Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 875
Filtrer
1.
Int J Biol Sci ; 20(9): 3570-3589, 2024.
Article de Anglais | MEDLINE | ID: mdl-38993556

RÉSUMÉ

Background: Cisplatin (DDP) based combination chemotherapy is a vital method for the treatment of bladder cancer (BLca). Chemoresistance easily occurs in the course of cisplatin chemotherapy, which is one of the important reasons for the unfavorable prognosis of BLca patients. Circular RNAs (circRNAs) are widely recognized for their role in the development and advancement of BLca. Nevertheless, the precise role of circRNAs in DDP resistance for BLca remains unclear. Methods: To study the properties of circATIC, sanger sequencing, agarose gel electrophoresis and treatment with RNase R/Actinomycin D were utilized. RT-qPCR assay was utilized to assess the expression levels of circRNA, miRNA and mRNA in BLca tissues and cells. Functional experiments were conducted to assess the function of circATIC in BLca progression and chemosensitivity in vitro. Various techniques such as FISH, Dual-luciferase reporter assay, TRAP, RNA digestion assay, RIP and ChIRP assay were used to investigate the relationships between PTBP1, circATIC, miR-1247-5p and RCC2. Orthotopic bladder cancer model, xenograft subcutaneous tumor model and xenograft lung metastasis tumor model were performed to indicate the function and mechanism of circATIC in BLca progression and chemosensitivity in vivo. Results: In our study, we observed that circATIC expression was significantly enhanced in BLca tissues and cells and DDP resistant cells. Patients with higher circATIC expression have larger tumor diameter, higher incidence of postoperative metastasis and lower overall survival rate. Further experiments showed that circATIC accelerated BLca cell growth and metastasis and induced DDP resistance. Mechanistically, alternative splicing enzyme PTBP1 mediated the synthesis of circATIC. circATIC could enhance RCC2 mRNA stability via sponging miR-1247-5p or constructing a circATIC/LIN28A/RCC2 RNA-protein ternary complex. Finally, circATIC promotes RCC2 expression to enhance Epithelial-Mesenchymal Transition (EMT) progression and activate JNK signal pathway, thus strengthening DDP resistance in BLca cells. Conclusion: Our study demonstrated that circATIC promoted BLca progression and DDP resistance, and could serve as a potential target for BLca treatment.


Sujet(s)
Cisplatine , Résistance aux médicaments antinéoplasiques , Ribonucléoprotéines nucléaires hétérogènes , Protéine PTB , ARN circulaire , Tumeurs de la vessie urinaire , Tumeurs de la vessie urinaire/métabolisme , Tumeurs de la vessie urinaire/traitement médicamenteux , Tumeurs de la vessie urinaire/anatomopathologie , Tumeurs de la vessie urinaire/génétique , Humains , ARN circulaire/génétique , ARN circulaire/métabolisme , Cisplatine/usage thérapeutique , Cisplatine/pharmacologie , Résistance aux médicaments antinéoplasiques/génétique , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/génétique , Protéine PTB/métabolisme , Protéine PTB/génétique , Animaux , Lignée cellulaire tumorale , Souris , Souris nude , microARN/métabolisme , microARN/génétique , Mâle , Femelle , Évolution de la maladie , Régulation de l'expression des gènes tumoraux , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Souris de lignée BALB C , Prolifération cellulaire/effets des médicaments et des substances chimiques
2.
Cell Death Dis ; 15(7): 504, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39009589

RÉSUMÉ

Abnormal epigenetic modifications are involved in the regulation of Warburg effect in tumor cells. Protein arginine methyltransferases (PRMTs) mediate arginine methylation and have critical functions in cellular responses. PRMTs are deregulated in a variety of cancers, but their precise roles in Warburg effect in cancer is largely unknown. Experiments from the current study showed that PRMT1 was highly expressed under conditions of glucose sufficiency. PRMT1 induced an increase in the PKM2/PKM1 ratio through upregulation of PTBP1, in turn, promoting aerobic glycolysis in non-small cell lung cancer (NSCLC). The PRMT1 level in p53-deficient and p53-mutated NSCLC remained relatively unchanged while the expression was reduced in p53 wild-type NSCLC under conditions of glucose insufficiency. Notably, p53 activation under glucose-deficient conditions could suppress USP7 and further accelerate the polyubiquitin-dependent degradation of PRMT1. Melatonin, a hormone that inhibits glucose intake, markedly suppressed cell proliferation of p53 wild-type NSCLC, while a combination of melatonin and the USP7 inhibitor P5091 enhanced the anticancer activity in p53-deficient NSCLC. Our collective findings support a role of PRMT1 in the regulation of Warburg effect in NSCLC. Moreover, combination treatment with melatonin and the USP7 inhibitor showed good efficacy, providing a rationale for the development of PRMT1-based therapy to improve p53-deficient NSCLC outcomes.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Protéines membranaires , Protein-arginine N-methyltransferases , , Hormones thyroïdiennes , Protéine p53 suppresseur de tumeur , Effet Warburg en oncologie , Protein-arginine N-methyltransferases/métabolisme , Protein-arginine N-methyltransferases/génétique , Humains , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/traitement médicamenteux , Effet Warburg en oncologie/effets des médicaments et des substances chimiques , Protéine p53 suppresseur de tumeur/métabolisme , Hormones thyroïdiennes/métabolisme , Lignée cellulaire tumorale , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Protéines de transport/métabolisme , Protéines de transport/génétique , Ubiquitin-specific peptidase 7/métabolisme , Ubiquitin-specific peptidase 7/génétique , Protéines de répression/métabolisme , Protéines de répression/génétique , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/génétique , Animaux , Glycolyse/effets des médicaments et des substances chimiques , Souris nude , Glucose/métabolisme , Souris , Régulation de l'expression des gènes tumoraux , Cellules A549 , Protéine PTB
3.
Exp Cell Res ; 440(2): 114147, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38944174

RÉSUMÉ

Coronary artery calcification (CAC) is a hallmark event in the pathogenesis of cardiovascular disease, involving the phenotypic transformation of vascular smooth muscle cells (VSMC) towards an osteogenic state. Despite this understanding, the molecular mechanisms governing the VSMC osteogenic switch remain incompletely elucidated. Here, we sought to examine the potential role of circular RNA (circRNA) in the context of CAC. Through transcriptome analysis of circRNA-seq, we identified circTOP1 as a potential candidate circRNA in individuals with CAC. Furthermore, we observed that overexpression of circTOP1 exacerbated vascular calcification in a CAC model. Subsequent pull-down assays revealed an interaction between circTOP1 and PTBP1, a putative target gene of circTOP1 in the context of CAC. In both in vivo and in vitro experiments, we observed heightened expression of circTOP1 and PTBP1 in the CAC model, and noted that reducing circTOP1 expression effectively reduced calcium salt deposits and mineralized nodules in model mice. Additionally, in vitro experiments demonstrated that overexpression of PTBP1 reversed the weakening of signaling caused by silencing circTOP1, thereby exacerbating the osteogenic transition and calcification of VSMC. Collectively, our findings suggested that circTOP1 promotes CAC by modulating PTBP1 expression to mediate VSMC transdifferentiation.


Sujet(s)
Ribonucléoprotéines nucléaires hétérogènes , Muscles lisses vasculaires , Myocytes du muscle lisse , Protéine PTB , ARN circulaire , Calcification vasculaire , Protéine PTB/génétique , Protéine PTB/métabolisme , Animaux , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/génétique , Souris , ARN circulaire/génétique , ARN circulaire/métabolisme , Humains , Calcification vasculaire/génétique , Calcification vasculaire/anatomopathologie , Calcification vasculaire/métabolisme , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Mâle , Maladie des artères coronaires/génétique , Maladie des artères coronaires/anatomopathologie , Maladie des artères coronaires/métabolisme , Souris de lignée C57BL , Vaisseaux coronaires/anatomopathologie , Vaisseaux coronaires/métabolisme , Ostéogenèse/génétique , Évolution de la maladie , Régulation de l'expression des gènes/génétique
4.
Sci Rep ; 14(1): 14584, 2024 06 25.
Article de Anglais | MEDLINE | ID: mdl-38918441

RÉSUMÉ

PTBP1 is an oncogene that regulates the splicing of precursor mRNA. However, the relationship between PTBP1 expression and gene methylation, cancer prognosis, and tumor microenvironment remains unclear. The expression profiles of PTBP1 across various cancers were derived from the TCGA, as well as the GTEx and CGGA databases. The CGGA mRNA_325, CGGA mRNA_301, and CGGA mRNA_693 datasets were utilized as validation cohorts. Immune cell infiltration scores were approximated using the TIMER 2.0 tool. Functional enrichment analysis for groups with high and low PTBP1 expression was conducted using Gene Set Enrichment Analysis (GSEA). Methylation data were predominantly sourced from the SMART and Mexpress databases. Linked-omics analysis was employed to perform functional enrichment analysis of genes related to PTBP1 methylation, as well as to conduct protein functional enrichment analysis. Single-cell transcriptome analysis and spatial transcriptome analysis were carried out using Seurat version 4.10. Compared to normal tissues, PTBP1 is significantly overexpressed and hypomethylated in various cancers. It is implicated in prognosis, immune cell infiltration, immune checkpoint expression, genomic variation, tumor neoantigen load, and tumor mutational burden across a spectrum of cancers, with particularly notable effects in low-grade gliomas. In the context of gliomas, PTBP1 expression correlates with WHO grade and IDH1 mutation status. PTBP1 expression and methylation play an important role in a variety of cancers. PTBP1 can be used as a marker of inflammation, progression and prognosis in gliomas.


Sujet(s)
Marqueurs biologiques tumoraux , Régulation de l'expression des gènes tumoraux , Gliome , Ribonucléoprotéines nucléaires hétérogènes , Protéine PTB , Microenvironnement tumoral , Humains , Protéine PTB/génétique , Protéine PTB/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/génétique , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Pronostic , Marqueurs biologiques tumoraux/génétique , Gliome/génétique , Gliome/anatomopathologie , Microenvironnement tumoral/génétique , Microenvironnement tumoral/immunologie , Méthylation de l'ADN , Analyse de profil d'expression de gènes , Inflammation/génétique , Transcriptome , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/mortalité , Évolution de la maladie , Multi-omique
5.
Genome Biol ; 25(1): 162, 2024 06 20.
Article de Anglais | MEDLINE | ID: mdl-38902825

RÉSUMÉ

BACKGROUND: The functional coupling between alternative pre-mRNA splicing (AS) and the mRNA quality control mechanism called nonsense-mediated decay (NMD) can modulate transcript abundance. Previous studies have identified several examples of such a regulation in developing neurons. However, the systems-level effects of AS-NMD in this context are poorly understood. RESULTS: We developed an R package, factR2, which offers a comprehensive suite of AS-NMD analysis functions. Using this tool, we conducted a longitudinal analysis of gene expression in pluripotent stem cells undergoing induced neuronal differentiation. Our analysis uncovers hundreds of AS-NMD events with significant potential to regulate gene expression. Notably, this regulation is significantly overrepresented in specific functional groups of developmentally downregulated genes. Particularly strong association with gene downregulation is detected for alternative cassette exons stimulating NMD upon their inclusion into mature mRNA. By combining bioinformatic analyses with CRISPR/Cas9 genome editing and other experimental approaches we show that NMD-stimulating cassette exons regulated by the RNA-binding protein PTBP1 dampen the expression of their genes in developing neurons. We also provided evidence that the inclusion of NMD-stimulating cassette exons into mature mRNAs is temporally coordinated with NMD-independent gene repression mechanisms. CONCLUSIONS: Our study provides an accessible workflow for the discovery and prioritization of AS-NMD targets. It further argues that the AS-NMD pathway plays a widespread role in developing neurons by facilitating the downregulation of functionally related non-neuronal genes.


Sujet(s)
Épissage alternatif , Régulation négative , Neurones , Dégradation des ARNm non-sens , Protéine PTB , Animaux , Souris , Neurones/métabolisme , Protéine PTB/métabolisme , Protéine PTB/génétique , Exons , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/génétique , ARN messager/génétique , ARN messager/métabolisme , Régulation de l'expression des gènes au cours du développement , Différenciation cellulaire/génétique , Neurogenèse/génétique
7.
Nat Commun ; 15(1): 5136, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38879605

RÉSUMÉ

Coordination of neuronal differentiation with expansion of the neuroepithelial/neural progenitor cell (NEPC/NPC) pool is essential in early brain development. Our in vitro and in vivo studies identify independent and opposing roles for two neural-specific and differentially expressed non-coding RNAs derived from the same locus: the evolutionarily conserved lncRNA Rncr3 and the embedded microRNA miR124a-1. Rncr3 regulates NEPC/NPC proliferation and controls the biogenesis of miR124a, which determines neuronal differentiation. Rncr3 conserved exons 2/3 are cytosine methylated and bound by methyl-CpG binding protein MeCP2, which restricts expression of miR124a embedded in exon 4 to prevent premature neuronal differentiation, and to orchestrate proper brain growth. MeCP2 directly binds cytosine-methylated Rncr3 through previously unrecognized lysine residues and suppresses miR124a processing by recruiting PTBP1 to block access of DROSHA-DGCR8. Thus, miRNA processing is controlled by lncRNA m5C methylation along with the defined m5C epitranscriptomic RNA reader protein MeCP2 to coordinate brain development.


Sujet(s)
Protéine-2 de liaison au CpG méthylé , microARN , Cellules souches neurales , Neurogenèse , ARN long non codant , microARN/métabolisme , microARN/génétique , Protéine-2 de liaison au CpG méthylé/métabolisme , Protéine-2 de liaison au CpG méthylé/génétique , Neurogenèse/génétique , Animaux , Souris , ARN long non codant/métabolisme , ARN long non codant/génétique , Cellules souches neurales/métabolisme , Cellules souches neurales/cytologie , Encéphale/métabolisme , Encéphale/embryologie , Humains , Différenciation cellulaire , Méthylation de l'ADN , Protéine PTB/métabolisme , Protéine PTB/génétique , Prolifération cellulaire , Souris de lignée C57BL , 5-Méthyl-cytosine/métabolisme , 5-Méthyl-cytosine/analogues et dérivés , Mâle , Exons/génétique , Neurones/métabolisme , Ribonuclease III
8.
Nat Commun ; 15(1): 4110, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38750024

RÉSUMÉ

Maturation of eukaryotic pre-mRNAs via splicing and polyadenylation is modulated across cell types and conditions by a variety of RNA-binding proteins (RBPs). Although there exist over 1,500 RBPs in human cells, their binding motifs and functions still remain to be elucidated, especially in the complex environment of tissues and in the context of diseases. To overcome the lack of methods for the systematic and automated detection of sequence motif-guided pre-mRNA processing regulation from RNA sequencing (RNA-Seq) data we have developed MAPP (Motif Activity on Pre-mRNA Processing). Applying MAPP to RBP knock-down experiments reveals that many RBPs regulate both splicing and polyadenylation of nascent transcripts by acting on similar sequence motifs. MAPP not only infers these sequence motifs, but also unravels the position-dependent impact of the RBPs on pre-mRNA processing. Interestingly, all investigated RBPs that act on both splicing and 3' end processing exhibit a consistently repressive or activating effect on both processes, providing a first glimpse on the underlying mechanism. Applying MAPP to normal and malignant brain tissue samples unveils that the motifs bound by the PTBP1 and RBFOX RBPs coordinately drive the oncogenic splicing program active in glioblastomas demonstrating that MAPP paves the way for characterizing pre-mRNA processing regulators under physiological and pathological conditions.


Sujet(s)
Polyadénylation , Précurseurs des ARN , Épissage des ARN , Protéines de liaison à l'ARN , Humains , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Précurseurs des ARN/métabolisme , Précurseurs des ARN/génétique , Régulation de l'expression des gènes tumoraux , Tumeurs/génétique , Tumeurs/métabolisme , Motifs nucléotidiques , Protéine PTB/métabolisme , Protéine PTB/génétique , Facteurs d'épissage des ARN/métabolisme , Facteurs d'épissage des ARN/génétique , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/génétique , ARN messager/métabolisme , ARN messager/génétique
9.
Cell Stem Cell ; 31(5): 754-771.e6, 2024 May 02.
Article de Anglais | MEDLINE | ID: mdl-38701759

RÉSUMÉ

Development of embryonic stem cells (ESCs) into neurons requires intricate regulation of transcription, splicing, and translation, but how these processes interconnect is not understood. We found that polypyrimidine tract binding protein 1 (PTBP1) controls splicing of DPF2, a subunit of BRG1/BRM-associated factor (BAF) chromatin remodeling complexes. Dpf2 exon 7 splicing is inhibited by PTBP1 to produce the DPF2-S isoform early in development. During neuronal differentiation, loss of PTBP1 allows exon 7 inclusion and DPF2-L expression. Different cellular phenotypes and gene expression programs were induced by these alternative DPF2 isoforms. We identified chromatin binding sites enriched for each DPF2 isoform, as well as sites bound by both. In ESC, DPF2-S preferential sites were bound by pluripotency factors. In neuronal progenitors, DPF2-S sites were bound by nuclear factor I (NFI), while DPF2-L sites were bound by CCCTC-binding factor (CTCF). DPF2-S sites exhibited enhancer modifications, while DPF2-L sites showed promoter modifications. Thus, alternative splicing redirects BAF complex targeting to impact chromatin organization during neuronal development.


Sujet(s)
Épissage alternatif , Différenciation cellulaire , Chromatine , Ribonucléoprotéines nucléaires hétérogènes , Neurones , Protéine PTB , Facteurs de transcription , Épissage alternatif/génétique , Protéine PTB/métabolisme , Protéine PTB/génétique , Animaux , Différenciation cellulaire/génétique , Chromatine/métabolisme , Souris , Neurones/métabolisme , Neurones/cytologie , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Transcription génétique , Cellules souches embryonnaires/métabolisme , Cellules souches embryonnaires/cytologie , Exons/génétique , Humains , Auto-renouvellement cellulaire/génétique
10.
Nat Commun ; 15(1): 4328, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38773155

RÉSUMÉ

Parental experiences can affect the phenotypic plasticity of offspring. In locusts, the population density that adults experience regulates the number and hatching synchrony of their eggs, contributing to locust outbreaks. However, the pathway of signal transmission from parents to offspring remains unclear. Here, we find that transcription factor Forkhead box protein N1 (FOXN1) responds to high population density and activates the polypyrimidine tract-binding protein 1 (Ptbp1) in locusts. FOXN1-PTBP1 serves as an upstream regulator of miR-276, a miRNA to control egg-hatching synchrony. PTBP1 boosts the nucleo-cytoplasmic transport of pre-miR-276 in a "CU motif"-dependent manner, by collaborating with the primary exportin protein exportin 5 (XPO5). Enhanced nuclear export of pre-miR-276 elevates miR-276 expression in terminal oocytes, where FOXN1 activates Ptbp1 and leads to egg-hatching synchrony in response to high population density. Additionally, PTBP1-prompted nuclear export of pre-miR-276 is conserved in insects, implying a ubiquitous mechanism to mediate transgenerational effects.


Sujet(s)
Transport nucléaire actif , Sauterelles , microARN , Protéine PTB , Animaux , microARN/métabolisme , microARN/génétique , Protéine PTB/métabolisme , Protéine PTB/génétique , Sauterelles/génétique , Sauterelles/métabolisme , Femelle , Facteurs de transcription Forkhead/métabolisme , Facteurs de transcription Forkhead/génétique , Ovule/métabolisme , Protéines d'insecte/métabolisme , Protéines d'insecte/génétique , Noyau de la cellule/métabolisme , Ovocytes/métabolisme
11.
Radiother Oncol ; 196: 110310, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38677328

RÉSUMÉ

BACKGROUND: Nasopharyngeal carcinoma (NPC) is a kind of malignant head and neck tumor with high mortality. lncRNAs are valuable diagnostic biomarkers and therapeutic targets for various tumors. This study investigated the effects and mechanism of LINC00313 in nasopharyngeal carcinoma. METHODS: Cell Counting Kit-8 (CCK-8) and immunohistochemistry were used for assessing cell proliferation. The levels of autophagy-related proteins, and stem cell markers were detected. Immunofluorescence assay was used for LC3 detection. Methylated RNA Immunoprecipitation (meRIP) of LINC00313 in NPC cells was assessed. The localization of LINC00313 was verified by luorescence in situ hybridization (FIHS). The interaction between LINC00313 and the downstream targets were analyzed and confirmed by immunoprecipitation (RIP). Besides, the tumorigenesis roles of LINC00313 were confirmed in tumor growth mice model. RESULTS: LINC00313 was increased in NPC tissues and cells. LINC00313 knockdown enhanced autophagy, and decreased stemness and cell viability of NPC cells through regulating STIM1. METTL3/IGF2BP1-mediated m6A modification promoted the stabilization and up-regulation of LINC00313. LINC00313 activated AKT/mTOR pathway in NPC cells through PTBP1/STIM1 axis. Moreover, LINC00313 promoted tumor growth and metastasis in xenograft model. CONCLUSION: Upregulation of LINC00313 suppressed autophagy and promoted stemness of NPC cells through PTBP1/STIM1 axis.


Sujet(s)
Autophagie , Ribonucléoprotéines nucléaires hétérogènes , Cancer du nasopharynx , Tumeurs du rhinopharynx , Protéine PTB , ARN long non codant , Humains , ARN long non codant/génétique , Cancer du nasopharynx/anatomopathologie , Cancer du nasopharynx/génétique , Cancer du nasopharynx/métabolisme , Souris , Animaux , Tumeurs du rhinopharynx/anatomopathologie , Tumeurs du rhinopharynx/génétique , Tumeurs du rhinopharynx/métabolisme , Protéine PTB/génétique , Protéine PTB/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/génétique , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Prolifération cellulaire , Lignée cellulaire tumorale , Cellules souches tumorales/métabolisme , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Souris nude
12.
Plant J ; 118(6): 2202-2218, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38578875

RÉSUMÉ

Alternative splicing (AS) is a complex process that generates transcript variants from a single pre-mRNA and is involved in numerous biological functions. Many RNA-binding proteins are known to regulate AS; however, little is known about the underlying mechanisms, especially outside the mammalian clade. Here, we show that polypyrimidine tract binding proteins (PTBs) from Arabidopsis thaliana regulate AS of cassette exons via pyrimidine (Py)-rich motifs close to the alternative splice sites. Mutational studies on three PTB-dependent cassette exon events revealed that only some of the Py motifs in this region are critical for AS. Moreover, in vitro binding of PTBs did not reflect a motif's impact on AS in vivo. Our mutational studies and bioinformatic investigation of all known PTB-regulated cassette exons from A. thaliana and human suggested that the binding position of PTBs relative to a cassette exon defines whether its inclusion or skipping is induced. Accordingly, exon skipping is associated with a higher frequency of Py stretches within the cassette exon, and in human also upstream of it, whereas exon inclusion is characterized by increased Py motif occurrence downstream of said exon. Enrichment of Py motifs downstream of PTB-activated 5' splice sites is also seen for PTB-dependent intron removal and alternative 5' splice site events from A. thaliana, suggesting this is a common step of exon definition. In conclusion, the position-dependent AS regulatory mechanism by PTB homologs has been conserved during the separate evolution of plants and mammals, while other critical features, in particular intron length, have considerably changed.


Sujet(s)
Épissage alternatif , Protéines d'Arabidopsis , Arabidopsis , Exons , Protéine PTB , Arabidopsis/génétique , Arabidopsis/métabolisme , Exons/génétique , Protéine PTB/génétique , Protéine PTB/métabolisme , Protéines d'Arabidopsis/génétique , Protéines d'Arabidopsis/métabolisme , Pyrimidines , Humains
13.
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
14.
Elife ; 132024 Apr 10.
Article de Anglais | MEDLINE | ID: mdl-38597390

RÉSUMÉ

Alternative RNA splicing is an essential and dynamic process in neuronal differentiation and synapse maturation, and dysregulation of this process has been associated with neurodegenerative diseases. Recent studies have revealed the importance of RNA-binding proteins in the regulation of neuronal splicing programs. However, the molecular mechanisms involved in the control of these splicing regulators are still unclear. Here, we show that KIS, a kinase upregulated in the developmental brain, imposes a genome-wide alteration in exon usage during neuronal differentiation in mice. KIS contains a protein-recognition domain common to spliceosomal components and phosphorylates PTBP2, counteracting the role of this splicing factor in exon exclusion. At the molecular level, phosphorylation of unstructured domains within PTBP2 causes its dissociation from two co-regulators, Matrin3 and hnRNPM, and hinders the RNA-binding capability of the complex. Furthermore, KIS and PTBP2 display strong and opposing functional interactions in synaptic spine emergence and maturation. Taken together, our data uncover a post-translational control of splicing regulators that link transcriptional and alternative exon usage programs in neuronal development.


Sujet(s)
Épissage alternatif , Exons , Neurones , Protéine PTB , Protein-Serine-Threonine Kinases , Animaux , Humains , Souris , Exons/génétique , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/génétique , Protéines de tissu nerveux/métabolisme , Protéines de tissu nerveux/génétique , Neurones/métabolisme , Phosphorylation , Protéine PTB/métabolisme , Protéine PTB/génétique , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Protéines et peptides de signalisation intracellulaire/métabolisme
15.
J Clin Invest ; 134(11)2024 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-38662454

RÉSUMÉ

Widespread alterations in RNA alternative splicing (AS) have been identified in adult gliomas. However, their regulatory mechanism, biological significance, and therapeutic potential remain largely elusive. Here, using a computational approach with both bulk and single-cell RNA-Seq, we uncover a prognostic AS signature linked with neural developmental hierarchies. Using advanced iPSC glioma models driven by glioma driver mutations, we show that this AS signature could be enhanced by EGFRvIII and inhibited by in situ IDH1 mutation. Functional validations of 2 isoform switching events in CERS5 and MPZL1 show regulations of sphingolipid metabolism and SHP2 signaling, respectively. Analysis of upstream RNA binding proteins reveals PTBP1 as a key regulator of the AS signature where targeting of PTBP1 suppresses tumor growth and promotes the expression of a neuron marker TUJ1 in glioma stem-like cells. Overall, our data highlights the role of AS in affecting glioma malignancy and heterogeneity and its potential as a therapeutic vulnerability for treating adult gliomas.


Sujet(s)
Épissage alternatif , Gliome , Protéine PTB , Gliome/génétique , Gliome/anatomopathologie , Gliome/métabolisme , Gliome/thérapie , Humains , Protéine PTB/génétique , Protéine PTB/métabolisme , Animaux , Souris , Ribonucléoprotéines nucléaires hétérogènes/génétique , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/thérapie , Adulte , Cellules souches pluripotentes induites/métabolisme , Lignée cellulaire tumorale , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Protéines tumorales/génétique , Protéines tumorales/métabolisme
16.
Nucleic Acids Res ; 52(7): 3971-3988, 2024 Apr 24.
Article de Anglais | MEDLINE | ID: mdl-38300787

RÉSUMÉ

The RAVER1 protein serves as a co-factor in guiding the polypyrimidine tract-binding protein (PTBP)-dependent control of alternative splicing (AS). Whether RAVER1 solely acts in concert with PTBPs and how it affects cancer cell fate remained elusive. Here, we provide the first comprehensive investigation of RAVER1-controlled AS in cancer cell models. This reveals a pro-oncogenic role of RAVER1 in modulating tumor growth and epithelial-mesenchymal-transition (EMT). Splicing analyses and protein-association studies indicate that RAVER1 guides AS in association with other splicing regulators, including PTBPs and SRSFs. In cancer cells, one major function of RAVER1 is the stimulation of proliferation and restriction of apoptosis. This involves the modulation of AS events within the miR/RISC pathway. Disturbance of RAVER1 impairs miR/RISC activity resulting in severely deregulated gene expression, which promotes lethal TGFB-driven EMT. Among others, RAVER1-modulated splicing events affect the insertion of protein interaction modules in factors guiding miR/RISC-dependent gene silencing. Most prominently, in all three human TNRC6 proteins, RAVER1 controls AS of GW-enriched motifs, which are essential for AGO2-binding and the formation of active miR/RISC complexes. We propose, that RAVER1 is a key modulator of AS events in the miR/RISC pathway ensuring proper abundance and composition of miR/RISC effectors. This ensures balanced expression of TGFB signaling effectors and limits TGFB induced lethal EMT.


Sujet(s)
Épissage alternatif , Transition épithélio-mésenchymateuse , microARN , Transition épithélio-mésenchymateuse/génétique , Humains , microARN/métabolisme , microARN/génétique , Lignée cellulaire tumorale , Protéine PTB/métabolisme , Protéine PTB/génétique , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Facteurs d'épissage riches en sérine-arginine/métabolisme , Facteurs d'épissage riches en sérine-arginine/génétique , Régulation de l'expression des gènes tumoraux , Prolifération cellulaire/génétique , Apoptose/génétique , Facteur de croissance transformant bêta/métabolisme , Animaux
17.
Carcinogenesis ; 45(6): 409-423, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38366384

RÉSUMÉ

In recent decades, considerable evidence has emerged indicating the involvement of tRNA-derived fragments (tRFs) in cancer progression through various mechanisms. However, the biological effects and mechanisms of tRFs in lung adenocarcinoma (LUAD) remain unclear. In this study, we screen out tRF-29-79, a 5'-tRF derived from tRNAGlyGCC, through profiling the tRF expressions in three pairs of LUAD tissues. We show that tRF-29-79 is downregulated in LUAD and downregulation of tRF-29-79 is associated with poorer prognosis. In vivo and in vitro assay reveal that tRF-29-79 inhibits proliferation, migration and invasion of LUAD cells. Mechanistically, we discovered that tRF-29-79 interacts with the RNA-binding protein PTBP1 and facilitates the transportation of PTBP1 from nucleus to cytoplasm, which regulates alternative splicing in the 3' untranslated region (UTR) of SLC1A5 pre-mRNA. Given that SLC1A5 is a core transporter of glutamine, we proved that tRF-29-79 mediate glutamine metabolism of LUAD through affecting the stability of SLC1A5 mRNA, thus exerts its anticancer function. In summary, our findings uncover the novel mechanism that tRF-29-79 participates in glutamine metabolism through interacting with PTBP1 and regulating alternative splicing in the 3' UTR of SLC1A5 pre-mRNA.


Sujet(s)
Adénocarcinome pulmonaire , Système ASC de transport d'acides aminés , Prolifération cellulaire , Évolution de la maladie , Régulation de l'expression des gènes tumoraux , Ribonucléoprotéines nucléaires hétérogènes , Tumeurs du poumon , Protéine PTB , Humains , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/métabolisme , Protéine PTB/métabolisme , Protéine PTB/génétique , Système ASC de transport d'acides aminés/métabolisme , Système ASC de transport d'acides aminés/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Souris , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/génétique , Animaux , Antigènes mineurs d'histocompatibilité/génétique , Antigènes mineurs d'histocompatibilité/métabolisme , Mouvement cellulaire , Pronostic , Lignée cellulaire tumorale , Épissage alternatif , Femelle , Glutamine/métabolisme , Mâle
18.
J Biol Chem ; 300(3): 105733, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38336291

RÉSUMÉ

RNA Binding Proteins regulate, in part, alternative pre-mRNA splicing and, in turn, gene expression patterns. Polypyrimidine tract binding proteins PTBP1 and PTBP2 are paralogous RNA binding proteins sharing 74% amino acid sequence identity. Both proteins contain four structured RNA-recognition motifs (RRMs) connected by linker regions and an N-terminal region. Despite their similarities, the paralogs have distinct tissue-specific expression patterns and can regulate discrete sets of target exons. How two highly structurally similar proteins can exert different splicing outcomes is not well understood. Previous studies revealed that PTBP2 is post-translationally phosphorylated in the unstructured N-terminal, Linker 1, and Linker 2 regions that share less sequence identity with PTBP1 signifying a role for these regions in dictating the paralog's distinct splicing activities. To this end, we conducted bioinformatics analysis to determine the evolutionary conservation of RRMs versus linker regions in PTBP1 and PTBP2 across species. To determine the role of PTBP2 unstructured regions in splicing activity, we created hybrid PTBP1-PTBP2 constructs that had counterpart PTBP1 regions swapped to an otherwise PTBP2 protein and assayed on differentially regulated exons. We also conducted molecular dynamics studies to investigate how negative charges introduced by phosphorylation in PTBP2 unstructured regions can alter their physical properties. Collectively, results from our studies reveal an important role for PTBP2 unstructured regions and suggest a role for phosphorylation in the differential splicing activities of the paralogs on certain regulated exons.


Sujet(s)
Épissage alternatif , Protéine PTB , Vertébrés , Animaux , Humains , Souris , Rats , Exons/génétique , Ribonucléoprotéines nucléaires hétérogènes/composition chimique , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Simulation de dynamique moléculaire , Protéines de tissu nerveux/composition chimique , Protéines de tissu nerveux/métabolisme , Spécificité d'organe , Phosphorylation , Protéine PTB/composition chimique , Protéine PTB/métabolisme , Spécificité d'espèce , Vertébrés/génétique , Poulets/génétique
19.
J Neuropathol Exp Neurol ; 83(4): 276-288, 2024 03 20.
Article de Anglais | MEDLINE | ID: mdl-38324733

RÉSUMÉ

Cerebral infarction (CI) is one of the leading causes of disability and death. LncRNAs are key factors in CI progression. Herein, we studied the function of long noncoding RNA KCNQ1OT1 in CI patient plasma samples and in CI models. Quantitative real-time PCR and Western blotting tested gene and protein expressions. The interactions of KCNQ1OT1/PTBP1 and miR-16-5p were analyzed using dual-luciferase reporter and RNA immunoprecipitation assays; MTT assays measured cell viability. Cell migration and angiogenesis were tested by wound healing and tube formation assays. Pathological changes were analyzed by triphenyltetrazolium chloride and routine staining. We found that KCNQ1OT1 and PTBP1 were overexpressed and miR-16-5p was downregulated in CI patient plasma and in oxygen-glucose deprived (OGD) induced mouse brain microvascular endothelial (bEnd.3) cells. KCNQ1OT1 knockdown suppressed pro-inflammatory cytokine production and stimulated angiogenic responses in OGD-bEnd.3 cells. KCNQ1OT1 upregulated PTBP1 by sponging miR-16-5p. PTBP1 overexpression or miR-16-5p inhibition attenuated the effects of KCNQ1OT1 knockdown. PTBP1 silencing protected against OGD-bEnd.3 cell injury by enhancing SIRT1. KCNQ1OT1 silencing or miR-16-5p overexpression also alleviated ischemic injury in a mice middle cerebral artery occlusion model. Thus, KCNQ1OT1 silencing alleviates CI by regulating the miR-16-5p/PTBP1/SIRT1 pathway, providing a theoretical basis for novel therapeutic strategies targeting CI.


Sujet(s)
microARN , ARN long non codant , Animaux , Humains , Souris , ARN long non codant/génétique , ARN long non codant/métabolisme , Cellules endothéliales/métabolisme , Sirtuine-1/génétique , Sirtuine-1/métabolisme , Apoptose/génétique , microARN/génétique , microARN/métabolisme , Infarctus du territoire de l'artère cérébrale moyenne/métabolisme , Oxygène , Ribonucléoprotéines nucléaires hétérogènes , Protéine PTB/génétique
20.
Thorac Cancer ; 15(10): 808-819, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38409914

RÉSUMÉ

BACKGROUND: Breast cancer (BC) is a common malignancy which threatens the health of women. Circular RNAs (circRNAs) are critical factors in multiple cancers, including BC. The aim of this experiment was to investigate the molecular mechanisms of circRNA Septin 9 (circSEPT9) in the progression of BC. METHODS: CircSEPT9, microRNA-625-5p (miR-625-5p) and polypyrimidine tract-binding protein 3 (PTBP3) levels were determined by quantitative real-time polymerase chain reaction (qRT-PCR). Western blot was performed to detect the protein levels of PTBP3, E-cadherin and vimentin. Cell counting kit-8 assay (CCK8) and thymidine analog 5-ethynyl-2'-deoxyuridine (EDU) was utilized for proliferation examination. Flow cytometry was conducted to measure apoptosis. Transwell assay and wound healing assay to investigate the migration of BC cells. Glucose uptake and lactate production were determined by specific kits. Additionally, dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were utilized to verify the interaction. A murine xenograft model was established to investigate the function of circSEPT9 in BC in vivo. RESULTS: Overexpression of circSEPT9 was found in BC tissues and cells. Silencing circSEPT9 impeded BC cell proliferation, migration, epithelial-mesenchymal transition (EMT) and glycolytic metabolism but facilitated cell apoptosis in vitro. Meanwhile, circSEPT9 knockdown constrained tumor growth in vivo. MiR-625-5p was targeted by circSEPT9. The influence of silencing circSEPT9 on BC cell function was regained by miR-625-5p inhibitor. Furthermore, miR-625-5p regulated BC cell malignant phenotypes via downregulating PTBP3. CONCLUSION: circSEPT9 contributed to the malignant progression of BC by up-regulating PTBP3 via sponging miR-625-5p.


Sujet(s)
Tumeurs du sein , microARN , Humains , Femelle , Animaux , Souris , Tumeurs du sein/génétique , Protéine PTB/génétique , Région mammaire , Apoptose , microARN/génétique , Prolifération cellulaire , Lignée cellulaire tumorale
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...