Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Clin Periodontol ; 44(5): 472-483, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28207941

RESUMO

AIM: We have previously shown that the secreted glycoprotein milk fat globule epidermal growth factor 8 (MFG-E8) has anti-inflammatory and anti-osteoclastogenic properties. Our objective was to investigate the potential of MFG-E8 as a diagnostic or therapeutic agent in periodontitis. MATERIALS AND METHODS: Periodontitis was induced in non-human primates (NHPs) by placing ligatures around posterior teeth on both halves of the mandible for a split-mouth design: one side was treated with MFG-E8-Fc and the other with Fc control. Disease was assessed by clinical periodontal examinations, radiographic analysis of bone loss, and analysis of cytokine mRNA expression in gingival biopsy samples. Gingival crevicular fluid (GCF) was collected from human healthy volunteers or subjects with gingivitis, chronic moderate periodontitis, or chronic severe periodontitis. Additionally, GCF was collected from a subset of severe periodontitis patients following scaling and root planing (SRP) and after pocket reduction surgery. GCF was analysed to quantify MFG-E8 and periodontitis-relevant cytokines using multiplex assays. RESULTS: In NHPs, sites treated with MFG-E8-Fc exhibited significantly less ligature-induced periodontal inflammation and bone loss than Fc control-treated sites. In humans, the GCF levels of MFG-E8 were significantly higher in health than in periodontitis, whereas the reverse was true for the proinflammatory cytokines tested. Consistently, MFG-E8 was elevated in GCF after both non-surgical (SRP) and surgical periodontal treatment of periodontitis patients. CONCLUSION: MFG-E8 is, in principle, a novel therapeutic agent and biomarker of periodontitis.


Assuntos
Antígenos de Superfície/uso terapêutico , Periodontite Crônica/diagnóstico , Periodontite Crônica/terapia , Líquido do Sulco Gengival/metabolismo , Proteínas do Leite/uso terapêutico , Animais , Antígenos de Superfície/metabolismo , Biomarcadores/metabolismo , Periodontite Crônica/metabolismo , Modelos Animais de Doenças , Feminino , Gengivite/diagnóstico , Gengivite/metabolismo , Gengivite/terapia , Humanos , Macaca fascicularis , Proteínas do Leite/metabolismo
2.
J Immunol ; 192(12): 6020-7, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24808362

RESUMO

Chronic periodontitis is induced by a dysbiotic microbiota and leads to inflammatory destruction of tooth-supporting connective tissue and bone. The third component of complement, C3, is a point of convergence of distinct complement activation mechanisms, but its involvement in periodontitis was not previously addressed. We investigated this question using two animal species models, namely, C3-deficient or wild-type mice and nonhuman primates (NHPs) locally treated with a potent C3 inhibitor (the compstatin analog Cp40) or an inactive peptide control. In mice, C3 was required for maximal periodontal inflammation and bone loss, and for the sustenance of the dysbiotic microbiota. The effect of C3 on the microbiota was therefore different from that reported for the C5a receptor, which is required for the initial induction of dysbiosis. C3-dependent bone loss was demonstrated in distinct models, including Porphyromonas gingivalis-induced periodontitis, ligature-induced periodontitis, and aging-associated periodontitis. Importantly, local treatment of NHPs with Cp40 inhibited ligature-induced periodontal inflammation and bone loss, which correlated with lower gingival crevicular fluid levels of proinflammatory mediators (e.g., IL-17 and RANKL) and decreased osteoclastogenesis in bone biopsy specimens, as compared with control treatment. To our knowledge, this is the first time, for any disease, that complement inhibition in NHPs was shown to inhibit inflammatory processes that lead to osteoclastogenesis and bone loss. These data strongly support the feasibility of C3-targeted intervention for the treatment of human periodontitis.


Assuntos
Infecções por Bacteroidaceae , Reabsorção Óssea , Complemento C3 , Periodontite , Porphyromonas gingivalis/imunologia , Piridonas/farmacologia , Animais , Infecções por Bacteroidaceae/tratamento farmacológico , Infecções por Bacteroidaceae/genética , Infecções por Bacteroidaceae/imunologia , Infecções por Bacteroidaceae/patologia , Reabsorção Óssea/tratamento farmacológico , Reabsorção Óssea/genética , Reabsorção Óssea/imunologia , Reabsorção Óssea/patologia , Complemento C3/antagonistas & inibidores , Complemento C3/genética , Complemento C3/imunologia , Modelos Animais de Doenças , Feminino , Humanos , Mediadores da Inflamação/imunologia , Macaca fascicularis , Masculino , Camundongos , Osteoclastos/imunologia , Osteoclastos/patologia , Peptídeos Cíclicos/farmacologia , Periodontite/tratamento farmacológico , Periodontite/genética , Periodontite/imunologia , Periodontite/patologia
3.
J Immunol ; 189(11): 5442-8, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23089394

RESUMO

When excessively activated or deregulated, complement becomes a major link between infection and inflammatory pathology including periodontitis. This oral inflammatory disease is associated with a dysbiotic microbiota, leads to the destruction of bone and other tooth-supporting structures, and exerts an adverse impact on systemic health. We have previously shown that mice deficient either in complement C5a receptor (C5aR; CD88) or TLR2 are highly and similarly resistant to periodontitis, suggesting that a cross-talk between the two receptors may be involved in the disease process. In this paper, we show that C5aR and TLR2 indeed synergize for maximal inflammatory responses in the periodontal tissue and uncover a novel pharmacological target to abrogate periodontitis. Using two different mouse models of periodontitis, we show that local treatments with a C5aR antagonist inhibited periodontal inflammation through downregulation of TNF, IL-1ß, IL-6, and IL-17 and further protected against bone loss, regardless of the presence of TLR2. These findings not only reveal a crucial cooperation between C5aR and TLR2 in periodontal inflammation but also provide proof-of-concept for local targeting of C5aR as a powerful candidate for the treatment of human periodontitis.


Assuntos
Infecções por Bacteroidaceae/tratamento farmacológico , Lipopeptídeos/administração & dosagem , Peptídeos Cíclicos/administração & dosagem , Periodontite/tratamento farmacológico , Porphyromonas gingivalis/efeitos dos fármacos , Receptor da Anafilatoxina C5a/antagonistas & inibidores , Animais , Infecções por Bacteroidaceae/imunologia , Infecções por Bacteroidaceae/microbiologia , Regulação para Baixo/efeitos dos fármacos , Sinergismo Farmacológico , Gengiva/imunologia , Imunidade Inata/efeitos dos fármacos , Injeções , Interleucina-17/biossíntese , Interleucina-17/imunologia , Interleucina-1beta/biossíntese , Interleucina-1beta/imunologia , Interleucina-6/biossíntese , Interleucina-6/imunologia , Camundongos , Periodontite/imunologia , Periodontite/microbiologia , Porphyromonas gingivalis/imunologia , Receptor Cross-Talk/efeitos dos fármacos , Receptor Cross-Talk/imunologia , Receptor da Anafilatoxina C5a/metabolismo , Receptor 2 Toll-Like/agonistas , Receptor 2 Toll-Like/metabolismo
4.
Cell Microbiol ; 14(7): 1085-96, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22381126

RESUMO

Porphyromonas gingivalis, a Gram-negative oral pathogen, has been shown to induce apoptosis in human gingival epithelial cells, yet the underlining cellular mechanisms controlling this process are poorly understood. We have previously shown that the P. gingivalis proteases arginine and lysine gingipains, are necessary and sufficient to induce host cell apoptosis. In the present study, we demonstrate that 'P. gingivalis-induced apoptosis' is mediated through degradation of actin leading to cytoskeleton collapse. Stimulation of human gingival epithelial cells with P. gingivalis strains 33277 and W50 at moi:100 induced ß-actin cleavage as early as 1 h and human serum inhibited this effect. By using gingipain-deficient mutants of P. gingivalis and purified gingipains, we demonstrate that lysine gingipain is involved in actin hydrolysis in a dose and time-dependent manner. Use of Jasplakinolide and cytochalasin D revealed that P. gingivalis internalization is necessary for actin cleavage. Further, we also show that lysine gingipain from P. gingivalis can cleave active caspase 3. Taken together, we have identified actin as a substrate for lysine gingipain and demonstrated a novel mechanism involved in microbial host cell invasion and apoptosis.


Assuntos
Actinas/metabolismo , Apoptose , Células Epiteliais/microbiologia , Porphyromonas gingivalis/patogenicidade , Adesinas Bacterianas/metabolismo , Células Cultivadas , Cisteína Endopeptidases/metabolismo , Cisteína Endopeptidases Gingipaínas , Humanos , Porphyromonas gingivalis/enzimologia
5.
J Immunol ; 186(2): 869-77, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21149611

RESUMO

The C5a anaphylatoxin receptor (C5aR; CD88) is activated as part of the complement cascade and exerts important inflammatory, antimicrobial, and regulatory functions, at least in part, via crosstalk with TLRs. However, the periodontal pathogen Porphyromonas gingivalis can control C5aR activation by generating C5a through its own C5 convertase-like enzymatic activity. In this paper, we show that P. gingivalis uses this mechanism to proactively and selectively inhibit TLR2-induced IL-12p70, whereas the same pathogen-instigated C5aR-TLR2 crosstalk upregulates other inflammatory and bone-resorptive cytokines (IL-1ß, IL-6, and TNF-α). In vivo, the ability of P. gingivalis to manipulate TLR2 activation via the C5a-C5aR axis allowed it to escape IL-12p70-dependent immune clearance and to cause inflammatory bone loss in a murine model of experimental periodontitis. In the latter regard, C5aR-deficient or TLR2-deficient mice were both resistant to periodontal bone loss, in stark contrast with wild-type control mice, which is consistent with the interdependent interactions of C5aR and TLR2 in P. gingivalis immune evasion and induction of bone-resorptive cytokines. In conclusion, P. gingivalis targets C5aR to promote its adaptive fitness and cause periodontal disease. Given the current availability of safe and effective C5aR antagonists, pharmacological blockade of C5aR could act therapeutically in human periodontitis and reduce associated systemic risks.


Assuntos
Perda do Osso Alveolar/imunologia , Perda do Osso Alveolar/microbiologia , Interleucina-12/antagonistas & inibidores , Interleucina-12/fisiologia , Periodontite/imunologia , Periodontite/microbiologia , Porphyromonas gingivalis/imunologia , Receptor da Anafilatoxina C5a/fisiologia , Perda do Osso Alveolar/metabolismo , Animais , Modelos Animais de Doenças , Evasão da Resposta Imune/genética , Evasão da Resposta Imune/imunologia , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/fisiologia , Interleucina-10/fisiologia , Interleucina-12/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/fisiologia , Periodontite/metabolismo , Porphyromonas gingivalis/crescimento & desenvolvimento , Receptor Cross-Talk/imunologia , Receptor da Anafilatoxina C5a/deficiência , Receptor da Anafilatoxina C5a/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptor 2 Toll-Like/antagonistas & inibidores , Receptor 2 Toll-Like/fisiologia
6.
Clin Dev Immunol ; 2013: 617809, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24416060

RESUMO

Developmental endothelial locus-1 (Del-1) is an endothelial cell-secreted protein that limits the recruitment of neutrophils by antagonizing the interaction between the LFA-1 integrin on neutrophils and the intercellular adhesion molecule (ICAM)-1 on endothelial cells. Mice with genetic or age-associated Del-1 deficiency exhibit increased neutrophil infiltration in the periodontium resulting in inflammatory bone loss. Here we investigated additional novel mechanisms whereby Del-1 could interfere with neutrophil recruitment and inflammation. Treatment of human endothelial cells with Del-1 did not affect the expression of endothelial molecules involved in the leukocyte adhesion cascade (ICAM-1, VCAM-1, and E-selectin). Moreover, genetic or age-associated Del-1 deficiency did not significantly alter the expression of these adhesion molecules in the murine periodontium, further ruling out altered adhesion molecule expression as a mechanism whereby Del-1 regulates leukocyte recruitment. Strikingly, Del-1 inhibited ICAM-1-dependent chemokine release (CXCL2, CCL3) by neutrophils. Therefore, Del-1 could potentially suppress the amplification of inflammatory cell recruitment mediated through chemokine release by infiltrating neutrophils. Interestingly, Del-1 was itself regulated by inflammatory stimuli, which generally exerted opposite effects on adhesion molecule expression. The reciprocal regulation between Del-1 and inflammation may contribute to optimally balance the protective and the potentially harmful effects of inflammatory cell recruitment.


Assuntos
Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Células Endoteliais/metabolismo , Periodonto/metabolismo , Animais , Proteínas de Ligação ao Cálcio , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Quimiocinas/biossíntese , Expressão Gênica , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Camundongos , Camundongos Knockout , Neutrófilos/metabolismo , Osteoporose/genética , Osteoporose/metabolismo , Periodontite/genética , Periodontite/imunologia , Molécula 1 de Adesão de Célula Vascular/metabolismo
7.
J Immunol ; 182(5): 2978-85, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19234193

RESUMO

The pentameric B subunit of type IIb Escherichia coli enterotoxin (LT-IIb-B(5)), a doughnut-shaped oligomeric protein from enterotoxigenic E. coli, activates the TLR2/TLR1 heterodimer (TLR2/1). We investigated the molecular basis of the LT-IIb-B(5) interaction with TLR2/1 to define the structure-function relationship of LT-IIb-B(5) and, moreover, to gain an insight into how TLR2/1 recognizes large, nonacylated protein ligands that cannot fit within its lipid-binding pockets, as previously shown for the Pam(3)CysSerLys(4) (Pam(3)CSK(4)) lipopeptide. We first identified four critical residues in the upper region of the LT-IIb-B(5) pore. Corresponding point mutants (M69E, A70D, L73E, S74D) were defective in binding TLR2 or TLR1 and could not activate APCs, despite retaining full ganglioside-binding capacity. Point mutations in the TLR2/1 dimer interface, as determined in the crystallographic structure of the TLR2/1-Pam(3)CSK(4) complex, resulted in diminished activation by both Pam(3)CSK(4) and LT-IIb-B(5). Docking analysis of the LT-IIb-B(5) interaction with this apparently predominant activation conformation of TLR2/1 revealed that LT-IIb-B(5) might primarily contact the convex surface of the TLR2 central domain. Although the TLR1/LT-IIb-B(5) interface is relatively smaller, the leucine-rich repeat motifs 9-12 in the central domain of TLR1 were found to be critical for cooperative TLR2-induced cell activation by LT-IIb-B(5). Moreover, the putative LT-IIb-B(5) binding site overlaps partially with that of Pam(3)CSK(4); consistent with this, Pam(3)CSK(4) suppressed TLR2 binding of LT-IIb-B(5), albeit not as potently as self-competitive inhibition. We identified the upper pore region of LT-IIb-B(5) as a TLR2/1 interactive domain, which contacts the heterodimeric receptor at a site that is distinct from, although it overlaps with, that of Pam(3)CSK(4).


Assuntos
Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Escherichia coli Enterotoxigênica/química , Enterotoxinas/química , Enterotoxinas/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Mapeamento de Peptídeos , Receptor 1 Toll-Like/metabolismo , Receptor 2 Toll-Like/metabolismo , Animais , Toxinas Bacterianas/genética , Linhagem Celular Tumoral , Células Cultivadas , Cristalografia por Raios X , Dimerização , Escherichia coli Enterotoxigênica/genética , Escherichia coli Enterotoxigênica/metabolismo , Enterotoxinas/genética , Proteínas de Escherichia coli/genética , Humanos , Lipopeptídeos/química , Lipopeptídeos/genética , Lipopeptídeos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Mapeamento de Peptídeos/métodos , Mutação Puntual , Ligação Proteica/genética , Ligação Proteica/imunologia , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Receptor 1 Toll-Like/antagonistas & inibidores , Receptor 1 Toll-Like/química , Receptor 2 Toll-Like/antagonistas & inibidores , Receptor 2 Toll-Like/química
8.
Sci Transl Med ; 6(229): 229ra40, 2014 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-24670684

RESUMO

Leukocyte adhesion deficiency type I (LAD-I), a disease syndrome associated with frequent microbial infections, is caused by mutations on the CD18 subunit of ß2 integrins. LAD-I is invariably associated with severe periodontal bone loss, which historically has been attributed to the lack of neutrophil surveillance of the periodontal infection. We provide an alternative mechanism by showing that the cytokine interleukin-17 (IL-17) plays a major role in the oral pathology of LAD-I. Defective neutrophil recruitment in LAD-I patients or in LFA-1 (CD11a/CD18)-deficient mice--which exhibit the LAD-I periodontal phenotype--was associated with excessive production of predominantly T cell-derived IL-17 in the periodontal tissue, although innate lymphoid cells also contributed to pathological IL-17 elevation in the LFA-1-deficient mice. Local treatment with antibodies to IL-17 or IL-23 in LFA-1-deficient mice not only blocked inflammatory periodontal bone loss but also caused a reduction in the total bacterial burden, suggesting that the IL-17-driven pathogenesis of LAD-I periodontitis leads to dysbiosis. Therefore, our findings support an IL-17-targeted therapy for periodontitis in LAD-I patients.


Assuntos
Reabsorção Óssea/complicações , Reabsorção Óssea/patologia , Inflamação/patologia , Interleucina-17/metabolismo , Síndrome da Aderência Leucocítica Deficitária/patologia , Infiltração de Neutrófilos , Adolescente , Animais , Adesão Celular , Criança , Perfilação da Expressão Gênica , Humanos , Inflamação/complicações , Inflamação/genética , Interleucina-23/metabolismo , Síndrome da Aderência Leucocítica Deficitária/complicações , Síndrome da Aderência Leucocítica Deficitária/genética , Antígeno-1 Associado à Função Linfocitária/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Periodontite/complicações , Periodontite/genética , Periodontite/microbiologia , Periodontite/patologia
9.
Mol Immunol ; 48(1-3): 294-304, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20728939

RESUMO

Recent studies implicate the mammalian target of rapamycin (mTOR) pathway in the control of inflammatory responses following Toll-like receptor (TLR) stimulation in myeloid cells but its role in non-myeloid cells such as human keratinocytes is unknown. Here we show that TLR3 signaling can induce robust cytokine secretion including interleukin 1 beta (IL-1ß), tumor necrosis factor alpha (TNFα), IL-12p70 and interferon beta (IFN-ß), and our data reveal for the first time that inhibiting mTOR with rapamycin, suppresses these TLR3 induced responses but actually enhances bioactive IL-12p70 production in human oral keratinocytes. Rapamycin inhibited the phosphorylation of the 70-kDa ribosomal protein S6 kinase (p70S6K) and the 4E binding protein 1 (4EBP-1), and suppressed the mitogen activated protein kinase (MAPK) pathway by decreasing phosphorylation of c-Jun N-terminal kinase (JNK). We also show that TLR3 induces interferon regulatory factor 3 (IRF3) activation by Akt via an mTOR-p70S6K-4EBP1 pathway. Furthermore, we provide evidence that Poly I:C induced expression of IL-1ß, TNFα, IL-12p70 and IFN-ß was blocked by JNK inhibitor SP600125. TLR3 preferentially phosphorylated IKKα through mTOR to activate nuclear factor kappa beta (NF-κB) in human oral keratinocytes. Taken together, these data demonstrate p70S6K, p4EBP1, JNK, NF-κB and IRF3 are involved in the regulation of inflammatory mediators by TLR3 via the mTOR pathway. mTOR is a novel pathway modulating TLR3 induced inflammatory and antiviral responses in human oral keratinocytes.


Assuntos
Queratinócitos/imunologia , Mucosa Bucal/imunologia , Transdução de Sinais/imunologia , Serina-Treonina Quinases TOR/imunologia , Receptor 3 Toll-Like/imunologia , Western Blotting , Citocinas/biossíntese , Citocinas/efeitos dos fármacos , Citocinas/imunologia , Humanos , Imunossupressores/farmacologia , Fator Regulador 3 de Interferon/efeitos dos fármacos , Fator Regulador 3 de Interferon/imunologia , Fator Regulador 3 de Interferon/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno/imunologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/imunologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mucosa Bucal/efeitos dos fármacos , Mucosa Bucal/metabolismo , Poli I-C/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Receptor 3 Toll-Like/efeitos dos fármacos , Receptor 3 Toll-Like/metabolismo
10.
Sci Signal ; 3(109): ra11, 2010 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-20159852

RESUMO

Crosstalk between complement and Toll-like receptors (TLRs) coordinates innate immunity. We report a previously unknown immune subversion mechanism involving microbial exploitation of communication between complement and TLRs. Porphyromonas gingivalis, a major oral and systemic pathogen with complement C5 convertase-like activity, synergizes with C5a (fragment of complement protein C5) to increase cyclic adenosine monophosphate (cAMP) concentrations, resulting in suppression of macrophage immune function and enhanced pathogen survival in vitro and in vivo. This synergy required TLR2 signaling, a pertussis toxin- and thapsigargin-sensitive C5a receptor pathway, with protein kinase A and glycogen synthase kinase-3beta as downstream effectors. Antagonistic blockade of the C5a receptor abrogated this evasive strategy and may thus have important therapeutic implications for periodontitis and atherosclerosis, diseases in which P. gingivalis is implicated. This first demonstration of complement-TLR crosstalk for immunosuppressive cAMP signaling indicates that pathogens may not simply undermine complement or TLRs (or both) as separate entities, but may also exploit their crosstalk pathways.


Assuntos
Complemento C5/metabolismo , Porphyromonas gingivalis/metabolismo , Receptor Cross-Talk , Receptores Toll-Like/metabolismo , Animais , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Macrófagos/imunologia , Camundongos , Transdução de Sinais
11.
Mech Ageing Dev ; 130(8): 538-46, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19559723

RESUMO

The impact of ageing in innate immunity is poorly understood. Studies in the mouse model have described altered innate immune functions in aged macrophages, although these were not generally linked to altered expression of receptors or regulatory molecules. Moreover, the influence of ageing in the expression of these molecules has not been systematically examined. We investigated age-dependent expression differences in selected Toll-like and other pattern-recognition receptors, receptors involved in inflammatory amplification, and in transmembrane and intracellular regulators of inflammatory signaling. Young and aged macrophages were examined under resting conditions or upon activation with Porphyromonas gingivalis, a major pathogen in periodontal disease, the prevalence and severity of which increase in old age. We detected a limited number of age-dependent alterations, involving both reduction and increase of immune activity. Interestingly, surface expression of receptors that amplify inflammation (C5a anaphylatoxin receptor and triggering receptor expressed on myeloid cells [TREM]-1) was elevated in aged macrophages. No significant age-dependent differences were observed regarding the phagocytosis and intracellular killing of P. gingivalis, consistent with lack of significant changes in phagocytic receptor expression and induction of antimicrobial molecules. Therefore, at least at the cellular level, certain aspects of innate immune function may not necessarily decline with age.


Assuntos
Envelhecimento , Macrófagos/imunologia , Macrófagos/microbiologia , Animais , Citometria de Fluxo/métodos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Imunidade Inata , Inflamação , Camundongos , Camundongos Endogâmicos BALB C , Células Mieloides/metabolismo , Doenças Periodontais/microbiologia , Fagocitose , Porphyromonas gingivalis/metabolismo , Receptores Toll-Like/metabolismo
12.
Vaccine ; 27(32): 4302-8, 2009 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-19450646

RESUMO

The pentameric B subunit of the Escherichia coli LT-IIb enterotoxin (LT-IIb-B(5)) activates TLR2 signaling in macrophages. Herein we demonstrate that LT-IIb-B(5), in contrast to a TLR2-nonbinding point mutant, induces functional activation of bone marrow-derived dendritic cells and stimulates CD4(+) T cell proliferation, activities which suggested that LT-IIb-B(5) might function as an adjuvant in vivo. Indeed, in an intranasal mouse immunization model, LT-IIb-B(5) augmented specific mucosal and serum antibody responses to a co-administered immunogen, at levels which were almost comparable to those induced by intact LT-IIb holotoxin, a potent but toxic adjuvant. Therefore, LT-IIb-B(5) displays useful adjuvant properties which, combined with lack of enterotoxicity and relative stability against degradation, may find application in mucosal vaccines.


Assuntos
Adjuvantes Imunológicos/farmacologia , Toxinas Bacterianas/farmacologia , Enterotoxinas/farmacologia , Proteínas de Escherichia coli/farmacologia , Administração Intranasal , Animais , Anticorpos/análise , Anticorpos/sangue , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Proliferação de Células , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Imunidade nas Mucosas , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Subunidades Proteicas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA