Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int Wound J ; 20(6): 2082-2086, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36756785

RESUMO

This study aimed to identify some risk factors for post-burn scarring in children aged 0-18 years. One hundred and eighty two participants were involved in this cohort study. Under the age of 18 who were admitted to the Department of Burn Reconstructive Surgery with a diagnosis of upper and lower extremity burns were followed for 6 months. A total of 182 participants (62.1% male, and 37.9% female participants) enrolled in this study. Age ranged from 1 to 17 and the average age was 3.95 ± 3.35. The degree of burn and the anatomical location of the burn had a statistically significant effect on the development of hypertrophic scars. The length of the patient's hospitalisation days and the area of ​​the burn were statistically correlated with wound healing (P = 000, P = .074). For example, the average length of hospitalisation days was 8 ± 5 days in the hypertrophic scars group of patients, and in the group with normal scars, average bed days were 6 ± 3 days (P = .000). Grade IIIb burns increased the risk of hypertrophic scar development by 4.9 times and grade IV burns increased it by 2.5 times. In addition, when the area of burns was 11% or more, the risk of hypertrophic scar development was increased by 58.8%. In the case of wound swab infection, the risk of hypertrophic scar development was 12.4% higher (B = 1.124, 95 EI = 0.55; 2.28, P = .748). Participants' age, burn area and degree of burn are statistically significant risk factors for post-burn scarring in children aged 0-18 years.


Assuntos
Cicatriz Hipertrófica , Procedimentos de Cirurgia Plástica , Infecção dos Ferimentos , Humanos , Masculino , Criança , Feminino , Lactente , Pré-Escolar , Recém-Nascido , Adolescente , Cicatriz Hipertrófica/etiologia , Cicatriz Hipertrófica/patologia , Estudos de Coortes , Mongólia , Cicatrização
2.
Int Immunol ; 28(10): 503-512, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27352793

RESUMO

Radioprotective 105 (RP105) is a type I transmembrane protein, which associates with a glycoprotein, MD-1. Monoclonal antibody (mAb)-mediated ligation of RP105/MD-1 robustly activates B cells. RP105/MD-1 is structurally similar to Toll-like receptor 4 (TLR4)/MD-2. B-cell responses to TLR2 and TLR4/MD-2 ligands are impaired in the absence of RP105 or MD-1. In addition to RP105/MD-1, MD-1 alone is secreted. The structure of MD-1 shows that MD-1 has a hydrophobic cavity that directly binds to phospholipids. Little is known, however, about a ligand for MD-1 and the role of MD-1 in vivo To study the role of RP105/MD-1 and MD-1 alone, specific mAbs against MD-1 are needed. Here, we report the establishment and characterization of two anti-MD-1 mAbs (JR2G9, JR7G1). JR2G9 detects soluble MD-1, whereas JR7G1 binds both soluble MD-1 and the cell surface RP105/MD-1 complex. With these mAbs, soluble MD-1 was detected in the serum and urine. The MD-1 concentration was altered by infection, diet and reperfusion injury. Serum MD-1 was rapidly elevated by TLR ligand injection in mice. The quantitative PCR and supernatant-precipitated data indicate that macrophages are one of the sources of serum soluble MD-1. These results suggest that soluble MD-1 is a valuable biomarker for inflammatory diseases.


Assuntos
Antígenos de Superfície/imunologia , Inflamação/imunologia , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Reações Antígeno-Anticorpo , Antígenos CD/imunologia , Antígenos de Superfície/sangue , Morte Celular/efeitos dos fármacos , Morte Celular/imunologia , Dexametasona/farmacologia , Feminino , Masculino , Glicoproteínas de Membrana/sangue , Glicoproteínas de Membrana/deficiência , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
3.
Immunopharmacol Immunotoxicol ; 36(3): 237-41, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24852317

RESUMO

The effect of spironolactone (SPIR) on lipopolysaccharide (LPS)-induced production of proinflammatory mediators was examined using RAW 264.7 macrophage-like cells and mouse peritoneal macrophages. SPIR significantly inhibited LPS-induced production of nitric oxide (NO), tumor necrosis factor-α and prostaglandin E2. The inhibition was not mediated by cell death. SPIR reduced the expression of an inducible NO synthase mRNA in response to LPS. SPIR significantly inhibited phosphorylation of p65 nuclear factor (NF)-κB in response to LPS. Furthermore, SPIR inhibited phosphorylation of IκB kinase (IKK) as an upstream molecule of NF-κB in response to LPS. LPS did not induce the production of aldosterone in RAW 264.7 cells. Taken together, SPIR is suggested to inhibit LPS-induced proinflammatory mediators via inactivation of IKK/NF-κB in LPS signaling.


Assuntos
Mediadores da Inflamação/antagonistas & inibidores , Lipopolissacarídeos/farmacologia , NF-kappa B/antagonistas & inibidores , Espironolactona/farmacologia , Aldosterona/biossíntese , Animais , Células Cultivadas , Dinoprostona/biossíntese , Quinase I-kappa B/metabolismo , Mediadores da Inflamação/metabolismo , Camundongos , Óxido Nítrico/biossíntese , Fosforilação , Fator de Necrose Tumoral alfa/biossíntese
4.
Immunopharmacol Immunotoxicol ; 36(2): 145-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24506665

RESUMO

The effect of lipopolysaccharide (LPS) on insulin sensitivity in adipocytes were examined by using differentiated 3T3-L1 adipocytes. Insulin-mediated activation of insulin receptor substrate (IRS) 1/2 was inhibited in LPS-pretreated adipocytes and IRS1/2-mediated Akt activation was also attenuated in those cells. LPS inhibited activation of glycogen synthase kinase 3 as a negative regulator of glycogenesis and impaired the glycogen synthesis in response to insulin. LPS-induced activation of phosphoinositide 3-kinase (PI3K) in adipocytes. Involvement of suppressor of cytokine signaling 3 (SOCS3) in LPS-induced IRS1/2 inhibition was excluded. Considering that both insulin and LPS were able to activate the PI3K/Akt signaling pathway, LPS was suggested to impair insulin sensitivity of adipocytes through down-regulating insulin-mediated PI3K/Akt activation.


Assuntos
Adipócitos/efeitos dos fármacos , Resistência à Insulina/fisiologia , Insulina/metabolismo , Lipopolissacarídeos/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células 3T3-L1 , Adipócitos/metabolismo , Animais , Linhagem Celular , Quinase 3 da Glicogênio Sintase/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Camundongos , Transdução de Sinais/efeitos dos fármacos , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/metabolismo
5.
Immunology ; 140(3): 352-61, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23826757

RESUMO

The effect of Pam3CSK4, a Toll-like receptor 2 (TLR2) ligand, on interferon-γ (IFN-γ) -induced nitric oxide (NO) production in mouse vascular endothelial END-D cells was studied. Pre-treatment or post-treatment with Pam3CSK4 augmented IFN-γ-induced NO production via enhanced expression of an inducible NO synthase (iNOS) protein and mRNA. Pam3CSK4 augmented phosphorylation of Janus kinase 1 and 2, followed by enhanced phosphorylation of signal transducer and activator of transcription 1 (STAT1) at tyrosine 701. Subsequently, the enhanced STAT1 activation augmented IFN-γ-induced IFN-regulatory factor 1 expression leading to the iNOS expression. Pam3CSK4 also induced the activation of p38 and subsequent phosphorylation of STAT1 at serine 727. A pharmacological p38 inhibitor abolished the augmentation of IFN-γ-induced NO production by Pam3CSK4. Surprisingly, Pam3CSK4 enhanced a physical association of MyD88 and IFN-γ receptor. Together, these findings suggest that Pam3CSK4 up-regulates IFN-γ signalling in vascular endothelial cells via the physical association between MyD88 and IFN-γ receptor α, and p38-dependent serine 727 STAT1 phosphorylation.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Lipopeptídeos/farmacologia , Fator 88 de Diferenciação Mieloide/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico/metabolismo , Receptores de Interferon/metabolismo , Receptor 2 Toll-Like/agonistas , Animais , Linhagem Celular , Endotélio Vascular/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Fator Gênico 3 Estimulado por Interferon/metabolismo , Interferon gama/imunologia , MAP Quinase Quinase 4/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Óxido Nítrico Sintase Tipo II/genética , Fosforilação/efeitos dos fármacos , Ligação Proteica , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Receptor de Interferon gama
6.
Cell Immunol ; 282(2): 100-5, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23770718

RESUMO

The effect of lipopolysaccharide (LPS) on valproic acid (VPA)-induced cell death was examined by using mouse RAW 264.7 macrophage cells. LPS inhibited the activation of caspase 3 and poly (ADP-ribose) polymerase and prevented VPA-induced apoptosis. LPS inhibited VPA-induced p53 activation and pifithrin-α as a p53 inhibitor as well as LPS prevented VPA-induced apoptosis. LPS abolished the increase of Bax/Bcl-2 ratio, which is a critical indicator of p53-mediated mitochondrial damage, in response to VPA. The nuclear factor (NF)-κB inhibitors, Bay 11-7082 and parthenolide, abolished the preventive action of LPS on VPA-induced apoptosis. A series of toll-like receptor ligands, Pam3CSK4, poly I:C, and CpG DNA as well as LPS prevented VPA-induced apoptosis. Taken together, LPS was suggested to prevent VPA-induced apoptosis via activation of anti-apoptotic NF-κB and inhibition of pro-apoptotic p53 activation. The detailed inhibitory mechanism of VPA-induced apoptosis by LPS is discussed.


Assuntos
Apoptose/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , NF-kappa B/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Ácido Valproico/farmacologia , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Immunoblotting , Lipopeptídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , NF-kappa B/antagonistas & inibidores , Nitrilas/farmacologia , Oligodesoxirribonucleotídeos/farmacologia , Poli I-C/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Sesquiterpenos/farmacologia , Sulfonas/farmacologia , Receptor Toll-Like 9/agonistas , Receptores Toll-Like/agonistas , Proteína X Associada a bcl-2/metabolismo
7.
Med ; 4(11): 778-796.e3, 2023 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-37683637

RESUMO

BACKGROUND: Unbalanced fatty acids intake is associated with a range of health outcomes; however, the impact on human health remains unclear globally. We aim to provide a comprehensive assessment of the health effect of unbalanced fatty acids intake on a global scale. METHODS: We analyzed the trends of summary exposure value (SEV) and the attributable burden of unbalanced fatty acids intake, including diet low in polyunsaturated fatty acids (low PUFAs), diet low in seafood omega-3 fatty acids (low seafood-(ω-3)-PUFAs), and diet high in trans fatty acids (high TFAs) from 1990 to 2019 using data from Global Burden of Disease Study 2019. FINDINGS: The global fatty acids intake was far from the optimal level. High-income North America had the highest SEV of diet of high TFAs, while less-developed regions located in Saharan Africa had the highest SEVs of low PUFAs and low seafood-(ω-3)-PUFAs. The attributable burden was unequally distributed to less-developed regions. Males had lower SEVs but higher attributable burden than females and this gender gap was particularly pronounced before the age of 59. The young population had a higher SEV of diet of low PUFAs, comparable SEV of low seafood-(ω-3)-PUFAs but lower SEV of high TFAs than the elderly population. CONCLUSIONS: This study underpinned the high prevalence of unbalanced fatty acids intake worldwide and provided evidence-based guidance for identifying at-risk populations and developing effective strategies to improve fatty acids intake in the future. FUNDING: The study was funded by Shanxi Province "136" Revitalization Medical Project Construction Funds and the Fundamental Research Funds for the Central Universities.


Assuntos
Ácidos Graxos Ômega-3 , Ácidos Graxos , Masculino , Feminino , Humanos , Idoso , Dieta , Ácidos Graxos Insaturados , Fatores de Risco
8.
Cancer Immunol Immunother ; 60(10): 1439-46, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21644032

RESUMO

An ADP ribosylation factor-GTPase activating protein (ASAP1) is highly expressed in a variety of tumor cells and is involved in the cell motility, invasion, and metastasis. In order to elucidate the involvement of ASAP1 in lipopolysaccharide (LPS)-mediated inflammatory response, the effect of ASAP1 silencing on LPS-induced proinflammatory mediators production was examined by using RAW 264.7 macrophage-like cells. ASAP1 was constitutively expressed in the cells and the expression was augmented by LPS stimulation. Silencing of ASAP1 with small interfering RNA enhanced the production of tumor necrosis factor-α, interleukin 6, interferon-ß, and nitric oxide in response to LPS. ASAP1 silencing augmented the activation of nuclear factor (NF)-κB and several mitogen-activated protein kinases (MAPKs). On the other hand, ASAP1 silencing did not affect the expression of IRAK4, TRAF6, and Akt as the upstream molecules of NF-κB signaling. A series of toll-like receptor ligands as well as LPS augmented the ASAP1 expression. Taken together, ASAP1 was suggested to negatively regulate LPS-induced proinflammatory mediators production through down-regulating LPS signaling. The feedback function of ASAP1 in LPS-mediated inflammatory response is discussed.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Lipopolissacarídeos/imunologia , Transdução de Sinais/imunologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Linhagem Celular , Citocinas/biossíntese , Citocinas/imunologia , Immunoblotting , Inflamação/imunologia , Inflamação/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Cell Immunol ; 270(1): 19-24, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21477797

RESUMO

Thalidomide is known as an anti-angiogenic, anti-tumor, and anti-proliferative agent, widely used in the treatment of some immunological disorders and cancers. The effect of thalidomide on interferon (IFN)-γ induced nitric oxide (NO) production in mouse vascular endothelial cells was examined in order to elucidate the anti-angiogenic or anti-inflammatory action. Thalidomide inhibited IFN-γ-induced NO production in mouse END-D cells via reduced expression of an inducible type of NO synthase (iNOS) protein and mRNA. Since thalidomide did not alter the cell surface expression of IFN-γ receptor, the NO inhibition was suggested to be due to the impairment of IFN-γ-induced intracellular event by thalidomide. Thalidomide inhibited the phosphorylation of IRF1, which was required for the iNOS expression. Moreover, it inhibited the phosphorylation of STAT1, an upstream molecule of IRF1, in IFN-γ signaling. Thalidomide did not inhibit the JAK activation in response to IFN-γ. A phosphatase inhibitor, sodium orthovanadate, abolished the inhibitory action of thalidomide. Therefore, thalidomide was suggested to inhibit IFN-γ-induced NO production via impaired STAT1 phosphorylation.


Assuntos
Células Endoteliais/metabolismo , Talidomida/farmacologia , Inibidores da Angiogênese/farmacologia , Animais , Linhagem Celular , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Interferon gama/metabolismo , Interferon gama/farmacologia , Camundongos , Óxido Nítrico/antagonistas & inibidores , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo II/metabolismo , Fosforilação , RNA Mensageiro/biossíntese , Proteínas Recombinantes , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais/efeitos dos fármacos
10.
Microbiol Immunol ; 55(3): 160-7, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21204955

RESUMO

Flavopiridol is a cyclin-dependent kinase inhibitor and inhibits the growth of various cancer cells. The effect of flavopiridol on lipopolysaccharide (LPS)-induced proinflammatory mediator production was examined in RAW 264.7 macrophage-like cells. Flavopiridol significantly reduced the production of tumor necrosis factor-α and, to a lesser extent, nitric oxide in LPS-stimulated cells. Flavopiridol inhibited the activation of nuclear factor-κB and IκB kinase in response to LPS. Flavopiridol also inhibited the activation of a series of mitogen-activated protein kinases, such as p38, stress-activated protein kinase/c-Jun N-terminal kinase and extracellular signal-regulated kinase 1/2 in response to LPS. However, flavopiridol did not alter the expression of tumor necrosis factor receptor-associated factor 6, myeloid differentiation factor 88 (MyD88) or CD14/toll-like receptor (TLR) 4. Flavopiridol inhibited nitric oxide production induced by a MyD88-dependent TLR2 ligand, but not a MyD88-independent TLR3 ligand. Further, flavopiridol did not alter the phosphorylation of interferon regulatory factor 3 in the MyD88-independent pathway. Therefore, it was suggested that flavopiridol exclusively inhibited the activation of nuclear factor-κB and mitogen-activated protein kinases in the MyD88-dependent pathway. Flavopiridol might be useful for the prevention of LPS-induced inflammatory response.


Assuntos
Flavonoides/farmacologia , Regulação da Expressão Gênica em Archaea/efeitos dos fármacos , Lipopolissacarídeos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/metabolismo , Piperidinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Fator de Necrose Tumoral alfa/biossíntese , Animais , Linhagem Celular , Receptores de Lipopolissacarídeos/metabolismo , Macrófagos/efeitos dos fármacos , Camundongos , Óxido Nítrico/biossíntese , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fator 6 Associado a Receptor de TNF/metabolismo , Receptores Toll-Like/metabolismo
11.
Microbes Infect ; 22(8): 322-330, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32032681

RESUMO

Sendai virus V protein is a known antagonist of RIG-I-like receptors (RLRs) RIG-I and MDA5, which activate transcription factors IRF3, leading to activation of ISGF3 and NF-κB. These transcription factors are known activators of inducible NO synthase (iNOS) and increase the production of nitric oxide (NO). By inhibiting ISGF3 and NF-κB, the V protein acts as an indirect negative regulator of iNOS and NO. Here we report that the V gene knockout Sendai virus [SeV V(-)] markedly enhanced iNOS expression and subsequent NO production in infected macrophages compared to wild-type SeV. The knockout of RIG-I in cells inhibited SeV V(-)-induced iNOS expression and subsequent NO production. To understand the underlying mechanism of the V protein-mediated negative regulation of iNOS activation, we transfected HEK293T cells with RIG-I and the RIG-I regulatory protein TRIM25. Our results demonstrated that the V protein inhibited iNOS activation via the RIG-I/TRIM25 pathway. Moreover, the V protein inhibited TRIM25-mediated K63-linked ubiquitination of RIG-I, as well as its CARD-dependent interaction with mitochondrial antiviral signaling (MAVS) molecules. These results suggest that the V protein downregulates iNOS activation and inhibits NO production by preventing the RIG-I-MAVS interaction, possibly through its effect on the ubiquitination status of RIG-I.


Assuntos
Proteína DEAD-box 58/metabolismo , Macrófagos/metabolismo , Óxido Nítrico/metabolismo , Vírus Sendai/metabolismo , Transdução de Sinais , Proteínas Virais/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteína DEAD-box 58/genética , Proteínas de Ligação a DNA/metabolismo , Células HEK293 , Humanos , Macrófagos/virologia , Camundongos , Óxido Nítrico Sintase Tipo II/metabolismo , Ligação Proteica , Células RAW 264.7 , Vírus Sendai/genética , Fatores de Transcrição/metabolismo , Ubiquitinação , Proteínas Virais/genética
12.
Innate Immun ; 24(7): 430-438, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30189760

RESUMO

To suppress virus multiplication, infected macrophages produce NO. However, it remains unclear how infecting viruses then overcome NO challenge. In the present study, we report the effects of accessory protein C from Sendai virus (SeV), a prototypical paramyxovirus, on NO output. We found that in RAW264.7 murine macrophages, a mutant SeV without C protein (4C(-)) significantly enhanced inducible NO synthase (iNOS) expression and subsequent NO production compared to wild type SeV (wtSeV). SeV 4C(-) infection caused marked production of IFN-ß, which is involved in induction of iNOS expression via the JAK-STAT pathway. Addition of anti-IFN-ß Ab, however, resulted in only marginal suppression of NO production. In contrast, NF-κB, a primarily important factor for transcription of the iNOS gene, was also activated by 4C(-) infection but not wtSeV infection. Induction of NO production and iNOS expression by 4C(-) was significantly suppressed in cells constitutively expressing influenza virus NS1 protein that can sequester double-stranded (ds)RNA, which triggers activation of signaling pathways leading to activation of NF-κB and IRF3. Therefore, C protein appears to suppress NF-κB activation to inhibit iNOS expression and subsequent NO production, possibly by limiting dsRNA generation in the context of viral infection.


Assuntos
Macrófagos/fisiologia , Infecções por Respirovirus/imunologia , Vírus Sendai/fisiologia , Proteínas Virais/metabolismo , Animais , Regulação da Expressão Gênica , Fator Regulador 3 de Interferon/metabolismo , Janus Quinases/metabolismo , Camundongos , Mutação/genética , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Células RAW 264.7 , RNA de Cadeia Dupla/metabolismo , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Proteínas não Estruturais Virais/metabolismo , Proteínas Virais/genética
13.
Innate Immun ; 21(2): 194-202, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24595208

RESUMO

Here we report that LPS induces osteoclast (OC) formation in murine RAW 264.7 macrophage cells in RPMI-1640 medium but not in α-minimum essential medium (α-MEM) as the original culture medium. LPS-induced OC formation in both media was examined to clarify the differential response. Receptor activator of NF-κB ligand induced OC formation in either α-MEM or RPMI-1640 medium. However, LPS-induced OC formation in RAW 264.7 cells maintained in RPMI-1640 medium, but not α-MEM, which was also supported by mouse bone marrow-derived macrophages, although they were less sensitive to LPS than RAW 264.7 cells. LPS augmented the expression of nuclear factor of activated T-cells (NFATc1) as a key transcription factor of osteoclastogenesis in cells maintained in RPMI-1640 medium, but reduced it in cells maintained in α-MEM. A high concentration of LPS was cytotoxic against cells maintained in α-MEM. Glutathione exclusively present in RPMI-1640 medium prevented LPS-induced cell death in α-MEM and augmented LPS-induced NFATc1 expression, followed by enhanced LPS-induced OC formation. LPS induced higher generation of reactive oxygen species in α-MEM than RPMI-1640 medium. An antioxidant enhanced LPS-induced OC formation, whereas a pro-oxidant reduced it. Taken together, redox balance in the culture condition was suggested to regulate in vitro LPS-induced OC formation.


Assuntos
Meios de Cultura/metabolismo , Glutationa/metabolismo , Lipopolissacarídeos/metabolismo , Macrófagos/fisiologia , Osteoclastos/fisiologia , Animais , Morte Celular , Diferenciação Celular , Linhagem Celular , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Fatores de Transcrição NFATC/metabolismo , Oxirredução , Espécies Reativas de Oxigênio/metabolismo
14.
Immunobiology ; 220(1): 136-41, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25172547

RESUMO

The effect of lipopolysaccharide (LPS) on the expression of p53 protein in RAW 264.7 macrophage cells was examined. LPS downregulated the expression of p53 protein 4-24 h after the stimulation. LPS-induced p53 inhibition was restored with pharmacological inhibitors of c-jun N-terminal kinase (JNK) and phosphatidylinositol 3-kinase (PI3K). It was also restored by inhibitors of MDM2 activation and proteasome. LPS-induced p53 inhibition corresponded to activation of MDM2. LPS-induced MDM2 activation was prevented by inhibitors of JNK and PI3K. The expression of p65 NF-κB at a late stage after LPS stimulation was downregulated in the presence of a MDM2 inhibitor. Nutlin-3 as a MDM2 inhibitor reduced LPS-induced production of nitric oxide but not tumor necrosis factor-α. Administration of LPS into mice downregulated the in vivo expression of p53 in the livers. Taken together, LPS was suggested to downregulate the expression of p53 via activation of MDM2 and enhance the activation of NF-κB at a late stage.


Assuntos
Lipopolissacarídeos/imunologia , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Linhagem Celular , Imidazóis/farmacologia , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Fígado/efeitos dos fármacos , Fígado/imunologia , Fígado/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Óxido Nítrico/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Piperazinas/farmacologia , Ligação Proteica , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética
15.
Int Immunopharmacol ; 26(1): 97-102, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25817178

RESUMO

High-mobility group box 1 (HMGB1) is suggested to participate in development of local and systemic inflammatory disorders. Irbesartan (IRB), an angiotensin II type1 receptor blocker, is widely used for treatment of hypertension, especially in patients with diabetic nephropathy. The effect of IRB on lipopolysaccharide (LPS)-induced HMGB1 and nitric oxide (NO) production was examined using RAW 264.7 macrophage-like cells. IRB inhibited LPS-induced HMGB1 production. IRB also reduced LPS-induced expression of an inducible NO synthase, and inhibited LPS-induced NO production. The expression levels of IFN-ß protein and mRNA, which is a key molecule in MyD88-independent pathway of LPS signaling, were exclusively inhibited by IRB. Peroxisome proliferator-activated receptor-γ and angiotensin II type 1 receptor were not involved in the inhibitory action of IRB on LPS-induced HMGB1 and NO production. Collectively, IRB was suggested to inhibit LPS-induced HMGB1 production via downregulation of IFN-ß production in the MyD88-independent pathway.


Assuntos
Anti-Inflamatórios/farmacologia , Compostos de Bifenilo/farmacologia , Proteína HMGB1/biossíntese , Interferon beta/biossíntese , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Tetrazóis/farmacologia , Animais , Técnicas de Cultura de Células , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Regulação para Baixo , Proteína HMGB1/antagonistas & inibidores , Interferon beta/antagonistas & inibidores , Irbesartana , Macrófagos/imunologia , Camundongos , Óxido Nítrico/antagonistas & inibidores , Óxido Nítrico/biossíntese , Reação em Cadeia da Polimerase em Tempo Real
16.
Int Immunopharmacol ; 23(1): 267-72, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25242386

RESUMO

The effect of Sendai virus (SeV) C protein on lipopolysaccharide (LPS)-induced nitric oxide (NO) production was examined using RAW 264.7 macrophage cells. Infection of SeV inhibited LPS-induced NO production via downregulating the expression of an inducible NO synthase protein (iNOS). On the other hand, C gene-knockout 4C(-) SeV inhibited neither NO production nor iNOS expression. Wild type and 4C(-) SeV did not affect LPS-induced production of tumor necrosis factor-α and interleukin-6, and further LPS-induced activation of nuclear factor (NF)-κB and mitogen-activated protein kinases. Although wild type and 4C(-) SeV did not inhibit LPS-induced interferon (IFN)-ß production, wild type SeV but not 4C(-) SeV inhibited the activation of STAT1/2 in the IFN-ß signaling. SeV C protein inhibited LPS-induced iNOS expression and NO production. C protein inhibited the promotor activation of IFN-ß and IFN-sensitive response element (ISRE) in response to LPS whereas the C mutant protein CF170S, which lacks the ability to block the STAT activation, did not inhibit it. Taken together, SeV C protein was suggested to inhibit LPS-induced NO production through impairing IFN-ß signaling.


Assuntos
Interferon beta/metabolismo , Macrófagos/imunologia , Infecções por Respirovirus/imunologia , Vírus Sendai/fisiologia , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Regulação Viral da Expressão Gênica/genética , Técnicas de Inativação de Genes , Lipopolissacarídeos/metabolismo , Macrófagos/virologia , Camundongos , Mutação/genética , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT2/metabolismo , Transdução de Sinais/genética , Ativação Transcricional/genética , Proteínas Virais/genética
17.
Innate Immun ; 20(1): 40-8, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23606517

RESUMO

The pyrin and HIN-domain (PYHIN) family member1 (pyhin1) is a member of PYHIN proteins and involved in transcriptional regulation of genes important for cell cycle control, differentiation and apoptosis. The regulatory action of mouse pyhin1 on LPS-induced inflammatory response was examined. LPS augmented the pyhin1 mRNA expression in murine RAW 264.7 macrophage cells and peritoneal macrophages. The augmentation of pyhin1 mRNA expression was abolished by parthenolide, a NF-κB inhibitor. Silencing of pyhin1 with small interfering RNA reduced the production of IFN-ß and NO. However, pyhin1 silencing did not affect the production of TNF-α, IL-6, IL-10 and prostaglandin E2. Reduced IFN-ß production by pyhin1 silencing caused inactivation of STAT1 and reduced expression of IRF1. Pyhin1 silencing inhibited the expression of TRAF6, TBK1 and TRIF, which trigger IFN-ß production in the MyD88-independent pathway. However, pyhin1 silencing did not affect the expression of MyD88, IRAK4 and several mitogen-activated protein kinases in the MyD88-dependent pathway. Taken together, mouse pyhin1 was suggested to be a NF-κB-responsible gene in response to LPS and positively regulate LPS-induced IFN-ß and NO production through up-regulating the MyD88-independent signaling pathway.


Assuntos
Inflamação , Interferon beta/metabolismo , Macrófagos Peritoneais/imunologia , Óxido Nítrico/metabolismo , Proteínas Nucleares/metabolismo , Animais , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/imunologia , Fator Regulador 1 de Interferon/genética , Fator Regulador 1 de Interferon/metabolismo , Lipopolissacarídeos/imunologia , Camundongos , NF-kappa B/antagonistas & inibidores , Proteínas Nucleares/genética , RNA Interferente Pequeno/genética , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Sesquiterpenos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fator 6 Associado a Receptor de TNF/genética , Fator 6 Associado a Receptor de TNF/metabolismo
18.
Innate Immun ; 20(8): 816-25, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24225655

RESUMO

The effect of LPS on the production of vascular endothelial growth factor (VEGF) was examined using RAW 264.7 macrophage cells. LPS induced VEGF production in RAW 264.7 cells and mouse peritoneal cells. LPS induced VEGF production via the expression of hypoxia inducible factor-1α and LPS-induced VEGF production was dependent on the activation of p38 MAPK and NF-κB activation· Transforming growth factor (TGF)-ß1 augmented LPS-induced VEGF production, although TGF-ß1 alone did not induce VEGF production. The augmentation of LPS-induced VEGF production by TGF-ß1 was inhibited by a p38 MAPK inhibitor and was correlated with the phosphorylation of Smad3. The enhancing effect of TGF-ß1 on LPS-induced VEGF production was observed in vivo in the skin lesions of mice receiving a subcutaneous injection of LPS. Taken together, it is suggested that LPS induced the VEGF production in macrophages and that it was augmented by TGF-ß1 in vitro and in vivo.


Assuntos
Lipopolissacarídeos/farmacologia , Macrófagos/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Linhagem Celular , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Macrófagos/efeitos dos fármacos , Camundongos , NF-kappa B/biossíntese , Fosforilação , Proteína Smad3/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/genética
19.
Immunol Lett ; 161(1): 31-7, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24792671

RESUMO

The effect of conophylline (CNP) on the receptor activator of nuclear factor-κB ligand (RANKL) or lipopolysaccharide (LPS)-induced osteoclast formation was studied in vitro using bone marrow-derived macrophages (BMMs) or the mouse macrophage-like cell line RAW 264.7. CNP inhibited RANKL-induced formation of osteoclasts identified as tartrate-resistant acid phosphatase (TRAP)-positive multinucleated cells in a culture of BMMs. It also inhibited RANKL- or LPS-induced osteoclast formation in RAW 264.7 cells. CNP lowered the osteoclast maturation markers such as calcitonin receptor, MMP9 and cathepsin K in BMMs, suggesting that CNP would inhibit the process of osteoclast differentiation. CNP inhibited the RANKL-induced expressions of c-Fos and nuclear factor of activated T cells (NFATc1), key transcription factors for osteoclastogenesis. On the other hand, CNP did not inhibit the signaling pathway of NF-κB and mitogen-activated protein kinases (MAPKs) in RANKL-stimulated BMMs. Interestingly, CNP inhibited RANKL-induced CREB activation that can mediate c-Fos and NFATc1. CNP also inhibited RANKL- or LPS-induced CREB, c-Fos and NFATc1 activation in RAW 264.7 cells. We have previously found that CNP directly binds to ADP-ribosylation-like factor-6 interacting protein (ARL6ip), although its role in osteoclastogenesis is not clear. Gene knockdown of ARL6ip by siRNA inhibited RANKL-induced c-Fos expression, suggesting that inactivation of ARL6ip may be involved in an inhibitory effect of CNP. Taken together, CNP was shown to inhibit osteoclast formation possibly via CREB inactivation following a decrease in c-Fos and NFATc1 expression.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Ligante RANK/farmacologia , Alcaloides de Vinca/farmacologia , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Linhagem Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Regulação para Baixo , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Fatores de Transcrição NFATC/genética , Proteínas Proto-Oncogênicas c-fos/genética , Transdução de Sinais/efeitos dos fármacos
20.
Int Immunopharmacol ; 20(1): 181-7, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24631367

RESUMO

The inhibitory effect of valproic acid (VPA) on lipopolysaccharide (LPS)-induced inflammatory response was studied by using mouse RAW 264.7 macrophage-like cells. VPA pretreatment attenuated LPS-induced phosphorylation of phosphatidylinositol 3-kinase (PI3K) and Akt, but not nuclear factor (NF)-κB and mitogen-activated protein kinases. VPA reduced phosphorylation of MDM2, an ubiquitin ligase and then prevented LPS-induced p53 degradation, followed by enhanced p53 expression. Moreover, p53 small interfering RNA (siRNA) abolished the inhibitory action of VPA on LPS-induced NF-κB p65 transcriptional activation and further LPS-induced tumor necrosis factor (TNF)-α and interleukin (IL)-6 production. VPA prevented LPS-induced degradation of phosphatase and tensin homologue deleted on chromosome ten (PTEN) and up-regulated the PTEN expression. Taken together, VPA was suggested to down-regulate LPS-induced NF-κB-dependent transcriptional activity via impaired PI3K/Akt/MDM2 activation and enhanced p53 expression. A detailed mechanism for inhibition of LPS-induced inflammatory response by VPA is discussed.


Assuntos
Anti-Inflamatórios/farmacologia , Interleucina-6/antagonistas & inibidores , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Ácido Valproico/farmacologia , Animais , Linhagem Celular , Interleucina-6/metabolismo , Lipopolissacarídeos , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA