Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 445
Filtrer
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
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
14.
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
15.
Biochim Biophys Acta Mol Basis Dis ; 1870(3): 167011, 2024 03.
Article de Anglais | MEDLINE | ID: mdl-38176460

RÉSUMÉ

Tamoxifen (TAM) is the primary drug for treating estrogen receptor alpha-positive (ER+) breast cancer (BC). However, resistance to TAM can develop in some patients, limiting its therapeutic efficacy. The ubiquitin-specific protease (USP) family has been associated with the development, progression, and drug resistance of various cancers. To explore the role of USPs in TAM resistance in BC, we used qRT-PCR to compare USP expression between TAM-sensitive (MCF-7 and T47D) and TAM-resistant cells (MCF-7R and T47DR). We then modulated USP46 expression and examined its impact on cell proliferation, drug resistance (via CCK-8 and EdU experiments), glycolysis levels (using a glycolysis detection assay), protein interactions (confirmed by co-IP), and protein changes (analyzed through Western blotting). Our findings revealed that USP46 was significantly overexpressed in TAM-resistant BC cells, leading to the inhibition of the ubiquitin degradation of polypyrimidine tract-binding protein 1 (PTBP1). Overexpression of PTBP1 increased the PKM2/PKM1 ratio, promoted glycolysis, and intensified TAM resistance in BC cells. Knockdown of USP46 induced downregulation of PTBP1 protein by promoting its K48-linked ubiquitination, resulting in a decreased PKM2/PKM1 ratio, reduced glycolysis, and heightened TAM sensitivity in BC cells. In conclusion, this study highlights the critical role of the USP46/PTBP1/PKM2 axis in TAM resistance in BC. Targeted therapy against USP46 may represent a promising strategy to improve the prognosis of TAM-resistant patients.


Sujet(s)
Tumeurs du sein , Tamoxifène , Humains , Femelle , Tamoxifène/pharmacologie , Tamoxifène/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Cellules MCF-7 , Résistance aux médicaments antinéoplasiques/génétique , Glycolyse , Ribonucléoprotéines nucléaires hétérogènes/génétique , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Protéine PTB/génétique , Protéine PTB/métabolisme
16.
Yi Chuan ; 46(1): 46-62, 2024 Jan 20.
Article de Anglais | MEDLINE | ID: mdl-38230456

RÉSUMÉ

Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer accounting for 90% of cases. It is a highly invasive and deadly cancer with a gradual onset. Polypyrimidine tract-binding protein 1 (PTBP1) is an important RNA-binding protein involved in RNA metabolism and has been linked to oncogenic splicing events. While the oncogenic role of PTBP1 in HCC cells has been established, the exact mechanism of action remains unclear. This study aimed to investigate the functional connection between PTBP1 and dysregulated splicing events in HCC. Through immunoprecipitation-mass spectrometry analyses, we discovered that the proteins bound to PTBP1 were significantly enriched in the complex responsible for the alternative splicing of FGFR2 (fibroblast growth factor receptor 2). Further RNA immunoprecipitation and quantitative PCR assays confirmed that PTBP1 down-regulated the FGFR2-IIIb isoform levels and up-regulated the FGFR2-IIIc isoform levels in HCC cells, leading to a switch from FGFR2-IIIb to FGFR2-IIIc isoforms. Subsequent functional evaluations using CCK-8, transwell, and plate clone formation assays in HCC cell lines HepG2 and Huh7 demonstrated that FGFR2-IIIb exhibited tumor-suppressive effects, while FGFR2-IIIc displayed tumor-promoting effects. In conclusion, this study provides insights into the PTBP1-mediated alternative splicing mechanism in HCC progression, offering a new theoretical basis for the prevention and treatment of this malignancy. Mechanistically, the isoform switch from FGFR2-IIIb to FGFR2-IIIc promoted epithelial-mesenchymal transformation (EMT) of HCC cells and activated the FGFR cascades ERK and AKT pathways.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , Humains , Récepteur FGFR2/génétique , Récepteur FGFR2/métabolisme , Carcinome hépatocellulaire/génétique , Tumeurs du foie/génétique , Isoformes de protéines/génétique , Épissage alternatif , ARN/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/génétique , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Protéine PTB/génétique , Protéine PTB/métabolisme
17.
Commun Biol ; 7(1): 130, 2024 01 25.
Article de Anglais | MEDLINE | ID: mdl-38273088

RÉSUMÉ

Colorectal cancer is a grievous health concern, we have proved long non-coding RNA LINC00689 is considered as a potential diagnosis biomarker for colorectal cancer, and it is necessary to further investigate its upstream and downstream mechanisms. Here, we show that KLF15, a transcription factor, exhibits the reduced expression in colorectal cancer. KLF15 suppresses the proliferative and metastatic capacities of colorectal cancer cells both in vitro and in vivo by transcriptionally activating LINC00689. Subsequently, LINC00689 recruits PTBP1 protein to enhance the stability of LATS2 mRNA in the cytoplasm. This stabilization causes the suppression of the YAP1/ß-catenin pathway and its target downstream genes. Our findings highlight a regulatory network involving KLF15, LINC00689, PTBP1, LATS2, and the YAP1/ß-catenin pathway in colorectal cancer, shedding light on potential therapeutic targets for colorectal cancer therapy.


Sujet(s)
Tumeurs colorectales , bêta-Caténine , Humains , bêta-Caténine/métabolisme , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Tumeurs colorectales/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Protéines suppresseurs de tumeurs/génétique , Facteurs de transcription Krüppel-like/génétique , Facteurs de transcription Krüppel-like/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/génétique , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Protéine PTB/génétique , Protéine PTB/métabolisme
18.
Cells ; 13(2)2024 01 12.
Article de Anglais | MEDLINE | ID: mdl-38247832

RÉSUMÉ

Gastric cancer (GC) is the most common type of malignant tumor within the gastrointestinal tract, and GC metastasis is associated with poor prognosis. Polypyrimidine tract binding protein 1 (PTBP1) is an RNA-binding protein implicated in various types of tumor development and metastasis. However, the role of PTBP1 in GC metastasis remains elusive. In this study, we verified that PTBP1 was upregulated in GC tissues and cell lines, and higher PTBP1 level was associated with poorer prognosis. It was shown that PTBP1 knockdown in vitro inhibited GC cell migration, whereas PTBP1 overexpression promoted the migration of GC cells. In vivo, the knockdown of PTBP1 notably reduced both the size and occurrence of metastatic nodules in a nude mice liver metastasis model. We identified phosphoglycerate kinase 1 (PGK1) as a downstream target of PTBP1 and found that PTBP1 increased the stability of PGK1 by directly binding to its mRNA. Furthermore, the PGK1/SNAIL axis could be required for PTBP1's function in the promotion of GC cell migration. These discoveries suggest that PTBP1 could be a promising therapeutic target for GC.


Sujet(s)
Phosphoglycerate kinase , Protéine PTB , Tumeurs de l'estomac , Animaux , Souris , Souris nude , ARN messager/génétique , Protéines de liaison à l'ARN , Tumeurs de l'estomac/génétique , Humains , Protéine PTB/génétique , Protéine PTB/métabolisme , Phosphoglycerate kinase/génétique
19.
Virology ; 592: 109986, 2024 04.
Article de Anglais | MEDLINE | ID: mdl-38290414

RÉSUMÉ

The large amount of viral RNA produced during infections has the potential to interact with and effectively sequester cellular RNA binding proteins, thereby influencing aspects of post-transcriptional gene regulation in the infected cell. Here we demonstrate that the abundant 5' leader RNA region of SARS-CoV-2 viral RNAs can interact with the cellular polypyrimidine tract binding protein (PTBP1). Interestingly, the effect of a knockdown of PTBP1 protein on cellular gene expression is also mimicked during SARS-CoV-2 infection, suggesting that this protein may be functionally sequestered by viral RNAs. Consistent with this model, the alternative splicing of mRNAs that is normally controlled by PTBP1 is dysregulated during SARS-CoV-2 infection. Collectively, these data suggest that the SARS-CoV-2 leader RNA sequesters the cellular PTBP1 protein during infection, resulting in significant impacts on the RNA biology of the host cell. These alterations in post-transcriptional gene regulation may play a role in SARS-CoV-2 mediated molecular pathogenesis.


Sujet(s)
COVID-19 , Ribonucléoprotéines nucléaires hétérogènes , Protéine PTB , SARS-CoV-2 , Humains , Épissage alternatif , COVID-19/métabolisme , COVID-19/virologie , Ribonucléoprotéines nucléaires hétérogènes/génétique , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Protéine PTB/génétique , Protéine PTB/métabolisme , ARN/métabolisme , Précurseurs des ARN/génétique , Précurseurs des ARN/métabolisme , Épissage des ARN , SARS-CoV-2/physiologie
20.
Nat Cancer ; 5(1): 30-46, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-37845485

RÉSUMÉ

In vitro-transcribed (IVT) mRNA has arisen as a rapid method for the production of nucleic acid drugs. Here, we have constructed an oncolytic IVT mRNA that utilizes human rhinovirus type 2 (HRV2) internal ribosomal entry sites (IRESs) to selectively trigger translation in cancer cells with high expression of EIF4G2 and PTBP1. The oncolytic effect was provided by a long hGSDMDc .825 T>A/c.884 A>G-F1LCT mutant mRNA sequence with mitochondrial inner membrane cardiolipin targeting toxicity that triggers mitophagy. Utilizing the permuted intron-exon (PIE) splicing circularization strategy and lipid nanoparticle (LNP) encapsulation reduced immunogenicity of the mRNA and enabled delivery to eukaryotic cells in vivo. Engineered HRV2 IRESs-GSDMDp.D275E/E295G-F1LCT circRNA-LNPs (GSDMDENG circRNA) successfully inhibited EIF4G2+/PTBP1+ pan-adenocarcinoma xenografts growth. Importantly, in a spontaneous tumor model with abnormal EIF4G2 and PTBP1 caused by KRAS G12D mutation, GSDMDENG circRNA significantly prevented the occurrence of pancreatic, lung and colon adenocarcinoma, improved the survival rate and induced persistent KRAS G12D tumor antigen-specific cytotoxic T lymphocyte responses.


Sujet(s)
Adénocarcinome , Tumeurs du côlon , Humains , ARN circulaire , Cardiolipides , Protéines proto-oncogènes p21(ras) , ARN messager/génétique , Facteur-4G d'initiation eucaryote/génétique , Facteur-4G d'initiation eucaryote/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/génétique , Protéine PTB/génétique , Protéine PTB/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...