Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cancer Sci ; 113(2): 373-381, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34812550

RESUMEN

There has been accumulating evidence that RNA splicing is frequently dysregulated in a variety of cancers and that hotspot mutations affecting key splicing factors, SF3B1, SRSF2 and U2AF1, are commonly enriched across cancers, strongly suggesting that aberrant RNA splicing is a new class of hallmark that contributes to the initiation and/or maintenance of cancers. In parallel, some studies have demonstrated that cancer cells with global splicing alterations are dependent on the transcriptional products derived from wild-type spliceosome for their survival, which potentially creates a therapeutic vulnerability in cancers with a mutant spliceosome. It has been c. 10 y since the frequent mutations affecting splicing factors were reported in cancers. Based on these surprising findings, there has been a growing interest in targeting altered splicing in the treatment of cancers, which has promoted a wide variety of investigations including genetic, molecular and biological studies addressing how altered splicing promotes oncogenesis and how cancers bearing alterations in splicing can be targeted therapeutically. In this mini-review we present a concise trajectory of what has been elucidated regarding the pathogenesis of cancers with aberrant splicing, as well as the development of therapeutic strategies to target global splicing alterations in cancers.


Asunto(s)
Neoplasias/genética , Empalme del ARN/genética , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Humanos , Mutación , Neoplasias/tratamiento farmacológico , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Oligonucleótidos Antisentido/uso terapéutico , Empalme del ARN/efectos de los fármacos , Factores de Empalme de ARN/antagonistas & inhibidores , Factores de Empalme de ARN/genética , Factores de Empalme de ARN/metabolismo , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Empalmosomas/efectos de los fármacos , Empalmosomas/genética , Empalmosomas/metabolismo
2.
Leukemia ; 35(12): 3542-3550, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34172893

RESUMEN

We conducted a phase I clinical trial of H3B-8800, an oral small molecule that binds Splicing Factor 3B1 (SF3B1), in patients with MDS, CMML, or AML. Among 84 enrolled patients (42 MDS, 4 CMML and 38 AML), 62 were red blood cell (RBC) transfusion dependent at study entry. Dose escalation cohorts examined two once-daily dosing regimens: schedule I (5 days on/9 days off, range of doses studied 1-40 mg, n = 65) and schedule II (21 days on/7 days off, 7-20 mg, n = 19); 27 patients received treatment for ≥180 days. The most common treatment-related, treatment-emergent adverse events included diarrhea, nausea, fatigue, and vomiting. No complete or partial responses meeting IWG criteria were observed; however, RBC transfusion free intervals >56 days were observed in nine patients who were transfusion dependent at study entry (15%). Of 15 MDS patients with missense SF3B1 mutations, five experienced RBC transfusion independence (TI). Elevated pre-treatment expression of aberrant transcripts of Transmembrane Protein 14C (TMEM14C), an SF3B1 splicing target encoding a mitochondrial porphyrin transporter, was observed in MDS patients experiencing RBC TI. In summary, H3B-8800 treatment was associated with mostly low-grade TAEs and induced RBC TI in a biomarker-defined subset of MDS.


Asunto(s)
Leucemia Mieloide Aguda/tratamiento farmacológico , Síndromes Mielodisplásicos/tratamiento farmacológico , Fosfoproteínas/antagonistas & inhibidores , Piperazinas/uso terapéutico , Piridinas/uso terapéutico , Factores de Empalme de ARN/antagonistas & inhibidores , Administración Oral , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Masculino , Persona de Mediana Edad , Mutación Missense , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/patología , Seguridad del Paciente , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Piperazinas/efectos adversos , Piridinas/efectos adversos , Factores de Empalme de ARN/genética , Factores de Empalme de ARN/metabolismo , Resultado del Tratamiento
3.
Cell Chem Biol ; 28(11): 1616-1627.e8, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34048672

RESUMEN

The proteolysis-targeting chimeras (PROTACs) are a new technology to degrade target proteins. However, their clinical application is limited currently by lack of chemical binders to target proteins. For instance, it is still unknown whether splicing factor 3B subunit 1 (SF3B1) is targetable by PROTACs. We recently identified a 2-aminothiazole derivative (herein O4I2) as a promoter in the generation of human pluripotent stem cells. In this work, proteomic analysis on the biotinylated O4I2 revealed that O4I2 targeted SF3B1 and positively regulated RNA splicing. Fusing thalidomide-the ligand of the cereblon ubiquitin ligase-to O4I2 led to a new PROTAC-O4I2, which selectively degraded SF3B1 and induced cellular apoptosis in a CRBN-dependent manner. In a Drosophila intestinal tumor model, PROTAC-O4I2 increased survival by interference with the maintenance and proliferation of stem cell. Thus, our finding demonstrates that SF3B1 is PROTACable by utilizing noninhibitory chemicals, which expands the list of PROTAC target proteins.


Asunto(s)
Fosfoproteínas/antagonistas & inhibidores , Factores de Empalme de ARN/antagonistas & inhibidores , Tiazoles/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Drosophila melanogaster , Humanos , Fosfoproteínas/metabolismo , Proteolisis/efectos de los fármacos , Empalme del ARN/efectos de los fármacos , Factores de Empalme de ARN/metabolismo , Tiazoles/síntesis química , Tiazoles/química
4.
Aging (Albany NY) ; 13(8): 11135-11149, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33819187

RESUMEN

Myocardial infarction (MI) is one of the leading causes of death. Wilms' tumor 1-associating protein (WTAP), one of the components of the m6A methyltransferase complex, has been shown to affect gene expression via regulating mRNA modification. Although WTAP has been implicated in various diseases, its role in MI is unclear. In this study, we found that hypoxia/reoxygenation (H/R) time-dependently increased WTAP expression, which in turn promoted endoplasmic reticulum (ER) stress and apoptosis, in human cardiomyocytes (AC16). H/R effects on ER stress and apoptosis were all blocked by silencing of WTAP, promoted by WTAP overexpression, and ameliorated by administration of ER stress inhibitor, 4-PBA. We then investigated the underlying molecular mechanism and found that WTAP affected m6A methylation of ATF4 mRNA to regulate its expression, and that the inhibitory effects of WTAP on ER stress and apoptosis were ATF4 dependent. Finally, WTAP's effects on myocardial I/R injury were confirmed in vivo. WTAP promoted myocardial I/R injury through promoting ER stress and cell apoptosis by regulating m6A modification of ATF4 mRNA. These findings highlight the importance of WTAP in I/R injury and provide new insights into therapeutic strategies for MI.


Asunto(s)
Factor de Transcripción Activador 4/genética , Proteínas de Ciclo Celular/metabolismo , Infarto del Miocardio/complicaciones , Daño por Reperfusión Miocárdica/genética , Factores de Empalme de ARN/metabolismo , Adenosina/análogos & derivados , Adenosina/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Butilaminas/farmacología , Butilaminas/uso terapéutico , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Línea Celular , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/genética , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Metilación , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/patología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Factores de Empalme de ARN/antagonistas & inhibidores , Factores de Empalme de ARN/genética , ARN Mensajero/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
5.
Cell Chem Biol ; 28(9): 1356-1365.e4, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-33784500

RESUMEN

RNA splicing, a highly conserved process in eukaryotic gene expression, is seen as a promising target for anticancer agents. Splicing is associated with other RNA processing steps, such as transcription and nuclear export; however, our understanding of the interaction between splicing and other RNA regulatory mechanisms remains incomplete. Moreover, the impact of chemical splicing inhibition on long non-coding RNAs (lncRNAs) has been poorly understood. Here, we demonstrate that spliceostatin A (SSA), a chemical splicing modulator that binds to the SF3B subcomplex of the U2 small nuclear ribonucleoprotein particle (snRNP), limits U1 snRNP availability in splicing, resulting in premature cleavage and polyadenylation of MALAT1, a nuclear lncRNA, as well as protein-coding mRNAs. Therefore, truncated transcripts are exported into the cytoplasm and translated, resulting in aberrant protein products. Our work demonstrates that active recycling of the splicing machinery maintains homeostasis of RNA processing beyond intron excision.


Asunto(s)
Fosfoproteínas/antagonistas & inhibidores , Piranos/farmacología , Factores de Empalme de ARN/antagonistas & inhibidores , ARN Largo no Codificante/metabolismo , Ribonucleoproteína Nuclear Pequeña U1/antagonistas & inhibidores , Compuestos de Espiro/farmacología , Femenino , Células HeLa , Humanos , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Poliadenilación/efectos de los fármacos , Piranos/química , Empalme del ARN/efectos de los fármacos , Factores de Empalme de ARN/química , Factores de Empalme de ARN/metabolismo , Ribonucleoproteína Nuclear Pequeña U1/química , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Compuestos de Espiro/química , Células Tumorales Cultivadas
6.
Commun Biol ; 4(1): 386, 2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-33753867

RESUMEN

APOBEC3A (A3A) and APOBEC3B (A3B) enzymes drive APOBEC-mediated mutagenesis. Identification of factors affecting the activity of these enzymes could help modulate mutagenesis and associated clinical outcomes. Here, we show that canonical and alternatively spliced A3A and A3B isoforms produce corresponding mutagenic and non-mutagenic enzymes. Increased expression of the mutagenic A3B isoform predicted shorter progression-free survival in bladder cancer. We demonstrate that the production of mutagenic vs. non-mutagenic A3B protein isoforms was considerably affected by inclusion/skipping of exon 5 in A3B. Furthermore, exon 5 skipping, resulting in lower levels of mutagenic A3B enzyme, could be increased in vitro. Specifically, we showed the effects of treatment with an SF3B1 inhibitor affecting spliceosome interaction with a branch point site in intron 4, or with splice-switching oligonucleotides targeting exon 5 of A3B. Our results underscore the clinical role of A3B and implicate alternative splicing of A3B as a mechanism that could be targeted to restrict APOBEC-mediated mutagenesis.


Asunto(s)
Empalme Alternativo , Biomarcadores de Tumor/genética , Citidina Desaminasa/genética , Antígenos de Histocompatibilidad Menor/genética , Mutagénesis , Proteínas/genética , Neoplasias de la Vejiga Urinaria/genética , Biomarcadores de Tumor/metabolismo , Citidina Desaminasa/metabolismo , Compuestos Epoxi/farmacología , Exones , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Isoenzimas , Macrólidos/farmacología , Antígenos de Histocompatibilidad Menor/metabolismo , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/metabolismo , Supervivencia sin Progresión , Proteínas/metabolismo , Factores de Empalme de ARN/antagonistas & inhibidores , Factores de Empalme de ARN/metabolismo , Neoplasias de la Vejiga Urinaria/enzimología , Neoplasias de la Vejiga Urinaria/mortalidad , Neoplasias de la Vejiga Urinaria/terapia
7.
ACS Chem Biol ; 16(3): 520-528, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33617218

RESUMEN

Small molecules that target the spliceosome SF3B complex are potent inhibitors of cancer cell growth. The compounds affect an early stage of spliceosome assembly when U2 snRNP first engages the branch point sequence of an intron. Employing an inactive herboxidiene analog (iHB) as a competitor, we investigated factors that influence inhibitor interactions with SF3B to interfere with pre-mRNA splicing in vitro. Order-of-addition experiments show that inhibitor interactions are long lasting and affected by both temperature and the presence of ATP. Our data are also consistent with the model that not all SF3B conformations observed in structural studies are conducive to productive inhibitor interactions. Notably, SF3B inhibitors do not impact an ATP-dependent rearrangement in U2 snRNP that exposes the branch binding sequence for base pairing. We also report extended structure-activity relationship analysis of the splicing inhibitor herboxidiene. We identified features of the tetrahydropyran ring that mediate its interactions with SF3B and its ability to interfere with splicing. In the context of recent structures of SF3B bound to inhibitor, our results lead us to extend the model for early spliceosome assembly and inhibitor mechanism. We postulate that interactions between a carboxylic acid substituent of herboxidiene and positively charged SF3B1 side chains in the inhibitor binding channel are needed to maintain inhibitor occupancy while counteracting the SF3B transition to a closed state that is required for stable U2 snRNP interactions with the intron.


Asunto(s)
Alcoholes Grasos/química , Fosfoproteínas/agonistas , Fosfoproteínas/antagonistas & inhibidores , Piranos/química , Factores de Empalme de ARN/agonistas , Factores de Empalme de ARN/antagonistas & inhibidores , Empalme del ARN/efectos de los fármacos , Ribonucleoproteína Nuclear Pequeña U2/química , Empalmosomas/química , Adenosina Trifosfato/química , Secuencia de Bases , Sitios de Unión , Alcoholes Grasos/metabolismo , Células HeLa , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Piranos/metabolismo , ARN Mensajero/química , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Empalmosomas/metabolismo , Relación Estructura-Actividad , Temperatura
8.
Curr Opin Hematol ; 28(2): 73-79, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33492002

RESUMEN

PURPOSE OF REVIEW: Mutations in components of the spliceosome are the most common acquired lesions in myelodysplastic syndromes (MDS) and are frequently identified in other myeloid malignancies with a high rate of progression to acute myeloid leukemia (AML) including chronic myelomonocytic leukemia and primary myelofibrosis. The only curative option for these disorders remains allogeneic stem-cell transplantation, which is associated with high morbidity and mortality in these patients. The purpose of this review is to highlight the recent therapeutic developments and strategies being pursued for clinical benefit in splicing factor mutant myeloid malignancies. RECENT FINDINGS: Cells harboring splicing factor mutations have increased aberrant splicing leading to R-loop formation and cell cycle stalling that create dependencies on Checkpoint kinase 1 (CHK1) activation and canonical splicing maintained by protein arginine methyltransferase activity. Both targeting of the spliceosome and targeting of the downstream consequences of splicing factor mutation expression show promise as selective strategies for the treatment of splicing factor-mutant myeloid malignancies. SUMMARY: An improved understanding of the therapeutic vulnerabilities in splicing factor-mutant MDS and AML has led to the development of clinical trials of small molecule inhibitors that target the spliceosome, ataxia telangectasia and Rad3 related (ATR)-CHK1 pathway, and methylation of splicing components.


Asunto(s)
Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Terapia Molecular Dirigida , Mutación , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/genética , Factores de Empalme de ARN/antagonistas & inhibidores , Factores de Empalme de ARN/genética , Animales , Biomarcadores , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Desarrollo de Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/diagnóstico , Síndromes Mielodisplásicos/diagnóstico , Empalme del ARN , Factores de Empalme de ARN/metabolismo , Transducción de Señal/efectos de los fármacos
9.
J Cereb Blood Flow Metab ; 41(3): 530-545, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32248729

RESUMEN

RNA-binding protein fox-1 homolog 1 (Rbfox-1), an RNA-binding protein in neurons, is thought to be associated with many neurological diseases. To date, the mechanism on which Rbfox-1 worsens secondary cell death in ICH remains poorly understood. In this study, we aimed to explore the role of Rbfox-1 in intracerebral hemorrhage (ICH)-induced secondary brain injury (SBI) and to identify its underlying mechanisms. We found that the expression of Rbfox-1 in neurons was significantly increased after ICH, which was accompanied by increases in the binding of Rbfox-1 to Ca2+/calmodulin-dependent protein kinase II (CaMKIIα) mRNA and the protein level of CaMKIIα. In addition, when exposed to exogenous upregulation or downregulation of Rbfox-1, the protein level of CaMKIIα showed a concomitant trend in brain tissue, which further suggested that CaMKIIα is a downstream-target protein of Rbfox-1. The upregulation of both proteins caused intracellular-Ca2+ overload and neuronal degeneration, which exacerbated brain damage. Furthermore, we found that Rbfox-1 promoted the expression of CaMKIIα via blocking the binding of micro-RNA-124 to CaMKIIα mRNA. Thus, Rbfox-1 is expected to be a promising therapeutic target for SBI after ICH.


Asunto(s)
Lesiones Encefálicas/patología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Hemorragias Intracraneales/complicaciones , MicroARNs/metabolismo , Factores de Empalme de ARN/metabolismo , Animales , Antagomirs/metabolismo , Apoptosis/efectos de los fármacos , Conducta Animal , Encéfalo/metabolismo , Encéfalo/patología , Lesiones Encefálicas/etiología , Lesiones Encefálicas/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Células Cultivadas , Disfunción Cognitiva/etiología , Modelos Animales de Enfermedad , Hemorragias Intracraneales/inducido químicamente , Masculino , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Neuronas/citología , Neuronas/metabolismo , Oxihemoglobinas/farmacología , Interferencia de ARN , Factores de Empalme de ARN/antagonistas & inhibidores , Factores de Empalme de ARN/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley
10.
Cells ; 9(12)2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-33317029

RESUMEN

Splicing factor 3b subunit 1 (SF3B1) is an essential protein in spliceosomes and mutated frequently in many cancers. While roles of SF3B1 in single intron splicing and roles of its cancer-linked mutant in aberrant splicing have been identified to some extent, regulatory functions of wild-type SF3B1 in alternative splicing (AS) are not well-understood yet. Here, we applied RNA sequencing (RNA-seq) to analyze genome-wide AS in SF3B1 knockdown (KD) cells and to identify a large number of skipped exons (SEs), with a considerable number of alternative 5' splice-site selection, alternative 3' splice-site selection, mutually exclusive exons (MXE), and retention of introns (RI). Among altered SEs by SF3B1 KD, survival motor neuron 2 (SMN2) pre-mRNA exon 7 splicing was a regulatory target of SF3B1. RT-PCR analysis of SMN exon 7 splicing in SF3B1 KD or overexpressed HCT116, SH-SY5Y, HEK293T, and spinal muscular atrophy (SMA) patient cells validated the results. A deletion mutation demonstrated that the U2 snRNP auxiliary factor 65 kDa (U2AF65) interaction domain of SF3B1 was required for its function in SMN exon 7 splicing. In addition, mutations to lower the score of the polypyrimidine tract (PPT) of exon 7, resulting in lower affinity for U2AF65, were not able to support SF3B1 function, suggesting the importance of U2AF65 in SF3B1 function. Furthermore, the PPT of exon 7 with higher affinity to U2AF65 than exon 8 showed significantly stronger interactions with SF3B1. Collectively, our results revealed SF3B1 function in SMN alternative splicing.


Asunto(s)
Empalme Alternativo , Fosfoproteínas/metabolismo , Factores de Empalme de ARN/metabolismo , Factor de Empalme U2AF/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Línea Celular , Exones , Humanos , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/patología , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/genética , Proteína de Unión al Tracto de Polipirimidina/genética , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Unión Proteica , Interferencia de ARN , Precursores del ARN/genética , Precursores del ARN/metabolismo , Factores de Empalme de ARN/antagonistas & inhibidores , Factores de Empalme de ARN/genética , ARN Interferente Pequeño/metabolismo , Factor de Empalme U2AF/química , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo
11.
Drug Resist Updat ; 53: 100728, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33070093

RESUMEN

Alternative splicing is a tightly regulated process whereby non-coding sequences of pre-mRNA are removed and protein-coding segments are assembled in diverse combinations, ultimately giving rise to proteins with distinct or even opposing functions. In the past decade, whole genome/transcriptome sequencing studies revealed the high complexity of splicing regulation, which occurs co-transcriptionally and is influenced by chromatin status and mRNA modifications. Consequently, splicing profiles of both healthy and malignant cells display high diversity and alternative splicing was shown to be widely deregulated in multiple cancer types. In particular, mutations in pre-mRNA regulatory sequences, splicing regulators and chromatin modifiers, as well as differential expression of splicing factors are important contributors to cancer pathogenesis. It has become clear that these aberrations contribute to many facets of cancer, including oncogenic transformation, cancer progression, response to anticancer drug treatment as well as resistance to therapy. In this respect, alternative splicing was shown to perturb the expression a broad spectrum of relevant genes involved in drug uptake/metabolism (i.e. SLC29A1, dCK, FPGS, and TP), activation of nuclear receptor pathways (i.e. GR, AR), regulation of apoptosis (i.e. MCL1, BCL-X, and FAS) and modulation of response to immunotherapy (CD19). Furthermore, aberrant splicing constitutes an important source of novel cancer biomarkers and the spliceosome machinery represents an attractive target for a novel and rapidly expanding class of therapeutic agents. Small molecule inhibitors targeting SF3B1 or splice factor kinases were highly cytotoxic against a wide range of cancer models, including drug-resistant cells. Importantly, these effects are enhanced in specific cancer subsets, such as splicing factor-mutated and c-MYC-driven tumors. Furthermore, pre-clinical studies report synergistic effects of spliceosome modulators in combination with conventional antitumor agents. These strategies based on the use of low dose splicing modulators could shift the therapeutic window towards decreased toxicity in healthy tissues. Here we provide an extensive overview of the latest findings in the field of regulation of splicing in cancer, including molecular mechanisms by which cancer cells harness alternative splicing to drive oncogenesis and evade anticancer drug treatment as well as splicing-based vulnerabilities that can provide novel treatment opportunities. Furthermore, we discuss current challenges arising from genome-wide detection and prediction methods of aberrant splicing, as well as unravelling functional relevance of the plethora of cancer-related splicing alterations.


Asunto(s)
Empalme Alternativo/genética , Antineoplásicos/farmacología , Carcinogénesis/genética , Resistencia a Antineoplásicos/genética , Neoplasias/tratamiento farmacológico , Empalme Alternativo/efectos de los fármacos , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Mutación , Neoplasias/genética , Factores de Empalme de ARN/antagonistas & inhibidores , Factores de Empalme de ARN/genética , Factores de Empalme de ARN/metabolismo
12.
ChemMedChem ; 15(22): 2098-2120, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33037739

RESUMEN

The pre-mRNA splicing factor SF3B1 shows recurrent mutations among hematologic malignancies and some solid tumors. In 2007, the identification of two cytotoxic natural products, which showed splicing inhibition by binding to SF3b, prompted the development of small-molecule splicing modulators of SF3B1 as therapeutics for cancer. Recent studies suggested that spliceosome-mutant cells are preferentially sensitive to pharmacologic splicing modulation; therefore, exploring the clinical utility of splicing modulator therapies in patients with spliceosome-mutant hematologic malignancies who have failed current therapies is greatly needed, as these patients have few treatment options. H3B-8800 had unique pharmacological activity and exhibited favorable data in phase I clinical trials to treat patients with advanced myeloid malignancies, indicating that further clinical trials are promising. The most established small-molecule modulators of SF3B1 can be categorized into three classes: the bicycles, the monopyranes, and the 12-membered macrolides. This review provides a comprehensive overview of the structure-activity relationships of small-molecule SF3B1 modulators, with a detailed analysis of interactions between modulators and protein binding pocket. The future strategy for splicing modulators development is also discussed.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , Fosfoproteínas/antagonistas & inhibidores , Factores de Empalme de ARN/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Antineoplásicos/química , Humanos , Neoplasias/metabolismo , Fosfoproteínas/metabolismo , Factores de Empalme de ARN/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad
13.
Recent Pat Anticancer Drug Discov ; 15(4): 293-305, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32900350

RESUMEN

BACKGROUND: RNA splicing, a fundamental step in gene expression, is aimed at intron removal and ordering of exons to form the protein's reading frame. OBJECTIVE: This review is focused on the role of RNA splicing in cancer biology; the splicing abnormalities that lead to tumor progression emerge as targets for therapeutic intervention. METHODS: We discuss the role of aberrant mRNA splicing in carcinogenesis and drug response. RESULTS AND CONCLUSION: Pharmacological modulation of RNA splicing sets the stage for treatment approaches in situations where mRNA splicing is a clinically meaningful mechanism of the disease.


Asunto(s)
Resistencia a Antineoplásicos , Mutación , Neoplasias/tratamiento farmacológico , Factores de Empalme de ARN/antagonistas & inhibidores , Empalme del ARN , ARN Mensajero , Exones , Humanos , Intrones , Neoplasias/genética , Neoplasias/patología , Patentes como Asunto , Fosfotransferasas , Factores de Empalme de ARN/metabolismo
14.
Hum Mol Genet ; 29(5): 756-765, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-31919495

RESUMEN

CRISPR/Cas and the high conservation of the spliceosome components facilitate the mimicking of human pathological mutations in splicing factors of model organisms. The degenerative retinal disease retinitis pigmentosa (RP) is caused by mutations in distinct types of genes, including missense mutations in splicing factors that provoke RP in an autosomal dominant form (s-adRP). Using CRISPR in Caenorhabditis elegans, we generated mutant strains to mimic s-adRP mutations reported in PRPF8 and SNRNP200. Whereas these inherited mutations are present in heterozygosis in patients, C. elegans allows the maintenance of these mutations as homozygotes, which is advantageous for genetic and drug screens. We found that snrp-200(cer23[V676L]) and prp-8(cer14[H2302del]) display pleiotropic phenotypes, including reduced fertility. However, snrp-200(cer24[S1080L]) and prp-8(cer22[R2303G]) are weak alleles suitable for RNAi screens for identifying genetic interactions, which could uncover potential disease modifiers. We screened a collection of RNAi clones for splicing-related genes and identified three splicing factors: isy-1/ISY1, cyn-15/PPWD1 and mog-2/SNRPA1, whose partial inactivation may modify the course of the disease. Interestingly, these three genes act as modifiers of prp-8(cer22) but not of snrp-200(cer24). Finally, a screen of the strong allele prp-8(cer14) with FDA-approved drugs did not identify molecules capable of alleviating the temperature-sensitive sterility. Instead, we detected drugs, such as dequalinium chloride, which exacerbated the phenotype, and therefore, are potentially harmful to s-adRP patients since they may accelerate the progression of the disease.


Asunto(s)
Mutación Missense , Preparaciones Farmacéuticas/administración & dosificación , Factores de Empalme de ARN/genética , Empalme del ARN , Proteínas de Unión al ARN/genética , Retinitis Pigmentosa/patología , Ribonucleoproteínas Nucleares Pequeñas/genética , Animales , Sistemas CRISPR-Cas , Caenorhabditis elegans , Genes Dominantes , Ensayos Analíticos de Alto Rendimiento , Humanos , Interferencia de ARN , Factores de Empalme de ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/antagonistas & inhibidores , Retinitis Pigmentosa/tratamiento farmacológico , Retinitis Pigmentosa/genética , Ribonucleoproteínas Nucleares Pequeñas/antagonistas & inhibidores
15.
Breast Cancer ; 27(3): 464-476, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-31919642

RESUMEN

BACKGROUND: Splicing factor 3b subunit 1 (SF3B1) was frequently reported to be significantly mutated in breast cancer. However, the status of SF3B1 expression, its function and molecular consequence in breast cancer remained unreported. METHODS: Immunohistochemistry was used to assess SF3B1expression in 110 breast cancer samples. SF3B1 knock­down in ZR-75-30 and MDA-MB-231 cells was performed by shRNA transfection. The expression of SF3B1 in cells was detected by quantitative real­time PCR and western blot. Cell proliferation ability was determined by MTT and colony formation assay. Migration and invasion were determined by transwell assay. Flow cytometry was performed to investigate cell cycle and apoptosis. RNA-sequencing was performed to examine differentially expressed genes and affected alternative splicing events. RESULTS: SF3B1 is overexpressed in breast cancer tissues compared with normal tissues. Overexpression of SF3B1 is associated with lymph node metastasis. SF3B1 knockdown in MDA-MB-231 and ZR-75-30 breast cancer cells significantly induced the suppression of proliferation, migration, invasion and also enhancement of apoptosis. RNA-sequencing data revealed that 860 genes were significantly up-regulated and 776 genes were significantly down-regulated upon SF3B1 knockdown. Differentially expressed genes enriched in the signaling pathways including Ras signaling pathway; cytokine receptor interaction; tight junction; MAPK signaling pathway, Glycine, serine and threonine metabolism. Alternative splicing analysis revealed that exon skipping (SKIP) and cassette exons (MSKIP) were the most common molecular effect upon SF3B1 knockdown. CONCLUSIONS: Our study suggests that SF3B1 may be an important molecular target for breast cancer treatment and provides a new clue for clinical treatment of breast cancer.


Asunto(s)
Empalme Alternativo , Biomarcadores de Tumor/genética , Neoplasias de la Mama/patología , Movimiento Celular , Proliferación Celular , Fosfoproteínas/antagonistas & inhibidores , Factores de Empalme de ARN/antagonistas & inhibidores , ARN Interferente Pequeño/genética , Apoptosis , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Invasividad Neoplásica , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Interferencia de ARN , Factores de Empalme de ARN/genética , Factores de Empalme de ARN/metabolismo , Células Tumorales Cultivadas
16.
Cancer Res ; 79(20): 5204-5217, 2019 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-31431456

RESUMEN

Androgen receptor splice variant-7 (AR-V7) is a constitutively active AR variant implicated in castration-resistant prostate cancers. Here, we show that the RNA splicing factor SF3B2, identified by in silico and CRISPR/Cas9 analyses, is a critical determinant of AR-V7 expression and is correlated with aggressive cancer phenotypes. Transcriptome and PAR-CLIP analyses revealed that SF3B2 controls the splicing of target genes, including AR, to drive aggressive phenotypes. SF3B2-mediated aggressive phenotypes in vivo were reversed by AR-V7 knockout. Pladienolide B, an inhibitor of a splicing modulator of the SF3b complex, suppressed the growth of tumors addicted to high SF3B2 expression. These findings support the idea that alteration of the splicing pattern by high SF3B2 expression is one mechanism underlying prostate cancer progression and therapeutic resistance. This study also provides evidence supporting SF3B2 as a candidate therapeutic target for treating patients with cancer. SIGNIFICANCE: RNA splicing factor SF3B2 is essential for the generation of an androgen receptor (AR) variant that renders prostate cancer cells resistant to AR-targeting therapy.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/20/5204/F1.large.jpg.


Asunto(s)
Adenocarcinoma/genética , Regulación Neoplásica de la Expresión Génica/genética , Proteínas de Neoplasias/fisiología , Neoplasias de la Próstata/genética , Factores de Empalme de ARN/fisiología , Empalme del ARN/fisiología , ARN Mensajero/metabolismo , ARN Neoplásico/metabolismo , Receptores Androgénicos/genética , Adenocarcinoma/patología , Secuencias de Aminoácidos , Animales , Simulación por Computador , Progresión de la Enfermedad , Compuestos Epoxi/farmacología , Exones/genética , Humanos , Macrólidos/farmacología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Invasividad Neoplásica , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Orquiectomía , Fenotipo , Neoplasias de la Próstata/patología , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Empalme del ARN/efectos de los fármacos , Empalme del ARN/genética , Factores de Empalme de ARN/antagonistas & inhibidores , Factores de Empalme de ARN/genética , ARN Mensajero/genética , ARN Neoplásico/genética , Receptores Androgénicos/biosíntesis , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Biochem Biophys Res Commun ; 516(3): 928-933, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31277943

RESUMEN

In Western societies where food is abundant, these excess nutrients are stored as fats mainly in adipose tissue. Fats are stored in structures known as lipid droplets, and a genome-wide screen performed in Drosophila cells has identified several genes that are important for the formation of these droplets. One group of genes found during this screen included those that regulate mRNA splicing. Previous work from our lab has identified some splicing factors that play a role in regulating fat storage; however, the full complement of splicing proteins that regulate lipid metabolism is still unknown. In this study, the levels of a number of serine-arginine (SR) domain containing splicing factors (RSF1, RBP1, RBP1-like, SF2 and Srp-54) were decreased using RNAi in the adult fat body to assess their role in the control of Drosophila metabolism. Decreasing SF2 and RBP1 showed increased triglycerides, while inducing RNAi towards RSF1, RBP1-Like and Srp-54 had no effect on triglycerides. Interestingly, the increased triglyceride phenotype in the SF2-RNAi flies was due to an increase in the amount of fat stored per cell while the RBP1-RNAi flies have more fat cells. In addition, the splicing of the ß-oxidation enzyme, CPT1, was altered in the SF2-RNAi flies potentially promoting the increased triglycerides in these animals. Together, this study identifies novel splicing factors responsible for the regulation of lipid storage in the Drosophila fat body and contributes to our understanding of the mechanisms, which influence the regulation of fat storage in adipose-like cells.


Asunto(s)
Empalme Alternativo , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Cuerpo Adiposo/metabolismo , Factores de Empalme de ARN/genética , Tejido Adiposo/metabolismo , Animales , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Gotas Lipídicas/metabolismo , Metabolismo de los Lípidos/genética , Longevidad/genética , Factores de Empalme de ARN/antagonistas & inhibidores , Factores de Empalme de ARN/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Triglicéridos/metabolismo
18.
Artif Cells Nanomed Biotechnol ; 47(1): 1273-1280, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30963795

RESUMEN

Pladienolide B is a potent cancer cell growth inhibitor that targets the SF3b1 subunit of the spliceosome. There is considerable interest in the compound as a tool to study SF3b1 function in cancer. However, so far little information is available on the molecular mechanism of SF3b1 eliciting apoptosis in cancer cells. Here, we investigated the molecular mechanism of SF3b1 eliciting apoptosis in human cervical carcinoma cells. We demonstrated that inhibition of SF3b1 by pladienolide B inhibited proliferation of HeLa cells at low nanomolar concentrations in a dose- and time-dependent manner. It also induced G2/M phase arrest and significant rise of apoptotic cells. Moreover, it is indicated that inhibition of SF3b1 by pladienolide B induced Tap73/ΔNp73 expression and consequently down-regulated Bax/Bcl-2 ratio, cytochrome c release and caspase-3 expression. Thus, our results showed that SF3b1 plays a pivotal role in cycle arrest, apoptosis induction, and p73 splicing in human cervical carcinoma cells, suggesting that SF3b1 could be used as a potential candidate for cervical cancer therapy.


Asunto(s)
Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Compuestos Epoxi/farmacología , Macrólidos/farmacología , Fosfoproteínas/antagonistas & inhibidores , Factores de Empalme de ARN/antagonistas & inhibidores , Empalme del ARN/efectos de los fármacos , Proteína Tumoral p73/genética , Neoplasias del Cuello Uterino/patología , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/patología
19.
Nat Commun ; 10(1): 1590, 2019 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-30962446

RESUMEN

Alternative splicing, a fundamental step in gene expression, is deregulated in many diseases. Splicing factors (SFs), which regulate this process, are up- or down regulated or mutated in several diseases including cancer. To date, there are no inhibitors that directly inhibit the activity of SFs. We designed decoy oligonucleotides, composed of several repeats of a RNA motif, which is recognized by a single SF. Here we show that decoy oligonucleotides targeting splicing factors RBFOX1/2, SRSF1 and PTBP1, can specifically bind to their respective SFs and inhibit their splicing and biological activities both in vitro and in vivo. These decoy oligonucleotides present an approach to specifically downregulate SF activity in conditions where SFs are either up-regulated or hyperactive.


Asunto(s)
Ribonucleoproteínas Nucleares Heterogéneas/genética , Oligonucleótidos/farmacología , Proteína de Unión al Tracto de Polipirimidina/genética , Factores de Empalme de ARN/genética , Factores de Empalme Serina-Arginina/genética , Empalme Alternativo , Animales , Animales Modificados Genéticamente , Sitios de Unión , Glioblastoma/genética , Glioblastoma/patología , Células HEK293 , Ribonucleoproteínas Nucleares Heterogéneas/antagonistas & inhibidores , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/genética , Músculo Esquelético/crecimiento & desarrollo , Degradación de ARNm Mediada por Codón sin Sentido , Oligonucleótidos/química , Oligonucleótidos/metabolismo , Proteína de Unión al Tracto de Polipirimidina/antagonistas & inhibidores , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Factores de Empalme de ARN/antagonistas & inhibidores , Factores de Empalme de ARN/metabolismo , Factores de Empalme Serina-Arginina/antagonistas & inhibidores , Factores de Empalme Serina-Arginina/metabolismo , Secuencias Repetidas en Tándem , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra/embriología , Pez Cebra/genética
20.
J Biol Chem ; 294(22): 8760-8772, 2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-31010829

RESUMEN

The cohesin complex regulates sister chromatid cohesion, chromosome organization, gene expression, and DNA repair. Cohesin is a ring complex composed of four core subunits and seven regulatory subunits. In an effort to comprehensively identify additional cohesin-interacting proteins, we used gene editing to introduce a dual epitope tag into the endogenous allele of each of 11 known components of cohesin in cultured human cells, and we performed MS analyses on dual-affinity purifications. In addition to reciprocally identifying all known components of cohesin, we found that cohesin interacts with a panoply of splicing factors and RNA-binding proteins (RBPs). These included diverse components of the U4/U6.U5 tri-small nuclear ribonucleoprotein complex and several splicing factors that are commonly mutated in cancer. The interaction between cohesin and splicing factors/RBPs was RNA- and DNA-independent, occurred in chromatin, was enhanced during mitosis, and required RAD21. Furthermore, cohesin-interacting splicing factors and RBPs followed the cohesin cycle and prophase pathway of cell cycle-regulated interactions with chromatin. Depletion of cohesin-interacting splicing factors and RBPs resulted in aberrant mitotic progression. These results provide a comprehensive view of the endogenous human cohesin interactome and identify splicing factors and RBPs as functionally significant cohesin-interacting proteins.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Mitosis , Proteómica , Factores de Empalme de ARN/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Microscopía Fluorescente , Unión Proteica , Mapas de Interacción de Proteínas , Interferencia de ARN , Factores de Empalme de ARN/antagonistas & inhibidores , Factores de Empalme de ARN/genética , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Cohesinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...