Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Genes Dev ; 34(15-16): 1051-1064, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32675324

RESUMEN

YAP1 is a transcriptional coactivator and the principal effector of the Hippo signaling pathway, which is causally implicated in human cancer. Several YAP1 gene fusions have been identified in various human cancers and identifying the essential components of this family of gene fusions has significant therapeutic value. Here, we show that the YAP1 gene fusions YAP1-MAMLD1, YAP1-FAM118B, YAP1-TFE3, and YAP1-SS18 are oncogenic in mice. Using reporter assays, RNA-seq, ChIP-seq, and loss-of-function mutations, we can show that all of these YAP1 fusion proteins exert TEAD-dependent YAP activity, while some also exert activity of the C'-terminal fusion partner. The YAP activity of the different YAP1 fusions is resistant to negative Hippo pathway regulation due to constitutive nuclear localization and resistance to degradation of the YAP1 fusion proteins. Genetic disruption of the TEAD-binding domain of these oncogenic YAP1 fusions is sufficient to inhibit tumor formation in vivo, while pharmacological inhibition of the YAP1-TEAD interaction inhibits the growth of YAP1 fusion-expressing cell lines in vitro. These results highlight TEAD-dependent YAP activity found in these gene fusions as critical for oncogenesis and implicate these YAP functions as potential therapeutic targets in YAP1 fusion-positive tumors.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinogénesis/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Ratones , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Señales de Localización Nuclear , Motivos de Nucleótidos , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteínas de Fusión Oncogénica/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Transcripción Genética
2.
Genes Dev ; 32(7-8): 512-523, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29632085

RESUMEN

Glioblastoma is the most frequently occurring and invariably fatal primary brain tumor in adults. The vast majority of glioblastomas is characterized by chromosomal copy number alterations, including gain of whole chromosome 7 and loss of whole chromosome 10. Gain of whole chromosome 7 is an early event in gliomagenesis that occurs in proneural-like precursor cells, which give rise to all isocitrate dehydrogenase (IDH) wild-type glioblastoma transcriptional subtypes. Platelet-derived growth factor A (PDGFA) is one gene on chromosome 7 known to drive gliomagenesis, but, given its location near the end of 7p, there are likely several other genes located along chromosome 7 that select for its increased whole-chromosome copy number within glioblastoma cells. To identify other potential genes that could select for gain of whole chromosome 7, we developed an unbiased bioinformatics approach that identified homeobox A5 (HOXA5) as a gene whose expression correlated with gain of chromosome 7 and a more aggressive phenotype of the resulting glioma. High expression of HOXA5 in glioblastoma was associated with a proneural gene expression pattern and decreased overall survival in both human proneural and PDGF-driven mouse glioblastoma. Furthermore, HOXA5 overexpression promoted cellular proliferation and potentiated radioresistance. We also found enrichment of HOXA5 expression in recurrent human and mouse glioblastoma at first recurrence after radiotherapy. Overall, this study implicates HOXA5 as a chromosome 7-associated gene-level locus that promotes selection for gain of whole chromosome 7 and an aggressive phenotype in glioblastoma.


Asunto(s)
Neoplasias Encefálicas/genética , Cromosomas Humanos Par 7 , Glioblastoma/genética , Proteínas de Homeodominio/metabolismo , Fosfoproteínas/metabolismo , Animales , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/radioterapia , Proliferación Celular , Duplicación Cromosómica , Glioblastoma/mortalidad , Glioblastoma/patología , Glioblastoma/radioterapia , Proteínas de Homeodominio/genética , Humanos , Isocitrato Deshidrogenasa/genética , Ratones , Recurrencia Local de Neoplasia , Fosfoproteínas/genética , Tolerancia a Radiación , Factores de Transcripción
3.
Kyobu Geka ; 76(2): 156-159, 2023 Feb.
Artículo en Japonés | MEDLINE | ID: mdl-36731853

RESUMEN

A 47-year-old man presented with hematuria. Computed tomography( CT) showed a posterior mediastinal cyst. Chest magnetic resonance imaging showed a well defined mass with high intensity on T2-weighted images. Echocardiogram revealed severe aortic regurgitation, moderate mitral regurgitation and no continuity between the cyst and the pericardium. We performed aortic valve replacement, mitral annuloplasty and cyst resection after confirming it was not malignant by intraoperative rapid pathological examination. We performed sufficient and safe cyst resection through full sternotomy under cardiac arrest. Pathological examination revealed that cyst was bronchogenic. Bronchogenic cyst has malignant potential and it is very difficult to resect after presenting symptoms. We need to consider the differential diagnosis, the timing of operation and operative strategy.


Asunto(s)
Quiste Broncogénico , Procedimientos Quirúrgicos Cardíacos , Quiste Mediastínico , Masculino , Humanos , Persona de Mediana Edad , Quiste Mediastínico/diagnóstico por imagen , Quiste Mediastínico/cirugía , Esternotomía , Quiste Broncogénico/cirugía , Mediastino
4.
Kyobu Geka ; 76(3): 193-196, 2023 Mar.
Artículo en Japonés | MEDLINE | ID: mdl-36861274

RESUMEN

We report the preoperative evaluation of saphenous vein (SV) graft using plain computed tomography (CT) for endoscopic saphenous vein harvesting (EVH). We made three-dimensional (3D) images of SV by using plain CT images. EVH was performed in 33 patients from July 2019 to September 2020. The mean age of the patients was 69±23 years, and 25 patients were men. The success rate of EVH was 93.9%. Hospital mortality was 0%. Postoperative wound complications was 0%. The early patency was 98.2% (55/56). 3D images of SV by plain CT are very important information for EVH because of surgical procedure in a closed space. Early patency is good and mid and long term patency of EVH may be improved due to safety and gentle technique by CT information.


Asunto(s)
Endoscopía , Vena Safena , Masculino , Humanos , Persona de Mediana Edad , Anciano , Anciano de 80 o más Años , Femenino , Vena Safena/diagnóstico por imagen , Vena Safena/cirugía , Complicaciones Posoperatorias , Tomografía Computarizada por Rayos X
5.
Kyobu Geka ; 75(6): 472-475, 2022 Jun.
Artículo en Japonés | MEDLINE | ID: mdl-35618695

RESUMEN

Giant cell myocarditis (GCM) is one of the rare diseases that cause fatal heart failure and suspected to be associated with autoimmune disorder. There are few reports that the patients of GCM live long because of their progressive heart failure. We report a rare case of patient who was suspected two long hospitalizations due to heart failure in her childhood and peripartum period, and incidentally diagnosed with GCM by myocardial biopsy performed at the aortic valve replacement and mitral annuloplasty for severe aortic valve regurgitation and moderate mitral regurgitation.


Asunto(s)
Insuficiencia Cardíaca , Anuloplastia de la Válvula Mitral , Insuficiencia de la Válvula Mitral , Miocarditis , Niño , Femenino , Células Gigantes/patología , Insuficiencia Cardíaca/complicaciones , Humanos , Insuficiencia de la Válvula Mitral/diagnóstico por imagen , Insuficiencia de la Válvula Mitral/etiología , Insuficiencia de la Válvula Mitral/cirugía , Miocarditis/complicaciones , Miocarditis/diagnóstico , Miocarditis/cirugía
6.
Kyobu Geka ; 74(9): 672-675, 2021 Sep.
Artículo en Japonés | MEDLINE | ID: mdl-34446620

RESUMEN

A 71-year-old man who hospitalized frequently for heart failure was referred to our hospital for severe coronary disease with mitral regurgitation. Transthoracic echocardiography revealed marked left ventricular dilatation, low ejection fraction (20%) and moderate mitral regurgitation with leaflet tethering. It also revealed myocardium with prominent trabeculations and deep intertrabecular recesses. Coronary angiography showed triple vessel disease. Coronary artery bypass grafting and mitral annuroplasty was performed. Coronary microcirculatory dysfunction by left ventricular noncompaction( LVNC) and myocardial ischemia made us pay more attention to myocardial protection. Aortic cross clamp time was 67 minnutes, total cardiopulmonary bypass time was 116 minnutes and operation time was 214 minnutes. The postoperative course was uneventful and the patient was discharged 15 days after the operation. Postoperative echocardiography revealed no mitral regurgitation and improving left ventricular function. Postoperative coronary computed tomography showed all grafts patent. Careful observation of cardiac function is vital because of the possibility of progression to heart failure in a patient with LVNC.


Asunto(s)
Enfermedad de la Arteria Coronaria , Anuloplastia de la Válvula Mitral , Insuficiencia de la Válvula Mitral , Anciano , Puente de Arteria Coronaria , Humanos , Masculino , Microcirculación , Insuficiencia de la Válvula Mitral/diagnóstico por imagen , Insuficiencia de la Válvula Mitral/cirugía , Resultado del Tratamiento
7.
Kyobu Geka ; 74(5): 362-365, 2021 May.
Artículo en Japonés | MEDLINE | ID: mdl-33980796

RESUMEN

Cardiac surgery for elder patients should be minimally invasive because of their frailty and concomitant multiple chronic diseases. We performed aortic valve replacement (AVR) via upper hemisternotomy (UHS) on an 85-year-old patient who suffered from severe aortic regurgitation with dobutamine support. The postoperative course was uneventful. AVR via UHS is safer than that via right axiallary thoracotomy AVR with shorter aortic cross-clamp time, shorter cardiopulmonary bypass time and fewer complications. AVR via UHS makes ambulation and rehabilitation easier than AVR via full sternotomy, because of thoracic stability. It is more effective and should be more prevalent as minimally invasive cardiac surgery for eldery patients with frailty.


Asunto(s)
Insuficiencia Cardíaca , Implantación de Prótesis de Válvulas Cardíacas , Prótesis Valvulares Cardíacas , Anciano , Anciano de 80 o más Años , Válvula Aórtica/cirugía , Humanos , Procedimientos Quirúrgicos Mínimamente Invasivos , Estudios Retrospectivos , Esternotomía , Toracotomía , Resultado del Tratamiento
8.
Genes Dev ; 24(19): 2205-18, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20889717

RESUMEN

Gene rearrangement in the form of an intragenic deletion is the primary mechanism of oncogenic mutation of the epidermal growth factor receptor (EGFR) gene in gliomas. However, the incidence of platelet-derived growth factor receptor-α (PDGFRA) gene rearrangement in these tumors is unknown. We investigated the PDGFRA locus in PDGFRA-amplified gliomas and identified two rearrangements, including the first case of a gene fusion between kinase insert domain receptor (KDR) (VEGFRII) and the PDGFRA gene, and six cases of PDGFRA(Δ8, 9), an intragenic deletion rearrangement. The PDGFRA(Δ8, 9) mutant was common, being present in 40% of the glioblastoma multiformes (GBMs) with PDGFRA amplification. Tumors with these two types of PDGFRA rearrangement displayed histologic features of oligodendroglioma, and the gene products of both rearrangements showed constitutively elevated tyrosine kinase activity and transforming potential that was reversed by PDGFR blockade. These results suggest the possibility that these PDGFRA mutants behave as oncogenes in this subset of gliomas, and that the prevalence of such rearrangements may have been considerably underestimated.


Asunto(s)
Reordenamiento Génico , Glioblastoma/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Benzamidas , Dosificación de Gen , Fusión Génica/genética , Glioblastoma/patología , Humanos , Mesilato de Imatinib , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , Mutación/genética , Oligodendroglioma/genética , Oligodendroglioma/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Ftalazinas/farmacología , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Piridinas/farmacología , Pirimidinas/farmacología , Transducción de Señal , Transformación Genética/efectos de los fármacos
9.
J Pharmacol Exp Ther ; 360(1): 215-224, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27821713

RESUMEN

Evidence suggests that the nonpsychotropic cannabis-derived compound, cannabidiol (CBD), has antineoplastic activity in multiple types of cancers, including glioblastoma multiforme (GBM). DNA-damaging agents remain the main standard of care treatment available for patients diagnosed with GBM. Here we studied the antiproliferative and cell-killing activity of CBD alone and in combination with DNA-damaging agents (temozolomide, carmustine, or cisplatin) in several human GBM cell lines and in mouse primary GBM cells in cultures. This activity was also studied in mouse neural progenitor cells (NPCs) in culture to assess for potential central nervous system toxicity. We found that CBD induced a dose-dependent reduction of both proliferation and viability of all cells with similar potencies, suggesting no preferential activity for cancer cells. Hill plot analysis indicates an allosteric mechanism of action triggered by CBD in all cells. Cotreatment regimens combining CBD and DNA-damaging agents produced synergistic antiproliferating and cell-killing responses over a limited range of concentrations in all human GBM cell lines and mouse GBM cells as well as in mouse NPCs. Remarkably, antagonistic responses occurred at low concentrations in select human GBM cell lines and in mouse GBM cells. Our study suggests limited synergistic activity when combining CBD and DNA-damaging agents in treating GBM cells, along with little to no therapeutic window when considering NPCs.


Asunto(s)
Antineoplásicos/efectos adversos , Antineoplásicos/farmacología , Cannabidiol/efectos adversos , Cannabidiol/farmacología , Daño del ADN , Glioblastoma/patología , Células-Madre Neurales/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Ratones , Células-Madre Neurales/citología , Transducción de Señal/efectos de los fármacos
10.
Brain ; 139(Pt 5): 1458-71, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27020328

RESUMEN

Glioblastoma is the most common and most aggressive primary brain tumour. Standard of care consists of surgical resection followed by radiotherapy and concomitant and maintenance temozolomide (temozolomide/radiotherapy→temozolomide). Corticosteroids are commonly used perioperatively to control cerebral oedema and are frequently continued throughout subsequent treatment, notably radiotherapy, for amelioration of side effects. The effects of corticosteroids such as dexamethasone on cell growth in glioma models and on patient survival have remained controversial. We performed a retrospective analysis of glioblastoma patient cohorts to determine the prognostic role of steroid administration. A disease-relevant mouse model of glioblastoma was used to characterize the effects of dexamethasone on tumour cell proliferation and death, and to identify gene signatures associated with these effects. A murine anti-VEGFA antibody was used in parallel as an alternative for oedema control. We applied the dexamethasone-induced gene signature to The Cancer Genome Atlas glioblastoma dataset to explore the association of dexamethasone exposure with outcome. Mouse experiments were used to validate the effects of dexamethasone on survival in vivo Retrospective clinical analyses identified corticosteroid use during radiotherapy as an independent indicator of shorter survival in three independent patient cohorts. A dexamethasone-associated gene expression signature correlated with shorter survival in The Cancer Genome Atlas patient dataset. In glioma-bearing mice, dexamethasone pretreatment decreased tumour cell proliferation without affecting tumour cell viability, but reduced survival when combined with radiotherapy. Conversely, anti-VEGFA antibody decreased proliferation and increased tumour cell death, but did not affect survival when combined with radiotherapy. Clinical and mouse experimental data suggest that corticosteroids may decrease the effectiveness of treatment and shorten survival in glioblastoma. Dexamethasone-induced anti-proliferative effects may confer protection from radiotherapy- and chemotherapy-induced genotoxic stress. This study highlights the importance of identifying alternative agents such as vascular endothelial growth factor antagonists for managing oedema in glioblastoma patients. Beyond the established adverse effect profile of protracted corticosteroid use, this analysis substantiates the request for prudent and restricted use of corticosteroids in glioblastoma.


Asunto(s)
Corticoesteroides/efectos adversos , Corticoesteroides/farmacología , Neoplasias Encefálicas/mortalidad , Glioblastoma/mortalidad , Animales , Anticuerpos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/radioterapia , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Terapia Combinada/efectos adversos , Dexametasona/efectos adversos , Dexametasona/farmacología , Femenino , Expresión Génica/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Glioblastoma/radioterapia , Humanos , Masculino , Ratones , Ratones Transgénicos , Radioterapia , Estudios Retrospectivos , Análisis de Supervivencia , Factor A de Crecimiento Endotelial Vascular/inmunología
11.
Nanomedicine ; 13(7): 2131-2139, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28614736

RESUMEN

Glioblastoma (GBM) remains incurable, and recurrent tumors rarely respond to standard-of-care radiation and chemo-therapies. Therefore, strategies that enhance the effects of these therapies should provide significant benefits to GBM patients. We have developed a nanoparticle delivery vehicle that can stably bind and protect nucleic acids for specific delivery into brain tumor cells. These nanoparticles can deliver therapeutic siRNAs to sensitize GBM cells to radiotherapy and improve GBM treatment via systemic administration. We show that nanoparticle-mediated knockdown of the DNA repair protein apurinic endonuclease 1 (Ape1) sensitizes GBM cells to radiotherapy and extend survival in a genetic mouse model of GBM. Specific knockdown of Ape1 activity by 30% in brain tumor tissue doubled the extended survival achieved with radiotherapy alone. Ape1 is a promising target for increasing the effectiveness of radiotherapy, and nanoparticle-mediated delivery of siRNA is a promising strategy for tumor specific knockdown of Ape1.


Asunto(s)
Neoplasias Encefálicas/radioterapia , Reparación del ADN , Portadores de Fármacos/química , Glioblastoma/radioterapia , Nanopartículas/química , ARN Interferente Pequeño/uso terapéutico , Animales , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/efectos de la radiación , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Línea Celular Tumoral , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Modelos Animales de Enfermedad , Glioblastoma/genética , Glioblastoma/patología , Glioblastoma/terapia , Ratones , ARN Interferente Pequeño/administración & dosificación , Tratamiento con ARN de Interferencia/métodos
12.
Proc Natl Acad Sci U S A ; 111(14): 5248-53, 2014 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-24706837

RESUMEN

Glioblastoma is the most common adult primary brain tumor and has a dismal median survival. Radiation is a mainstay of treatment and significantly improves survival, yet recurrence is nearly inevitable. Better understanding the radiation response of glioblastoma will help improve strategies to treat this devastating disease. Here, we present a comprehensive study of the in vivo radiation response of glioma cells in a mouse model of proneural glioblastoma. These tumors are a heterogeneous mix of cell types with differing radiation sensitivities. To explicitly study the gene expression changes comprising the radiation response of the Olig2(+) tumor bulk cells, we used translating ribosome affinity purification (TRAP) from Olig2-TRAP transgenic mice. Comparing both ribosome-associated and total pools of mRNA isolated from Olig2(+) cells indicated that the in vivo gene expression response to radiation occurs primarily at the total transcript level. Genes related to apoptosis and cell growth were significantly altered. p53 and E2F were implicated as major regulators of the radiation response, with p53 activity needed for the largest gene expression changes after radiation. Additionally, radiation induced a marked shift away from a proneural expression pattern toward a mesenchymal one. This shift occurs in Olig2(+) cells within hours and in multiple genetic backgrounds. Targets for Stat3 and CEBPB, which have been suggested to be master regulators of a mesenchymal shift, were also up-regulated by radiation. These data provide a systematic description of the events following radiation and may be of use in identifying biological processes that promote glioma radioresistance.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Mesodermo/metabolismo , Neuronas/metabolismo , Tolerancia a Radiación/genética , Transcripción Genética , Proteína p53 Supresora de Tumor/fisiología , Animales , Animales Recién Nacidos , Factores de Transcripción E2F/fisiología , Mesodermo/citología , Ratones , Ratones Transgénicos , Neuronas/citología , ARN Mensajero/genética , Proteína p53 Supresora de Tumor/genética
13.
Proc Natl Acad Sci U S A ; 109(8): 3041-6, 2012 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-22323597

RESUMEN

Glioblastoma (GBM) is distinguished by a high degree of intratumoral heterogeneity, which extends to the pattern of expression and amplification of receptor tyrosine kinases (RTKs). Although most GBMs harbor RTK amplifications, clinical trials of small-molecule inhibitors targeting individual RTKs have been disappointing to date. Activation of multiple RTKs within individual GBMs provides a theoretical mechanism of resistance; however, the spectrum of functional RTK dependence among tumor cell subpopulations in actual tumors is unknown. We investigated the pattern of heterogeneity of RTK amplification and functional RTK dependence in GBM tumor cell subpopulations. Analysis of The Cancer Genome Atlas GBM dataset identified 34 of 463 cases showing independent focal amplification of two or more RTKs, most commonly platelet-derived growth factor receptor α (PDGFRA) and epidermal growth factor receptor (EGFR). Dual-color fluorescence in situ hybridization was performed on eight samples with EGFR and PDGFRA amplification, revealing distinct tumor cell subpopulations amplified for only one RTK; in all cases these predominated over cells amplified for both. Cell lines derived from coamplified tumors exhibited genotype selection under RTK-targeted ligand stimulation or pharmacologic inhibition in vitro. Simultaneous inhibition of both EGFR and PDGFR was necessary for abrogation of PI3 kinase pathway activity in the mixed population. DNA sequencing of isolated subpopulations establishes a common clonal origin consistent with late or ongoing divergence of RTK genotype. This phenomenon is especially common among tumors with PDGFRA amplification: overall, 43% of PDGFRA-amplified GBM were found to have amplification of EGFR or the hepatocyte growth factor receptor gene (MET) as well.


Asunto(s)
Receptores ErbB/genética , Amplificación de Genes , Heterogeneidad Genética , Glioblastoma/enzimología , Glioblastoma/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Proliferación Celular , Segregación Cromosómica/genética , Simulación por Computador , Genoma Humano/genética , Glioblastoma/patología , Humanos , Hibridación Fluorescente in Situ
14.
Sci Adv ; 8(40): eabo6789, 2022 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-36206341

RESUMEN

Temporally regulated alternative splicing choices are vital for proper development, yet the wrong splice choice may be detrimental. Here, we highlight a previously unidentified role for the neurotrophin receptor splice variant TrkB.T1 in neurodevelopment, embryogenesis, transformation, and oncogenesis across multiple tumor types in humans and mice. TrkB.T1 is the predominant NTRK2 isoform across embryonic organogenesis, and forced overexpression of this embryonic pattern causes multiple solid and nonsolid tumors in mice in the context of tumor suppressor loss. TrkB.T1 also emerges as the predominant NTRK isoform expressed in a wide range of adult and pediatric tumors, including those harboring tropomyosin receptor kinase fusions. Affinity purification-mass spectrometry proteomic analysis reveals distinct interactors with known developmental and oncogenic signaling pathways such as Wnt, transforming growth factor-ß, Sonic Hedgehog, and Ras. From alterations in splicing factors to changes in gene expression, the discovery of isoform specific oncogenes with embryonic ancestry has the potential to shape the way we think about developmental systems and oncology.

15.
Nat Commun ; 13(1): 4478, 2022 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-35918310

RESUMEN

Intracranial germ cell tumors (IGCTs) are rare brain neoplasms that mainly occur in children and adolescents with a particularly high incidence in East Asian populations. Here, we conduct a genome-wide association study (GWAS) of 133 patients with IGCTs and 762 controls of Japanese ancestry. A common 4-bp deletion polymorphism in an enhancer adjacent to BAK1 is significantly associated with the disease risk (rs3831846; P = 2.4 × 10-9, odds ratio = 2.46 [95% CI: 1.83-3.31], minor allele frequency = 0.43). Rs3831846 is in strong linkage disequilibrium with a testicular GCTs susceptibility variant rs210138. In-vitro reporter assays reveal rs3831846 to be a functional variant attenuating the enhancer activity, suggesting its contribution to IGCTs predisposition through altering BAK1 expression. Risk alleles of testicular GCTs derived from the European GWAS show significant positive correlations in the effect sizes with the Japanese IGCTs GWAS (P = 1.3 × 10-4, Spearman's ρ = 0.48). These results suggest the shared genetic susceptibility of GCTs beyond ethnicity and primary sites.


Asunto(s)
Neoplasias Encefálicas , Neoplasias de Células Germinales y Embrionarias , Neoplasias Testiculares , Adolescente , Alelos , Neoplasias Encefálicas/genética , Niño , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Neoplasias de Células Germinales y Embrionarias/genética , Polimorfismo de Nucleótido Simple , Neoplasias Testiculares/genética , Proteína Destructora del Antagonista Homólogo bcl-2/genética
16.
Acta Neuropathol Commun ; 9(1): 36, 2021 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-33685520

RESUMEN

Recurrent C11orf95-RELA fusions (RELAFUS) are the hallmark of supratentorial ependymomas. The presence of RELA as the fusion partner indicates a close association of aberrant NF-κB activity with tumorigenesis. However, the oncogenic role of the C11orf95 has not been determined. Here, we performed ChIP-seq analyses to explore genomic regions bound by RELAFUS and H3K27ac proteins in human 293T and mouse ependymoma cells. We then utilized published RNA-Seq data from human and mouse RELAFUS tumors and identified target genes that were directly regulated by RELAFUS in these tumors. Subsequent transcription factor motif analyses of RELAFUS target genes detected a unique GC-rich motif recognized by the C11orf95 moiety, that is present in approximately half of RELAFUS target genes. Luciferase assays confirmed that a promoter carrying this motif is sufficient to drive RELAFUS-dependent gene expression. Further, the RELAFUS target genes were found to be overlapped with Rela target genes primarily via non-canonical NF-κB binding sites. Using a series of truncation and substitution mutants of RELAFUS, we also show that the activation domain in the RELAFUS moiety is necessary for the regulation of gene expression of these RELAFUS target genes. Lastly, we performed an anti-cancer drug screening with mouse ependymoma cells and identified potential anti-ependymoma drugs that are related to the oncogenic mechanism of RELAFUS. These findings suggested that RELAFUS might induce ependymoma formation through oncogenic pathways orchestrated by both C11orf95 and RELA target genes. Thus, our study unveils a complex gene function of RELAFUS as an oncogenic transcription factor in RELAFUS positive ependymomas.


Asunto(s)
Proteínas de Unión al ADN/genética , Ependimoma/genética , Epigénesis Genética , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteínas/genética , Factor de Transcripción ReIA/genética , Animales , Neoplasias Encefálicas/genética , Expresión Génica , Regulación de la Expresión Génica , Técnicas Genéticas , Células HEK293 , Humanos , Ratones , FN-kappa B/metabolismo , Neoplasias Supratentoriales/genética
17.
Hum Cell ; 34(1): 271-278, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32997328

RESUMEN

Atypical teratoid/rhabdoid tumor (AT/RT) is a rare intracranial tumor occurring predominantly in young children. The prognosis is poor, and no effective treatment is currently available. To develop novel effective therapies, there is a need for experimental models for AT/RT. In this research, we established a cell line from a patient's AT/RT tissue (designated ATRT_OCGH) and performed drug screening using 164 FDA-approved anti-cancer agents, to identify candidates for therapeutic options. We found that bortezomib, a proteasome inhibitor, was among the agents for which the cell line showed high sensitivity, along with tyrosine kinase inhibitors, topoisomerase inhibitors, and histone deacetylase inhibitors, which are known to exert anti-AT/RT effects. Concomitant use of panobinostat potentiated the inhibitory effect of bortezomib on AT/RT cell proliferation. Our findings may provide a rationale for considering combination therapy of panobinostat and bortezomib for treatment of AT/RT.


Asunto(s)
Antineoplásicos/farmacología , Bortezomib/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Ensayos de Selección de Medicamentos Antitumorales/métodos , Inhibidores de Proteasoma/farmacología , Tumor Rabdoide/tratamiento farmacológico , Tumor Rabdoide/patología , Teratoma/tratamiento farmacológico , Teratoma/patología , Antineoplásicos/administración & dosificación , Bortezomib/administración & dosificación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , Panobinostat/administración & dosificación , Panobinostat/farmacología , Pronóstico , Inhibidores de Proteasoma/administración & dosificación
18.
Acta Neuropathol Commun ; 8(1): 203, 2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-33228790

RESUMEN

Recurrent RELA and YAP1 fusions are intimately associated with tumorigenesis in supratentorial ependymomas. Chromothripsis and focal copy number alterations involving 11q are hallmarks of these tumors. However, it is unknown whether the chromosomal alterations are a direct causal event resulting in fusion transcripts. In addition, the biological significance of the RELA fusion variants and YAP1 fusions is not yet fully characterized. In this study, we generated gene rearrangements on 11q with the CRISPR/Cas9 system and investigated the formation of oncogenic ependymoma fusion genes. Further, we examined the oncogenic potential of RELA fusion variants and YAP1 fusions in a lentiviral gene transfer model. We observed that endogenous RELA fusion events were successfully induced by CRISPR/Cas9-mediated genome rearrangement in cultured cells. In vivo genome editing in mouse brain resulted in the development of ependymoma-like brain tumors that harbored the Rela fusion gene. All RELA fusion variants tested, except a variant lacking the Rel homology domain, were able to induce tumor formation, albeit with different efficacy. Furthermore, expression of YAP1-FAM118B and YAP1-MAMLD1 fusions induced the formation of spindle-cell-like tumors at varying efficacy. Our results indicate that chromosomal rearrangements involving the Rela locus are the causal event for the formation of Rela fusion-driven ependymomas in mice. Furthermore, the type of RELA. fusion might affect the aggressiveness of tumors and that the Rel homology domain is essential for the oncogenic functions of RELA. fusions. The YAP1 fusion genes are also oncogenic when expressed in mice.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Neoplasias Encefálicas/genética , Carcinogénesis/genética , Proteínas de Unión al ADN/genética , Ependimoma/genética , Fusión de Oncogenes/genética , Factor de Transcripción ReIA/genética , Factores de Transcripción/genética , Animales , Sistemas CRISPR-Cas , Células Cultivadas , Cromotripsis , Variaciones en el Número de Copia de ADN , Edición Génica , Técnicas de Transferencia de Gen , Ratones , Fenotipo , Proteínas Señalizadoras YAP
19.
Nat Commun ; 11(1): 2977, 2020 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-32532995

RESUMEN

Independent scientific achievements have led to the discovery of aberrant splicing patterns in oncogenesis, while more recent advances have uncovered novel gene fusions involving neurotrophic tyrosine receptor kinases (NTRKs) in gliomas. The exploration of NTRK splice variants in normal and neoplastic brain provides an intersection of these two rapidly evolving fields. Tropomyosin receptor kinase B (TrkB), encoded NTRK2, is known for critical roles in neuronal survival, differentiation, molecular properties associated with memory, and exhibits intricate splicing patterns and post-translational modifications. Here, we show a role for a truncated NTRK2 splice variant, TrkB.T1, in human glioma. TrkB.T1 enhances PDGF-driven gliomas in vivo, augments PDGF-induced Akt and STAT3 signaling in vitro, while next generation sequencing broadly implicates TrkB.T1 in the PI3K signaling cascades in a ligand-independent fashion. These TrkB.T1 findings highlight the importance of expanding upon whole gene and gene fusion analyses to include splice variants in basic and translational neuro-oncology research.


Asunto(s)
Neoplasias Encefálicas/genética , Glioma/genética , Glicoproteínas de Membrana/genética , Oncogenes/genética , Isoformas de ARN/genética , Empalme del ARN , Receptor trkB/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Carcinogénesis/genética , Células Cultivadas , Perfilación de la Expresión Génica , Ontología de Genes , Glioma/metabolismo , Glioma/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Glicoproteínas de Membrana/metabolismo , Ratones , Células 3T3 NIH , Células-Madre Neurales/metabolismo , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Isoformas de ARN/metabolismo , Receptor trkB/metabolismo , Transducción de Señal/genética
20.
Brain Tumor Pathol ; 36(1): 14-19, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30350109

RESUMEN

The proto-oncogene tyrosine-protein kinase ROS1 (ROS1) is a tyrosine kinase that is closely related to anaplastic lymphoma kinase receptor (ALK). We describe a novel KLC1-ROS1 fusion identified in a case of pediatric low-grade glioma. This was detected by RNA sequencing and confirmed by reverse-transcription PCR and fluorescent in situ hybridization. Immunohistochemical staining for ROS1 was positive in the tumor cytoplasm. In vitro analysis demonstrated the oncogenic activity of this fusion, which was suppressed by the ALK/ROS1 inhibitor, crizotinib. Our case and others suggest that various ROS1 fusions might be present in a subset of pediatric gliomas, which could be targeted for therapy.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Fusión Génica , Glioma/genética , Glioma/patología , Proteínas Asociadas a Microtúbulos/genética , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Neoplasias Encefálicas/tratamiento farmacológico , Preescolar , Crizotinib/farmacología , Femenino , Fusión Génica/efectos de los fármacos , Glioma/tratamiento farmacológico , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Cinesinas , Terapia Molecular Dirigida , Estadificación de Neoplasias , Proteínas Tirosina Quinasas/metabolismo , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA