Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(22): e2123202119, 2022 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-35617425

RESUMO

p73, a p53 family member, undergoes alternative splicing at the 3' end to produce multiple isoforms, but their expression and activity are largely unknown. Thus, CRISPR was used to knock out exon 12 (E12) in human cancer cell lines and mice, leading to isoform switch from p73α to isoform p73α1. We found that p73α1 is naturally expressed and induced by DNA damage. We also found that knockout of E12 suppresses cell growth and migration in H1299 and MIA PaCa-2 cells and promotes cellular senescence in mouse embryonic fibroblasts. Similarly, ectopic expression of p73α1 suppresses cell proliferation, whereas knockdown of p73α1 restores the cell proliferative and migratory capacities of E12−/− cells. Consistently, we found that E12+/− mice are not prone to spontaneous tumors. Instead, E12+/− mice are prone to systemic inflammation and exhibit elevated TNFα expression in inflamed tissues. Moreover, we found that Notch1, a master regulator of the inflammatory response, is regulated by p73α1 and highly expressed in E12−/− cells and inflamed E12+/− mouse tissues. Furthermore, through knockdown of p73α1 and/or Notch1 in E12−/− cells, we found that Notch1 is necessary for p73α1-mediated growth suppression. Together, these data suggest that p73α1 plays a critical role in tumor suppression and the inflammatory response via Notch1.


Assuntos
Genes Supressores de Tumor , Inflamação , Neoplasias , Receptor Notch1 , Proteína Tumoral p73 , Animais , Linhagem Celular Tumoral , Dano ao DNA , Éxons/genética , Técnicas de Inativação de Genes , Humanos , Inflamação/genética , Camundongos , Camundongos Knockout , Neoplasias/genética , Neoplasias/patologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo , Proteína Tumoral p73/genética , Proteína Tumoral p73/metabolismo
2.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 44(5): 777-784, 2022 Oct.
Artigo em Zh | MEDLINE | ID: mdl-36325774

RESUMO

Objective To explore the effect and mechanism of hesperidin in treating the lung injury in the mouse model of respiratory syncytial virus (RSV)-induced bronchiolitis. Methods A mouse model of RSV-induced bronchiolitis was established,and 60 BALB/c mice were assigned into a control group,a model group,a low-dose hesperidin (18 mg/kg) group,a high-dose hesperidin (36 mg/kg) group,and a high-dose hesperidin (36 mg/kg)+Jagged1(1 mg/kg) group by random number table method,with 12 mice in each group. Corresponding doses of drugs were administrated for intervention,and the control group and model group were administrated with the same amount of saline.The bronchoalveolar lavage fluid (BALF) samples were collected and alveolar macrophages were isolated.ELISA was employed to detect the levels of interleukin (IL)-4,IL-6,tumor necrosis factor-α (TNF-α),and IL-10 in BALF,and flow cytometry to detect the M1/M2 polarization of macrophages.qRT-PCR and Western blotting were respectively conducted to detect the mRNA and protein levels of inducible nitric oxide synthase (iNOS),arginase 1 (Arg-1),Jagged1,and Notch1 in the lung tissue. Results Compared with the control group,the modeling of RSV-induced bronchiolitis elevated the IL-4,IL-6,and TNF-α levels,increased the proportion of M1-type macrophages and the lung inflammation and mucus secretion scores,and up-regulated the mRNA and protein levels of iNOS,Jagged1,and Notch1 in BALF (all P<0.001).Meanwhile,the modeling lowered the IL-10 level,decreased the proportion of M2-type macrophages,and down-regulated the mRNA and protein levels of Arg-1 (all P<0.001).Compared with the model group,low- and high-dose hesperidin lowered the IL-4,IL-6,TNF-α levels,decreased the proportion of M1-type macrophages and the lung inflammation and mucus secretion scores,and down-regulated the mRNA and protein levels of iNOS,Jagged1,and Notch1 in BALF (all P<0.05).Moreover,hesperidin elevated the IL-10 level,increased the proportion of M2-type macrophages,and up-regulated the mRNA and protein levels of Arg-1 (all P<0.001).Using recombinant Jagged1 protein to activate Notch1 signaling pathway can significantly attenuate the promotion of high-dose hesperidin on M2 macrophage polarization and amelioration of lung inflammation damage (all P<0.01). Conclusion Hesperidin may alleviate the lung inflammation damage in mice with RSV-induced bronchiolitis by inhibiting the Jagged1/Notch1 signaling pathway and promoting the M2-type polarization of macrophages.


Assuntos
Bronquiolite , Hesperidina , Lesão Pulmonar , Animais , Camundongos , Bronquiolite/metabolismo , Hesperidina/farmacologia , Hesperidina/uso terapêutico , Hesperidina/metabolismo , Interleucina-10/metabolismo , Interleucina-10/farmacologia , Interleucina-4/metabolismo , Interleucina-4/farmacologia , Interleucina-6/metabolismo , Proteína Jagged-1/metabolismo , Proteína Jagged-1/farmacologia , Lesão Pulmonar/metabolismo , Macrófagos , Camundongos Endogâmicos BALB C , RNA Mensageiro/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
3.
BMC Ophthalmol ; 21(1): 451, 2021 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-34961513

RESUMO

BACKGROUND: Diabetic retinopathy (DR) is a common and potentially devastating microvascular complication of diabetes mellitus (DM). The main features of DR are inflammation and oxidative damage. Gliquidone (GLI) is confirmed to be a hypoglycemic drug by oral administration. The current study is aimed to investigate the role and mechanism of GLI on the pathogenesis of DR. METHODS: High glucose (HG)-induced human retinal endothelial cells (HRECs) were used to explore the anti-inflammatory and anti-oxidant effects of GLI on DR in vitro. Streptozotocin (STZ)-induced DM rats were used to investigate the effects of GLI on retinal structures, inflammation, and oxidative stress. The levels of SIRT1/Notch1 pathway-related proteins were determined by western blotting. RESULTS: GLI treatment promoted the viability and inhibited the apoptosis of HG-induced HRECs. Meanwhile, the levels of interleukin (IL)-6, IL-1ß, tumour necrosis factor alpha and reactive oxygen species were suppressed, while both catalase and superoxide dismutase were elevated after GLI treatment in HG-induced HRECs. Furthermore, we found that Silencing information regulator 2 related enzyme 1 (SIRT1) silencing reversed the inhibiting effects of GLI on the levels of protein Notch1 and effector genes Hes1 and Hey2. Similar anti-inflammatory and anti-oxidant effects of GLI in STZ-induced DM rats were observed. Additionally, GLI administration also repressed vascular hyperpermeability in vivo. CONCLUSION: GLI may be an effective agent to improve DR through repression of inflammation and oxidative stress via SIRT1/Notch1 pathway.


Assuntos
Diabetes Mellitus , Retinopatia Diabética , Animais , Retinopatia Diabética/tratamento farmacológico , Células Endoteliais/metabolismo , Estresse Oxidativo , Ratos , Receptor Notch1 , Retina/metabolismo , Sirtuína 1/metabolismo , Compostos de Sulfonilureia
4.
Dig Dis Sci ; 65(6): 1748-1760, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31655908

RESUMO

BACKGROUND AND AIMS: Taurine-upregulated gene 1 (TUG1) is reported to be upregulated and contributes to the progression of Pancreatic cancer (PC) by serving as an oncogene. Our aims were to explore the precise mechanism of TUG1 involved in PC pathogenesis. METHODS: TUG1 and miR-299-3p expression profiles were measured by qRT-PCR. The direct interaction between TUG1 and miR-299-3p was explored by luciferase reporter assay. MTT assay, flow cytometry analysis, caspase-3 activity assay, Transwell invasion assay and wound healing assay were performed to evaluate cell proliferative ability, apoptosis, caspase-3 activity, invasion and migration, respectively. Western blot was conducted to examine the expressions of Ki67, Bax, Bcl-2, matrix metalloproteinase-2 (MMP-2), MMP-9, E-cadherin, N-cadherin, Snail, Notch1, Survivin, and CyclinD1. In addition, animal experiments were also implemented. RESULTS: TUG1 was highly expressed, while miR-299-3p was underexpressed in PC tissues and PC cells. Furthermore, the significant increase of TUG1 in PC tissues of advanced patients (stage 3/4) was observed compared to patients (stage 1/2). TUG1 was negatively correlated with miR-299-3p expression in PC tissues. Moreover, TUG1 functioned as a molecular sponge of miR-299-3p to repress its expression. TUG1 knockdown suppressed cell proliferation, invasion, migration, and epithelial-mesenchymal transition (EMT), and induced apoptosis in PC cells, and repressed tumor growth and EMT in PC xenograft models, which were reversed following reintroduction with anti-miR-299-3p. Furthermore, we found that TUG1 silencing inactivated the Notch1 pathway in PC by upregulating miR-299-3p. CONCLUSIONS: The results reported that inhibition of TUG1/miR-299-3p axis suppressed PC malignant progression via suppression of the Notch1 pathway.


Assuntos
MicroRNAs/metabolismo , Neoplasias Pancreáticas/metabolismo , RNA Longo não Codificante/metabolismo , Receptor Notch1/metabolismo , Animais , Apoptose , Linhagem Celular , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Humanos , Camundongos , Camundongos Nus , MicroRNAs/genética , Neoplasias Experimentais , RNA Longo não Codificante/genética , Receptor Notch1/genética , Regulação para Cima
5.
Phytother Res ; 34(7): 1629-1637, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32067269

RESUMO

Curcumin (CUR) is a kind of polyphenolic compound and widely used in the treatment of diseases. However, the involvement of CUR in thymic carcinoma remains unknown. The object of our research is to clarify the role of CUR and related regulatory mechanism in thymic carcinoma cells. After treatment with CUR for 24 hr, cell viability, apoptosis, migration, and invasion of TC1889 cells were measured. Real-time polymerase chain reaction was executed to examine the expression of microRNA-27a (miR-27a) in thymic carcinoma tissues and TC1889 cells. After miR-27a mimic transfection, whether miR-27a is involved in CUR-modulated cell behaviors was measured. Finally, western blot was utilized to detect mTOR and Notch 1 pathways-linked proteins. CUR restrained cell viability and increased cell apoptosis of TC1889 cells. In addition, cell migration and invasion were restrained by CUR. Meanwhile, miR-27a expression was positively regulated in thymic carcinoma tissues and downregulated by CUR in TC1889 cells. Overexpressed miR-27a reversed the CUR-induced reduction of growth, migration, and invasion in TC1889 cells. Furthermore, CUR blocked mTOR and Notch 1 pathways via downregulating miR-27a. We demonstrated that CUR blocked mTOR and Notch 1 pathways via downregulating miR-27a, thereby suppressing cell growth, migration, and invasion of thymic carcinoma cells.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Curcumina/uso terapêutico , Neoplasias do Timo/tratamento farmacológico , Adulto , Anti-Inflamatórios não Esteroides/farmacologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Curcumina/farmacologia , Regulação para Baixo , Humanos , Pessoa de Meia-Idade , Transfecção
6.
Ren Fail ; 42(1): 381-390, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32338120

RESUMO

Transforming growth factor-ß1 (TGF-ß1) is a crucial factor implicated in the development of renal inflammation and tubulointerstitial fibrosis (TIF). The cytokine interleukin 22 (IL-22) was previously reported to involve in the pathogenesis of chronic inflammatory diseases, however recent studies showed that IL-22 could reduced inflammatory responses and tissue damage. In the present study, we aim to investigate the role and mechanisms of IL-22 in renal tubular cells inflammation and fibrosis induced by TGF-ß1. HK-2 cells were treated with TGF-ß1 in the presence of IL-22 or the Notch pathway inhibitor dibenzazepine (DBZ) for 48 h. Collagen I (Col I), fibronectin (FN), α-smooth muscle actin (α-SMA), vimentin and E-Cadherin were detected by western blot, proinflammatory factors (TNF-α, IL-6) and chemokines (MCP-1, RANTES) were evaluated by ELISA. Jagged1, Notch1, NICD1, and Hes1 were also detected by western blot. We found TGF-ß1 increased the levels of Col I, FN, α-SMA and vimentin in HK-2 cells compared with control, and decreased E-Cadherin level, however, IL-22 restored their expressions partly. IL-22 reduced overexpression of proinflammatory factors (TNF-α, IL-6) and chemokines (MCP-1, RANTES) levels induced by TGF-ß1, along with down-regulation of Jagged1, Notch, NICD1 and Hes1. Fibrosis and inflammation in renal tubular cells induced by TGF-ß1 could be attenuated by IL-22, and the effects were similar to DBZ treatment. Collectively, our study shows that IL-22 exerts a protective role in renal fibrotic and inflammatory responses induced by TGF-ß1 in vitro, which may be through inhibiting Jagged1/Notch1 signaling pathway activation.


Assuntos
Células Epiteliais/citologia , Interleucinas/farmacologia , Túbulos Renais/citologia , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta1/metabolismo , Linhagem Celular , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Fibrose/metabolismo , Humanos , Inflamação/metabolismo , Proteína Jagged-1/metabolismo , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/metabolismo , Receptor Notch1/metabolismo , Interleucina 22
7.
Genes Chromosomes Cancer ; 58(6): 396-401, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30578688

RESUMO

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematopoietic malignancy with few molecular alterations showing a consensual prognostic value. CRLF2 overexpression was recently identified in high-risk T-ALL patients. For these cases, no genomic abnormality was found to be associated with CRLF2 overexpression. IKZF1 has been recently shown to be a direct transcriptional regulator of CRLF2 expression. Moreover, it is known that NOTCH1 antagonizes IKZF1 in T-ALL. In light of these pieces of evidence, we reasoned that IKZF1 binding perturbation and CRLF2 upregulation could be associated in T-ALL. We evaluated two independent series of pediatric T-ALL cases (PHOP, n = 57 and TARGET, n = 264) for the presence of common T-ALL molecular abnormalities, such as NOTCH1/FBXW7 mutations. We also assessed CRLF2 and IKZF1 gene expression. CRLF2 overexpression was observed in 14% (PHOP) and 16% (TARGET) of T-ALL patients. No correlation was found between mRNA expression of CRLF2 and IKZF1 in both cohorts. Interestingly, we show that patients with mutations affecting NOTCH1-PEST domain and/or FBXW7 had higher CRLF2 expression (P = .04). In summary, we demonstrate for the first time that only mutations resulting in ICN1 (intracellular domain of NOTCH1) stabilization are associated with CRLF2 overexpression.


Assuntos
Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Receptor Notch1/genética , Receptores de Citocinas/genética , Humanos , Fator de Transcrição Ikaros/genética , Fator de Transcrição Ikaros/metabolismo , Mutação , Ligação Proteica , Domínios Proteicos , Estabilidade Proteica , Receptor Notch1/química , Receptor Notch1/metabolismo , Receptores de Citocinas/metabolismo
8.
J Neuroinflammation ; 16(1): 181, 2019 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-31526384

RESUMO

BACKGROUND: Remote ischemic preconditioning (RIPC) initiates endogenous protective pathways in the brain from a distance and represents a new, promising paradigm in neuroprotection against cerebral ischemia-reperfusion (I/R) injury. However, the underlying mechanism of RIPC-mediated cerebral ischemia tolerance is complicated and not well understood. We reported previously that preactivation of Notch1 mediated the neuroprotective effects of cerebral ischemic preconditioning in rats subjected to cerebral I/R injury. The present study seeks to further explore the role of crosstalk between the Notch1 and NF-κB signaling pathways in the process of RIPC-induced neuroprotection. METHODS: Middle cerebral artery occlusion and reperfusion (MCAO/R) in adult male rats and oxygen-glucose deprivation and reoxygenation (OGD/R) in primary hippocampal neurons were used as models of I/R injury in vivo and in vitro, respectively. RIPC was induced by a 3-day procedure with 4 cycles of 5 min of left hind limb ischemia followed by 5 min of reperfusion each day before MCAO/R. Intracerebroventricular DAPT injection and sh-Notch1 lentivirus interference were used to inhibit the Notch1 signaling pathway in vivo and in vitro, respectively. After 24 h of reperfusion, neurological deficit scores, infarct volume, neuronal apoptosis, and cell viability were assessed. The protein expression levels of NICD, Hes1, Phospho-IKKα/ß (p-IKK α/ß), Phospho-NF-κB p65 (p-NF-κB p65), Bcl-2, and Bax were assessed by Western blotting. RESULTS: RIPC significantly improved neurological scores and reduced infarct volume and neuronal apoptosis in rats subjected to I/R injury. OGD preconditioning significantly reduced neuronal apoptosis and improved cell viability after I/R injury on days 3 and 7 after OGD/R. However, the neuroprotective effect was reversed by DAPT in vivo and attenuated by Notch1-RNAi in vitro. RIPC significantly upregulated the expression of proteins related to the Notch1 and NF-κB pathways. NF-κB signaling pathway activity was suppressed by a Notch1 signaling pathway inhibitor and Notch1-RNAi. CONCLUSIONS: The neuroprotective effect of RIPC against cerebral I/R injury was associated with preactivation of the Notch1 and NF-κB pathways in neurons. The NF-κB pathway is a downstream target of the Notch1 pathway in RIPC and helps protect focal cerebral I/R injury.


Assuntos
Precondicionamento Isquêmico/métodos , NF-kappa B/metabolismo , Receptor Notch1/metabolismo , Traumatismo por Reperfusão/metabolismo , Animais , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Masculino , Neurônios/metabolismo , Neurônios/patologia , Ratos , Ratos Sprague-Dawley , Receptor Cross-Talk/fisiologia , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/patologia , Transdução de Sinais
9.
Neurochem Res ; 43(9): 1855-1861, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30066161

RESUMO

Malignant astrocytoma is the most common malignant tumor with strong invasion in the central nervous system. Tanshinone IIA is an effective compound to suppress cell proliferation and promote cell apoptosis. However, there is little research about the role of tanshinone IIA in the treatment of astrocytoma. This study aimed to investigate the effect of tanshinone IIA on migration, proliferation and apoptosis of astrocytoma cells. The efficacy of tanshinone IIA on migration, proliferation and apoptosis of astrocytoma cells were evaluated by flow cytometry and the assays of plate clone formation, CCK-8, wound healing and transwell migration. The protein molecule and signaling pathway were detected by western blot. High-dose tanshinone IIA suppressed migration and proliferation of astrocytoma cells while promoting apoptosis of astrocytoma cells. The western blot results showed that there were high Notch-1 protein expression and low c-Myc, MMP-9 and Bcl-2 activation in the high-dose tanshinone IIA group compared with the control group. High-dose tanshinone IIA suppresses astrocytoma cell proliferation, migration while promoting apoptosis through Notch-1 pathway. Tanshinone IIA may be used to develop new drugs for the treatment of astrocytoma.


Assuntos
Abietanos/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Receptor Notch1/efeitos dos fármacos , Abietanos/administração & dosagem , Proteínas Reguladoras de Apoptose/metabolismo , Astrocitoma/tratamento farmacológico , Linhagem Celular Tumoral , Humanos , Transdução de Sinais/efeitos dos fármacos
10.
Biochim Biophys Acta ; 1860(7): 1417-30, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27091611

RESUMO

BACKGROUND: The rapid development of multi-drug resistance (MDR) process has hindered the effectiveness of advanced hepatocellular carcinoma (HCC) treatments. Notch-1 pathway, which mediates the stress-response, promotes cell survival, EMT (epithelial-mesenchymal transition) process and induces anti-apoptosis in cancer cells, would be a potential target for overcoming MDR process. This study investigated the potential application of rhamnetin, a specific inhibitor of Notch-1 pathway, in anti-tumor drug sensitization of HCC treatment. METHODS: The expression of miR-34a, proteins belonging to Notch-1 signaling pathway or MDR-related proteins was detected by quantitative polymerase chain reaction (qPCR) and western blot assay. To identify whether rhamnetin induces the chemotherapeutic sensitization in HCC cells, the MTT-assays, flow cytometry, soft agar, trans-well and nude mice assays were performed. RESULTS: The endogenous expression of miR-34a was significantly increased and the expression of Notch-1 and Survivin was downregulated after rhamnetin treatment. Treatment of rhamnetin also reduced the expression of MDR related proteins P-GP (P-glycoprotein) and BCRP (breast cancer resistance protein). Rhamnetin increased the susceptibility of HCC cells and especially HepG2/ADR, a MDR HCC cell line, to a small molecular kinase inhibitor sorafenib or chemotherapeutic drugs etoposide and paclitaxel. The IC(50) value of those drugs correspondingly decreased. CONCLUSIONS: Together, our findings suggest that rhamnetin treatment may attenuate the MDR process in HCC cells. These findings may contribute to more effective strategies for HCC therapy. GENERAL SIGNIFICANCE: Rhamnetin acts as a promising sensitizer to chemotherapy and may be a novel approach to overcome the MDR process of HCC.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Etoposídeo/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Niacinamida/análogos & derivados , Paclitaxel/farmacologia , Compostos de Fenilureia/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Quercetina/análogos & derivados , Animais , Carcinoma Hepatocelular/enzimologia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica , Células Hep G2 , Humanos , Concentração Inibidora 50 , Neoplasias Hepáticas/enzimologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Masculino , Camundongos SCID , MicroRNAs/genética , MicroRNAs/metabolismo , Niacinamida/farmacologia , Quercetina/farmacologia , Receptor Notch1/antagonistas & inibidores , Receptor Notch1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sorafenibe , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
11.
J Neurosci Res ; 95(9): 1850-1857, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28084632

RESUMO

Stroke is a leading cause of morbidity and mortality worldwide. Administration of Netrin-1 during the peri-infarct period has been shown to decrease infarct size in rats; however, the underlying mechanism is unclear. We addressed this question in the present study by inducing stroke in rats via middle cerebral artery occlusion (MCAO), and evaluating the effects of Netrin-1 treatment by neurobehavioral testing, immunocytochemistry, and western blotting. Netrin-1 overexpression increased neurobehavioral test scores and reduced cerebral infarct volume following MCAO via inhibition of the Notch1 signaling pathway. These results demonstrate that early administration of Netrin-1 can is an effective therapeutic approach for improving outcome after stroke. © 2017 Wiley Periodicals, Inc.


Assuntos
Netrina-1/metabolismo , Receptor Notch1/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Comportamento Animal/fisiologia , Masculino , Ratos , Ratos Wistar , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/patologia , Regulação para Cima
12.
Exp Cell Res ; 343(2): 148-158, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-27090014

RESUMO

PURPOSE: The aim of this study was to investigate the effect of downregulation of HIF-1α gene on human U251 glioma cells and examine the consequent changes of TMZ induced effects and explore the molecular mechanisms. METHODS: U251 cell line stably expressing HIF-1α shRNA was acquired via lentiviral vector transfection. The mRNA and protein expression alterations of genes involved in our study were determined respectively by qRT-PCR and Western blot. Cell proliferation was measured by MTT assay and colony formation assay, cell invasion/migration capacity was determined by transwell invasion assay/wound healing assay, and cell apoptosis was detected by flow cytometry. RESULTS: We successfully established a U251 cell line with highly efficient HIF-1α knockdown. HIF-1a downregulation sensitized U251 cells to TMZ treatment and enhanced the proliferation-inhibiting, invasion/migration-suppressing, apoptosis-inducing and differentiation-promoting effects exerted by TMZ. The related molecular mechanisms demonstrated that expression of O(6)-methylguanine DNA methyltransferase gene (MGMT) and genes of Notch1 pathway were significantly upregulated by TMZ treatment. However, this upregulation was abrogated by HIF-1α knockdown. We further confirmed important regulatory roles of HIF-1α in the expression of MGMT and activation of Notch1 pathways. CONCLUSION: HIF-1α downregulation sensitizes U251 glioma cells to the temozolomide treatment via inhibiting MGMT expression and Notch1 pathway activation.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Dacarbazina/análogos & derivados , Regulação para Baixo/efeitos dos fármacos , Glioma/tratamento farmacológico , Glioma/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dacarbazina/farmacologia , Dacarbazina/uso terapêutico , Regulação para Baixo/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma/patologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Invasividade Neoplásica , Proteínas de Neoplasias/metabolismo , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Temozolomida , Transfecção
13.
Int Immunopharmacol ; 128: 111521, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38246005

RESUMO

Otitis media with effusion (OME) is a recurrent middle ear inflammatory condition. It may be complicated by acquired hearing loss and speech impairment especially in children. Accordingly, the current study aimed to assess the role of cytokines and the imbalance of Th17/Tregs in the pathogenesis of OME. Additionally, the protective effect of astaxanthin and its mechanisms related to Notch1/ Hes1/mTORC1/S6K1 signalling were investigated. METHODS: Forty-eight children were grouped as follow: G1: control healthy group G2: acute otitis media (AOM) group, G3: OME group. In the lipopolysaccharide (LPS) induced OME rat model, 15 rats were randomised into: G1: normal control group, G2: LPS group, and G3: astaxanthin treated group. RESULTS: Biochemical analysis of the children's peripheral blood samples showed that IL1ß, IL-2, IL-4, IL-6, IL-17, and IL-23 were significantly elevated, while TGF-ß was significantly decreased in AOM and OME patients (group 2 and 3). In the LPS- induced OME rat model, astaxanthin treatment resulted in suppression of IL-17, IL-6, TNF-α, Muc5A, TFF3, NICD, Hes1, mTORC1, and S6K1 in rat middle ear mucosa. Furthermore, astaxanthin significantly downregulated RORγ while upregulating FoxP3 and restored the balance between Th17/Tregs. Moreover, astaxanthin improved the histopathological picture of the inflamed middle ear mucosa. CONCLUSIONS: Proinflammatory cytokines as well as Th17/Tregs imbalance play a crucial role in the pathogenesis of AOM and OME. Additionally, astaxanthin alleviated LPS- induced OME in rats through suppression of Notch1/ Hes1/mTORC1/S6K1 pathway, and regulation of Th17/Tregs.


Assuntos
Otite Média com Derrame , Otite Média , Humanos , Criança , Ratos , Animais , Citocinas/metabolismo , Otite Média com Derrame/etiologia , Otite Média com Derrame/metabolismo , Interleucina-17 , Interleucina-6 , Lipopolissacarídeos , Otite Média/complicações , Fatores de Transcrição HES-1 , Receptor Notch1 , Xantofilas
14.
Tissue Cell ; 91: 102554, 2024 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-39316936

RESUMO

Liver fibrosis is characterized by an excessive reparative response to various etiological factors, with the activated hepatic stellate cells (aHSCs) leading to extracellular matrix (ECM) accumulation. Senescence is a stable growth arrest, and the senescence of aHSCs is associated with the degradation of ECM and the regression of hepatic fibrosis, making it a promising approach for managing hepatic fibrosis. The role and specific mechanisms by which V-Type Proton ATPase Subunit G 3 (ATP6V1G3) influences senescence in activated HSCs during liver fibrosis remain unclear. Our preliminary results reveal upregulation of ATP6V1G3 in both human fibrotic livers and murine liver fibrosis models. Additionally, ATP6V1G3 inhibition induced senescence in aHSCs in vitro. Moreover, suppressing Notch1 reversed the senescence caused by ATP6V1G3 inhibition in HSCs. Thus, targeting ATP6V1G3, which appears to drive HSCs senescence through the Notch1 pathway, emerges as a potential therapeutic strategy for hepatic fibrosis.

15.
Transl Cancer Res ; 13(6): 2847-2859, 2024 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-38988940

RESUMO

Background: Osteosarcoma (OS) is a malignancy originating from mesenchymal tissue. Microfibril-associated protein 2 (MFAP2) plays a crucial role in cancer, notably promoting epithelial-mesenchymal transition (EMT). However, its involvement in OS remains unexplored. Methods: MFAP2 was silenced in U2OS cells using shRNA targeting MFAP2 (sh-MFAP2) and validated by quantitative real-time polymerase chain reaction (qRT-PCR). We extracted gene chip data of MFAP2 from multiple databases (GSE28424, GSE42572, and GSE126209). Correlation analyses between MFAP2 and the Notch1 pathway identified through the gene set variation analysis (GSVA) enrichment analysis were conducted using the Pearson correlation method. Cellular behaviors (viability, migration, and invasion) were assessed via the Cell Counting Kit-8 (CCK-8), wound healing, and Transwell assays. EMT markers (N-cadherin, vimentin, and ß-catenin) and Notch1 levels were examined by western blotting and qRT-PCR. Cell morphology was observed microscopically to evaluate EMT. Finally, the role of MFAP2 in OS was validated through a xenograft tumor model. Results: OS cell lines exhibited higher MFAP2 mRNA expression than normal osteoblasts. MFAP2 knockdown in U2OS cells significantly reduced viability, migration, and invasion, along with downregulation of N-cadherin and vimentin, as well as upregulation of ß-catenin. MFAP2 significantly correlated with the Notch1 pathway in OS and its knockdown inhibited Notch1 protein expression. Furthermore, Notch1 activation reversed the inhibitory effects of MFAP2 knockdown on the malignant characteristic of U2OS cells. Additionally, MFAP2 knockdown inhibited tumor growth, expression levels of EMT markers, and Notch1 expression in OS tumor tissues. Conclusions: Our study revealed that MFAP2 was an upstream regulator of the Notch1 signaling pathway to promote EMT in OS. These findings suggested MFAP2 as a potential OS therapy target.

16.
Artigo em Inglês | MEDLINE | ID: mdl-38242426

RESUMO

The core clinical characteristics of autism, which is a neurodevelopmental disease, involve repetitive behavior and impaired social interactions. Studies have shown that the Notch and Neuregulin1 (NRG1) signaling pathways are abnormally activated in autism, but the mechanism by which these two signaling pathways interact to contribute to the progression of autism has not been determined. Our results suggest that the levels of Notch1, Hes1, NRG1, and phosphorylated ErbB4 in the cerebellum (CB), hippocampus (HC), and prefrontal cortex (PFC) were increased in rats with valproic acid (VPA)-induced autism compared to those in the Con group. However, 3, 5-difluorophenyl-L-alanyl-L-2-phenylglycine tert-butyl (DAPT), which is a Notch pathway inhibitor, ameliorated autism-like behavioral abnormalities and decreased the protein levels of NRG1 and phosphorylated ErbB4 in rats with VPA-induced autism; these results demonstrated that the Notch1/Hes1 pathway could participate in the pathogenesis of autism by regulating the NRG1/ErbB4 signaling pathway. Studies have shown that the Notch pathway regulates microglial differentiation and activation during the onset of neurological disorders and that microglia affect autism-like behavior via synaptic pruning. Therefore, we hypothesized that the Notch1/Hes1 pathway could regulate the NRG1/ErbB4 pathway and thus participate in the development of autism by regulating microglial functions. The present study showed that AG1478, which is an ErbB4 inhibitor, ameliorated the autism-like behaviors in a VPA-induced autism rat model, reduced abnormal microglial activation, and decreased NRG1 and Iba-1 colocalization; however, AG1478 did not alter Notch1/Hes1 activity. These results demonstrated that Notch1/Hes1 may participate in the microglial activation in autism by regulating NRG1/ErbB4, revealing a new mechanism underlying the pathogenesis of autism.


Assuntos
Transtorno Autístico , Quinazolinas , Tirfostinas , Animais , Ratos , Transtorno Autístico/induzido quimicamente , Neuregulina-1 , Microglia , Ácido Valproico , Fatores de Transcrição HES-1 , Receptor Notch1
17.
Neuropharmacology ; 239: 109682, 2023 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-37543138

RESUMO

As a pervasive neurodevelopmental disease, autism spectrum disorder (ASD) is caused by both hereditary and environmental elements. Research has demonstrated the functions of the Notch pathway and DNA methylation in the etiology of ASD. DNA methyltransferases DNMT3 and DNMT1 are responsible for methylation establishment and maintenance, respectively. In this study, we aimed to explore the association of DNA methyltransferases with the Notch pathway in ASD. Our results showed Notch1 and Hes1 were upregulated, while DNMT3A and DNMT3B were downregulated at the protein level in the prefrontal cortex (PFC), hippocampus (HC) and cerebellum (CB) of VPA-induced ASD rats compared with Control (Con) group. However, the protein levels of DNMT3A and DNMT3B were augmented after treatment with 3,5-difluorophenacetyl-L-alanyl-S-phenylglycine-2-butyl ester (DAPT), suggesting that abnormal Notch pathway activation may affect the expression of DNMT3A and DNMT3B. Besides, our previous findings revealed that the Notch pathway may participate in development of ASD by influencing autophagy. Therefore, we hypothesized the Notch pathway adjusts autophagy and contributes to ASD by affecting DNA methyltransferases. Our current results showed that after receiving the DNA methyltransferase inhibitor 5-Aza-2'-deoxycytidine (5-Aza-2'dc), the VPA + DAPT+5-Aza-2'dc (V + D + Aza) group exhibited reduced social interaction ability and increased stereotyped behaviors, and decreased expression of DNMT3A, DNMT3B and autophagy-related proteins, but did not show changes in Notch1 and Hes1 protein levels. Our results indicated that the Notch1/Hes1 pathway may adjust DNMT3A and DNMT3B expression and subsequently affect autophagy in the occurrence of ASD, providing new insight into the pathogenesis of ASD.


Assuntos
Transtorno do Espectro Autista , Ácido Valproico , Ratos , Animais , Ácido Valproico/farmacologia , Transtorno do Espectro Autista/induzido quimicamente , Transtorno do Espectro Autista/genética , Metilação de DNA , Transdução de Sinais , Metilases de Modificação do DNA/metabolismo , DNA/metabolismo , Autofagia , Fatores de Transcrição HES-1/genética , Fatores de Transcrição HES-1/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo
18.
Exp Biol Med (Maywood) ; 248(11): 966-978, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37377100

RESUMO

N-acetylcysteine (NAC) has been reported to improve social interaction behavior, irritability, self-injury, and anxiety-like behavior in autism. However, the molecular mechanism underlying the therapeutic roles of NAC in autism remains unknown. This study mainly aimed to investigate the therapeutic effect of NAC on valproic acid (VPA)-induced autism model and the underlying mechanisms. Our results showed that NAC ameliorated the deficits in sociability and the anxiety- and repetitive-like behaviors displayed by VPA-exposed rats. In addition, VPA exposure induced autophagic deficiency and enhanced Notch-1/Hes-1 pathway activity based on lowered Beclin-1 and LC3B levels, while increased expression of p62, Notch-1, and Hes-1 expression at the protein level. However, NAC recovered VPA-induced autophagic deficiency and reduced Notch-1/Hes-1 pathway activity in a VPA-exposed autism rat model and SH-SY5Y neural cells. The present results demonstrated that NAC improves autism-like behavioral abnormalities by inactivating Notch-1/Hes-1 signaling pathway and recovering autophagic deficiency. Taken together, this study helps to elucidate a novel molecular mechanism that underlies the therapeutic actions of NAC in autism and suggests its potential to ameliorate behavioral abnormalities in neurodevelopmental disorders.


Assuntos
Transtorno Autístico , Neuroblastoma , Efeitos Tardios da Exposição Pré-Natal , Ratos , Humanos , Animais , Feminino , Transtorno Autístico/tratamento farmacológico , Acetilcisteína/farmacologia , Comportamento Animal , Ácido Valproico/efeitos adversos , Modelos Animais de Doenças , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente
19.
Inflammation ; 45(6): 2449-2464, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35705831

RESUMO

Ulcerative colitis (UC) is closely associated with disruption of intestinal epithelial tight junction proteins. A variety of studies have confirmed that resveratrol (RSV), a natural polyphenolic compound, has a potential anti-inflammatory effect and can regulate the expression of tight junction proteins. However, the mechanism by which RSV regulates the expression of tight junction proteins in the intestinal epithelium remains unclear. Therefore, we investigated the potential effect of RSV on tight junction proteins in an HT-29 cell model of inflammation induced by lipopolysaccharide (LPS) and explored its mechanism of action. First, the downregulated expression of the tight junction proteins occludin, ZO-1, and claudin-1 in the HT-29 cell model of inflammation induced by LPS was reversed by incubation with RSV, accompanied by a decrease in the expression of tumor necrosis factor α-converting enzyme (TACE). Additionally, the Notch1 pathway was attenuated and the expression of the inflammatory factors IL-6 and TNF-α was decreased by treatment with RSV. Second, after Jagged-1 was used in combination with RSV to reactivate the Notch1 pathway, the protective effects of RSV against the LPS-induced reductions in the expression of the tight junction proteins occludin, ZO-1, and claudin-1 and the decreases in the levels of the inflammatory factors IL-6 and TNF-α were abolished. These results suggest that RSV might regulate the expression of tight junction proteins by attenuating the Notch1 pathway.


Assuntos
Inflamação , Receptor Notch1 , Resveratrol , Proteínas de Junções Íntimas , Humanos , Claudina-1/metabolismo , Células HT29 , Inflamação/induzido quimicamente , Inflamação/metabolismo , Interleucina-6/metabolismo , Mucosa Intestinal/metabolismo , Lipopolissacarídeos , Ocludina/metabolismo , Receptor Notch1/metabolismo , Resveratrol/farmacologia , Proteínas de Junções Íntimas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
20.
Acta Pharm ; 72(4): 575-585, 2022 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-36651368

RESUMO

177Lu-EDTMP (Ethylenediamine tetramethylene phosphonic acid) is the most used radioactive agent for pain palliation in bone cancer patients. The present study aims to study the impact of relaxin-2 on the 177Lu-EDTMP associated cell toxicity and death in osteosarcoma cells. MG63 and Saos-2 cells were cultured with 177Lu-EDTMP (37 MBq) for 24 h with and without pretreatment of recombinant relaxin 2 (RLXH2) for 12 and 24 h. 177Lu-EDTMP associated cellular deterioration and death was determined by LDH, MTT, and trypan blue dye assays. ELISA-based kit was used to determine apoptotic DNA fragmentation. Western blotting was used to determine expression levels of apoptotic-related signalling pathway proteins like bcl2, poly(ADP-ribose) polymerase (PARP), and MAPK (mitogen-activated protein kinase). Our results found that RLXH2 counters 177Lu-EDTMP associated cellular toxicity. Similarly, RLXH2 was able to counter 177Lu-EDTMP induced cell death in a concentration and time--dependent manner. Furthermore, it was found that RLXH2 treatment prevents apoptosis in 177Lu-EDTMP challenged cells through activation of the notch-1 pathway in a concentration- and time-dependent manner. We reported that RLXH2 significantly declined cellular toxicity and apoptosis associated with 177Lu-EDTMP in MG63 and Saos-2 cells through the notch-1 pathway.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Relaxina , Humanos , Apoptose , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/complicações , Neoplasias Ósseas/metabolismo , Morte Celular , Osteossarcoma/tratamento farmacológico , Osteossarcoma/complicações , Relaxina/farmacologia , Linhagem Celular Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA