Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Gastroenterology ; 159(6): 2130-2145.e5, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32805279

RESUMO

BACKGROUND & AIMS: After birth, the immune system matures via interactions with microbes in the gut. The S100 calcium binding proteins S100A8 and S100A9, and their extracellular complex form, S100A8-A9, are found in high amounts in human breast milk. We studied levels of S100A8-A9 in fecal samples (also called fecal calprotectin) from newborns and during infancy, and their effects on development of the intestinal microbiota and mucosal immune system. METHODS: We collected stool samples (n = 517) from full-term (n = 72) and preterm infants (n = 49) at different timepoints over the first year of life (days 1, 3, 10, 30, 90, 180, and 360). We measured levels of S100A8-A9 by enzyme-linked immunosorbent assay and analyzed fecal microbiomes by 16S sRNA gene sequencing. We also obtained small and large intestine biopsies from 8 adults and 10 newborn infants without inflammatory bowel diseases (controls) and 8 infants with necrotizing enterocolitis and measured levels of S100A8 by immunofluorescence microscopy. Children were followed for 2.5 years and anthropometric data and medical information on infections were collected. We performed studies with newborn C57BL/6J wild-type and S100a9-/- mice (which also lack S100A8). Some mice were fed or given intraperitoneal injections of S100A8 or subcutaneous injections of Staphylococcus aureus. Blood and intestine, mesenterial and celiac lymph nodes were collected; cells and cytokines were measured by flow cytometry and studied in cell culture assays. Colon contents from mice were analyzed by culture-based microbiology assays. RESULTS: Loss of S100A8 and S100A9 in mice altered the phenotypes of colonic lamina propria macrophages, compared with wild-type mice. Intestinal tissues from neonatal S100-knockout mice had reduced levels of CX3CR1 protein, and Il10 and Tgfb1 mRNAs, compared with wild-type mice, and fewer T-regulatory cells. S100-knockout mice weighed 21% more than wild-type mice at age 8 weeks and a higher proportion developed fatal sepsis during the neonatal period. S100-knockout mice had alterations in their fecal microbiomes, with higher abundance of Enterobacteriaceae. Feeding mice S100 at birth prevented the expansion of Enterobacteriaceae, increased numbers of T-regulatory cells and levels of CX3CR1 protein and Il10 mRNA in intestine tissues, and reduced body weight and death from neonatal sepsis. Fecal samples from term infants, but not preterm infants, had significantly higher levels of S100A8-A9 during the first 3 months of life than fecal samples from adults; levels decreased to adult levels after weaning. Fecal samples from infants born by cesarean delivery had lower levels of S100A8-A9 than from infants born by vaginal delivery. S100 proteins were expressed by lamina propria macrophages in intestinal tissues from infants, at higher levels than in intestinal tissues from adults. High fecal levels of S100 proteins, from 30 days to 1 year of age, were associated with higher abundance of Actinobacteria and Bifidobacteriaceae, and lower abundance of Gammaproteobacteria-particularly opportunistic Enterobacteriaceae. A low level of S100 proteins in infants' fecal samples associated with development of sepsis and obesity by age 2 years. CONCLUSION: S100A8 and S100A9 regulate development of the intestinal microbiota and immune system in neonates. Nutritional supplementation with these proteins might aide in development of preterm infants and prevent microbiota-associated disorders in later years.


Assuntos
Calgranulina A/metabolismo , Calgranulina B/metabolismo , Disbiose/imunologia , Microbioma Gastrointestinal/imunologia , Adulto , Animais , Biópsia , Calgranulina A/administração & dosagem , Calgranulina A/análise , Calgranulina B/análise , Calgranulina B/genética , Pré-Escolar , Colo/microbiologia , Colo/patologia , DNA Bacteriano/genética , DNA Bacteriano/isolamento & purificação , Disbiose/microbiologia , Disbiose/prevenção & controle , Enterocolite Necrosante/epidemiologia , Enterocolite Necrosante/imunologia , Enterocolite Necrosante/microbiologia , Enterocolite Necrosante/prevenção & controle , Fezes/química , Fezes/microbiologia , Feminino , Seguimentos , Microbioma Gastrointestinal/genética , Humanos , Imunidade nas Mucosas , Lactente , Recém-Nascido , Recém-Nascido Prematuro/imunologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos Knockout , Obesidade/epidemiologia , Obesidade/imunologia , Obesidade/microbiologia , Obesidade/prevenção & controle , RNA Ribossômico 16S/genética , Sepse/epidemiologia , Sepse/imunologia , Sepse/microbiologia , Sepse/prevenção & controle
2.
Int Immunopharmacol ; 67: 239-247, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30562685

RESUMO

Activation of the inflammatory signaling pathway is the most vital part of the pre-metastatic events of breast cancer. Platycodin D (PlaD) shows favorable pharmacological activities in anti-inflammatory and anti-tumor effect. The main purpose of this study was to survey the effects of PlaD on S100A8/A9-induced inflammation in mouse mammary carcinoma 4T1 cells. S100A8/A9 immunolocalization and expression in pre-metastatic lung tissue were assessed by immunofluorescence staining and ELISA. 4T1 cells were treated with 2.5 µg/mL recombinant S100A8/A9 heterodimer and 7.5, 10, or 12.5 µM of PlaD. After 24 h of incubation, cell viability, migration, and invasion were evaluated by CCK-8, wound-healing, and transwell assay, respectively. Nuclear translocation of NF-κB p65 was determined by immunostaining and western blot. The levels of pro-inflammatory cytokines including IL-1ß, IL-6, and TNF-α were detected by ELISA. The results showed that S100A8/A9 was actively increased and released into the extracellular space during the pre-metastatic phase of breast cancer. PlaD treatment attenuated S100A8/A9-induced growth, migration, and invasion of 4T1 cells. Furthermore, PlaD decreased the levels of IL-1ß, IL-6, and TNF-α by inhibiting nuclear translocation of NF-κB p65. In conclusion, this study demonstrated that PlaD inhibited S100A8/A9-induced inflammatory response in 4T1 cells by suppressing the expression of IL-6, IL-1ß, and TNF-α via inhibition of NF-κB signaling pathways.


Assuntos
Calgranulina A/administração & dosagem , Calgranulina B/toxicidade , Neoplasias Mamárias Animais/metabolismo , Neoplasias Experimentais/tratamento farmacológico , Saponinas/farmacologia , Triterpenos/farmacologia , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citocinas/genética , Citocinas/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Molecular , Neoplasias Experimentais/metabolismo , Saponinas/química , Triterpenos/química
3.
Int J Mol Sci ; 18(7)2017 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-28672877

RESUMO

Pseudomonas aeruginosa biofilm maintains and perturbs local host defense, hindering timely wound healing. Previously, we showed that P. aeruginosa suppressed S100A8/A9 of the murine innate host defense. We assessed the potential antimicrobial effect of S100A8/A9 on biofilm-infected wounds in a murine model and P. aeruginosa growth in vitro. Seventy-six mice, inflicted with a full-thickness burn wound were challenged subcutaneously (s.c.) by 106 colony-forming units (CFUs) of P. aeruginosa biofilm. Mice were subsequently randomized into two treatment groups, one group receiving recombinant murine S100A8/A9 and a group of vehicle controls (phosphate-buffered saline, PBS) all treated with s.c. injections daily for up to five days. Wounds were analyzed for quantitative bacteriology and contents of key inflammatory markers. Count of blood polymorphonuclear leukocytes was included. S100A8/A9-treatment ameliorated wound infection, as evaluated by quantitative bacteriology (p ≤ 0.05). In vitro, growth of P. aeruginosa was inhibited dose-dependently by S100A8/A9 in concentrations from 5 to 40 µg/mL, as determined by optical density-measurement (OD-measurement) and quantitative bacteriology. Treatment slightly augmented key inflammatory cytokine Tumor Necrosis Factor-α (TNF-α), but dampened interferon-γ (IFN-γ) levels and blood polymorphonuclear count. In conclusion, topical S100A8/A9 displays remarkable novel immune stimulatory and anti-infective properties in vivo and in vitro. Importantly, treatment by S100A8/A9 provides local infection control. Implications for a role as adjunctive treatment in healing of chronic biofilm-infected wounds are discussed.


Assuntos
Antibacterianos/administração & dosagem , Biofilmes/efeitos dos fármacos , Calgranulina A/administração & dosagem , Calgranulina B/administração & dosagem , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/imunologia , Infecção dos Ferimentos/imunologia , Infecção dos Ferimentos/microbiologia , Administração Tópica , Animais , Biomarcadores , Doença Crônica , Citocinas/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Fatores Imunológicos , Mediadores da Inflamação/metabolismo , Contagem de Leucócitos , Camundongos , Neutrófilos/imunologia , Neutrófilos/metabolismo , Infecções por Pseudomonas/tratamento farmacológico , Infecções por Pseudomonas/metabolismo , Infecção dos Ferimentos/tratamento farmacológico , Infecção dos Ferimentos/metabolismo
4.
Biol Res ; 50(1): 23, 2017 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-28637501

RESUMO

BACKGROUND: Airway remodeling is a key feature of asthma, characterized by increased proliferation of airway smooth muscle cells (ASMCs). S100A8 is a calcium-binding protein with a potential to regulate cell proliferation. Here, the effect of exogenous S100A8 protein on the proliferation of ASMCs induced by platelet-derived growth factor (PDGF) and the underlying molecular mechanism was investigated. METHODS: Rat ASMCs were cultured with or without a neutralizing antibody to the receptor for advanced glycation end-products (RAGE), a potential receptor for S100A8 protein. Purified recombinant rat S100A8 protein was then added into the cultured cells, and the proliferation of ASMCs induced by PDGF was detected by colorimetric-based WST-8 assay and ampedance-based xCELLigence proliferation assay. The expression levels of RAGE in ASMCs were analyzed using western blotting assay. RESULTS: Results showed that exogenous S100A8 inhibited the PDGF-induced proliferation of rat ASMCs in a dose-dependent manner with the maximal effect at 1 µg/ml in vitro. Furthermore, when ASMCs was pre-treated with anti-RAGE neutralizing antibody, the inhibitory effect of S100A8 on PDGF-induced proliferation was significantly suppressed. In addition, neither the treatment with S100A8 or PDGF alone nor the pre-treatment with rS100A8 followed by PDGF stimulation affected the expression levels of RAGE. CONCLUSIONS: Our study demonstrated that S100A8 inhibits PDGF-induced ASMCs proliferation in a manner dependent on membrane receptor RAGE.


Assuntos
Calgranulina A/administração & dosagem , Proliferação de Células/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/agonistas , Receptor para Produtos Finais de Glicação Avançada/efeitos dos fármacos , Animais , Células Cultivadas , Ratos
5.
Biol. Res ; 50: 23, 2017. graf
Artigo em Inglês | LILACS | ID: biblio-950874

RESUMO

BACKGROUND: Airway remodeling is a key feature of asthma, characterized by increased proliferation of airway smooth muscle cells (ASMCs). S100A8 is a calcium-binding protein with a potential to regulate cell proliferation. Here, the effect of exogenous S100A8 protein on the proliferation of ASMCs induced by platelet-derived growth factor (PDGF) and the underlying molecular mechanism was investigated. METHODS: Rat ASMCs were cultured with or without a neutralizing antibody to the receptor for advanced glycation end-products (RAGE), a potential receptor for S100A8 protein. Purified recombinant rat S100A8 protein was then added into the cultured cells, and the proliferation of ASMCs induced by PDGF was detected by colorimetric-based WST-8 assay and ampedance-based xCELLigence proliferation assay. The expression levels of RAGE in ASMCs were analyzed using western blotting assay. RESULTS: Results showed that exogenous S100A8 inhibited the PDGF-induced proliferation of rat ASMCs in a dose- dependent manner with the maximal effect at 1 µg/ml in vitro. Furthermore, when ASMCs was pre-treated with anti-RAGE neutralizing antibody, the inhibitory effect of S100A8 on PDGF-induced proliferation was significantly suppressed. In addition, neither the treatment with S100A8 or PDGF alone nor the pre-treatment with rS100A8 followed by PDGF stimulation affected the expression levels of RAGE. CONCLUSIONS: Our study demonstrated that S100A8 inhibits PDGF-induced ASMCs proliferation in a manner dependent on membrane receptor RAGE.


Assuntos
Animais , Ratos , Fator de Crescimento Derivado de Plaquetas/agonistas , Miócitos de Músculo Liso/efeitos dos fármacos , Calgranulina A/administração & dosagem , Proliferação de Células/efeitos dos fármacos , Receptor para Produtos Finais de Glicação Avançada/efeitos dos fármacos , Células Cultivadas
6.
Biochem Biophys Res Commun ; 472(1): 243-9, 2016 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-26920052

RESUMO

S100A8 is an important member of the S100 protein family, which is involved in intracellular and extracellular regulatory activities. We previously reported that the S100A8 protein was differentially expressed in the asthmatic respiratory tracts. To understand the potential role of S100A8 in asthma, we investigated the effect of recombinant S100A8 protein on the platelet-derived growth factor (PDGF)-induced migration of airway smooth muscle cells (ASMCs) and the underlying molecular mechanism by using multiple methods, such as impedance-based xCELLigence migration assay, transwell migration assays and wound-healing assays. We found that exogenous S100A8 protein significantly inhibited PDGF-induced ASMC migration. Furthermore, the migration inhibition effect of S100A8 was blocked by neutralizing antibody against the receptor for advanced glycation end-products (RAGE), a potential receptor for the S100A8 protein. These findings provide direct evidence that exogenous S100A8 protein inhibits the PDGF-induced migration of ASMCs through the membrane receptor RAGE. Our study highlights a novel role of S100A8 as a potential means of counteracting airway remodeling in chronic airway diseases.


Assuntos
Calgranulina A/fisiologia , Movimento Celular/fisiologia , Miócitos de Músculo Liso/fisiologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Traqueia/patologia , Traqueia/fisiologia , Animais , Anticorpos Neutralizantes , Asma/patologia , Asma/fisiopatologia , Calgranulina A/administração & dosagem , Calgranulina A/genética , Células Cultivadas , Modelos Animais de Doenças , Ratos , Receptor para Produtos Finais de Glicação Avançada/antagonistas & inibidores , Receptor para Produtos Finais de Glicação Avançada/imunologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Cicatrização
7.
Arthritis Rheumatol ; 67(11): 2933-43, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26245312

RESUMO

OBJECTIVE: To determine whether selected damage-associated molecular patterns (DAMPs) present in the osteoarthritic (OA) joints of mice excite nociceptors through Toll-like receptor 4 (TLR-4). METHODS: The ability of S100A8 and α2 -macroglobulin to excite nociceptors was determined by measuring the release of monocyte chemoattractant protein 1 (MCP-1) by cultured dorsal root ganglion (DRG) cells as well as by measuring the intracellular calcium concentration ([Ca(2+) ]i ) in cultured DRG neurons from naive mice or from mice that had undergone surgical destabilization of the medial meniscus (DMM) 8 weeks previously. The role of TLR-4 was assessed using TLR-4(-/-) cells or a TLR-4 inhibitor. The [Ca(2+) ]i in neurons within ex vivo intact DRGs was measured in samples from Pirt-GCaMP3 mice. Neuronal expression of the Tlr4 gene was determined by in situ hybridization. DMM surgery was performed in wild-type and TLR-4(-/-) mice; mechanical allodynia was monitored, and joint damage was assessed histologically after 16 weeks. RESULTS: DRG neurons from both naive and DMM mice expressed Tlr4. Both S100A8 and α2 -macroglobulin stimulated release of the proalgesic chemokine MCP-1 in DRG cultures, and the neurons rapidly responded to S100A8 and α2 -macroglobulin with increased [Ca(2+) ]i . Blocking TLR-4 inhibited these effects. Neurons within intact DRGs responded to the TLR-4 agonist lipopolysaccharide. In both of the calcium-imaging assays, it was primarily the nociceptor population of neurons that responded to TLR-4 ligands. TLR-4(-/-) mice were not protected from mechanical allodynia or from joint damage associated with DMM. CONCLUSION: Our experiments suggest a role of TLR-4 signaling in the excitation of nociceptors by selected DAMPs. Further research is needed to delineate the importance of this pathway in relation to OA pain.


Assuntos
Neurônios/metabolismo , Nociceptores/metabolismo , Osteoartrite/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Cálcio/metabolismo , Calgranulina A/administração & dosagem , Células Cultivadas , Quimiocina CCL2/metabolismo , Modelos Animais de Doenças , Feminino , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Masculino , Camundongos , Camundongos Knockout , Neurônios/efeitos dos fármacos , Receptor 4 Toll-Like/genética , alfa-Macroglobulinas/farmacologia
8.
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
9.
Mol Immunol ; 53(4): 443-9, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23127860

RESUMO

Polymorphonuclear neutrophils (PMNs) produce and release copious amounts of reactive oxygen species (ROS) which target potential bacterial invaders but also contribute to the inflammation-associated organ injuries seen in sepsis. Calprotectin is an immune regulatory protein complex made of S100A8 and S100A9 that inhibits the oxidative metabolism of PMNs in vitro, an effect that can be potentiated by the controlled activation of the protease activated receptor-2 (PAR2). The aim of this study was to test the use of a dual strategy of calprotectin and PAR2 administration to mitigate the deleterious inflammation seen in sepsis. We hypothesized that exogenous calprotectin would protect against the injuries produced by lipopolysaccharides (LPS)-induced endotoxemia and that the controlled activation of PAR2 would potentiate this beneficial effect. Exogenous S100A8 and/or a PAR2 activating peptide (PAR2 AP) were administered in a mouse model of LPS induced endotoxemia. The survival rates as well as markers of inflammation and oxidative damage were measured in the lungs, kidneys, and livers of endotoxemic mice. Mice treated with S100A8 following LPS had less PMN infiltration and less severe histological changes in their lungs, kidneys, and livers. A significantly lower score of oxidative damage in the livers and lungs of S100A8/LPS treated mice was also noted when compared to mice treated with LPS alone. This protective and anti-inflammatory effect of S100A8 was potentiated by the controlled activation of PAR2. Finally, in further support to our hypothesis, the survival rate was almost doubled from 33% to 65% and 63% in mice treated by, respectively, S100A8 and PAR2 AP, whereas 85% of the mice treated with both PAR2 AP and S100A8 survived, a statistically significant higher rate. These results support an anti-inflammatory, anti-oxidative, and protective effect of S100A8 in sepsis, and warrant further studies on the role of PAR2.


Assuntos
Anti-Inflamatórios/farmacologia , Calgranulina A/administração & dosagem , Endotoxemia/tratamento farmacológico , Neutrófilos/efeitos dos fármacos , Oligopeptídeos/farmacologia , Sepse/tratamento farmacológico , Animais , Endotoxemia/induzido quimicamente , Endotoxemia/imunologia , Endotoxemia/mortalidade , Feminino , Rim/efeitos dos fármacos , Rim/imunologia , Rim/metabolismo , Lipopolissacarídeos , Fígado/efeitos dos fármacos , Fígado/imunologia , Fígado/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/metabolismo , Camundongos , Neutrófilos/imunologia , Neutrófilos/metabolismo , Oxirredução , Sepse/induzido quimicamente , Sepse/imunologia , Sepse/mortalidade , Taxa de Sobrevida
10.
Inflammation ; 35(2): 409-19, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21487906

RESUMO

The immunological properties of rat S100A8 (r-S100A8) and S100A9 (r-S100A9) in immune cells are poorly understood. Enzyme-linked immunosorbent assay (ELISA) for r-S100A9 enabled us to discuss the differential functional roles of the two proteins, and their localization in the cells was observed microscopically. Recombinant human S100A8 (rh-S100A8) or S100A9 (rh-S100A9) were intravenously administrated into rats with LPS-induced liver damage. ELISA was used to measure the serum concentration of S100A9 in the rats. Western blotting and a preparative ELISA were used to prove specificity and avidity of monoclonal antibodies for r-S100A8 and r-S100A9. Immunohistochemical staining was carried out to visualize intracellular localization of the two proteins in the immune cells using the antibodies. When rh-S100A8 was intravenously injected in the rats (B group), the serum concentration of r-S100A9 apparently decreased as compared with that of the positive control rats (A group). The activities of AST, ALT, and LD in the rat sera (B group) also significantly went down in comparison with those of the rats (A group). Although both the S100A8 and S100A9 were abundantly expressed in activated immune cells, quite difference of not only their intracellular localization but also distribution of the cells expressing the two proteins was microscopically observed. In the rats (B group), less number of the immune cells or less amount of r-S100A8 and r-S100A9 in the cells than those of the rats (A group) was also seen. The r-S100A8 could serve as a regulator of acute inflammatory reaction in the rats with LPS-induced damage.


Assuntos
Calgranulina A/imunologia , Calgranulina B/imunologia , Inflamação/imunologia , Animais , Calgranulina A/administração & dosagem , Calgranulina A/biossíntese , Calgranulina B/biossíntese , Calgranulina B/sangue , Doença Hepática Induzida por Substâncias e Drogas , Humanos , Mediadores da Inflamação , Lipopolissacarídeos/imunologia , Macrófagos/imunologia , Masculino , Neutrófilos/imunologia , Ratos , Ratos Wistar , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia
11.
Arthritis Rheum ; 64(5): 1477-87, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22127564

RESUMO

OBJECTIVE: S100A8 and S100A9 are two Ca(2+) binding proteins classified as damage-associated molecular patterns or alarmins that are found in high amounts in the synovial fluid of osteoarthritis (OA) patients. The purpose of this study was to investigate whether S100A8 and/or S100A9 can interact with chondrocytes from OA patients to increase catabolic mediators. METHODS: Using immunohistochemistry, we stained for S100A8 and S100A9 protein, matrix metalloproteinases (MMPs), and a cartilage-breakdown epitope specific for MMPs (VDIPEN) in cartilage from OA donors. Isolated chondrocytes or explants from OA and non-OA donors were stimulated with S100A8 and/or S100A9. Messenger RNA and protein levels of MMPs, cytokines, and cartilage matrix molecules were determined with quantitative reverse transcription-polymerase chain reaction and Luminex techniques, respectively. For receptor blocking studies, specific inhibitors for Toll-like receptor 4 (TLR-4), receptor for advanced glycation end products (RAGE), and carboxylated glycans were used. RESULTS: In cartilage from OA patients, the expression of S100A8 and S100A9 protein close to chondrocytes was associated with proteoglycan depletion and expression of MMP-1, MMP-3, and VDIPEN. Stimulation of chondrocytes with S100A8 and S100A9 caused a strong up-regulation of catabolic markers (MMPs 1, 3, 9, and 13, interleukin-6 [IL-6], IL-8, and monocyte chemotactic protein 1) and down-regulation of anabolic markers (aggrecan and type II collagen), thereby favoring cartilage breakdown. Blocking TLR-4, but not carboxylated glycans or RAGE, inhibited the S100 effect. The catabolic S100 effect was significantly more pronounced in chondrocytes from OA patients as compared to those from non-OA patients, possibly due to higher TLR-4 expression. CONCLUSION: S100A8 and S100A9 have a catabolic effect on human chondrocytes that is TLR-4 dependent. OA chondrocytes are more sensitive than normal chondrocytes to S100 stimulation.


Assuntos
Calgranulina A/metabolismo , Calgranulina B/metabolismo , Cartilagem Articular/metabolismo , Condrócitos/metabolismo , Osteoartrite/metabolismo , Receptor 4 Toll-Like/metabolismo , Biomarcadores/metabolismo , Calgranulina A/administração & dosagem , Calgranulina B/farmacologia , Proteína de Matriz Oligomérica de Cartilagem , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/patologia , Células Cultivadas , Condrócitos/efeitos dos fármacos , Condrócitos/patologia , Citocinas/genética , Citocinas/metabolismo , Epitopos/metabolismo , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Expressão Gênica , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Proteínas Matrilinas , Metaloproteinases da Matriz/genética , Metaloproteinases da Matriz/metabolismo , Oligopeptídeos/metabolismo , Osteoartrite/patologia , Fragmentos de Peptídeos/metabolismo , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/antagonistas & inibidores , Proteínas Recombinantes , Receptor 4 Toll-Like/antagonistas & inibidores
12.
Clin Exp Rheumatol ; 29(5): 768-75, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21961943

RESUMO

OBJECTIVES: In our previous proteomic surveillance, we found that at least 11 proteins in neutrophils were increased more than 2.5-fold by the stimulation of GM-CSF. In this paper, focusing on one of the 11 proteins, S100 calcium binding protein A8 (S100A8), we tried to elucidate the effect of S100A8 and the cooperative effect of S100A8 and GM-CSF on production and secretion of cytokines of neutrophils. METHODS: S100A8 in neutrophil was detected by western blotting, and concentrations of S100A8 in synovial fluid (SF) from patients with rheumatoid arthritis (RA) and osteoarthritis (OA) were measured by ELISA. Cytokine levels in the culture medium of neutrophils incubated with and without S100A8 were measured by an antibody array. IL-8 and IL-16 levels in the culture medium of neutrophils stimulated with S100A8, GM-CSF, and the combination of S100A8 and GM-CSF were measured by ELISA. The mRNA levels of IL-8 and IL-16 in the stimulated neutrophils were analysed by real-time PCR. RESULTS: The western blotting analysis confirmed that S100A8 is up-regulated in neutrophil by the stimulation of GM-CSF. Furthermore, the ELISA analysis confirmed that S100A8 was significantly elevated in SF of patients with RA compared to SF of patients with OA. S100A8 induced mRNA expression and secretion of IL-8 and IL-16. S100A8 further enhanced production of IL-8 by GM-CSF but not that of IL-16. CONCLUSIONS: These data suggest that S100A8 may be involved in the exacerbation of RA, and that S100A8 may be a therapeutic target of RA.


Assuntos
Artrite Reumatoide/fisiopatologia , Calgranulina A/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Interleucina-16/genética , Interleucina-8/genética , Neutrófilos/fisiologia , Idoso , Idoso de 80 Anos ou mais , Artrite Reumatoide/imunologia , Artrite Reumatoide/metabolismo , Calgranulina A/administração & dosagem , Células Cultivadas , Sinergismo Farmacológico , Feminino , Humanos , Interleucina-16/metabolismo , Interleucina-8/metabolismo , Masculino , Pessoa de Meia-Idade , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , RNA Mensageiro/metabolismo , Proteínas Recombinantes/farmacologia , Líquido Sinovial/metabolismo
13.
Rinsho Byori ; 58(7): 651-7, 2010 Jul.
Artigo em Japonês | MEDLINE | ID: mdl-20715507

RESUMO

We have described a possible mechanism for the regulation of excessive inflammatory responses with S100A8/A9 protein in damaged rat livers. Recombinant human S100A8(r-S100A8) and S100A9 (r-S100A9) were expressed in E. coli cells, and their heterodimer (r-S100A8/A9) with 90% approximate purity was also prepared successfully. The effect of the r-S100A8/A9 on suppression of acute inflammatory changes in rat livers with LPS-induced damage was microscopically observed. Indeed, the liver damage diminished as the dose of the r-S100A8/A9 increased, and the minimum requirement of the protein was estimated to be 1,000 microg/rat in this study. Observation of superoxide anions was positively observed in control rats treated with LPS alone, but almost not in the livers of rats treated with the r-S 100A8/A9 1h after injection of LPS. This fact strongly suggests that the r-S100A8/A9 could indirectly suppress production of such internal oxidants according to unknown pathway (s) in acute inflammation. Expression of mRNAs of several kinds of inflammatory cytokines, such as TNF-alpha, IL-6 and IL-1beta, was also significantly suppressed, which was of much note. Therefore, the possibility that the r-S100A8/A9 partly inhibits the process of signal transduction of inflammatory responses in the immunological cells leading to down regulation of inflammatory changes in vivo was suggested in this study. Conclusively, S100A8/A9 is not necessarily an inflammatory-induced factor, and preferably effective on suppression of excessive inflammatory reaction in vivo dose-dependently, although the mechanism is still unclear.


Assuntos
Calgranulina A/administração & dosagem , Calgranulina B/administração & dosagem , Inflamação/prevenção & controle , Doença Aguda , Animais , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Regulação para Baixo , Inflamação/induzido quimicamente , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos , Fígado/metabolismo , Fígado/patologia , Macrófagos , Masculino , Neutrófilos , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Superóxidos/metabolismo
14.
Arthritis Rheum ; 62(11): 3353-64, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20662072

RESUMO

OBJECTIVE: The levels of both Fcγ receptor (FcγR) and the alarmins S100A8 and S100A9 are correlated with the development and progression of cartilage destruction during antigen-induced arthritis (AIA). This study was undertaken to study the active involvement of S100A8, S100A9, and S100A8/S100A9 in FcγR regulation in murine macrophages and synovium during AIA. METHODS: Recombinant murine S100A8 (rS100A8) was injected into normal mouse knee joints, and the synovium was isolated for analysis of FcγR messenger RNA (mRNA) expression by reverse transcription-polymerase chain reaction (RT-PCR). Macrophages, including bone marrow macrophages derived from Toll-like receptor 4-deficient (TLR-4(-/-)) mice, and polymorphonuclear cells (PMNs) were stimulated with S100 proteins, and levels of FcγR mRNA and protein were measured using RT-PCR and fluorescence-activated cell sorting analyses. AIA was induced in the knee joints of S100A9-deficient (S100A9(-/-)) mice, compared with wild-type (WT) controls, and the extent of cartilage destruction was determined using immunohistochemical analysis. RESULTS: Intraarticular injection of rS100A8 into the knee joints of normal mice caused a strong up-regulation of mRNA levels of activating FcγRI (64-fold increase) and FcγRIV (256-fold increase) in the synovium. Stimulation of macrophages with rS100A8 led to significant up-regulation of mRNA and protein levels of FcγRI and FcγRIV, but not FcγRIII, while the effects of S100A9 or S100A8/S100A9 complexes were less potent. Stimulation of PMNs (32Dcl3 cell line) with S100 proteins had no effect on FcγR expression. Up-regulation of FcγRI and FcγRIV was abrogated in rS100A8-stimulated macrophages from TLR-4(-/-) mice, indicating that the induction of FcγR expression by S100A8 is mediated by TLR-4. FcγR expression in the inflamed synovium of S100A9(-/-) mice was significantly lower on day 14 after arthritis induction when compared with WT controls, and these findings correlated with reduced severity of matrix metalloproteinase-mediated cartilage destruction. CONCLUSION: S100A8 is a strong promoter of activating FcγRI and FcγRIV in macrophages through the activation of TLR-4, and acts as a regulator of FcγR expression in inflamed synovium in chronic experimental arthritis.


Assuntos
Artrite Experimental/metabolismo , Calgranulina A/metabolismo , Macrófagos/metabolismo , Receptores de IgG/metabolismo , Membrana Sinovial/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Artrite Experimental/genética , Artrite Experimental/imunologia , Calgranulina A/administração & dosagem , Calgranulina A/imunologia , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/imunologia , Cartilagem Articular/metabolismo , Ensaio de Imunoadsorção Enzimática , Injeções Intra-Articulares , Articulação do Joelho/efeitos dos fármacos , Articulação do Joelho/imunologia , Articulação do Joelho/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de IgG/genética , Receptores de IgG/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estatísticas não Paramétricas , Membrana Sinovial/efeitos dos fármacos , Membrana Sinovial/imunologia , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia , Regulação para Cima
15.
J Immunol ; 170(6): 3233-42, 2003 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-12626582

RESUMO

S100A8 and S100A9 are small calcium-binding proteins that are highly expressed in neutrophil and monocyte cytosol and are found at high levels in the extracellular milieu during inflammatory conditions. Although reports have proposed a proinflammatory role for these proteins, their extracellular activity remains controversial. In this study, we report that S100A8, S100A9, and S100A8/A9 caused neutrophil chemotaxis at concentrations of 10(-12)-10(-9) M. S100A8, S100A9, and S100A8/A9 stimulated shedding of L-selectin, up-regulated and activated Mac-1, and induced neutrophil adhesion to fibrinogen in vitro. Neutralization with Ab showed that this adhesion was mediated by Mac-1. Neutrophil adhesion was also associated with an increase in intracellular calcium levels. However, neutrophil activation by S100A8, S100A9, and S100A8/A9 did not induce actin polymerization. Finally, injection of S100A8, S100A9, or S100A8/A9 into a murine air pouch model led to rapid, transient accumulation of neutrophils confirming their activities in vivo. These studies 1) show that S100A8, S100A9, and S100A8/A9 are potent stimulators of neutrophils and 2) strongly suggest that these proteins are involved in neutrophil migration to inflammatory sites.


Assuntos
Calgranulina A/fisiologia , Calgranulina B/fisiologia , Fatores Quimiotáticos/fisiologia , Quimiotaxia de Leucócito/fisiologia , Mediadores da Inflamação/fisiologia , Neutrófilos/fisiologia , Adulto , Animais , Antígeno CD11b/biossíntese , Antígeno CD11b/metabolismo , Cálcio/metabolismo , Calgranulina A/administração & dosagem , Calgranulina A/biossíntese , Calgranulina B/administração & dosagem , Calgranulina B/biossíntese , Adesão Celular/fisiologia , Fatores Quimiotáticos/administração & dosagem , Fatores Quimiotáticos/biossíntese , Dimerização , Feminino , Fibrinogênio/metabolismo , Vetores Genéticos , Humanos , Mediadores da Inflamação/administração & dosagem , Injeções Subcutâneas , Integrina alfaVbeta3/fisiologia , Selectina L/metabolismo , Antígeno de Macrófago 1/biossíntese , Antígeno de Macrófago 1/metabolismo , Antígeno de Macrófago 1/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Infiltração de Neutrófilos/fisiologia , Neutrófilos/metabolismo , Ligação Proteica/fisiologia , Proteínas Recombinantes/biossíntese , Regulação para Cima/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...