Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Cell Biochem ; 119(2): 1591-1603, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28771806

RESUMO

Accumulation of advanced glycation end-products (AGEs) in periodontal tissues of patients with diabetes mellitus aggravates periodontitis, but the mechanisms are unknown. Calprotectin, a heterocomplex of S100A8 and S100A9 proteins, is a constitutive cytoplasmic component of healthy gingival epithelial cells. This study aimed at investigating the effects of AGE and Porphyromonas gingivalis lipopolysaccharide (PgLPS) on calprotectin expression in the human gingival epithelial cell line OBA-9. AGE and PgLPS increased the expression of S100A8 and S100A9 mRNAs, and AGE+PgLPS co-stimulation amplified their expression in OBA-9 cells. A higher concentration of calprotectin in cell lysates was also induced by stimulation with AGE and/or PgLPS. S100A8 was mainly translocated from the nucleus to the cytoplasm by AGE stimulation, while cytoplasmic localization of S100A9 was not altered following stimulation with AGE and/or PgLPS. Calprotectin was found in the cytoplasm of BSA-treated cells, but cytoplasmic and nuclear localization was observed following stimulation with AGE and/or PgLPS. AGE-induced S100A8, and S100A9 mRNA expression was partially suppressed by RAGE-specific siRNA. In contrast, PgLPS-induced S100A8 and S100A9 mRNA expression was strongly suppressed by TLR2-specific siRNA. Furthermore, the inhibition of p38, JNK MAPK, and NF-κB attenuated AGE- and PgLPS-induced S100A8 and S100A9 mRNA expression. Taken together, these results demonstrate that AGE acts in synergy with PgLPS to stimulate RAGE and TLR2 expression and activate p38, JNK MAPK, and NF-κB signaling pathways, resulting in increased activation of calprotectin (S100A8/S100A9) in human gingival epithelial cells. Our results suggest that calprotectin may be involved in the pathogenesis of diabetic periodontitis.


Assuntos
Calgranulina A/genética , Calgranulina B/genética , Gengiva/metabolismo , Produtos Finais de Glicação Avançada/efeitos adversos , Lipopolissacarídeos/efeitos adversos , Porphyromonas gingivalis/metabolismo , Calgranulina A/metabolismo , Calgranulina B/metabolismo , Linhagem Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Citoplasma/genética , Citoplasma/metabolismo , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Gengiva/citologia , Gengiva/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases , Periodontite/genética , Periodontite/metabolismo , Regulação para Cima
2.
J Cell Sci ; 129(6): 1198-209, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26826187

RESUMO

Inhibitory proteins, particularly Nogo 66, a highly conserved 66-amino-acid loop of Nogo A (an isoform of RTN4), play key roles in limiting the intrinsic capacity of the central nervous system (CNS) to regenerate after injury. Ligation of surface Nogo receptors (NgRs) and/or leukocyte immunoglobulin-like receptor B2 (LILRB2) and its mouse orthologue the paired immunoglobulin-like receptor B (PIRB) by Nogo 66 transduces inhibitory signals that potently inhibit neurite outgrowth. Here, we show that soluble leukocyte immunoglobulin-like receptor A3 (LILRA3) is a high-affinity receptor for Nogo 66, suggesting that LILRA3 might be a competitive antagonist to these cell surface inhibitory receptors. Consistent with this, LILRA3 significantly reversed Nogo-66-mediated inhibition of neurite outgrowth and promoted synapse formation in primary cortical neurons through regulation of the ERK/MEK pathway. LILRA3 represents a new antagonist to Nogo-66-mediated inhibition of neurite outgrowth in the CNS, a function distinct from its immune-regulatory role in leukocytes. This report is also the first to demonstrate that a member of LILR family normally not expressed in rodents exerts functions on mouse neurons through the highly homologous Nogo 66 ligand.


Assuntos
Neuritos/metabolismo , Neurônios/citologia , Proteínas Nogo/metabolismo , Receptores Imunológicos/metabolismo , Sinapses/metabolismo , Animais , Células Cultivadas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese , Crescimento Neuronal , Neurônios/metabolismo , Proteínas Nogo/genética , Ligação Proteica , Receptores Imunológicos/genética , Sinapses/genética
3.
Immunol Cell Biol ; 95(5): 461-472, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28074060

RESUMO

S100A8 and S100A9 are myeloid cell-derived proteins that are elevated in several types of inflammatory lung disorders. Pro- and anti-inflammatory properties are reported and these proteins are proposed to activate TLR4. S100A8 and S100A9 can function separately, likely through distinct receptors but a systematic comparison of their effects in vivo are limited. Here we assess inflammation in murine lung following S100A9 and S100A8/A9 inhalation. Unlike S100A8, S100A9 promoted mild neutrophil and lymphocyte influx, possibly mediated in part, by increased mast cell degranulation and selective upregulation of some chemokine genes, particularly CXCL-10. S100 proteins did not significantly induce proinflammatory mediators including TNF-α, interleukin-1ß (IL-1ß), IL-6 or serum amyloid A3 (SAA3). In contrast to S100A8, neither preparation induced S100A8 or IL-10 mRNA/protein in airway epithelial cells, or in tracheal epithelial cells in vitro. Like S100A8, S100A9 and S100A8/A9 reduced neutrophil influx in acute lung injury provoked by lipopolysaccharide (LPS) challenge but were somewhat less inhibitory, possibly because of differential effects on expression of some chemokines, IL-1ß, SAA3 and IL-10. Novel common pathways including increased induction of an NAD+-dependent protein deacetylase sirtuin-1 that may reduce NF-κB signalling, and increased STAT3 activation may reduce LPS activation. Results suggest a role for these proteins in normal homeostasis and protective mechanisms in the lung.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Proteínas S100/metabolismo , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/patologia , Animais , Líquido da Lavagem Broncoalveolar , Quimiocina CXCL10/metabolismo , Feminino , Regulação da Expressão Gênica , Inflamação/genética , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos , Pulmão/metabolismo , Pulmão/patologia , Linfócitos/patologia , Camundongos Endogâmicos BALB C , Infiltração de Neutrófilos , Fosforilação , Transdução de Sinais/genética
4.
J Immunol ; 192(6): 2800-11, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24532576

RESUMO

S100A8 is considered proinflammatory by activating TLR4 and/or the receptor for advanced glycation end products. The aim was to investigate inflammatory effects of S100A8 in murine lung. S100A8 was administered to BALB/c mice by nasal inhalation and genes induced over a time-course assessed. LPS was introduced intranasally either alone or 2 h after pretreatment of mice with intranasal application of S100A8 or dexamethasone. A Cys(42)-Ala(42) mutant S100A8 mutant was used to assess whether S100A8's effects were via pathways that were dependent on reactive oxygen species. S100A8 induced IL-10 mRNA, and expression was apparent only in airway epithelial cells. Importantly, it suppressed acute lung injury provoked by LPS inhalation by suppressing mast-cell activation and induction of mediators orchestrating leukocyte recruitment, possibly by reducing NF-κB activation via an IκBα/Akt pathway and by downmodulating pathways generating oxidative stress. The Cys(42)-Ala(42) S100A8 mutant did not induce IL-10 and was less immunosuppressive, indicating modulation by scavenging oxidants. S100A8 inhibition of LPS-mediated injury was as potent, and outcomes were remarkably similar to immunosuppression by dexamethasone. We challenge the notion that S100A8 is an agonist for TLR4 or the receptor for advanced glycation end products. S100A8 induced IL-10 in vivo and initiates a feedback loop that attenuates acute lung injury.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Calgranulina A/administração & dosagem , Regulação da Expressão Gênica/efeitos dos fármacos , Interleucina-10/genética , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/metabolismo , Administração Intranasal , Animais , Anti-Inflamatórios/administração & dosagem , Western Blotting , Calgranulina A/genética , Análise por Conglomerados , Dexametasona/administração & dosagem , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Imuno-Histoquímica , Interleucina-10/metabolismo , Lipopolissacarídeos/administração & dosagem , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Mutantes/administração & dosagem , Proteínas Mutantes/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transcriptoma/efeitos dos fármacos , Transcriptoma/genética
5.
Mediators Inflamm ; 2015: 405629, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25767333

RESUMO

Glucocorticoids are commonly used for treating asthma and its exacerbations but have well-recognised adverse effects and are not always effective. Few alternative treatments exist. Using a murine model of an acute exacerbation of asthma, we assessed the ability of ISU201, a novel protein drug, to suppress the inflammatory response when administered after induction of an exacerbation. Sensitised mice were chronically challenged with a low mass concentration of aerosolised ovalbumin, and then received a single moderate-level challenge to simulate an allergen-induced exacerbation. ISU201 was administered to mice 2 and 8 hours later, while pulmonary inflammation and expression of mRNA for chemokines and proinflammatory cytokines were assessed after 4, 12, and 24 hours. Relative to vehicle-treated controls, ISU201 suppressed accumulation of pulmonary neutrophils and eosinophils, while accelerating the decline in CXCL1, TNF-α, and IL-6 in lavage fluid and lung tissue. ISU201 significantly reduced peak expression of mRNA for the chemokines Cxcl9 and Cxcl10, the adhesion molecules Icam1 and Vcam1, and the proinflammatory cytokines Il1b, Il12p40, and Csf1. The ability of ISU201 to promote resolution of inflammation suggests that it may have potential as an alternative to glucocorticoids in the management of asthma, including when administered after the onset of an acute exacerbation.


Assuntos
Antígenos CD/uso terapêutico , Asma/tratamento farmacológico , Asma/imunologia , Inflamação/tratamento farmacológico , Inflamação/imunologia , Pulmão/imunologia , Pulmão/metabolismo , Fragmentos de Peptídeos/uso terapêutico , Animais , Asma/metabolismo , Quimiocina CXCL10/genética , Quimiocina CXCL9/genética , Modelos Animais de Doenças , Feminino , Técnicas Imunoenzimáticas , Inflamação/metabolismo , Molécula 1 de Adesão Intercelular/genética , Interleucina-1beta/genética , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética
6.
Circ J ; 78(2): 271-80, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24389598

RESUMO

S100A8, S100A9 and S100A12 are considered proinflammatory mediators of atherosclerosis. Known as calgranulins, they are major components of neutrophils and are upregulated in macrophages and foam cells. They influence leukocyte recruitment, and may propagate inflammation by binding TLR4 and/or receptor for advanced glycation endproducts (RAGE). However, the receptors for calgranulins remain an enigma; we have no evidence for TLR4 or RAGE activation by S100A8 or S100A12. Moreover, gene regulation studies suggest antiinflammatory functions for S100A8 and emerging reports indicate pleiotropic roles. Unlike S100A9, S100A8 effectively scavenges oxidants generated by the myeloperoxidase system in vivo, forming novel thiol modifications. S100A8 is also readily S-nitrosylated, stabilizing nitric oxide and transporting it to hemoglobin. S100A8-SNO reduces leukocyte transmigration in the vasculature. S-glutathionylation of S100A9 modifies its effects on leukocyte adhesion. Both S100A8 forms inhibit mast cell activation, at least partially by scavenging reactive oxygen species required for signaling. Conversely, S100A12 activates and sequesters mast cells. However S100A12 suppresses proinflammatory cytokine induction by SAA-activated monocytes and macrophages, and inhibits matrix metalloprotease activity. We propose that the abundance and types of cells expressing calgranulins in particular microenvironments, their relative concentrations and post-translational modifications may have distinct functional outcomes, including those that are protective, at different stages of atherogenesis.


Assuntos
Aterosclerose/metabolismo , Sequestradores de Radicais Livres/metabolismo , Complexo Antígeno L1 Leucocitário/metabolismo , Transdução de Sinais , Aterosclerose/genética , Aterosclerose/patologia , Células Espumosas/metabolismo , Células Espumosas/patologia , Humanos , Complexo Antígeno L1 Leucocitário/genética , Oxidantes/metabolismo , Peroxidase/genética , Peroxidase/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
7.
bioRxiv ; 2023 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-37873248

RESUMO

Atherosclerosis is a chronic inflammatory disease which is driven in part by the aberrant trans -differentiation of vascular smooth muscle cells (SMCs). No therapeutic drug has been shown to reverse detrimental SMC-derived cell phenotypes into protective phenotypes, a hypothesized enabler of plaque regression and improved patient outcome. Herein, we describe a novel function of colchicine in the beneficial modulation of SMC-derived cell phenotype, independent of its conventional anti-inflammatory effects. Using SMC fate mapping in an advanced atherosclerotic lesion model, colchicine induced plaque regression by converting pathogenic SMC-derived macrophage-like and osteoblast-like cells into protective myofibroblast-like cells which thickened, and thereby stabilized, the fibrous cap. This was dependent on Notch3 signaling in SMC-derived plaque cells. These findings may help explain the success of colchicine in clinical trials relative to other anti-inflammatory drugs. Thus, we demonstrate the potential of regulating SMC phenotype in advanced plaque regression through Notch3 signaling, in addition to the canonical anti-inflammatory actions of drugs to treat atherosclerosis.

8.
Front Immunol ; 13: 826391, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35655772

RESUMO

Lung cancer is the leading cause of cancer-related death worldwide. Increasing evidence indicates a critical role for chronic inflammation in lung carcinogenesis. S100A8 is a protein with reported pro- and anti-inflammatory functions. It is highly expressed in myeloid-derived suppressor cells (MDSC) that accumulate in the tumor microenvironment and abrogate effective anti-cancer immune responses. Mechanisms of MDSC-mediated immunosuppression include production of reactive oxygen species and nitric oxide, and depletion of L-arginine required for T cell function. Although S100A8 is expressed in MDSC, its role in the lung tumor microenvironment is largely unknown. To address this, mouse recombinant S100A8 was repeatedly administered intranasally to mice bearing orthotopic lung cancers. S100A8 treatment prolonged survival from 19 days to 28 days (p < 0.001). At midpoint of survival, whole lungs and bronchoalveolar lavage fluid (BALF) were collected and relevant genes/proteins measured. We found that S100A8 significantly lowered expression of cytokine genes and proteins that promote expansion and activation of MDSC in lungs and BALF from cancer-bearing mice. Moreover, S100A8 enhanced activities of antioxidant enzymes and suppressed production of nitrite to create a lung microenvironment conducive to cytotoxic lymphocyte expansion and function. In support of this, we found decreased MDSC numbers, and increased numbers of CD4+ T cells and natural killer T (NK-T) cells in lungs from cancer-bearing mice treated with S100A8. Ex-vivo treatment of splenocytes with S100A8 protein activated NK cells. Our results indicate that treatment with S100A8 may favourably modify the lung microenvironment to promote an effective immune response in lungs, thereby representing a new strategy that could complement current immunotherapies in lung cancer.


Assuntos
Calgranulina A , Neoplasias Pulmonares , Animais , Calgranulina A/genética , Calgranulina A/metabolismo , Pulmão/metabolismo , Camundongos , Proteínas/metabolismo , Tórax , Microambiente Tumoral
9.
J Biol Chem ; 285(19): 14377-88, 2010 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-20223829

RESUMO

Reactive oxygen species generated by activated neutrophils can cause oxidative stress and tissue damage. S100A8 (A8) and S100A9 (A9), abundant in neutrophil cytoplasm, are exquisitely sensitive to oxidation, which may alter their functions. Murine A8 is a neutrophil chemoattractant, but it suppresses leukocyte transmigration in the microcirculation when S-nitrosylated. Glutathione (GSH) modulates intracellular redox, and S-glutathionylation can protect susceptible proteins from oxidative damage and regulate function. We characterized S-glutathionylation of A9; GSSG and GSNO generated S-glutathionylated A8 (A8-SSG) and A9 (A9-SSG) in vitro, whereas only A9-SSG was detected in cytosol of neutrophils activated with phorbol myristate acetate (PMA) but not with fMLP or opsonized zymosan. S-Glutathionylation exposed more hydrophobic regions in Zn(2+)-bound A9 but did not alter Zn(2+) binding affinity. A9-SSG had reduced capacity to form heterocomplexes with A8, but the arachidonic acid binding capacities of A8/A9 and A8/A9-SSG were similar. A9 and A8/A9 bind endothelial cells; S-glutathionylation reduced binding. We found little effect of A9 or A9-SSG on neutrophil CD11b/CD18 expression or neutrophil adhesion to endothelial cells. However, A9, A9-SSG and A8/A9 promoted neutrophil adhesion to fibronectin but, in the presence of A8, A9-mediated adhesion was abrogated by glutathionylation. S-Glutathionylation of A9 may protect its oxidation to higher oligomers and reduce neutrophil binding to the extracellular matrix. This may regulate the magnitude of neutrophil migration in the extravasculature, and together with the functional changes we reported for S-nitrosylated A8, particular oxidative modifications of these proteins may limit tissue damage in acute inflammation.


Assuntos
Calgranulina A/metabolismo , Calgranulina B/metabolismo , Adesão Celular , Glutationa/metabolismo , Inflamação , Neutrófilos/metabolismo , Calgranulina A/química , Calgranulina B/química , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Fibronectinas/metabolismo , Imunofluorescência , Glutationa/química , Humanos , Integrinas/metabolismo , Neutrófilos/citologia , Espectrometria de Massas por Ionização por Electrospray , Compostos de Sulfidrila/química , Compostos de Sulfidrila/metabolismo
10.
Amino Acids ; 41(4): 821-42, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20213444

RESUMO

This review focuses on new aspects of extracellular roles of the calgranulins. S100A8, S100A9 and S100A12 are constitutively expressed in neutrophils and induced in several cell types. The S100A8 and S100A9 genes are regulated by pro- and anti-inflammatory mediators and their functions may depend on cell type, mediators within a particular inflammatory milieu, receptors involved in their recognition and their post-translational modification. The S100A8 gene induction in macrophages is dependent on IL-10 and potentiated by immunosuppressive agents. S100A8 and S100A9 are oxidized by peroxide, hypochlorite and nitric oxide (NO). HOCl generates intra-chain sulfinamide bonds; stronger oxidation promotes cross-linked forms that are seen in human atheroma. S100A8 is >200-fold more sensitive to oxidative cross-linking than low-density lipoprotein and may reduce oxidative damage. S100A8 and S100A9 can be S-nitrosylated. S100A8-SNO suppresses mast cell activation and inflammation in the microcirculation and may act as an NO transporter to regulate vessel tone in inflammatory lesions. S100A12 activates mast cells and is a monocyte and mast cell chemoattractant; a G-protein-coupled mechanism may be involved. Structure-function studies are discussed in relation to conservation and divergence of functions in S100A8. S100A12 induces cytokines in mast cells, but not monocytes/macrophages. It forms complexes with Zn(2+) and, by chelating Zn(2+), S100A12 significantly inhibits MMPs. Zn(2+) in S100A12 complexes co-localize with MMP-9 in foam cells in atheroma. In summary, S100A12 has pro-inflammatory properties that are likely to be stable in an oxidative environment, because it lacks Cys and Met residues. Conversely, S100A8 and S100A9 oxidation and S-nitrosylation may have important protective mechanisms in inflammation.


Assuntos
Calgranulina A/genética , Calgranulina A/metabolismo , Calgranulina B/genética , Calgranulina B/metabolismo , Inflamação/metabolismo , Sequência de Aminoácidos , Animais , Movimento Celular , Regulação da Expressão Gênica , Interleucina-10/metabolismo , Leucócitos/citologia , Leucócitos/metabolismo , Mastócitos/metabolismo , Camundongos , Dados de Sequência Molecular , Processamento de Proteína Pós-Traducional , Proteínas S100/metabolismo , Proteína S100A12 , Relação Estrutura-Atividade , Receptor 4 Toll-Like/metabolismo , Zinco/metabolismo
11.
J Immunol ; 182(4): 2258-68, 2009 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19201880

RESUMO

The S100 calcium-binding proteins S100A8 and S100A9 are elevated systemically in patients with viral infections. The S100A8-S100A9 complex facilitated viral replication in human CD4(+) T lymphocytes latently infected with HIV-1- and S100A8-induced HIV-1 transcriptional activity. Mechanisms inducing the S100 genes and the potential source of these proteins following viral activation are unknown. In this study, we show that S100A8 was induced in murine macrophages, and S100A8 and S100A9 in human monocytes and macrophages, by polyinosinic:polycytidylic acid, a dsRNA mimetic. Induction was at the transcriptional level and was IL-10 dependent. Similar to LPS-induced S100A8, induction by dsRNA was dependent on p38 and ERK MAPK. Protein kinase R (PKR) mediates antiviral defense and participates in MyD88-dependent/independent signaling triggered by TLR4 or TLR3. Like IL-10, S100 induction by polyinosinic:polycytidylic acid and by LPS was inhibited by the specific PKR inhibitor 2-aminopurine, indicating a novel IL-10, PKR-dependent pathway. Other mediators such as IFN-beta, which synergized with dsRNA, may also be involved. C/EBPbeta bound the defined promoter region in response to dsRNA. S100A8 was expressed in lungs of mice infected with influenza virus and was maximal at day 8 with strong immunoreactivity in epithelial cells lining the airways and in mononuclear cells and declined early in the recovery phase, implying down-regulation by mediator(s) up-regulated during resolution of the infection. IL-10 is implicated in viral persistence. Since S100A8/S100A9 levels are likely to be maintained in conditions where IL-10 is raised, these proteins may contribute to viral persistence in patients infected by some RNA viruses.


Assuntos
Calgranulina A/genética , Regulação da Expressão Gênica/imunologia , Macrófagos/imunologia , Monócitos/imunologia , RNA de Cadeia Dupla/imunologia , Animais , Calgranulina A/metabolismo , Calgranulina B/imunologia , Calgranulina B/metabolismo , Ensaio de Imunoadsorção Enzimática , Expressão Gênica , Humanos , Macrófagos/virologia , Camundongos , Monócitos/virologia , Análise de Sequência com Séries de Oligonucleotídeos , Poli I-C/imunologia , Infecções por Vírus de RNA/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/imunologia , eIF-2 Quinase/imunologia , eIF-2 Quinase/metabolismo
12.
J Immunol ; 183(1): 593-603, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19542470

RESUMO

Macrophages, cytokines, and matrix metalloproteinases (MMP) play important roles in atherogenesis. The Ca(2+)-binding protein S100A12 regulates monocyte migration and may contribute to atherosclerosis by inducing proinflammatory cytokines in macrophages. We found significantly higher S100A12 levels in sera from patients with coronary artery disease than controls and levels correlated positively with C-reactive protein. S100A12 was released into the coronary circulation from ruptured plaque in acute coronary syndrome, and after mechanical disruption by percutaneous coronary intervention in stable coronary artery disease. In contrast to earlier studies, S100A12 did not stimulate proinflammatory cytokine production by human monocytes or macrophages. Similarly, no induction of MMP genes was found in macrophages stimulated with S100A12. Because S100A12 binds Zn(2+), we studied some functional aspects that could modulate atherogenesis. S100A12 formed a hexamer in the presence of Zn(2+); a novel Ab was generated that specifically recognized this complex. By chelating Zn(2+), S100A12 significantly inhibited MMP-2, MMP-9, and MMP-3, and the Zn(2+)-induced S100A12 complex colocalized with these in foam cells in human atheroma. S100A12 may represent a new marker of this disease and may protect advanced atherosclerotic lesions from rupture by inhibiting excessive MMP-2 and MMP-9 activities by sequestering Zn(2+).


Assuntos
Aterosclerose/metabolismo , Doença das Coronárias/metabolismo , Proteínas S100/fisiologia , Adulto , Idoso , Aterosclerose/patologia , Biomarcadores/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Doença das Coronárias/patologia , Feminino , Humanos , Mediadores da Inflamação/sangue , Mediadores da Inflamação/fisiologia , Macrófagos/enzimologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz , Pessoa de Meia-Idade , Ruptura Espontânea/enzimologia , Ruptura Espontânea/metabolismo , Ruptura Espontânea/prevenção & controle , Proteínas S100/sangue , Proteína S100A12 , Zinco/fisiologia
13.
Free Radic Biol Med ; 166: 255-264, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33539947

RESUMO

During vascular inflammation, the leukocyte-derived enzyme myeloperoxidase (MPO) is transcytosed across the endothelium and into the sub-endothelial extracellular matrix, where it promotes endothelial dysfunction by catalytically consuming nitric oxide (NO) produced by endothelial NO synthase (eNOS). In the presence of chloride ions and hydrogen peroxide (H2O2), MPO forms the oxidant hypochlorous acid (HOCl). Here we examined the short-term implications of HOCl produced by endothelial-transcytosed MPO for eNOS activity. Incubation of MPO with cultured aortic endothelial cells (ECs) resulted in its transport into the sub-endothelium. Exposure of MPO-containing ECs to low micromolar concentrations of H2O2 yielded enhanced rates of H2O2 consumption that correlated with HOCl formation and increased eNOS enzyme activity. The MPO-dependent activation of eNOS occurred despite reduced cellular uptake of the eNOS substrate l-arginine, which involved a decrease in the maximal activity (Vmax), but not substrate affinity (Km), of the major endothelial l-arginine transporter, cationic amino acid transporter-1. Activation of eNOS in MPO-containing ECs exposed to H2O2 involved a rapid elevation in cytosolic calcium and increased eNOS phosphorylation at Ser-1179 and de-phosphorylation at Thr-497. These signaling events were attenuated by intracellular calcium chelation, removal of extracellular calcium and inhibition of phospholipase C. This study shows that stimulation of endothelial-transcytosed MPO activates eNOS by promoting phospholipase C-dependent calcium signaling and altered eNOS phosphorylation at Ser-1179 and Thr-497. This may constitute a compensatory signaling response of ECs aimed at maintaining eNOS activity and NO production in the face of MPO-catalyzed oxidative stress.


Assuntos
Óxido Nítrico Sintase Tipo III , Peroxidase , Cálcio/metabolismo , Sinalização do Cálcio , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Peróxido de Hidrogênio/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Peroxidase/metabolismo , Fosfolipases Tipo C/metabolismo
14.
Immunol Cell Biol ; 88(1): 41-9, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19935766

RESUMO

Calgranulins comprise three proteins, S100A8 (Calgranulin A), S100A9 (Calgranulin B) and S100A12 (Calgranulin C) that are predominantly expressed by neutrophils, monocytes and activated macrophages. These S100 calcium-binding proteins are important molecular mediators in a range of diseases, including inflammatory arthritis, atherosclerosis and microbial infections. Much of the literature has focused on the pro-inflammatory functions of calgranulins, and this has tended to underplay important regulatory, anti-oxidant and protective properties. S100A8 and S100A9 are particularly complex in their actions because they exert intracellular and extracellular functions, they form a heterocomplex, S100A8-A9 (calprotectin), and have actions that are independent of or dependent on heterocomplex formation. In some circumstances S100A9 appears to regulate, rather than synergize with the actions of S100A8 and vice versa. Moreover, these calgranulins also bind zinc and other divalent cations and are sensitive to post-translational oxidative modifications, properties that also affect some functions. It is important to note that S100A8 has potent anti-oxidant activity, which could be important in host protection. Furthermore, although the genes for S100A8 and S100A9 are induced by activation of the toll-like receptor/interleukin-1 pathway, their expression is enhanced by interleukin-10 and glucocorticoids, thus suggesting a regulatory role in inflammation. On the other hand, S100A12 appears to be predominantly pro-inflammatory, particularly by its ability to activate mast cells. Measurement of S100A12 levels may be a highly sensitive biomarker for inflammatory disease activity. This review summarizes the current understanding of the biology of calgranulins, with a focus on their pleiotropic roles in inflammatory arthritis.


Assuntos
Artrite/imunologia , Complexo Antígeno L1 Leucocitário/imunologia , Animais , Antioxidantes/metabolismo , Artrite/genética , Artrite/metabolismo , Biomarcadores/metabolismo , Regulação da Expressão Gênica , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Complexo Antígeno L1 Leucocitário/genética , Complexo Antígeno L1 Leucocitário/metabolismo
15.
J Immunol ; 181(8): 5627-36, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18832721

RESUMO

S100A8 and S100A9, highly expressed by neutrophils, activated macrophages, and microvascular endothelial cells, are secreted during inflammatory processes. Our earlier studies showed S100A8 to be an avid scavenger of oxidants, and, together with its dependence on IL-10 for expression in macrophages, we postulated that this protein has a protective role. S-nitrosylation is an important posttranslational modification that regulates NO transport, cell signaling, and homeostasis. Relatively few proteins are targets of S-nitrosylation. To date, no inflammation-associated proteins with NO-shuttling capacity have been identified. We used HPLC and mass spectrometry to show that S100A8 and S100A9 were readily S-nitrosylated by NO donors. S-nitrosylated S100A8 (S100A8-SNO) was the preferred nitrosylated product. No S-nitrosylation occurred when the single Cys residue in S100A8 was mutated to Ala. S100A8-SNO in human neutrophils treated with NO donors was confirmed by the biotin switch assay. The stable adduct transnitrosylated hemoglobin, indicating a role in NO transport. S100A8-SNO suppressed mast cell activation by compound 48/80; intravital microscopy was used to demonstrate suppression of leukocyte adhesion and extravasation triggered by compound 48/80 in the rat mesenteric microcirculation. Although S100A8 is induced in macrophages by LPS or IFN-gamma, the combination, which activates inducible NO synthase, did not induce S100A8. Thus, the antimicrobial functions of NO generated under these circumstances would not be compromised by S100A8. Our results suggest that S100A8-SNO may regulate leukocyte-endothelial cell interactions in the microcirculation, and suppression of mast cell-mediated inflammation represents an additional anti-inflammatory property for S100A8.


Assuntos
Anti-Inflamatórios não Esteroides/imunologia , Calgranulina A/imunologia , Calgranulina B/imunologia , Sequestradores de Radicais Livres/imunologia , Leucócitos/imunologia , Processamento de Proteína Pós-Traducional/imunologia , Animais , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/farmacologia , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/imunologia , Calgranulina A/química , Calgranulina B/química , Cisteína/química , Cisteína/imunologia , Células Endoteliais/imunologia , Sequestradores de Radicais Livres/química , Sequestradores de Radicais Livres/farmacologia , Hemoglobinas/imunologia , Humanos , Inflamação/imunologia , Interferon gama/farmacologia , Interleucina-10/imunologia , Lipopolissacarídeos/farmacologia , Mastócitos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Microcirculação/imunologia , Óxido Nítrico/imunologia , Óxido Nítrico Sintase Tipo II/imunologia , Oxidantes/imunologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Ratos , p-Metoxi-N-metilfenetilamina/farmacologia
16.
J Leukoc Biol ; 83(2): 334-43, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17998301

RESUMO

The leukocyte Ig-like receptors (LILRs) comprise a family of cell-surface immunoregulatory receptors with activating and inhibitory members. The inhibitory LILRs possess cytoplasmic ITIMs that down-regulate signaling by nonreceptor tyrosine kinase cascades. The activating members have a truncated cytoplasmic domain and signal through the FcR gamma chain. We examined the expression of LILRs on human mast cells during their development in vitro. Progenitor mast cells expressed cell surface inhibitory LILRB1, -B2, -B3, and -B4 and activating LILRA1. However, although mature cord blood-derived mast cells (hMCs) had detectable mRNA encoding multiple LILRs, none were expressed on the cell surface. Culture of progenitor mast cells or hMCs with various cytokine combinations failed to retain or induce cell surface expression of the LILRs. It is interesting that hMCs expressed LILRB5 in cytoplasmic granules and upon cross-linking of the high-affinity IgE receptor, released LILRB5 into the culture medium. Our results demonstrate that LILRs are developmentally regulated in human mast cells and that LILRB5 is expressed in mast cell granules and the release of soluble LILRB5 following IgE FcR-dependent stimulation, which has potential for amplification of mast cell-dependent, inflammatory responses.


Assuntos
Antígenos CD/biossíntese , Sangue Fetal/citologia , Células-Tronco Hematopoéticas/metabolismo , Mastócitos/metabolismo , Glicoproteínas de Membrana/biossíntese , Receptores de Superfície Celular/biossíntese , Receptores Imunológicos/biossíntese , Animais , Anticorpos Monoclonais/imunologia , Antígenos CD/genética , Diferenciação Celular/genética , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Citocinas/farmacologia , Grânulos Citoplasmáticos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Hematopoéticas/citologia , Humanos , Receptor B1 de Leucócitos Semelhante a Imunoglobulina , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/biossíntese , Agregação de Receptores , Receptores de Superfície Celular/genética , Receptores de IgE/fisiologia , Receptores Imunológicos/genética , Proteínas Recombinantes/farmacologia , Fator de Células-Tronco/farmacologia
17.
Front Immunol ; 10: 380, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30899260

RESUMO

Elevated serum amyloid A (SAA) levels may promote endothelial dysfunction, which is linked to cardiovascular and renal pathologies. We investigated the effect of SAA on vascular and renal function in apolipoprotein E-deficient (ApoE-/-) mice. Male ApoE-/- mice received vehicle (control), low-level lipopolysaccharide (LPS), or recombinant human SAA by i.p. injection every third day for 2 weeks. Heart, aorta and kidney were harvested between 3 days and 18 weeks after treatment. SAA administration increased vascular cell adhesion molecule (VCAM)-1 expression and circulating monocyte chemotactic protein (MCP)-1 and decreased aortic cyclic guanosine monophosphate (cGMP), consistent with SAA inhibiting nitric oxide bioactivity. In addition, binding of labeled leukocytes to excised aorta increased as monitored using an ex vivo leukocyte adhesion assay. Renal injury was evident 4 weeks after commencement of SAA treatment, manifesting as increased plasma urea, urinary protein, oxidized lipids, urinary kidney injury molecule (KIM)-1 and multiple cytokines and chemokines in kidney tissue, relative to controls. Phosphorylation of nuclear-factor-kappa-beta (NFκB-p-P65), tissue factor (TF), and macrophage recruitment increased in kidneys from ApoE-/- mice 4 weeks after SAA treatment, confirming that SAA elicited a pro-inflammatory and pro-thrombotic phenotype. These data indicate that SAA impairs endothelial and renal function in ApoE-/- mice in the absence of a high-fat diet.


Assuntos
Vasos Sanguíneos/metabolismo , Nefropatias/etiologia , Nefropatias/metabolismo , Animais , Aorta/metabolismo , Aorta/patologia , Aorta/fisiopatologia , Apolipoproteínas E/deficiência , Biomarcadores , Vasos Sanguíneos/patologia , Vasos Sanguíneos/fisiopatologia , Citocinas/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Células Endoteliais/metabolismo , Imuno-Histoquímica , Mediadores da Inflamação/metabolismo , Nefropatias/patologia , Nefropatias/fisiopatologia , Testes de Função Renal , Lipídeos/sangue , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Peroxidase/metabolismo , Proteína Amiloide A Sérica/metabolismo
18.
Can J Gastroenterol ; 22(5): 461-4, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18478131

RESUMO

The expression of the inflammatory S100 calgranulin proteins (S100A8, S100A9 and S100A12) in normal and Helicobacter pylori-infected gastric mucosa of children were examined. S100A8, S100A9 and S100A12, which were virtually absent in normal gastric mucosa, were highly expressed in H pylori-infected mucosa. This expression correlated with the severity of gastritis (r=0.9422, P<0.05). S100 calgranulins may be involved in bacterial-induced gastritis and may limit bacterial growth.


Assuntos
Gastrite/metabolismo , Gastrite/microbiologia , Infecções por Helicobacter/metabolismo , Helicobacter pylori , Proteínas S100/metabolismo , Adolescente , Estudos de Casos e Controles , Criança , Endoscopia do Sistema Digestório , Feminino , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Gastrite/patologia , Infecções por Helicobacter/patologia , Humanos , Masculino , Índice de Gravidade de Doença
19.
Innate Immun ; 23(4): 381-391, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28409541

RESUMO

The leukocyte Ig-like receptor B4 (LILRB4) is an inhibitory cell surface receptor, primarily expressed on mono-myeloid cells. It contains 2 C-type Ig-like extracellular domains and a long cytoplasmic domain that contains three intracellular immunoreceptor tyrosine-based inhibitory motifs (ITIMs). Data suggest that LILRB4 suppresses Fc receptor-dependent monocyte functions via its ITIMs, but relative contributions of the three ITIMs are not characterised. To address this, tyrosine (Tyr) residues at positions 337, 389 and 419 were single, double or triple mutated to phenylalanine and stably transfected into a human monocytic cell line, THP-1. Intact Tyr389 was sufficient to maximally inhibit FcγRI-mediated TNF-α production in THP-1 cells, but, paradoxically, Tyr337 significantly enhanced TNF-α production. In contrast, bactericidal activity was significantly enhanced in mutants containing Tyr419, while Tyr337 markedly inhibited bacteria killing. Taken together, these results indicate that LILRB4 might have dual inhibitory and activating functions, depending on the position of the functional tyrosine residues in its ITIMs and/or the nature of the stimuli.


Assuntos
Monócitos/fisiologia , Mutação/genética , Células Mieloides/fisiologia , Domínios Proteicos/genética , Receptores de Superfície Celular/metabolismo , Tirosina/genética , Bacteriólise/genética , Linhagem Celular , Humanos , Imunomodulação , Glicoproteínas de Membrana , Mutagênese Sítio-Dirigida , Fosforilação , Receptores de Superfície Celular/genética , Receptores de IgG/metabolismo , Receptores Imunológicos , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
20.
Sci Rep ; 6: 35085, 2016 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-27725776

RESUMO

FcγRI cross-linking on monocytes may trigger clathrin-mediated endocytosis, likely through interaction of multiple intracellular molecules that are controlled by phosphorylation and dephosphorylation events. However, the identity of phospho-proteins and their regulation are unknown. We proposed the leukocyte immunoglobulin-like receptor B4 (LILRB4) that inhibits FcγRI-mediated cytokine production via Tyr dephosphorylation of multiple kinases, may also regulate endocytosis/phagocytosis through similar mechanisms. FcγRI and/or LILRB4 were antibody-ligated on THP-1 cells, lysates immunoprecipitated using anti-pTyr antibody and peptides sequenced by mass spectrometry. Mascot Search identified 25 Tyr phosphorylated peptides with high confidence. Ingenuity Pathway Analysis revealed that the most significantly affected pathways were clathrin-mediated endocytosis and Fc-receptor dependent phagocytosis. Tyr phosphorylation of key candidate proteins in these pathways included common γ-chain of the Fc receptors, Syk, clathrin, E3 ubiquitin protein ligase Cbl, hepatocyte growth factor-regulated tyrosine kinase substrate, tripartite motif-containing 21 and heat shock protein 70. Importantly, co-ligation of LILRB4 with FcγRI caused significant dephosphorylation of these proteins and was associated with suppression of Fc receptor-dependent uptake of antibody-opsonised bacterial particles, indicating that LILRB4. These results suggest that Tyr phosphorylation may be critical in FcγRI-dependent endocytosis/phagocytosis that may be regulated by LILRB4 by triggering dephosphorylation of key signalling proteins.


Assuntos
Clatrina/metabolismo , Endocitose , Monócitos/imunologia , Receptores de Superfície Celular/metabolismo , Receptores de IgG/metabolismo , Humanos , Glicoproteínas de Membrana , Receptores Imunológicos , Células THP-1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA