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










Base de datos
Intervalo de año de publicación
1.
Genes (Basel) ; 12(9)2021 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-34573320

RESUMEN

Pulmonary arterial hypertension (PAH) is a rare cardiovascular disease with very high mortality rate. The currently available therapeutic strategies, which improve symptoms, cannot fundamentally reverse the condition. Thus, new therapeutic strategies need to be established. Our research analyzed three microarray datasets of lung tissues from human PAH samples retrieved from the Gene Expression Omnibus (GEO) database. We combined two datasets for subsequent analyses, with the batch effects removed. In the merged dataset, 542 DEGs were identified and the key module relevant to PAH was selected using WGCNA. GO and KEGG analyses of DEGs and the key module indicated that the pre-ribosome, ribosome biogenesis, centriole, ATPase activity, helicase activity, hypertrophic cardiomyopathy, melanoma, and dilated cardiomyopathy pathways are involved in PAH. With the filtering standard (|MM| > 0.95 and |GS| > 0.90), 70 hub genes were identified. Subsequently, five candidate marker genes (CDC5L, AP3B1, ZFYVE16, DDX46, and PHAX) in the key module were found through overlapping with the top thirty genes calculated by two different methods in CytoHubb. Two of them (CDC5L and DDX46) were found to be significantly upregulated both in the merged dataset and the validating dataset in PAH patients. Meanwhile, expression of the selected genes in lung from PAH chicken measured by qRT-PCR and the ROC curve analyses further verified the potential marker genes' predictive value for PAH. In conclusion, CDC5L and DDX46 may be marker genes and potential therapeutic targets for PAH.


Asunto(s)
Proteínas de Ciclo Celular/genética , ARN Helicasas DEAD-box/genética , Hipertensión Arterial Pulmonar/diagnóstico , Proteínas de Unión al ARN/genética , Ribonucleoproteína Nuclear Pequeña U2/genética , Animales , Biomarcadores/análisis , Biomarcadores/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo , Pollos , Biología Computacional , ARN Helicasas DEAD-box/antagonistas & inhibidores , ARN Helicasas DEAD-box/metabolismo , Conjuntos de Datos como Asunto , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Redes Reguladoras de Genes/efectos de los fármacos , Humanos , Pulmón/patología , Análisis por Micromatrices , Terapia Molecular Dirigida/métodos , Valor Predictivo de las Pruebas , Mapas de Interacción de Proteínas/efectos de los fármacos , Mapas de Interacción de Proteínas/genética , Hipertensión Arterial Pulmonar/tratamiento farmacológico , Hipertensión Arterial Pulmonar/genética , Hipertensión Arterial Pulmonar/patología , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/metabolismo , Curva ROC , Ribonucleoproteína Nuclear Pequeña U2/antagonistas & inhibidores , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Regulación hacia Arriba/efectos de los fármacos
2.
Curr Hematol Malig Rep ; 11(6): 408-415, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27492253

RESUMEN

Genome sequencing of primary cells from patients with myelodysplastic syndromes (MDS) led to the identification of recurrent heterozygous mutations in gene encoding components of the spliceosome, the cellular machinery which processes pre-messenger RNA (mRNA) to mature mRNA during gene transcription. Splicing mutations are mutually exclusive with one another and collectively represent the most common mutation class in MDS, occurring in approximately 60 % of patients overall and more than 80 % of those with ring sideroblasts. Evidence from animal models suggests that homozygous splicing mutations are lethal, and that in heterozygously mutated models, any further disruption of splicing triggers apoptosis and cell death. MDS cells with spliceosome mutations are thus uniquely vulnerable to therapies targeting splicing, which may be tolerated by healthy cells. The spliceosome is emerging as a novel therapeutic target in MDS and related myeloid neoplasms, with the first clinical trial of a splicing modulator opening in 2016.


Asunto(s)
Neoplasias Hematológicas/patología , Síndromes Mielodisplásicos/patología , Trastornos Mieloproliferativos/patología , Empalme del ARN , Animales , Compuestos Epoxi/uso terapéutico , Neoplasias Hematológicas/tratamiento farmacológico , Neoplasias Hematológicas/genética , Humanos , Indoles/farmacología , Macrólidos/uso terapéutico , Mutación , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/genética , Trastornos Mieloproliferativos/tratamiento farmacológico , Trastornos Mieloproliferativos/genética , Empalme del ARN/efectos de los fármacos , Empalme del ARN/genética , Factores de Empalme de ARN/genética , Factores de Empalme de ARN/metabolismo , Ribonucleoproteína Nuclear Pequeña U2/antagonistas & inhibidores , Ribonucleoproteína Nuclear Pequeña U2/genética , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Empalmosomas/efectos de los fármacos , Empalmosomas/metabolismo
3.
RNA ; 22(3): 350-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26742993

RESUMEN

The protein SF3B1 is a core component of the spliceosome, the large ribonucleoprotein complex responsible for pre-mRNA splicing. Interest in SF3B1 intensified when tumor exome sequencing revealed frequent specific SF3B1 mutations in a variety of neoplasia and when SF3B1 was identified as the target of three different cancer cell growth inhibitors. A better mechanistic understanding of SF3B1's role in splicing is required to capitalize on these discoveries. Using the inhibitor compounds, we probed SF3B1 function in the spliceosome in an in vitro splicing system. Formerly, the inhibitors were shown to block early steps of spliceosome assembly, consistent with a previously determined role of SF3B1 in intron recognition. We now report that SF3B1 inhibitors also interfere with later events in the spliceosome cycle, including exon ligation. These observations are consistent with a requirement for SF3B1 throughout the splicing process. Additional experiments aimed at understanding how three structurally distinct molecules produce nearly identical effects on splicing revealed that inactive analogs of each compound interchangeably compete with the active inhibitors to restore splicing. The competition indicates that all three types of compounds interact with the same site on SF3B1 and likely interfere with its function by the same mechanism, supporting a shared pharmacophore model. It also suggests that SF3B1 inhibition does not result from binding alone, but is consistent with a model in which the compounds affect a conformational change in the protein. Together, our studies reveal new mechanistic insight into SF3B1 as a principal player in the spliceosome and as a target of inhibitor compounds.


Asunto(s)
Fosfoproteínas/antagonistas & inhibidores , Precursores del ARN/genética , Empalme del ARN , ARN Mensajero/genética , Ribonucleoproteína Nuclear Pequeña U2/antagonistas & inhibidores , Adenosina Trifosfato/metabolismo , Sitios de Unión , Exones , Humanos , Factores de Empalme de ARN
4.
Cell Rep ; 14(3): 598-610, 2016 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-26776507

RESUMEN

Ewing sarcoma cells depend on the EWS-FLI1 fusion transcription factor for cell survival. Using an assay of EWS-FLI1 activity and genome-wide RNAi screening, we have identified proteins required for the processing of the EWS-FLI1 pre-mRNA. We show that Ewing sarcoma cells harboring a genomic breakpoint that retains exon 8 of EWSR1 require the RNA-binding protein HNRNPH1 to express in-frame EWS-FLI1. We also demonstrate the sensitivity of EWS-FLI1 fusion transcripts to the loss of function of the U2 snRNP component, SF3B1. Disrupted splicing of the EWS-FLI1 transcript alters EWS-FLI1 protein expression and EWS-FLI1-driven expression. Our results show that the processing of the EWS-FLI1 fusion RNA is a potentially targetable vulnerability in Ewing sarcoma cells.


Asunto(s)
Proteínas de Fusión Oncogénica/metabolismo , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína EWS de Unión a ARN/metabolismo , Secuencia de Bases , Sitios de Unión , Proteínas de Unión a Calmodulina/antagonistas & inhibidores , Proteínas de Unión a Calmodulina/genética , Proteínas de Unión a Calmodulina/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Exones , Regulación Neoplásica de la Expresión Génica , Ribonucleoproteína Heterogénea-Nuclear Grupo F-H/antagonistas & inhibidores , Ribonucleoproteína Heterogénea-Nuclear Grupo F-H/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo F-H/metabolismo , Humanos , Proteínas de Microfilamentos/antagonistas & inhibidores , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteínas de Fusión Oncogénica/genética , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteína Proto-Oncogénica c-fli-1/antagonistas & inhibidores , Proteína Proto-Oncogénica c-fli-1/genética , Interferencia de ARN , Precursores del ARN/metabolismo , Empalme del ARN , Factores de Empalme de ARN , ARN Interferente Pequeño/metabolismo , Proteína EWS de Unión a ARN/antagonistas & inhibidores , Proteína EWS de Unión a ARN/genética , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Ribonucleoproteína Nuclear Pequeña U2/antagonistas & inhibidores , Ribonucleoproteína Nuclear Pequeña U2/genética , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Sarcoma de Ewing/patología , Transactivadores , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
5.
Leukemia ; 30(2): 351-60, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26488112

RESUMEN

The pro-survival Bcl-2 family member Mcl-1 is expressed in chronic lymphocytic leukaemia (CLL), with high expression correlated with progressive disease. The spliceosome inhibitor spliceostatin A (SSA) is known to regulate Mcl-1 and so here we assessed the ability of SSA to elicit apoptosis in CLL. SSA induced apoptosis of CLL cells at low nanomolar concentrations in a dose- and time-dependent manner, but independently of SF3B1 mutational status, IGHV status and CD38 or ZAP70 expression. However, normal B and T cells were less sensitive than CLL cells (P=0.006 and P<0.001, respectively). SSA altered the splicing of anti-apoptotic MCL-1(L) to MCL-1(s) in CLL cells coincident with induction of apoptosis. Overexpression studies in Ramos cells suggested that Mcl-1 was important for SSA-induced killing since its expression inversely correlated with apoptosis (P=0.001). IL4 and CD40L, present in patient lymph nodes, are known to protect tumour cells from apoptosis and significantly inhibited SSA, ABT-263 and ABT-199 induced killing following administration to CLL cells (P=0.008). However, by combining SSA with the Bcl-2/Bcl-x(L) antagonists ABT-263 or ABT-199, we were able to overcome this pro-survival effect. We conclude that SSA combined with Bcl-2/Bcl-x(L) antagonists may have therapeutic utility for CLL.


Asunto(s)
Apoptosis/efectos de los fármacos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Fosfoproteínas/antagonistas & inhibidores , Piranos/farmacología , Ribonucleoproteína Nuclear Pequeña U2/antagonistas & inhibidores , Compuestos de Espiro/farmacología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Humanos , Interleucina-4/farmacología , Leucemia Linfocítica Crónica de Células B/patología , Mutación , Fosfoproteínas/genética , Empalme del ARN , Factores de Empalme de ARN , Ribonucleoproteína Nuclear Pequeña U2/genética , Microambiente Tumoral , Proteína bcl-X/antagonistas & inhibidores
7.
PLoS Genet ; 11(2): e1004932, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25658809

RESUMEN

The innate immune response plays a key role in fighting infection by activating inflammation and stimulating the adaptive immune response. However, chronic activation of innate immunity can contribute to the pathogenesis of many diseases with an inflammatory component. Thus, various negatively acting factors turn off innate immunity subsequent to its activation to ensure that inflammation is self-limiting and to prevent inflammatory disease. These negatively acting pathways include the production of inhibitory acting alternate proteins encoded by alternative mRNA splice forms of genes in Toll-like receptor (TLR) signaling pathways. We previously found that the SF3a mRNA splicing complex was required for a robust innate immune response; SF3a acts to promote inflammation in part by inhibiting the production of a negatively acting splice form of the TLR signaling adaptor MyD88. Here we inhibit SF3a1 using RNAi and subsequently perform an RNAseq study to identify the full complement of genes and splicing events regulated by SF3a in murine macrophages. Surprisingly, in macrophages, SF3a has significant preference for mRNA splicing events within innate immune signaling pathways compared with other biological pathways, thereby affecting the splicing of specific genes in the TLR signaling pathway to modulate the innate immune response.


Asunto(s)
Inmunidad Adaptativa/inmunología , Empalme Alternativo/genética , Inmunidad Innata/genética , Ribonucleoproteína Nuclear Pequeña U2/genética , Receptor Toll-Like 4/genética , Empalme Alternativo/inmunología , Animales , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Intrones/genética , Macrófagos/inmunología , Macrófagos/patología , Ratones , Empalme del ARN/genética , Factores de Empalme de ARN , ARN Mensajero/genética , ARN Interferente Pequeño , Ribonucleoproteína Nuclear Pequeña U2/antagonistas & inhibidores , Ribonucleoproteína Nuclear Pequeña U2/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptor Toll-Like 4/inmunología
8.
ACS Chem Biol ; 10(4): 914-24, 2015 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-25560473

RESUMEN

Alternative splicing is a critical step where a limited number of human genes generate a complex and diverse proteome. Various diseases, including inherited diseases with abnormalities in the "genome code," have been found to result in an aberrant mis-spliced "transcript code" with correlation to the resulting phenotype. Chemical compound-based and nucleic acid-based strategies are trying to target this mis-spliced "transcript code". We will briefly mention about how to obtain splicing-modifying-compounds by high-throughput screening and overview of what is known about compounds that modify splicing pathways. The main focus will be on RNA-binding protein kinase inhibitors. In the main text, we will refer to diseases where splicing-modifying-compounds have been intensively investigated, with comparison to nucleic acid-based strategies. The information on their involvement in mis-splicing as well as nonsplicing events will be helpful in finding better compounds with less off-target effects for future implications in mis-splicing therapy.


Asunto(s)
Empalme Alternativo , Terapia Molecular Dirigida/métodos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas de Unión al ARN/antagonistas & inhibidores , Empalme Alternativo/efectos de los fármacos , Animales , Citocininas/farmacología , Síndrome de Down/tratamiento farmacológico , Disautonomía Familiar/tratamiento farmacológico , Disautonomía Familiar/fisiopatología , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/genética , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Atrofia Muscular Espinal/tratamiento farmacológico , Atrofia Muscular Espinal/genética , Distrofia Muscular de Duchenne/tratamiento farmacológico , Síndromes Mielodisplásicos/tratamiento farmacológico , Fosfoproteínas/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Factores de Empalme de ARN , Ribonucleoproteína Nuclear Pequeña U2/antagonistas & inhibidores , Quinasas DyrK
9.
J Pathol ; 235(4): 571-80, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25424858

RESUMEN

Mutations in genes encoding proteins involved in RNA splicing have been found to occur at relatively high frequencies in several tumour types including myelodysplastic syndromes, chronic lymphocytic leukaemia, uveal melanoma, and pancreatic cancer, and at lower frequencies in breast cancer. To investigate whether dysfunction in RNA splicing is implicated in the pathogenesis of breast cancer, we performed a re-analysis of published exome and whole genome sequencing data. This analysis revealed that mutations in spliceosomal component genes occurred in 5.6% of unselected breast cancers, including hotspot mutations in the SF3B1 gene, which were found in 1.8% of unselected breast cancers. SF3B1 mutations were significantly associated with ER-positive disease, AKT1 mutations, and distinct copy number alterations. Additional profiling of hotspot mutations in a panel of special histological subtypes of breast cancer showed that 16% and 6% of papillary and mucinous carcinomas of the breast harboured the SF3B1 K700E mutation. RNA sequencing identified differentially spliced events expressed in tumours with SF3B1 mutations including the protein coding genes TMEM14C, RPL31, DYNL11, UQCC, and ABCC5, and the long non-coding RNA CRNDE. Moreover, SF3B1 mutant cell lines were found to be sensitive to the SF3b complex inhibitor spliceostatin A and treatment resulted in perturbation of the splicing signature. Albeit rare, SF3B1 mutations result in alternative splicing events, and may constitute drivers and a novel therapeutic target in a subset of breast cancers.


Asunto(s)
Adenocarcinoma Mucinoso/genética , Empalme Alternativo/genética , Neoplasias de la Mama/genética , Carcinoma Papilar/genética , Mutación , Fosfoproteínas/genética , Ribonucleoproteína Nuclear Pequeña U2/genética , Adenocarcinoma Mucinoso/tratamiento farmacológico , Adenocarcinoma Mucinoso/metabolismo , Adenocarcinoma Mucinoso/patología , Empalme Alternativo/efectos de los fármacos , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinoma Papilar/tratamiento farmacológico , Carcinoma Papilar/metabolismo , Carcinoma Papilar/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Terapia Molecular Dirigida , Fenotipo , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/metabolismo , Piranos/farmacología , Interferencia de ARN , Factores de Empalme de ARN , Receptores de Estrógenos/metabolismo , Ribonucleoproteína Nuclear Pequeña U2/antagonistas & inhibidores , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Compuestos de Espiro/farmacología , Transfección
10.
Org Lett ; 16(21): 5560-3, 2014 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-25376106

RESUMEN

A total synthesis of the natural product 6-deoxypladienolide D (1) has been achieved. Two noteworthy attributes of the synthesis are (1) a late-stage allylic oxidation which proceeds with full chemo-, regio-, and diastereoselectivity and (2) the development of a scalable and cost-effective synthetic route to support drug discovery efforts. 6-Deoxypladienolide D (1) demonstrates potent growth inhibition in a mutant SF3B1 cancer cell line, high binding affinity to the SF3b complex, and inhibition of pre-mRNA splicing.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Línea Celular Tumoral/química , Línea Celular Tumoral/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Compuestos Epoxi/síntesis química , Compuestos Epoxi/metabolismo , Macrólidos/síntesis química , Macrólidos/metabolismo , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/química , Empalme del ARN/efectos de los fármacos , Ribonucleoproteína Nuclear Pequeña U2/antagonistas & inhibidores , Ribonucleoproteína Nuclear Pequeña U2/química , Antineoplásicos/química , Sitios de Unión , Compuestos Epoxi/química , Humanos , Macrólidos/química , Factores de Empalme de ARN
11.
EMBO Rep ; 15(9): 948-55, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25092791

RESUMEN

Sister chromatid cohesion, which depends on cohesin, is essential for the faithful segregation of replicated chromosomes. Here, we report that splicing complex Prp19 is essential for cohesion in both G2 and mitosis, and consequently for the proper progression of the cell through mitosis. Inactivation of splicing factors SF3a120 and U2AF65 induces similar cohesion defects to Prp19 complex inactivation. Our data indicate that these splicing factors are all required for the accumulation of cohesion factor Sororin, by facilitating the proper splicing of its pre-mRNA. Finally, we show that ectopic expression of Sororin corrects defective cohesion caused by Prp19 complex inactivation. We propose that the Prp19 complex and the splicing machinery contribute to the establishment of cohesion by promoting Sororin accumulation during S phase, and are, therefore, essential to the maintenance of genome stability.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas de Ciclo Celular/genética , Enzimas Reparadoras del ADN/genética , Proteínas Nucleares/genética , Precursores del ARN/genética , Empalme del ARN/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cromátides/genética , Segregación Cromosómica/genética , Enzimas Reparadoras del ADN/antagonistas & inhibidores , Enzimas Reparadoras del ADN/biosíntesis , Regulación de la Expresión Génica , Inestabilidad Genómica , Células HeLa , Humanos , Mitosis/genética , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/biosíntesis , Factores de Empalme de ARN , Ribonucleoproteína Nuclear Pequeña U2/antagonistas & inhibidores , Ribonucleoproteínas/antagonistas & inhibidores , Factor de Empalme U2AF
12.
Sci Rep ; 4: 6098, 2014 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-25139387

RESUMEN

ABT-737 inhibits the anti-apoptotic proteins B-cell lymphoma 2 (BCL-2) and BCL-X(L). Meayamycin B switches the splicing pattern of myeloid cell leukemia factor 1 (MCL1) pre-mRNA. Specifically, inhibition of splicing factor 3B subunit 1 (SF3B1) with meayamycin B promotes the generation of the proapoptotic, short splicing variant (MCL1-S) and diminishes the antiapoptotic, long variant (MCL1-L). This action was previously associated with the cytotoxicity of meayamycin B in non-small cell lung carcinoma cell lines. ABT-737 induced apoptosis in response to an ablation of MCL1-L by meayamycin B. In this study, we further exploited this synergistic combination in head and neck squamous cell carcinoma (HNSCC), up to 90% of which overexpress MCL1 and BCL-X(L). In a panel of seven HNSCC cell lines, the combination of meayamycin B and ABT-737 rapidly triggered a Bax/Bak-mediated apoptosis that overcame the resistance from HPV16-positive HNSCC against each agent alone. Both RT-PCR and Western blotting showed that meayamycin B up-regulated MCL1-S and down-regulated MCL1-L. Significantly, we discovered that SF3B1 was involved in the splicing of oncogenic HPV16 E6 to produce non-oncogenic HPV16 E6*, indicating that SF3B1 may inhibit HPV16-induced tumorigenesis.


Asunto(s)
Papillomavirus Humano 16/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Fosfoproteínas/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Ribonucleoproteína Nuclear Pequeña U2/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Compuestos de Bifenilo/toxicidad , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Línea Celular Tumoral , Sinergismo Farmacológico , Células HeLa , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Neoplasias de Cabeza y Cuello/virología , Humanos , Morfolinas/toxicidad , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Nitrofenoles/toxicidad , Proteínas Oncogénicas Virales/genética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Piperazinas/toxicidad , Piranos/toxicidad , Interferencia de ARN , Empalme del ARN/efectos de los fármacos , Factores de Empalme de ARN , ARN Interferente Pequeño/metabolismo , Proteínas Represoras/genética , Ribonucleoproteína Nuclear Pequeña U2/genética , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Sulfonamidas/toxicidad , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Proteína bcl-X/metabolismo
13.
J Biol Chem ; 289(4): 1938-47, 2014 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-24302718

RESUMEN

Pladienolide B (PB) is a potent cancer cell growth inhibitor that targets the SF3B1 subunit of the spliceosome. There is considerable interest in the compound as a potential chemotherapeutic, as well as a tool to study SF3B1 function in splicing and cancer development. The molecular structure of PB, a bacterial natural product, contains a 12-member macrolide ring with an extended epoxide-containing side chain. Using a novel concise enantioselective synthesis, we created a series of PB structural analogs and the structurally related compound herboxidiene. We show that two methyl groups in the PB side chain, as well as a feature of the macrolide ring shared with herboxidiene, are required for splicing inhibition in vitro. Unexpectedly, we find that the epoxy group contributes only modestly to PB potency and is not absolutely necessary for activity. The orientations of at least two chiral centers off the macrolide ring have no effect on PB activity. Importantly, the ability of analogs to inhibit splicing in vitro directly correlated with their effects in a series of cellular assays. Those effects likely arise from inhibition of some, but not all, endogenous splicing events in cells, as previously reported for the structurally distinct SF3B1 inhibitor spliceostatin A. Together, our data support the idea that the impact of PB on cells is derived from its ability to impair the function of SF3B1 in splicing and also demonstrate that simplification of the PB scaffold is feasible.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Epoxi/química , Macrólidos/química , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/metabolismo , Fosfoproteínas/antagonistas & inhibidores , Empalme del ARN/efectos de los fármacos , Ribonucleoproteína Nuclear Pequeña U2/antagonistas & inhibidores , Antineoplásicos/química , Células HeLa , Humanos , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Piranos/química , Piranos/farmacología , Factores de Empalme de ARN , Ribonucleoproteína Nuclear Pequeña U2/química , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Compuestos de Espiro/química , Compuestos de Espiro/farmacología
14.
ACS Chem Biol ; 8(5): 895-900, 2013 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-23485022

RESUMEN

The myeloid cell leukemia-1 (MCL1) gene encodes antiapoptotic Mcl-1(L) and proapoptotic Mcl-1(S) proteins. In cancer, the Mcl-1(L)/Mcl-1(S) ratio is very high, accounting for the antiapoptotic nature of cancer cells. As such, reducing this ratio can render the cancer cells prone to apoptosis. The Mcl-1(L)/Mcl-1(S) ratio is determined in the alternative pre-mRNA splicing step that is regulated by splicing factor 3B1 (SF3B1). Here, we report that meayamycin B, a potent inhibitor of SF3B1, reversed the dominant isoform from Mcl-1(L) to Mcl-1(S) at the mRNA and protein levels. The resulting proapoptotic cellular environment was further exploited; when meayamycin B was combined with Bcl-x(L) inhibitor ABT-737, the combination treatment triggered apoptosis in nonsmall cell lung cancer A549 and H1299 cells that were otherwise resistant to ABT-737. These results demonstrate that perturbation of the MCL1 splicing with small molecule inhibitors of SF3B1 provides a means to sensitize cancer cells toward Bcl-x(L) inhibitors.


Asunto(s)
Empalme Alternativo/efectos de los fármacos , Compuestos Epoxi/farmacología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Piranos/farmacología , Apoptosis/genética , Compuestos de Bifenilo/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Morfolinas/farmacología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Nitrofenoles/farmacología , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/metabolismo , Piperazinas/farmacología , Isoformas de Proteínas , Factores de Empalme de ARN , Ribonucleoproteína Nuclear Pequeña U2/antagonistas & inhibidores , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Sulfonamidas/farmacología , Proteína bcl-X/antagonistas & inhibidores
15.
Chembiochem ; 14(1): 49-52, 2013 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-23172726

RESUMEN

Name your splice: FR901464 analogues and herboxidiene inhibit constitutive splicing, most likely by inhibiting spliceosomal subunit SF3b. A parallel comparison of these compounds in a cell-based assay system showed meayamycin B as the most potent splicing inhibitor among these small molecules.


Asunto(s)
Morfolinas/farmacología , Piranos/farmacología , Empalme del ARN/efectos de los fármacos , Ribonucleoproteína Nuclear Pequeña U2/antagonistas & inhibidores , Animales , Exones/genética , Genes Reporteros/genética , Células HEK293 , Humanos , Intrones/genética , Luciferasas de Luciérnaga/genética , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Triosa-Fosfato Isomerasa/genética
16.
ACS Chem Biol ; 6(3): 229-33, 2011 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-21138297

RESUMEN

GEX1A is a microbial product with antitumor activity. HeLa cells cultured with GEX1A accumulated p27(Kip) and its C-terminally truncated form p27*. GEX1A inhibited the pre-mRNA splicing of p27, producing p27* from the unspliced mRNA containing the first intron. p27* lacked the site required for E3 ligase-mediated proteolysis of p27, leading to its accumulation in GEX1A-treated cells. The accumulated p27* was able to bind to and inhibit the cyclin E-Cdk2 complex that causes E3 ligase-mediated degradation of p27, which probably triggers the accumulation of p27. By using a series of photoaffinity-labeling derivatives of GEX1A, we found that GEX1A targeted SAP155 protein, a subunit of SF3b responsible for pre-mRNA splicing. The linker length between the GEX1A pharmacophore and the photoreactive group was critical for detection of the GEX1A-binding protein. GEX1A serves as a novel splicing inhibitor that specifically impairs the SF3b function by binding to SAP155.


Asunto(s)
Antineoplásicos/farmacología , Productos Biológicos/farmacología , Alcoholes Grasos/farmacología , Fosfoproteínas/antagonistas & inhibidores , Piranos/farmacología , Ribonucleoproteína Nuclear Pequeña U2/antagonistas & inhibidores , Antineoplásicos/química , Sitios de Unión/efectos de los fármacos , Productos Biológicos/química , Células Cultivadas , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/antagonistas & inhibidores , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/biosíntesis , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Alcoholes Grasos/química , Células HeLa , Humanos , Estructura Molecular , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Piranos/química , Precursores del ARN/antagonistas & inhibidores , Precursores del ARN/genética , Empalme del ARN/efectos de los fármacos , Empalme del ARN/genética , Factores de Empalme de ARN , Ribonucleoproteína Nuclear Pequeña U2/química , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Relación Estructura-Actividad
17.
Oncogene ; 28(20): 2065-73, 2009 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-19377511

RESUMEN

Ddx42p is a recently characterized mammalian DEAD box protein with unknown cellular function. We found that in human cells Ddx42p physically interacts with ASPP2, a major apoptosis inducer known to enhance p53 transactivation of proapoptotic genes. The proteins interact via a domain within the carboxy-terminal part of Ddx42p and a mid-amino-terminal sequence as well as the ankyrin-SH3 region of ASPP2. Overexpression of Ddx42p interferes with apoptosis induction by ASPP2, whereas Ddx42p knockdown reduces the survival rate of cultured human cells. In addition, ASPP2 is found in cytoplasm and nucleus at low Ddx42p level, and predominantly in cytoplasm at high concentration of Ddx42p, respectively. Our results show that Ddx42p is capable of modulating ASPP2 function.


Asunto(s)
Apoptosis/fisiología , Proteínas Portadoras/metabolismo , ARN Helicasas DEAD-box/metabolismo , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Ancirinas/metabolismo , Proteínas Reguladoras de la Apoptosis , Proteínas Portadoras/genética , Núcleo Celular/metabolismo , Citoplasma/metabolismo , ARN Helicasas DEAD-box/antagonistas & inhibidores , ARN Helicasas DEAD-box/genética , Técnica del Anticuerpo Fluorescente , Biblioteca de Genes , Células HeLa , Humanos , Inmunoprecipitación , Transporte de Proteínas , ARN Interferente Pequeño/farmacología , Ribonucleoproteína Nuclear Pequeña U2/antagonistas & inhibidores , Ribonucleoproteína Nuclear Pequeña U2/genética , Activación Transcripcional , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Técnicas del Sistema de Dos Híbridos , Dominios Homologos src
18.
RNA ; 15(1): 153-75, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19029308

RESUMEN

The removal of intervening sequences from a primary RNA transcript is catalyzed by the spliceosome, a large complex consisting of five small nuclear (sn) RNAs and more than 150 proteins. At the start of the splicing cycle, the spliceosome assembles anew onto each pre-mRNA intron in an ordered process. Here, we show that several small-molecule inhibitors of protein acetylation/deacetylation block the splicing cycle: by testing a small number of bioactive compounds, we found that three small-molecule inhibitors of histone acetyltransferases (HATs), as well as three small-molecule inhibitors of histone deacetylases (HDACs), block pre-mRNA splicing in vitro. By purifying and characterizing the stalled spliceosomes, we found that the splicing cycle is blocked at distinct stages by different inhibitors: two inhibitors allow only the formation of A-like spliceosomes (as determined by the size of the stalled complexes and their snRNA composition), while the other compounds inhibit activation for catalysis after incorporation of all U snRNPs into the spliceosome. Mass-spectrometric analysis of affinity-purified stalled spliceosomes indicated that the intermediates differ in protein composition both from each other and from previously characterized native A and B splicing complexes. This suggests that the stalled complexes represent hitherto unobserved intermediates of spliceosome assembly.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Histona Acetiltransferasas/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas , Empalme del ARN/efectos de los fármacos , Empalmosomas/metabolismo , Acetilación , Catálisis , Células HeLa , Histona Acetiltransferasas/metabolismo , Histona Desacetilasas/metabolismo , Humanos , Precursores del ARN/metabolismo , Ribonucleoproteína Nuclear Pequeña U2/antagonistas & inhibidores , Ribonucleoproteína Nuclear Pequeña U2/genética , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/antagonistas & inhibidores , Ribonucleoproteínas Nucleares Pequeñas/genética , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Empalmosomas/efectos de los fármacos
20.
Nat Chem Biol ; 3(9): 576-83, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17643111

RESUMEN

The removal of intervening sequences from transcripts is catalyzed by the spliceosome, a multicomponent complex that assembles on the newly synthesized pre-mRNA. Pre-mRNA translation in the cytoplasm leads to the generation of aberrant proteins that are potentially harmful. Therefore, tight control to prevent undesired pre-mRNA export from the nucleus and its subsequent translation is an essential requirement for reliable gene expression. Here, we show that the natural product FR901464 (1) and its methylated derivative, spliceostatin A (2), inhibit in vitro splicing and promote pre-mRNA accumulation by binding to SF3b, a subcomplex of the U2 small nuclear ribonucleoprotein in the spliceosome. Importantly, treatment of cells with these compounds resulted in leakage of pre-mRNA to the cytoplasm, where it was translated. Knockdown of SF3b by small interfering RNA induced phenotypes similar to those seen with spliceostatin A treatment. Thus, the inhibition of pre-mRNA splicing during early steps involving SF3b allows unspliced mRNA leakage and translation.


Asunto(s)
Transporte Activo de Núcleo Celular/efectos de los fármacos , Antineoplásicos/farmacología , Fosfoproteínas/antagonistas & inhibidores , Precursores del ARN/efectos de los fármacos , Empalme del ARN/efectos de los fármacos , Ribonucleoproteína Nuclear Pequeña U2/antagonistas & inhibidores , Línea Celular Tumoral , Humanos , Fosfoproteínas/genética , Piranos/farmacología , Precursores del ARN/metabolismo , Factores de Empalme de ARN , ARN Interferente Pequeño/farmacología , Proteínas de Unión al ARN , Ribonucleoproteína Nuclear Pequeña U2/genética , Compuestos de Espiro/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...