Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Acta Neuropathol ; 146(3): 527-541, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37450044

RESUMEN

Atypical teratoid/rhabdoid tumors (AT/RT) are the most common malignant brain tumors manifesting in infancy. They split into four molecular types. The major three (AT/RT-SHH, AT/RT-TYR, and AT/RT-MYC) all carry mutations in SMARCB1, the fourth quantitatively smaller type is characterized by SMARCA4 mutations (AT/RT-SMARCA4). Molecular characteristics of disease recurrence or metastatic spread, which go along with a particularly dismal outcome, are currently unclear. Here, we investigated tumor tissue from 26 patients affected by AT/RT to identify signatures of recurrences in comparison with matched primary tumor samples. Microscopically, AT/RT recurrences demonstrated a loss of architecture and significantly enhanced mitotic activity as compared to their related primary tumors. Based on DNA methylation profiling, primary tumor and related recurrence were grossly similar, but three out of 26 tumors belonged to a different molecular type or subtype after second surgery compared to related primary lesions. Copy number variations (CNVs) differed in six cases, showing novel gains on chromosome 1q or losses of chromosome 10 in recurrences as the most frequent alterations. To consolidate these observations, our cohort was combined with a data set of unmatched primary and recurrent AT/RT, which demonstrated chromosome 1q gain and 10 loss in 18% (n = 7) and 11% (n = 4) of the recurrences (n = 38) as compared to 7% (n = 3) and 0% (n = 0) in the primary tumors (n = 44), respectively. Similar to the observations made by DNA methylation profiling, RNA sequencing of our cohort revealed AT/RT primary tumors and matched recurrences clustering closely together. However, a number of genes showed significantly altered expression in AT/RT-SHH recurrences. Many of them are known tumor driving growth factors, involved in embryonal development and tumorigenesis, or are cell-cycle-associated. Overall, our work identifies subtle molecular changes that occur in the course of the disease and that may help define novel therapeutic targets for AT/RT recurrences.


Asunto(s)
Variaciones en el Número de Copia de ADN , Progresión de la Enfermedad , Epigénesis Genética , Perfilación de la Expresión Génica , Recurrencia , Tumor Rabdoide , Teratoma , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Cromosomas Humanos Par 1/genética , Cromosomas Humanos Par 10/genética , Estudios de Cohortes , Células Dendríticas , Variaciones en el Número de Copia de ADN/genética , Metilación de ADN , Histología , Mitosis , Tumor Rabdoide/clasificación , Tumor Rabdoide/genética , Tumor Rabdoide/inmunología , Tumor Rabdoide/patología , Análisis de Secuencia de ARN , Teratoma/clasificación , Teratoma/genética , Teratoma/inmunología , Teratoma/patología , Factores de Transcripción/genética , Regulación Neoplásica de la Expresión Génica/genética
2.
Childs Nerv Syst ; 34(3): 581-584, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29167993

RESUMEN

Glycogen storage disease (GSD) 0a is a rare congenital metabolic disease with symptoms in infancy and childhood caused by biallelic GYS2 germline variants. A predisposition to cancer has not been described yet. We report here a boy with GSD 0a, who developed a malignant brain tumor at the age of 4.5 years. The tumor was classified as a group 3 medulloblastoma, and the patient died from cancer 27 months after initial tumor diagnosis. This case appears interesting as group 3 medulloblastoma is so far not known to arise in hereditary syndromes and the biology of sporadic group 3 medulloblastoma is largely unknown.


Asunto(s)
Neoplasias Cerebelosas/diagnóstico por imagen , Mutación de Línea Germinal , Enfermedad del Almacenamiento de Glucógeno/diagnóstico por imagen , Meduloblastoma/diagnóstico por imagen , Neoplasias Cerebelosas/complicaciones , Neoplasias Cerebelosas/genética , Preescolar , Resultado Fatal , Mutación de Línea Germinal/genética , Enfermedad del Almacenamiento de Glucógeno/complicaciones , Enfermedad del Almacenamiento de Glucógeno/genética , Humanos , Masculino , Meduloblastoma/complicaciones , Meduloblastoma/genética
4.
Pediatr Hematol Oncol ; 34(1): 17-23, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28085536

RESUMEN

Neuroblastoma (NBL) stage 4s is an incompletely understood phenomenon with variable clinical course. While the majority of patients may undergo spontaneous regression and achieve complete resolution without intensive therapy, a small proportion is at increased risk of developing secondary complications. One such situation is liver insufficiency due to diffuse metastases. We report a patient suffering from NBL 4S who required double lifesaving liver transplantation. Abdominal and respiratory complications due to hepatomegaly are crucial determinants for treatment intensity and duration in 4S NBL [1,2] . We provide an algorithm in order to facilitate the clinical decision when dealing with similar potentially life-threatening events.


Asunto(s)
Algoritmos , Toma de Decisiones , Neoplasias Hepáticas/cirugía , Trasplante de Hígado , Neuroblastoma/cirugía , Adulto , Humanos , Neoplasias Hepáticas/secundario , Masculino , Metástasis de la Neoplasia , Estadificación de Neoplasias
5.
Int J Cancer ; 135(4): 989-95, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24420698

RESUMEN

Rhabdoid tumors are highly aggressive tumors occurring in infants and very young children. Despite multimodal and intensive therapy prognosis remains poor. Molecular analyses have uncovered several deregulated pathways, among them the CDK4/6-Rb-, the WNT- and the Sonic hedgehog (SHH) pathways. The SHH pathway is activated in rhabdoid tumors by GLI1 overexpression. Here, we demonstrate that arsenic trioxide (ATO) inhibits tumor cell growth of malignant rhabdoid tumors in vitro and in a mouse xenograft model by suppressing Gli1. Our data uncover ATO as a promising therapeutic approach to improve prognosis for rhabdoid tumor patients.


Asunto(s)
Antineoplásicos/farmacología , Arsenicales/farmacología , Regulación Neoplásica de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/metabolismo , Óxidos/farmacología , Tumor Rabdoide/tratamiento farmacológico , Factores de Transcripción/metabolismo , Animales , Apoptosis , Trióxido de Arsénico , Ciclo Celular , Proliferación Celular , Biología Computacional , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Proteínas Hedgehog/metabolismo , Humanos , Ratones , Ratones SCID , Trasplante de Neoplasias , Pronóstico , Transducción de Señal , Proteína con Dedos de Zinc GLI1
6.
Pediatr Hematol Oncol ; 30(7): 587-604, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23848359

RESUMEN

Rhabdoid tumors are rare but highly aggressive tumors with a predilection for infants and young children. The majority of these tumors harbor biallelic mutations in SMARCB1/INI1/hSNF5. Rather rare cases with mutations in other SWI/SNF core members such as BRG1 are on record. Rhabdoid tumors have only recently been registered and treated according to specifically designed treatment recommendations and in the framework of clinical trials. Within the last decade, prognosis has improved significantly but at least 50% of patients still relapse and subsequently almost inevitably succumb to their disease. This review summarizes past and current clinical approaches and presents an overview of the rationales for targeted therapy with potential for future clinical treatment trials for rhabdoid tumors.


Asunto(s)
Proteínas Cromosómicas no Histona/genética , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Mutación , Proteínas Nucleares/genética , Tumor Rabdoide/genética , Tumor Rabdoide/terapia , Factores de Transcripción/genética , Ensayos Clínicos como Asunto , Humanos , Tumor Rabdoide/metabolismo , Tumor Rabdoide/patología , Proteína SMARCB1
7.
Nat Commun ; 13(1): 1544, 2022 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-35318328

RESUMEN

Rhabdoid tumors (RT) are rare and highly aggressive pediatric neoplasms. Their epigenetically-driven intertumoral heterogeneity is well described; however, the cellular origin of RT remains an enigma. Here, we establish and characterize different genetically engineered mouse models driven under the control of distinct promoters and being active in early progenitor cell types with diverse embryonic onsets. From all models only Sox2-positive progenitor cells give rise to murine RT. Using single-cell analyses, we identify distinct cells of origin for the SHH and MYC subgroups of RT, rooting in early stages of embryogenesis. Intra- and extracranial MYC tumors harbor common genetic programs and potentially originate from fetal primordial germ cells (PGCs). Using PGC specific Smarcb1 knockout mouse models we validate that MYC RT originate from these progenitor cells. We uncover an epigenetic imbalance in MYC tumors compared to PGCs being sustained by epigenetically-driven subpopulations. Importantly, treatments with the DNA demethylating agent decitabine successfully impair tumor growth in vitro and in vivo. In summary, our work sheds light on the origin of RT and supports the clinical relevance of DNA methyltransferase inhibitors against this disease.


Asunto(s)
Tumor Rabdoide , Animales , Células Germinativas/patología , Humanos , Ratones , Tumor Rabdoide/genética , Tumor Rabdoide/patología , Proteína SMARCB1/genética , Análisis de la Célula Individual , Transcriptoma
8.
Brain Pathol ; 31(1): 33-44, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32633004

RESUMEN

The molecular biology of ependymomas is not well understood and this is particularly true for ependymoma relapses. We aimed at finding out if and to which extent, relapses differ from their corresponding primary tumors on the morphological, chromosomal and epigenetic level. We investigated 24 matched ependymoma primary and relapsed tumor samples and, as a first step, compared cell density, necrosis, vessel proliferation, Ki67 proliferative index, trimethylation at H3K27 and expression of CXorf67. For the investigation of global methylation profiles, we used public data in order to analyze copy number variation profiles, differential methylation, methylation status and fractions of hypo- and hypermethylated CpGs in different epigenomic substructures. Morphologically, we found a significant increase with relapse in cell density and proliferation. H3K27 trimethylation and CXorf67 expression remained stable between primary and relapse tumor samples, and the analysis of DNA methylation profiles neither revealed significant differences in copy number variations nor differentially methylated regions. Significant differences in the methylation status were found for CpG islands, but also in N Shelves or S Shelves, depending on the molecular subgroup. The fraction of probes changing their methylation in the epigenomic substructures appeared subgroup-specific. Most changes occur in CpG islands, for which relapsed tumors demonstrate higher methylation values than primary tumors. The morphological differences reflect increased aggressiveness upon ependymoma relapse, but, despite slight changes, this observation does not appear to be sufficiently explained by epigenetic changes.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Ependimoma/genética , Ependimoma/patología , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Adolescente , Adulto , Niño , Epigenoma , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Adulto Joven
9.
J Neuropathol Exp Neurol ; 80(1): 52-59, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33270865

RESUMEN

Central nervous system neuroblastoma with FOXR2 activation (CNS NB FOXR2) has recently been described as a class of brain tumors sharing common genetic events and a highly similar DNA methylation profile. Most of these tumors have previously been diagnosed as primitive neuroectodermal tumor (PNET). Whereas the entity of PNET has been removed from the WHO classification of brain tumors in its current edition, CNS neuroblastoma was kept as an entity, but still lacks any molecular detail. Here, we describe 8 cases of CNS NB FOXR2 focusing on histomorphological and immunohistochemical features and include magnetic resonance imaging (MRI) for 2 of these cases. MRI revealed large supratentorial masses in superficial location with prominent cysts and necrosis, but little edema. Diffusion and enhancement characteristics were variable. Histological analyses showed that most of the cases displayed neuronal differentiation with necrosis, endothelial proliferation, and high vascularity. Immunohistochemistry revealed strong expression of synaptophysin, MAP2, and OLIG2 as well as moderate proliferation. These findings suggest that tumors with the molecular diagnosis of CNS NB FOXR2 may fit well into the WHO entity of CNS neuroblastoma. Our findings may be helpful when establishing an integrated diagnosis and may be indispensable if molecular data are unavailable.


Asunto(s)
Neoplasias Encefálicas/patología , Encéfalo/patología , Neuroblastoma/patología , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Niño , Preescolar , Variaciones en el Número de Copia de ADN , Metilación de ADN , Femenino , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Humanos , Lactante , Imagen por Resonancia Magnética , Masculino , Neuroblastoma/diagnóstico por imagen , Neuroblastoma/genética , Neuroblastoma/metabolismo
10.
Cancer Lett ; 477: 10-18, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32112900

RESUMEN

Medulloblastoma (MB) is the most frequent malignant brain tumour in children with a poor outcome. Divided into four molecular subgroups, MB of the Sonic hedgehog (SHH) subgroup accounts for approximately 25% of the cases and is driven by mutations within components of the SHH pathway, such as its receptors PTCH1 or SMO. A fraction of these cases additionally harbour PIK3CA mutations, the relevance of which is so far unknown. To unravel the role of Pik3ca mutations alone or in combination with a constitutively activated SHH signalling pathway, transgenic mice were used. These mice show mutated variants within Smo, Ptch1 or Pik3ca genes in cerebellar granule neuron precursors, which represent the cellular origin of SHH MB. Our results show that Pik3ca mutations alone are insufficient to cause developmental alterations or to initiate MB. However, they significantly accelerate the growth of Shh MB, induce tumour spread throughout the cerebrospinal fluid, and result in lower survival rates of mice with a double Pik3caH1047R/SmoM2 or Pik3caH1047R/Ptch1 mutation. Therefore, PIK3CA mutations in SHH MB may represent a therapeutic target for first and second line combination treatments.


Asunto(s)
Neoplasias Cerebelosas/genética , Fosfatidilinositol 3-Quinasa Clase I/genética , Meduloblastoma/genética , Mutación , Animales , Neoplasias Cerebelosas/patología , Modelos Animales de Enfermedad , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Meduloblastoma/patología , Ratones Transgénicos , Neoplasias Experimentales/diagnóstico por imagen , Neoplasias Experimentales/genética , Neoplasias Experimentales/mortalidad , Receptor Patched-1/genética , Receptor Smoothened/genética , Neoplasias de la Médula Espinal/secundario , Tasa de Supervivencia , Secuenciación Completa del Genoma
11.
Eur J Hum Genet ; 26(8): 1083-1093, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29706634

RESUMEN

Germline variants that affect function are found in seven genes of the BAF chromatin-remodeling complex. They are linked to a broad range of diseases that, according to the gene affected, range from non-syndromic or syndromic neurodevelopmental disorders to low-grade tumors and malignancies. In the current meta-analysis, we evaluate genetic and clinical data from more than 400 families and 577 patients affected by BAF germline alterations. We focus on SMARCB1, including 43 unpublished patients from the EU-RHAB registry and our institution. For this gene, we further demonstrate whole gene as well as exon deletions and truncating variants to be associated with malignancy and early-onset disease. In contrast, non-truncating variants are associated with non-malignant disorders, such as Coffin-Siris syndrome or late-onset tumors like schwannoma or meningioma (p < 0.0001). SMARCB1 germline variants are distributed across the gene with variants in exons 1, 2, 8, and 9 being associated with low-grade entities, and single-nucleotide variants or indels outside of exon 9 that appear in patients with malignancies (p < 0.001). We attribute variants in specific BAF genes to certain disease entities. Finally, single-nucleotide variants and indels are sometimes detected in the healthy relatives of tumor patients, while Coffin-Siris syndrome and Nicolaides-Baraitser syndrome generally seem to appear de novo. Our findings add further information on the genotype-phenotype association of germline variants detected in genes of the BAF complex. Functional studies are urgently needed for a deeper understanding of BAF-related disorders and may take advantage from the comprehensive information gathered in this article.


Asunto(s)
Anomalías Múltiples/genética , Cara/anomalías , Mutación de Línea Germinal , Deformidades Congénitas de la Mano/genética , Discapacidad Intelectual/genética , Neoplasias Meníngeas/genética , Meningioma/genética , Micrognatismo/genética , Cuello/anomalías , Neurilemoma/genética , Proteína SMARCB1/genética , Anomalías Múltiples/patología , Cara/patología , Deformidades Congénitas de la Mano/patología , Humanos , Discapacidad Intelectual/patología , Neoplasias Meníngeas/patología , Meningioma/patología , Micrognatismo/patología , Cuello/patología , Neurilemoma/patología , Fenotipo , Polimorfismo de Nucleótido Simple
12.
Oncotarget ; 8(49): 84986-84995, 2017 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-29156698

RESUMEN

Rhabdoid tumors are caused by the deletion of SMARCB1, whose protein encodes the SMARCB1 subunit of the chromatin remodeling complex SWI/SNF that is involved in global chromatin organization and gene expression control. Simultaneously inhibiting the main players involved in the deregulated transcription machinery is a promising option for preventing exaggerated tumor cell proliferation and survival as it may bypass compensatory mechanisms. In support of this hypothesis, we report efficient impairment of cellular proliferation and strong induction of cell death elicited by inhibition of bromodomain protein BRD4 and transcription kinase CDK9 using small molecular compounds. Combination of both compounds efficiently represses antiapoptotic genes and the oncogene MYC. Our results provide a novel approach for the treatment of RT.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA