Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 175
Filtrer
1.
Int J Oncol ; 65(2)2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38873993

RÉSUMÉ

Genes encoding subunits of SWI/SNF (BAF) chromatin­remodeling complexes are recurrently mutated in a broad array of tumor types, and among the subunits, ARID1A is the most frequent target with mutations. In the present study, it was reported that ARID1A inhibits the epithelial­mesenchymal transition (EMT) and stemness of ovarian cancer cells, accompanied by reduced cell viability, migration and colony formation, suggesting that ARID1A acts as a tumor suppressor in ovarian cancer. Mechanistically, ARID1A exerts its inhibitory effects on ovarian cancer cells by activating the Hippo signaling pathway. Conversely, the overexpression of a gain­of­function transcriptional co­activator with PDZ­binding motif (TAZ) mutant (TAZ­Ser89) effectively reverses the effects induced by ARID1A. In addition, activation of Hippo signaling apparently upregulates ARID1A protein expression, whereas ectopic expression of TAZ­Ser89 results in the markedly decreased ARID1A levels, indicating a feedback of ARID1A­TAZ in regulating ovarian cancer cell EMT and stemness. Thus, the present study uncovered the role of ARID1A through the Hippo/TAZ pathway in modulating EMT and stemness of ovarian cancer cells, and providing with evidence that TAZ inhibitors could effectively prevent initiation and metastasis of ovarian cancer cases where ARID1A is lost or mutated.


Sujet(s)
Protéines de liaison à l'ADN , Transition épithélio-mésenchymateuse , Régulation de l'expression des gènes tumoraux , Voie de signalisation Hippo , Cellules souches tumorales , Tumeurs de l'ovaire , Protein-Serine-Threonine Kinases , Transduction du signal , Facteurs de transcription , Humains , Femelle , Tumeurs de l'ovaire/anatomopathologie , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/métabolisme , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Lignée cellulaire tumorale , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Mouvement cellulaire , Prolifération cellulaire , Transcriptional coactivator with PDZ-binding motif proteins/métabolisme , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique
2.
Neuromolecular Med ; 26(1): 9, 2024 Apr 03.
Article de Anglais | MEDLINE | ID: mdl-38568291

RÉSUMÉ

Aggregation of α-synuclein (α-syn) and α-syn cytotoxicity are hallmarks of sporadic and familial Parkinson's disease (PD). Nuclear factor (erythroid-derived 2)-like 2 (Nrf2)-dependent enhancement of the expression of the 20S proteasome core particles (20S CPs) and regulatory particles (RPs) increases proteasome activity, which can promote α-syn clearance in PD. Activation of peroxisome proliferator-activated receptor γ co-activator 1α (PGC-1α) may reduce oxidative stress by strongly inducing Nrf2 gene expression. In the present study, tetramethylpyrazine nitrone (TBN), a potent-free radical scavenger, promoted α-syn clearance by the ubiquitin-proteasome system (UPS) in cell models overexpressing the human A53T mutant α-syn. In the α-syn transgenic mice model, TBN improved motor impairment, decreased the products of oxidative damage, and down-regulated the α-syn level in the serum. TBN consistently up-regulated PGC-1α and Nrf2 expression in tested models of PD. Additionally, TBN similarly enhanced the proteasome 20S subunit beta 8 (Psmb8) expression, which is linked to chymotrypsin-like proteasome activity. Furthermore, TBN increased the mRNA levels of both the 11S RPs subunits Pa28αß and a proteasome chaperone, known as the proteasome maturation protein (Pomp). Interestingly, specific siRNA targeting of Nrf2 blocked TBN's effects on Psmb8, Pa28αß, Pomp expression, and α-syn clearance. In conclusion, TBN promotes the clearance of α-syn via Nrf2-mediated UPS activation, and it may serve as a potentially disease-modifying therapeutic agent for PD.


Sujet(s)
Facteur-2 apparenté à NF-E2 , Proteasome endopeptidase complex , Pyrazines , Humains , Animaux , Souris , Facteur-2 apparenté à NF-E2/génétique , alpha-Synucléine/génétique , Souris transgéniques , Ubiquitines
3.
Apoptosis ; 29(7-8): 1198-1210, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38553612

RÉSUMÉ

A number of studies have confirmed that Yes-associated protein (YAP)/transcriptional co-activator with PDZ-binding motif (TAZ)-transcriptional enhanced associate domain (TEAD) activity is the driver of cancer development. However, the role and mechanism of the YAP/TAZ-TEAD pathway in cervical intraepithelial neoplasia (CIN) remain to be clarified. Therefore, this study was designed to observe the effect of YAP/TAZ-TEAD activity on the development of CIN and provide new ideas for the diagnosis and treatment of CIN. Firstly, cervical tissues were collected from CIN patients in different stages [CIN grade 1 (CIN1) tissue, CIN grade 2/3 (CIN 2/3) and squamous cell carcinoma (SCC)] and healthy volunteers. Next, the expression levels of YAP, TAZ and TEAD in cervical tissues and cells were observed by immunohistochemistry, qRT-PCR and western blot. Besides, Z172 and Z183 cells were transfected with siRNA-YAP/TAZ (si-YAP/TAZ) and YAP/TAZ overexpression vector (YAP-5SA). Also, Z172 cells were co-transfected with YAP-5SA and si-TEAD2/4. Subsequently, the stemness characteristics, glycolysis level and malignant transformation of cells in each group were observed by sphere-formation assay, commercial kit, MTT, Transwell, scratch experiment, xenotransplantation and western blot.The expression of YAP, TAZ and TEAD increased significantly in cervical cancer tissue and cell line at the stage of CIN2/3 and SCC. When YAP/TAZ was knocked down, the stemness characteristics, glycolysis level and malignant transformation of cancer cells were notably inhibited; while activating YAP/TAZ exhibited a completely opposite result. In addition, activating YAP/TAZ and knocking down the TEAD expression at the same time significant weakened the effect of activated YAP/TAZ signal on precancerous cells and reduced inhibitory effect of knocking down TEAD alone. YAP/TAZ-TEAD signal activates the characteristics and Warburg effect of cancer stem cells, thereby promoting the malignant transformation of CIN.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Transformation cellulaire néoplasique , Cellules souches tumorales , Transactivateurs , Facteurs de transcription , Transcriptional coactivator with PDZ-binding motif proteins , Dysplasie du col utérin , Tumeurs du col de l'utérus , Protéines de signalisation YAP , Humains , Femelle , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Protéines de signalisation YAP/métabolisme , Protéines de signalisation YAP/génétique , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Tumeurs du col de l'utérus/génétique , Tumeurs du col de l'utérus/métabolisme , Tumeurs du col de l'utérus/anatomopathologie , Transcriptional coactivator with PDZ-binding motif proteins/métabolisme , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/métabolisme , Dysplasie du col utérin/anatomopathologie , Dysplasie du col utérin/génétique , Dysplasie du col utérin/métabolisme , Animaux , Transactivateurs/génétique , Transactivateurs/métabolisme , Facteurs de transcription à domaine TEA/métabolisme , Lignée cellulaire tumorale , Souris , Effet Warburg en oncologie , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Prolifération cellulaire/génétique , Souris nude , Régulation de l'expression des gènes tumoraux , Carcinome épidermoïde/génétique , Carcinome épidermoïde/métabolisme , Carcinome épidermoïde/anatomopathologie
4.
Behav Brain Res ; 465: 114972, 2024 May 08.
Article de Anglais | MEDLINE | ID: mdl-38552744

RÉSUMÉ

The hippocampal salt-inducible kinase 2 (SIK2)-CREB-regulated transcription co-activator 1 (CRTC1) system has been demonstrated to participate in not only the pathogenesis of depression but also the antidepressant mechanisms of several antidepressant medications including fluoxetine, paroxetine, and mirtazapine. Like fluoxetine, paroxetine is also a widely used selective serotonin (5-HT) reuptake inhibitor (SSRI). Recent studies have indicated that paroxetine also modulates several pharmacological targets other than the 5-HT system. Here, we speculate that paroxetine regulates the hippocampal SIK2-CRTC1 system. Chronic stress models of depression, various behavioral tests, western blotting, co-immunoprecipitation, quantitative real-time reverse transcription PCR, and genetic knockdown were used together in the present study. Our results show that the antidepressant actions of paroxetine in mice models of depression were accompanied by its preventing effects against chronic stress on hippocampal SIK2, CRTC1, and CRTC1-CREB binding. In contrast, genetic knockdown of hippocampal CRTC1 notably abrogated the antidepressant effects of paroxetine in mice. In summary, regulating hippocampal SIK2 and CRTC1 participates in the antidepressant mechanism of paroxetine, extending the knowledge of its pharmacological targets.


Sujet(s)
Fluoxétine , Paroxétine , Animaux , Souris , Antidépresseurs/pharmacologie , Fluoxétine/pharmacologie , Hippocampe/métabolisme , Paroxétine/pharmacologie , Sérotonine/métabolisme
5.
BMC Cancer ; 24(1): 229, 2024 Feb 19.
Article de Anglais | MEDLINE | ID: mdl-38373978

RÉSUMÉ

Transcriptional Co-Activator with PDZ-Binding Motif (TAZ, also known as WWTR1) is a downstream effector of the Hippo pathway, involved in the regulation of organ regeneration and cell differentiation in processes such as development and regeneration. TAZ has been shown to play a tumor-promoting role in various cancers. Currently, many studies focus on the role of TAZ in the process of mitophagy. However, the molecular mechanism and biological function of TAZ in renal clear cell carcinoma (KIRC) are still unclear. Therefore, we systematically analyzed the mRNA expression profile and clinical data of KIRC in The Cancer Genome Atlas (TCGA) dataset. We found that TAZ expression was significantly upregulated in KIRC compared with normal kidney tissue and was closely associated with poor prognosis of patients. Combined with the joint analysis of 36 mitophagy genes, it was found that TAZ was significantly negatively correlated with the positive regulators of mitophagy. Finally, our results confirmed that high expression of TAZ in KIRC inhibits mitophagy and promotes KIRC cell proliferation. In conclusion, our findings reveal the important role of TAZ in KIRC and have the potential to be a new target for KIRC therapy.


Sujet(s)
Néphrocarcinome , Tumeurs du rein , Mitophagie , Transcriptional coactivator with PDZ-binding motif proteins , Humains , Néphrocarcinome/génétique , Néphrocarcinome/anatomopathologie , Prolifération cellulaire/génétique , Analyse de profil d'expression de gènes , Protéines et peptides de signalisation intracellulaire/génétique , Tumeurs du rein/génétique , Tumeurs du rein/anatomopathologie , Mitophagie/génétique , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Transcriptional coactivator with PDZ-binding motif proteins/génétique
6.
Curr Eye Res ; 49(5): 524-532, 2024 05.
Article de Anglais | MEDLINE | ID: mdl-38305219

RÉSUMÉ

PURPOSE: Diabetic retinopathy (DR) is a major cause of irreversible blindness in the working-age population. Neovascularization is an important hallmark of advanced DR. There is evidence that Yes-associated protein (YAP)/transcriptional co-activator with a PDZ binding domain (TAZ) plays an important role in angiogenesis and that its activity is regulated by vascular endothelial growth factor (VEGF). Therefore, the aim of this study was to investigate the effect of YAP/TAZ-VEGF crosstalk on the angiogenic capacity of human retinal microvascular endothelial cells (hRECs) in a high-glucose environment. METHODS: The expression of YAP and TAZ of hRECs under normal conditions, hypertonic conditions and high glucose were observed. YAP overexpression (OE-YAP), YAP silencing (sh-YAP), VEGF overexpression (OE-VEGF) and VEGF silencing (sh-VEGF) plasmids were constructed. Cell counting kit-8 assay was performed to detect cells proliferation ability, transwell assay to detect cells migration ability, and tube formation assay to detect tube formation ability. The protein expression of YAP, TAZ, VEGF, matrix metalloproteinase (MMP)-8, MMP-13, vessel endothelium (VE)-cadherin and alpha smooth muscle actin (α-SMA) was measured by western blot. RESULTS: The proliferation of hRECs was significantly higher in the high glucose group compared with the normal group, as well as the protein expression of YAP and TAZ (p < 0.01). YAP and VEGF promoted the proliferation, migration and tube formation of hRECs in the high glucose environment (p < 0.01), and increased the expression of TAZ, VEGF, MMP-8, MMP-13 and α-SMA while reducing the expression of VE-cadherin (p < 0.01). Knockdown of YAP effectively reversed the above promoting effects of OE-VEGF (p < 0.01) and overexpression of YAP significantly reversed the inhibition effects of sh-VEGF on above cell function (p < 0.01). CONCLUSION: In a high-glucose environment, YAP/TAZ can significantly promote the proliferation, migration and tube formation ability of hRECs, and the mechanism may be related to the regulation of VEGF expression.


Sujet(s)
, Rétinopathie diabétique , Transcriptional coactivator with PDZ-binding motif proteins , Facteur de croissance endothéliale vasculaire de type A , Protéines de signalisation YAP , Humains , /métabolisme , Prolifération cellulaire , Rétinopathie diabétique/génétique , Rétinopathie diabétique/métabolisme , Cellules endothéliales/métabolisme , Glucose/pharmacologie , Glucose/métabolisme , Matrix Metalloproteinase 13/métabolisme , Matrix Metalloproteinase 13/pharmacologie , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Protéines de signalisation YAP/métabolisme , Transcriptional coactivator with PDZ-binding motif proteins/métabolisme , Rétine/métabolisme , Rétine/anatomopathologie
7.
BMC Pharmacol Toxicol ; 25(1): 7, 2024 01 03.
Article de Anglais | MEDLINE | ID: mdl-38173037

RÉSUMÉ

BACKGROUND: To comprehend the influences of fenofibrate on hepatic lipid accumulation and mitochondrial function-related signaling pathways in mice with non-alcoholic fatty liver disease (NAFLD) secondary to high-fat diets together with free fatty acids-influenced HepG2 cells model. MATERIALS AND METHODS: A random allocation of male 6-week C57BL/6J mice into three groups was done, including controls, model (14 weeks of a high-fat diet), and fenofibrate [similar to the model one with administered 0.04 g/(kg.d) fenofibrate by gavage at 11 weeks for 4 weeks] groups, which contained 10 mice each. This study verified NAFLD pathogenesis via mitochondrial functions in hepatic pathological abnormalities, liver index and weight, body weight, serum biochemical indexes, oxidative stress indicators, mitochondrial function indexes, and related signaling pathways. The effect of fenofibrate intervention was investigated in NAFLD model mice. In vitro, four groups based on HepG2 cells were generated, including controls, the FFA model (1.5 mmol/L FFA incubation for 24 h), LV-PGC-1α intervention (similar to the FFA model one after PPARGC1A lentivirus transfection), and LV control intervention (similar to the FFA model one after negative control lentivirus transfection) groups. The study investigated the mechanism of PGC-1α related to lipid decomposition and mitochondrial biosynthesis by Oil red O staining, colorimetry and western blot. RESULTS: In vivo experiments, a high-fat diet achieved remarkable changes regarding liver weight, liver index, serum biochemical indicators, oxidative stress indicators, liver pathological changes, mitochondrial function indicators, and body weight of the NAFLD model mice while fenofibrate improved the objective indicators. In the HepG2 cells model, the lipid accumulation increased significantly within the FFA model group, together with aggravated hepatocytic damage and boosted oxidative stress levels. Moreover, FFA induced excessive mitosis into fragmented in mitochondrial morphology, ATP content in cells decreased, mtDNA replication fold decreased, the expression of lipid decomposition protein PPARα reduced, mitochondrial biosynthesis related protein PGC-1α, NRF-1 and TFAM decreased. PGC-1α overexpression inhibited lipid deposition by improving mitochondrial biosynthesis and lipid decomposition. CONCLUSION: Fenofibrate up-regulated PPARα/PGC-1α signaling pathway, promoted mitochondrial ß-oxidation, reduced oxidative stress damage and lipid accumulation of liver. PGC-1α overexpression enhanced mitochondrial biosynthesis and ATP production, and reduced HepG2 intracellular accumulation of lipids and oxidative stress.


Sujet(s)
Fénofibrate , Stéatose hépatique non alcoolique , Mâle , Souris , Animaux , Stéatose hépatique non alcoolique/traitement médicamenteux , Stéatose hépatique non alcoolique/métabolisme , Fénofibrate/pharmacologie , Fénofibrate/usage thérapeutique , Récepteur PPAR alpha/génétique , Récepteur PPAR alpha/métabolisme , Souris de lignée C57BL , Foie , Mitochondries/métabolisme , Transduction du signal , Poids , Lipides , Adénosine triphosphate/métabolisme , Alimentation riche en graisse/effets indésirables
8.
Biochem Biophys Res Commun ; 692: 149323, 2024 Jan 15.
Article de Anglais | MEDLINE | ID: mdl-38043154

RÉSUMÉ

Osteoporosis is a metabolic bone loss disorder usually accompanied by overactivated osteoclast formation and increased bone resorption. Transcriptional co-activator with PDZ-binding motif (TAZ) is an emerging potential target for the treatment of osteoporosis. Our previous research showed that TAZ overexpression inhibited osteoclast formation while TAZ silencing had the opposite effect. In addition, TAZ knockout in mouse osteoclasts induced osteoporosis in animal experiments. XMU-MP-1 (XMU) is a selective MST1/2 inhibitor that can theoretically activate TAZ; however, its effect on osteoporosis remains unknown. In this study, we found that XMU treatment significantly increased TAZ expression in osteoclasts and inhibited osteoclast formation in vitro; however, this inhibitory effect was eliminated after the deletion of TAZ. Furthermore, XMU treatment upregulated TAZ expression in osteoclasts and alleviated ovariectomy (OVX)-induced osteoporosis in bilateral OVX mouse models. These findings suggest that XMU can effectively activate TAZ and that pharmacological activation of TAZ may be a promising option for the treatment of osteoporosis.


Sujet(s)
Ostéogenèse , Ostéoporose , Souris , Animaux , Femelle , Humains , Os spongieux , Ostéoporose/étiologie , Ostéoporose/induit chimiquement , Facteurs de transcription/génétique , Facteurs de transcription/pharmacologie , Ovariectomie
9.
Basic & Clinical Medicine ; (12): 141-146, 2024.
Article de Chinois | WPRIM (Pacifique Occidental) | ID: wpr-1018586

RÉSUMÉ

Objective To investigate the role and regulatory mechanism of stress-inducing protein 1(SESN1)in liver gluconeogenesis of fasting mice.Methods RT-qPCR was used to detect mRNA expression of SESN1 in liver tissues of C57BL/6J mice and primary mouse hepatocytes treated with forskolin(Fsk)and dexamethasone(Dex).HepG2 cells were transfected with plasmids and the effects of SESN1 overexpression on mRNA expression of gluconeogenesis related genes PGC-1α,PEPCK and G6Pase was detected by RT-qPCR.The effect of SESN1 on the promoter activity of PGC-1α in HepG2 cells was studied using a dual luciferase reporter system.The effect of SESN1 on PGC-1α deacetylation was detected by overexpression of SESN1 and inhibition of SIRT1 expression.By knocking down SIRT1 expression,we detected whether it mediated the changes in mRNA levels of SESN1 in-duced gluconeogenesis related genes.Results The mRNA expression of SESN1 was significantly increased in liver tissues of starved C57BL/6J mice and in primary hepatocytes treated with Fsk and Dex(P<0.001).Over-expression of SESN1 in HepG2 cells promoted mRNA expression of PGC-1α,PEPCK and G6Pase(P<0.001)and promoter activity of PGC-1α(P<0.001).Over-expression of SESN1 decreased the acetylation level of PGC-1α in primary hepatocytes.Sirt family inhibitors NAM and shRNA adenovirus interfered with SIRT1 expression respective-ly,and antagonized the deacetylation effect of SESN1 on PGC-1α.The expression of PGC-1α,PEPCK and G6Pase induced by SIRT1 was also significantly impaired(P<0.000 1).Conclusions SESN1 regulates liver gluconeogene-sis in mice with a SIRT1-dependent mechanism.

10.
Article de Chinois | WPRIM (Pacifique Occidental) | ID: wpr-1021910

RÉSUMÉ

BACKGROUND:Peroxisome proliferators-activated receptor gamma co-activator 1α(PGC-1α)is closely related to aging and plays an important regulatory role in exercise anti-aging.However,there is a lack of comprehensive reviews on the role of PGC-1α in exercise anti-aging from the perspective of different tissues and organs. OBJECTIVE:To provide a detailed overview of the role of PGC-1α in exercise anti-aging and discuss its regulation from the perspective of different tissues and organs. METHODS:A literature search was conducted from May 1,2023 to July 1,2023.The search covered self-built databases up to July 2023,as well as databases such as Web of Science,PubMed,China National Knowledge Infrastructure(CNKI),WanFang,and VIP.The Chinese search terms included"PGC-1α,peroxisome proliferators-activated receptor gamma co-activator 1α,PPARGC1A,aging,exercise,older adults".The English search terms were"PGC-1α,aging,exercise,exercise training,older adults".Boolean logical operators were used to connect the search terms,and corresponding search strategies were developed.Based on inclusion and exclusion criteria,83 articles were included in the review. RESULTS AND CONCLUSION:(1)PGC-1α is an important transcriptional coactivator that plays a key regulatory role in maintaining mitochondrial function,regulating energy metabolism,and adapting to different metabolic demands.(2)PGC-1α has a significant regulatory role in mitochondrial aging and various functions in multiple cell types,and is associated with inflammatory pathways,redox control,protein modifications,and epigenetic changes.(3)The expression level of PGC-1α can be increased by exercise training,and it exerts positive effects through regulating mitochondrial biogenesis,energy metabolism,and anti-oxidative stress pathways.It plays an important role in exercise-induced improvement of adipose tissue aging,cardiovascular aging,neurosystem aging,renal aging,skeletal muscle aging,and liver aging.(4)The expert group recommends future research directions including exploring the regulatory effects of different types,intensities,and durations of exercise on PGC-1α expression,studying the regulatory mechanisms of protein modifications and epigenetic changes in PGC-1α, and strengthening the research on the mechanisms of PGC-1α in different aging-related diseases.

11.
Article de Chinois | WPRIM (Pacifique Occidental) | ID: wpr-1031487

RÉSUMÉ

ObjectiveTo investigate the possible mechanism of Quyu Jiedu Formula (祛瘀解毒方) in the treatment of endometriosis in terms of iron autophagy mediated by nuclear receptor coactivator 4/nuclear factor κB (NCOA4/NF-κB) signalling pathway. MethodsFifty female SD rats were randomly divided into sham surgery group, model group, mifepristone group, low- and high-dose Quyu Jiedu Formula group, with 10 rats in each group. In the sham surgery group, only operation of opening and closing abdomen was performed, and in the remaining groups, the rat with endometriosis was modelled by autotransplantation. On the next day after successful modelling, saline 2 ml/d was given by gavage to the sham surgery group and the model group; mifepristone 1.05 mg/(kg·d) was given by gavage to the mifepristone group; Quyu Jiedu Formula 12.23 g/(kg·d) and 48.92 g/(kg·d) were given to the low- and high-dosage Quyu Jiedu Formula groups, respectively administered for 4 weeks consecutively. In the remaining 4 groups, all ectopic endometrial tissues were removed from the rats. The volume of ectopic lesions was measured in the model group, the mifepristone group, and the low- and high-dose Quyu Jiedu Formula groups, and the pathological changes of endometrial/ectopic tissues were observed by HE staining, and the protein expression and expression of NCOA4, Ferritin Heavy Chain 1 (FTH1), Panax quinquefolium (P62), Microtubule-associated Protein 1 Light Chain 3β (LC3B), and P-NF-κB protein expression and NCOA4, FTH1, LC3B, P62 mRNA expression were detected in the endometrium and ectopic tissues; the co-localisation of NCOA4 and LC3B, free iron content, and levels of interleukin 6 (IL-6) and tumour necrosis factor α (TNF-α) in endometrial/eutopic endometrial tissues were also detected. ResultsNo ectopic lesions were seen in the sham surgery group. The ectopic tissues of rats in the model group showed obvious pathological damage, while the pathological damage of the ectopic tissues of rats in each admi-nistration group was reduced to different degrees. Compared with the model group, the volume of ectopic lesions was reduced in the mifepristone group and the high- and low-dose Quyu Jiedu Formula groups, and the volume of ectopic lesions in the high-dose Quyu Jiedu Formula group and the mifepristone group was significantly smaller than that in the low-dose Quyu Jiedu Formula group (P<0.01). Compared with the sham surgery group, the ectopic tissues of the model group showed up-regulation of LC3BⅡ/LC3B I values, NCOA4, and P-NF-κB protein expression, down-regulation of P62 and FTH1 protein expression, increase in free iron content and IL-6 and TNF-α levels, and increase in the co-localisation positivity rate and co-localised cell density of NCOA4 and LC3B (P<0.05 or P<0.01). Compared with the model group, the ectopic endothelial tissue LC3BⅡ/LC3BⅠ values and the expression of NCOA4 and P-NF-κB proteins were down-regulated in the low- and high-dose Quyu Jiedu Formula group and mifepristone group, the colocalisation positivity rate of NCOA4 and LC3B significantly reduced, and the content of free iron and the level of IL-6 decreased (P<0.05 or P<0.01). Compared with the mifepristone group, P62 more obvious up-regulated and TNF-α level reduced in the high-dose Quyu Jiedu Formula group (P<0.05). Compared with the low-dose Quyu Jiedu Formula group, the free iron content of ectopic tissues and the levels of IL-6 and TNF-α reduced in the high-dose Quyu Jiedu Formula group (P<0.01). ConclusionThe mechanism of endometriosis treatment by Quyu Jiedu Formula may be related to the inhibition of iron autophagy mediated by the NCOA4/NF-κB signalling pathway in endometriotic tissues, which improves endometrial inflammation.

12.
J Adv Res ; 2023 Nov 19.
Article de Anglais | MEDLINE | ID: mdl-37989471

RÉSUMÉ

INTRODUCTION: Parkinson's disease (PD) is common neurodegenerative disease where oxidative stress and mitochondrial dysfunction play important roles in its progression. Tetramethylpyrazine nitrone (TBN), a potent free radical scavenger, has shown protective effects in various neurological conditions. However, the neuroprotective mechanisms of TBN in PD models remain unclear. OBJECTIVES: We aimed to investigate TBN's neuroprotective effects and mechanisms in PD models. METHODS: TBN's neuroprotection was initially measured in MPP+/MPTP-induced PD models. Subsequently, a luciferase reporter assay was used to detect peroxisome proliferator-activated receptor γ co-activator 1α (PGC-1α) promoter activity. Effects of TBN on antioxidant damage and the PGC-1α/Nuclear factor erythroid-2-related factor 2 (Nrf2) pathway were thoroughly investigated. RESULTS: In MPP+-induced cell model, TBN (30-300 µM) increased cell survival by 9.95 % (P < 0.05), 16.63 % (P < 0.001), and 24.09 % (P < 0.001), respectively. TBN enhanced oxidative phosphorylation (P < 0.05) and restored PGC-1α transcriptional activity suppressed by MPP+ (84.30 % vs 59.03 %, P < 0.01). In MPTP-treated mice, TBN (30 mg/kg) ameliorated motor impairment, increased striatal dopamine levels (16.75 %, P < 0.001), dopaminergic neurons survival (27.12 %, P < 0.001), and tyrosine hydroxylase expression (28.07 %, P < 0.01). Selegiline, a positive control, increased dopamine levels (15.35 %, P < 0.001) and dopaminergic neurons survival (25.34 %, P < 0.001). Additionally, TBN reduced oxidative products and activated the PGC-1α/Nrf2 pathway. PGC-1α knockdown diminished TBN's neuroprotective effects, decreasing cell viability from 73.65 % to 56.87 % (P < 0.001). CONCLUSION: TBN has demonstrated consistent effectiveness in MPP+-induced midbrain neurons and MPTP-induced mice. Notably, the therapeutic effect of TBN in mitigating motor deficits and neurodegeneration is superior to selegiline. The neuroprotective mechanisms of TBN are associated with activation of the PGC-1α/Nrf2 pathway, thereby reducing oxidative stress and maintaining mitochondrial function. These findings suggest that TBN may be a promising therapeutic candidate for PD, warranting further development and investigation.

13.
Mol Cell ; 83(19): 3421-3437.e11, 2023 10 05.
Article de Anglais | MEDLINE | ID: mdl-37751740

RÉSUMÉ

The nuclear receptor co-repressor (NCoR) complex mediates transcriptional repression dependent on histone deacetylation by histone deacetylase 3 (HDAC3) as a component of the complex. Unexpectedly, we found that signaling by the receptor activator of nuclear factor κB (RANK) converts the NCoR/HDAC3 co-repressor complex to a co-activator of AP-1 and NF-κB target genes that are required for mouse osteoclast differentiation. Accordingly, the dominant function of NCoR/HDAC3 complexes in response to RANK signaling is to activate, rather than repress, gene expression. Mechanistically, RANK signaling promotes RNA-dependent interaction of the transcriptional co-activator PGC1ß with the NCoR/HDAC3 complex, resulting in the activation of PGC1ß and inhibition of HDAC3 activity for acetylated histone H3. Non-coding RNAs Dancr and Rnu12, which are associated with altered human bone homeostasis, promote NCoR/HDAC3 complex assembly and are necessary for RANKL-induced osteoclast differentiation in vitro. These findings may be prototypic for signal-dependent functions of NCoR in other biological contexts.


Sujet(s)
Ostéoclastes , ARN , Humains , Souris , Animaux , Protéines corépressives/génétique , Ostéoclastes/métabolisme , Ligand de RANK/génétique , Corépresseur-1 de récepteur nucléaire/génétique , Corépresseur-1 de récepteur nucléaire/métabolisme , Expression des gènes
14.
Oncol Rep ; 50(4)2023 Oct.
Article de Anglais | MEDLINE | ID: mdl-37594135

RÉSUMÉ

Resistance to radiation therapy remains a treatment obstacle for patients with high­risk colorectal cancer. Neuromedin U (NMU) has been identified as a potential predictor of the response to radiation therapy by RNA sequencing analysis of colorectal cancer tissues obtained from patients. However, the role of NMU in colorectal cancer remains unknown. In order to investigate role of NMU in colorectal cancer, NMU expression was regulated using small interfering RNA or an NMU­expression pCMV3 vector, and cell counting, wound­healing and clonogenic assays were subsequently performed. NMU knockdown decreased colorectal cancer cell proliferation and migration, and sensitized the cells to radiation. Conversely, NMU overexpression increased radiation resistance, proliferation and migration of colorectal cancer cells. Furthermore, by western blotting and nuclear fractionation experiments, NMU knockdown inhibited the nuclear translocation of yes­associated protein (YAP) and transcriptional co­activator with PDZ­binding motif (TAZ), resulting from the phosphorylation of these proteins. By contrast, the nuclear translocation of YAP and TAZ was increased following NMU overexpression in colorectal cancer cells. Recombinant NMU regulated YAP and TAZ activity, and the expression of the YAP and TAZ transcriptional target genes AXL, connective tissue growth factor and cysteine­rich angiogenic inducer 61 in an NMU receptor 1 activity­dependent manner. These results suggested that NMU may contribute to the acquisition of radioresistance in colorectal cancer by enhancing the Hippo signaling pathway via YAP and TAZ activation.


Sujet(s)
Tumeurs colorectales , Neuropeptides , Radiotolérance , Humains , Tumeurs colorectales/génétique , Tumeurs colorectales/radiothérapie , Phosphorylation , Transduction du signal
15.
J Affect Disord ; 338: 228-238, 2023 10 01.
Article de Anglais | MEDLINE | ID: mdl-37257779

RÉSUMÉ

BACKGROUND: The salt-inducible kinase 1 (SIK1)-CREB-regulated transcription co-activator 1 (CRTC1) system in the paraventricular nucleus (PVN) of the hypothalamus has been demonstrated to participate in not only depression neurobiology but also the antidepressant mechanisms of fluoxetine, paroxetine, venlafaxine, and duloxetine. Like fluoxetine and paroxetine, escitalopram is also a well-known selective serotonin (5-HT) reuptake inhibitor (SSRI). However, recently it has been found that escitalopram can modulate a lot of targets other than the 5-HT system. Here, we speculate that escitalopram produces effects on the SIK1-CRTC1 system in the PVN. METHODS: Two mice models of depression (chronic social defeat stress (CSDS) and chronic unpredictable mild stress (CUMS)), various behavioral tests, enzyme linked immunosorbent assay (ELISA), western blotting, co-immunoprecipitation (Co-IP), quantitative real-time reverse transcription PCR (qRT-PCR), immunofluorescence, and adeno-associated virus (AAV)-mediated gene transfer were used together in the present study. RESULTS: It was found that escitalopram administration not only significantly prevented the hyperactivity of the hypothalamic-pituitary-adrenal (HPA) axis induced by CSDS and CUMS, but also notably reversed the effects of CSDS and CUMS on SIK1, CRTC1, and CRTC1-CREB binding in the PVN of mice. AAV-based genetic knock-down of SIK1 in PVN neurons evidently abolished the antidepressant-like effects of escitalopram in mice. LIMITATION: A shortage of this study is that only rodent models of depression were used, while human samples were not included. CONCLUSIONS: In summary, regulating the SIK1-CRTC1 system in the PVN participates in the antidepressant mechanism of escitalopram, which extends the knowledge of the pharmacological actions of escitalopram.


Sujet(s)
Escitalopram , Noyau paraventriculaire de l'hypothalamus , Souris , Humains , Animaux , Paroxétine , Fluoxétine , Sérotonine , Antidépresseurs/pharmacologie , Antidépresseurs/usage thérapeutique , Stress psychologique/traitement médicamenteux , Stress psychologique/métabolisme , Dépression/traitement médicamenteux , Dépression/génétique , Dépression/métabolisme
16.
Life Sci ; 322: 121671, 2023 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-37023953

RÉSUMÉ

In diabetic kidney disease (DKD), the long-term hyperactivation of yes-associated protein (YAP)/transcriptional coactivator PDZ-binding motif (TAZ) in renal proximal tubule epithelial cells (RPTCs) plays an important role in progressive tubulointerstitial fibrosis. Sodium-glucose cotransporter 2 (SGLT2) is highly expressed in RPTCs, but its relationship with YAP/TAZ in tubulointerstitial fibrosis in DKD is still unknown. The purpose of this study was to clarify whether the SGLT2 inhibitor (SGLT2i) dapagliflozin could alleviate renal tubulointerstitial fibrosis in DKD by regulating YAP/TAZ. We examined 58 patients with DKD confirmed by renal biopsy and found that the expression and nuclear translocation of YAP/TAZ increased with the exacerbation of chronic kidney disease classification. In models of DKD, dapagliflozin showed similar effects to verteporfin, an inhibitor of YAP/TAZ, in reducing the activation of YAP/TAZ and downregulating the expression of their target genes, connective tissue growth factor (CTGF) and amphiregulin in vivo and in vitro. Silencing SGLT2 also confirmed this effect. Importantly, dapagliflozin showed a better effect than verteporfin in inhibiting inflammation, oxidative stress and fibrosis in the kidney in DKD rats. Taken together, this study proved for the first time that dapagliflozin delayed tubulointerstitial fibrosis at least partly by inhibiting YAP/TAZ activation, which further enriched the antifibrotic effect of SGLT2i.


Sujet(s)
Diabète , Néphropathies diabétiques , Rats , Animaux , Protéines adaptatrices de la transduction du signal/métabolisme , Néphropathies diabétiques/traitement médicamenteux , Transduction du signal , Transporteur-2 sodium-glucose/métabolisme , Protéines de signalisation YAP , Vertéporfine/pharmacologie , Protéines du cycle cellulaire/métabolisme , Fibrose
17.
Methods Mol Biol ; 2576: 155-169, 2023.
Article de Anglais | MEDLINE | ID: mdl-36152184

RÉSUMÉ

Peroxisome proliferator-activated receptors are a family of nuclear hormone receptors that control the expression of genes involved in a variety of physiologic processes, through heterodimerization with retinoid X receptor and complex formation with various cofactors. The specific cofactors recruited to PPAR-RXR complexes in response to different ligands lead to major differences in the transactivation of target genes. We developed a cofactor recruitment assay that is based on an europium-labeled anti-GST antibody and streptavidin-APC leading to a fluorescence resonance energy transfer signal. This assay allows for the determination of unique agonistic profiles in terms of potency and co-activator motif. Hence, it is a valuable drug discovery tool to support hit finding and lead optimization campaigns, enabling the characterization of next generation PPAR agonists.


Sujet(s)
Récepteur PPAR alpha , Récepteur PPAR gamma , Europium , Transfert d'énergie par résonance de fluorescence , Récepteur PPAR alpha/agonistes , Récepteur PPAR alpha/génétique , Récepteur PPAR alpha/métabolisme , Récepteur PPAR gamma/métabolisme , Récepteurs X des rétinoïdes , Streptavidine
18.
Front Physiol ; 13: 917510, 2022.
Article de Anglais | MEDLINE | ID: mdl-36091380

RÉSUMÉ

Human adipose-derived stem cells (hASCs) have multi-directional differentiation potential including osteogenic differentiation. Mechanical stimulation is thought to be a key regulator of bone remodeling and has been proved to promote osteogenic differentiation of mesenchymal stem cells. However, the mechanism how mechanical tension-induced osteogenesis of hASCs still remains poor understood. Polycystin-2 (PC2), a member of the transient receptor potential polycystic (TRPP) family, is involved in cilia-mediated mechanical transduction. To understand the role of PC2 in osteogenic differentiation under mechanical stimuli in hASCs, PKD2 gene was stably silenced by using lentivirus-mediated shRNA technology. The results showed that mechanical tension sufficiently enhanced osteogenic differentiation but hardly affected proliferation of hASCs. Silencing PKD2 gene caused hASCs to lose the ability of sensing mechanical stimuli and subsequently promoting osteogenesis. PC2 knock-out also reduced the cilia population frequency and cilia length in hASCs. TAZ (transcriptional coactivator with PDZ-binding motif, also known as Wwtr1) could mediate the genes regulation and biological functions of mechanotransduction signal pathway. Here, mechanical tension also enhanced TAZ nuclear translocation of hASCs. PC2 knock-out blocked tension-induced upregulation of nuclear TAZ and suppress tension-induced osteogenesis. TAZ could directly interact with Runx2, and inhibiting TAZ could suppress tension-induced upregulation of Runx2 expression. In summary, our findings demonstrated that PC2 mediate mechanical tension-induced osteogenic differentiation of hASCs by activating TAZ.

19.
Acta Pharm Sin B ; 12(4): 1856-1870, 2022 Apr.
Article de Anglais | MEDLINE | ID: mdl-35847510

RÉSUMÉ

In most acute promyelocytic leukemia (APL) cells, promyelocytic leukemia (PML) fuses to retinoic acid receptor α (RARα) due to chromosomal translocation, thus generating PML/RARα oncoprotein, which is a relatively stable oncoprotein for degradation in APL. Elucidating the mechanism regulating the stability of PML/RARα may help to degrade PML/RARα and eradicate APL cells. Here, we describe a deubiquitinase (DUB)-involved regulatory mechanism for the maintenance of PML/RARα stability and develop a novel pharmacological approach to degrading PML/RARα by inhibiting DUB. We utilized a DUB siRNA library to identify the ovarian tumor protease (OTU) family member deubiquitinase YOD1 as a critical DUB of PML/RARα. Suppression of YOD1 promoted the degradation of PML/RARα, thus inhibiting APL cells and prolonging the survival time of APL cell-bearing mice. Subsequent phenotypic screening of small molecules allowed us to identify ubiquitin isopeptidase inhibitor I (G5) as the first YOD1 pharmacological inhibitor. As expected, G5 notably degraded PML/RARα protein and eradicated APL, particularly drug-resistant APL cells. Importantly, G5 also showed a strong killing effect on primary patient-derived APL blasts. Overall, our study not only reveals the DUB-involved regulatory mechanism on PML/RARα stability and validates YOD1 as a potential therapeutic target for APL, but also identifies G5 as a YOD1 inhibitor and a promising candidate for APL, particularly drug-resistant APL treatment.

20.
Front Immunol ; 13: 889564, 2022.
Article de Anglais | MEDLINE | ID: mdl-35603192

RÉSUMÉ

The transcriptional co-activator BOB.1/OBF.1 is expressed in both B and T cells. The main characteristic of conventional BOB.1/OBF.1 deficient mice is the complete absence of germinal centers (GCs). This defect was mainly attributed to the defective B cell compartment. However, it is unknown whether and how BOB.1/OBF.1 expression in T cells contributes to the GC reaction. To finally clarify this question, we studied the in vivo function of BOB.1/OBF.1 in CD4+ T and follicular T helper (TFH) cell subpopulations by conditional mutagenesis, in the presence of immunocompetent B lymphocytes. BOB.1/OBF.1 deletion in CD4+ T as well as TFH cells resulted in impaired GC formation demonstrating that the impaired GC reaction described for conventional BOB.1/OBF.1-deficient mice cannot exclusively be traced back to the B cell compartment. Furthermore, we show a requirement of BOB.1/OBF.1 for T helper (TH) cell subsets, particularly for TFH cell differentiation.


Sujet(s)
Centre germinatif , Activation des lymphocytes , Animaux , Lymphocytes B/métabolisme , Différenciation cellulaire , Souris , Transactivateurs , Facteurs de transcription/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE