Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 117(22): 12375-12386, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32424080

RESUMEN

Tooth decay (dental caries) is a widespread human disease caused by microbial biofilms. Streptococcus mutans, a biofilm-former, has been consistently associated with severe childhood caries; however, how this bacterium is spatially organized with other microorganisms in the oral cavity to promote disease remains unknown. Using intact biofilms formed on teeth of toddlers affected by caries, we discovered a unique 3D rotund-shaped architecture composed of multiple species precisely arranged in a corona-like structure with an inner core of S. mutans encompassed by outer layers of other bacteria. This architecture creates localized regions of acidic pH and acute enamel demineralization (caries) in a mixed-species biofilm model on human teeth, suggesting this highly ordered community as the causative agent. Notably, the construction of this architecture was found to be an active process initiated by production of an extracellular scaffold by S. mutans that assembles the corona cell arrangement, encapsulating the pathogen core. In addition, this spatial patterning creates a protective barrier against antimicrobials while increasing bacterial acid fitness associated with the disease-causing state. Our data reveal a precise biogeography in a polymicrobial community associated with human caries that can modulate the pathogen positioning and virulence potential in situ, indicating that micron-scale spatial structure of the microbiome may mediate the function and outcome of host-pathogen interactions.


Asunto(s)
Caries Dental/microbiología , Microbiota , Boca/microbiología , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Fenómenos Fisiológicos Bacterianos , Biopelículas , Niño , Preescolar , Femenino , Humanos , Masculino , Streptococcus mutans/genética , Streptococcus mutans/aislamiento & purificación , Streptococcus mutans/fisiología
2.
PLoS Pathog ; 10(2): e1004647, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25679217

RESUMEN

Signaling via pattern recognition receptors (PRRs) expressed on professional antigen presenting cells, such as dendritic cells (DCs), is crucial to the fate of engulfed microbes. Among the many PRRs expressed by DCs are Toll-like receptors (TLRs) and C-type lectins such as DC-SIGN. DC-SIGN is targeted by several major human pathogens for immune-evasion, although its role in intracellular routing of pathogens to autophagosomes is poorly understood. Here we examined the role of DC-SIGN and TLRs in evasion of autophagy and survival of Porphyromonas gingivalis in human monocyte-derived DCs (MoDCs). We employed a panel of P. gingivalis isogenic fimbriae deficient strains with defined defects in Mfa-1 fimbriae, a DC-SIGN ligand, and FimA fimbriae, a TLR2 agonist. Our results show that DC-SIGN dependent uptake of Mfa1+P. gingivalis strains by MoDCs resulted in lower intracellular killing and higher intracellular content of P. gingivalis. Moreover, Mfa1+P. gingivalis was mostly contained within single membrane vesicles, where it survived intracellularly. Survival was decreased by activation of TLR2 and/or autophagy. Mfa1+P. gingivalis strain did not induce significant levels of Rab5, LC3-II, and LAMP1. In contrast, P. gingivalis uptake through a DC-SIGN independent manner was associated with early endosomal routing through Rab5, increased LC3-II and LAMP-1, as well as the formation of double membrane intracellular phagophores, a characteristic feature of autophagy. These results suggest that selective engagement of DC-SIGN by Mfa-1+P. gingivalis promotes evasion of antibacterial autophagy and lysosome fusion, resulting in intracellular persistence in myeloid DCs; however TLR2 activation can overcome autophagy evasion and pathogen persistence in DCs.


Asunto(s)
Autofagia/inmunología , Moléculas de Adhesión Celular/metabolismo , Células Dendríticas/metabolismo , Lectinas Tipo C/metabolismo , Células Mieloides/metabolismo , Porphyromonas gingivalis/metabolismo , Receptores de Superficie Celular/metabolismo , Receptor Toll-Like 2/metabolismo , Dendritas/ultraestructura , Células Dendríticas/inmunología , Células Dendríticas/ultraestructura , Fimbrias Bacterianas , Humanos , Espacio Intracelular/inmunología , Espacio Intracelular/metabolismo , Monocitos/inmunología , Monocitos/ultraestructura , Células Mieloides/inmunología , Receptor Toll-Like 2/inmunología
3.
Infect Immun ; 82(1): 101-11, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24126519

RESUMEN

Several intracellular pathogens, including a key etiological agent of chronic periodontitis, Porphyromonas gingivalis, infect blood myeloid dendritic cells (mDCs). This infection results in pathogen dissemination to distant inflammatory sites (i.e., pathogen trafficking). The alteration in chemokine-chemokine receptor expression that contributes to this pathogen trafficking function, particularly toward sites of neovascularization in humans, is unclear. To investigate this, we utilized human monocyte-derived DCs (MoDCs) and primary endothelial cells in vitro, combined with ex vivo-isolated blood mDCs and serum from chronic periodontitis subjects and healthy controls. Our results, using conditional fimbria mutants of P. gingivalis, show that P. gingivalis infection of MoDCs induces an angiogenic migratory profile. This profile is enhanced by expression of DC-SIGN on MoDCs and minor mfa-1 fimbriae on P. gingivalis and is evidenced by robust upregulation of CXCR4, but not secondary lymphoid organ (SLO)-homing CCR7. This disruption of SLO-homing capacity in response to respective chemokines closely matches surface expression of CXCR4 and CCR7 and is consistent with directed MoDC migration through an endothelial monolayer. Ex vivo-isolated mDCs from the blood of chronic periodontitis subjects, but not healthy controls, expressed a similar migratory profile; moreover, sera from chronic periodontitis subjects expressed elevated levels of CXCL12. Overall, we conclude that P. gingivalis actively "commandeers" DCs by reprogramming the chemokine receptor profile, thus disrupting SLO homing, while driving migration toward inflammatory vascular sites.


Asunto(s)
Infecciones por Bacteroidaceae/metabolismo , Movimiento Celular/fisiología , Periodontitis Crónica/metabolismo , Células Dendríticas/microbiología , Células Mieloides/microbiología , Porphyromonas gingivalis/fisiología , Receptores de Quimiocina/metabolismo , Infecciones por Bacteroidaceae/inmunología , Infecciones por Bacteroidaceae/microbiología , Estudios de Casos y Controles , Moléculas de Adhesión Celular/metabolismo , Quimiocina CXCL12/metabolismo , Quimiotaxis/fisiología , Periodontitis Crónica/inmunología , Periodontitis Crónica/microbiología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Endoteliales/metabolismo , Fimbrias Bacterianas/genética , Fimbrias Bacterianas/fisiología , Humanos , Lectinas Tipo C/metabolismo , Lipopolisacáridos/farmacología , Células Mieloides/efectos de los fármacos , Células Mieloides/inmunología , Células Mieloides/metabolismo , Neovascularización Patológica/microbiología , Fenotipo , Receptores CCR7/metabolismo , Receptores CXCR4/metabolismo , Receptores de Superficie Celular/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
4.
J Leukoc Biol ; 94(2): 281-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23729500

RESUMEN

Maintenance of blood DC homeostasis is essential to preventing autoimmunity while controlling chronic infection. However, the ability of bacteremic pathogens to directly regulate blood DC homeostasis has not been defined. One such bacteremic pathogen, Porphyromonas gingivalis, is shown by our group to survive within mDCs under aerobic conditions and therein, metastasize from its oral mucosal niche. This is accompanied by expansion of the blood mDC pool in vivo, independently of canonical DC poietins. We presently know little of how this bacteremic pathogen causes blood DC expansion and the pathophysiological significance. This work shows that optimum differentiation of MoDCs from primary human monocytes, with or without GM-CSF/IL-4, is dependent on infection with P. gingivalis strains expressing the DC-SIGN ligand mfa-1. DC differentiation is lost when DC-SIGN is blocked with its ligand HIV gp120 or knocked out by siRNA gene silencing. Thus, we have identified a novel, noncanonical pathway of DC differentiation. We term these PDDCs and show that PDDCs are bona fide DCs, based on phenotype and phagocytic activity when immature and the ability to up-regulate accessory molecules and stimulate allo-CD4(+) T cell proliferation when matured. The latter is dependent on the P. gingivalis strain used to initially "educate" PDDCs. Moreover, we show that P. gingivalis-infected, conventional MoDCs become resistant to apoptosis and inflammatory pyroptosis, as determined by levels of Annexin V and caspase-8, -3/7, and -1. Taken together, we provide new insights into how a relatively asymptomatic bacteremia may influence immune homeostasis and promote chronic inflammation.


Asunto(s)
Bacteriemia/inmunología , Proteínas Bacterianas/inmunología , Moléculas de Adhesión Celular/inmunología , Células Dendríticas/patología , Proteínas Fimbrias/inmunología , Lectinas Tipo C/inmunología , Porphyromonas gingivalis/fisiología , Receptores de Superficie Celular/inmunología , Aerobiosis , Anexina A5/inmunología , Apoptosis , Linfocitos T CD4-Positivos/inmunología , Caspasas/fisiología , Moléculas de Adhesión Celular/antagonistas & inhibidores , Moléculas de Adhesión Celular/genética , Diferenciación Celular , Supervivencia Celular , Células Cultivadas/inmunología , Células Cultivadas/patología , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Proteínas Fimbrias/deficiencia , Proteínas Fimbrias/genética , Fimbrias Bacterianas/inmunología , Proteína gp120 de Envoltorio del VIH/farmacología , Homeostasis , Interacciones Huésped-Patógeno/inmunología , Humanos , Lectinas Tipo C/antagonistas & inhibidores , Lectinas Tipo C/genética , Monocitos/citología , Mucosa Bucal/inmunología , Mucosa Bucal/microbiología , Fagocitosis , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/inmunología , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/genética
5.
J Immunol ; 189(6): 3178-87, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22891282

RESUMEN

The low-grade oral infection chronic periodontitis (CP) has been implicated in coronary artery disease risk, but the mechanisms are unclear. In this study, a pathophysiological role for blood dendritic cells (DCs) in systemic dissemination of oral mucosal pathogens to atherosclerotic plaques was investigated in humans. The frequency and microbiome of CD19(-)BDCA-1(+)DC-SIGN(+) blood myeloid DCs (mDCs) were analyzed in CP subjects with or without existing acute coronary syndrome and in healthy controls. FACS analysis revealed a significant increase in blood mDCs in the following order: healthy controls < CP < acute coronary syndrome/CP. Analysis of the blood mDC microbiome by 16S rDNA sequencing showed Porphyromonas gingivalis and other species, including (cultivable) Burkholderia cepacia. The mDC carriage rate with P. gingivalis correlated with oral carriage rate and with serologic exposure to P. gingivalis in CP subjects. Intervention (local debridement) to elicit a bacteremia increased the mDC carriage rate and frequency in vivo. In vitro studies established that P. gingivalis enhanced by 28% the differentiation of monocytes into immature mDCs; moreover, mDCs secreted high levels of matrix metalloproteinase-9 and upregulated C1q, heat shock protein 60, heat shock protein 70, CCR2, and CXCL16 transcripts in response to P. gingivalis in a fimbriae-dependent manner. Moreover, the survival of the anaerobe P. gingivalis under aerobic conditions was enhanced when within mDCs. Immunofluorescence analysis of oral mucosa and atherosclerotic plaques demonstrate infiltration with mDCs, colocalized with P. gingivalis. Our results suggest a role for blood mDCs in harboring and disseminating pathogens from oral mucosa to atherosclerosis plaques, which may provide key signals for mDC differentiation and atherogenic conversion.


Asunto(s)
Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Placa Aterosclerótica/inmunología , Placa Aterosclerótica/microbiología , Adulto , Anciano , Anciano de 80 o más Años , Infecciones por Bacteroidaceae/sangre , Infecciones por Bacteroidaceae/inmunología , Infecciones por Bacteroidaceae/microbiología , Infecciones por Burkholderia/sangre , Infecciones por Burkholderia/inmunología , Infecciones por Burkholderia/microbiología , Portador Sano/sangre , Portador Sano/inmunología , Portador Sano/microbiología , Enfermedad Crónica , Células Dendríticas/patología , Femenino , Humanos , Inmunofenotipificación , Masculino , Persona de Mediana Edad , Monocitos/inmunología , Monocitos/microbiología , Monocitos/patología , Mucosa Bucal/inmunología , Mucosa Bucal/microbiología , Mucosa Bucal/patología , Células Mieloides/inmunología , Células Mieloides/microbiología , Células Mieloides/patología , Periodontitis , Placa Aterosclerótica/sangre , Porphyromonas gingivalis
6.
J Bacteriol ; 192(16): 4103-10, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20562309

RESUMEN

We recently reported that the oral mucosal pathogen Porphyromonas gingivalis, through its 67-kDa Mfa1 (minor) fimbria, targets the C-type lectin receptor DC-SIGN for invasion and persistence within human monocyte-derived dendritic cells (DCs). The DCs respond by inducing an immunosuppressive and Th2-biased CD4(+) T-cell response. We have now purified the native minor fimbria by ion-exchange chromatography and sequenced the fimbria by tandem mass spectrometry (MS/MS), confirming its identity and revealing two putative N-glycosylation motifs as well as numerous putative O-glycosylation sites. We further show that the minor fimbria is glycosylated by ProQ staining and that glycosylation is partially removed by treatment with beta(1-4)-galactosidase, but not by classic N- and O-linked deglycosidases. Further monosaccharide analysis by gas chromatography-mass spectrometry (GC-MS) confirmed that the minor fimbria contains the DC-SIGN-targeting carbohydrates fucose (1.35 nmol/mg), mannose (2.68 nmol/mg), N-acetylglucosamine (2.27 nmol/mg), and N-acetylgalactosamine (0.652 nmol/mg). Analysis by transmission electron microscopy revealed that the minor fimbria forms fibers approximately 200 nm in length that could be involved in targeting or cross-linking DC-SIGN. These findings shed further light on molecular mechanisms of invasion and immunosuppression by this unique mucosal pathogen.


Asunto(s)
Proteínas Bacterianas/química , Moléculas de Adhesión Celular/metabolismo , Proteínas Fimbrias/química , Glicoproteínas/química , Lectinas Tipo C/metabolismo , Porphyromonas gingivalis/química , Receptores de Superficie Celular/metabolismo , Factores de Virulencia/química , Acetilgalactosamina/análisis , Acetilglucosamina/análisis , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/aislamiento & purificación , Proteínas Bacterianas/metabolismo , Proteínas Fimbrias/genética , Proteínas Fimbrias/aislamiento & purificación , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/ultraestructura , Fucosa/análisis , Cromatografía de Gases y Espectrometría de Masas , Glicoproteínas/genética , Glicoproteínas/aislamiento & purificación , Glicoproteínas/metabolismo , Manosa/análisis , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Peso Molecular , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/ultraestructura , Unión Proteica , Análisis de Secuencia de Proteína , Espectrometría de Masas en Tándem , Factores de Virulencia/genética , Factores de Virulencia/aislamiento & purificación , Factores de Virulencia/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...