM rather than the cytoplasmic membrane. The production of the OMV and tubes (OMV/T) by F. novicida was coordinately regulated and responsive to both growth medium and growth phase. Proteomic analysis of purified OMV/T identified known Francisella virulence factors among the constituent proteins, suggesting roles for the vesicles in pathogenesis. In support of this, production of OM tubes by F. novicida was stimulated during infection of macrophages and addition of purified OMV/T to macrophages elicited increased release of proinflammatory cytokines. Finally, vaccination with purified OMV/T protected mice from subsequent challenge with highly lethal doses of F. novicida.


Asunto(s)
Extensiones de la Superficie Celular/metabolismo , Francisella/metabolismo , Francisella/ultraestructura , Vesículas Transportadoras/metabolismo , Vesículas Transportadoras/ultraestructura , Animales , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas de la Membrana Bacteriana Externa/metabolismo , Vacunas Bacterianas/inmunología , Comunicación Celular , Extensiones de la Superficie Celular/inmunología , Extensiones de la Superficie Celular/ultraestructura , Medios de Cultivo , Citocinas/biosíntesis , Francisella/inmunología , Francisella/patogenicidad , Infecciones por Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/prevención & control , Macrófagos/microbiología , Ratones , Ratones Endogámicos BALB C , Proteómica , Vesículas Transportadoras/inmunología , Vacunación , Factores de Virulencia/metabolismo
18.
PLoS One ; 7(11): e50214, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23189190

RESUMEN

Outer membrane vesicles (OMVs) released by some gram-negative bacteria have been shown to exert immunomodulatory effects that favor the establishment of the infection. The aim of the present study was to assess the interaction of OMVs from Brucella abortus with human epithelial cells (HeLa) and monocytes (THP-1), and the potential immunomodulatory effects they may exert. Using confocal microscopy and flow cytometry, FITC-labeled OMVs were shown to be internalized by both cell types. Internalization was shown to be partially mediated by clathrin-mediated endocytosis. Pretreatment of THP-1 cells with Brucella OMVs inhibited some cytokine responses (TNF-α and IL-8) to E. coli LPS, Pam3Cys or flagellin (TLR4, TLR2 and TLR5 agonists, respectively). Similarly, pretreatment with Brucella OMVs inhibited the cytokine response of THP-1 cells to B. abortus infection. Treatment of THP-1 cells with OMVs during IFN-γ stimulation reduced significantly the inducing effect of this cytokine on MHC-II expression. OMVs induced a dose-dependent increase of ICAM-1 expression on THP-1 cells and an increased adhesion of these cells to human endothelial cells. The addition of OMVs to THP-1 cultures before the incubation with live B. abortus resulted in increased numbers of adhered and internalized bacteria as compared to cells not treated with OMVs. Overall, these results suggest that OMVs from B. abortus exert cellular effects that promote the internalization of these bacteria by human monocytes, but also downregulate the innate immune response of these cells to Brucella infection. These effects may favor the persistence of Brucella within host cells.


Asunto(s)
Brucella abortus/inmunología , Brucella abortus/metabolismo , Inmunidad Innata , Monocitos/inmunología , Fagocitosis/inmunología , Vesículas Transportadoras/metabolismo , Adhesión Bacteriana/inmunología , Moléculas de Adhesión Celular/metabolismo , Línea Celular , Clatrina/metabolismo , Citocinas/metabolismo , Endocitosis/fisiología , Células HeLa , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Interferón gamma/farmacología , Monocitos/efectos de los fármacos , Monocitos/microbiología , Receptores Toll-Like/agonistas , Receptores Toll-Like/metabolismo , Vesículas Transportadoras/inmunología
19.
Immunity ; 37(5): 854-66, 2012 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-23084031

RESUMEN

Immunological control of infections or tumors depends on the release of effector cytokines and polarized secretion of cytotoxic granules from T cells and natural killer cells. Here we show that the sorting receptor Sortilin controlled both processes. In murine Sortilin-deficient cytotoxic T lymphocytes, regulated secretion of granzyme A and cytotoxic killing was enhanced and correlated with increased vesicle-associated membrane protein 7 availability. In contrast, loss of Sortilin reduced the release of interferon-γ upon infections and in autoimmune colitis. Exit of interferon-γ from the Golgi apparatus required the presence of Sortilin. Furthermore, we tracked the transport route of interferon-γ beyond this Sortilin-dependent Golgi to early endosome step. In wild-type T cells, trafficking of interferon-γ from the endosomal sorting platform to the plasma membrane proceeded independently of recycling endosomes, and interferon-γ remained excluded from late endosomes. Our results suggest that Sortilin modulates systemic immune responses through exocytic sorting of immunological effector molecules.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Granzimas/metabolismo , Interferón gamma/metabolismo , Linfocitos T/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Antígenos CD4/inmunología , Antígenos CD4/metabolismo , Membrana Celular/inmunología , Membrana Celular/metabolismo , Colitis/inmunología , Colitis/metabolismo , Endosomas/inmunología , Endosomas/metabolismo , Exocitosis/inmunología , Aparato de Golgi/inmunología , Aparato de Golgi/metabolismo , Granzimas/inmunología , Interferón gamma/inmunología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ratones , Ratones Endogámicos C57BL , Transporte de Proteínas , Proteínas R-SNARE/inmunología , Proteínas R-SNARE/metabolismo , Linfocitos T/inmunología , Vesículas Transportadoras/inmunología , Vesículas Transportadoras/metabolismo
20.
Infect Immun ; 80(12): 4089-98, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22966047

RESUMEN

Campylobacter jejuni is the most prevalent cause of food-borne gastroenteritis in the developed world; however, the molecular basis of pathogenesis is unclear. Secretion of virulence factors is a key mechanism by which enteric bacterial pathogens interact with host cells to enhance survival and/or damage the host. However, C. jejuni lacks the virulence-associated secretion systems possessed by other enteric pathogens. Many bacterial pathogens utilize outer membrane vesicles (OMVs) for delivery of virulence factors into host cells. In the absence of prototypical virulence-associated secretion systems, OMVs could be an important alternative for the coordinated delivery of C. jejuni proteins into host cells. Proteomic analysis of C. jejuni 11168H OMVs identified 151 proteins, including periplasmic and outer membrane-associated proteins, but also many determinants known to be important in survival and pathogenesis, including the cytolethal distending toxin (CDT). C. jejuni OMVs contained 16 N-linked glycoproteins, indicating a delivery mechanism by which these periplasm-located yet immunogenic glycoproteins can interact with host cells. C. jejuni OMVs possess cytotoxic activity and induce a host immune response from T84 intestinal epithelial cells (IECs), which was not reduced by OMV pretreatment with proteinase K or polymyxin B prior to coincubation with IECs. Pretreatment of IECs with methyl-beta-cyclodextrin partially blocks OMV-induced host immune responses, indicating a role for lipid rafts in host cell plasma membranes during interactions with C. jejuni OMVs. OMVs isolated from a C. jejuni 11168H cdtA mutant induced interleukin-8 (IL-8) to the same extent as did wild-type OMVs, suggesting OMV induction of IL-8 is independent of CDT.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Campylobacter jejuni/patogenicidad , Células Epiteliales/microbiología , Interacciones Huésped-Patógeno , Intestinos/microbiología , Vesículas Transportadoras/fisiología , Animales , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/inmunología , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Células CACO-2 , Campylobacter jejuni/genética , Campylobacter jejuni/metabolismo , Línea Celular Tumoral , Células Epiteliales/inmunología , Humanos , Interleucina-8/metabolismo , Intestinos/citología , Intestinos/inmunología , Microscopía Electrónica de Transmisión , Proteómica , Vesículas Transportadoras/inmunología , Vesículas Transportadoras/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...