Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Cell Biochem ; 112(10): 2891-901, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21618593

RESUMEN

Periodontal ligament fibroblasts (PLF) sense and respond to mechanical stimuli and participate in alveolar bone resorption during orthodontic treatments. This study examined how PLF influence osteoclastogenesis from bone marrow-derived macrophages (BMM) after application of tension or compression force. We also investigated whether lymphocytes could be a primary stimulator of osteoclastic activation during alveolar bone remodeling. We found that mechanical forces inhibited osteoclastic differentiation from BMM in co-cultures with PLF, with PLF producing predominantly osteoprotegerin (OPG) rather than receptor activator of nuclear factor-kappaB (NF-κB) ligand (RANKL). In particular, PLF increased the expression of tumor necrosis factor (TNF)-α in response to compression. Additional experiments showed the presence of CD4- and B220-positive cells with a subsequent increase in tartrate-resistant acid phosphatase (TRAP)-positive cells and RANKL expression only at the compression side of the force-subjected periodontal tissues. Exogenous TNF-α increased the number of TRAP-positive cells and pit formation in the co-cultures of BMM with Jurkat, but not with BJAB cells and this effect was almost completely inhibited by antibodies to TNF-α or TNF receptor. Collectively, the current findings suggest that PLF secrete relatively higher levels of TNF-α at the compression side than at the tension side and this imbalance leads to RANKL expression by activating CD4+ T cells, thereby facilitating bone resorption during orthodontic tooth movement.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Fibroblastos/metabolismo , Osteoclastos/citología , Osteoclastos/metabolismo , Osteogénesis/fisiología , Ligamento Periodontal/citología , Estrés Mecánico , Factor de Necrosis Tumoral alfa/metabolismo , Adulto , Animales , Western Blotting , Diferenciación Celular , Células Cultivadas , Citocinas/metabolismo , Fibroblastos/fisiología , Líquido del Surco Gingival , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Adulto Joven
2.
Microbiol Immunol ; 55(3): 199-210, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21223367

RESUMEN

FimA of Porphyromonas gingivalis, a major pathogen in periodontitis, is known to be closely related to the virulence of these bacteria and has been suggested as a candidate for development of a vaccine against periodontal disease. In order to develop a passive immunization method for inhibiting the establishment of periodontal disease, B hybridoma clones 123-123-10 and 256-265-9, which produce monoclonal antibodies (Mabs) specific to purified fimbriae, were established. Both mAbs reacted with the conformational epitopes displayed by partially dissociated oligomers of FimA, but not with the 43 kDa FimA monomer. Gene sequence analyses of full-length cDNAs encoding heavy and light chain immunoglobulins enabled classification of the genes of mAb 123-123-10 as members of the mVh II (A) and mVκ I subgroups, and those of mAb 256-265-9 as members of the mVh III (D) and mVκ I subgroups. More importantly, 50 ng/mL of antibodies purified from the culture supernatant of antibody gene-transfected CHO cells inhibited, by approximately 50%, binding of P. gingivalis to saliva-coated hydroxyapatite bead surfaces. It is expected that these mAbs could be used as a basis for passive immunization against P. gingivalis-mediated periodontitis.


Asunto(s)
Anticuerpos Monoclonales/genética , Proteínas Fimbrias/genética , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/genética , Porphyromonas gingivalis/genética , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos/inmunología , Adhesión Bacteriana/genética , Secuencia de Bases , Células CHO , Cricetinae , Cricetulus , Proteínas Fimbrias/inmunología , Hibridomas/inmunología , Ratones , Datos de Secuencia Molecular , Periodontitis/inmunología , Periodontitis/microbiología , Porphyromonas gingivalis/inmunología , Unión Proteica/inmunología
3.
BMC Immunol ; 11: 65, 2010 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-21194475

RESUMEN

BACKGROUND: Immunization with the spike protein (S) of severe acute respiratory syndrome (SARS)-coronavirus (CoV) in mice is known to produce neutralizing antibodies and to prevent the infection caused by SARS-CoV. Polyethylenimine 25K (PEI) is a cationic polymer which effectively delivers the plasmid DNA. RESULTS: In the present study, the immune responses of BALB/c mice immunized via intranasal (i.n.) route with SARS DNA vaccine (pci-S) in a PEI/pci-S complex form have been examined. The size of the PEI/pci-S nanoparticles appeared to be around 194.7 ± 99.3 nm, and the expression of the S mRNA and protein was confirmed in vitro. The mice immunized with i.n. PEI/pci-S nanoparticles produced significantly (P < 0.05) higher S-specific IgG1 in the sera and mucosal secretory IgA in the lung wash than those in mice treated with pci-S alone. Compared to those in mice challenged with pci-S alone, the number of B220+ cells found in PEI/pci-S vaccinated mice was elevated. Co-stimulatory molecules (CD80 and CD86) and class II major histocompatibility complex molecules (I-Ad) were increased on CD11c+ dendritic cells in cervical lymph node from the mice after PEI/pci-S vaccination. The percentage of IFN-γ-, TNF-α- and IL-2-producing cells were higher in PEI/pci-S vaccinated mice than in control mice. CONCLUSION: These results showed that intranasal immunization with PEI/pci-S nanoparticles induce antigen specific humoral and cellular immune responses.


Asunto(s)
ADN/inmunología , Inmunidad/inmunología , Inmunización/métodos , Glicoproteínas de Membrana/inmunología , Nanopartículas/química , Plásmidos/inmunología , Polietileneimina/farmacología , Proteínas del Envoltorio Viral/inmunología , Administración Intranasal , Animales , Anticuerpos Antivirales/inmunología , Formación de Anticuerpos/efectos de los fármacos , Antígenos de Superficie/inmunología , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/virología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , ADN/administración & dosificación , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/virología , Epítopos/inmunología , Inmunidad/efectos de los fármacos , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/inmunología , Inmunidad Humoral/efectos de los fármacos , Inmunidad Humoral/inmunología , Ratones , Ratones Endogámicos BALB C , Plásmidos/administración & dosificación , Glicoproteína de la Espiga del Coronavirus , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/virología
4.
Mol Biotechnol ; 41(2): 157-64, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18807220

RESUMEN

The gram-negative anaerobic oral bacterium Porphyromonas gingivalis initiates periodontal disease through fimbrial attachment to saliva-coated oral surfaces. To study the effects of immunomodulation on enhancement of subunit vaccination, the expression in E. coli and immunogenicity of P. gingivalis fimbrial protein (FimA) linked to the C-terminus of the cholera toxin B subunit (CTB) were investigated. Complementary DNAs encoding the P. gingivalis 381 fimbrillin protein sequence FimA1 (amino acid residues 1-200) and FimA2 (amino acid residues 201-337) were cloned into an E. coli expression vector downstream of a cDNA fragment encoding the immunostimulatory CTB. CTB-FimA1 and CTB-FimA2 fusion proteins synthesized in E. coli BL21 (DE3) cells were purified under denaturing conditions by Ni2+-NTA affinity column chromatography. Renaturation of the CTB-FimA1 and CTB-FimA2 fusion proteins, permitted identification of CTB-FimA pentamers and restored CTB binding activity to GM1-ganglioside to provide a biologically active CTB-FimA fusion protein. Mice orally inoculated with purified CTB-FimA1 or CTB-FimA2 fusion proteins generated measurable FimA1 and FimA2 IgG antibody titers, while no serum fimbrial IgG antibodies were detected when mice were inoculated with FimA1 or FimA2 proteins alone. Immunoblot analysis confirmed that sera from mice immunized with CTB linked to FimA1 or FimA2 contained antibodies specific for P. gingivalis fimbrial proteins. In addition, mice immunized with FimA2 or CTB-FimA2 generated measurable intestinal IgA titers indicating the presence of fimbrial antibody class switching. Further, mice orally immunized with CTB-FimA1 generated higher IgA antibody titers than mice inoculated with FimA1 alone. The experimental data show that the immunostimulatory molecule CTB enhances B cell-mediated immunity against linked P. gingivalis FimA fusion proteins, in comparison to immunization with FimA protein alone. Thus, linkage of CTB to P. gingivalis fimbrial antigens can increase subunit vaccine immunogenicity to provide enhanced protection against periodontal disease.


Asunto(s)
Toxina del Cólera/inmunología , Proteínas Fimbrias/inmunología , Porphyromonas gingivalis/genética , Proteínas Recombinantes de Fusión/inmunología , Administración Oral , Anticuerpos Antibacterianos/análisis , Anticuerpos Antibacterianos/sangre , Vacunas Bacterianas/genética , Vacunas Bacterianas/inmunología , Vacunas Bacterianas/metabolismo , Infecciones por Bacteroidaceae/inmunología , Infecciones por Bacteroidaceae/microbiología , Toxina del Cólera/genética , Toxina del Cólera/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Heces/química , Proteínas Fimbrias/genética , Proteínas Fimbrias/metabolismo , Inmunización , Inmunoglobulina A/análisis , Inmunoglobulina G/sangre , Porphyromonas gingivalis/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
5.
Int Immunopharmacol ; 43: 91-98, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27987467

RESUMEN

Studies have been focused on natural products with antibacterial and anti-inflammatory activities, such as fucoidan. Many in vivo studies have evaluated the effect of fucoidan on tumor growth, diabetes, obesity, ischemia reperfusion, and oxidative stress. However, the effects of fucoidan on bacteria-induced gingival inflammation and periodontitis have not been reported. We previously characterized the anti-inflammatory effect of fucoidan in vitro. Here, we confirmed the anti-inflammatory activity of fucoidan in a macrophage cell line in terms of its inhibition of the expression of inflammatory mediators and pro-inflammatory cytokines. Additionally, we confirmed the ability of fucoidan to inhibit gingival inflammation, expression of pro-inflammatory cytokines, and neutrophil recruitment in the gingival tissue of mice injected with LPS prepared from P. gingivalis. Interestingly, however, fucoidan did not inhibit the expression of pro-inflammatory cytokines in a P. gingivalis-infected mouse model of periodontitis. Additionally, fucoidan treatment did not lead to clearance of P. gingivalis or improvement of P. gingivalis infection-mediated bone loss in the periodontitis model. We conclude that fucoidan exerts anti-inflammatory effects in vitro and in vivo, together with a limited antibacterial effect in vivo.


Asunto(s)
Antiinflamatorios/uso terapéutico , Infecciones por Bacteroidaceae/tratamiento farmacológico , Encía/efectos de los fármacos , Gingivitis/tratamiento farmacológico , Neutrófilos/efectos de los fármacos , Periodontitis/tratamiento farmacológico , Polisacáridos/uso terapéutico , Porphyromonas gingivalis/inmunología , Animales , Movimiento Celular/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encía/inmunología , Encía/microbiología , Gingivitis/inducido químicamente , Humanos , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos , Neutrófilos/inmunología , Células RAW 264.7
6.
J Biotechnol ; 96(3): 205-11, 2002 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-12044549

RESUMEN

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a glycoprotein that stimulates the production of granulocytes, macrophages, and white blood cells. Secretion of human GM-CSF from cell suspension cultures of genetically modified tobacco has been facilitated using natural mammalian leader sequences. At the mid-exponential growth phase (day 4 after the initiation of cell suspension culture), GM-CSF was detected in the medium at a maximum concentration of 180 microg l(-1). However, the secreted GM-CSF was unstable in the medium, and rapidly degraded after day 5. In order to stabilize the secreted GM-CSF, three stabilizing polymers were tested, polyethylene glycol, polyvinylpyrrolidone and gelatin. Gelatin was the most effective in stabilizing the secreted GM-CSF. Following the addition of 5% (w/v) gelatin, the maximum GM-CSF concentration reached 783 microg l(-1), a 4.6-fold increase over control.


Asunto(s)
Gelatina/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Nicotiana/citología , Nicotiana/metabolismo , Polietilenglicoles/metabolismo , Povidona/metabolismo , Agrobacterium tumefaciens/química , Agrobacterium tumefaciens/genética , División Celular/fisiología , ADN Complementario/genética , Endopeptidasas/análisis , Endopeptidasas/metabolismo , Excipientes/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Nicotiana/genética , Nicotiana/microbiología , Transformación Bacteriana
7.
Immune Netw ; 13(4): 157-62, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24009543

RESUMEN

Application of vaccine materials through oral mucosal route confers great economical advantage in animal farming industry due to much less vaccination cost compared with that of injection-based vaccination. In particular, oral administration of recombinant protein antigen against foot-and-mouth disease virus (FMDV) is an ideal strategy because it is safe from FMDV transmission during vaccine production and can induce antigen-specific immune response in mucosal compartments, where FMDV infection has been initiated, which is hardly achievable through parenteral immunization. Given that effective delivery of vaccine materials into immune inductive sites is prerequisite for effective oral mucosal vaccination, M cell-targeting strategy is crucial in successful vaccination since M cells are main gateway for luminal antigen influx into mucosal lymphoid tissue. Here, we applied previously identified M cell-targeting ligand Co1 to VP1 of FMDV in order to test the possible oral mucosal vaccination against FMDV infection. M cell-targeting ligand Co1-conjugated VP1 interacted efficiently with M cells of Peyer's patch. In addition, oral administration of ligand-conjugated VP1 enhanced the induction of VP1-specific IgG and IgA responses in systemic and mucosal compartments, respectively, in comparison with those from oral administration of VP1 alone. In addition, the enhanced VP1-specific immune response was found to be due to antigen-specific Th2-type cytokine production. Collectively, it is suggested that the M cell-targeting strategy could be applied to develop efficient oral mucosal vaccine against FMDV infection.

8.
Biomaterials ; 32(26): 6254-63, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21620470

RESUMEN

We report the fabrication of a one-pot antigen system that delivers antigen to dendritic cells (DCs) and tracks their in vivo migration after injection. Multifunctional polymer nanoparticles containing ovalbumin protein, magnetic resonance imaging contrast agents (iron oxide nanoparticles), and near-infrared fluorophores (indocyanine green, ICG), MPN-OVA, were prepared using a double emulsion method. The MPN-OVA was efficiently taken up by the dendritic cells and subsequently localized in the lysosome. Flow cytometry analysis revealed an increase in the uptake of OVA antigen by MPN-OVA at 37 °C, when compared with soluble OVA protein. We found that MPN-OVA had no effect on DC surface expression of MHC class I, costimulatory (CD80, CD86) or adhesion (CD54) molecules or the ability of DCs to mature in response to LPS. Following the uptake of MPN-OVA, exogenous OVA antigen was delivered to the cytoplasm, and OVA peptides were presented on MHC class I molecules, which enhanced OVA antigen-specific cross-presentation to OT-1 T cells and CD8OVA1.3 T cell hybridoma in vitro. The immunization of mice with MPN-OVA-treated DCs induced OVA-specific CTL activity in draining lymph nodes. The presence of MPN allowed us to monitor the migration of DCs via lymphatic drainage using NIR fluorescence imaging, and the homing of DCs into the lymph nodes was imaged using MRI. This system has potential for use as a delivery system to induce T cell priming and to image DC-based immunotherapies.


Asunto(s)
Células Dendríticas/inmunología , Nanopartículas/química , Animales , Línea Celular , Proliferación Celular , Supervivencia Celular/fisiología , Células Cultivadas , Citometría de Flujo , Verde de Indocianina/química , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ovalbúmina/química , Ovalbúmina/inmunología , Polímeros/química , Linfocitos T/citología , Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA