Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Cell Death Dis ; 15(8): 616, 2024 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-39183343

RESUMEN

Glioblastoma (GBM) represents a primary malignant brain tumor. Temozolomide resistance is a major hurdle in GBM treatment. Proteins encoded by circular RNAs (circRNAs) can modulate the sensitivity of multiple tumor chemotherapies. However, the impact of circRNA-encoded proteins on GBM sensitivity to temozolomide remains unknown. Herein, we discover a circRNA (circCOPA) through the circRNA microarray profile in GBM samples, which can encode a novel 99 amino acid protein (COPA-99aa) through its internal ribosome entry site. Functionally, circCOPA overexpression in GBM cells inhibits cell proliferation, migration, and invasion in vitro and growth in vivo. Rather than itself, circCOPA mainly functions as a suppressive effector by encoding COPA-99aa. Moreover, we reveal that circCOPA is downregulated in GBM tissues and high expression of circCOPA is related to a better prognosis in GBM patients. Mechanistically, a heteromer of SFPQ and NONO is required for double-strand DNA break repair. COPA-99aa disrupts the dimerization of NONO and SFPQ by separately binding with the NONO and SFPQ proteins, thus resulting in the inhibition of proliferation or invasion and the increase of temozolomide-induced DNA damage in GBM cells. Collectively, our data suggest that circCOPA mainly contributes to inhibiting the GBM malignant phenotype through its encoded COPA-99aa and that COPA-99aa increases temozolomide-induced DNA damage by interfering with the dimerization of NONO and SFPQ. Restoring circCOPA or COPA-99aa may increase the sensitivity of patients to temozolomide.


Asunto(s)
Neoplasias Encefálicas , Proliferación Celular , Glioblastoma , ARN Circular , Proteínas de Unión al ARN , Temozolomida , Temozolomida/farmacología , Temozolomida/uso terapéutico , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , ARN Circular/genética , ARN Circular/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Proliferación Celular/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/metabolismo , Animales , Factor de Empalme Asociado a PTB/metabolismo , Factor de Empalme Asociado a PTB/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Fenotipo , Movimiento Celular/efectos de los fármacos , Ratones , Reparación del ADN/efectos de los fármacos , Ratones Desnudos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Antineoplásicos Alquilantes/farmacología
2.
Oncogene ; 43(28): 2199-2214, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38802648

RESUMEN

The MUC1 gene evolved in mammals for adaptation of barrier tissues in response to infections and damage. Paraspeckles are nuclear bodies formed on the NEAT1 lncRNA in response to loss of homeostasis. There is no known intersection of MUC1 with NEAT1 or paraspeckles. Here, we demonstrate that the MUC1-C subunit plays an essential role in regulating NEAT1 expression. MUC1-C activates the NEAT1 gene with induction of the NEAT1_1 and NEAT1_2 isoforms by NF-κB- and MYC-mediated mechanisms. MUC1-C/MYC signaling also induces expression of the SFPQ, NONO and FUS RNA binding proteins (RBPs) that associate with NEAT1_2 and are necessary for paraspeckle formation. MUC1-C integrates activation of NEAT1 and RBP-encoding genes by recruiting the PBAF chromatin remodeling complex and increasing chromatin accessibility of their respective regulatory regions. We further demonstrate that MUC1-C and NEAT1 form an auto-inductive pathway that drives common sets of genes conferring responses to inflammation and loss of homeostasis. Of functional significance, we find that the MUC1-C/NEAT1 pathway is of importance for the cancer stem cell (CSC) state and anti-cancer drug resistance. These findings identify a previously unrecognized role for MUC1-C in the regulation of NEAT1, RBPs, and paraspeckles that has been co-opted in promoting cancer progression.


Asunto(s)
Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Mucina-1 , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , Mucina-1/genética , Mucina-1/metabolismo , Animales , Línea Celular Tumoral , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Ratones , Neoplasias/genética , Neoplasias/patología , Neoplasias/metabolismo , FN-kappa B/metabolismo , FN-kappa B/genética , Factor de Empalme Asociado a PTB/genética , Factor de Empalme Asociado a PTB/metabolismo , Transducción de Señal/genética , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteína FUS de Unión a ARN/genética , Proteína FUS de Unión a ARN/metabolismo , Proteínas de Unión al ADN
3.
Theriogenology ; 225: 107-118, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38805993

RESUMEN

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.


Asunto(s)
Exosomas , Líquido Folicular , Técnicas de Maduración In Vitro de los Oocitos , Sistema de Señalización de MAP Quinasas , MicroARNs , Oocitos , Animales , Porcinos , MicroARNs/metabolismo , MicroARNs/genética , Oocitos/metabolismo , Oocitos/fisiología , Técnicas de Maduración In Vitro de los Oocitos/veterinaria , Exosomas/metabolismo , Femenino , Líquido Folicular/metabolismo , Factor de Empalme Asociado a PTB/metabolismo , Factor de Empalme Asociado a PTB/genética , Regulación de la Expresión Génica
4.
Nat Commun ; 15(1): 4156, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38755141

RESUMEN

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.


Asunto(s)
Herpesvirus Humano 4 , Histonas , Factor de Empalme Asociado a PTB , Activación Viral , Latencia del Virus , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/fisiología , Humanos , Histonas/metabolismo , Activación Viral/genética , Latencia del Virus/genética , Factor de Empalme Asociado a PTB/metabolismo , Factor de Empalme Asociado a PTB/genética , Regulación Viral de la Expresión Génica , Linfocitos B/virología , Linfocitos B/metabolismo , Infecciones por Virus de Epstein-Barr/virología , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/metabolismo , Sistemas CRISPR-Cas , Regiones Promotoras Genéticas/genética , Transactivadores/metabolismo , Transactivadores/genética , Genoma Viral
5.
Neuron ; 112(15): 2558-2580.e13, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-38761794

RESUMEN

Neurodegenerative diseases are commonly classified as proteinopathies that are defined by the aggregation of a specific protein. Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are classified as synucleinopathies since α-synuclein (α-syn)-containing inclusions histopathologically define these diseases. Unbiased biochemical analysis of PD and DLB patient material unexpectedly revealed novel pathological inclusions in the nucleus comprising adenosine-to-inosine (A-to-I)-edited mRNAs and NONO and SFPQ proteins. These inclusions showed no colocalization with Lewy bodies and accumulated at levels comparable to α-syn. NONO and SFPQ aggregates reduced the expression of the editing inhibitor ADAR3, increasing A-to-I editing mainly within human-specific, Alu-repeat regions of axon, synaptic, and mitochondrial transcripts. Inosine-containing transcripts aberrantly accumulated in the nucleus, bound tighter to recombinant purified SFPQ in vitro, and potentiated SFPQ aggregation in human dopamine neurons, resulting in a self-propagating pathological state. Our data offer new insight into the inclusion composition and pathophysiology of PD and DLB.


Asunto(s)
Enfermedad por Cuerpos de Lewy , Factor de Empalme Asociado a PTB , Enfermedad de Parkinson , Edición de ARN , Humanos , Enfermedad por Cuerpos de Lewy/metabolismo , Enfermedad por Cuerpos de Lewy/patología , Enfermedad por Cuerpos de Lewy/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Factor de Empalme Asociado a PTB/metabolismo , Factor de Empalme Asociado a PTB/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Inosina/metabolismo , Adenosina/metabolismo , Núcleo Celular/metabolismo , Masculino , Anciano , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Femenino , ARN Mensajero/metabolismo , alfa-Sinucleína/metabolismo , alfa-Sinucleína/genética , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Anciano de 80 o más Años
6.
Biochimie ; 222: 9-17, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38373651

RESUMEN

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.


Asunto(s)
Integrasa de VIH , VIH-1 , Factor de Empalme Asociado a PTB , Integración Viral , Replicación Viral , VIH-1/metabolismo , VIH-1/fisiología , VIH-1/genética , Humanos , Integrasa de VIH/metabolismo , Integrasa de VIH/genética , Factor de Empalme Asociado a PTB/metabolismo , Factor de Empalme Asociado a PTB/genética , Unión Proteica , Mutación , Células HEK293
7.
Medicine (Baltimore) ; 102(45): e35837, 2023 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-37960731

RESUMEN

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.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Regiones no Traducidas 3' , Antígeno B7-H1/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/terapia , Carcinoma de Pulmón de Células no Pequeñas/patología , Glutamina , Leucocitos Mononucleares/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Neoplasias Pulmonares/metabolismo , Factores de Empalme de ARN/genética , Linfocitos T Citotóxicos/metabolismo , Factor de Empalme Asociado a PTB/genética , Factor de Empalme Asociado a PTB/metabolismo
8.
Biochemistry ; 61(17): 1723-1734, 2022 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-35998361

RESUMEN

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.


Asunto(s)
ARN Largo no Codificante , Proteínas de Unión al ARN , Animales , Humanos , Ratones , Factor de Empalme Asociado a PTB/genética , Factor de Empalme Asociado a PTB/metabolismo , Empalme del ARN , Factores de Empalme de ARN/genética , Factores de Empalme de ARN/metabolismo , ARN Largo no Codificante/metabolismo , Proteínas de Unión al ARN/metabolismo
10.
Cancer Res ; 81(13): 3495-3508, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33975881

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Epigénesis Genética , Factor de Empalme Asociado a PTB/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Procesamiento Postranscripcional del ARN , ARN Largo no Codificante/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Apoptosis , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Factor de Empalme Asociado a PTB/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , ARN Largo no Codificante/genética , Transcripción Genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
J Mol Med (Berl) ; 99(7): 967-980, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33770188

RESUMEN

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.


Asunto(s)
Factor de Empalme Asociado a PTB/metabolismo , Neovascularización Retiniana/metabolismo , Factor de Transcripción Activador 4/metabolismo , Aldehídos/farmacología , Animales , Células Cultivadas , Estrés del Retículo Endoplásmico , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Factor 2 Eucariótico de Iniciación/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hemo-Oxigenasa 1/metabolismo , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Microvasos/citología , Mitocondrias/metabolismo , Factor de Empalme Asociado a PTB/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de LDL/genética , Retina/citología , Retina/metabolismo , Retina/patología , Neovascularización Retiniana/genética , eIF-2 Quinasa/metabolismo
12.
Nat Commun ; 12(1): 1918, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33771997

RESUMEN

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.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Codón sin Sentido , Exones/genética , Factor de Empalme Asociado a PTB/genética , Animales , Secuencia de Bases , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Hibridación in Situ/métodos , Intrones/genética , Ratones , Neuronas/metabolismo , Pez Cebra/embriología , Pez Cebra/genética
13.
Hum Mol Genet ; 30(11): 971-984, 2021 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-33729478

RESUMEN

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.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Ciclinas/genética , Demencia Frontotemporal/genética , Factor de Empalme Asociado a PTB/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Proteínas de Unión al ADN/genética , Demencia Frontotemporal/metabolismo , Demencia Frontotemporal/patología , Células HEK293 , Humanos , Agregado de Proteínas/genética , Mapas de Interacción de Proteínas/genética , Proteolisis , ARN/genética , ARN/metabolismo , Procesamiento Postranscripcional del ARN/genética , Proteínas de Unión al ARN/genética
14.
Cell Commun Signal ; 19(1): 14, 2021 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-33573690

RESUMEN

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.


Asunto(s)
Hipoxia/metabolismo , Mitocondrias/metabolismo , Factor de Empalme Asociado a PTB/metabolismo , Enfermedades de la Retina/metabolismo , Animales , Movimiento Celular , Células Cultivadas , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Humanos , Hipoxia/complicaciones , Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Empalme Asociado a PTB/genética , Receptores de LDL/genética , Retina/metabolismo , Enfermedades de la Retina/etiología , Enfermedades de la Retina/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Elife ; 102021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33476259

RESUMEN

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.


Asunto(s)
Intrones , Factor de Empalme Asociado a PTB/genética , Empalme del ARN , ARN Circular/metabolismo , Animales , Células HEK293 , Células Hep G2 , Humanos , Ratones , Factor de Empalme Asociado a PTB/metabolismo
16.
Int J Mol Sci ; 21(19)2020 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-32998269

RESUMEN

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.


Asunto(s)
Enfermedades Neurodegenerativas/genética , Neuronas/metabolismo , Factor de Empalme Asociado a PTB/genética , Empalme del ARN , ARN Mensajero/genética , Animales , Sitios de Unión , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Gránulos Citoplasmáticos/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Humanos , Modelos Moleculares , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuronas/citología , Factor de Empalme Asociado a PTB/química , Factor de Empalme Asociado a PTB/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , ARN Mensajero/metabolismo
17.
Int J Mol Sci ; 21(16)2020 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-32764370

RESUMEN

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.


Asunto(s)
Empalme Alternativo/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al GTP Monoméricas/genética , Factor de Empalme Asociado a PTB/genética , ARN Largo no Codificante/genética , Proteínas de Unión al ARN/genética , Animales , Codón sin Sentido/genética , Regulación del Desarrollo de la Expresión Génica , Hepatocitos/metabolismo , Ratones , Complejos Multiproteicos/genética , Degradación de ARNm Mediada por Codón sin Sentido/genética , Transcriptoma/genética
18.
Oncogene ; 39(34): 5616-5632, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32661324

RESUMEN

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.


Asunto(s)
Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Carcinoma Nasofaríngeo/genética , Neoplasias Nasofaríngeas/genética , Factor de Empalme Asociado a PTB/genética , Proteínas Proto-Oncogénicas c-fos/genética , ARN Largo no Codificante/genética , Receptores Androgénicos/genética , Animales , Línea Celular , Línea Celular Tumoral , Células HEK293 , Humanos , Estimación de Kaplan-Meier , Metástasis Linfática , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Carcinoma Nasofaríngeo/metabolismo , Carcinoma Nasofaríngeo/terapia , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/terapia , Factor de Empalme Asociado a PTB/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Interferencia de ARN , Receptores Androgénicos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
19.
Genes Chromosomes Cancer ; 59(9): 540-543, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32306475

RESUMEN

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.


Asunto(s)
Proteínas de Fusión Oncogénica/genética , Factor de Empalme Asociado a PTB/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas Proto-Oncogénicas c-abl/genética , Adolescente , Antineoplásicos/uso terapéutico , Niño , Preescolar , Femenino , Humanos , Factor de Transcripción Ikaros/genética , Lactante , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Inhibidores de Proteínas Quinasas/uso terapéutico
20.
Cancer Res ; 80(11): 2230-2242, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32213542

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/genética , Receptor alfa de Estrógeno/genética , Factor de Empalme Asociado a PTB/genética , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Receptor alfa de Estrógeno/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Xenoinjertos , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Factor de Empalme Asociado a PTB/metabolismo , Pronóstico , Procesamiento Postranscripcional del ARN , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Factores de Empalme Serina-Arginina/genética , Factores de Empalme Serina-Arginina/metabolismo , Tamoxifeno/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA