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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Immunol ; 20(1): 40-49, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30455459

RESUMO

Resolution of inflammation is essential for tissue homeostasis and represents a promising approach to inflammatory disorders. Here we found that developmental endothelial locus-1 (DEL-1), a secreted protein that inhibits leukocyte-endothelial adhesion and inflammation initiation, also functions as a non-redundant downstream effector in inflammation clearance. In human and mouse periodontitis, waning of inflammation was correlated with DEL-1 upregulation, whereas resolution of experimental periodontitis failed in DEL-1 deficiency. This concept was mechanistically substantiated in acute monosodium-urate-crystal-induced inflammation, where the pro-resolution function of DEL-1 was attributed to effective apoptotic neutrophil clearance (efferocytosis). DEL-1-mediated efferocytosis induced liver X receptor-dependent macrophage reprogramming to a pro-resolving phenotype and was required for optimal production of at least certain specific pro-resolving mediators. Experiments in transgenic mice with cell-specific overexpression of DEL-1 linked its anti-leukocyte-recruitment action to endothelial cell-derived DEL-1 and its efferocytic/pro-resolving action to macrophage-derived DEL-1. Thus, the compartmentalized expression of DEL-1 facilitates distinct homeostatic functions in an appropriate context that can be harnessed therapeutically.


Assuntos
Proteínas de Transporte/metabolismo , Inflamação/imunologia , Macrófagos/fisiologia , Neutrófilos/imunologia , Periodontite/imunologia , Adulto , Animais , Proteínas de Ligação ao Cálcio , Proteínas de Transporte/genética , Moléculas de Adesão Celular , Reprogramação Celular , Citocinas/metabolismo , Regulação da Expressão Gênica , Humanos , Inflamação/induzido quimicamente , Peptídeos e Proteínas de Sinalização Intercelular , Células K562 , Receptores X do Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose
2.
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
3.
Mol Psychiatry ; 20(7): 880-888, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25385367

RESUMO

Inflammation in the central nervous system (CNS) and disruption of its immune privilege are major contributors to the pathogenesis of multiple sclerosis (MS) and of its rodent counterpart, experimental autoimmune encephalomyelitis (EAE). We have previously identified developmental endothelial locus-1 (Del-1) as an endogenous anti-inflammatory factor, which inhibits integrin-dependent leukocyte adhesion. Here we show that Del-1 contributes to the immune privilege status of the CNS. Intriguingly, Del-1 expression decreased in chronic-active MS lesions and in the inflamed CNS in the course of EAE. Del-1-deficiency was associated with increased EAE severity, accompanied by increased demyelination and axonal loss. As compared with control mice, Del-1(-/-) mice displayed enhanced disruption of the blood-brain barrier and increased infiltration of neutrophil granulocytes in the spinal cord in the course of EAE, accompanied by elevated levels of inflammatory cytokines, including interleukin-17 (IL-17). The augmented levels of IL-17 in Del-1-deficiency derived predominantly from infiltrated CD8(+) T cells. Increased EAE severity and neutrophil infiltration because of Del-1-deficiency was reversed in mice lacking both Del-1 and IL-17 receptor, indicating a crucial role for the IL-17/neutrophil inflammatory axis in EAE pathogenesis in Del-1(-/-) mice. Strikingly, systemic administration of Del-1-Fc ameliorated clinical relapse in relapsing-remitting EAE. Therefore, Del-1 is an endogenous homeostatic factor in the CNS protecting from neuroinflammation and demyelination. Our findings provide mechanistic underpinnings for the previous implication of Del-1 as a candidate MS susceptibility gene and suggest that Del-1-centered therapeutic approaches may be beneficial in neuroinflammatory and demyelinating disorders.


Assuntos
Axônios/metabolismo , Barreira Hematoencefálica/metabolismo , Proteínas de Transporte/metabolismo , Bainha de Mielina/metabolismo , Neuroimunomodulação/fisiologia , Medula Espinal/metabolismo , Animais , Axônios/efeitos dos fármacos , Axônios/patologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/patologia , Proteínas de Ligação ao Cálcio , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/fisiologia , Proteínas de Transporte/genética , Moléculas de Adesão Celular , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Feminino , Granulócitos/efeitos dos fármacos , Granulócitos/metabolismo , Granulócitos/patologia , Homeostase/efeitos dos fármacos , Homeostase/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular , Interleucina-17/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Bainha de Mielina/efeitos dos fármacos , Bainha de Mielina/patologia , Neuroimunomodulação/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Neutrófilos/patologia , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/metabolismo , Índice de Gravidade de Doença , Medula Espinal/efeitos dos fármacos , Medula Espinal/patologia
4.
J Immunol ; 193(3): 1383-91, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24958900

RESUMO

The glycoprotein milk fat globule-epidermal growth factor factor 8 (MFG-E8) is expressed in several tissues and mediates diverse homeostatic functions. However, whether it plays a role in bone homeostasis has not been established. In this study, we show for the first time, to our knowledge, that osteoclasts express and are regulated by MFG-E8. Bone marrow-derived osteoclast precursors from MFG-E8-deficient (Mfge8(-/-)) mice underwent increased receptor activator of NF-κB ligand-induced osteoclastogenesis, leading to enhanced resorption pit formation compared with wild-type controls. Consistently, exogenously added MFG-E8 inhibited receptor activator of NF-κB ligand-induced osteoclastogenesis from mouse or human osteoclast precursors. Upon induction of experimental periodontitis, an oral inflammatory disease characterized by loss of bone support of the dentition, Mfge8(-/-) mice exhibited higher numbers of osteoclasts and more bone loss than did wild-type controls. Accordingly, local microinjection of anti-MFG-E8 mAb exacerbated periodontal bone loss in wild-type mice. Conversely, microinjection of MFG-E8 inhibited bone loss in experimental mouse periodontitis. In comparison with wild-type controls, Mfge8(-/-) mice also experienced >60% more naturally occurring chronic periodontal bone loss. In conclusion, MFG-E8 is a novel homeostatic regulator of osteoclasts that could be exploited therapeutically to treat periodontitis and perhaps other immunological disorders associated with inflammatory bone loss.


Assuntos
Antígenos de Superfície/fisiologia , Reabsorção Óssea/imunologia , Homeostase/imunologia , Mediadores da Inflamação/fisiologia , Osteoclastos/imunologia , Osteoclastos/patologia , Animais , Antígenos de Superfície/biossíntese , Antígenos de Superfície/genética , Reabsorção Óssea/genética , Reabsorção Óssea/patologia , Linhagem Celular , Homeostase/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Leite/biossíntese , Proteínas do Leite/genética , Periodontite/genética , Periodontite/imunologia , Periodontite/patologia , Ligante RANK/administração & dosagem , Ligante RANK/efeitos adversos , Células-Tronco/imunologia , Células-Tronco/metabolismo , Células-Tronco/patologia
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
Artigo em Inglês | MEDLINE | ID: mdl-28848717

RESUMO

Porphyromonas gingivalis is a gram-negative anaerobic periodontal pathogen that persists in dysbiotic mixed-species biofilms alongside a dense inflammatory infiltrate of neutrophils and other leukocytes in the subgingival areas of the periodontium. Toll-like receptor 2 (TLR2) mediates the inflammatory response to P. gingivalis and TLR2-deficient mice resist alveolar bone resorption following oral challenge with this organism. Although, MyD88 is an adaptor protein considered necessary for TLR2-induced inflammation, we now report for the first time that oral challenge with P. gingivalis leads to alveolar bone resorption in the absence of MyD88. Indeed, in contrast to prototypical TLR2 agonists, such as the lipopeptide Pam3CSK4 that activates TLR2 in a strictly MyD88-dependent manner, P. gingivalis strikingly induced TLR2 signaling in neutrophils and macrophages regardless of the presence or absence of MyD88. Moreover, genetic or antibody-mediated inactivation of TLR2 completely reduced cytokine production in P. gingivalis-stimulated neutrophils or macrophages, suggesting that TLR2 plays a non-redundant role in the host response to P. gingivalis. In the absence of MyD88, inflammatory TLR2 signaling in P. gingivalis-stimulated neutrophils or macrophages depended upon PI3K. Intriguingly, TLR2-PI3K signaling was also critical to P. gingivalis evasion of killing by macrophages, since their ability to phagocytose this pathogen was reduced in a TLR2 and PI3K-dependent manner. Moreover, within those cells that did phagocytose bacteria, TLR2-PI3K signaling blocked phago-lysosomal maturation, thereby revealing a novel mechanism whereby P. gingivalis can enhance its intracellular survival. Therefore, P. gingivalis uncouples inflammation from bactericidal activity by substituting TLR2-PI3K in place of TLR2-MyD88 signaling. These findings further support the role of P. gingivalis as a keystone pathogen, which manipulates the host inflammatory response in a way that promotes bone loss but not bacterial clearance. Modulation of these host response factors may lead to novel therapeutic approaches to improve outcomes in disease conditions associated with P. gingivalis.


Assuntos
Perda do Osso Alveolar/microbiologia , Infecções por Bacteroidaceae/imunologia , Infecções por Bacteroidaceae/microbiologia , Fator 88 de Diferenciação Mieloide/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Porphyromonas gingivalis/patogenicidade , Receptor 2 Toll-Like/metabolismo , Animais , Citocinas/análise , Humanos , Inflamação/imunologia , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide/genética , Neutrófilos/imunologia , Neutrófilos/microbiologia , Fosfatidilinositol 3-Quinases/genética , Porphyromonas gingivalis/genética , Células RAW 264.7 , Receptor 2 Toll-Like/genética
12.
J Clin Invest ; 127(10): 3624-3639, 2017 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-28846069

RESUMO

Hematopoietic stem cells (HSCs) remain mostly quiescent under steady-state conditions but switch to a proliferative state following hematopoietic stress, e.g., bone marrow (BM) injury, transplantation, or systemic infection and inflammation. The homeostatic balance between quiescence, self-renewal, and differentiation of HSCs is strongly dependent on their interactions with cells that constitute a specialized microanatomical environment in the BM known as the HSC niche. Here, we identified the secreted extracellular matrix protein Del-1 as a component and regulator of the HSC niche. Specifically, we found that Del-1 was expressed by several cellular components of the HSC niche, including arteriolar endothelial cells, CXCL12-abundant reticular (CAR) cells, and cells of the osteoblastic lineage. Del-1 promoted critical functions of the HSC niche, as it regulated long-term HSC (LT-HSC) proliferation and differentiation toward the myeloid lineage. Del-1 deficiency in mice resulted in reduced LT-HSC proliferation and infringed preferentially upon myelopoiesis under both steady-state and stressful conditions, such as hematopoietic cell transplantation and G-CSF- or inflammation-induced stress myelopoiesis. Del-1-induced HSC proliferation and myeloid lineage commitment were mediated by ß3 integrin on hematopoietic progenitors. This hitherto unknown Del-1 function in the HSC niche represents a juxtacrine homeostatic adaptation of the hematopoietic system in stress myelopoiesis.


Assuntos
Proteínas de Transporte/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Mielopoese , Nicho de Células-Tronco , Estresse Fisiológico , Animais , Proteínas de Ligação ao Cálcio , Proteínas de Transporte/genética , Moléculas de Adesão Celular , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Células Endoteliais/metabolismo , Humanos , Integrina beta3/genética , Integrina beta3/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Camundongos Knockout
13.
Thromb Haemost ; 115(4): 781-8, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26676803

RESUMO

Platelet-monocyte interactions are strongly implicated in thrombo-inflammatory injury by actively contributing to intravascular inflammation, leukocyte recruitment to inflamed sites, and the amplification of the procoagulant response. Instant blood-mediated inflammatory reaction (IBMIR) represents thrombo-inflammatory injury elicited upon pancreatic islet transplantation (islet-Tx), thereby dramatically affecting transplant survival and function. Developmental endothelial locus-1 (Del-1) is a functionally versatile endothelial cell-derived homeostatic factor with anti-inflammatory properties, but its potential role in IBMIR has not been previously addressed. Here, we establish Del-1 as a novel inhibitor of IBMIR using a whole blood-islet model and a syngeneic murine transplantation model. Indeed, Del-1 pre-treatment of blood before addition of islets diminished coagulation activation and islet damage as assessed by C-peptide release. Consistently, intraportal islet-Tx in transgenic mice with endothelial cell-specific overexpression of Del-1 resulted in a marked decrease of monocytes and platelet-monocyte aggregates in the transplanted tissues, relative to those in wild-type recipients. Mechanistically, Del-1 decreased platelet-monocyte aggregate formation, by specifically blocking the interaction between monocyte Mac-1-integrin and platelet GPIb. Our findings reveal a hitherto unknown role of Del-1 in the regulation of platelet-monocyte interplay and the subsequent heterotypic aggregate formation in the context of IBMIR. Therefore, Del-1 may represent a novel approach to prevent or mitigate the adverse reactions mediated through thrombo-inflammatory pathways in islet-Tx and perhaps other inflammatory disorders involving platelet-leukocyte aggregate formation.


Assuntos
Plaquetas/fisiologia , Proteínas de Transporte/metabolismo , Inflamação/genética , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/metabolismo , Monócitos/fisiologia , Trombose/genética , Animais , Coagulação Sanguínea/genética , Proteínas de Ligação ao Cálcio , Proteínas de Transporte/genética , Moléculas de Adesão Celular , Células Cultivadas , Humanos , Ilhotas Pancreáticas/patologia , Antígeno de Macrófago 1/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Agregação Plaquetária/genética , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Trombose/imunologia
14.
Sci Transl Med ; 7(307): 307ra155, 2015 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-26424570

RESUMO

DEL-1 (developmental endothelial locus-1) is an endothelial cell-secreted protein that regulates LFA-1 (lymphocyte function-associated antigen-1) integrin-dependent leukocyte recruitment and inflammation in various tissues. We identified a novel regulatory mechanism of DEL-1 in osteoclast biology. Specifically, we showed that DEL-1 is expressed by human and mouse osteoclasts and regulates their differentiation and resorptive function. Mechanistically, DEL-1 inhibited the expression of NFATc1, a master regulator of osteoclastogenesis, in a Mac-1 integrin-dependent manner. In vivo mechanistic analysis has dissociated the anti-inflammatory from the anti-bone-resorptive action of DEL-1 and identified structural components thereof mediating these distinct functions. Locally administered human DEL-1 blocked inflammatory periodontal bone loss in nonhuman primates-a relevant model of human periodontitis. The ability of DEL-1 to regulate both upstream (inflammatory cell recruitment) and downstream (osteoclastogenesis) events that lead to inflammatory bone loss paves the way to a new class of endogenous therapeutics for treating periodontitis and perhaps other inflammatory disorders.


Assuntos
Reabsorção Óssea/complicações , Proteínas de Transporte/metabolismo , Inflamação/complicações , Osteoclastos/metabolismo , Osteogênese , Motivos de Aminoácidos , Animais , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Reabsorção Óssea/prevenção & controle , Proteínas de Ligação ao Cálcio , Moléculas de Adesão Celular , Diferenciação Celular , Movimento Celular , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Líquido do Sulco Gengival/metabolismo , Humanos , Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intercelular , Antígeno-1 Associado à Função Linfocitária/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/patologia , Periodontite/patologia , Primatas , Estrutura Terciária de Proteína
15.
Nat Commun ; 6: 8272, 2015 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-26374165

RESUMO

Del-1 is an endothelial cell-secreted anti-inflammatory protein. In humans and mice, Del-1 expression is inversely related to that of IL-17, which inhibits Del-1 through hitherto unidentified mechanism(s). Here we show that IL-17 downregulates human endothelial cell expression of Del-1 by targeting a critical transcription factor, C/EBPß. Specifically, IL-17 causes GSK-3ß-dependent phosphorylation of C/EBPß, which is associated with diminished C/EBPß binding to the Del-1 promoter and suppressed Del-1 expression. This inhibitory action of IL-17 can be reversed at the GSK-3ß level by PI3K/Akt signalling induced by D-resolvins. The biological relevance of this regulatory network is confirmed in a mouse model of inflammatory periodontitis. Intriguingly, resolvin-D1 (RvD1) confers protection against IL-17-driven periodontal bone loss in a Del-1-dependent manner, indicating an RvD1-Del-1 axis against IL-17-induced pathological inflammation. The dissection of signalling pathways regulating Del-1 expression provides potential targets to treat inflammatory diseases associated with diminished Del-1 expression, such as periodontitis and multiple sclerosis.


Assuntos
Perda do Osso Alveolar/imunologia , Proteína beta Intensificadora de Ligação a CCAAT/imunologia , Proteínas de Transporte/imunologia , Quinase 3 da Glicogênio Sintase/imunologia , Interleucina-17/imunologia , Periodontite/imunologia , Perda do Osso Alveolar/genética , Animais , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Proteínas de Ligação ao Cálcio , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Moléculas de Adesão Celular , Imunoprecipitação da Cromatina , Modelos Animais de Doenças , Ácidos Docosa-Hexaenoicos/farmacologia , Regulação para Baixo , Ensaio de Imunoadsorção Enzimática , Gengiva/metabolismo , Quinase 3 da Glicogênio Sintase/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta , Células Endoteliais da Veia Umbilical Humana , Humanos , Immunoblotting , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Camundongos Knockout , Periodontite/genética , Peroxidase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais
16.
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
17.
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
18.
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
19.
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
20.
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