Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Theriogenology ; 225: 107-118, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38805993

ABSTRACT

In this study, we aimed to investigate cytoplasmic maturation and miRNA expression of mature oocytes cultured in porcine follicular fluid exosomes. We also examined the effect of miR-339-5p on oocyte maturation. Twenty eight differentially expressed miRNAs were detected using miRNA-seq. We then transfected cumulus oocyte complexes with miR-339-5p mimics and inhibitor during culture. The results showed that exosomes increased endoplasmic reticulum levels and the amount of lipid droplets, and decreased ROS levels, lipid droplet size, and percentage of oocytes with abnormal cortical granule distribution. Overexpressing miR-339-5p significantly decreased cumulus expansion genes, oocyte maturation-related genes, target gene proline/glutamine-rich splicing factor (SFPQ), ERK1/2 phosphorylation levels, oocyte maturation rate, blastocyst rate, and lipid droplet number, but increased lipid droplet size and the ratio of oocytes with abnormal cortical granule distribution. Inhibiting miR-339-5p reversed the decrease observed during overexpression. Mitochondrial membrane potential and ROS levels did not differ significantly between groups. In summary, exosomes promote oocyte cytoplasmic maturation and miR-339-5p regulating ERK1/2 activity through SFPQ expression, thereby elevating oocyte maturation and blastocyst formation rate in vitro.


Subject(s)
Exosomes , Follicular Fluid , In Vitro Oocyte Maturation Techniques , MAP Kinase Signaling System , MicroRNAs , Oocytes , Animals , Swine , MicroRNAs/metabolism , MicroRNAs/genetics , Oocytes/metabolism , Oocytes/physiology , In Vitro Oocyte Maturation Techniques/veterinary , Exosomes/metabolism , Female , Follicular Fluid/metabolism , PTB-Associated Splicing Factor/metabolism , PTB-Associated Splicing Factor/genetics , Gene Expression Regulation
2.
Nat Commun ; 15(1): 4156, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755141

ABSTRACT

Epstein-Barr virus (EBV) uses a biphasic lifecycle of latency and lytic reactivation to infect >95% of adults worldwide. Despite its central role in EBV persistence and oncogenesis, much remains unknown about how EBV latency is maintained. We used a human genome-wide CRISPR/Cas9 screen to identify that the nuclear protein SFPQ was critical for latency. SFPQ supported expression of linker histone H1, which stabilizes nucleosomes and regulates nuclear architecture, but has not been previously implicated in EBV gene regulation. H1 occupied latent EBV genomes, including the immediate early gene BZLF1 promoter. Upon reactivation, SFPQ was sequestered into sub-nuclear puncta, and EBV genomic H1 occupancy diminished. Enforced H1 expression blocked EBV reactivation upon SFPQ knockout, confirming it as necessary downstream of SFPQ. SFPQ knockout triggered reactivation of EBV in B and epithelial cells, as well as of Kaposi's sarcoma-associated herpesvirus in B cells, suggesting a conserved gamma-herpesvirus role. These findings highlight SFPQ as a major regulator of H1 expression and EBV latency.


Subject(s)
Herpesvirus 4, Human , Histones , PTB-Associated Splicing Factor , Virus Activation , Virus Latency , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Humans , Histones/metabolism , Virus Activation/genetics , Virus Latency/genetics , PTB-Associated Splicing Factor/metabolism , PTB-Associated Splicing Factor/genetics , Gene Expression Regulation, Viral , B-Lymphocytes/virology , B-Lymphocytes/metabolism , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/metabolism , CRISPR-Cas Systems , Promoter Regions, Genetic/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , Genome, Viral
3.
Biochimie ; 222: 9-17, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38373651

ABSTRACT

The cellular SFPQ protein is involved in several stages of the HIV-1 life cycle, but the detailed mechanism of its involvement is not yet fully understood. Here, the role of SFPQ in the early stages of HIV-1 replication has been studied. It is found that changes in the intracellular level of SFPQ affect the integration of viral DNA, but not reverse transcription, and SFPQ is a positive factor of integration. A study of the SFPQ interaction with HIV-1 integrase (IN) has revealed two diRGGX1-4 motifs in the N-terminal region of SFPQ, which are involved in IN binding. Substitution of a single amino acid residue in any of these regions led to a decrease in binding efficiency, while mutations in both motifs almost completely disrupted the SFPQ interaction with IN. The effect of the SFPQ mutants with impaired ability to bind IN on viral replication has been analyzed. Unlike the wild-type protein, the SFPQ mutants did not affect viral integration. This confirms that SFPQ influences the integration stage through direct interaction with IN. Our results indicate that the SFPQ/IN complex can be considered as a potential therapeutic target for the development of new inhibitors of HIV replication.


Subject(s)
HIV Integrase , HIV-1 , PTB-Associated Splicing Factor , Virus Integration , Virus Replication , HIV-1/metabolism , HIV-1/physiology , HIV-1/genetics , Humans , HIV Integrase/metabolism , HIV Integrase/genetics , PTB-Associated Splicing Factor/metabolism , PTB-Associated Splicing Factor/genetics , Protein Binding , Mutation , HEK293 Cells
4.
Medicine (Baltimore) ; 102(45): e35837, 2023 Nov 10.
Article in English | MEDLINE | ID: mdl-37960731

ABSTRACT

Splicing factor proline- and glutamine-rich (SFPQ) can interact with RNAs to regulate gene expression. The function of SFPQ in the immunotherapy of non-small cell lung cancer (NSCLC) is investigated in this study. H1299 and A549 cells were transfected with shSFPQ plasmid. Cell counting kit-8 (CCK-8) and cell clone formation were utilized to detect survival and proliferation. Programmed death-ligand 1 (PD-L1) and SFPQ were detected in NSCLC patients treated with anti-PD-L1 antibody. Dual-luciferase assays, RNA immunoblotting, RNA pull-down, and mRNA stability assay were applied to verify the regulation of PD-L1 with SFPQ. Human peripheral blood mononuclear cells (PBMC)-derived dendritic cells were loaded with irradiated A549 and H1299 cells, which were cultured with autologous CD8+T cells and tumor cells to perform in vitro tumor-specific cytotoxic T lymphocytes (CTL) cytotoxicity analysis. SFPQ silencing inhibited the survival and proliferation of H1299 and A549 cells with down-regulated PD-L1 expression. PD-L1 and SFPQ expression were markedly higher in anti-PD-L1 antibody treatment responders compared to non-responders, which showed a positive Pearson correlation (R = 0.76, P < .001). SFPQ up-regulated the relative mRNA and protein expression of PD-L1 by binding to the PD-L1 3'UTR to slow the decay of PD-L1 mRNA. SFPQ silencing promoted the killing effect of CTL on A549 and H1299 cells. SFPQ up-regulates PD-L1 expression by binding with PD-L1 3'UTR to slow the decay of PD-L1 mRNA, and SFPQ silencing promotes CTL-mediated cytotoxicity on NSCLC cells.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , 3' Untranslated Regions , B7-H1 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Non-Small-Cell Lung/pathology , Glutamine , Leukocytes, Mononuclear/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Lung Neoplasms/metabolism , RNA Splicing Factors/genetics , T-Lymphocytes, Cytotoxic/metabolism , PTB-Associated Splicing Factor/genetics , PTB-Associated Splicing Factor/metabolism
5.
Biochemistry ; 61(17): 1723-1734, 2022 09 06.
Article in English | MEDLINE | ID: mdl-35998361

ABSTRACT

Human polypyrimidine-binding splicing factor (PSF/SFPQ) is a tumor suppressor protein that regulates the gene expression of several proto-oncogenes and binds to the 5'-polyuridine negative-sense template (5'-PUN) of some RNA viruses. The activity of PSF is negatively regulated by long-noncoding RNAs, human metastasis associated in lung adenocarcinoma transcript-1 and murine virus-like 30S transcript-1 (VL30-1). PSF is a 707-amino acid protein that has a DNA-binding domain and two RNA recognition motifs (RRMs). Although the structure of the apo-truncated PSF is known, how PSF recognizes RNA remains elusive. Here, we report the 2.8 Å and 3.5 Å resolution crystal structures of a biologically active truncated construct of PSF (sPSF, consisting of residues 214-598) alone and in a complex with a 30mer fragment of VL30-1 RNA, respectively. The structure of the complex reveals how the 30mer RNA is recognized at two U-specific induced-fit binding pockets, located at the previously unrecognized domain-swapped, inter-subunit RRM1 (of the first subunit)-RRM2 (of the second subunit) interfaces that do not exist in the apo structure. Thus, the sPSF dimer appears to have two conformations in solution: one in a low-affinity state for RNA binding, as seen in the apo-structure, and the other in a high-affinity state for RNA binding, as seen in the sPSF-RNA complex. PSF undergoes an all or nothing transition between having two or no RNA-binding pockets. We predict that the RNA binds with a high degree of positive cooperativity. These structures provide an insight into a new regulatory mechanism that is likely involved in promoting malignancies and other human diseases.


Subject(s)
RNA, Long Noncoding , RNA-Binding Proteins , Animals , Humans , Mice , PTB-Associated Splicing Factor/genetics , PTB-Associated Splicing Factor/metabolism , RNA Splicing , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/metabolism
7.
Cancer Res ; 81(13): 3495-3508, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33975881

ABSTRACT

RNA-binding protein PSF functions as an epigenetic modifier by interacting with long noncoding RNAs and the corepressor complex. PSF also promotes RNA splicing events to enhance oncogenic signals. In this study, we conducted an in vitro chemical array screen and identified multiple small molecules that interact with PSF. Several molecules inhibited RNA binding by PSF and decreased prostate cancer cell viability. Among these molecules and its derivatives was a promising molecule, No. 10-3 [7,8-dihydroxy-4-(4-methoxyphenyl)chromen-2-one], that was the most effective at blocking PSF RNA-binding ability and suppressing treatment-resistant prostate and breast cancer cell proliferation. Exposure to No. 10-3 inhibited PSF target gene expression at the mRNA level. Treatment with No. 10-3 reversed epigenetically repressed PSF downstream targets, such as cell-cycle inhibitors, at the transcriptional level. Chromatin immunoprecipitation sequencing in prostate cancer cells revealed that No. 10-3 enhances histone acetylation to induce expression of apoptosis as well as cell-cycle inhibitors. Furthermore, No. 10-3 exhibited antitumor efficacy in a hormone therapy-resistant prostate cancer xenograft mouse model, suppressing treatment-resistant tumor growth. Taken together, this study highlights the feasibility of targeting PSF-mediated epigenetic and RNA-splicing activities for the treatment of aggressive cancers. SIGNIFICANCE: This study identifies small molecules that target PSF-RNA interactions and suppress hormone therapy-refractory cancer growth, suggesting the potential of targeting PSF-mediated gene regulation for cancer treatment.


Subject(s)
Breast Neoplasms/drug therapy , Epigenesis, Genetic , PTB-Associated Splicing Factor/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , RNA Processing, Post-Transcriptional , RNA, Long Noncoding/metabolism , Small Molecule Libraries/pharmacology , Animals , Apoptosis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , PTB-Associated Splicing Factor/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , RNA, Long Noncoding/genetics , Transcription, Genetic , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
J Mol Med (Berl) ; 99(7): 967-980, 2021 07.
Article in English | MEDLINE | ID: mdl-33770188

ABSTRACT

Reactive oxygen species (ROS), a by-product of oxygen metabolism mainly originating from mitochondria, participate in many pathological processes related to ophthalmopathy. Excessive production of ROS leads to oxidative stress, which influences the permeability, proliferation, migration, and tube formation of human retinal microcapillary endothelial cells (HRMECs). The molecular mechanisms underlying the effects of ROS are not clear. In Vldlr-/- mice, we used fundus fluorescein angiography and retinal flat mount staining to observe the effect of polypyrimidine tract-binding protein-associated splicing factor (PSF) on pathological retinal neovascularization in vivo. Additionally, in human retinal microvascular endothelial cells treated with 4-HNE, cell viability, tube formation, wound healing, and Transwell assays were performed to study the effect of PSF on the proliferation, migration, and tube formation of retinal vascular endothelial cells in vitro. Moreover, reactive oxygen species assay, real-time PCR, and Western blot were included to analyze the potential mechanism of PSF in the above series of effects. PSF ameliorated intraretinal neovascularization (IRNV) in vivo in Vldlr-/- mice. Under 4-hydroxynonenal (4-HNE) conditions in vitro, PSF reprogrammed mitochondrial bioenergetic and glycolytic profiles. It also reduced ROS levels and inhibited 4-HNE-induced angiogenesis, which involves the proliferation, migration, and tube formation of HRMECs. These results suggest that PSF participates in the regulation of HRMECs proliferation and migration during the development of pathological angiogenesis. We demonstrated that PSF enhanced Nrf2 activation and heme oxygenase-1 (HO-1) expression via extracellular signal-regulated kinase (ERK) and Akt signaling in HRMECs, which subsequently resulted in intracellular ROS scavenging. PSF restored endoplasmic reticulum (ER) redox homeostasis, which was indicated by an increase in protein disulfide isomerase (PDI) and Ero-1α and a reduction in GRP78 and C/EBP homologous protein (CHOP). PSF also attenuated ER stress via regulation of the protein kinase R (PKR)-like endoplasmic reticulum kinase PERK/eukaryotic translation factor 2 alpha (eIF2α)/activating transcription factor 4 (ATF4) pathway in 4-HNE-treated HRMECs. Our research shows that PSF may be a potential antioxidant that regulates pathological angiogenesis through ERK-AKT/Nrf2/HO-1 and PERK/eIF2α/ATF4 signal regulation. KEY MESSAGES: Reactive oxygen species (ROS) mainly originating from mitochondria is a by-product of oxygen metabolism in the body and participates in the pathological process related to multiple blindness-related ophthalmopathy. Moreover , excessive production of ROS will lead to oxidative stress. Consequently, oxidative stress influences the permeability, proliferation, migration, and tube formation of human retinal microcapillary endothelial cells (HRMECs). The molecular mechanisms underlying the effects of ROS remain unclear. Here, we reveal that Polypyrimidine tract-binding protein-associated splicing factor (PSF) ameliorates intraretinal neovascularization (IRNV) in vivo in Vldlr-/- mice. Furthermore, under 4-HNE conditions in vitro, PSF reprograms mitochondrial bioenergetic and glycolytic profiles, reduces ROS levels, and inhibits 4-HNE-induced angiogenesis, which involves the proliferation, migration, and tube formation of HRMECs, suggesting that it participates in regulating the proliferation and migration of HRMECs during the development of pathological angiogenesis. Furthermore, PSF enhances Nrf2 activation and HO-1 expression through ERK and AKT signaling in HRMECs, resulting in intracellular ROS scavenging. PSF restores endoplasmic reticulum (ER) redox homeostasis, as indicated by an increase in PDI and Ero-1α and a reduction in GRP78 and CHOP. PSF also attenuates ER stress by regulating the PERK/eIF2α/ATF4 pathway in 4-HNE-treated HRMECs.


Subject(s)
PTB-Associated Splicing Factor/metabolism , Retinal Neovascularization/metabolism , Activating Transcription Factor 4/metabolism , Aldehydes/pharmacology , Animals , Cells, Cultured , Endoplasmic Reticulum Stress , Endothelial Cells/drug effects , Endothelial Cells/physiology , Eukaryotic Initiation Factor-2/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Heme Oxygenase-1/metabolism , Humans , Mice, Inbred C57BL , Mice, Knockout , Microvessels/cytology , Mitochondria/metabolism , PTB-Associated Splicing Factor/genetics , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Receptors, LDL/genetics , Retina/cytology , Retina/metabolism , Retina/pathology , Retinal Neovascularization/genetics , eIF-2 Kinase/metabolism
9.
Nat Commun ; 12(1): 1918, 2021 03 26.
Article in English | MEDLINE | ID: mdl-33771997

ABSTRACT

The RNA-binding protein SFPQ plays an important role in neuronal development and has been associated with several neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), and Alzheimer's disease. Here, we report that loss of sfpq leads to premature termination of multiple transcripts due to widespread activation of previously unannotated cryptic last exons (CLEs). These SFPQ-inhibited CLEs appear preferentially in long introns of genes with neuronal functions and can dampen gene expression outputs and/or give rise to short peptides interfering with the normal gene functions. We show that one such peptide encoded by the CLE-containing epha4b mRNA isoform is responsible for neurodevelopmental defects in the sfpq mutant. The uncovered CLE-repressive activity of SFPQ is conserved in mouse and human, and SFPQ-inhibited CLEs are found expressed across ALS iPSC-derived neurons. These results greatly expand our understanding of SFPQ function and uncover a gene regulation mechanism with wide relevance to human neuropathologies.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Codon, Nonsense , Exons/genetics , PTB-Associated Splicing Factor/genetics , Animals , Base Sequence , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Humans , In Situ Hybridization/methods , Introns/genetics , Mice , Neurons/metabolism , Zebrafish/embryology , Zebrafish/genetics
10.
Hum Mol Genet ; 30(11): 971-984, 2021 05 31.
Article in English | MEDLINE | ID: mdl-33729478

ABSTRACT

Previously, we identified missense mutations in CCNF that are causative of familial and sporadic amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Hallmark features of these diseases include the build-up of insoluble protein aggregates as well as the mislocalization of proteins such as transactive response DNA binding protein 43 kDa (TDP-43). In recent years, the dysregulation of SFPQ (splicing factor proline and glutamine rich) has also emerged as a pathological hallmark of ALS/FTD. CCNF encodes for the protein cyclin F, a substrate recognition component of an E3 ubiquitin ligase. We have previously shown that ALS/FTD-linked mutations in CCNF cause disruptions to overall protein homeostasis that leads to a build-up of K48-linked ubiquitylated proteins as well as defects in autophagic machinery. To investigate further processes that may be affected by cyclin F, we used a protein-proximity ligation method, known as Biotin Identification (BioID), standard immunoprecipitations and mass spectrometry to identify novel interaction partners of cyclin F and infer further process that may be affected by the ALS/FTD-causing mutation. Results demonstrate that cyclin F closely associates with proteins involved with RNA metabolism as well as a number of RNA-binding proteins previously linked to ALS/FTD, including SFPQ. Notably, the overexpression of cyclin F(S621G) led to the aggregation and altered subcellular distribution of SFPQ in human embryonic kidney (HEK293) cells, while leading to altered degradation in primary neurons. Overall, our data links ALS/FTD-causing mutations in CCNF to converging pathological features of ALS/FTD and provides a link between defective protein degradation systems and the pathological accumulation of a protein involved in RNA processing and metabolism.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Cyclins/genetics , Frontotemporal Dementia/genetics , PTB-Associated Splicing Factor/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , DNA-Binding Proteins/genetics , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , HEK293 Cells , Humans , Protein Aggregates/genetics , Protein Interaction Maps/genetics , Proteolysis , RNA/genetics , RNA/metabolism , RNA Processing, Post-Transcriptional/genetics , RNA-Binding Proteins/genetics
11.
Cell Commun Signal ; 19(1): 14, 2021 02 11.
Article in English | MEDLINE | ID: mdl-33573690

ABSTRACT

BACKGROUND: Abnormal neovascularization is the most common cause of blindness, and hypoxia alters tissue metabolism, function, and morphology. HIF-1α, the transcriptional activator of VEGF, has intricate mechanisms of nuclear translocation and activation, but its signal termination mechanisms remain unclear. METHODS: We investigated the role of polypyrimidine tract-binding protein-associated splicing factor (PSF) in cellular energy production, migration, and proliferation by targeting HIF-1α in vivo and in vitro PSF plasmids were transfected with liposome 2000 transfection reagent. Young C57/BL6J mice were kept in a hyperoxia environment, followed by indoor air, resulting in oxygen-induced retinopathy. Oxygen-induced retinopathy (OIR) animals were randomly divided into three groups: OIR group, OIR + vector group (OIR cubs treated with rAAV vector) and OIR + PSF group (OIR cubs treated with rAAV-PSF). Age-matched C57/BL6J mice were used as controls and exposed to constant normoxic conditions. The animals were executed and their pupils were subjected to subsequent experiments. The metabolic spectrum was analyzed by Seahorse XFe96 flux analyzer, and OCR and extracellular acidification rate were quantified at the same time. RESULTS: PSF ameliorated retinal neovascularization and corrected abnormal VEGF expression in mice with oxygen-induced retinopathy and reduced intra-retinal neovascularization in Vldlr - / - mice. PSF reprogrammed mitochondrial bioenergetics and inhibited the transition of endothelial cells after hypoxia, suggesting its involvement in pathological angiogenesis.Ectopic PSF expression inhibited hypoxia-induced HIF-1α activation in the nucleus by recruiting Hakai to the PSF/HIF-1α complex, causing HIF-1α inhibition. PSF knockdown increased hypoxia-stimulated HIF-1α reactions. These hypoxia-dependent processes may play a vital role in cell metabolism, migration, and proliferation. Thus, PSF is a potential treatment target in neovascularization-associated ophthalmopathy. CONCLUSION: This is the first study showing that PSF inhibits HIF-1α via recruitment of Hakai, modulates mitochondrial oxidation and glycolysis, and downregulates VEGF expression under hypoxia. We propose a new HIF-1 α/Hakai regulatory mechanism that may play a vital role in the pathogenesis of neovascularization in ophthalmopathy. PSF-Hakai-HIF-1α signaling pathway under hypoxia condition. Schematic diagram showing that the PSF-Hakai-HIF-1α signaling pathway. Under hypoxia condition, PSF-Hakai complex regulate HIF-1α signaling, thus inhibiting downstream target gene VEGF, cell metabolism and angiogenesis eventually. Video Abstract: Detailed information of Materials and Methods.


Subject(s)
Hypoxia/metabolism , Mitochondria/metabolism , PTB-Associated Splicing Factor/metabolism , Retinal Diseases/metabolism , Animals , Cell Movement , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/physiology , Humans , Hypoxia/complications , Hypoxia/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice, Inbred C57BL , Mice, Knockout , PTB-Associated Splicing Factor/genetics , Receptors, LDL/genetics , Retina/metabolism , Retinal Diseases/etiology , Retinal Diseases/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
12.
Elife ; 102021 01 21.
Article in English | MEDLINE | ID: mdl-33476259

ABSTRACT

Circular RNAs (circRNAs) represent an abundant and conserved entity of non-coding RNAs; however, the principles of biogenesis are currently not fully understood. Here, we identify two factors, splicing factor proline/glutamine rich (SFPQ) and non-POU domain-containing octamer-binding protein (NONO), to be enriched around circRNA loci. We observe a subclass of circRNAs, coined DALI circRNAs, with distal inverted Alu elements and long flanking introns to be highly deregulated upon SFPQ knockdown. Moreover, SFPQ depletion leads to increased intron retention with concomitant induction of cryptic splicing, premature transcription termination, and polyadenylation, particularly prevalent for long introns. Aberrant splicing in the upstream and downstream regions of circRNA producing exons are critical for shaping the circRNAome, and specifically, we identify missplicing in the immediate upstream region to be a conserved driver of circRNA biogenesis. Collectively, our data show that SFPQ plays an important role in maintaining intron integrity by ensuring accurate splicing of long introns, and disclose novel features governing Alu-independent circRNA production.


Subject(s)
Introns , PTB-Associated Splicing Factor/genetics , RNA Splicing , RNA, Circular/metabolism , Animals , HEK293 Cells , Hep G2 Cells , Humans , Mice , PTB-Associated Splicing Factor/metabolism
13.
Int J Mol Sci ; 21(19)2020 Sep 28.
Article in English | MEDLINE | ID: mdl-32998269

ABSTRACT

RNA-binding proteins (RBPs) are a class of proteins known for their diverse roles in RNA biogenesis, from regulating transcriptional processes in the nucleus to facilitating translation in the cytoplasm. With higher demand for RNA metabolism in the nervous system, RBP misregulation has been linked to a wide range of neurological and neurodegenerative diseases. One of the emerging RBPs implicated in neuronal function and neurodegeneration is splicing factor proline- and glutamine-rich (SFPQ). SFPQ is a ubiquitous and abundant RBP that plays multiple regulatory roles in the nucleus such as paraspeckle formation, DNA damage repair, and various transcriptional regulation processes. An increasing number of studies have demonstrated the nuclear and also cytoplasmic roles of SFPQ in neurons, particularly in post-transcriptional regulation and RNA granule formation. Not surprisingly, the misregulation of SFPQ has been linked to pathological features shown by other neurodegenerative disease-associated RBPs such as aberrant RNA splicing, cytoplasmic mislocalization, and aggregation. In this review, we discuss recent findings on the roles of SFPQ with a particular focus on those in neuronal development and homeostasis as well as its implications in neurodegenerative diseases.


Subject(s)
Neurodegenerative Diseases/genetics , Neurons/metabolism , PTB-Associated Splicing Factor/genetics , RNA Splicing , RNA, Messenger/genetics , Animals , Binding Sites , Cell Nucleus/metabolism , Cytoplasm/metabolism , Cytoplasmic Granules/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Humans , Models, Molecular , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurons/cytology , PTB-Associated Splicing Factor/chemistry , PTB-Associated Splicing Factor/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , RNA, Messenger/metabolism
14.
Int J Mol Sci ; 21(16)2020 Aug 05.
Article in English | MEDLINE | ID: mdl-32764370

ABSTRACT

The coupling of alternative splicing with the nonsense-mediated decay (NMD) pathway maintains quality control of the transcriptome in eukaryotes by eliminating transcripts with premature termination codons (PTC) and fine-tunes gene expression. Long noncoding RNA (lncRNA) can regulate multiple cellular processes, including alternative splicing. Previously, murine Morrbid (myeloid RNA repressor of Bcl2l11 induced death) lncRNA was described as a locus-specific controller of the lifespan of short-living myeloid cells via transcription regulation of the apoptosis-related Bcl2l11 protein. Here, we report that murine Morrbid lncRNA in hepatocytes participates in the regulation of proto-oncogene NRAS (neuroblastoma RAS viral oncogene homolog) splicing, including the formation of the isoform with PTC. We observed a significant increase of the NRAS isoform with PTC in hepatocytes with depleted Morrbid lncRNA. We demonstrated that the NRAS isoform with PTC is degraded via the NMD pathway. This transcript is presented almost only in the nucleus and has a half-life ~four times lower than other NRAS transcripts. Additionally, in UPF1 knockdown hepatocytes (the key NMD factor), we observed a significant increase of the NRAS isoform with PTC. By a modified capture hybridization (CHART) analysis of the protein targets, we uncovered interactions of Morrbid lncRNA with the SFPQ (splicing factor proline and glutamine rich)-NONO (non-POU domain-containing octamer-binding protein) splicing complex. Finally, we propose the regulation mechanism of NRAS splicing in murine hepatocytes by alternative splicing coupled with the NMD pathway with the input of Morrbid lncRNA.


Subject(s)
Alternative Splicing/genetics , DNA-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/genetics , PTB-Associated Splicing Factor/genetics , RNA, Long Noncoding/genetics , RNA-Binding Proteins/genetics , Animals , Codon, Nonsense/genetics , Gene Expression Regulation, Developmental , Hepatocytes/metabolism , Mice , Multiprotein Complexes/genetics , Nonsense Mediated mRNA Decay/genetics , Transcriptome/genetics
15.
Oncogene ; 39(34): 5616-5632, 2020 08.
Article in English | MEDLINE | ID: mdl-32661324

ABSTRACT

Increasing evidence indicates that long non-coding RNAs (lncRNAs) play vital roles in the tumorigenesis and progression of cancers. However, the functions and regulatory mechanisms of lncRNAs in nasopharyngeal carcinoma (NPC) are still largely unknown. Our previous lncRNA expression profiles identified that LINC01503 was overexpressed in NPC. Here, we verified that LINC01503 was highly expressed in NPC and correlated with poor prognosis. LINC01503 promoted NPC cell proliferation, migration, and invasion in vitro, and facilitated tumor growth and metastasis in vivo. Mechanistically, LINC01503 recruited splicing factor proline-and glutamine-rich (SFPQ) to activate Fos like 1 (FOSL1) transcription, and ectopic expression of FOSL1 reversed the suppressive effect of LINC01503 knockdown on NPC progression. Moreover, androgen receptor (AR)-mediated transcription activation was responsible for the overexpression of LINC01503, and AR ligand-dependent cell growth, migration, and invasion in NPC cells. Taken together, our findings reveal that AR-induced LINC01503 can promote NPC progression through the SFPQ-FOSL1 axis, which represents a novel prognostic biomarker and therapeutic target for NPC patients.


Subject(s)
Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Neoplasms/genetics , PTB-Associated Splicing Factor/genetics , Proto-Oncogene Proteins c-fos/genetics , RNA, Long Noncoding/genetics , Receptors, Androgen/genetics , Animals , Cell Line , Cell Line, Tumor , HEK293 Cells , Humans , Kaplan-Meier Estimate , Lymphatic Metastasis , Male , Mice, Inbred BALB C , Mice, Nude , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/therapy , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/therapy , PTB-Associated Splicing Factor/metabolism , Proto-Oncogene Proteins c-fos/metabolism , RNA Interference , Receptors, Androgen/metabolism , Xenograft Model Antitumor Assays/methods
16.
Genes Chromosomes Cancer ; 59(9): 540-543, 2020 09.
Article in English | MEDLINE | ID: mdl-32306475

ABSTRACT

In recent years, a subgroup of B-cell precursor acute lymphoblastic leukemia (BCP ALL) without an established abnormality ("B-other") has been shown to be characterized by rearrangements of ABL1, ABL2, CSF1R, or PDGFRB (a.k.a. ABL-class genes). Using FISH with probes for these genes, we screened 55 pediatric and 50 adult B-other cases. Three (6%) of the adult but none of the childhood B-other cases were positive for ABL-class aberrations. RT-PCR and sequencing confirmed a rare SFPQ-ABL1 fusion in one adult B-other case with t(1;9)(p34;q34). Only six SFPQ-ABL1-positive BCP ALLs have been reported, present case included. A review of these shows that all harbored fusions between exon 9 of SFPQ and exon 4 of ABL1, that the fusion is typically found in adolescents/younger adults without hyperleukocytosis, and that IKZF1 deletions are recurrent. The few patients not treated with tyrosine kinase inhibitors (TKIs) and/or allogeneic stem cell transplantation relapsed, strengthening the notion that TKI should be added to the therapy of SFPQ-ABL1-positive BCP ALL.


Subject(s)
Oncogene Proteins, Fusion/genetics , PTB-Associated Splicing Factor/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-abl/genetics , Adolescent , Antineoplastic Agents/therapeutic use , Child , Child, Preschool , Female , Humans , Ikaros Transcription Factor/genetics , Infant , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Kinase Inhibitors/therapeutic use
17.
Cancer Res ; 80(11): 2230-2242, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32213542

ABSTRACT

Endocrine therapy is standard treatment for estrogen receptor (ER)-positive breast cancer, yet long-term treatment often causes acquired resistance, which results in recurrence and metastasis. Recent studies have revealed that RNA-binding proteins (RBP) are involved in tumorigenesis. Here, we demonstrate that PSF/SFPQ is an RBP that potentially predicts poor prognosis of patients with ER-positive breast cancer by posttranscriptionally regulating ERα (ESR1) mRNA expression. Strong PSF immunoreactivity correlated with shorter overall survival in patients with ER-positive breast cancer. PSF was predominantly expressed in a model of tamoxifen-resistant breast cancer cells, and depletion of PSF attenuated proliferation of cultured cells and xenografted tumors. PSF expression was significantly associated with estrogen signaling. PSF siRNA downregulated ESR1 mRNA by inhibiting nuclear export of the RNA. Integrative analyses of microarray and RNA immunoprecipitation sequencing also identified SCFD2, TRA2B, and ASPM as targets of PSF. Among the PSF targets, SCFD2 was a poor prognostic indicator of breast cancer and SCFD2 knockdown significantly suppressed breast cancer cell proliferation. Collectively, this study shows that PSF plays a pathophysiologic role in ER-positive breast cancer by posttranscriptionally regulating expression of its target genes such as ESR1 and SCFD2. Overall, PSF and SCFD2 could be potential diagnostic and therapeutic targets for primary and hormone-refractory breast cancers. SIGNIFICANCE: This study defines oncogenic roles of RNA-binding protein PSF, which exhibits posttranscriptional regulation in ER-positive breast cancer.


Subject(s)
Breast Neoplasms/genetics , Estrogen Receptor alpha/genetics , PTB-Associated Splicing Factor/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Disease Progression , Drug Resistance, Neoplasm , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , PTB-Associated Splicing Factor/metabolism , Prognosis , RNA Processing, Post-Transcriptional , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/metabolism , Tamoxifen/pharmacology
18.
Nucleic Acids Res ; 48(6): 3356-3365, 2020 04 06.
Article in English | MEDLINE | ID: mdl-32034402

ABSTRACT

SFPQ is a ubiquitous nuclear RNA-binding protein implicated in many aspects of RNA biogenesis. Importantly, nuclear depletion and cytoplasmic accumulation of SFPQ has been linked to neuropathological conditions such as Alzheimer's disease (AD) and amyotrophic lateral sclerosis (ALS). Here, we describe a molecular mechanism by which SFPQ is mislocalized to the cytoplasm. We report an unexpected discovery of the infinite polymerization of SFPQ that is induced by zinc binding to the protein. The crystal structure of human SFPQ in complex with zinc at 1.94 Å resolution reveals intermolecular interactions between SFPQ molecules that are mediated by zinc. As anticipated from the crystal structure, the application of zinc to primary cortical neurons induced the cytoplasmic accumulation and aggregation of SFPQ. Mutagenesis of the three zinc-coordinating histidine residues resulted in a significant reduction in the zinc-binding affinity of SFPQ in solution and the zinc-induced cytoplasmic aggregation of SFPQ in cultured neurons. Taken together, we propose that dysregulation of zinc availability and/or localization in neuronal cells may represent a mechanism for the imbalance in the nucleocytoplasmic distribution of SFPQ, which is an emerging hallmark of neurodegenerative diseases including AD and ALS.


Subject(s)
Neurons/metabolism , PTB-Associated Splicing Factor/ultrastructure , RNA-Binding Proteins/ultrastructure , RNA/genetics , Alzheimer Disease/genetics , Amyotrophic Lateral Sclerosis/genetics , Cell Nucleus/genetics , Crystallography, X-Ray , Cytoplasm/genetics , Humans , Neurons/pathology , PTB-Associated Splicing Factor/chemistry , PTB-Associated Splicing Factor/genetics , Polymerization , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Zinc/metabolism
19.
Biochem J ; 477(2): 509-524, 2020 01 31.
Article in English | MEDLINE | ID: mdl-31930351

ABSTRACT

DNA hemicatenanes (HCs) are four-way junctions in which one strand of a double-stranded helix is catenated with one strand of another double-stranded DNA. Frequently mentioned as DNA replication, recombination and repair intermediates, they have been proposed to participate in the spatial organization of chromosomes and in the regulation of gene expression. To explore potential roles of HCs in genome metabolism, we sought to purify proteins capable of binding specifically HCs by fractionating nuclear extracts from HeLa cells. This approach identified three RNA-binding proteins: the Tudor-staphylococcal nuclease domain 1 (SND1) protein and two proteins from the Drosophila behavior human splicing family, the paraspeckle protein component 1 and the splicing factor proline- and glutamine-rich protein. Since these proteins were partially pure after fractionation, truncated forms of these proteins were expressed in Escherichia coli and purified to near homogeneity. The specificity of their interaction with HCs was re-examined in vitro. The two truncated purified SND1 proteins exhibited specificity for HCs, opening the interesting possibility of a link between the basic transcription machinery and HC structures via SND1.


Subject(s)
Catenanes/metabolism , DNA/genetics , Endonucleases/genetics , Transcription, Genetic , Animals , Catenanes/chemistry , Chromosomes/genetics , DNA Replication/genetics , DNA-Binding Proteins/genetics , Endonucleases/metabolism , HeLa Cells , Humans , PTB-Associated Splicing Factor/genetics , Protein Binding/genetics , RNA-Binding Proteins/genetics , Recombination, Genetic/genetics
20.
Genes Cells ; 24(11): 719-730, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31541592

ABSTRACT

Amyloid precursor protein (APP) is a representative gene related to Alzheimer's disease (AD). Androgens function by binding to the androgen receptor (AR). Both androgen and RNA-binding protein PSF play a role in the pathology of AD. However, the involvement of AR and PSF in APP regulation in neuron has not been investigated. Here, we explored the regulatory mechanism of APP expression by AR and PSF using neuron-derived cells. We demonstrated that androgen up-regulates the production of APP at the mRNA and protein levels. This induction is enhanced by AR over-expression and inhibited by its silencing. One candidate AR-binding region was identified in the intron region of APP and validated its activity as AR-dependent enhancer by the luciferase assay. Furthermore, the public transcriptome data of brain tissues of mice indicated that APP is regulated by PSF post-transcriptionally. We observed a decreased expression of APP after PSF knockdown and interaction of PSF with the APP transcript. Moreover, we revealed that silencing of PSF inhibited the stability of the APP mRNA. Thus, these results presented a new regulatory mechanism of APP expression by androgen through AR-mediated transcription and PSF at the post-transcriptional level that might be associated with the occurrence of AD.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Androgens/genetics , Androgens/metabolism , Neurons/metabolism , RNA-Binding Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Brain/metabolism , Brain/pathology , Cell Line , Disease Models, Animal , Gene Knockdown Techniques , Humans , Mice , PTB-Associated Splicing Factor/genetics , Protein Binding , RNA, Messenger/metabolism , Receptors, Androgen , Transcriptome , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...