Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros

Banco de datos
Tipo del documento
Publication year range
1.
Inflamm Res ; 71(10-11): 1315-1325, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35962797

RESUMEN

BACKGROUND: Sepsis is a global fatal disease and leads to severe lung injury due to dysfunction of inflammation response. TRIM27 is closely related to the diseased with dysfunction of inflammation response. The aim of this study was to clarify the role and mechanism of TRIM27 in sepsis-induced lung injury. METHODS: The lipopolysaccharide (LPS)-induced septic mouse model was successfully established. The lung injury was evaluated by lung wet/dry (W/D) ratio and hematoxylin-eosin (H&E) staining. The cell apoptosis was evaluated by TUNEL assay. The inflammatory cytokines were measured by quantitative real time-PCR (qRT-PCR) assay and commercial enzyme-linked immunosorbent assay (ELISA). The oxidative stress was assessed by the contents of superoxide dismutase (SOD) and malondialdehyde (MDA), and the expression of dihydroethidium (DHE). RESULTS: In this study, we demonstrated that TRIM27 was up-regulated in LPS-induced septic mice. In loss-of-function experiments, knockdown of TRIM27 alleviated sepsis-induced lung injury, inflammation, apoptosis, and oxidative stress. More importantly, knockdown of TRIM27 was observed to reduce p-p65/NOX4 expression via suppressing ubiquitination of PPARγ. In rescue experiments, overexpression of NOX4 abolished the effect of sh-TRIM27 on alleviating sepsis-induced inflammation, apoptosis, and oxidative stress. CONCLUSION: These findings highlighted that knockdown of TRIM27 alleviated sepsis-induced inflammation, oxidative stress and apoptosis via suppressing ubiquitination of PPARγ and reducing NOX4 expression, which supports the potential utility of TRIM27 as a therapeutic target in septic lung injury.


Asunto(s)
Lesión Pulmonar Aguda , Sepsis , Ratones , Animales , Lipopolisacáridos/farmacología , PPAR gamma/genética , PPAR gamma/metabolismo , Estrés Oxidativo , Inflamación/tratamiento farmacológico , Sepsis/complicaciones , Sepsis/genética , Apoptosis , Lesión Pulmonar Aguda/tratamiento farmacológico , Ubiquitinación , NADPH Oxidasa 4/genética , NADPH Oxidasa 4/metabolismo , NADPH Oxidasa 4/farmacología , Proteínas de Unión al ADN/metabolismo , Ubiquitina-Proteína Ligasas
2.
J Periodontal Res ; 57(6): 1219-1226, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36205057

RESUMEN

OBJECTIVE AND BACKGROUND: Gingival overgrowth (GO) is a common side effect of some drugs such as anticonvulsants, immunosuppressant, and calcium channel blockers. Among them, the antiepileptic agent phenytoin is the most common agent related to this condition due to its high incidence. Transforming growth factor ß (TGFß) importantly contributes to the pathogenesis of GO. Connective tissue growth factor (CTGF or CCN2) is a key mediator of tissue fibrosis and is positively associated with the degree of fibrosis in GO. We previously showed that Src, c-jun N-terminal kinase, and Smad3 mediate TGFß1-induced CCN2 protein expression in human gingival fibroblasts (HGFs). This study investigates whether phenytoin can induce CCN2 synthesis through activated latent TGFß in HGFs and its mechanisms. METHODS: CCN2 synthesis, latent TGFß1 activation, and cellular reactive oxygen species (ROS) generation in HGFs were studied using western blot analysis, a TGFß1 Emax® ImmunoAssay System, and 2',7'-dichlorodihydrofluorescein diacetate (an oxidation-sensitive fluorescent probe), respectively. RESULTS: Phenytoin significantly stimulated CCN2 synthesis, latent TGFß1 activation, and ROS generation in HGFs. Addition of an TGFß-neutralizing antibody, TGFß receptor kinase inhibitor SB431542, and Smad3 inhibitor SIS3 completely inhibited phenytoin-induced CCN2 synthesis. General antioxidant N-acetylcysteine, NADPH oxidase (NOX) inhibitor diphenylene iodonium, and specific NOX4 inhibitor plumbagin almost completely suppressed phenytoin-induced total cellular ROS and latent TGFß1 activation. Curcumin dose-dependently decreased phenytoin-induced TGFß1 activation and CCN2 synthesis in HGFs. CONCLUSIONS: Our findings indicated that NOX4-derived ROS play pivotal roles in phenytoin-induced latent TGFß1 activation. Molecular targeting the phenytoin/NOX4/ROS/TGFß1 pathway may provide promising strategies for the prevention and treatment of GO. Curcumin-inhibited phenytoin-induced CCN2 synthesis is caused by the suppression of latent TGFß1 activation.


Asunto(s)
Curcumina , Sobrecrecimiento Gingival , Humanos , Encía/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/farmacología , Curcumina/farmacología , NADPH Oxidasa 4/metabolismo , NADPH Oxidasa 4/farmacología , Fenitoína/efectos adversos , Especies Reactivas de Oxígeno/metabolismo , Células Cultivadas , Fibroblastos , Factor de Crecimiento Transformador beta1/metabolismo , Sobrecrecimiento Gingival/inducido químicamente , Fibrosis
3.
J Nanobiotechnology ; 20(1): 241, 2022 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-35606794

RESUMEN

Periprosthetic osteolysis (PPO) triggered by wear particles is the most severe complication of total joint replacement (TJR) surgeries, representing the major cause of implant failure, which is public health concern worldwide. Previous studies have confirmed the specialized role of osteoclast-induced progressive bone destruction in the progression of PPO. Additionally, the reactive oxygen species (ROS) induced by wear particles can promote excessive osteoclastogenesis and bone resorption. Nicotinamide adenine dinucleotide phosphate oxidase 4 (NOX4), a cellular enzyme, is considered to be responsible for the production of ROS and the formation of mature osteoclasts. However, NOX4 involvement in PPO has not yet been elucidated. Therefore, we investigated the mechanism by which NOX4 regulates osteoclast differentiation and the therapeutic effects on titanium nanoparticle-induced bone destruction. We found that NOX4 blockade suppressed osteoclastogenesis and enhanced the scavenging of intracellular ROS. Our rescue experiment revealed that nuclear factor-erythroid 2-related factor 2 (Nrf2) silencing reversed the effects of NOX4 blockade on ROS production and osteoclast differentiation. In addition, we found increased expression levels of NOX4 in PPO tissues, while NOX4 inhibition in vivo exerted protective effects on titanium nanoparticle-induced osteolysis through antiosteoclastic and antioxidant effects. Collectively, these findings suggested that NOX4 blockade suppresses titanium nanoparticle-induced bone destruction via activation of the Nrf2 signaling pathway and that NOX4 blockade may be an attractive therapeutic approach for preventing PPO.


Asunto(s)
Nanopartículas , Osteólisis , Animales , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasa 4/metabolismo , NADPH Oxidasa 4/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Osteogénesis , Osteólisis/inducido químicamente , Osteólisis/tratamiento farmacológico , Osteólisis/metabolismo , Especies Reactivas de Oxígeno , Transducción de Señal , Titanio/farmacología
4.
Pharm Biol ; 60(1): 1449-1457, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35938505

RESUMEN

CONTEXT: A novobiocin derivative, XN4, has been shown to promote cell apoptosis in chronic myeloid leukaemia. OBJECTIVE: This study explores the mechanism by which XN4 promotes ferroptosis of gastric cancer (GC) cells. MATERIALS AND METHODS: Human GC SGC-7901 and BGC-823 cells were treated with different XN4 concentrations (0, 0.1, 0.5, 1.0, 5.0, and 10.0 µmol/L) to evaluate effects of XN4. Additionally, cells were pre-treated for 24 h with si-NOX4, for 1 h with the iron chelator deferoxamine mesylate (DFO) or for 1 h with the lipid peroxidation inhibitor liproxstatin-1 before being treated with XN4 to analyse the mechanism of XN4. RESULTS: XN4 increased cell death (IC50 values of XN4 on SGC-7901 and BGC-823 cells: 1.592 ± 0.14 µmol/L and 2.022 ± 0.19 µmol/L) and Fe2+ levels in SGC-7901 and BGC-823 cells. These effects of 2.0 µmol/L XN4 were abolished by 100 µmol/L DFO treatment. XN4 enhanced transferrin and transferrin receptor expression to induce Fe2+ accumulation. XN4 decreased mitochondrial membrane potentials in GC cells, similar to erastin. Additionally, XN4 increased MDA, hydrogen peroxide, and ROS levels, but diminished total glutathione levels. Liproxstatin-1 (200 nmol/L) nullified the effects of XN4 (2.0 µmol/L) on MDA levels and cell death. Moreover, GPX4 levels decreased, but NOX4 and ferroptosis-related protein PTGS2 levels increased in GC cells following XN4 treatment, which was nullified by NOX4 knockdown. DISCUSSION AND CONCLUSIONS: The pro-ferroptotic role of XN4 in GC might enable it to become a promising drug for GC treatment in the future despite the need for extensive research.


Asunto(s)
Ferroptosis , Neoplasias Gástricas , Apoptosis , Muerte Celular , Humanos , Peroxidación de Lípido , NADPH Oxidasa 4/metabolismo , NADPH Oxidasa 4/farmacología , Novobiocina/farmacología , Especies Reactivas de Oxígeno/metabolismo , Neoplasias Gástricas/tratamiento farmacológico
5.
Wei Sheng Yan Jiu ; 46(4): 615-620, 2017 Jul.
Artículo en Zh | MEDLINE | ID: mdl-29903185

RESUMEN

OBJECTIVE: To explore the role of NADPH oxidase 4( NOX4) in transforming growth factor-ß( TGF-ß)-induced A549 cells migration. METHODS: The A549 cells were allocated into five groups: TGF-ß( stimulation) group, Normal control group, DPI( NOX4 inhibitor) group, TGF-ß + DPI group, and DMSO( solvent control)group. The level of ROS were detected by flow cytometry instrument. The level of NOX4, snail and E-cadherin protein were detected by western blot. Use scratches experiment to express the change of A549 cells migration. RESULTS: After the quantification by Quantity One software, the expression of NOX4 in TGF-ß group is( 1. 80 ± 0. 07), the TGF-ß +DPI group is( 0. 49 ± 0. 03)( F = 327. 071, P < 0. 001). The change of EMT related protein: the expression of snail protein in TGF-ß group is( 9. 0 ± 0. 6), the TGF-ß + DPI group is( 1. 8 ± 0. 3)( F = 119. 097, P < 0. 001), the expression of E-cadherin protein in TGF-ß group is( 0. 5 ± 0. 1), the TGF-ß + DPI group is( 3. 3 ± 0. 3)( F = 71. 063, P <0. 001). These aboveresult indicate that DPI can inhibit the expression of NOX4 and EMTprogress in A549 cells. Then TGF-ß + DPI group compared with the TGF-ß group, the scratch healing rate is decreased( F = 33. 899, P < 0. 001). It illustrates that DPI can inhibit migration ability of A549 cells. CONCLUSION: After the NOX4 was inhibited by DPI, TGF-ß-induced migration of A549 cells was inhibited. And this phenomenon is associated with the progress of TGF-ß-induced EMT.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , NADPH Oxidasa 4/farmacología , Factor de Crecimiento Transformador beta1/fisiología , Línea Celular Tumoral , Humanos , Oxidorreductasas , Fosfatos , Factores de Crecimiento Transformadores
6.
Bioengineered ; 13(4): 8657-8666, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35331079

RESUMEN

Hyperoside (Hyp) is a flavonoid active compound deriving from Chinese herbal medicines. Increasing studies have implicated that Hyp may serve as a predominant promoting factor in osteoblast differentiation. This paper investigates whether Hyp could relieve glucocorticoid-induced osteonecrosis of the femoral head (GONFH) via promoting osteoblast survival and differentiation as well as to uncover its potential mechanism. GONFH cell model was induced by treating MC3T3-E1 cells with dexamethasone (DEX). The viability, apoptosis, and osteogenic differentiation of DEX-induced cells with the presence or absence of Hyp were assessed by CCK-8, Tunel, ALP assay, and ARS staining, respectively. The NADPH Oxidase 4 (NOX4) overexpression was performed by transfection with overexpression vector. Besides, western blot was used to determine the levels of apoptosis-, osteogenic differentiation-, and c-Jun N-terminal kinase (JNK) signaling-related proteins. It was noticed that Hyp caused no significant effects on the viability of MC3T3-E1 cells without any treatment but significantly enhanced the viability of DEX-induced cells. Besides, Hyp inhibited the apoptosis in DEX-induced cells but enhanced ALP activity and calcium nodule formation. Additionally, Hyp declined NOX4 expression in DEX-induced cells. However, NOX4 overexpression partially reversed the impacts of Hyp on DEX-exposed MC3T3-E1 cells. Finally, Hyp suppressed the activation of ROS/JNK pathway in DEX-induced cells, which was then counteracted by NOX4 overexpression. In conclusion, Hyp could promote the survival and differentiation of DEX-induced osteoblasts by targeting NOX4 to inhibit the ROS/JNK pathway. These results provide evidence for the application of Hyp in treating GONFH.


Asunto(s)
Proteínas Quinasas JNK Activadas por Mitógenos , Osteogénesis , Apoptosis , Línea Celular , Dexametasona/metabolismo , Dexametasona/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/farmacología , NADPH Oxidasa 4/genética , NADPH Oxidasa 4/metabolismo , NADPH Oxidasa 4/farmacología , Osteoblastos , Quercetina/análogos & derivados , Especies Reactivas de Oxígeno/metabolismo
7.
BMC Complement Med Ther ; 22(1): 257, 2022 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-36192741

RESUMEN

BACKGROUND: Baicalin is a generally available flavonoid with potent biological activity. The present study aimed to assess the underlying mechanism of baicalin in treatment of atherosclerosis (AS) with the help of network pharmacology, molecular docking and experimental validation. METHODS: The target genes of baicalin and AS were identified from public databases, and the overlapping results were considered to be baicalin-AS targets. Core target genes of baicalin were obtained through the PPI network and validated by a clinical microarray dataset (GSE132651). Human aortic endothelial cells (HAECs) were treated with Lipopolysaccharide (LPS) to construct an endothelial injury model. The expression of NOX4 was examined by real-time qPCR and western blot. Flow cytometry was used to detect intracellular levels of reactive oxygen species (ROS). Furthermore, HAECs were transfected with NOX4-specific siRNA and then co-stimulated with baicalin and LPS to investigate whether NOX4 was involved in the anti-oxidative stress effects of baicalin. RESULTS: In this study, baicalin had 45 biological targets against AS. Functional enrichment analysis demonstrated that most targets were involved in oxidative stress. Using the CytoHubba plug-in, we obtained the top 10 genes in the PPI network ranked by the EPC algorithm. Molecular docking and microarray dataset validation indicated that NOX4 may be an essential target of baicalin, and its expression was significantly suppressed in AS samples compared to controls. In endothelial injury model, intervention of HAECs with baicalin increased the expression levels of NOX4 and NOS3 (eNOS), and decreased LPS-induced ROS generation. After inhibition of NOX4, the anti-ROS-generating effect of baicalin was abolished. CONCLUSION: Collectively, we combined network pharmacology and endothelial injury models to investigate the anti-AS mechanism of baicalin. The results demonstrate that baicalin may exert anti-oxidative stress effects by targeting NOX4, providing new mechanisms and insights to baicalin for the treatment of AS.


Asunto(s)
Aterosclerosis , Células Endoteliales , Aterosclerosis/tratamiento farmacológico , Células Cultivadas , Flavonoides/farmacología , Humanos , Lipopolisacáridos , Simulación del Acoplamiento Molecular , NADPH Oxidasa 4/genética , NADPH Oxidasa 4/metabolismo , NADPH Oxidasa 4/farmacología , Farmacología en Red , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/farmacología , Especies Reactivas de Oxígeno/metabolismo
8.
Transpl Immunol ; 72: 101537, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35031454

RESUMEN

OBJECTIVE: To explore the effect of erythropoietin (EPO) on the AMP-activated protein kinase (AMPK)/nicotinamide adenine dinucleotide phosphatase oxidase 4 (NOX4) signaling pathway during renal ischemia reperfusion injury (RIRI) in rats. METHODS: A rat model of RIRI was established by clamping the left renal pedicle and removing the right kidney. The rats in the sham group did not have their left renal pedicle clamped. Rats with a model of RIRI were randomly divided into RIRI alone (control), erythropoietin treatment (EPO/RIRI), and Compound C treatment (CPC/RIRI) groups. Hematoxylin-eosin (H&E) staining was used to examine pathological kidney damage. Serum creatinine and urea nitrogen levels were measured to evaluate renal function. Western blotting was performed to detect the expression levels of phosphorylated p-AMPK and total AMPK protein in the kidneys. RT-PCR was used to evaluate the mRNA levels of Nox4 and p22 in the kidneys. Oxidative stress-related indices (ROS, CAT, GSH, SOD, and MDA) were also measured. RESULTS: EPO treatment improved kidney function by preventing kidney damage induced by the RIRI model. Preventing ischemia/reperfusion injury in the RIRI model was correlated with an increased p-AMPK/AMPK ratio and elevated activity of CAT, GSH, and SOD, which ameliorated the expression of NOX4, p22, ROS, and MDA. Moreover, treatment with CPC (an AMPK inhibitor) reduced the effects of EPO in the RIRI model. CONCLUSION: EPO treatment protected rats against RIRI in the RIRI model by alleviating oxidative stress by triggering the AMPK/NOX4/ROS pathway.


Asunto(s)
Eritropoyetina , Daño por Reperfusión , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Quinasas Activadas por AMP/farmacología , Animales , Eritropoyetina/metabolismo , Eritropoyetina/farmacología , Eritropoyetina/uso terapéutico , Riñón/metabolismo , NADPH Oxidasa 4/metabolismo , NADPH Oxidasa 4/farmacología , Estrés Oxidativo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Especies Reactivas de Oxígeno/farmacología , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa/farmacología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda