Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Lancet Oncol ; 25(7): 922-932, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38936379

RESUMEN

BACKGROUND: Neuroblastoma is the most common extracranial solid tumour in children. Relapsed or refractory neuroblastoma is associated with a poor outcome. We assessed the combination of irinotecan-temozolomide and dasatinib-rapamycin (RIST) in patients with relapsed or refractory neuroblastoma. METHODS: The multicentre, open-label, randomised, controlled, phase 2, RIST-rNB-2011 trial recruited from 40 paediatric oncology centres in Germany and Austria. Patients aged 1-25 years with high-risk relapsed (defined as recurrence of all stage IV and MYCN amplification stages, after response to treatment) or refractory (progressive disease during primary treatment) neuroblastoma, with Lansky and Karnofsky performance status at least 50%, were assigned (1:1) to RIST (RIST group) or irinotecan-temozolomide (control group) by block randomisation, stratified by MYCN status. We compared RIST (oral rapamycin [loading 3 mg/m2 on day 1, maintenance 1 mg/m2 on days 2-4] and oral dasatinib [2 mg/kg per day] for 4 days with 3 days off, followed by intravenous irinotecan [50 mg/m2 per day] and oral temozolomide [150 mg/m2 per day] for 5 days with 2 days off; one course each of rapamycin-dasatinib and irinotecan-temozolomide for four cycles over 8 weeks, then two courses of rapamycin-dasatinib followed by one course of irinotecan-temozolomide for 12 weeks) with irinotecan-temozolomide alone (with identical dosing as experimental group). The primary endpoint of progression-free survival was analysed in all eligible patients who received at least one course of therapy. The safety population consisted of all patients who received at least one course of therapy and had at least one post-baseline safety assessment. This trial is registered at ClinicalTrials.gov, NCT01467986, and is closed to accrual. FINDINGS: Between Aug 26, 2013, and Sept 21, 2020, 129 patients were randomly assigned to the RIST group (n=63) or control group (n=66). Median age was 5·4 years (IQR 3·7-8·1). 124 patients (78 [63%] male and 46 [37%] female) were included in the efficacy analysis. At a median follow-up of 72 months (IQR 31-88), the median progression-free survival was 11 months (95% CI 7-17) in the RIST group and 5 months (2-8) in the control group (hazard ratio 0·62, one-sided 90% CI 0·81; p=0·019). Median progression-free survival in patients with amplified MYCN (n=48) was 6 months (95% CI 4-24) in the RIST group versus 2 months (2-5) in the control group (HR 0·45 [95% CI 0·24-0·84], p=0·012); median progression-free survival in patients without amplified MYCN (n=76) was 14 months (95% CI 9-7) in the RIST group versus 8 months (4-15) in the control group (HR 0·84 [95% CI 0·51-1·38], p=0·49). The most common grade 3 or worse adverse events were neutropenia (54 [81%] of 67 patients given RIST vs 49 [82%] of 60 patients given control), thrombocytopenia (45 [67%] vs 41 [68%]), and anaemia (39 [58%] vs 38 [63%]). Nine serious treatment-related adverse events were reported (five patients given control and four patients given RIST). There were no treatment-related deaths in the control group and one in the RIST group (multiorgan failure). INTERPRETATION: RIST-rNB-2011 demonstrated that targeting of MYCN-amplified relapsed or refractory neuroblastoma with a pathway-directed metronomic combination of a multkinase inhibitor and an mTOR inhibitor can improve progression-free survival and overall survival. This exclusive efficacy in MYCN-amplified, relapsed neuroblastoma warrants further investigation in the first-line setting. FUNDING: Deutsche Krebshilfe.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Dasatinib , Irinotecán , Recurrencia Local de Neoplasia , Neuroblastoma , Sirolimus , Temozolomida , Humanos , Temozolomida/administración & dosificación , Temozolomida/uso terapéutico , Irinotecán/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Masculino , Femenino , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/mortalidad , Neuroblastoma/patología , Neuroblastoma/genética , Preescolar , Niño , Dasatinib/administración & dosificación , Dasatinib/uso terapéutico , Dasatinib/efectos adversos , Adolescente , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/patología , Lactante , Adulto , Sirolimus/administración & dosificación , Sirolimus/uso terapéutico , Adulto Joven , Alemania , Resistencia a Antineoplásicos , Supervivencia sin Progresión
2.
Biol Chem ; 404(11-12): 1123-1136, 2023 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-37632732

RESUMEN

Small non-coding RNAs (sncRNA) are involved in many steps of the gene expression cascade and regulate processing and expression of mRNAs by the formation of ribonucleoprotein complexes (RNP) such as the RNA-induced silencing complex (RISC). By analyzing small RNA Seq data sets, we identified a sncRNA annotated as piR-hsa-1254, which is likely derived from the 3'-end of 7SL RNA2 (RN7SL2), herein referred to as snc7SL RNA. The 7SL RNA is an abundant long non-coding RNA polymerase III transcript and serves as structural component of the cytoplasmic signal recognition particle (SRP). To evaluate a potential functional role of snc7SL RNA, we aimed to define its cellular localization by live cell imaging. Therefore, a Molecular Beacon (MB)-based method was established to compare the subcellular localization of snc7SL RNA with its precursor 7SL RNA. We designed and characterized several MBs in vitro and tested those by live cell fluorescence microscopy. Using a multiplex approach, we show that 7SL RNA localizes mainly to the endoplasmic reticulum (ER), as expected for the SRP, whereas snc7SL RNA predominately localizes to the nucleus. This finding suggests a fundamentally different function of 7SL RNA and its derivate snc7SL RNA.


Asunto(s)
ARN Citoplasmático Pequeño , Partícula de Reconocimiento de Señal , Partícula de Reconocimiento de Señal/genética , ARN , ARN Citoplasmático Pequeño/genética , ARN Citoplasmático Pequeño/metabolismo , ARN Mensajero
3.
Cancers (Basel) ; 15(6)2023 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-36980635

RESUMEN

BACKGROUND: The outcome for patients with high-risk neuroblastoma remains poor and novel treatment strategies are urgently needed. The RIST protocol represents a novel metronomic and multimodal treatment strategy for high-risk neuroblastoma combining molecular-targeted drugs as 'pre-treatment' with a conventional chemotherapy backbone, currently evaluated in a phase II clinical trial. For preclinical drug testing, cancer cell growth as spheroid compared to mo-nolayer cultures is of advantage since it reproduces a wide range of tumor characteristics, including the three-dimensional architecture and cancer stem cell (CSC) properties. The objective of this study was to establish a neuroblastoma spheroid model for the rigorous assessment of the RIST treatment protocol. METHODS: Evaluation of CSC marker expression was performed by mRNA and protein analysis and spheroid viability by luminescence-based assays. Aberrant expression of RNA-binding protein La in neuroblastoma was assessed by tissue microarray analysis and patients' data mining. RESULTS: Spheroid cultures showed increased expression of a subgroup of CSC-like markers (CXCR4, NANOG and BMI) and higher Thr389 phosphorylation of the neuroblastoma-associated RNA-binding protein La when compared to monolayer cultures. Molecular-targeted 'pre-treatment' of spheroids decreased neoplastic signaling and CSC marker expression. CONCLUSIONS: The RIST treatment protocol efficiently reduced the viability of neuroblastoma spheroids characterized by advanced CSC properties.

4.
Hematol Oncol Stem Cell Ther ; 16(3): 217-229, 2023 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-34848216

RESUMEN

Some of the early complications of hematopoietic stem cell transplantation (HSCT) concerning the small vessels can be summarized as transplant-associated systemic vasculopathies (TASV). One enzyme known to play a major role in inflammation, tissue remodeling, and repair processes as well as tumor metastasis is heparanase (HPSE). HPSE genetic variants have recently been associated with significant influence on the risk of developing certain TASV such as a sinusoidal obstruction syndrome. This study aimed to validate the two known HPSE single nucleotide polymorphisms (SNPs)-rs4693608 and rs4364254-as a genetic predictor of TASV in a cohort of 494 patients and were correlated retrospectively with the clinical course post-HSCT. Significant association was revealed for rs4364254, showing that the incidence of TASV (38.0% vs. 57.8%, p = .009) and in particular of acute graft-versus-host disease (aGvHD) (36.3% vs. 54.0%, p = .0138) was lower in wildtype CC carriers than in TC/TT carriers. Moreover, compared with all other genotypes, the allelic combination GG-CC had the lowest incidence of TASV (34.9% vs. 57.4%, p = .0109) and aGvHD in particular (34.9% vs. 53.5%, p = .0315). A competing risk regression analysis confirmed a significantly reduced risk for a TASV in patients with GG (subhazard ratio [SHR] = 0.670, p = .043) and CC (SHR = 0.598, p = .041) compared with the corresponding homozygote SNP as well as for allelic combinations correlated with low HPSE gene expression (SHR = 0.630, p = .016) and in correlation with clinical risk factors. In summary, our study emphasizes an association of HPSE gene SNPs with TASV, in particular with aGvHD, which could be implementable as pre-transplant risk stratification if validated prospectively.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Humanos , Estudios Retrospectivos , Incidencia , Genotipo , Polimorfismo de Nucleótido Simple , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Enfermedad Injerto contra Huésped/genética
5.
Int J Med Sci ; 18(1): 137-149, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33390782

RESUMEN

The prognosis for patients with relapsed or refractory high-risk neuroblastoma remains dismal and novel therapeutic options are urgently needed. The RIST treatment protocol has a multimodal metronomic therapy design combining molecular-targeted drugs (Rapamycin and Dasatinib) with chemotherapy backbone (Irinotecan and Temozolomide), which is currently verified in a phase II clinical trial (NCT01467986). With the availability of novel and more potent ATP competitive mTOR inhibitors, we expect to improve the RIST combination therapy. By comparing the IC50 values of Torin-1, Torin-2, AZD3147 and PP242 we established that only Torin-2 inhibited cell viability of all three MycN-amplified neuroblastoma cell lines tested at nanomolar concentration. Single treatment of both mTOR inhibitors induced a significant G1 cell cycle arrest and combination treatment with Dasatinib reduced the expression of cell cycle regulator cyclin D1 or increased the expression of cell cycle inhibitor p21. The combinatorial index depicted for both mTOR inhibitors a synergistic effect with Dasatinib. Interestingly, compared to Rapamycin, the combination treatment with Torin-2 resulted in a broader mTOR pathway inhibition as indicated by reduced phosphorylation of AKT (Thr308, Ser473), 4E-BP (Ser65), and S6K (Thr389). Furthermore, substituting Rapamycin in the modified multimodal RIST protocol with Torin-2 reduced cell viability and induced apoptosis despite a significant lower Torin-2 drug concentration applied. The efficacy of nanomolar concentrations may significantly reduce unwanted immunosuppression associated with Rapamycin. However, at this point we cannot rule out that Torin-2 has increased toxicity due to its potency in more complex systems. Nonetheless, our results suggest that including Torin-2 as a substitute for Rapamycin in the RIST protocol may represent a valid option to be evaluated in prospective clinical trials for relapsed or treatment-refractory high-risk neuroblastoma.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Naftiridinas/administración & dosificación , Neuroblastoma/tratamiento farmacológico , Sirolimus/administración & dosificación , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Administración Metronómica , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Dasatinib/administración & dosificación , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Humanos , Indoles/administración & dosificación , Concentración 50 Inhibidora , Irinotecán/administración & dosificación , Neuroblastoma/patología , Purinas/administración & dosificación , Pirimidinas/administración & dosificación , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Temozolomida/administración & dosificación , Tiourea/administración & dosificación , Tiourea/análogos & derivados
6.
Cancers (Basel) ; 13(2)2021 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-33477794

RESUMEN

BACKGROUND: the aberrant overexpression of predominantly nuclear localizing RNA-binding protein (RBP) La contributes to proliferation, mobility, and chemoresistance of cancer cells and tumor growth in mice. METHODS: studies included cancer tissue microarrays (TMAs) analyses, cancer tissue data mining, transforming growth factor ß (TGFß)-induced cancer cell plasticity studies, three dimensional sphere growth, epithelial to mesenchymal transition (EMT) assays, analysis of cancer stem cell (CSC) marker expression, and post-translational modification of cancer-associated La protein. RESULTS: we demonstrated that significant overexpression of RBP La in lung and head and neck cancer tissue correlates with poor overall survival. Furthermore, small interfering RNA-mediated depletion of La reduced proliferation and migration of cancer cells, blocked TGFß-induced EMT, and diminished both EMT and CSC marker expression. Rescue experiments with La wildtype but not RNA chaperone domain activity-defective La mutant increased the expression of those cancer progression markers, suggesting a critical role of La's RNA chaperone activity in this process. La depletion in cancer cells also significantly decreased sphere growth in the presence of TGFß. Interestingly, TGFß treatment induced phosphorylation of La at threonine 389 (pLaT389) only in adherents but not in 3D growing cultures. CONCLUSION: our study suggests that the TGFß/AKT/pLaT389 signaling pathway regulates cancer cell plasticity.

7.
RNA Biol ; 18(2): 218-236, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32687431

RESUMEN

RNA-binding proteins are important regulators of RNA metabolism and are of critical importance in all steps of the gene expression cascade. The role of aberrantly expressed RBPs in human disease is an exciting research field and the potential application of RBPs as a therapeutic target or a diagnostic marker represents a fast-growing area of research.Aberrant overexpression of the human RNA-binding protein La has been found in various cancer entities including lung, cervical, head and neck, and chronic myelogenous leukaemia. Cancer-associated La protein supports tumour-promoting processes such as proliferation, mobility, invasiveness and tumour growth. Moreover, the La protein maintains the survival of cancer cells by supporting an anti-apoptotic state that may cause resistance to chemotherapeutic therapy.The human La protein represents a multifunctional post-translationally modified RNA-binding protein with RNA chaperone activity that promotes processing of non-coding precursor RNAs but also stimulates the translation of selective messenger RNAs encoding tumour-promoting and anti-apoptotic factors. In our model, La facilitates the expression of those factors and helps cancer cells to cope with cellular stress. In contrast to oncogenes, able to initiate tumorigenesis, we postulate that the aberrantly elevated expression of the human La protein contributes to the non-oncogenic addiction of cancer cells. In this review, we summarize the current understanding about the implications of the RNA-binding protein La in cancer progression and therapeutic resistance. The concept of exploiting the RBP La as a cancer drug target will be discussed.


Asunto(s)
Susceptibilidad a Enfermedades , Neoplasias/etiología , Neoplasias/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Animales , Apoptosis/genética , Biomarcadores de Tumor , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Fosfoproteínas/antagonistas & inhibidores , Unión Proteica , Procesamiento Proteico-Postraduccional , ARN/genética , ARN/metabolismo , Procesamiento Postranscripcional del ARN , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Proteínas de Unión al ARN/antagonistas & inhibidores , Ribonucleoproteínas/antagonistas & inhibidores , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo
8.
Methods Mol Biol ; 2106: 121-136, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31889254

RESUMEN

It is well established that the RNA-binding protein La has RNA chaperone activity. Recent work suggests that the La protein has two distinct RNA chaperone domains (RCD-A and RCD-B) assisting structural changes in diverse groups of RNA molecules such as RNA polymerase III transcripts (e.g., pre-tRNA, U6 snRNA), cellular messenger, and viral RNAs. In this protocol we focus on the RNA chaperone domain RCD-B, which is located in the carboxy-terminal domain of La. It has been shown that this RNA chaperone domain assists structural changes in predicted RNA hairpins folded in the 5'-untranslated regions of cyclin D1 and Bcl2 mRNAs. Besides RNA helicases, which are implicated in melting RNA hairpin structures in an ATP-dependent manner, RNA chaperones fulfil a similar function in an ATP-independent manner. Aiming to study the RNA chaperon activity of La, we established a La-dependent molecular beacon-based RNA chaperone assay and systematically tested the various salt conditions. Herein we describe the assay format and design to study the salt dependency of RNA chaperones. This protocol can be easily adapted to test the RNA chaperone activity of other RNA-binding proteins and to optimize assay conditions.


Asunto(s)
Ensayo de Cambio de Movilidad Electroforética/métodos , Chaperonas Moleculares/metabolismo , Estabilidad del ARN , Proteínas de Unión al ARN/metabolismo , Salinidad , Animales , Ciclina D1/genética , Humanos , Proteínas Intrínsecamente Desordenadas/química , Secuencias Invertidas Repetidas , Chaperonas Moleculares/química , Dominios Proteicos , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN/química , ARN/metabolismo , Sondas ARN/química , ARN Mensajero/química , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/química
9.
Mol Cell Biol ; 38(2)2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29084811

RESUMEN

The cancer-associated RNA-binding protein La is posttranslationally modified by phosphorylation and sumoylation. Sumoylation of La regulates not only the trafficking of La in neuronal axons but also its association with specific mRNAs. Depletion of La in various types of cancer cell lines impairs cell proliferation; however, the molecular mechanism whereby La supports cell proliferation is not clearly understood. In this study, we address the question of whether sumoylation of La contributes to cell proliferation of HEK293 cells. We show that HEK293 cells stably expressing green fluorescent protein (GFP)-tagged wild-type La (GFP-LaWT) grow faster than cells expressing a sumoylation-deficient mutant La (GFP-LaSD), suggesting a proproliferative function of La in HEK293 cells. Further, we found that STAT3 protein levels were reduced in GFP-LaSD cells due to an increase in STAT3 ubiquitination and that overexpression of STAT3 partially restored cell proliferation. Finally, we present RNA sequencing data from RNA immunoprecipitations (RIPs) and report that mRNAs associated with the cell cycle and ubiquitination are preferentially bound by GFP-LaWT and are less enriched in GFP-LaSD RIPs. Taken together, results of our study support a novel mechanism whereby sumoylation of La promotes cell proliferation by averting ubiquitination-mediated degradation of the STAT3 protein.


Asunto(s)
Fosfoproteínas/metabolismo , Factor de Transcripción STAT3/metabolismo , Proliferación Celular , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunoprecipitación , Fosfoproteínas/genética , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factor de Transcripción STAT3/genética , Sumoilación , Ubiquitinación
10.
PLoS One ; 12(3): e0173246, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28291789

RESUMEN

The RNA-binding protein La is overexpressed in a number of tumor tissues and is thought to support tumorigenesis by binding to and facilitating the expression of mRNAs encoding tumor-promoting and anti-apoptotic factors. Hence, small molecules able to block the binding of La to specific RNAs could have a therapeutic impact by reducing the expression of tumor-promoting and anti-apoptotic factors. Toward this novel therapeutic strategy, we aimed to develop a high-throughput fluorescence polarization assay to screen small compound libraries for molecules blocking the binding of La to an RNA element derived from cyclin D1 mRNA. Herein, we make use of a robust fluorescence polarization assay and the validation of primary hits by electrophoretic mobility shift assays. We showed recently that La protects cells against cisplatin treatment by stimulating the protein synthesis of the anti-apoptotic factor Bcl2. Here, we show by RNA immunoprecipitation experiments that one small compound specifically impairs the association of La with Bcl2 mRNA in cells and sensitizes cells for cipslatin-induced cell death. In summary, we report the application of a high-throughput fluorescence polarization assay to identify small compounds that impair the binding of La to target RNAs in vitro and in cells.


Asunto(s)
Polarización de Fluorescencia/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Sondas Moleculares/química , Proteínas de Unión al ARN/antagonistas & inhibidores , ARN/antagonistas & inhibidores , Línea Celular , Ensayo de Cambio de Movilidad Electroforética , Humanos , Inmunoprecipitación , Técnicas In Vitro , Sondas Moleculares/farmacología , Unión Proteica , ARN/metabolismo , Proteínas de Unión al ARN/metabolismo
11.
PLoS One ; 11(5): e0156365, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27224031

RESUMEN

The RNA-binding protein La is involved in several aspects of RNA metabolism including the translational regulation of mRNAs and processing of pre-tRNAs. Besides its well-described phosphorylation by Casein kinase 2, the La protein is also posttranslationally modified by the Small Ubiquitin-like MOdifier (SUMO), but the functional outcome of this modification has not been defined. The objective of this study was to test whether sumoylation changes the RNA-binding activity of La. Therefore, we established an in vitro sumoylation assay for recombinant human La and analyzed its RNA-binding activity by electrophoretic mobility shift assays. We identified two novel SUMO-acceptor sites within the La protein located between the RNA recognition motif 1 and 2 and we demonstrate for the first time that sumoylation facilitates the RNA-binding of La to small RNA oligonucleotides representing the oligopyrimidine tract (TOP) elements from the 5' untranslated regions (UTR) of mRNAs encoding ribosomal protein L22 and L37 and to a longer RNA element from the 5' UTR of cyclin D1 (CCND1) mRNA in vitro. Furthermore, we show by RNA immunoprecipitation experiments that a La mutant deficient in sumoylation has impaired RNA-binding activity in cells. These data suggest that modulating the RNA-binding activity of La by sumoylation has important consequences on its functionality.


Asunto(s)
Ciclina D1/genética , Fosfoproteínas/metabolismo , ARN Mensajero/metabolismo , Proteínas Ribosómicas/genética , Regiones no Traducidas 3' , Sitios de Unión , Ensayo de Cambio de Movilidad Electroforética , Humanos , Fosfoproteínas/química , Unión Proteica , Procesamiento Proteico-Postraduccional , Sumoilación
12.
Oncotarget ; 7(20): 29664-76, 2016 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-27105491

RESUMEN

Up-regulation of anti-apoptotic factors is a critical mechanism of cancer cell resistance and often counteracts the success of chemotherapeutic treatment. Herein, we identified the cancer-associated RNA-binding protein La as novel factor contributing to cisplatin resistance. Our data demonstrate that depletion of the RNA-binding protein La in head and neck squamous cell carcinoma cells (HNSCC) increases the sensitivity toward cisplatin-induced cell death paralleled by reduced expression of the anti-apoptotic factor Bcl2. Furthermore, it is shown that transient expression of Bcl2 in La-depleted cells protects against cisplatin-induced cell death. By dissecting the underlying mechanism we report herein, that the La protein is required for Bcl2 protein synthesis in cisplatin-treated cells. The RNA chaperone La binds in close proximity to the authentic translation start site and unwinds a secondary structure embedding the authentic AUG. Altogether, our data support a novel model, whereby cancer-associated La protein contributes to cisplatin resistance by stimulating the translation of anti-apoptotic factor Bcl2 in HNSCC cells.


Asunto(s)
Autoantígenos/metabolismo , Carcinoma de Células Escamosas/patología , Resistencia a Antineoplásicos/fisiología , Neoplasias de Cabeza y Cuello/patología , Fragmentos de Péptidos/metabolismo , Biosíntesis de Proteínas/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Antineoplásicos/farmacología , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Cisplatino/farmacología , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello , Regulación hacia Arriba
13.
Nucleic Acids Res ; 43(1): 581-94, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25520193

RESUMEN

The cellular function of the cancer-associated RNA-binding protein La has been linked to translation of viral and cellular mRNAs. Recently, we have shown that the human La protein stimulates IRES-mediated translation of the cooperative oncogene CCND1 in cervical cancer cells. However, there is little known about the underlying molecular mechanism by which La stimulates CCND1 IRES-mediated translation, and we propose that its RNA chaperone activity is required. Herein, we show that La binds close to the CCND1 start codon and demonstrate that La's RNA chaperone activity can change the folding of its binding site. We map the RNA chaperone domain (RCD) within the C-terminal region of La in close proximity to a novel AKT phosphorylation site (T389). Phosphorylation at T389 by AKT-1 strongly impairs its RNA chaperone activity. Furthermore, we demonstrate that the RCD as well as T389 is required to stimulate CCND1 IRES-mediated translation in cells. In summary, we provide a model whereby a novel interplay between RNA-binding, RNA chaperoning and AKT phosphorylation of La protein regulates CCND1 IRES-mediated translation.


Asunto(s)
Autoantígenos/metabolismo , Ciclina D1/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN/metabolismo , Ribonucleoproteínas/metabolismo , Regiones no Traducidas 5' , Secuencias de Aminoácidos , Autoantígenos/química , Sitios de Unión , Codón Iniciador , Ciclina D1/biosíntesis , Células HEK293 , Humanos , Fosforilación , Biosíntesis de Proteínas , Estructura Terciaria de Proteína , Ribonucleoproteínas/química , Treonina/metabolismo , Antígeno SS-B
14.
PLoS One ; 7(9): e45197, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23028842

RESUMEN

BACKGROUND: Suppressor of cytokine signaling 3 (SOCS3) is an inducible endogenous negative regulator of signal transduction and activator of transcription 3 (STAT3). Epigenetic silencing of SOCS3 has been shown in head and neck squamous cell carcinoma (HNSCC), which is associated with increased activation of STAT3. There is scarce information on the functional role of the reduction of SOCS3 expression and no information on altered subcellular localization of SOCS3 in HNSCC. METHODOLOGY/PRINCIPAL FINDINGS: We assessed endogenous SOCS3 expression in different HNSCC cell lines by RT-qPCR and western blot. Immunofluorescence and western blot were used to study the subcellular localization of endogenous SOCS3 induced by IL-6. Overexpression of SOCS3 by CMV-driven plasmids and siRNA-mediated inhibition of endogenous SOCS3 were used to verify the role of SOCS3 on tumor cell proliferation, viability, invasion and migration in vitro. In vivo relevance of SOCS3 expression in HNSCC was studied by quantitative immunohistochemistry of commercially-available tissue microarrays. Endogenous expression of SOCS3 was heterogeneous in four HNSCC cell lines and surprisingly preserved in most of these cell lines. Subcellular localization of endogenous SOCS3 in the HNSCC cell lines was predominantly nuclear as opposed to cytoplasmic in non-neoplasic epithelial cells. Overexpression of SOCS3 produced a relative increase of the protein in the cytoplasmic compartment and significantly inhibited proliferation, migration and invasion, whereas inhibition of endogenous nuclear SOCS3 did not affect these events. Analysis of tissue microarrays indicated that loss of SOCS3 is an early event in HNSCC and was correlated with tumor size and histological grade of dysplasia, but a considerable proportion of cases presented detectable expression of SOCS3. CONCLUSION: Our data support a role for SOCS3 as a tumor suppressor gene in HNSCC with relevance on proliferation and invasion processes and suggests that abnormal subcellular localization impairs SOCS3 function in HNSCC cells.


Asunto(s)
Carcinoma de Células Escamosas/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/genética , Invasividad Neoplásica/genética , Factor de Transcripción STAT3/genética , Proteínas Supresoras de la Señalización de Citocinas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Proliferación Celular , Citoplasma/genética , Citoplasma/metabolismo , Citoplasma/ultraestructura , Silenciador del Gen , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , ARN Interferente Pequeño , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
15.
Proc Natl Acad Sci U S A ; 109(34): E2276-83, 2012 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-22652566

RESUMEN

Most gastrointestinal stromal tumors (GISTs) harbor a gain-of-function mutation in the Kit receptor. GIST patients treated with the tyrosine kinase inhibitor imatinib frequently develop imatinib resistance as a result of second-site Kit mutations. To investigate the consequences of second-site Kit mutations on GIST development and imatinib sensitivity, we engineered a mouse model carrying in the endogenous Kit locus both the Kit(V558Δ) mutation found in a familial case of GIST and the Kit(T669I) (human KIT(T670I)) "gatekeeper" mutation found in imatinib-resistant GIST patients. Similar to Kit(V558/+) mice, Kit(V558;T669I/+) mice developed gastric and colonic interstitial cell of Cajal hyperplasia as well as cecal GIST. In contrast to the single-mutant Kit(V558/+) control mice, treatment of the Kit(V558;T669I/+) mice with either imatinib or dasatinib failed to inhibit oncogenic Kit signaling and GIST growth. However, this resistance could be overcome by treatment of Kit(V558;T669I/+) mice with sunitinib or sorafenib. Although tumor lesions were smaller in Kit(V558;T669I/+) mice than in single-mutant mice, both interstitial cell of Cajal hyperplasia and mast cell hyperplasia were exacerbated in Kit(V558;T669I/+) mice. Strikingly, the Kit(V558;T669I/+) mice developed a pronounced polycythemia vera-like erythrocytosis in conjunction with microcytosis. This mouse model should be useful for preclinical studies of drug candidates designed to overcome imatinib resistance in GIST and to investigate the consequences of oncogenic KIT signaling in hematopoietic as well as other cell lineages.


Asunto(s)
Eritrocitos/citología , Tumores del Estroma Gastrointestinal/genética , Mutación , Piperazinas/farmacología , Policitemia/genética , Proteínas Proto-Oncogénicas c-kit/genética , Pirimidinas/farmacología , Animales , Antineoplásicos/farmacología , Benzamidas , Linaje de la Célula , Dasatinib , Modelos Animales de Enfermedad , Resistencia a Medicamentos , Resistencia a Antineoplásicos/genética , Exones , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Mesilato de Imatinib , Ratones , Fenotipo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Tiazoles/farmacología
16.
PLoS One ; 6(10): e25402, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22016766

RESUMEN

The 5-year survival rate for oral cavity cancer is poorer than for breast, colon or prostate cancer, and has improved only slightly in the last three decades. Hence, new therapeutic strategies are urgently needed. Here we demonstrate by tissue micro array analysis for the first time that RNA-binding protein La is significantly overexpressed in oral squamous cell carcinoma (SCC). Within this study we therefore addressed the question whether siRNA-mediated depletion of the La protein may interfere with known tumor-promoting characteristics of head and neck SCC cells. Our studies demonstrate that the La protein promotes cell proliferation, migration and invasion of lymph node-metastasized hypopharyngeal SCC cells. We also reveal that La is required for the expression of ß-catenin as well as matrix metalloproteinase type 2 (MMP-2) within these cells. Taken together these data suggest a so far unknown function of the RNA-binding protein La in promoting tumor progression of head and neck SCC.


Asunto(s)
Autoantígenos/metabolismo , Carcinoma de Células Escamosas/patología , Movimiento Celular , Neoplasias Hipofaríngeas/patología , Ribonucleoproteínas/metabolismo , Autoantígenos/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Ciclo Celular , Proliferación Celular , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Células HeLa , Humanos , Neoplasias Hipofaríngeas/genética , Neoplasias Hipofaríngeas/metabolismo , Metástasis Linfática , Metaloproteinasa 2 de la Matriz/metabolismo , Invasividad Neoplásica , Ribonucleoproteínas/genética , beta Catenina/metabolismo , Antígeno SS-B
17.
Front Biosci ; 13: 5533-47, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18508603

RESUMEN

Acute and chronic hepatitis B virus (HBV) infection is the cause of about 1 million deaths each year worldwide. Although a vaccine for HBV is available, new HBV infections are appearing at alarming rates and maternal-fetal transmission is a major cause of viral spread. Thus, new and effective antiviral strategies are necessary for the 300 million chronically HBV-infected individuals to reduce their morbidity and mortality. Precise processing of viral RNAs is essential for the hepatitis viral life cycle, and, to our knowledge, the processing, nuclear export, and stabilization/degradation of viral RNAs are exclusively mediated by host factors. Thus, identification of host factors required for viral RNA metabolism and subsequent molecular analysis of the interactions between viral RNAs and corresponding host factors might represent novel avenues for the development of new antiviral strategies. In this review, we summarize the current knowledge about the posttranscriptional control of HBV RNA metabolism.


Asunto(s)
Regulación Viral de la Expresión Génica , Virus de la Hepatitis B/genética , Procesamiento Postranscripcional del ARN , ARN Viral/genética , Núcleo Celular/virología
18.
Proc Natl Acad Sci U S A ; 103(34): 12843-8, 2006 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-16908864

RESUMEN

Kit receptor-activating mutations are critical in the pathogenesis of gastrointestinal stromal tumors (GIST). We investigated mechanisms of oncogenic Kit signaling and the consequences of therapeutic intervention in a mouse model of human GIST. Treatment of GIST mice with imatinib decreased cell proliferation and increased apoptosis in the tumor. Analysis of tumor tissue from imatinib-treated mice showed diminished phosphatidylinositol 3-kinase (PI3-kinase) and mammalian target of rapamycin (mTOR) signaling suggesting that oncogenic Kit signaling critically contributes to the translational response in GIST. Treatment with RAD001 (everolimus), an mTOR inhibitor, diminished the translational response and cell proliferation in tumor lesions, pointing to mTOR inhibition as a therapeutic approach for imatinib-resistant GIST. Analysis of RNA expression profiles in GIST lesions with and without imatinib treatment showed changes in expression of IFN-inducible genes and cell cycle regulators. These results convincingly show that KitV558Delta/+ mice represent a unique faithful mouse model of human familial GIST, and they demonstrate the utility of these mice for preclinical investigations and to elucidate oncogenic signaling mechanisms by using genetic approaches and targeted pharmacological intervention.


Asunto(s)
Modelos Animales de Enfermedad , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Transducción de Señal , Animales , Benzamidas , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo , Everolimus , Tumores del Estroma Gastrointestinal/genética , Tumores del Estroma Gastrointestinal/patología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Mesilato de Imatinib , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación , Piperazinas/uso terapéutico , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-kit/genética , Pirimidinas/uso terapéutico , Sirolimus/análogos & derivados , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR
19.
Nucleic Acids Res ; 34(1): 353-63, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16410615

RESUMEN

The posttranscriptional regulatory element (PRE) is considered to enhance hepatitis B virus (HBV) gene expression by facilitating the nuclear export of intronless viral subgenomic RNAs. Its role in the RNA metabolism of the viral pregenomic RNA (pgRNA) is currently unknown. We identified a positively cis-acting splicing regulatory element (SRE-1) and present two lines of evidence for its functionality. Firstly, in a heterologous context SRE-1 functionally substitutes for a retroviral bidirectional exonic splicing enhancer (ESE). As expected, SRE-1 is a splicing enhancer also in its natural viral sequence context, since deletion of SRE-1 reduces splicing of pgRNA in cell culture experiments. Secondly, we show that stimulation of HBV RNA splicing by the splicing factor PSF was repressed by the PRE. Analysis of a variety of PSF mutants indicated that RNA-binding and protein-protein interaction were required to enhance splicing. In addition, we show that the PRE contributed to pgRNA stability, but has little influence on its nuclear export. Herein, we report for the first time that the PRE harbors splicing stimulating and inhibiting regulatory elements controlling processing of the viral pregenome. We discuss a model in which the regulation of pgRNA splicing depends on cellular factors interacting with the PRE.


Asunto(s)
Regulación Viral de la Expresión Génica , Virus de la Hepatitis B/genética , Empalme del ARN , ARN Viral/química , ARN Viral/metabolismo , Secuencias Reguladoras de Ácido Ribonucleico , Línea Celular Tumoral , Virus de la Hepatitis B/metabolismo , Humanos , Factor de Empalme Asociado a PTB , Proteínas de Unión al ARN/metabolismo
20.
Blood ; 106(12): 3958-61, 2005 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16081693

RESUMEN

Multiple genetic alterations are required to induce acute myelogenous leukemia (AML). Mutations in the extracellular domain of the KIT receptor are almost exclusively found in patients with AML carrying translocations or inversions affecting members of the core binding factor (CBF) gene family and correlate with a high risk of relapse. We demonstrate that these complex insertion and deletion mutations lead to constitutive activation of the KIT receptor, which induces factor-independent growth of interleukin-3 (IL-3)-dependent cells. Mutation of the evolutionary conserved amino acid D419 within the extracellular domain was sufficient to constitutively activate the KIT receptor, although high expression levels were required. Dose-dependent growth inhibition and apoptosis were observed using either the protein tyrosine kinase inhibitor imatinib mesylate (STI571, Gleevec) or by blocking the phosphoinositide-3-kinase (PI3K)-AKT pathway. Our data show that the addition of kinase inhibitors to conventional chemotherapy might be a new therapeutic option for CBF-AML expressing mutant KIT.


Asunto(s)
Antineoplásicos/farmacología , Factores de Unión al Sitio Principal/genética , Leucemia Mieloide Aguda/genética , Piperazinas/farmacología , Pirimidinas/farmacología , Factor de Células Madre/genética , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Benzamidas , Humanos , Mesilato de Imatinib , Leucemia Mieloide Aguda/tratamiento farmacológico , Ratones , Mutagénesis Sitio-Dirigida , Mutación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...