Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Altern Ther Health Med ; 30(2): 171-177, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37856812

RESUMEN

Tumor necrosis factor alpha-induced protein-3, also called A20, is a zinc-finger protein that participates in various inflammatory responses; however, the putative relationship between A20 and hepatic fibrosis remains unelucidated. Therefore, we investigated the role and mechanism of action of A20 in activating hepatic stellate cells (HSC) during the progression of hepatic fibrosis. Cell counting kit-8 (CCK8), colony growth, transwell assays, cell cycle analysis, and apoptosis assays were performed to explore the effect of A20 on cell function in vitro. An interspecies intravenous injection of the adeno-associated virus was used to assess the in vivo role of A20. The regulation of A20 on p65 was detected using mass spectrometry and immunoprecipitation. Our findings revealed that A20 was highly expressed in the liver tissues of patients with hepatic fibrosis and that the expression level of A20 in the liver tissue was closely correlated with the stage of liver fibrosis. In the LX-2 cell line, the downregulation of A20 upregulated the expression of fibrosis-related proteins and increased the expression of inflammatory factors, indicating the activation of HSC and vice versa. In addition, overexpression of A20 in mice reduced the degree of liver fibrosis in thioacetamide model mice. Finally, co-immunoprecipitation demonstrated that A20 could interact with p65. Hence, A20 inhibits HSC activation by binding to p65.


Asunto(s)
FN-kappa B , Factor de Necrosis Tumoral alfa , Humanos , Ratones , Animales , FN-kappa B/metabolismo , FN-kappa B/farmacología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Transducción de Señal , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/patología
2.
Stem Cells ; 42(4): 360-373, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38153253

RESUMEN

Recent investigations have shown that the necroptosis of tissue cells in joints is important in the development of osteoarthritis (OA). This study aimed to investigate the potential effects of exogenous skeletal stem cells (SSCs) on the necroptosis of subchondral osteoblasts in OA. Human SSCs and subchondral osteoblasts isolated from human tibia plateaus were used for Western blotting, real-time PCR, RNA sequencing, gene editing, and necroptosis detection assays. In addition, the rat anterior cruciate ligament transection OA model was used to evaluate the effects of SSCs on osteoblast necroptosis in vivo. The micro-CT and pathological data showed that intra-articular injections of SSCs significantly improved the microarchitecture of subchondral trabecular bones in OA rats. Additionally, SSCs inhibited the necroptosis of subchondral osteoblasts in OA rats and necroptotic cell models. The results of bulk RNA sequencing of SSCs stimulated or not by tumor necrosis factor α suggested a correlation of SSCs-derived tumor necrosis factor α-induced protein 3 (TNFAIP3) and cell necroptosis. Furthermore, TNFAIP3-derived from SSCs contributed to the inhibition of the subchondral osteoblast necroptosis in vivo and in vitro. Moreover, the intra-articular injections of TNFAIP3-overexpressing SSCs further improved the subchondral trabecular bone remodeling of OA rats. Thus, we report that TNFAIP3 from SSCs contributed to the suppression of the subchondral osteoblast necroptosis, which suggests that necroptotic subchondral osteoblasts in joints may be possible targets to treat OA by stem cell therapy.


Asunto(s)
Osteoartritis , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Animales , Humanos , Ratas , Necroptosis , Osteoartritis/metabolismo , Osteoartritis/patología , Osteoartritis/terapia , Osteoblastos/metabolismo , Osteoblastos/patología , Células Madre/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/farmacología
3.
Arch Gerontol Geriatr ; 117: 105274, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37995648

RESUMEN

BACKGROUND: The aging inflammatory microenvironment surrounding Leydig cells is linked to reduced testosterone levels in males. Tumor necrosis factor alpha-induced protein 3 (TNFAIP3) acts as a critical anti-inflammatory factor in various aging-related diseases. This study aims to investigate the protective effect of TNFAIP3 on testosterone production in Leydig cells under an aging inflammatory microenvironment. METHODS: Bioinformatics analysis examined TNFAIP3 expression differences in aging rat testes and validated the findings in aging mouse testes. In vitro models of inflammation were established using two Leydig cell lines, with tumor necrosis factor alpha (TNF-α) as the inflammatory factor. Lentiviral transduction was utilized to manipulate TNFAIP3 expression in these cell lines. Transcriptomic sequencing identified differentially expressed genes in TNFAIP3-overexpressing cells. RESULTS: Bioinformatics analysis and validation experiments revealed increased inflammatory signaling and elevated TNFAIP3 expression in aging rat and mouse testes. TNFAIP3 knockdown worsened testosterone synthesis inhibition and apoptosis in cells, while TNFAIP3 overexpression reversed these effects. Transcriptome analysis identified alterations in the P38MAPK pathway following TNFAIP3 overexpression. TNFAIP3 knockdown enhanced TNF-induced P38MAPK signaling, whereas its overexpression attenuated this effect. TNFAIP3 was found to regulate testosterone synthesis by upregulating CEBPB expression. CONCLUSIONS: TNFAIP3 exhibits inhibitory effects on apoptosis and promotes testosterone production in Leydig cells. The protective influence of TNFAIP3 on Leydig cells within an inflammatory microenvironment is likely mediated through by inhibiting the P38MAPK pathway and upregulating CEBPB expression.


Asunto(s)
Células Intersticiales del Testículo , Testosterona , Animales , Humanos , Masculino , Ratones , Ratas , Envejecimiento/fisiología , Células Intersticiales del Testículo/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
4.
Immun Inflamm Dis ; 11(10): e970, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37904691

RESUMEN

BACKGROUND: Endometritis seriously affects the health of women, and it is important to identify new targets for its treatment. OBJECTIVE: This study aimed to explore the role of TNFAIP3 interacting protein 2 (TNIP2) in endometritis through human endometrial epithelial cells (hEECs) stimulated by lipopolysaccharide (LPS). METHODS: hEECs were induced with LPS to build a cellular model of endometritis. Cell growth and apoptosis were detected by cell counting kit-8 and flow cytometry. The TNIP2 mRNA and protein levels were measured using reverse transcription quantitative polymerase chain reaction (RT-qPCR) and western blot analysis, respectively. The caspase3 activity was calculated using a Caspase3 activity kit. Interleukin (IL)-1ß, IL-6, and tumor necrosis factor-alpha (TNF-α) levels were determined by enzyme-linked-immunosorbent-assay. The reactive oxygen species (ROS), lactate dehydrogenase (LDH), catalase (CAT), and superoxide dismutase (SOD) levels were determined using the corresponding kits. Nuclear factor-kappaB (NF-κB) pathway was determined by western blot assay. RESULTS: TNIP2 was downregulated in the LPS-induced endometritis cell model. Cell viability was reduced, apoptosis was enhanced, and IL-6, IL-1ß, and TNF-α levels increased in LPS-induced hEECs. Additionally, LDH activity and ROS concentration were upregulated, whereas CAT and SOD activities were downregulated in LPS-induced hEECs. These results were reversed by TNIP2 overexpression. Moreover, the results hinted that NF-κB was involved in the effects of TNIP2 on the LPS-induced endometritis cell model. CONCLUSION: TNIP2 alleviated endometritis by inhibiting the NF-κB pathway, suggesting a potential therapeutic target for endometritis.


Asunto(s)
Endometritis , FN-kappa B , Humanos , Femenino , FN-kappa B/metabolismo , Endometritis/inducido químicamente , Endometritis/metabolismo , Lipopolisacáridos/toxicidad , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-6/farmacología , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/efectos adversos , Superóxido Dismutasa/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/farmacología , Proteínas Adaptadoras Transductoras de Señales/efectos adversos , Proteínas Adaptadoras Transductoras de Señales/metabolismo
5.
Neurosci Lett ; 805: 137223, 2023 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-37019273

RESUMEN

This study aimed to probe the function of tumor necrosis factor α-induced protein 3 (TNFAIP3) in the pathogenesis of Parkinson disease (PD) with its association with autophagy and inflammatory response. TNFAIP3 was reduced in the SN of PD patients (the GSE54282 dataset) and mice and in the MPP+-treated SK-N-SH cells. TNFAIP3 inhibited inflammatory response and enhanced autophagy, thereby alleviating PD in mice. NFκB and mTOR pathways were activated in the SN of PD mice and MPP+-treated cells. TNFAIP3 blocked the two pathways by preventing the p65 nuclear translocation and stabilizing DEPTOR, an endogenous inhibitor of mTOR. NFκB activator LPS and mTOR activator MHY1485 reversed the effects of TNFAIP3 on mitigation of injury in PD mice and in SK-N-SH cells induced with MPP+. Altogether, TNFAIP3 played a neuroprotective role in MPTP-induced mice by restricting NFκB and mTOR pathways.


Asunto(s)
Enfermedad de Parkinson , Ratones , Animales , Enfermedad de Parkinson/metabolismo , Transducción de Señal , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/farmacología , Serina-Treonina Quinasas TOR/metabolismo , FN-kappa B/metabolismo , Inflamación , Autofagia , Ratones Endogámicos C57BL , Línea Celular Tumoral
6.
Sci Total Environ ; 878: 163069, 2023 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-36996991

RESUMEN

The threat to public health posed by rapidly increasing levels of cadmium (Cd) in the environment is receiving worldwide attention. Although, Cd is known to be absorbed into the body and causes non-negligible damage to the liver, the detailed mechanisms underlying its hepatoxicity are incompletely understood. In the present study, investigated the effect of TNFAIP3 and α-ketoglutarate (AKG) on Cd-induced liver inflammation and hepatocyte death. Male C57BL/6 mice were exposed to cadmium chloride (1.0 mg/kg) while being fed a diet with 2 % AKG for two weeks. We found that Cd induced hepatocyte injury and inflammatory infiltration. In addition, TNFAIP3 expression was inhibited in the liver tissues and cells of CdCl2-treated mice. Mouse hepatocyte-specific TNFAIP3 overexpression by tail vein injection of an adeno-associated virus (AAV) vector effectively alleviated Cd-induced hepatic necrosis and inflammation, which was mediated by the NF-κB signaling pathway. Notably, this inhibitory effect of TNFAIP3 on Cd-induced liver injury was dependent on AKG. Exogenous addition of AKG prevented Cd exposure-induced increases in serum ALT, AST and LDH levels, production of pro-inflammatory cytokines, activation of the NF-κB signaling pathway, and even significantly reduced Cd-induced oxidative stress and hepatocyte death. Mechanistically, AKG exerted its anti-inflammatory effect by promoting the hydroxylation and degradation of HIF1A to reduce its Cd-induced overexpression in vivo and in vitro, avoiding the inhibition of the TNFAIP3 promoter by HIF1A. Moreover, the protective effect of AKG was significantly weaker in Cd-treated primary hepatocytes transfected with HIF1A pcDNA. Overall, our results reveal a novel mechanism of Cd-induced hepatotoxicity.


Asunto(s)
Cadmio , FN-kappa B , Masculino , Ratones , Animales , Cadmio/metabolismo , Ácidos Cetoglutáricos/metabolismo , Ácidos Cetoglutáricos/farmacología , Ratones Endogámicos C57BL , Hepatocitos , Inflamación/inducido químicamente , Hígado/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/farmacología
7.
Apoptosis ; 28(3-4): 498-513, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36587050

RESUMEN

Osteoblasts are important regulators of bone formation, but their roles in ankylosing spondylitis (AS) remain unclear. This study aims to explore the role of long non-coding RNA (lncRNA) maternally expressed 3 (MEG3) MEG3 in AS. Serum from AS patients as well as AS mesenchymal stem cells (ASMSCs) and healthy donors mesenchymal stem cells (HDMSCs) was collected. Accordingly, poorly expressed MEG3 and TNF alpha induced protein 3 (TNFAIP3) as well as overexpressed microRNA-125a-5p (miR-125a-5p) were noted in the serum of AS patients and in ASMSCs during the osteogenic induction process. Meanwhile, the interaction among MEG3, miR-125a-5p, and TNFAIP3 was determined and their effect on osteoblast activity was examined in vitro and in vivo. Overexpression of MEG3 and TNFAIP3 or inhibition of miR-125a-5p was found to inactivate the Wnt/ß-catenin pathway, thus suppressing osteogenic differentiation of MSCs. MEG3 competitively bound to miR-125a-5p to increase TNFAIP3 expression, thereby inactivating the Wnt/ß-catenin pathway and repressing the osteogenic differentiation of MSCs. In proteoglycan (PG)-induced AS mouse models, MEG3 also reduced osteogenic activity of MSCs to inhibit AS progression through the miR-125a-5p/TNFAIP3/Wnt/ß-catenin axis. Therefore, up-regulation of MEG3 or depletion of miR-125a-5p holds potential of alleviating AS, which sheds light on a new therapeutic strategy for AS treatment.


Asunto(s)
Células Madre Mesenquimatosas , MicroARNs , Espondilitis Anquilosante , Animales , Ratones , Apoptosis , beta Catenina/metabolismo , Diferenciación Celular/genética , MicroARNs/metabolismo , Osteogénesis/genética , Espondilitis Anquilosante/genética , Espondilitis Anquilosante/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/farmacología , Vía de Señalización Wnt/genética
8.
Biol Trace Elem Res ; 201(5): 2442-2457, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-35871203

RESUMEN

Apoptosis of kidney tubular epithelial cells contributes to diabetic kidney disease (DKD) pathophysiology, but the mechanisms are not fully understood. Zinc transporter protein member 8 (ZnT8, SLC30A8) is a susceptive gene in diabetes. Here, we aim to investigate whether ZnT8 has effects on pathophysiology of DKD. The animal groups include control, ZnT8KO mice, STZ-induced, and ZnT8-KO-STZ. STZ-induced DKD was performed in male C57BL/6 J mice and in ZnT8-KO mice. High glucose (HG)-induced apoptosis in a normal rat kidney tubular epithelial cell line (NRK-52E cells) was performed in vitro. Transfection of hZnT8-EGFP or TNFAIP3 siRNA was done in NRK-52E cells. Flow cytometry with Annexin V-FITC/PI double staining and TUNEL analysis was performed for the detection of apoptosis. Gene expression at mRNA and protein levels was examined with real-time RT-PCR and Western blot. Urine albumin to creatinine ratio, proinflammatory cytokines, and apoptosis were enhanced in kidneys of STZ and ZnT8-KO-STZ mice compared to control or ZnT8-KO mice. ZnT8 overexpression after hZnT8-EGFP transfection decreased HG-stimulated inflammatory activity and apoptosis in NRK-52E cells. Furthermore, treatment with ZnSO4 blunted HG-induced apoptosis and NF-κB activation. ZnSO4 increased the abundance of zinc-finger protein TNF-α-induced protein 3 (TNFAIP3). Also, ZnT8 over-expression after hZnT8-EGFP transfection significantly ameliorates HG-induced NF-κB-dependent transcriptional activity and apoptotic protein expressions in NRK-52E cells, but the inhibitory effect of ZnT8 was significantly abolished with TNFAIP3 siRNA. Our study provides evidence that ZnT8 has protective effects against apoptosis of renal tubular epithelial cells through induction of TNFAIP3 and subsequent suppression of the NF-κB pathway.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Ratas , Ratones , Masculino , Animales , FN-kappa B/metabolismo , Nefropatías Diabéticas/metabolismo , Ratones Endogámicos C57BL , Transducción de Señal , Células Epiteliales/metabolismo , Riñón/metabolismo , Túbulos Renales , Diabetes Mellitus/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/farmacología
9.
Cell Death Dis ; 10(10): 692, 2019 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-31534131

RESUMEN

The cytokine TNF promotes inflammation either directly by activating the MAPK and NF-κB signaling pathways, or indirectly by triggering cell death. A20 is a potent anti-inflammatory molecule, and mutations in the gene encoding A20 are associated with a wide panel of inflammatory pathologies, both in human and in the mouse. Binding of TNF to TNFR1 triggers the NF-κB-dependent expression of A20 as part of a negative feedback mechanism preventing sustained NF-κB activation. Apart from acting as an NF-κB inhibitor, A20 is also well-known for its ability to counteract the cytotoxic potential of TNF. However, the mechanism by which A20 mediates this function and the exact cell death modality that it represses have remained incompletely understood. In the present study, we provide in vitro and in vivo evidences that deletion of A20 induces RIPK1 kinase-dependent and -independent apoptosis upon single TNF stimulation. We show that constitutively expressed A20 is recruited to TNFR1 signaling complex (Complex I) via its seventh zinc finger (ZF7) domain, in a cIAP1/2-dependent manner, within minutes after TNF sensing. We demonstrate that Complex I-recruited A20 protects cells from apoptosis by stabilizing the linear (M1) ubiquitin network associated to Complex I, a process independent of its E3 ubiquitin ligase and deubiquitylase (DUB) activities and which is counteracted by the DUB CYLD, both in vitro and in vivo. In absence of linear ubiquitylation, A20 is still recruited to Complex I via its ZF4 and ZF7 domains, but this time protects the cells from death by deploying its DUB activity. Together, our results therefore demonstrate two distinct molecular mechanisms by which constitutively expressed A20 protect cells from TNF-induced apoptosis.


Asunto(s)
Receptores Tipo I de Factores de Necrosis Tumoral/efectos adversos , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/uso terapéutico , Ubiquitina/efectos de los fármacos , Animales , Apoptosis , Humanos , Ratones , Transducción de Señal , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/farmacología
10.
Neurosci Lett ; 712: 134480, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31493550

RESUMEN

LPS-induced microglial activation has a major influence on neuronal damage in the inflammatory process. Integral to this is the cellular and molecular interaction between microglia and neurons. Exosomes, a mediator of communication between cells, can transfer lipids, proteins and nucleic acids, affecting many donor and recipient cells. To investigate the mechanism by which microglial exosomes regulate neuronal inflammation after traumatic brain injury, this study primarily analyzed the effect of microglial exosomes on neuronal apoptosis. Exosomes derived from lipopolysaccharide (LPS)-activated microglial cultures were identified and purified. Neurons treated with these exosomes underwent apoptosis. A20 (also known as TNF-inducible protein 3, TNFAIP3) is a deubiquitinating enzyme with key anti-inflammatory functions. A20 is of huge significance to the degeneration and development of neuron. Importantly, A20 protects the exosomes-induced neuronal death, while A20 knockdown increases neuronal death. This study shows that exosomes may be critical for communication between microglia and neurons.


Asunto(s)
Apoptosis/efectos de los fármacos , Exosomas/efectos de los fármacos , Lipopolisacáridos/farmacología , Microglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/farmacología , Animales , Muerte Celular/efectos de los fármacos , Línea Celular , Exosomas/metabolismo , Ratones , Microglía/metabolismo , Neuronas/metabolismo , Ratas
11.
Med Sci Monit ; 23: 3536-3545, 2017 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-28724884

RESUMEN

BACKGROUND The aim of this study was to investigate the effects of zinc finger protein A20 on lipopolysaccharide (LPS)-induced pulmonary inflammation/anti-inflammatory mediators in an acute lung injury/acute respiratory distress syndrome (ALI/ARDS) rat model. MATERIAL AND METHODS Forty-eight ALI/ARDS rats were selected and assigned into normal saline (NS) (injected with NS), LPS (injected with LPS), LPS-C1 (injected with pEGFP-C1, NS and LPS), and A20 groups (injected with pEGFP-C1-A20, NS, and LPS). The wet/dry (W/D) ratio of rat lung tissues and total protein concentration and the number of neutrophils in bronchoalveolar lavage fluid (BALF) were detected. Enzyme-linked immunosorbent assay (ELISA) and qRT-PCR were applied to detect the protein and mRNA expressions of A20, IL-10, and TNF-α, respectively. Western blotting was employed to detect the protein expressions of A20, nuclear factor-kappa B (NF-κB) p65 and NF-κB p-P65 in rat lung tissues. RESULTS Compared with the NS group, the W/D ratio of rat lung tissues and total protein concentration and the number of neutrophils in BALF in the other 3 groups increased significantly. The protein and mRNA expressions of A20, IL-10, and TNF-α were significantly higher in the LPS group than in the NS group. The protein and mRNA expressions of A20 and IL-10 were significantly up-regulated and the expression of TNF-α, NF-κB p65, and NF-κB p-P65 was significantly down-regulated in rats injected with A20 compared to those in the LPS group. CONCLUSIONS The study provided evidence that zinc finger protein A20 can alleviate pulmonary inflammation by inhibiting TNF-α, NF-κB p65, and NF-κB p-P65 expressions and promoting IL-10 expression.


Asunto(s)
Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/farmacología , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/metabolismo , Animales , Antiinflamatorios/uso terapéutico , Líquido del Lavado Bronquioalveolar , Modelos Animales de Enfermedad , Mediadores de Inflamación , Interleucina-10/metabolismo , Lipopolisacáridos/farmacología , Pulmón/metabolismo , Masculino , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Neutrófilos/metabolismo , Proteínas de Transporte Nucleocitoplasmático/efectos de los fármacos , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Neumonía/inducido químicamente , Neumonía/tratamiento farmacológico , Neumonía/metabolismo , Ratas , Ratas Sprague-Dawley , Síndrome de Dificultad Respiratoria/metabolismo , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
12.
J Periodontal Res ; 51(4): 529-39, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26548452

RESUMEN

BACKGROUND AND OBJECTIVE: Although overexpression of the nuclear factor κB inhibitory and ubiquitin-editing enzyme A20 is thought to be involved in the pathogenesis of inflammatory diseases, its function in periodontal disease remains unknown. The aims of the present study were to evaluate A20 expression in patients with periodontitis and to study the effects of A20 overexpression, using a recombinant adenovirus encoding A20 (Ad-A20), on the inflammatory response and on osteoclastic differentiation in lipopolysaccharide (LPS)- and nicotine-stimulated human periodontal ligament cells (hPDLCs). MATERIAL AND METHODS: The concentration of prostaglandin E2 was measured by radioimmunoassay. Reverse transcription-polymerase chain reactions and western blot analyses were used to measure mRNA and protein levels, respectively. Osteoclastic differentiation was assessed in mouse bone marrow-derived macrophages using conditioned medium from LPS- and nicotine-treated hPDLCs. RESULTS: A20 was upregulated in the gingival tissues and neutrophils from patients with periodontitis and in LPS- and nicotine-exposed hPDLCs. Pretreatment with A20 overexpression by Ad-A20 markedly attenuated LPS- and nicotine-induced production of prostaglandin E2 , as well as expression of cyclooxygenase-2 and proinflammatory cytokines. Moreover, A20 overexpression inhibited the number and size of tartrate-resistant acid phosphatase-stained osteoclasts, and downregulated osteoclast-specific gene expression. LPS- and nicotine-induced p38 phosphorylation and nuclear factor κB activation were blocked by Ad-A20. Ad-A20 inhibited the effects of nicotine and LPS on the activation of pan-protein kinase C, Akt, GSK-3ß and protein kinase Cα. CONCLUSIONS: This study is the first to demonstrate that A20 overexpression has anti-inflammatory effects and blocks osteoclastic differentiation in a nicotine- and LPS-stimulated hPDLC model. Thus, A20 overexpression may be a potential therapeutic target in inflammatory bone loss diseases, such as periodontal disease.


Asunto(s)
Antiinflamatorios/farmacología , Encía/metabolismo , Osteoclastos/efectos de los fármacos , Ligamento Periodontal/efectos de los fármacos , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/farmacología , Animales , Antiinflamatorios/metabolismo , Western Blotting , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Dinoprostona/metabolismo , Perfilación de la Expresión Génica , Humanos , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Ratones , FN-kappa B/antagonistas & inhibidores , Nicotina/farmacología , Ligamento Periodontal/citología , Porphyromonas gingivalis , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...