Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 7 de 7
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Folia Histochem Cytobiol ; 62(1): 25-36, 2024.
Article de Anglais | MEDLINE | ID: mdl-38563050

RÉSUMÉ

INTRODUCTION: Endometriosis (EMs), manifested by pain and infertility, is a chronic inflammatory disease. The precise pathophysiology of this disease remains uncertain. Insulin-like growth factor-2 mRNA-binding protein 1 (IGF2BP1) and polypyrimidine tract-binding protein 1 (PTBP1) have both been found to regulate proliferation, apoptosis, and invasion. This study aimed to investigate the effects of IGF2BP1/PTBP1 in treating EMs. MATERIALS AND METHODS: qRT-PCR and western blotting were employed to quantify IGF2BP1 and PTBP1 expression in six patients with EMs (mean age 33.83 years). The correlation analysis, STRING database prediction, and RNA immunoprecipitation were utilized to identify the relationship between IGF2BP1 and PTBP1. Ectopic endometrial volume, weight, HE staining, and IGF2BP1 silencing were utilized to estimate the effects of IGF2BP1 in EMs model rats. qRT-PCR, CCK-8, 5-ethynyl-2'-deoxyuridine (EDU) labeling, Transwell assay, and flow cytometry were utilized to assess the effects of IGF2BP1/PTBP1 on the proliferation, migration, invasion, and apoptosis of ectopic endometrial stromal cells (eESCs). Furthermore, western blotting was employed to evaluate expressions of PCNA, VEGF, and E-cadherin in EMs rats and eESCs. RESULTS: The mRNA and protein levels of IGF2BP1 and PTBP1 in the ectopic and eutopic endometrium of EMs patients were significantly increased. RNA immunoprecipitation revealed a close interaction of IGF2BP1 with PTBP1. Additionally, the endometrial volume, weight, and histopathologic scores in rats were significantly reduced after IGF2BP1 silencing. IGF2BP1 silencing also decreased the expression of PCNA and VEGF, and increased E-cadherin expression in endometrial tissues of EMs rats. Moreover, IGF2BP1 silencing inhibited proliferation, migration, and invasion and promoted apoptosis through PTBP1 in eESCs. CONCLUSIONS: IGF2BP1 exhibits potential beneficial properties in the management of EMs by interacting with PTBP1, thereby highlighting IGF2BP1 as a promising therapeutic target for EMs.


Sujet(s)
Endométriose , Adulte , Animaux , Femelle , Humains , Rats , Cadhérines/métabolisme , Prolifération cellulaire , Endométriose/anatomopathologie , Endomètre/anatomopathologie , Ribonucléoprotéines nucléaires hétérogènes/génétique , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Protéine PTB/génétique , Protéine PTB/métabolisme , Protéine PTB/pharmacologie , Antigène nucléaire de prolifération cellulaire/métabolisme , ARN messager/métabolisme , Facteur de croissance endothéliale vasculaire de type A/métabolisme
2.
Chem Biol Drug Des ; 103(1): e14380, 2024 01.
Article de Anglais | MEDLINE | ID: mdl-37890873

RÉSUMÉ

Labeled with pluripotent potential, the transplantation of bone marrow mesenchymal stem cells (BMSCs) is considered as a promising strategy for treating osteoporosis (OP). Melatonin (MEL) has been investigated to be an essential regulator involved in bone metabolism, as well as BMSCs differentiation. Circular RNAs (circRNAs) are a unique kind of non-coding RNA and play an important regulatory role in OP. However, whether circRNAs are implicated in the effects of MEL on BMSCs osteogenic differentiation remains largely indeterminate. Expression of circ_0005753 in human BMSCs with MEL treatment, clinical specimens diagnosed with OP, either with ovariectomy (OVX)-induced mice, was measured by RT-qPCR. Western blot was conducted to analyze protein levels of osteogenesis-related molecules (Opg, RUNX2, ALP, BMP4) and TXNIP. RNA immunoprecipitation (RIP) and RNA pull-down assays were performed to validate the binding relationship among circ_0005753, PTBP1, and TXNIP. Alkaline phosphatase (ALP) and alizarin red staining (ARS) were performed to evaluate osteogenic capacity of BMSCs. OP mouse model was established by ovariectomy, as evaluated pathologic changes via hematoxylin-eosin (HE), Masson, and Immunohistochemistry (IHC) staining. Expression of circ_0005753 was remarkably decreased during MEL-induced osteogenic differentiation of BMSCs. Interestingly, not only circ_0005753 knockdown significantly promoted osteogenic differentiation of BMSCs, but circ_0005753 overexpression also weakened osteogenic differentiation induced by MEL treatment. Mechanistically, circ_0005753 maintained the stabilization of TXNIP mRNA via recruiting PTBP1. Additionally, reinforced circ_0005753 abrogated MEL-mediated protective effects on OP pathogenesis in a mouse model. This work shows that MEL facilitates osteogenic differentiation of BMSCs via the circ_0005753/PTBP1/TXNIP axis, which may shed light on the development of a novel therapeutic strategy to prevent OP.


Sujet(s)
Mélatonine , Cellules souches mésenchymateuses , microARN , Ostéoporose , Femelle , Souris , Humains , Animaux , Ostéogenèse , Mélatonine/pharmacologie , ARN circulaire/génétique , ARN circulaire/analyse , ARN circulaire/métabolisme , Cellules cultivées , Ostéoporose/traitement médicamenteux , Ostéoporose/génétique , Différenciation cellulaire , Cellules souches mésenchymateuses/métabolisme , Modèles animaux de maladie humaine , microARN/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/analyse , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Protéine PTB/analyse , Protéine PTB/métabolisme , Protéine PTB/pharmacologie , Protéines de transport/métabolisme
3.
Exp Mol Med ; 55(12): 2596-2607, 2023 12.
Article de Anglais | MEDLINE | ID: mdl-38036735

RÉSUMÉ

Exposure to nanomicroplastics (nano-MPs) can induce lung damage. The gut microbiota is a critical modulator of the gut-lung axis. However, the mechanisms underlying these interactions have not been elucidated. This study explored the role of lactate, a key metabolite of the microbiota, in the development of lung damage induced by nano-MPs (LDMP). After 28 days of exposure to nano-MPs (50-100 nm), mice mainly exhibited damage to the lungs and intestinal mucosa and dysbiosis of the gut microbiota. Lactate accumulation was observed in the lungs, intestines and serum and was strongly associated with the imbalance in lactic acid bacteria in the gut. Furthermore, no lactate accumulation was observed in germ-free mice, while the depletion of the gut microbiota using a cocktail of antibiotics produced similar results, suggesting that lactate accumulation in the lungs may have been due to changes in the gut microbiota components. Mechanistically, elevated lactate triggers activation of the HIF1a/PTBP1 pathway, exacerbating nano-MP-induced lung damage through modulation of the epithelial-mesenchymal transition (EMT). Conversely, mice with conditional knockout of Ptbp1 in the lungs (Ptbp1flfl) and PTBP1-knockout (PTBP1-KO) human bronchial epithelial (HBE) cells showed reversal of the effects of lactate through modulation of the HIF1a/PTBP1 signaling pathway. These findings indicate that lactate is a potential target for preventing and treating LDMP.


Sujet(s)
Microbiome gastro-intestinal , Microbiote , Humains , Animaux , Souris , Acide lactique/métabolisme , Muqueuse intestinale/métabolisme , Poumon , Souris de lignée C57BL , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Protéine PTB/métabolisme , Protéine PTB/pharmacologie
4.
ESC Heart Fail ; 10(3): 1677-1688, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-36788730

RÉSUMÉ

AIMS: The activation of cardiac fibroblasts (CFs) leads to overproduction of collagens and subsequently cardiac fibrosis. However, the regulatory mechanism of CF function in the process of cardiac fibrosis remains unclear. This work investigated the function of polypyrimidine tract binding protein 1 (PTBP1)/nuclear receptor NR4A1 (Nur77)/fatty acid-binding protein 5 (FABP5) axis in myocardial fibrosis. METHODS AND RESULTS: Cardiac fibrosis was induced in mice suffered left anterior descending ligation. In parallel, neonatal mouse CFs were isolated and stimulated with transforming growth factor-ß1 (TGF-ß1). Cardiac fibrosis was evaluated by Masson's trichrome staining. Expression of PTBP1, Nur77, FABP5, collagen I, and collagen III was measured by quantitative real-time PCR and western blotting. Proliferation of CFs was assessed by 5-ethynyl-2'-deoxyuridine assay. Molecular interaction was validated by RNA-binding protein immunoprecipitation, chromatin immunoprecipitation, and dual luciferase reporter assay. PTBP1 was up-regulated (P < 0.05), whereas Nur77 (P < 0.05) and FABP5 (P < 0.05) were down-regulated in the fibrotic hearts of mice and TGF-ß1-exposed CFs. PTBP1 overexpression facilitated proliferation (P < 0.05) and collagen I (P < 0.05) and collagen III (P < 0.05) expression of CFs after stimulation with TGF-ß1. PTBP1 reduced Nur77 stability (P < 0.05) to inhibit Nur77 expression (P < 0.05) in CFs. Nur77 bound to FABP5 promoter to promote the transcription (P < 0.05) and expression (P < 0.05) of FABP5. Silencing of Nur77 or FABP5 abolished the inhibitory effect of PTBP1 knockdown on proliferation (P < 0.05) and collagen I (P < 0.05) and collagen III (P < 0.05) expression of CFs in vitro. PTBP1 depletion ameliorated cardiac fibrosis (P < 0.05), α-smooth muscle actin (P < 0.05), and collagen I (P < 0.05) expression in myocardial infarction mice through regulating Nur77/FABP5 pathway (P < 0.05) in vivo. CONCLUSIONS: PTBP1 contributed to cardiac fibrosis via promoting CF proliferation and collagen deposition through Nur77 mRNA decay and subsequent transcription inhibition of FABP5. Our findings suggest that PTBP1/Nur77/FABP5 axis may be potential targets for cardiac fibrosis therapy.


Sujet(s)
Myocarde , Facteur de croissance transformant bêta-1 , Animaux , Souris , Collagène/métabolisme , Collagène de type I , Protéines de liaison aux acides gras/génétique , Fibrose , Myocarde/anatomopathologie , Protéine PTB/génétique , Protéine PTB/métabolisme , Protéine PTB/pharmacologie , Facteur de croissance transformant bêta-1/pharmacologie , Facteur de croissance transformant bêta-1/métabolisme
5.
Leuk Res ; 123: 106967, 2022 12.
Article de Anglais | MEDLINE | ID: mdl-36270092

RÉSUMÉ

BACKGROUND: Bortezomib resistance hampers the long-term survival of multiple myeloma (MM) patients. Our previous study has proved that downregulated lncRNA MEG3 is associated with the poor clinical outcome in MM. However, the effect of MEG3 on the sensitivity of bortezomib in MM and its possible molecular mechanism remains muddled. METHODS: In this study, CCK8 and flow cytometry techniques were used to assess cell viability in MEG3 overexpressed MM cells after bortezomib treatment. The expression of autophagy-related protein LC3 and p62 were distinguished by Western blot, and the mCherry-GFP-LC3 puncta reflecting autophagy level was observed under fluorescence microscope. RNA immunoprecipitation (RIP) technology was used to detect the binding relationship of MEG3 and ATG2B to PTBP1. RESULTS: Increased toxicity of bortezomib and diminished autophagy level were found in MEG3 overexpressed MM cells. Mechanistically, we discovered that RNA-binding protein PTBP1 could bind to MEG3 and ATG2B by RIP assay. Upregulation of MEG3 promoted PTBP1 expression and inhibited the expression level of ATG2B, suggesting that MEG3 recruited PTBP1 and then decayed ATG2B expression. CONCLUSION: In summary, our study illustrated that MEG3 increased bortezomib sensitivity by hindering autophagy through the PTBP1/ATG2B axis, providing a new therapeutic target for bortezomib-resistant MM patients.


Sujet(s)
Autophagie , Bortézomib , microARN , Myélome multiple , ARN long non codant , Humains , Apoptose , Protéines associées à l'autophagie/génétique , Protéines associées à l'autophagie/métabolisme , Protéines associées à l'autophagie/pharmacologie , Bortézomib/pharmacologie , Ribonucléoprotéines nucléaires hétérogènes , microARN/génétique , Myélome multiple/traitement médicamenteux , Myélome multiple/génétique , Protéine PTB/génétique , Protéine PTB/pharmacologie , ARN long non codant/génétique , Protéines du transport vésiculaire/métabolisme , Protéines du transport vésiculaire/pharmacologie
6.
Mol Cell ; 19(4): 475-84, 2005 Aug 19.
Article de Anglais | MEDLINE | ID: mdl-16109372

RÉSUMÉ

Fas exon 6 can be included or skipped to generate mRNAs encoding, respectively, a membrane bound form of the receptor that promotes apoptosis or a soluble isoform that prevents programmed cell death. We report that the apoptosis-inducing protein TIA-1 promotes U1 snRNP binding to the 5' splice site of intron 6, which in turn facilitates exon definition by enhancing U2AF binding to the 3' splice site of intron 5. The polypyrimidine tract binding protein (PTB) promotes exon skipping by binding to an exonic splicing silencer and inhibiting the association of U2AF and U2 snRNP with the upstream 3' splice site, without affecting recognition of the downstream 5' splice site by U1. Remarkably, U1 snRNP-mediated recognition of the 5' splice site is required both for efficient U2AF binding and for U2AF inhibition by PTB. We propose that TIA-1 and PTB regulate Fas splicing and possibly Fas-mediated apoptosis by targeting molecular events that lead to exon definition.


Sujet(s)
Épissage alternatif , Exons , Protéine PTB/métabolisme , Protéines/métabolisme , Antigènes CD95/génétique , Antigènes CD95/métabolisme , Apoptose , Séquence nucléotidique , Régulation de l'expression des gènes , Extinction de l'expression des gènes , Cellules HeLa , Humains , Modèles moléculaires , Données de séquences moléculaires , Protéines nucléaires/métabolisme , Protéines de liaison au poly(A) , Protéine PTB/pharmacologie , Protéines/pharmacologie , Protéines de liaison à l'ARN , Ribonucléoprotéines/métabolisme , Facteur d'épissage U2AF , Antigène intracellulaire-1 des lymphocytes T
7.
Virus Res ; 98(2): 141-9, 2003 Dec.
Article de Anglais | MEDLINE | ID: mdl-14659561

RÉSUMÉ

The 5' untranslated region (5'UTR) of the hepatitis A virus (HAV) genomic RNA contains an internal ribosome entry site (IRES) which interacts with various cellular proteins and facilitates cap-independent translation. We report the interaction of a 25kDa protein (p25), present in certain murine tissues and most abundantly in mouse kidney, with the HAV 5'UTR. This protein was found to be a cleavage product of the polypyrimidine tract-binding protein (PTB) and competed with it for binding to the HAV 5'UTR RNA. The binding site of p25 overlapped with the reported binding site of PTB. Exogenous addition of partially purified p25 to in vitro translation reactions resulted in the inhibition of HAV IRES-mediated translation, which could be rescued by the addition of purified PTB. These results suggest that p25 is a cleavage product of PTB which binds to the HAV IRES and antagonizes the translation-stimulating activity of PTB. The presence of the 25kDa cleavage product of PTB may therefore play a role in the inhibition of HAV IRES-mediated translation in mouse tissues.


Sujet(s)
Régions 5' non traduites/effets des médicaments et des substances chimiques , Virus de l'hépatite A/effets des médicaments et des substances chimiques , Protéine PTB/pharmacologie , Biosynthèse des protéines/effets des médicaments et des substances chimiques , Animaux , Cellules HeLa , Virus de l'hépatite A/génétique , Humains , Souris , Masse moléculaire , Fragments peptidiques/pharmacologie , Protéines de liaison à l'ARN/métabolisme , Séquences d'acides nucléiques régulatrices
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...