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










Base de datos
Intervalo de año de publicación
1.
Med ; 4(11): 778-796.e3, 2023 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-37683637

RESUMEN

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.


Asunto(s)
Ácidos Grasos Omega-3 , Ácidos Grasos , Masculino , Femenino , Humanos , Anciano , Dieta , Ácidos Grasos Insaturados , Factores de Riesgo
2.
Int Wound J ; 20(6): 2082-2086, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36756785

RESUMEN

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.


Asunto(s)
Cicatriz Hipertrófica , Procedimientos de Cirugía Plástica , Infección de Heridas , Humanos , Masculino , Niño , Femenino , Lactante , Preescolar , Recién Nacido , Adolescente , Cicatriz Hipertrófica/etiología , Cicatriz Hipertrófica/patología , Estudios de Cohortes , Mongolia , Cicatrización de Heridas
3.
Microbes Infect ; 22(8): 322-330, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32032681

RESUMEN

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.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , Macrófagos/metabolismo , Óxido Nítrico/metabolismo , Virus Sendai/metabolismo , Transducción de Señal , Proteínas Virales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteína 58 DEAD Box/genética , Proteínas de Unión al ADN/metabolismo , Células HEK293 , Humanos , Macrófagos/virología , Ratones , Óxido Nítrico Sintasa de Tipo II/metabolismo , Unión Proteica , Células RAW 264.7 , Virus Sendai/genética , Factores de Transcripción/metabolismo , Ubiquitinación , Proteínas Virales/genética
4.
Innate Immun ; 24(7): 430-438, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30189760

RESUMEN

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.


Asunto(s)
Macrófagos/fisiología , Infecciones por Respirovirus/inmunología , Virus Sendai/fisiología , Proteínas Virales/metabolismo , Animales , Regulación de la Expresión Génica , Factor 3 Regulador del Interferón/metabolismo , Quinasas Janus/metabolismo , Ratones , Mutación/genética , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Células RAW 264.7 , ARN Bicatenario/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Proteínas no Estructurales Virales/metabolismo , Proteínas Virales/genética
5.
Int Immunol ; 28(10): 503-512, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27352793

RESUMEN

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.


Asunto(s)
Antígenos de Superficie/inmunología , Inflamación/inmunología , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Reacciones Antígeno-Anticuerpo , Antígenos CD/inmunología , Antígenos de Superficie/sangre , Muerte Celular/efectos de los fármacos , Muerte Celular/inmunología , Dexametasona/farmacología , Femenino , Masculino , Glicoproteínas de Membrana/sangre , Glicoproteínas de Membrana/deficiencia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
6.
Int Immunopharmacol ; 26(1): 97-102, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25817178

RESUMEN

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.


Asunto(s)
Antiinflamatorios/farmacología , Compuestos de Bifenilo/farmacología , Proteína HMGB1/biosíntesis , Interferón beta/biosíntesis , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Tetrazoles/farmacología , Animales , Técnicas de Cultivo de Célula , Línea Celular , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo , Proteína HMGB1/antagonistas & inhibidores , Interferón beta/antagonistas & inhibidores , Irbesartán , Macrófagos/inmunología , Ratones , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/biosíntesis , Reacción en Cadena en Tiempo Real de la Polimerasa
7.
Innate Immun ; 21(2): 194-202, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24595208

RESUMEN

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.


Asunto(s)
Medios de Cultivo/metabolismo , Glutatión/metabolismo , Lipopolisacáridos/metabolismo , Macrófagos/fisiología , Osteoclastos/fisiología , Animales , Muerte Celular , Diferenciación Celular , Línea Celular , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Factores de Transcripción NFATC/metabolismo , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo
8.
Immunobiology ; 220(1): 136-41, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25172547

RESUMEN

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.


Asunto(s)
Lipopolisacáridos/inmunología , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular , Imidazoles/farmacología , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/farmacología , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Óxido Nítrico/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Piperazinas/farmacología , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/genética
9.
Int Immunopharmacol ; 23(1): 267-72, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25242386

RESUMEN

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.


Asunto(s)
Interferón beta/metabolismo , Macrófagos/inmunología , Infecciones por Respirovirus/inmunología , Virus Sendai/fisiología , Proteínas Virales/metabolismo , Animales , Línea Celular , Regulación Viral de la Expresión Génica/genética , Técnicas de Inactivación de Genes , Lipopolisacáridos/metabolismo , Macrófagos/virología , Ratones , Mutación/genética , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/metabolismo , Transducción de Señal/genética , Activación Transcripcional/genética , Proteínas Virales/genética
10.
Immunopharmacol Immunotoxicol ; 36(3): 237-41, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24852317

RESUMEN

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.


Asunto(s)
Mediadores de Inflamación/antagonistas & inhibidores , Lipopolisacáridos/farmacología , FN-kappa B/antagonistas & inhibidores , Espironolactona/farmacología , Aldosterona/biosíntesis , Animales , Células Cultivadas , Dinoprostona/biosíntesis , Quinasa I-kappa B/metabolismo , Mediadores de Inflamación/metabolismo , Ratones , Óxido Nítrico/biosíntesis , Fosforilación , Factor de Necrosis Tumoral alfa/biosíntesis
11.
Immunol Lett ; 161(1): 31-7, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24792671

RESUMEN

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.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Regulación de la Expresión Génica/efectos de los fármacos , Lipopolisacáridos/farmacología , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Ligando RANK/farmacología , Alcaloides de la Vinca/farmacología , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Línea Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Regulación hacia Abajo , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Factores de Transcripción NFATC/genética , Proteínas Proto-Oncogénicas c-fos/genética , Transducción de Señal/efectos de los fármacos
12.
Int Immunopharmacol ; 20(1): 181-7, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24631367

RESUMEN

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.


Asunto(s)
Antiinflamatorios/farmacología , Interleucina-6/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Ácido Valproico/farmacología , Animales , Línea Celular , Interleucina-6/metabolismo , Lipopolisacáridos , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
13.
Immunopharmacol Immunotoxicol ; 36(2): 145-9, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24506665

RESUMEN

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.


Asunto(s)
Adipocitos/efectos de los fármacos , Resistencia a la Insulina/fisiología , Insulina/metabolismo , Lipopolisacáridos/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células 3T3-L1 , Adipocitos/metabolismo , Animales , Línea Celular , Glucógeno Sintasa Quinasa 3/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Ratones , Transducción de Señal/efectos de los fármacos , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
14.
Innate Immun ; 20(1): 40-8, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23606517

RESUMEN

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.


Asunto(s)
Inflamación , Interferón beta/metabolismo , Macrófagos Peritoneales/inmunología , Óxido Nítrico/metabolismo , Proteínas Nucleares/metabolismo , Animales , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/inmunología , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/metabolismo , Lipopolisacáridos/inmunología , Ratones , FN-kappa B/antagonistas & inhibidores , Proteínas Nucleares/genética , ARN Interferente Pequeño/genética , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Sesquiterpenos/farmacología , Transducción de Señal/efectos de los fármacos , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo
15.
Innate Immun ; 20(6): 639-46, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24055878

RESUMEN

The effect of LPS on the production of prostaglandin E2 (PGE2) in mouse HEI-OC1 auditory cells was examined. HEI-OC1 auditory cells constitutively produce a small amount of PGE2. LPS augmented the PGE2 production via enhanced cyclooxygenase 2 (COX2) expression. LPS-induced augmentation of COX2 expression was dependent on up-regulation of COX2 mRNA expression. LPS induced the production of TNF-α, but not IL-1ß· An anti-TNF-α neutralizing Ab significantly inhibited PGE2 production and COX2 mRNA expression in response to LPS. LPS-induced PGE2 production was prevented by a series of pharmacological signaling inhibitors to NF-κB and MAPKs. Pam3CSK4 as a TLR2 ligand, as well as LPS as a TLR4 ligand, augmented the PGE2 production. However, poly I:C as a TLR3 ligand, imiquimod as a TLR7 ligand and CpG DNA as a TLR9 ligand did not augment it. HEI-OC1 cells expressed TLR2, TLR4 and TLR9, but not TLR3 or TLR7. The putative role of LPS-induced PGE2 production in auditory cells is discussed.


Asunto(s)
Cóclea/inmunología , Dinoprostona/inmunología , Lipopolisacáridos/farmacología , Animales , Línea Celular , Cóclea/citología , Ciclooxigenasa 2/biosíntesis , Interleucina-1beta/biosíntesis , Ratones , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , FN-kappa B/antagonistas & inhibidores , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptor Toll-Like 2/biosíntesis , Receptor Toll-Like 4/biosíntesis , Receptor Toll-Like 9/biosíntesis , Factor de Necrosis Tumoral alfa/biosíntesis , Regulación hacia Arriba/efectos de los fármacos
16.
Innate Immun ; 20(8): 816-25, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24225655

RESUMEN

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.


Asunto(s)
Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Animales , Línea Celular , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Macrófagos/efectos de los fármacos , Ratones , FN-kappa B/biosíntesis , Fosforilación , Proteína smad3/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/genética
17.
Oncol Res ; 21(1): 59-65, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24330853

RESUMEN

A toll-like receptor 4 (TLR-4) ligand, lipopolysaccharide (LPS) not only activates expression and secretion of inflammatory cytokines, but it also often shows toxicity in monocytes. Whether an oncogenic protein, ß-catenin, is positively involved in LPS-induced cytotoxicity in a mouse leukemic monocyte cell line, RAW 264.7, was examined. TWS119, a GSK-3ß inhibitor, increased LPS-induced ß-catenin accumulation in the nucleus and augmented LPS-induced cytotoxicity. Cardamonin, a ß-catenin inhibitor, inhibited LPS-induced ß-catenin accumulation in the nucleus and reduced LPS-induced cytotoxicity. To confirm that ß-catenin is involved in LPS-induced cytotoxicity, silencing of ß-catenin expression by siRNA was carried out. The results were that knockdown of ß-catenin reduced LPS-induced cytotoxicity. Interestingly, Cardamonin treatment or ß-catenin silencing reduced LPS-induced endoplasmic reticulum (ER) stress responses such as PERK and e1F-2α phosphorylation and CHOP expression. Moreover, TWS119 increased LPS-induced ER stress responses. On the basis of these results, the oncogenic protein ß-catenin is considered to be positively involved in LPS-induced cytotoxicity, possibly by downregulating ER stress responses.


Asunto(s)
Lipopolisacáridos/toxicidad , Monocitos/efectos de los fármacos , Monocitos/metabolismo , beta Catenina/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Chalconas/farmacología , Interacciones Farmacológicas , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/fisiología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Leucemia , Ratones , Fosforilación , Pirimidinas/farmacología , Pirroles/farmacología , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Transfección , beta Catenina/biosíntesis , beta Catenina/genética
18.
Immunology ; 140(3): 352-61, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23826757

RESUMEN

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.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Lipopéptidos/farmacología , Factor 88 de Diferenciación Mieloide/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico/metabolismo , Receptores de Interferón/metabolismo , Receptor Toll-Like 2/agonistas , Animales , Línea Celular , Endotelio Vascular/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Interferón gamma/inmunología , MAP Quinasa Quinasa 4/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Óxido Nítrico Sintasa de Tipo II/genética , Fosforilación/efectos de los fármacos , Unión Proteica , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Receptor de Interferón gamma
19.
Cell Immunol ; 282(2): 100-5, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23770718

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Lipopolisacáridos/farmacología , FN-kappa B/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ácido Valproico/farmacología , Animales , Línea Celular , Relación Dosis-Respuesta a Droga , Immunoblotting , Lipopéptidos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , FN-kappa B/antagonistas & inhibidores , Nitrilos/farmacología , Oligodesoxirribonucleótidos/farmacología , Poli I-C/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Sesquiterpenos/farmacología , Sulfonas/farmacología , Receptor Toll-Like 9/agonistas , Receptores Toll-Like/agonistas , Proteína X Asociada a bcl-2/metabolismo
20.
Int Immunopharmacol ; 15(4): 671-8, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23454130

RESUMEN

The effect of pifithrin (PFT)-α, a pharmacological inhibitor of p53, on lipopolysaccharide (LPS)-induced nitric oxide (NO) production in RAW 264.7 macrophage-like cells was examined. PFT-α inhibited the production of NO but not tumor necrosis factor (TNF)-α in response to LPS. PFT-α inhibited LPS-induced NO production via reduced expression of an inducible NO synthase (iNOS). Moreover, PFT-α inhibited LPS-induced iNOS expression in p53-silenced cells. PFT-α inhibited the production of interferon (IFN)-ß, characteristic of the MyD88-independent pathway of LPS signaling, whereas it did not affect the activation of nuclear factor (NF)-κB and mitogen-activated protein kinases in the MyD88-dependent pathway. PFT-α inhibited poly I:C-induced NO production whereas it did not inhibit IFN-ß-induced NO production. Further, PFT-α reduced the expression of IFN regulatory factor 3 that leads to the IFN-ß production in the MyD88-independent pathway. The most upstream event impaired by PFT-α was the reduced expression of TNF receptor-associated factor (TRAF) 3 in the MyD88-independent pathway. PFT-α also reduced the in vivo expression of iNOS in the livers of mice injected with LPS. Taken together, PFT-α was suggested to inhibit LPS-induced NO production via impairment of the MyD88-independent pathway and attenuated LPS-mediated inflammatory response.


Asunto(s)
Benzotiazoles/farmacología , Lipopolisacáridos/toxicidad , Macrófagos Peritoneales/efectos de los fármacos , Factor 88 de Diferenciación Mieloide/metabolismo , Óxido Nítrico/antagonistas & inhibidores , Tolueno/análogos & derivados , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Animales , Benzotiazoles/administración & dosificación , Benzotiazoles/uso terapéutico , Técnicas de Cultivo de Célula , Línea Celular , Supervivencia Celular/efectos de los fármacos , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Silenciador del Gen , Interferón beta/inmunología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tolueno/administración & dosificación , Tolueno/farmacología , Tolueno/uso terapéutico , Factor de Necrosis Tumoral alfa/inmunología , Proteína p53 Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA