Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
Más filtros












Intervalo de año de publicación
1.
Toxicology ; 440: 152487, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32418911

RESUMEN

Renal toxicity is the primary factor that limits clinical use of cisplatin (CP). A previous study showed that omeprazole (OME) protected against CP-induced toxicity in human renal tubular HK-2 cells and rat kidneys. However, the protective mechanisms of OME have not been characterized. We evaluated the ability of OME to inhibit CP-induced inflammation, and characterized the pathways responsible for this effect. Rats were randomly divided into five groups (n = 10/group). The OME groups were intraperitoneally injected with 1.8 or 3.6 mg OME /kg body weight once daily for 5 days. One hour after final administration of vehicle or OME, all rats (except those in control group and OME alone group) were intraperitoneally injected with 15 mg/kg CP. Twenty-four hours after CP injection, the surgery was applied. The time points and dosing of OME and CP were calculated based on previous studies and the therapeutic dose for patients. Omeprazole attenuated CP-induced apoptosis and damage in vivo and in vitro, as evidenced by increased cell viability and prevention of structural damage. Omeprazole ameliorated CP-induced renal injury through inhibition of NF-κB activation and IκBα degradation, and down-regulation of toll-like receptor 4 (TLR4) and Nod-like receptor protein 3 (NLRP3). Lipopolysaccharide, a TLR4 agonist, was used to verify this mechanism. The results indicated that OME inhibited CP-induced expression of inflammatory proteins, and this effect was blunted by co-treatment with LPS in HK-2 cells. These findings suggested that the protective effects of OME against CP-induced kidney damage may occur through inhibition of the TLR4/NF-κB/NLRP3 signaling pathway. This study provided evidence that OME may be a promising agent to inhibit CP-induced nephrotoxicity.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/prevención & control , Antineoplásicos , Cisplatino , FN-kappa B/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/efectos de los fármacos , Omeprazol/farmacología , Inhibidores de la Bomba de Protones/farmacología , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/efectos de los fármacos , Animales , Antiinflamatorios/farmacología , Apoptosis/efectos de los fármacos , Línea Celular , Humanos , Proteínas I-kappa B/efectos de los fármacos , Pruebas de Función Renal , Masculino , Ratas , Ratas Sprague-Dawley , Cicatrización de Heridas/efectos de los fármacos
2.
Glia ; 68(3): 561-573, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31647138

RESUMEN

We used mouse microglial cells in culture activated by lipopolysaccharide (LPS, 10 ng/ml) to study the anti-inflammatory potential of cannabidiol (CBD), the major nonpsychoactive component of cannabis. Under LPS stimulation, CBD (1-10 µM) potently inhibited the release of prototypical proinflammatory cytokines (TNF-α and IL-1ß) and that of glutamate, a noncytokine mediator of inflammation. The effects of CBD were predominantly receptor-independent and only marginally blunted by blockade of CB2 receptors. We established that CBD inhibited a mechanism involving, sequentially, NADPH oxidase-mediated ROS production and NF-κB-dependent signaling events. In line with these observations, active concentrations of CBD demonstrated an intrinsic free-radical scavenging capacity in the cell-free DPPH assay. Of interest, CBD also prevented the rise in glucose uptake observed in microglial cells challenged with LPS, as did the inhibitor of NADPH oxidase apocynin and the inhibitor of IκB kinase-2, TPCA-1. This indicated that the capacity of CBD to prevent glucose uptake also contributed to its anti-inflammatory activity. Supporting this view, the glycolytic inhibitor 2-deoxy-d-glucose (2-DG) mimicked the antioxidant/immunosuppressive effects of CBD. Interestingly, CBD and 2-DG, as well as apocynin and TPCA-1 caused a reduction in glucose-derived NADPH, a cofactor required for NADPH oxidase activation and ROS generation. These different observations suggest that CBD exerts its anti-inflammatory effects towards microglia through an intrinsic antioxidant effect, which is amplified through inhibition of glucose-dependent NADPH synthesis. These results also further confirm that CBD may have therapeutic utility in conditions where neuroinflammatory processes are prominent.


Asunto(s)
Cannabidiol/farmacología , Glucosa/metabolismo , Inflamación/prevención & control , Microglía/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Animales , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Citocinas/farmacología , Proteínas I-kappa B/efectos de los fármacos , Inflamación/tratamiento farmacológico , Lipopolisacáridos/farmacología , Ratones , Microglía/metabolismo , Transducción de Señal/efectos de los fármacos
3.
J Appl Oral Sci ; 27: e20180150, 2019 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-30624466

RESUMEN

OBJECTIVES: This investigation aimed to assess the differentiation inhibitory effects of ProRoot MTA® (PMTA) and Biodentine® (BIOD) on osteoclasts originated from murine bone marrow macrophages (BMMs) and compare these effects with those of alendronate (ALD). MATERIALS AND METHODS: Mouse BMMs were cultured to differentiate into osteoclasts with macrophage colony-stimulating factor and receptor activator of NF-κB (RANKL), treated with lipopolysaccharide. After application with PMTA, BIOD, or ALD, cell toxicities were examined using WST-1 assay kit, and RANKL-induced osteoclast differentiation and activities were determined by resorption pit formation assay and tartrate-resistant acid phosphate (TRAP) staining. The mRNA levels of osteoclast activity-related genes were detected with quantitative real time polymerase chain reaction. Expressions of molecular signaling pathways were assessed by western blot. All data were statistically analyzed with one-way ANOVA and Tukey's post-hoc test (p<0.05). RESULTS: Mouse BMMs applied with PMTA, BIOD, or ALD showed highly reduced levels of TRAP-positive osteoclasts. The BIOD treated specimens suppressed mRNA expressions of cathepsin K, TRAP, and c-Fos. Nonetheless, it showed a lower effect than PMTA or ALD applications. Compared with ALD, PMTA and BIOD decreased RANKL-mediated phosphorylation of ERK1/2 and IκBα. CONCLUSIONS: PMTA and BIOD showed the inhibitory effect on osteoclast differentiation and activities similar to that of ALD through IκB phosphorylation and suppression of ERK signaling pathways.


Asunto(s)
Alendronato/farmacología , Conservadores de la Densidad Ósea/farmacología , Células de la Médula Ósea/efectos de los fármacos , Compuestos de Calcio/farmacología , Diferenciación Celular/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Materiales de Obturación del Conducto Radicular/farmacología , Silicatos/farmacología , Animales , Western Blotting , Células de la Médula Ósea/citología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Proteínas I-kappa B/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Osteoclastos/fisiología , Osteogénesis/efectos de los fármacos , Fosforilación/efectos de los fármacos , Ligando RANK/análisis , Ligando RANK/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Reproducibilidad de los Resultados , Resorción Radicular/prevención & control , Fosfatasa Ácida Tartratorresistente , Factores de Tiempo
4.
Inflamm Res ; 68(2): 147-155, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30506262

RESUMEN

BACKGROUND: The current study was intended to investigate the effect of ketamine (KET) on complete Freund's adjuvant (CFA)-induced arthritis in rats. METHODS: The CFA was administered in the hind paw of the rats for the induction of adjuvant-induced arthritis. The paw swelling of experimental animals was measured as hind paw volume. Hematoxylin and eosin staining was estimated and pathological changes in the joint tissues were observed under a light microscope. Furthermore, the bicinchoninic acid assay was used for protein quantification. The antibody-reactive bands were visualized using enhanced chemiluminescence. RESULTS: The present study showed that KET significantly reduces the severity of arthritis in CFA mice. The therapeutic effects were linked with reduced joint swelling and destruction, as evidenced by analyzing rat paws. The KET also revealed to attenuate the expression of pro-inflammatory cytokines (TNF-α, IL-1ß, IL-6). In western blot analysis, KET inhibit phosphorylation of MAPKs, IκBα and nuclear translocation NF-κB in the inflammatory joints of AIA rats. Moreover, KET showed to induce apoptosis via mitochondrial signalling pathways (Bcl2, Bax, cytochrome C, cleaved caspase-3 and cleaved caapse-9). CONCLUSION: Taken together, KET show significant anti-rheumatoid arthritis activity via multiple mechanisms and may thus have therapeutic benefits for RA.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Antagonistas de Aminoácidos Excitadores/uso terapéutico , Ketamina/uso terapéutico , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , FN-kappa B/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Administración Intravenosa , Animales , Apoptosis/efectos de los fármacos , Artritis Experimental/inducido químicamente , Artritis Experimental/patología , Citocinas/antagonistas & inhibidores , Citocinas/sangre , Edema/tratamiento farmacológico , Edema/patología , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Pie , Adyuvante de Freund/administración & dosificación , Proteínas I-kappa B/efectos de los fármacos , Articulaciones/patología , Ketamina/administración & dosificación , Masculino , Fosforilación , Ratas , Ratas Sprague-Dawley
5.
J. appl. oral sci ; 27: e20180150, 2019. graf
Artículo en Inglés | LILACS, BBO | ID: biblio-975883

RESUMEN

Abstract Objectives This investigation aimed to assess the differentiation inhibitory effects of ProRoot MTA® (PMTA) and Biodentine® (BIOD) on osteoclasts originated from murine bone marrow macrophages (BMMs) and compare these effects with those of alendronate (ALD). Materials and Methods Mouse BMMs were cultured to differentiate into osteoclasts with macrophage colony-stimulating factor and receptor activator of NF-κB (RANKL), treated with lipopolysaccharide. After application with PMTA, BIOD, or ALD, cell toxicities were examined using WST-1 assay kit, and RANKL-induced osteoclast differentiation and activities were determined by resorption pit formation assay and tartrate-resistant acid phosphate (TRAP) staining. The mRNA levels of osteoclast activity-related genes were detected with quantitative real time polymerase chain reaction. Expressions of molecular signaling pathways were assessed by western blot. All data were statistically analyzed with one-way ANOVA and Tukey's post-hoc test (p<0.05). Results Mouse BMMs applied with PMTA, BIOD, or ALD showed highly reduced levels of TRAP-positive osteoclasts. The BIOD treated specimens suppressed mRNA expressions of cathepsin K, TRAP, and c-Fos. Nonetheless, it showed a lower effect than PMTA or ALD applications. Compared with ALD, PMTA and BIOD decreased RANKL-mediated phosphorylation of ERK1/2 and IκBα. Conclusions PMTA and BIOD showed the inhibitory effect on osteoclast differentiation and activities similar to that of ALD through IκB phosphorylation and suppression of ERK signaling pathways.


Asunto(s)
Animales , Ratones , Osteoclastos/efectos de los fármacos , Materiales de Obturación del Conducto Radicular/farmacología , Células de la Médula Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Silicatos/farmacología , Compuestos de Calcio/farmacología , Alendronato/farmacología , Conservadores de la Densidad Ósea/farmacología , Osteoclastos/fisiología , Osteogénesis/efectos de los fármacos , Fosforilación/efectos de los fármacos , Resorción Radicular/prevención & control , Factores de Tiempo , Células de la Médula Ósea/citología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Western Blotting , Reproducibilidad de los Resultados , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas I-kappa B/efectos de los fármacos , Ligando RANK/análisis , Ligando RANK/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Fosfatasa Ácida Tartratorresistente
6.
Exp Cell Res ; 370(2): 490-497, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30017935

RESUMEN

To investigate the role of tumor necrosis factor-associated factor 6 (TRAF6) in high glucose-induced endothelial cell dysfunction. Human aortic endothelial cells (HAECs) were cultured in high glucose medium, and TRAF6 expression was assayed by quantitative real-time Polymerase Chain Reaction (PCR) and western blotting. The effect of TRAF6 on in vitro endothelial cell viability, apoptosis, migration, and endothelial-monocyte adhesion was investigated by gene knockdown. The expression of TRAF6 and related adhesion molecules was assayed in a mouse streptozotocin-induced type I diabetes model. The signaling pathways associated with TRAF6 effects on endothelial cells were investigated in high glucose HAEC cultures. Culture of HAECs in high glucose medium significantly increased TRAF6 mRNA and protein expression in a time dependent manner. High glucose markedly reduced HAEC viability, apoptosis, and migration, and these effects was significantly reversed by TRAF6 knockdown. High glucose significantly increased intercellular adhesion of THP-1 monocytic cells and HAECs via upregulation of ICAM-1 and VCAM-1 expression, and TRAF6 knockdown attenuated the effect on THP-1 cell adhesion. TRAF6, ICAM-1, and VCAM-1 expression were increased in aorta tissue of mice with streptozotocin-induced diabetes. The free radical scavenger N-acetyl-L-cysteine attenuated TRAF6 expression in HAECs cultured in high glucose medium, and TRAF6 knockdown inhibited high glucose-induced IκB-α degradation and JNK phosphorylation. TRAF6 mediated high glucose-induced endothelial dysfunction via NF-κB- and AP-1-dependent signaling. Targeting TRAF6 may delay progression of vascular diseases during diabetes mellitus and atherosclerosis.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Glucosa/farmacología , Factor 6 Asociado a Receptor de TNF/efectos de los fármacos , Animales , Moléculas de Adhesión Celular/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Glucosa/metabolismo , Humanos , Proteínas I-kappa B/efectos de los fármacos , Proteínas I-kappa B/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Ratones , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo
7.
Am J Physiol Endocrinol Metab ; 315(4): E543-E551, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29584445

RESUMEN

Several studies have demonstrated that protectins, ω-3 fatty acid-derived proresolution mediators, may ameliorate inflammation. Recently, protectin DX (PDX) was also reported to attenuate inflammation and insulin resistance in several cell types. However, the effects of PDX on inflammation in adipocytes remain ambiguous. In this study, we found that PDX treatment suppressed adipogenesis and lipid accumulation during 3T3-L1 differentiation. Treatment of differentiated 3T3-L1 cells with PDX stimulated AMP-activated protein kinase (AMPK) phosphorylation in a dose-dependent manner. PDX-induced AMPK phosphorylation blocked lipopolysaccharide (LPS)-induced secretion of proinflammatory cytokines, such as tumor necrosis factor-α and monocyte chemoattractant protein-1. Treatment of 3T3-L1 cells with PDX alleviated LPS-induced NF-κB and inhibitory factor κB phosphorylation. Furthermore, PDX treatment diminished LPS-induced impairment of insulin signaling and insulin-stimulated glucose uptake, as well as fatty acid oxidation. These effects were decreased by silencing AMPK expression with small-interfering RNA. In conclusion, the current findings suggest that PDX attenuates inflammation and insulin resistance in adipocytes via an AMPK-dependent pathway, which in turn provides evidence that PDX has anti-inflammatory and antidiabetic effects in adipocytes.


Asunto(s)
Adenilato Quinasa/efectos de los fármacos , Ácidos Docosahexaenoicos/farmacología , Inflamación/inmunología , Resistencia a la Insulina , FN-kappa B/efectos de los fármacos , Células 3T3-L1 , Adenilato Quinasa/metabolismo , Animales , Quimiocina CCL2/efectos de los fármacos , Quimiocina CCL2/inmunología , Glucosa/metabolismo , Proteínas I-kappa B/efectos de los fármacos , Proteínas I-kappa B/metabolismo , Insulina/metabolismo , Lipopolisacáridos/farmacología , Ratones , FN-kappa B/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/inmunología
8.
Brain Behav Immun ; 70: 141-156, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29458194

RESUMEN

Acute alcohol intoxication induces significant alterations in brain cytokines. Since stress challenges also profoundly impact central cytokine expression, these experiments examined the influence of acute and chronic stress on ethanol-induced brain cytokine responses. In Experiment 1, adult male rats were exposed to acute footshock. After a post-stress recovery interval of 0, 2, 4, or 24 h, rats were administered ethanol (4 g/kg; intragastric), with trunk blood and brains collected 3 h later. In non-stressed controls, acute ethanol increased expression of Il-6 and IκBα in the hippocampus. In contrast, rats exposed to footshock 24 h prior to ethanol demonstrated potentiation of hippocampal Il-6 and IκBα expression relative to ethanol-exposed non-stressed controls. Experiment 2 subsequently examined the effects of chronic stress on ethanol-related cytokine expression. Following a novel chronic escalating stress procedure, rats were intubated with ethanol. As expected, acute ethanol increased Il-6 expression in all structures examined, yet the Il-6 response was attenuated exclusively in the hippocampus in chronically stressed rats. Later experiments determined that neither acute nor chronic stress affected ethanol pharmacokinetics. When ethanol hypnosis was examined, however, rats exposed to chronic stress awoke at significantly lower blood ethanol levels compared to acutely stressed rats, despite similar durations of ethanol-induced sedation. These data indicate that chronic stress may increase sensitivity to ethanol hypnosis. Together, these experiments demonstrate an intriguing interaction between recent stress history and ethanol-induced increases in hippocampal Il-6, and may provide insight into novel pharmacotherapeutic targets for prevention and treatment of alcohol-related health outcomes based on stress susceptibility.


Asunto(s)
Etanol/metabolismo , Estrés Psicológico/metabolismo , Animales , Encéfalo/metabolismo , Enfermedad Crónica , Corticosterona/sangre , Citocinas/metabolismo , Etanol/farmacocinética , Etanol/farmacología , Hipocampo/metabolismo , Proteínas I-kappa B/efectos de los fármacos , Interleucina-1beta/efectos de los fármacos , Interleucina-6/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Estrés Psicológico/fisiopatología
9.
Drug Discov Ther ; 11(4): 206-211, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28867753

RESUMEN

We have reported that 3a,4,5,9b-tetrahydro-4-(1-naphthalenyl)-3H-cyclopentan[c]quinoline-8-sulfonamide (TQS), α7 nicotinic acetylcholine receptor (nAChR) positive allosteric modulator (PAM) reduces lipopolysaccharide (LPS)-induced hyperalgesia and allodynia in mice. The objective of the present study was to determine the effects of TQS on LPS-induced activation of hippocampal inhibitor of κB (IκB) and cluster of differentiation 11b (CD11b) gene expression involving hyperalgesia and allodynia in mice. We also examined the effects of TQS on microglial phenotype following LPS administration. Pretreatment of TQS (4 mg/kg) reduced the expressions of IκB and CD11b mRNA. Pretreatment of methyllycaconitine (3 mg/kg), an α7 nAChR antagonist, reversed TQS-induced decrease in IκB and CD11b mRNA expressions in the hippocampus indicating the involvement of α7 nAChR. In addition, TQS (4 mg/kg) reversed the LPS-induced microglial morphological changes. These results suggest that TQS reduces LPS-induced IκB and CD11b gene expression and microglial activation associated with hyperalgesia and allodynia by targeting microglial α7 nAChR in the hippocampus.


Asunto(s)
Antígeno CD11b/efectos de los fármacos , Hipocampo/efectos de los fármacos , Proteínas I-kappa B/efectos de los fármacos , Lipopolisacáridos/farmacología , Microglía/efectos de los fármacos , Naftalenos/farmacología , Quinolinas/farmacología , Sulfonamidas/farmacología , Receptor Nicotínico de Acetilcolina alfa 7/efectos de los fármacos , Aconitina/análogos & derivados , Aconitina/farmacología , Regulación Alostérica , Animales , Antígeno CD11b/genética , Expresión Génica/efectos de los fármacos , Hipocampo/metabolismo , Proteínas I-kappa B/genética , Masculino , Ratones , Microglía/patología , Antagonistas Nicotínicos/farmacología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/antagonistas & inhibidores
10.
Mar Drugs ; 15(9)2017 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-28832545

RESUMEN

Pseudopterosins are a group of marine diterpene glycosides which possess an array of biological activities including anti-inflammatory effects. However, despite the striking in vivo anti-inflammatory potential, the underlying in vitro molecular mode of action remains elusive. To date, few studies have examined pseudopterosin effects on cancer cells. However, to our knowledge, no studies have explored their ability to block cytokine release in breast cancer cells and the respective bidirectional communication with associated immune cells. The present work demonstrates that pseudopterosins have the ability to block the key inflammatory signaling pathway nuclear factor κB (NF-κB) by inhibiting the phosphorylation of p65 and IκB (nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor) in leukemia and in breast cancer cells, respectively. Blockade of NF-κB leads to subsequent reduction of the production of the pro-inflammatory cytokines interleukin-6 (IL-6), tumor necrosis factor alpha (TNFα) and monocyte chemotactic protein 1 (MCP-1). Furthermore, pseudopterosin treatment reduces cytokine expression induced by conditioned media in both cell lines investigated. Interestingly, the presence of pseudopterosins induces a nuclear translocation of the glucocorticoid receptor. When knocking down the glucocorticoid receptor, the natural product loses the ability to block cytokine expression. Thus, we hypothesize that pseudopterosins inhibit NF-κB through activation of the glucocorticoid receptor in triple negative breast cancer.


Asunto(s)
Productos Biológicos/farmacología , Citocinas/efectos de los fármacos , Diterpenos/farmacología , Glicósidos/farmacología , Proteínas I-kappa B/metabolismo , Leucemia Monocítica Aguda/tratamiento farmacológico , FN-kappa B/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Antiinflamatorios/farmacología , Linfocitos B/efectos de los fármacos , Productos Biológicos/química , Recuento de Células , Quimiocina CCL2/metabolismo , Citocinas/metabolismo , Diterpenos/química , Femenino , Glicósidos/química , Humanos , Proteínas I-kappa B/efectos de los fármacos , Interleucina-6/metabolismo , Biología Marina , FN-kappa B/metabolismo , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
11.
Inflammation ; 40(6): 1959-1966, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28761990

RESUMEN

In the present study, we investigated the anti-inflammatory effect of roburic acid on production of nitric oxide (NO) and interlukin-6 (IL-6) in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophage cells. We found that roburic acid reduced production of NO and IL-6, and the expression of inducible nitric oxide synthases (iNOS). Meanwhile, phosphorylation of inhibitor of κBα (IκBα) and IκB kinase α/ß (IKKα/ß), as well as translocation of nuclear factor-κB (NF-κB) to the nucleus, was suppressed by roburic acid treatment. In addition, phosphorylation of mitogen-activated protein kinase (MAPKs) including p38 and c-Jun-NH2-terminal kinase (JNK) was inhibited. Roburic acid exhibited inhibitory activities on production of NO and IL-6 via blocking IKK/IκB/NF-κB and MAPKs pathway, suggesting the potential application as a drug candidate for therapy of inflammatory diseases.


Asunto(s)
Interleucina-6/biosíntesis , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , FN-kappa B/efectos de los fármacos , Óxido Nítrico/biosíntesis , Triterpenos/farmacología , Animales , Gentianaceae/química , Quinasa I-kappa B/efectos de los fármacos , Proteínas I-kappa B/efectos de los fármacos , Interleucina-6/antagonistas & inhibidores , Lipopolisacáridos , Macrófagos/metabolismo , Ratones , Óxido Nítrico/antagonistas & inhibidores , Células RAW 264.7
12.
Inflammation ; 40(1): 13-20, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27696333

RESUMEN

Triggering receptors expressed on myeloid cell-1 (TREM-1) is a superimmunoglobulin receptor expressed on myeloid cells. TREM-1 amplifies the inflammatory response. Epoxyeicosatrienoic acids (EETs), the metabolites of arachidonic acid derived from the cytochrome P450 enzyme, have anti-inflammatory properties. However, the effects of EETs on TREM-1 expression under inflammatory stimulation remain unclear. Therefore, inhibition of soluble epoxide hydrolase (sEH) with a highly selective inhibitor [1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl) urea, TPPU] was used to stabilize EETs. LPS was intratracheally injected into mice to induce pulmonary inflammation, after TPPU treatment for 3 h. Histological examination showed TPPU treatment-alleviated LPS-induced pulmonary inflammation. TPPU decreased TREM-1 expression, but not DAP12 or MyD88 expression. Murine peritoneal macrophages were challenged with LPS in vitro. We found that TPPU reduced LPS-induced TREM-1 expression in a dose-dependent manner, but not DAP12 or MyD88 expression. TPPU also decreased downstream signal from TREM-1, reducing pro-inflammatory cytokine TNF-α and IL-1ß mRNA expression. Furthermore, TPPU treatment inhibited IkB degradation in vivo and in vitro. Our results indicate that the inhibition of sEH suppresses LPS-induced TREM-1 expression and inflammation via inhibiting NF-kB activation in murine macrophage.


Asunto(s)
Epóxido Hidrolasas/antagonistas & inhibidores , Glicoproteínas de Membrana/metabolismo , Células Mieloides/metabolismo , FN-kappa B/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Células Cultivadas , Citocinas/efectos de los fármacos , Citocinas/metabolismo , Expresión Génica/efectos de los fármacos , Proteínas I-kappa B/efectos de los fármacos , Proteínas I-kappa B/metabolismo , Macrófagos/metabolismo , Glicoproteínas de Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Compuestos de Fenilurea/farmacología , Piperidinas/farmacología , Neumonía/tratamiento farmacológico , Receptores Inmunológicos/efectos de los fármacos , Solubilidad , Receptor Activador Expresado en Células Mieloides 1
13.
Pharmazie ; 72(1): 5-9, 2017 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-29441890

RESUMEN

MicroRNAs (miRs) are reported to play key roles in various disease models. In this study, the functional role of miR-300 in the regulation of lung injury was explored to assess the feasibility of serum miR-300 as a potential biomarker for lung injury. Firstly, the expression of miR-300 was studied in the serum of 50 lung injury patients and 50 healthy controls. And the expression of miR-300 was also explored in the serum and lung tissues of mouse models. To further explore the possible mechanism in which miR-300 may contribute to lung injury, the target genes of miR-300 were predicted by TargetScan and validated using dual luciferase reporter assay. Moreover, the expression of inflammation factors was studied after transfection of miR-300 mimics and inhibitors into A549 cells. Here, we first identified that the level of miR-300 was significantly upregulated in the blood samples of acute lung injury patients compared with healthy control. Meanwhile, miR-300 was also found to be enhanced in the blood samples and lung tissues of LPS-induced mouse models. Further study showed that miR-300 significantly suppressed the expression of IκBα and luciferase reporter assay showed that IκBα was a target gene of miR-300. More importantly, the levels of inflammatory factors, such as TNFα, COX-2, iNOS, IL-6 and IL8, were significantly upregulated accompanied by overexpression of miR-300 in A549 cells. In summary, enhanced miR-300 expression in the peripheral blood contributed to the lung injury mainly by inhibiting the expression of IκBα.


Asunto(s)
Lesión Pulmonar Aguda/sangre , Lesión Pulmonar Aguda/inducido químicamente , Biomarcadores/análisis , Proteínas I-kappa B/efectos de los fármacos , Lipopolisacáridos/toxicidad , MicroARNs/sangre , Regiones no Traducidas 3'/efectos de los fármacos , Células A549 , Animales , Femenino , Humanos , Pulmón/química , Pulmón/metabolismo , Masculino , Ratones , Persona de Mediana Edad , FN-kappa B/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transfección
14.
Pharmazie ; 71(4): 213-7, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27209702

RESUMEN

1-O-acetylbritannilactone (ABL), a natural chemical component obtained from Chinese traditional medicine, Inula britannica, has been demonstrated to have anticancer activities. In the present study, we evaluated the anti-proliferative and the pro-apoptotic abilities of ABL alone or in combination with gemcitabine in human NSCLC cell line. A549 cells were treated, in vitro, with ABL, gemcitabine, and the combination of ABL and gemcitabine for 72 h. Our results showed ABL and gemcitabine inhibited cell growth and induced apoptosis of A549 cells. These effects after the combination of ABL and gemcitabine were superior to those of each alone. Furthermore, signal transduction analysis revealed NF-κB expression was significantly decreased by ABL and the combination treatment. IκBα and Bax levels were up regulated whereas Bcl-2 was substantially downregulated after all treatments. Our findings suggest that ABL combined with gemcitabine elicits a potent apoptosis of lung cancer cell and hence ABL has the potential to be developed as a chemotherapeutic agent.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antimetabolitos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Desoxicitidina/análogos & derivados , Lactonas/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Desoxicitidina/administración & dosificación , Humanos , Proteínas I-kappa B/efectos de los fármacos , Neoplasias Pulmonares/patología , Transducción de Señal , Proteína X Asociada a bcl-2/biosíntesis , Gemcitabina
15.
Acta Odontol Scand ; 74(4): 307-14, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26763602

RESUMEN

OBJECTIVE: The aim of this work is to investigate the possible role of Toll-like receptor 4 (TLR4) during the development of mouse tooth germ. TLR4 is well known to inhibit mineralization and cause inflammation in mature odontoblasts and dental pulp cells. However, unlike these pathological functions of TLR4, little is known about the developmental function(s) of TLR4 during tooth development. MATERIALS AND METHODS: TLR4 expression was studied via Western blot in developing lower mouse incisors from E13.5 to E18.5. To generate functional data about the effects of TLR4, a specific agonist (LPS) was applied to the medium of in vitro tooth germ cultures, followed by Western blot, histochemical staining, ELISA assay, in situ hybridization and RT-qPCR. RESULTS: Increased accumulation of biotin-labelled LPS was detected in the enamel organ and in preodontoblasts. LPS treatment induced degradation of the inhibitor molecule (IκB) of the NF-κB signalling pathway. However, no morphological alterations were detected in cultured tissue after LPS addition at the applied dosage. Activation of TLR4 inhibited the mineralization of enamel and dentin, as demonstrated by alizarin red staining and as decreased levels of collagen type X. mRNA expression of ameloblastin was elevated after LPS administration. CONCLUSION: These results demonstrate that TLR4 may decrease the mineralization of hard tissues of the tooth germ and may trigger the maturation of ameloblasts; it can give valuable information to understand better congenital tooth abnormalities.


Asunto(s)
Transducción de Señal/fisiología , Receptor Toll-Like 4/fisiología , Calcificación de Dientes/fisiología , Germen Dentario/fisiología , Ameloblastos/efectos de los fármacos , Animales , Colágeno Tipo X/análisis , Colágeno Tipo X/efectos de los fármacos , Esmalte Dental/efectos de los fármacos , Esmalte Dental/metabolismo , Proteínas del Esmalte Dental/análisis , Proteínas del Esmalte Dental/efectos de los fármacos , Dentina/efectos de los fármacos , Dentina/metabolismo , Órgano del Esmalte/efectos de los fármacos , Órgano del Esmalte/metabolismo , Proteínas I-kappa B/análisis , Proteínas I-kappa B/efectos de los fármacos , Lipopolisacáridos/farmacología , Ratones , Odontoblastos/efectos de los fármacos , Odontoblastos/metabolismo , Odontogénesis/efectos de los fármacos , Odontogénesis/fisiología , Técnicas de Cultivo de Órganos , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/efectos de los fármacos , Calcificación de Dientes/efectos de los fármacos , Germen Dentario/efectos de los fármacos
16.
Osteoarthritis Cartilage ; 24(1): 178-187, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26256766

RESUMEN

OBJECTIVE: To determine whether mandibular condylar cartilage degradation induced by experimentally abnormal occlusion could be ameliorated via systemic administration of strontium or NBD peptide. METHODS: Six-week-old female C57BL/6J mice were used. From the seventh day after mock operation or unilateral anterior crossbite (UAC) treatment, the control and UAC mice were further respectively pharmacologically treated for 2 weeks or 4 weeks of saline (CON + Saline and UAC + Saline groups), SrCl2 (CON + SrCl2 and UAC + SrCl2 groups) or NBD peptide (CON + NBD peptide and UAC + NBD peptide groups). Changes in condylar cartilage and subchondral bone were assessed 21 and 35 days after mock operation or UAC procedure by histology and micro-CT. Real-time PCR and/or immunohistochemistry (IHC) were performed to evaluate changes in expression levels of col2a1, aggrecan, ADAMTS-5, tnf-α, il-1ß, nfkbia, nuclear factor-kappaB phospho-p65 in condylar cartilage, and rankl/rank/opg in both condylar cartilage and subchondral bone. RESULTS: Cartilage degradation with decreased col2a1 and aggrecan expression, and increased ADAMTS-5, tnf-α/il1-ß, nfkbia and NF-κB phospho-p65 was observed in UAC + Saline groups. Subchondral bone loss with increased osteoclast numbers and decreased opg/rankl ratio was found in UAC + Saline groups compared to age-match CON + Saline groups. Cartilage degradation and subchondral bone loss were reversed by treatment of SrCl2 or NBD peptide while the same dosage in control mice induced few changes in condylar cartilage and subchondral bone. CONCLUSIONS: The results demonstrate reverse effect of systemic administration of strontium or NBD peptide on UAC-induced condylar cartilage degradation and subchondral bone loss.


Asunto(s)
Cartílago Articular/efectos de los fármacos , Maloclusión , Cóndilo Mandibular/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Péptidos/farmacología , ARN Mensajero/efectos de los fármacos , Estroncio/farmacología , Proteínas ADAM/efectos de los fármacos , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAMTS5 , Agrecanos/efectos de los fármacos , Agrecanos/genética , Agrecanos/metabolismo , Animales , Cartílago Articular/metabolismo , Colágeno Tipo II/efectos de los fármacos , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Oclusión Dental , Femenino , Proteínas I-kappa B/efectos de los fármacos , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Inmunohistoquímica , Interleucina-1beta/efectos de los fármacos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Cóndilo Mandibular/metabolismo , Cóndilo Mandibular/patología , Ratones , Ratones Endogámicos C57BL , Inhibidor NF-kappaB alfa , Osteoclastos/metabolismo , Osteoprotegerina/efectos de los fármacos , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Proteoglicanos/efectos de los fármacos , Proteoglicanos/metabolismo , Ligando RANK/efectos de los fármacos , Ligando RANK/genética , Ligando RANK/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Activador del Factor Nuclear kappa-B/efectos de los fármacos , Receptor Activador del Factor Nuclear kappa-B/genética , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Factor de Transcripción ReIA/efectos de los fármacos , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
17.
Diab Vasc Dis Res ; 13(1): 56-68, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26408644

RESUMEN

BACKGROUND: Glucagon-like peptide-1 receptor agonists may have a role in modulation of cardiac fibrosis. Our study aimed to determine the effect of the glucagon-like peptide-1 receptor agonist liraglutide in obesity, hypertension and age-induced murine models of cardiac fibrosis and identify associated molecular mechanisms. METHODS: C57Bl/6J mice on a high-fat diet and C57Bl/6J mice on a normal chow diet treated with angiotensin II were used to induce obesity and hypertension-mediated cardiac fibrosis, respectively. C57Bl/6J mice 20 months old were used to study age-induced cardiac fibrosis. Liraglutide treatment of 30 µg/kg/day-300 µg/kg s.c. twice daily was administered for 4 weeks. RESULTS: Liraglutide treatment attenuated obesity, hypertension and age-induced increases in interstitial cardiac fibrosis and expression of inflammatory and oxidative stress markers. CONCLUSIONS: These observations identify a potential role for liraglutide in the prevention of cardiac fibrosis and identify molecular mechanisms associated with these effects.


Asunto(s)
Envejecimiento/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón/agonistas , Cardiopatías/patología , Corazón/efectos de los fármacos , Incretinas/farmacología , Liraglutida/farmacología , Miocardio/patología , Estrés Oxidativo/efectos de los fármacos , Angiotensina II/farmacología , Animales , Aorta/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Quimiocina CCL2/efectos de los fármacos , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Fibrosis , Cardiopatías/metabolismo , Hipertensión/inducido químicamente , Hipertensión/metabolismo , Proteínas I-kappa B/efectos de los fármacos , Proteínas I-kappa B/metabolismo , Inmunohistoquímica , Inflamación , Interleucina-10/genética , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Subunidad p50 de NF-kappa B/efectos de los fármacos , Subunidad p50 de NF-kappa B/genética , Obesidad/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Vasoconstrictores/farmacología , Vimentina/efectos de los fármacos , Vimentina/metabolismo
18.
Arch Oral Biol ; 60(9): 1327-32, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26123747

RESUMEN

OBJECTIVE: Periodontal disease is one of the most prevalent oral diseases, which is associated with inflammation of the tooth-supporting tissues. Tormentic acid (TA), a triterpene isolated from Rosa rugosa, has been reported to exert anti-inflammatory effects. The aim of this study was to investigate the anti-inflammatory effects of TA on lipopolysaccharide (LPS)-stimulated human gingival fibroblasts (HGFs). METHODS: The levels of inflammatory cytokines such as interleukin (IL)-6 and chemokines such as IL-8 were detected by enzyme-linked immunosorbent assay (ELISA). The expression of Toll-like receptor 4 (TLR4), nuclear factor kappa B (NF-κB), IκBα, p38, extracellular signal-regulated kinase (ERK), and c-Jun N-terminal kinase (JNK) was determined by Western blotting. RESULTS: The results showed that Porphyromonas gingivalis LPS significantly upregulated the expression of IL-6 and IL-8. TA inhibited the LPS-induced production of IL-6 and IL-8 in a dose-dependent manner. Furthermore, TA inhibited LPS-induced TLR4 expression; NF-κB activation; IκBα degradation; and phosphorylation of ERK, JNK, and P38. CONCLUSION: TA inhibits the LPS-induced inflammatory response in HGFs by suppressing the TLR4-mediated NF-κB and mitogen-activated protein kinase (MAPK) signalling pathway.


Asunto(s)
Fibroblastos/metabolismo , Encía/citología , Triterpenos/farmacología , Western Blotting , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Proteínas I-kappa B/efectos de los fármacos , Proteínas I-kappa B/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lipopolisacáridos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , FN-kappa B/metabolismo , Porphyromonas gingivalis , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Triterpenos/administración & dosificación
19.
Inflamm Bowel Dis ; 21(9): 2005-15, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26076056

RESUMEN

BACKGROUND: An important role has emerged for calpain enzymes in regulating inflammation with one isoform, calpain-2, particularly important for macrophage activation. The goal of this study was to determine the therapeutic potential of a synthetic calpain-2 inhibitor, zLLY-CH2F, for colitis and inflammation-associated colorectal cancer. METHODS: Mice were then subjected to the azoxymethane/dextran sulfate sodium model of colitis and colitis-associated cancer incorporating intervention with daily injections of 0.75 mg/kg calpain-2 inhibitor beginning after the first signs of colitis. RESULTS: Calpain-2 inhibitor treatment alleviated weight loss and bloody diarrhea, and reduced inflammatory infiltration into colon tissues and inflammatory cytokine mRNA. Calpain-2 inhibitor intervention also reduced total colitis-associated cancer tumor volume by up to 70% in vehicle control mice and decreased cancer pathology scores of blinded histological colon tissue analyses. Mechanistic investigations showed that calpain-2 inhibition during macrophage activation reduced inhibitor of kappa beta (IκB) degradation and nuclear factor kappa beta (NFκB) nuclear localization as well as secretion of specific inflammatory cytokines. In addition, calpain-2 inhibitor treatment of CT26.WT mouse and HT-29 human colorectal cancer cells decreased proliferation and reduced IκB degradation and NFκB translocation. CONCLUSIONS: Overall, these findings suggest that intervention with a calpain-2 inhibitor may reduce colitis and colitis-associated cancer through a two-hit process of limiting macrophage activation and inhibiting growth of the colorectal cancer cells themselves.


Asunto(s)
Colitis/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Activación de Macrófagos/efectos de los fármacos , Oligopéptidos/farmacología , Animales , Azoximetano , Proliferación Celular , Colitis/inducido químicamente , Colitis/patología , Neoplasias del Colon/etiología , Neoplasias del Colon/patología , Citocinas/metabolismo , Sulfato de Dextran , Modelos Animales de Enfermedad , Células HT29/efectos de los fármacos , Humanos , Proteínas I-kappa B/efectos de los fármacos , Proteínas I-kappa B/genética , Inflamación/complicaciones , Inflamación/patología , Inyecciones Intraperitoneales , Ratones , FN-kappa B/efectos de los fármacos , FN-kappa B/genética , Translocación Genética
20.
Neurochem Res ; 40(6): 1220-31, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25998883

RESUMEN

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) has been recently shown to elicit inflammatory response in a number of cell-types. However, whether TCDD could provoke inflammation in astrocytes, the most abundant glial cells in central nervous system (CNS), remains virtually unknown. In the present study, we showed that TCDD exposure could induce evident astrocyte activation both in vivo and in vitro. Further, we found that TGF-ß-activated kinase 1 (TAK1), a critical regulator of NF-κB signaling, was rapidly phosphorylated in the process of TCDD-induced reactive astroglia. Exposure to TCDD led to rapid TAK1 and NF-κB p65 phosphorylation, as well as IKBα degradation. Moreover, blockage of TAK1 using siRNA oligos or TAK1 inhibitor 5Z-7-oxozeaenol significantly attenuated TCDD-induced astrocyte activation as well as the release of TNF-α. Finally, we showed that the conditioned medium of TCDD-treated astrocytes promoted the apoptosis of PC12 neuronal cells, which could be blocked with the pre-treatment of TAK1 inhibitor. Taken together, these findings suggested that TCDD could promote the inflammatory activation of astrocytes through modulating TAK1-NF-κB cascade, implicating that reactive astrocytes might contribute to TCDD-induced adverse effects on CNS system.


Asunto(s)
Astrocitos/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Quinasas Quinasa Quinasa PAM/efectos de los fármacos , FN-kappa B/efectos de los fármacos , Neuronas/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Animales , Muerte Celular/efectos de los fármacos , Células Cultivadas , Medios de Cultivo Condicionados , Femenino , Proteínas I-kappa B/efectos de los fármacos , Proteínas I-kappa B/metabolismo , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Células PC12 , Fosforilación , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Factor de Transcripción ReIA/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...