Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(29): e2117090119, 2022 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35858306

RESUMEN

Retinal photoreceptors have a distinct transcriptomic profile compared to other neuronal subtypes, likely reflecting their unique cellular morphology and function in the detection of light stimuli by way of the ciliary outer segment. We discovered a layer of this molecular specialization by revealing that the vertebrate retina expresses the largest number of tissue-enriched microexons of all tissue types. A subset of these microexons is included exclusively in photoreceptor transcripts, particularly in genes involved in cilia biogenesis and vesicle-mediated transport. This microexon program is regulated by Srrm3, a paralog of the neural microexon regulator Srrm4. Despite the fact that both proteins positively regulate retina microexons in vitro, only Srrm3 is highly expressed in mature photoreceptors. Its deletion in zebrafish results in widespread down-regulation of microexon inclusion from early developmental stages, followed by other transcriptomic alterations, severe photoreceptor defects, and blindness. These results shed light on the transcriptomic specialization and functionality of photoreceptors, uncovering unique cell type-specific roles for Srrm3 and microexons with implications for retinal diseases.


Asunto(s)
Proteínas , Segmento Externo de las Células Fotorreceptoras Retinianas , Factores de Empalme Serina-Arginina , Visión Ocular , Animales , Exones , Eliminación de Gen , Humanos , Proteínas/genética , Proteínas/fisiología , Segmento Externo de las Células Fotorreceptoras Retinianas/metabolismo , Factores de Empalme Serina-Arginina/genética , Factores de Empalme Serina-Arginina/fisiología , Transcriptoma , Visión Ocular/genética , Visión Ocular/fisiología , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/genética
2.
J Clin Invest ; 131(22)2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34779408

RESUMEN

The tumorigenic mechanism for pancreatic ductal adenocarcinoma (PDAC) is not clear, although chronic inflammation is implicated. Here, we identified an inflammatory cytokine-regulated transfer RNA-derived (tRNA-derived) fragment, tRF-21-VBY9PYKHD (tRF-21), as a tumor suppressor in PDAC progression. We found that the biogenesis of tRF-21 could be inhibited by leukemia inhibitory factor and IL-6 via the splicing factor SRSF5. Reduced tRF-21 promoted AKT2/1-mediated heterogeneous nuclear ribonucleoprotein L (hnRNP L) phosphorylation, enhancing hnRNP L to interact with dead-box helicase 17 (DDX17) to form an alternative splicing complex. The provoked hnRNP L-DDX17 activity preferentially spliced Caspase 9 and mH2A1 pre-mRNAs to form Caspase 9b and mH2A1.2, promoting PDAC cell malignant phenotypes. The tRF-21 levels were significantly lower in PDACs than in normal tissues, and patients with low tRF-21 levels had a poor prognosis. Treatment of mouse PDAC xenografts or patient-derived xenografts (PDXs) with tRF-21 mimics repressed tumor growth and metastasis. These results demonstrate that tRF-21 has a tumor-suppressive effect and is a potential therapeutic agent for PDAC.


Asunto(s)
Carcinoma Ductal Pancreático/prevención & control , Citocinas/fisiología , Neoplasias Pancreáticas/prevención & control , ARN de Transferencia/fisiología , Empalme Alternativo , Carcinoma Ductal Pancreático/genética , Línea Celular Tumoral , ARN Helicasas DEAD-box/metabolismo , Progresión de la Enfermedad , Humanos , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas c-akt/fisiología , Ribonucleoproteínas/metabolismo , Factores de Empalme Serina-Arginina/fisiología , Proteínas Supresoras de Tumor , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cell Death Dis ; 12(6): 587, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099633

RESUMEN

Long noncoding RNAs (lncRNAs) emerge as essential roles in the regulation of alternative splicing (AS) in various malignancies. Serine- and arginine-rich splicing factor 1 (SRSF1)-mediated AS events are the most important molecular hallmarks in cancer. Nevertheless, the biological mechanism underlying tumorigenesis of lncRNAs correlated with SRSF1 in esophageal squamous cell carcinoma (ESCC) remains elusive. In this study, we found that lncRNA DiGeorge syndrome critical region gene 5 (DGCR5) was upregulated in ESCC clinical samples, which associated with poor prognosis. Through RNA interference and overexpression approaches, we confirmed that DGCR5 contributed to promote ESCC cell proliferation, migration, and invasion while inhibited apoptosis in vitro. Mechanistically, DGCR5 could directly bind with SRSF1 to increase its stability and thus stimulate alternative splicing events. Furthermore, we clarified that SRSF1 regulated the aberrant splicing of myeloid cell leukemia-1 (Mcl-1) and initiated a significant Mcl-1L (antiapoptotic) isoform switch, which contributed to the expression of the full length of Mcl-1. Moreover, the cell-derived xenograft (CDX) model was validated that DGCR5 could facilitate the tumorigenesis of ESCC in vivo. Collectively, our findings identified that the key biological role of lncRNA DGCR5 in alternative splicing regulation and emphasized DGCR5 as a potential biomarker and therapeutic target for ESCC.


Asunto(s)
Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas de Esófago/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , ARN Largo no Codificante/fisiología , Factores de Empalme Serina-Arginina/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Empalme Alternativo/genética , Animales , Apoptosis/genética , Carcinogénesis/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Células Tumorales Cultivadas
4.
Biochim Biophys Acta Mol Cell Res ; 1868(7): 119045, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33872670

RESUMEN

PGAM5 is a protein phosphatase located in the inner mitochondrial membrane through its transmembrane (TM) domain and is cleaved within the TM domain upon mitochondrial dysfunction. We found previously that cleaved PGAM5 is released from mitochondria, following proteasome-mediated rupture of the outer mitochondrial membrane during mitophagy, a selective form of autophagy specific to mitochondria. Here, we examined the role of cleaved PGAM5 outside mitochondria. Deletion mutants that mimic cleaved PGAM5 existed not only in the cytosol but also in the nucleus, and a fraction of cleaved PGAM5 translocated to the nucleus during mitophagy induced by the uncoupler CCCP. We identified serine/arginine-related nuclear matrix protein of 160 kDa (SRm160)/SRRM1, which contains a highly phosphorylated domain rich in arginine/serine dipeptides, called the RS domain, as a nuclear protein that interacts with PGAM5. PGAM5 dephosphorylated SRm160, and incubation of lysates from WT cells, but not of those from PGAM5-deficient cells, induced dephosphorylation of SRm160 and another RS domain-containing protein SRSF1, one of the most characterized serine/arginine-rich (SR) proteins. Moreover, phosphorylation of these proteins and other SR proteins, which are commonly reactive toward the 1H4 monoclonal antibody that detects phosphorylated SR proteins, decreased during mitophagy, largely because of PGAM5 activity. These results suggest that PGAM5 regulates phosphorylation of these nuclear proteins during mitophagy. Because SRm160 and SR proteins play critical roles in mRNA metabolism, PGAM5 may coordinate cellular responses to mitochondrial stress at least in part through post-transcriptional and pre-translational events.


Asunto(s)
Proteínas Mitocondriales/metabolismo , Mitofagia/genética , Fosfoproteínas Fosfatasas/metabolismo , Factores de Empalme Serina-Arginina/metabolismo , Antígenos Nucleares/metabolismo , Núcleo Celular/metabolismo , Citosol/metabolismo , Células HeLa , Humanos , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/genética , Mitofagia/fisiología , Proteínas Asociadas a Matriz Nuclear/metabolismo , Fosfoproteínas Fosfatasas/genética , Fosforilación , Proteínas de Unión al ARN/metabolismo , Factores de Empalme Serina-Arginina/fisiología , Ubiquitina-Proteína Ligasas/metabolismo
5.
FASEB J ; 35(5): e21544, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33819356

RESUMEN

Serine-rich splicing factor 3 (SRSF3) was recently reported as being necessary to preserve RNA stability via an mTOR mechanism in a cardiac mouse model in adulthood. Here, we demonstrate the link between Srsf3 and mitochondrial integrity in an embryonic cardiomyocyte-specific Srsf3 conditional knockout (cKO) mouse model. Fifteen-day-old Srsf3 cKO mice showed dramatically reduced (below 50%) survival and reduced the left ventricular systolic performance, and histological analysis of these hearts revealed a significant increase in cardiomyocyte size, confirming the severe remodeling induced by Srsf3 deletion. RNA-seq analysis of the hearts of 5-day-old Srsf3 cKO mice revealed early changes in expression levels and alternative splicing of several transcripts related to mitochondrial integrity and oxidative phosphorylation. Likewise, the levels of several protein complexes of the electron transport chain decreased, and mitochondrial complex I-driven respiration of permeabilized cardiac muscle fibers from the left ventricle was impaired. Furthermore, transmission electron microscopy analysis showed disordered mitochondrial length and cristae structure. Together with its indispensable role in the physiological maintenance of mouse hearts, these results highlight the previously unrecognized function of Srsf3 in regulating the mitochondrial integrity.


Asunto(s)
Regulación de la Expresión Génica , Mitocondrias Cardíacas/patología , Miocitos Cardíacos/patología , Factores de Empalme Serina-Arginina/fisiología , Empalme Alternativo , Animales , Perfilación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias Cardíacas/metabolismo , Miocitos Cardíacos/metabolismo , Fosforilación Oxidativa , RNA-Seq
6.
Am J Hematol ; 96(4): 462-470, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33502020

RESUMEN

The ASXL1 and SRSF2 mutations in AML are frequently found in patients with preexisting myeloid malignancies and are individually associated with poor outcomes. In this multi-institutional retrospective analysis, we assessed the genetic features and clinical outcomes of 43 patients with ASXL1mut SRSF2mut AML and compared outcomes to patients with either ASXL1 (n = 57) or SRSF2 (n = 70) mutations. Twenty-six (60%) had secondary-AML (s-AML). Variant allele fractions suggested that SRSF2 mutations preceded ASXL1 mutational events. Median overall survival (OS) was 7.0 months (95% CI:3.8,15.3) and was significantly longer in patients with de novo vs s-AML (15.3 vs 6.4 months, respectively; P = .04 on adjusted analysis). Compared to ASXL1mut SRSF2wt and ASXL1wt SRSF2mut , co-mutated patients had a 1.4 and 1.6 times increase in the probability of death, respectively (P = .049), with a trend towards inferior OS (median OS = 7.0 vs 11.5 vs 10.9 months, respectively; P = .10). Multivariable analysis suggests this difference in OS is attributable to the high proportion of s-AML patients in the co-mutated cohort (60% vs 32% and 23%, respectively). Although this study is limited by the retrospective data collection and the relatively small sample size, these data suggest that ASXL1mut SRSF2mut AML is a distinct subgroup of AML frequently associated with s-AML and differs from ASXL1mut SRSF2wt /ASXL1wt SRSF2mut with respect to etiology and leukemogenesis.


Asunto(s)
Leucemia Mieloide Aguda/genética , Mutación , Proteínas Represoras/genética , Factores de Empalme Serina-Arginina/genética , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Transformación Celular Neoplásica/genética , Cocarcinogénesis/genética , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Estimación de Kaplan-Meier , Leucemia Mieloide Aguda/clasificación , Leucemia Mieloide Aguda/mortalidad , Masculino , Persona de Mediana Edad , Pronóstico , Proteínas Represoras/fisiología , Estudios Retrospectivos , Factores de Empalme Serina-Arginina/fisiología
7.
Oncogene ; 39(22): 4390-4403, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32332923

RESUMEN

In epithelial ovarian cancer (EOC), response to platinum (PT)-based chemotherapy dictates subsequent treatments and predicts patients' prognosis. Alternative splicing is often deregulated in human cancers and can be altered by chemotherapy. Whether and how changes in alternative splicing regulation could impact on the response of EOC to PT-based chemotherapy is still not clarified. We identified the splicing factor proline and glutamine rich (SFPQ) as a critical mediator of response to PT in an unbiased functional genomic screening in EOC cells and, using a large cohort of primary and recurrent EOC samples, we observed that it is frequently overexpressed in recurrent PT-treated samples and that its overexpression correlates with PT resistance. At mechanistic level, we show that, under PT treatment, SFPQ, in complex with p54nrb, binds and regulates the activity of the splicing factor SRSF2. SFPQ/p54nrb complex decreases SRSF2 binding to caspase-9 RNA, favoring the expression of its alternative spliced antiapoptotic form. As a consequence, SFPQ/p54nrb protects cells from PT-induced death, eventually contributing to chemoresistance. Overall, our work unveils a previously unreported SFPQ/p54nrb/SRSF2 pathway that in EOC cells plays a central role in regulating alternative splicing and PT-induced apoptosis and that could result in the design of new possible ways of intervention to overcome PT resistance.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Cisplatino/farmacología , Proteínas de Unión al ADN/fisiología , Proteínas de Neoplasias/fisiología , Neoplasias Ováricas/tratamiento farmacológico , Factor de Empalme Asociado a PTB/fisiología , Proteínas de Unión al ARN/fisiología , Factores de Empalme Serina-Arginina/fisiología , Animales , Antineoplásicos Alquilantes/uso terapéutico , Apoptosis , Caspasa 8/metabolismo , Caspasa 9/genética , Caspasa 9/metabolismo , Inhibidores de Caspasas/farmacología , Línea Celular Tumoral , Cisplatino/uso terapéutico , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Neoplasias Ováricas/metabolismo , Empalme del ARN , ARN Mensajero/metabolismo , ARN Neoplásico/metabolismo , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Recurrencia , Empalmosomas/metabolismo
8.
Mol Immunol ; 119: 69-82, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31986311

RESUMEN

SRSF1, a member of the SR protein family, is an important splicing factor and regulator of splicing. Multiple splicing isoforms have been reported for this gene. SRSF1-3, a splicing isoform of SRSF1, is necessary for AID-dependent SHM of IgV genes. However, its precise role in SHM remains enigmatic. Transcriptomic analysis of SRSF1-3 reconstituted cells shows upregulation of transcription factor SATB2 and chromatin regulator UBN1. The increased SATB2 and UBN1 are strikingly enriched in the MAR and promoter regions of the IgL gene, respectively. Furthermore, UBN1 enrichment at the promoter region was coupled with a hundred-fold enhanced occupancy of the histone variant H3.3 at the IgL promoter, that is a hallmark of efficient SHM. The enhanced occupancy of SATB2 at the MAR, UBN1 and histone variant H3.3 at the IgL promoter leads to an increase in IgL transcription, revealing a role of SRSF1-3 in SHM. Thus, SRSF1-3 is likely involved in the regulation of SHM, via upregulation of a crucial transcription factor SATB2, as well as, by overexpression of a chromatin modulator of Ig genes, UBN1, which further assists in the recruitment of the histone variant H3.3. Furthermore, the splicing isoform SRSF1-3 regulates alternate splicing pattern of splicing isoforms for various crucial genes. The present study provides the first evidence that a splicing isoform of an SR protein can regulate the post-transcriptional processing of RNA in vivo.


Asunto(s)
Regulación de la Expresión Génica , Genes de Inmunoglobulinas , Histonas/fisiología , Región Variable de Inmunoglobulina/genética , Empalme del ARN/fisiología , Factores de Empalme Serina-Arginina/fisiología , Factores de Transcripción/fisiología , Empalme Alternativo , Animales , Linfocitos B/fisiología , Línea Celular , Pollos , Activación Transcripcional
9.
J Clin Invest ; 129(12): 5411-5423, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31487268

RESUMEN

Systemic lupus erythematosus (SLE) is a devastating autoimmune disease in which hyperactive T cells play a critical role. Understanding molecular mechanisms underlying the T cell hyperactivity will lead to identification of specific therapeutic targets. Serine/arginine-rich splicing factor 1 (SRSF1) is an essential RNA-binding protein that controls posttranscriptional gene expression. We have demonstrated that SRSF1 levels are aberrantly decreased in T cells from patients with SLE and that they correlate with severe disease, yet the role of SRSF1 in T cell physiology and autoimmune disease is largely unknown. Here we show that T cell-restricted Srsf1-deficient mice develop systemic autoimmunity and lupus-nephritis. Mice exhibit increased frequencies of activated/effector T cells producing proinflammatory cytokines, and an elevated T cell activation gene signature. Mechanistically, we noted increased activity of the mechanistic target of rapamycin (mTOR) pathway and reduced expression of its repressor PTEN. The mTOR complex 1 (mTORC1) inhibitor rapamycin suppressed proinflammatory cytokine production by T cells and alleviated autoimmunity in Srsf1-deficient mice. Of direct clinical relevance, PTEN levels correlated with SRSF1 in T cells from patients with SLE, and SRSF1 overexpression rescued PTEN and suppressed mTORC1 activation and proinflammatory cytokine production. Our studies reveal the role of a previously unrecognized molecule, SRSF1, in restraining T cell activation, averting the development of autoimmune disease, and acting as a potential therapeutic target for lupus.


Asunto(s)
Autoinmunidad , Factores de Empalme Serina-Arginina/fisiología , Linfocitos T/inmunología , Animales , Citocinas/biosíntesis , Humanos , Nefritis Lúpica/etiología , Activación de Linfocitos , Diana Mecanicista del Complejo 1 de la Rapamicina/fisiología , Ratones , Ratones Endogámicos C57BL , Fosfohidrolasa PTEN/análisis , Fosfohidrolasa PTEN/genética , Transducción de Señal , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/fisiología
10.
Cancer Res ; 79(20): 5288-5301, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31462429

RESUMEN

Misregulated alternative RNA splicing (AS) contributes to the tumorigenesis and progression of human cancers, including glioblastoma (GBM). Here, we showed that a major splicing factor, serine and arginine rich splicing factor 3 (SRSF3), was frequently upregulated in clinical glioma specimens and that elevated SRSF3 was associated with tumor progression and a poor prognosis for patients with glioma. In patient-derived glioma stem-like cells (GSC), SRSF3 expression promoted cell proliferation, self-renewal, and tumorigenesis. Transcriptomic profiling identified more than 1,000 SRSF3-affected AS events, with a preference for exon skipping in genes involved with cell mitosis. Motif analysis identified the sequence of CA(G/C/A)CC(C/A) as a potential exonic splicing enhancer for these SRSF3-regulated exons. To evaluate the biological impact of SRSF3-affected AS events, four candidates were selected whose AS correlated with SRSF3 expression in glioma tissues, and their splicing pattern was modified using a CRISPR/Cas9 approach. Two functionally validated AS candidates were further investigated for the mechanisms underlying their isoform-specific functions. Specifically, following knockout of SRSF3, transcription factor ETS variant 1 (ETV1) gene showed exon skipping at exon 7, while nudE neurodevelopment protein 1 (NDE1) gene showed replacement of terminal exon 9 with a mutually exclusive exon 9'. SRSF3-regulated AS of these two genes markedly increased their oncogenic activity in GSCs. Taken together, our data demonstrate that SRSF3 is a key regulator of AS in GBM and that understanding mechanisms of misregulated AS could provide critical insights for developing effective therapeutic strategies against GBMs. SIGNIFICANCE: SRSF3 is a significant regulator of glioma-associated alternative splicing, implicating SRSF3 as an oncogenic factor that contributes to the tumor biology of GBM.


Asunto(s)
Empalme Alternativo , Neoplasias Encefálicas/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Proteínas de Neoplasias/fisiología , ARN Mensajero/biosíntesis , Factores de Empalme Serina-Arginina/fisiología , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Sistemas CRISPR-Cas , División Celular , Línea Celular Tumoral , Autorrenovación de las Células , Proteínas de Unión al ADN/genética , Progresión de la Enfermedad , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Glioblastoma/metabolismo , Glioblastoma/patología , Células HEK293 , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Fosforilación , Pronóstico , Isoformas de Proteínas/fisiología , Procesamiento Proteico-Postraduccional , ARN Mensajero/genética , Factores de Empalme Serina-Arginina/antagonistas & inhibidores , Factores de Empalme Serina-Arginina/genética , Huso Acromático/metabolismo , Factores de Transcripción/genética
11.
Gene ; 706: 140-145, 2019 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-31078657

RESUMEN

BACKGROUND: Alternative splicing regulates most of protein-coding genes by producing diverse messenger RNA transcripts; and mis-splicing events can induce aberrant protein isoforms that contribute to cancer development. It is possible that genetic variations in splicing associated genes may regulate the formation of transcripts and multiple protein isoforms by affecting the splice regulatory elements. In this study, we aimed to determine whether genetic variations in the crucial alternative-splicing genes were associated with breast cancer risk. MATERIALS AND METHODS: A case-control study was conducted with 1064 breast cancer cases and 1073 healthy controls from China. A total of 16 tagging polymorphisms within three splicing factor-associated genes (SFRS3, ESRP1 and ESRP2) were genotyped by using Infinium BeadChip. The association between the polymorphisms and risk of breast cancer was evaluated by computing odds ratios (OR) and 95% confidence intervals (CIs). RESULTS: The genotype distribution of rs2145048 in SFRS3 was different between cases and controls (Bonferroni corrected P = 0.022). After adjusting for age, age at menarche and menopausal status, the A allele of rs2145048 showed an inverse association with breast cancer risk in the additive model (adjusted OR = 0.81, 95% CI = 0.71-0.92, P = 0.001, Bonferroni corrected P = 0.016). In the stratification analysis, the association between rs2145048 A allele and breast cancer remained significant in subgroups of earlier menarche, older first born, premenopausal status, and ER/PR negative status. CONCLUSIONS: This study provided the first evidence that SFRS3 rs2145048 was associated with breast cancer susceptibility in Chinese women, which might represent a biomarker to improve the identification of individuals at high risk of this malignancy.


Asunto(s)
Neoplasias de la Mama/genética , Factores de Empalme Serina-Arginina/genética , Adulto , Alelos , Empalme Alternativo/genética , Pueblo Asiatico/genética , Estudios de Casos y Controles , China , Femenino , Frecuencia de los Genes/genética , Predisposición Genética a la Enfermedad/genética , Variación Genética/genética , Genotipo , Humanos , Persona de Mediana Edad , Oportunidad Relativa , Polimorfismo de Nucleótido Simple/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/fisiología , Factores de Riesgo , Factores de Empalme Serina-Arginina/fisiología
12.
Med Sci Monit Basic Res ; 25: 53-62, 2019 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-30773530

RESUMEN

BACKGROUND Radio-resistance is an important barrier in nasopharyngeal carcinoma treatment. MicroRNAs are gene expression core regulators in various biological procedures containing cancer radio-resistance. Nevertheless, the clinical association between nasopharyngeal carcinoma and miR-193a-3p/SRSF2 remains unclear. MATERIAL AND METHODS We examined the miR-193a-3p level in radio-sensitive CNE-2 and radio-resistant CNE-1 NPC cell lines, and, based on a literature review, predicted SRSF2 to be the target gene of miR-193a-3p. We explored the expression of SRSF2 at protein and mRNA levels by transfecting either miR-193a-3p-mimic or antagomiR. Finally, we performed signaling pathway analysis to assess the possible role of miR-193a-3p/SRSF2 in signaling pathways. RESULTS miR-193a-3p promotes NPC radio-resistance, and the SRSF2 gene is the direct target for miR-193a-3p in NPC, and thus is negatively correlated with NPC radio-resistance. The hypoxia signaling pathway activity is strongly affected, and it is possible to use the downstream activity of the SRSF2 gene to show the effect of miR-193a-3p on radio-resistance in NPC cells. CONCLUSIONS miR-193a-3p mediates promotion of NPC radio-resistance.


Asunto(s)
MicroARNs/genética , Neoplasias Nasofaríngeas/genética , Tolerancia a Radiación/genética , Animales , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Marcación de Gen , Humanos , Hipoxia/genética , Ratones , Ratones Desnudos , MicroARNs/metabolismo , MicroARNs/fisiología , Neoplasias Nasofaríngeas/radioterapia , Factores de Empalme Serina-Arginina/genética , Factores de Empalme Serina-Arginina/fisiología , Transducción de Señal/genética , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Hepatology ; 69(1): 179-195, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30028541

RESUMEN

Deregulation of alternative splicing contributes to the malignant progression of cancer. Little is known about the significant alternative splicing events in hepatocellular carcinoma (HCC). High-throughput sequencing revealed that coiled-coil domain containing 50 (CCDC50) pre-mRNA is aberrantly spliced in 50% of our HCC cases. A BaseScope assay was performed to examine the expression of CCDC50S (a truncated oncogenic splice variant) in HCC tissues. Compared with benign liver tumors and several other types of solid tumors, CCDC50S mRNA was up-regulated in HCC, with a diagnostic potential (sensitivity, 0.711; specificity, 0.793). High expression of CCDC50S mRNA in HCC was significantly correlated with poor tumor differentiation, advanced tumor node metastasis (TNM) stage, and unfavorable prognosis. Overexpression of CCDC50S exerted tumorigenic activities that promoted HCC growth and metastasis by activation of Ras/forkhead box protein O4 (Foxo4) signaling. Either suppression of mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) phosphorylation or overexpression of Foxo4 markedly attenuated CCDC50S-mediated phenotypes. Furthermore, serine- and arginine-rich splicing factor 3 (SRSF3) directly bound to CCDC50S mRNA to maintain its stability in the cytoplasm. The cytosolic retention of SRSF3 was mediated by the interaction of hepatitis B virus-encoded X protein (HBx) and 14-3-3ß. Ectopic HBx expression induced expression of cytosolic SRSF3 and CCDC50S. Conclusion: Our study provided compelling evidence that up-regulation of CCDC50S was modulated by HBx/SRSF3/14-3-3ß complex and enhanced oncogenic progression of HCC through the Ras/Foxo4 signaling pathway. These data suggest that CCDC50S may serve as a diagnostic and prognostic biomarker and probably a promising therapeutic target in HCC.


Asunto(s)
Carcinoma Hepatocelular/etiología , Neoplasias Hepáticas/etiología , Proteínas Proto-Oncogénicas p21(ras)/fisiología , Factores de Empalme Serina-Arginina/fisiología , Transducción de Señal/fisiología , Animales , Masculino , Ratones , Ratones Endogámicos BALB C
15.
Blood ; 132(6): 608-621, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-29903888

RESUMEN

Mutations in SRSF2 occur in myelodysplastic syndromes (MDS) and MDS/myeloproliferative neoplasms (MPN). SRSF2 mutations cluster at proline 95, with the most frequent mutation being a histidine (P95H) substitution. They undergo positive selection, arise early in the course of disease, and have been identified in age-related clonal hemopoiesis. It is not clear how mutation of SRSF2 modifies hemopoiesis or contributes to the development of myeloid bias or MDS/MPN. Two prior mouse models of Srsf2P95H mutation have been reported; however, these models do not recapitulate many of the clinical features of SRSF2-mutant disease and relied on bone marrow (BM) transplantation stress to elicit the reported phenotypes. We describe a new conditional murine Srsf2P95H mutation model, where the P95H mutation is expressed physiologically and heterozygously from its endogenous locus after Cre activation. Using multiple Cre lines, we demonstrate that during native hemopoiesis (ie, no BM transplantation), the Srsf2P95H mutation needs to occur within the hemopoietic stem-cell-containing populations to promote myelomonocytic bias and expansion with corresponding transcriptional and RNA splicing changes. With age, nontransplanted Srsf2P95H animals developed a progressive, transplantable disease characterized by myeloid bias, morphological dysplasia, and monocytosis, hallmarks of MDS/MPN in humans. Analysis of cooccurring mutations within the BM demonstrated the acquisition of additional mutations that are recurrent in humans with SRSF2 mutations. The tractable Srsf2P95H/+ knock-in model we have generated is highly relevant to human disease and will serve to elucidate the effect of SRSF2 mutations on initiation and maintenance of MDS/MPN.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Síndromes Mielodisplásicos/genética , Células Mieloides/metabolismo , Mielopoyesis/genética , Trastornos Mieloproliferativos/genética , Factores de Empalme Serina-Arginina/genética , Envejecimiento/genética , Animales , Trasplante de Médula Ósea , Modelos Animales de Enfermedad , Exoma , Perfilación de la Expresión Génica , Técnicas de Sustitución del Gen , Genes p53 , Células Madre Hematopoyéticas/patología , Ratones , Ratones Endogámicos C57BL , Células Mieloides/patología , Empalme del ARN , Quimera por Radiación , Proteínas Recombinantes/metabolismo , Factores de Empalme Serina-Arginina/fisiología , Especificidad de la Especie
16.
Biomed Res Int ; 2018: 3819719, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29789787

RESUMEN

Serine/arginine-rich splicing factors (SRSFs) have one or two RNA recognition motifs in the N terminal and a serine/arginine-enriched domain in the C terminal. SRSFs are essential components of spliceosomes and are involved in alternative splicing, spliceosome assembly, mRNA export, and nonsense-mediated mRNA decay. The maintenance of cellular and tissue homeostasis relies on accurate alternative splicing, and various patterns of abnormal alternative splicing can cause different diseases. SRSF4 is associated with many physiological and pathological processes and has applications in the diagnosis and prognosis of specific diseases. In this review, we discuss knowledge of SRSF4 in physiological and pathological processes and highlight the applications of SRSF4 in the regulation of gene expression and associated diseases.


Asunto(s)
Factores de Empalme Serina-Arginina/genética , Factores de Empalme Serina-Arginina/fisiología , Azoospermia/genética , Azoospermia/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Regulación de la Expresión Génica/genética , Humanos , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Masculino , Polimorfismo de Nucleótido Simple/genética , Empalmosomas/genética , Empalmosomas/fisiología
17.
Mol Cells ; 41(5): 465-475, 2018 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-29764005

RESUMEN

The advent of massively parallel sequencing, also called next-generation sequencing (NGS), has dramatically influenced cancer genomics by accelerating the identification of novel molecular alterations. Using a whole genome sequencing (WGS) approach, we identified somatic coding and noncoding variants that may contribute to leukemogenesis in 11 adult Korean acute myeloid leukemia (AML) patients, with serial tumor samples (primary and relapse) available for 5 of them; somatic variants were identified in 187 AML-related genes, including both novel (SIN3A, C10orf53, PTPRR, and RERGL) and well-known (NPM1, RUNX1, and CEPBA) AML-related genes. Notably, SIN3A expression shows prognostic value in AML. A newly designed method, referred to as "hot-zone" analysis, detected two putative functional noncoding variants that can alter transcription factor binding affinity near PPP1R10 and SRSF1. Moreover, the functional importance of the SRSF1 noncoding variant was further investigated by luciferase assays, which showed that the variant is critical for the regulation of gene expression leading to leukemogenesis. We expect that further functional investigation of these coding and noncoding variants will contribute to a more in-depth understanding of the underlying molecular mechanisms of AML and the development of targeted anti-cancer drugs.


Asunto(s)
Proteínas de Unión al ADN/genética , Leucemia Mieloide Aguda/genética , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Proteínas de Unión al ARN/genética , Proteínas Represoras/genética , Factores de Empalme Serina-Arginina/genética , Adulto , Anciano , Análisis Mutacional de ADN , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/fisiología , Femenino , Regulación Leucémica de la Expresión Génica , Ontología de Genes , Genes Reporteros , Mutación de Línea Germinal , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/química , Proteínas de Neoplasias/fisiología , Proteínas Nucleares/química , Proteínas Nucleares/fisiología , Nucleofosmina , Pronóstico , Unión Proteica , Estructura Secundaria de Proteína , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/fisiología , Proteínas Represoras/química , Proteínas Represoras/fisiología , Factores de Empalme Serina-Arginina/química , Factores de Empalme Serina-Arginina/fisiología , Complejo Correpresor Histona Desacetilasa y Sin3 , Factores de Transcripción/metabolismo , Secuenciación Completa del Genoma , Adulto Joven
18.
Proc Natl Acad Sci U S A ; 115(16): E3808-E3816, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29610341

RESUMEN

The zinc finger CCCH-type containing 11A (ZC3H11A) gene encodes a well-conserved zinc finger protein that may function in mRNA export as it has been shown to associate with the transcription export (TREX) complex in proteomic screens. Here, we report that ZC3H11A is a stress-induced nuclear protein with RNA-binding capacity that localizes to nuclear splicing speckles. During an adenovirus infection, the ZC3H11A protein and splicing factor SRSF2 relocalize to nuclear regions where viral DNA replication and transcription take place. Knockout (KO) of ZC3H11A in HeLa cells demonstrated that several nuclear-replicating viruses are dependent on ZC3H11A for efficient growth (HIV, influenza virus, herpes simplex virus, and adenovirus), whereas cytoplasmic replicating viruses are not (vaccinia virus and Semliki Forest virus). High-throughput sequencing of ZC3H11A-cross-linked RNA showed that ZC3H11A binds to short purine-rich ribonucleotide stretches in cellular and adenoviral transcripts. We show that the RNA-binding property of ZC3H11A is crucial for its function and localization. In ZC3H11A KO cells, the adenovirus fiber mRNA accumulates in the cell nucleus. Our results suggest that ZC3H11A is important for maintaining nuclear export of mRNAs during stress and that several nuclear-replicating viruses take advantage of this mechanism to facilitate their replication.


Asunto(s)
Núcleo Celular/virología , Proteínas Nucleares/fisiología , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Proteínas de Unión al ARN/fisiología , Replicación Viral , Dedos de Zinc/fisiología , Adenovirus Humanos/genética , Adenovirus Humanos/fisiología , Sitios de Unión , Transporte Biológico , Sistemas CRISPR-Cas , Núcleo Celular/metabolismo , Citoplasma/virología , Técnicas de Inactivación de Genes , Células HeLa , Respuesta al Choque Térmico/genética , Respuesta al Choque Térmico/fisiología , Secuenciación de Nucleótidos de Alto Rendimiento , Interacciones Huésped-Patógeno , Humanos , Proteínas Nucleares/antagonistas & inhibidores , Dominios Proteicos , Estabilidad Proteica , Proteínas de Unión al ARN/antagonistas & inhibidores , Factores de Empalme Serina-Arginina/fisiología
19.
Mol Biol Cell ; 29(9): 1111-1124, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29496966

RESUMEN

C3G (Crk SH3 domain binding guanine nucleotide releasing factor) (Rap guanine nucleotide exchange factor 1), essential for mammalian embryonic development, is ubiquitously expressed and undergoes regulated nucleocytoplasmic exchange. Here we show that C3G localizes to SC35-positive nuclear speckles and regulates splicing activity. Reversible association of C3G with speckles was seen on inhibition of transcription and splicing. C3G shows partial colocalization with SC35 and is recruited to a chromatin and RNase-sensitive fraction of speckles. Its presence in speckles is dependent on intact cellular actin cytoskeleton and is lost on expression of the kinase Clk1. Rap1, a substrate of C3G, is also present in nuclear speckles, and inactivation of Rap signaling by expression of GFP-Rap1GAP alters speckle morphology and number. Enhanced association of C3G with speckles is seen on glycogen synthase kinase 3 beta inhibition or differentiation of C2C12 cells to myotubes. CRISPR/Cas9-mediated knockdown of C3G resulted in altered splicing activity of an artificial gene as well as endogenous CD44. C3G knockout clones of C2C12 as well as MDA-MB-231 cells showed reduced protein levels of several splicing factors compared with control cells. Our results identify C3G and Rap1 as novel components of nuclear speckles and a role for C3G in regulating cellular RNA splicing activity.


Asunto(s)
Factor 2 Liberador de Guanina Nucleótido/metabolismo , Factor 2 Liberador de Guanina Nucleótido/fisiología , Empalme del ARN/fisiología , Animales , Diferenciación Celular , Línea Celular , Línea Celular Tumoral , Estructuras del Núcleo Celular/fisiología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Proteínas Nucleares , Unión Proteica , Proteínas Serina-Treonina Quinasas , Proteínas Tirosina Quinasas , Procesamiento Postranscripcional del ARN/fisiología , ARN Mensajero/metabolismo , Factores de Empalme Serina-Arginina/metabolismo , Factores de Empalme Serina-Arginina/fisiología , Complejo Shelterina , Transducción de Señal , Empalmosomas , Proteínas de Unión a Telómeros/metabolismo
20.
Oncogene ; 37(18): 2394-2409, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29429992

RESUMEN

High-risk human papillomavirus oncoproteins E6 and E7 are the major etiological factors of cervical cancer but are insufficient for malignant transformation of cervical cancer. Dysregulated alternative splicing, mainly ascribed to aberrant splicing factor levels and activities, contributes to most cancer hallmarks. However, do E6 and E7 regulate the expression of splicing factors? Does alternative splicing acts as an "accomplice" of E6E7 to promote cervical cancer progression? Here, we identified that the splicing factor SRSF10, which promotes tumorigenesis of cervix, was upregulated by E6E7 via E2F1 transcriptional activation. SRSF10 modulates the alternate terminator of interleukin-1 receptor accessory protein exon 13 to increase production of the membrane form of interleukin-1 receptor accessory protein. SRSF10-mediated mIL1RAP upregulates the expression of the "don't eat me" signal CD47 to inhibit macrophage phagocytosis by promoting nuclear factor-κB activation, which is pivotal in inflammatory, immune, and tumorigenesis processes. Altogether, these data reveal a close relationship among HPV infection, alternative splicing and tumor immune evasion, and also suggests that the SRSF10-mIL1RAP-CD47 axis could be an attractive therapeutic target for the treatment of cervical cancer.


Asunto(s)
Empalme Alternativo/genética , Carcinogénesis/genética , Proteínas de Ciclo Celular/fisiología , Proteína Accesoria del Receptor de Interleucina-1/genética , Proteínas Represoras/fisiología , Factores de Empalme Serina-Arginina/fisiología , Neoplasias del Cuello Uterino/genética , Animales , Antígeno CD47/metabolismo , Carcinogénesis/metabolismo , Carcinogénesis/patología , Transformación Celular Viral/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Papillomavirus Humano 16/fisiología , Papillomavirus Humano 18/fisiología , Humanos , Ratones , Ratones Desnudos , FN-kappa B/metabolismo , Isoformas de Proteínas/genética , Transducción de Señal/genética , Células Tumorales Cultivadas , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA