Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 136
Filtrer
1.
Nat Commun ; 15(1): 4985, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38862515

RÉSUMÉ

Hyperglycemia accelerates calcification of atherosclerotic plaques in diabetic patients, and the accumulation of advanced glycation end products (AGEs) is closely related to the atherosclerotic calcification. Here, we show that hyperglycemia-mediated AGEs markedly increase vascular smooth muscle cells (VSMCs) NF90/110 activation in male diabetic patients with atherosclerotic calcified samples. VSMC-specific NF90/110 knockout in male mice decreases obviously AGEs-induced atherosclerotic calcification, along with the inhibitions of VSMC phenotypic changes to osteoblast-like cells, apoptosis, and matrix vesicle release. Mechanistically, AGEs increase the activity of NF90, which then enhances ubiquitination and degradation of AGE receptor 1 (AGER1) by stabilizing the mRNA of E3 ubiquitin ligase FBXW7, thus causing the accumulation of more AGEs and atherosclerotic calcification. Collectively, our study demonstrates the effects of VSMC NF90 in mediating the metabolic imbalance of AGEs to accelerate diabetic atherosclerotic calcification. Therefore, inhibition of VSMC NF90 may be a potential therapeutic target for diabetic atherosclerotic calcification.


Sujet(s)
Athérosclérose , Protéine-7 contenant une boite F et des répétitions WD , Produits terminaux de glycation avancée , Souris knockout , Muscles lisses vasculaires , Myocytes du muscle lisse , Facteurs nucléaires-90 , Récepteur spécifique des produits finaux de glycosylation avancée , Animaux , Mâle , Souris , Produits terminaux de glycation avancée/métabolisme , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Athérosclérose/métabolisme , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Humains , Protéine-7 contenant une boite F et des répétitions WD/métabolisme , Protéine-7 contenant une boite F et des répétitions WD/génétique , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Facteurs nucléaires-90/métabolisme , Facteurs nucléaires-90/génétique , Récepteur spécifique des produits finaux de glycosylation avancée/métabolisme , Récepteur spécifique des produits finaux de glycosylation avancée/génétique , Calcification vasculaire/métabolisme , Calcification vasculaire/anatomopathologie , Calcification vasculaire/génétique , Souris de lignée C57BL , Ubiquitination , Diabète expérimental/métabolisme , Diabète expérimental/complications , Diabète expérimental/génétique , Diabète expérimental/anatomopathologie , Hyperglycémie/métabolisme , Hyperglycémie/génétique , Plaque d'athérosclérose/métabolisme , Plaque d'athérosclérose/anatomopathologie , Plaque d'athérosclérose/génétique , Apoptose
2.
J Mol Cell Cardiol ; 190: 62-75, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38583797

RÉSUMÉ

Intimal hyperplasia is a complicated pathophysiological phenomenon attributable to in-stent restenosis, and the underlying mechanism remains unclear. Interleukin enhancer-binding factor 3 (ILF3), a double-stranded RNA-binding protein involved in regulating mRNA stability, has been recently demonstrated to assume a crucial role in cardiovascular disease; nevertheless, its impact on intimal hyperplasia remains unknown. In current study, we used samples of human restenotic arteries and rodent models of intimal hyperplasia, we found that vascular smooth muscle cell (VSMC) ILF3 expression was markedly elevated in human restenotic arteries and murine ligated carotid arteries. SMC-specific ILF3 knockout mice significantly suppressed injury induced neointimal formation. In vitro, platelet-derived growth factor type BB (PDGF-BB) treatment elevated the level of VSMC ILF3 in a dose- and time-dependent manner. ILF3 silencing markedly inhibited PDGF-BB-induced phenotype switching, proliferation, and migration in VSMCs. Transcriptome sequencing and RNA immunoprecipitation sequencing depicted that ILF3 maintained its stability upon binding to the mRNA of the high-mobility group box 1 protein (HMGB1), thereby exerting an inhibitory effect on the transcription of dual specificity phosphatase 16 (DUSP16) through enhanced phosphorylation of signal transducer and activator of transcription 3 (STAT3). Therefore, the results both in vitro and in vivo indicated that the loss of ILF3 in VSMC ameliorated neointimal hyperplasia by regulating the STAT3/DUSP16 axis through the degradation of HMGB1 mRNA. Our findings revealed that vascular injury activates VSMC ILF3, which in turn promotes intima formation. Consequently, targeting specific VSMC ILF3 may present a potential therapeutic strategy for ameliorating cardiovascular restenosis.


Sujet(s)
Protéine HMGB1 , Hyperplasie , Souris knockout , Muscles lisses vasculaires , Myocytes du muscle lisse , Facteurs nucléaires-90 , Stabilité de l'ARN , Facteur de transcription STAT-3 , Tunique intime , Animaux , Humains , Mâle , Souris , Mouvement cellulaire , Prolifération cellulaire , Modèles animaux de maladie humaine , Régulation de l'expression des gènes , Protéine HMGB1/métabolisme , Protéine HMGB1/génétique , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/métabolisme , Néointima/métabolisme , Néointima/anatomopathologie , Facteurs nucléaires-90/métabolisme , Facteurs nucléaires-90/génétique , ARN messager/génétique , ARN messager/métabolisme , Transduction du signal , Facteur de transcription STAT-3/métabolisme , Tunique intime/métabolisme , Tunique intime/anatomopathologie
3.
Mol Med ; 30(1): 30, 2024 Feb 23.
Article de Anglais | MEDLINE | ID: mdl-38395749

RÉSUMÉ

BACKGROUND: Sepsis is a systemic inflammatory response which is frequently associated with acute lung injury (ALI). Activating transcription factor 3 (ATF3) promotes M2 polarization, however, the biological effects of ATF3 on macrophage polarization in sepsis remain undefined. METHODS: LPS-stimulated macrophages and a mouse model of cecal ligation and puncture (CLP)-induced sepsis were generated as in vitro and in vivo models, respectively. qRT-PCR and western blot were used to detect the expression of ATF3, ILF3, NEAT1 and other markers. The phenotypes of macrophages were monitored by flow cytometry, and cytokine secretion was measured by ELISA assay. The association between ILF3 and NEAT1 was validated by RIP and RNA pull-down assays. RNA stability assay was employed to assess NEAT1 stability. Bioinformatic analysis, luciferase reporter and ChIP assays were used to study the interaction between ATF3 and ILF3 promoter. Histological changes of lung tissues were assessed by H&E and IHC analysis. Apoptosis in lungs was monitored by TUNEL assay. RESULTS: ATF3 was downregulated, but ILF3 and NEAT1 were upregulated in PBMCs of septic patients, as well as in LPS-stimulated RAW264.7 cells. Overexpression of ATF3 or silencing of ILF3 promoted M2 polarization of RAW264.7 cells via regulating NEAT1. Mechanistically, ILF3 was required for the stabilization of NEAT1 through direct interaction, and ATF3 was a transcriptional repressor of ILF3. ATF3 facilitated M2 polarization in LPS-stimulated macrophages and CLP-induced septic lung injury via ILF3/NEAT1 axis. CONCLUSION: ATF3 triggers M2 macrophage polarization to protect against the inflammatory injury of sepsis through ILF3/NEAT1 axis.


Sujet(s)
Facteur de transcription ATF-3 , Macrophages , ARN long non codant , Sepsie , Animaux , Humains , Souris , Facteur de transcription ATF-3/génétique , Facteur de transcription ATF-3/métabolisme , Lipopolysaccharides , Macrophages/métabolisme , Facteurs nucléaires-90/génétique , Facteurs nucléaires-90/métabolisme , Cellules RAW 264.7 , Sepsie/métabolisme , ARN long non codant/génétique , ARN long non codant/métabolisme
4.
J Med Chem ; 66(24): 16843-16868, 2023 12 28.
Article de Anglais | MEDLINE | ID: mdl-38079530

RÉSUMÉ

Survivin is a novel attractive target for cancer therapy; however, it is considered undruggable because it lacks enzymatic activities. Herein, we describe our efforts toward the discovery of a novel series of 4,11-dioxo-4,11-dihydro-1H-anthra[2,3-d]imidazol-3-ium derivatives as survivin inhibitors by targeting ILF3/NF110. Intensive structural modifications led us to identify a lead compound AQIM-I, which remarkably inhibited nonsmall cell lung cancer cells A549 with an IC50 value of 9 nM and solid tumor cell proliferation with more than 700-fold selectivity against human normal cells. Further biological studies revealed that compound AQIM-I significantly inhibited survivin expression and colony formation and induced ROS production, apoptosis, cell cycle arrest, DNA damage, and autophagy. Furthermore, the promoter-luciferase reporter assay showed that AQIM-I attenuated the survivin promoter activity enhanced by the overexpression of ILF3/NF110 in a concentration-dependent manner, and specific binding (KD = 163 nM) of AQIM-I to ILF3/NF110 was detected by surface plasmon resonance.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Survivine/métabolisme , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Tumeurs du poumon/traitement médicamenteux , Apoptose , Protéines IAP , Lignée cellulaire tumorale , Prolifération cellulaire , Facteurs nucléaires-90/génétique , Facteurs nucléaires-90/métabolisme
5.
Biol Direct ; 18(1): 33, 2023 06 19.
Article de Anglais | MEDLINE | ID: mdl-37337223

RÉSUMÉ

BACKGROUND: Proteasome 26S subunit, non-ATPase 3 (PSMD3) has been reported to participate in various human cancers. Nevertheless, the function of PSMD3 in lung cancer (LC) remains unclear. METHODS: RT-qPCR and western blot were used to detect the expression of PSMD3 in LC tissues form TCGA database and clinical samples, and LC cell lines. To study the effect of PSMD3 on LC cell proliferation, migration, invasion, and apoptosis, siRNAs targeting PSMD3 were synthesized and overexpressed plasmids were constructed. CCK-8 assay, Transwell assay, and etc. were used to evaluate the results. Tumor xenograft model was used to evaluate the function of PSMD3 on tumor growth. CO-IP and MS were used to scan the proteins that bind with PSMD3. The interaction between PSMD3 and ILF3 in lung cancer cells were studied using IF staining, CHX protein stability, and ubiquitination assay. Additionally, the effect of ILF3 on cell progression and LC tumor growth was demonstrated by conducting a recovery assay using siILF3 and an ILF3 inhibitor YM155. RESULTS: We observed that PSMD3 was significantly overexpressed in LC tissues and cells, which indicated a poor prognosis. Meanwhile, we found that PSMD3 promoted cell proliferation, migration, and invasion of LC cells. We also determined that PSMD3 stabilized the protein expression of ILF3 and the deubiquitination of ILF3 in lung cancer cells. Furthermore, animal experiments showed that the ILF3 inhibitor YM155 could suppress tumor growth with the presence of PSMD3. CONCLUSIONS: PSMD3 collectively regulated the stability of ILF3 protein and facilitated the ubiquitination of endogenous ILF3 in LC, which ultimately promoted the progression of LC cells. The PSMD3/ ILF3 axis could potentially be used as a novel strategy for both diagnosis and treatment of LC.


Sujet(s)
Tumeurs du poumon , Facteurs nucléaires-90 , Proteasome endopeptidase complex , Animaux , Humains , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Tumeurs du poumon/génétique , Facteurs nucléaires-90/génétique , Facteurs nucléaires-90/métabolisme , Transduction du signal , Proteasome endopeptidase complex/génétique , Proteasome endopeptidase complex/métabolisme
6.
Kaohsiung J Med Sci ; 39(8): 811-823, 2023 Aug.
Article de Anglais | MEDLINE | ID: mdl-37132584

RÉSUMÉ

Skin is the first line of the body to resist pathogen invasion. A potentially fatal infection may result from problems with wound healing. Small molecule drugs like astragaloside IV (AS-IV) show pro-healing activities, but the mechanisms are not fully understood. Using real-time quantitative PCR and a western blot assay, the amount of gene expression was evaluated. The proliferation and migration of keratinocytes were determined by MTS and wound healing assay, respectively. The binding of lncRNA H19 to RBP protein ILF3 and the binding of ILF3 protein to CDK4 mRNA were confirmed by RNA immunoprecipitation. Treatment with AS-IV enhanced the expression of lncRNA H19, ILF3, and CDK4 and improved the proliferation and migration of keratinocytes HaCaT. Additionally, apoptosis of keratinocytes was attenuated by AS-IV. Further studies showed that both lncRNA H19 and ILF3 were important for AS-IV-mediated keratinocyte growth and migration. In addition, lncRNA H19 recruited ILF3 to increase CDK4 mRNA level and enhanced cell proliferation. We discovered a lncRNA H19/ILF3/CDK4 axis that is activated by AS-IV to promote keratinocyte migration and proliferation. These results elucidate the mechanism of action of AS-IV and justify its application in further application in wound healing treatment.


Sujet(s)
Kinase-4 cycline-dépendante , Kératinocytes , Facteurs nucléaires-90 , ARN long non codant , Prolifération cellulaire/génétique , Kératinocytes/métabolisme , ARN long non codant/génétique , ARN long non codant/métabolisme , ARN messager/génétique , Cellules HaCaT , Humains , Facteurs nucléaires-90/génétique , Facteurs nucléaires-90/métabolisme , Kinase-4 cycline-dépendante/génétique
7.
Nat Cell Biol ; 25(5): 754-764, 2023 05.
Article de Anglais | MEDLINE | ID: mdl-37037994

RÉSUMÉ

The mechanistic target of rapamycin complex 1 (mTORC1) is an essential hub that integrates nutrient signals and coordinates metabolism to control cell growth. Amino acid signals are detected by sensor proteins and relayed to the GATOR2 and GATOR1 complexes to control mTORC1 activity. Here we perform genome-wide CRISPR/Cas9 screens, coupled with an assay for mTORC1 activity based on fluorescence-activated cell sorting analysis of pS6, to identify potential regulators of mTORC1-dependent amino acid sensing. We then focus on interleukin enhancer binding factor 3 (ILF3), one of the candidate genes from the screen. ILF3 tethers the GATOR complexes to lysosomes to control mTORC1. Adding a lysosome-targeting sequence to the GATOR2 component WDR24 bypasses the requirement for ILF3 to modulate amino-acid-dependent mTORC1 signalling. ILF3 plays an evolutionarily conserved role in human and mouse cells, and in worms to regulate the mTORC1 pathway, control autophagy activity and modulate the ageing process.


Sujet(s)
Acides aminés , Sérine-thréonine kinases TOR , Animaux , Humains , Souris , Acides aminés/métabolisme , Lysosomes/métabolisme , Complexe-1 cible mécanistique de la rapamycine/génétique , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Complexes multiprotéiques/métabolisme , Facteurs nucléaires-90/génétique , Facteurs nucléaires-90/métabolisme , Transduction du signal , Sérine-thréonine kinases TOR/génétique , Sérine-thréonine kinases TOR/métabolisme
8.
J Biomed Sci ; 30(1): 20, 2023 Mar 23.
Article de Anglais | MEDLINE | ID: mdl-36959575

RÉSUMÉ

BACKGROUND: Although immune checkpoint blockade (ICB) therapy has brought survival benefits to patients with specific cancer types, most of cancer patients remain refractory to the ICB therapy, which is largely attributed to the immunosuppressive tumor microenvironment. Thereby, it is urgent to profile key molecules and signal pathways responsible for modification of tumor microenvironment. METHODS: Multiple databases of esophageal squamous cell carcinoma (ESCC) were integratively analyzed to screen candidate genes responsible for infiltration of CD8+ T cells. Expression of pescadillo ribosomal biogenesis factor 1 (PES1) in clinical ESCC samples was examined by qRT-PCR, western blotting, and immunohistochemistry. The mechanisms of PES1 were investigated via RNA sequencing and mass spectrometry followed by immunoprecipitation and proximity ligation assay. The clinical and therapeutic significance of PES1 in ESCC was comprehensively investigated using ESCC cells and mouse model. RESULTS: PES1 was significantly upregulated and correlated with poor prognosis in ESCC patients. PES1 knockdown decreased ESCC cell growth in vitro and in vivo and enhanced the efficacy of ICB therapy in mouse model, which was established through subcutaneous inoculation with ESCC cells. Analyses on RNA sequencing and mass spectrometry suggested that PES1 expression was negatively correlated with IL15 and ILF3 was one of the PES1-associated proteins. It has been known that ILF3 interacts with and stabilizes IL15 mRNA to increase IL15 protein level. Our data further indicated that PES1 interfered with the interaction between ILF3 and IL15 mRNA and impaired ILF3-mediated stabilization of IL15 mRNA, which eventually reduced the protein level of IL15. Interestingly, the inhibitory effect of ICB therapy boosted by PES1 knockdown dramatically antagonized by knockdown of IL15, which suppressed the tumor-infiltrated CD8+ T cells in ESCC. Finally, we confirmed the relationships among PES1, IL15, and CD8+ T cell infiltration in 10 locally advanced ESCC patients receiving ICB neoadjuvant therapy and demonstrated that ICB therapy would be more effective in those with low expression of PES1. CONCLUSIONS: Altogether, our findings herein provided novel insights on biological function and clinical significance of PES1 and suggested that high expression of PES1 could suppress ILF3-IL15 axis-mediated immunosurveillance and promote resistance to ICB through restraining tumor-infiltrated CD8+ T cells.


Sujet(s)
Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , Animaux , Souris , Lymphocytes T CD8+ , Tumeurs de l'oesophage/traitement médicamenteux , Tumeurs de l'oesophage/génétique , Carcinome épidermoïde de l'oesophage/génétique , Carcinome épidermoïde de l'oesophage/thérapie , Immunothérapie , Interleukine-15/pharmacologie , Interleukine-15/usage thérapeutique , Microenvironnement tumoral , Protéines de liaison à l'ARN/métabolisme , Facteurs nucléaires-90/métabolisme
9.
Cancer Gene Ther ; 30(7): 1007-1017, 2023 07.
Article de Anglais | MEDLINE | ID: mdl-36973424

RÉSUMÉ

Hepatocellular carcinoma (HCC) is a gastrointestinal tumor with high clinical incidence. Long non-coding RNAs (lncRNAs) play vital roles in modulating the growth and epithelial-mesenchymal transition (EMT) of HCC. However, the underlying mechanism of lncRNA KDM4A antisense RNA 1 (KDM4A-AS1) in HCC remains elusive. In our study, the role of KDM4A-AS1 in HCC was systematically investigated. The levels of KDM4A-AS1, interleukin enhancer-binding factor 3 (ILF3), Aurora kinase A (AURKA), and E2F transcription factor 1 (E2F1) were determined by RT-qPCR or western blot. ChIP and dual luciferase reporter experiments were performed to detect the binding relationship between E2F1 and KDM4A-AS1 promoter sequence. RIP and RNA-pull down confirmed the interaction of ILF3 with KDM4A-AS1/AURKA. Cellular functions were analyzed by MTT, flow cytometry, wound healing and transwell assays. IHC was performed to detect Ki67 in vivo. We found that KDM4A-AS1 was increased in HCC tissues and cells. Elevated KDM4A-AS1 level was correlated to poor prognosis of HCC. Knockdown of KDM4A-AS1 inhibited the proliferation, migration, invasion and EMT of HCC cells. ILF3 bound to KDM4A-AS1 and AURKA. KDM4A-AS1 maintained the stability of AURKA mRNA by recruiting ILF3. E2F1 transcriptionally activated KDM4A-AS1. Overexpressed KDM4A-AS1 reversed the contribution of E2F1 depletion to AURKA expression and EMT in HCC cells. KDM4A-AS1 promoted tumor formation in vivo through the PI3K/AKT pathway. These results revealed that E2F1 transcriptionally activated KDM4A-AS1 to regulate HCC progression via the PI3K/AKT pathway. E2F1 and KDM4A-AS1 may serve as good prognostic targets for HCC treatment.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , microARN , ARN long non codant , Humains , Carcinome hépatocellulaire/anatomopathologie , Tumeurs du foie/anatomopathologie , Aurora kinase A/génétique , Aurora kinase A/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Régulation positive , ARN messager , Transition épithélio-mésenchymateuse/génétique , Phosphatidylinositol 3-kinases/génétique , Prolifération cellulaire/génétique , microARN/génétique , Lignée cellulaire tumorale , ARN long non codant/métabolisme , Régulation de l'expression des gènes tumoraux , Facteurs nucléaires-90/génétique , Facteurs nucléaires-90/métabolisme
10.
Cell Death Dis ; 14(1): 59, 2023 01 25.
Article de Anglais | MEDLINE | ID: mdl-36697384

RÉSUMÉ

The regulatory role of circRNAs in cancer metastasis has become a focused issue in recent years. To date, however, the discovery of novel functional circRNAs and their regulatory mechanisms via binding with RBPs in bladder cancer (BC) are still lacking. Here, we screened out circSLC38A1 based on our sequencing data and followed validation with clinical tissue samples and cell lines. Functional assays showed that circSLC38A1 promoted BC cell invasion in vitro and lung metastasis of mice in vivo. By conducting RNA pull-down, mass spectrum, and RIP assays, circSLC38A1 was found to interact with Interleukin enhancer-binding factor 3 (ILF3), and stabilize ILF3 protein via modulating the ubiquitination process. By integrating our CUT&Tag-seq and RNA-seq data, TGF-ß2 was identified as the functional target of the circSLC38A1-ILF3 complex. In addition, m6A methylation was enriched in circSLC38A1 and contributed to its upregulation. Clinically, circSLC38A1 was identified in serum exosomes of BC patients and could distinguish BC patients from healthy individuals with a diagnostic accuracy of 0.878. Thus, our study revealed an essential role and clinical significance of circSLC38A1 in BC via activating the transcription of TGF-ß2 in an ILF3-dependent manner, extending the understanding of the importance of circRNA-mediated transcriptional regulation in BC metastasis.


Sujet(s)
Facteurs nucléaires-90 , ARN circulaire , Facteur de croissance transformant bêta-2 , Tumeurs de la vessie urinaire , Animaux , Souris , Lignée cellulaire tumorale , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Facteurs nucléaires-90/génétique , Facteurs nucléaires-90/métabolisme , Oncogènes , ARN circulaire/génétique , Facteur de croissance transformant bêta-2/métabolisme , Tumeurs de la vessie urinaire/anatomopathologie , Humains
11.
Int J Mol Sci ; 23(21)2022 Nov 05.
Article de Anglais | MEDLINE | ID: mdl-36362366

RÉSUMÉ

Double-stranded RNA-binding proteins (dsRBPs) are major players in the regulation of gene expression patterns. Among them, Nuclear Factor 90 (NF90) has a plethora of well-known functions in viral infection, transcription, and translation as well as RNA stability and degradation. In addition, NF90 has been identified as a regulator of microRNA (miRNA) maturation by competing with Microprocessor for the binding of pri-miRNAs in the nucleus. NF90 was recently shown to control the biogenesis of a subset of human miRNAs, which ultimately influences, not only the abundance, but also the expression of the host gene and the fate of the mRNA target repertoire. Moreover, recent evidence suggests that NF90 is also involved in RNA-Induced Silencing Complex (RISC)-mediated silencing by binding to target mRNAs and controlling their translation and degradation. Here, we review the many, and growing, functions of NF90 in RNA biology, with a focus on the miRNA pathway and RISC-mediated gene silencing.


Sujet(s)
microARN , Facteurs nucléaires-90 , Humains , Facteurs nucléaires-90/génétique , Facteurs nucléaires-90/métabolisme , microARN/métabolisme , ARN messager/génétique , ARN messager/métabolisme , Stabilité de l'ARN , Biologie
12.
Am J Physiol Regul Integr Comp Physiol ; 323(6): R861-R874, 2022 12 01.
Article de Anglais | MEDLINE | ID: mdl-36222883

RÉSUMÉ

Hypertension is characterized by sympathetic hyperactivity, which is related to the overexcitation of the presympathetic neurons in the rostral ventrolateral medulla (RVLM). Nitric oxide (NO) has been reported to be a vital neuromodulator involved in central cardiovascular regulation. However, the mechanism of interleukin-enhanced binding factor 3 (ILF3) participating in blood pressure (BP) regulation is still unclear. Therefore, this study aims to clarify the role of ILF3 within the rostral ventrolateral medulla (RVLM) in regulating NO in hypertension. It was found that the expression level of ILF3 was significantly increased in the RVLM of spontaneously hypertensive rats (SHR) compared with Wistar-Kyoto (WKY) rats through microarray gene expression analysis, Western blot, and immunofluorescence. Overexpression of ILF3 by injecting constructed adenovirus into the RVLM increased the BP and renal sympathetic nerve activity (RSNA) of the WKY rats, significantly decreasing NO production and neuronal nitric oxide synthase (nNOS) expression. Knockdown of ILF3 in the RVLM of SHR significantly reduced BP but increased NO production and the neuronal nitric oxide synthase (nNOS) expression. Furthermore, it was found that the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway was activated via Western blotting in the RVLM after overexpression of ILF3, whereas it was attenuated after knockdown of ILF3 in SHR. In addition, inhibition of PI3K by intracisternal infusion of the PI-103 attenuated the increase in Akt phosphorylation and decrease in nNOS expression and NO production caused by overexpressing ILF3, which ultimately blunted high BP induced by overexpressing ILF3. Taken together, this current study suggests that ILF3 participates in high BP via reducing NO production in the RVLM through PI3K/Akt pathway.


Sujet(s)
Hypertension artérielle , Protéines proto-oncogènes c-akt , Rats , Animaux , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Monoxyde d'azote/métabolisme , Nitric oxide synthase type I/génétique , Nitric oxide synthase type I/métabolisme , Rats de lignée WKY , Phosphatidylinositol 3-kinase/métabolisme , Moelle allongée/métabolisme , Pression sanguine , Rats de lignée SHR , Interleukines/métabolisme , Facteurs nucléaires-90/métabolisme
13.
Chem Biol Interact ; 368: 110208, 2022 Dec 01.
Article de Anglais | MEDLINE | ID: mdl-36208777

RÉSUMÉ

The regulatory network between arsenic, genes and signaling pathways has been reported in arsenic carcinogenesis. Studies on circRNA represent a growing field, but the extent to circRNA potential mechanisms remains poorly understood. So this study we explore the systematic function of hsa_circ_0005050 in mediating the cell apoptosis and proliferation. We demonstrated that hsa_circ_0005050 was highly expressed in subjects who are long-term exposed to arsenic, and could be induced by NaAs2O3 in A549 and 16HBE. Knockdown of hsa_circ_0005050 promotes A549 cell viability, whereas exerts the opposite effects in 16HBE. Mechanistically, hsa_circ_0005050 regulates the p53 and NF-κB signaling pathway involved in the apoptosis and proliferation. And we found that hsa_circ_0005050 could directly bind to the RNA binding protein ILF3 and mutually influence each other's formation. Upon si-hsa_circ_0005050, ILF3 export to the cytoplasm resulting the formation of a ternary complex ILF3-p65-IκBA, breaks the balance of p53 and NF-κB pathway and induces A549 apoptosis and leads to 16HBE proliferation. As a result of these investigations, suggestions were identified for future research.


Sujet(s)
Arsenic , Humains , Apoptose/génétique , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Régulation de l'expression des gènes tumoraux , Facteur de transcription NF-kappa B/métabolisme , Facteurs nucléaires-90/génétique , Facteurs nucléaires-90/métabolisme , ARN circulaire/génétique , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Carcinogenèse/induit chimiquement , Intoxication par l'arsenic
14.
BMC Biol ; 20(1): 194, 2022 09 01.
Article de Anglais | MEDLINE | ID: mdl-36050755

RÉSUMÉ

BACKGROUND: Nuclear factor 90 (NF90) is a double-stranded RNA-binding protein involved in a multitude of different cellular mechanisms such as transcription, translation, viral infection, and mRNA stability. Recent data suggest that NF90 might influence the abundance of target mRNAs in the cytoplasm through miRNA- and Argonaute 2 (Ago2)-dependent activity. RESULTS: Here, we identified the interactome of NF90 in the cytoplasm, which revealed several components of the RNA-induced silencing complex (RISC) and associated factors. Co-immunoprecipitation analysis confirmed the interaction of NF90 with the RISC-associated RNA helicase, Moloney leukemia virus 10 (MOV10), and other proteins involved in RISC-mediated silencing, including Ago2. Furthermore, NF90 association with MOV10 and Ago2 was found to be RNA-dependent. Glycerol gradient sedimentation of NF90 immune complexes indicates that these proteins occur in the same protein complex. At target RNAs predicted to bind both NF90 and MOV10 in their 3' UTRs, NF90 association was increased upon loss of MOV10 and vice versa. Interestingly, loss of NF90 led to an increase in association of Ago2 as well as a decrease in the abundance of the target mRNA. Similarly, during hypoxia, the binding of Ago2 to vascular endothelial growth factor (VEGF) mRNA increased after loss of NF90, while the level of VEGF mRNA decreased. CONCLUSIONS: These findings reveal that, in the cytoplasm, NF90 can associate with components of RISC such as Ago2 and MOV10. In addition, the data indicate that NF90 and MOV10 may compete for the binding of common target mRNAs, suggesting a role for NF90 in the regulation of RISC-mediated silencing by stabilizing target mRNAs, such as VEGF, during cancer-induced hypoxia.


Sujet(s)
Protéines Argonaute/métabolisme , Facteurs nucléaires-90/métabolisme , ARN messager/métabolisme , Complexe réprimant l'expression de l'ARN , Régions 3' non traduites , Protéines Argonaute/génétique , Humains , Hypoxie/génétique , microARN/métabolisme , Facteurs nucléaires-90/génétique , RNA helicases/génétique , RNA helicases/métabolisme , Complexe réprimant l'expression de l'ARN/génétique , Complexe réprimant l'expression de l'ARN/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Facteur de croissance endothéliale vasculaire de type A/métabolisme
15.
Biol Chem ; 403(10): 907-915, 2022 09 27.
Article de Anglais | MEDLINE | ID: mdl-36040368

RÉSUMÉ

Protein-arginine methylation is a common posttranslational modification, crucial to various cellular processes, such as protein-protein interactions or binding to nucleic acids. The central enzyme of symmetric protein arginine methylation in mammals is the protein arginine methyltransferase 5 (PRMT5). While the methylation reaction itself is well understood, recruitment and differentiation among substrates remain less clear. One mechanism to regulate the diversity of PRMT5 substrate recognition is the mutual binding to the adaptor proteins pICln or RioK1. Here, we describe the specific interaction of Nuclear Factor 90 (NF90) with the PRMT5-WD45-RioK1 complex. We show for the first time that NF90 is symmetrically dimethylated by PRMT5 within the RG-rich region in its C-terminus. Since upregulation of PRMT5 is a hallmark of many cancer cells, the characterization of its dimethylation and modulation by specific commercial inhibitors in vivo presented here may contribute to a better understanding of PRMT5 function and its role in cancer.


Sujet(s)
Facteurs nucléaires-90 , Protein-arginine N-methyltransferases , Animaux , Arginine/métabolisme , Mammifères/métabolisme , Méthylation , Facteurs nucléaires-90/génétique , Facteurs nucléaires-90/métabolisme , Maturation post-traductionnelle des protéines , Protein-arginine N-methyltransferases/génétique , Protein-arginine N-methyltransferases/métabolisme
16.
Aging (Albany NY) ; 14(9): 3887-3909, 2022 05 04.
Article de Anglais | MEDLINE | ID: mdl-35507914

RÉSUMÉ

BACKGROUND: This study aimed to investigate the relationship of dyslipidemia and interleukin-enhancer binding factor 3 (ILF3) in gastric cancer, and provide insights into the potential application of statins as an agent to prevent and treat gastric cancer. METHODS: The expression levels of ILF3 in gastric cancer were examined with publicly available datasets such as TCGA, and western blotting and immunohistochemistry were performed to determine the expression of ILF3 in clinical specimens. The effects of ox-LDL on expression of ILF3 were further verified with western blot analyses. RNA sequencing, Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Ontology (GO), and Gene Set Enrichment Analysis (GSEA) pathway analyses were performed to reveal the potential downstream signaling pathway targets of ILF3. The effects of statins and ILF3 on PI3K/AKT/mTOR signaling pathway, cell proliferation, cell cycle, migration and invasion of gastric cancer cells were investigated with Edu assay, flow cytometry and transwell assay. RESULTS: Immunohistochemistry and western blot demonstrated that the positive expression rates of ILF3 in gastric cancer tissues were higher than adjacent mucosa tissues. The ox-LDL promoted the expression of ILF3 in a time-concentration-dependent manner. ILF3 promoted the proliferation, cell cycle, migration and invasion by activating the PI3K/AKT/mTOR signaling pathway. Statins inhibited the proliferation, cell cycle, migration and invasion of gastric cancer by inhibiting the expression of ILF3. CONCLUSIONS: These findings demonstrate that ox-LDL promotes ILF3 overexpression to regulate gastric cancer progression by activating the PI3K/AKT/mTOR signaling pathway. Statins inhibits the expression of ILF3, which might be a new targeted therapy for gastric cancer.


Sujet(s)
Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase , Tumeurs de l'estomac , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire , Humains , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/pharmacologie , Lipoprotéines LDL , Facteurs nucléaires-90/génétique , Facteurs nucléaires-90/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/métabolisme , Sérine-thréonine kinases TOR/métabolisme
17.
Sci Rep ; 12(1): 8837, 2022 05 25.
Article de Anglais | MEDLINE | ID: mdl-35614067

RÉSUMÉ

The Nuclear Factor 90 (NF90)-NF45 complex has been known to regulate the progression of transcription, mRNA stability, translational inhibition, RNA export and microRNA biogenesis. However, the physiological functions of the NF90-NF45 complex remain unclear. We newly discovered that the NF90-NF45 complex was expressed in primary ß cells and established cell lines. Therefore, in this study, we focused on the function of the endogenous NF90-NF45 complex in the ß cells. To investigate this issue, we generated ß-cell-specific NF90-NF45 deficient mice. These mice exhibited hyperglycaemia and lower plasma insulin levels under a high fat diet together with decreased islet mass. To uncover this mechanism, we performed a whole-genome expression microarray of the total RNA prepared from ß cell lines treated with siRNAs targeting both NF90 and NF45. In this result, we found an activation of p53 signaling in the NF90-NF45-knockdown cells. This activation was supported by elevation of luciferase activity derived from a reporter plasmid harboring p53 binding sites in the NF90-NF45-knockdown cells. Furthermore, the knockdown of NF90-NF45 resulted in a significant retardation of the ß cell line growth rates. Importantly, a dominant negative form of p53 rescues the growth retardation in BTC6 cells depleted of NF90-NF45, suggesting that NF90-NF45 would be positively involved in ß cell proliferation through suppression of p53 signal pathway. Taken together, NF90-NF45 is essential for ß cell compensation under obesity-inducing metabolic stress via repression of p53 signaling.


Sujet(s)
Facteur nucléaire-45 , Facteurs nucléaires-90 , Protéine p53 suppresseur de tumeur , Animaux , Souris , Facteur nucléaire-45/métabolisme , Facteurs nucléaires-90/métabolisme , Obésité/génétique , ARN , Transduction du signal , Stress physiologique , Protéine p53 suppresseur de tumeur/métabolisme
18.
J Mol Biol ; 434(7): 167469, 2022 04 15.
Article de Anglais | MEDLINE | ID: mdl-35120969

RÉSUMÉ

MicroRNAs (miRNAs) play important roles in regulated gene expression and miRNA biogenesis is also subject to regulation, together constituting critical regulatory circuitries in numerous physiological and pathological processes. As a dsRNA binding protein, interleukin enhancer binding factor 3 (ILF3) has been implicated as a negative regulator in miRNA biogenesis, but the mechanism and specificity have remained undefined. Here, combining small-RNA-seq and CLIP-seq, we showed that ILF3 directly represses many miRNAs or perhaps other types of small RNAs annotated in both miRBase and MirGeneDB. We demonstrated that ILF3 preferentially binds to A/U-enriched motifs, which tend to lengthen and/or stabilize the stem-loop in pri-miRNAs, thereby effectively competing with the Microprocessor to block miRNA biogenesis. Focusing on the biological function of ILF3-suppressed miR-582-3p, we discovered that this LINE-derived miRNA targets a critical interferon-inducible gene RIG-I for repression, thus establishing a novel ILF3/miR-582/RIG-I axis in the antiviral response.


Sujet(s)
Protéine-58 à domaine DEAD , Interféron de type I , microARN , Facteurs nucléaires-90 , Récepteurs immunologiques , Protéine-58 à domaine DEAD/génétique , Régulation de l'expression des gènes , Cellules HeLa , Humains , Interféron de type I/métabolisme , microARN/génétique , microARN/métabolisme , Facteurs nucléaires-90/métabolisme , Récepteurs immunologiques/génétique
19.
Sci Rep ; 12(1): 364, 2022 01 10.
Article de Anglais | MEDLINE | ID: mdl-35013429

RÉSUMÉ

RNA-binding proteins (RBPs) interact with and determine the fate of many cellular RNAs directing numerous essential roles in cellular physiology. Nuclear Factor 90 (NF90) is an RBP encoded by the interleukin enhancer-binding factor 3 (ILF3) gene that has been found to influence RNA metabolism at several levels, including pre-RNA splicing, mRNA turnover, and translation. To systematically identify the RNAs that interact with NF90, we carried out iCLIP (individual-nucleotide resolution UV crosslinking and immunoprecipitation) analysis in the human embryonic fibroblast cell line HEK-293. Interestingly, many of the identified RNAs encoded proteins involved in the response to viral infection and RNA metabolism. We validated a subset of targets and investigated the impact of NF90 on their expression levels. Two of the top targets, IRF3 and IRF9 mRNAs, encode the proteins IRF3 and IRF9, crucial regulators of the interferon pathway involved in the SARS-CoV-2 immune response. Our results support a role for NF90 in modulating key genes implicated in the immune response and offer insight into the immunological response to the SARS-CoV-2 infection.


Sujet(s)
COVID-19/métabolisme , Immunoprécipitation/méthodes , Facteurs nucléaires-90/métabolisme , Protéines de liaison à l'ARN/métabolisme , ARN/métabolisme , SARS-CoV-2/métabolisme , COVID-19/virologie , Cellules cultivées , Cellules HEK293 , Humains , Facteur-3 de régulation d'interféron/génétique , Facteur-3 de régulation d'interféron/métabolisme , Sous-unité gamma du complexe ISGF3/génétique , Sous-unité gamma du complexe ISGF3/métabolisme , Facteurs nucléaires-90/génétique , Liaison aux protéines , ARN/génétique , Interférence par ARN , Stabilité de l'ARN , ARN messager/génétique , ARN messager/métabolisme , Protéines de liaison à l'ARN/génétique , RNA-Seq/méthodes , SARS-CoV-2/génétique , SARS-CoV-2/physiologie
20.
Hum Cell ; 34(6): 1843-1854, 2021 Nov.
Article de Anglais | MEDLINE | ID: mdl-34491544

RÉSUMÉ

Hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide. Increasing evidences have demonstrated that ILF3 antisense RNA 1 (ILF3-AS1) acts as an oncogenic long noncoding RNA (lncRNA) in several types of human cancers. However, the expression pattern, functional role and underlying mechanism of ILF3-AS1 in HCC remains largely unclear. Here, we found that ILF3-AS1 expression was significantly elevated in HCC tissues and also associated with prognosis of patients with HCC. Functional assays demonstrated that knockdown of ILF3-AS1 expression resulted in the suppression of proliferation, migration and invasion in HCC cells, whereas overexpression of ILF3-AS1 exerted opposite effects. Additionally, knockdown of IFL3-AS1 attenuated HCC tumorigenesis and metastasis in vivo. Mechanistically, ILF3-AS1 associated with ILF3 mRNA and inhibited its degradation. ILF3-AS1 increased ILF3 m6A level via recruiting N6-methyladenosine (m6A) RNA methyltransferase METTL3. Moreover, IFL3-AS1 enhanced the interaction between ILF3 mRNA and m6A reader IGF2BP1. Overall, our study revealed the function and mechanism of ILF3-AS1 in the malignant phenotypes of HCC cells, which provides a novel therapeutic target for HCC.


Sujet(s)
Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Régulation de l'expression des gènes tumoraux/génétique , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Methyltransferases/métabolisme , Facteurs nucléaires-90/génétique , Facteurs nucléaires-90/physiologie , ARN long non codant/génétique , ARN long non codant/physiologie , ARN messager/génétique , ARN messager/métabolisme , Carcinome hépatocellulaire/thérapie , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Évolution de la maladie , Expression des gènes/génétique , Humains , Tumeurs du foie/thérapie , Thérapie moléculaire ciblée , Invasion tumorale/génétique , Facteurs nucléaires-90/métabolisme , ARN long non codant/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...