Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Oral Maxillofac Surg ; 81(8): 1021-1024, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37220870

RESUMEN

BACKGROUND: Superior orbital rim fractures are challenging fractures as they often concomitantly occur with additional calvaria fractures. Virtual surgical planning (VSP) has been underutilized in this area of craniomaxillofacial trauma for reconstruction. PURPOSE: The purpose of this study is to qualitatively describe the use of VSP and anatomically perfected stereolithic models in treatment of superior orbital rim fractures in combined neurosurgery/oral and maxillofacial surgery cases. STUDY DESIGN, SETTING, SAMPLE: This study is a retrospective case series of subjects who were treated at the Massachusetts General Hospital (July 2022 to November 2022). Inclusion criteria include subjects who had both calvaria and maxillofacial injuries requiring concurrent operative intervention on their superior orbital rim fractures and the use of VSP. PREDICTOR/EXPOSURE/INDEPENDENT VARIABLE: Not applicable. MAIN OUTCOME VARIABLE: The outcome variable of interest is the difference in the planned position of the orbital rim repair compared to the actual position achieved. COVARIATES: None. ANALYSES: Heat map analysis was used to compare the difference in the planned position versus the actual position achieved. RESULTS: There were six orbits (five subjects, mean age 33.8 ± 21.49 years) that met the criteria. The mean difference in planned versus actual orbital volume achieved was 2.52 ± 2.48 cm3. The superimposition of the postoperative scan to the planned simulation revealed 84% ± 3.27% of the voxel surface was within +2 and -2 millimeters of its planned position. CONCLUSION AND RELEVANCE: This study has demonstrated the use of VSP in combined neurosurgery and oral and maxillofacial surgery procedures in the fixation of superior orbital rim fractures. This case series highlights that the postoperative position achieved in the six orbits was within 84% of the planned position.


Asunto(s)
Traumatismos Maxilofaciales , Fracturas Orbitales , Procedimientos de Cirugía Plástica , Humanos , Niño , Adolescente , Adulto Joven , Adulto , Persona de Mediana Edad , Estudios Retrospectivos , Órbita/cirugía , Traumatismos Maxilofaciales/cirugía , Fracturas Orbitales/diagnóstico por imagen , Fracturas Orbitales/cirugía
2.
Genes Chromosomes Cancer ; 54(9): 542-54, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26032282

RESUMEN

As subsets of pheochromocytomas (PCCs) lack a defined molecular etiology, we sought to characterize the mutational landscape of PCCs to identify novel gene candidates involved in disease development. A discovery cohort of 15 PCCs wild type for mutations in PCC susceptibility genes underwent whole-exome sequencing, and an additional 83 PCCs served as a verification cohort for targeted sequencing of candidate mutations. A low rate of nonsilent single nucleotide variants (SNVs) was detected (6.1/sample). Somatic HRAS and EPAS1 mutations were observed in one case each, whereas the remaining 13 cases did not exhibit variants in established PCC genes. SNVs aggregated in apoptosis-related pathways, and mutations in COSMIC genes not previously reported in PCCs included ZAN, MITF, WDTC1, and CAMTA1. Two somatic mutations and one constitutional variant in the well-established cancer gene lysine (K)-specific methyltransferase 2D (KMT2D, MLL2) were discovered in one sample each, prompting KMT2D screening using focused exome-sequencing in the verification cohort. An additional 11 PCCs displayed KMT2D variants, of which two were recurrent. In total, missense KMT2D variants were found in 14 (11 somatic, two constitutional, one undetermined) of 99 PCCs (14%). Five cases displayed somatic mutations in the functional FYR/SET domains of KMT2D, constituting 36% of all KMT2D-mutated PCCs. KMT2D expression was upregulated in PCCs compared to normal adrenals, and KMT2D overexpression positively affected cell migration in a PCC cell line. We conclude that KMT2D represents a recurrently mutated gene with potential implication for PCC development.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/genética , Proteínas de Unión al ADN/genética , Exoma , Mutación , Proteínas de Neoplasias/genética , Feocromocitoma/genética , Neoplasias de las Glándulas Suprarrenales/etiología , Línea Celular Tumoral , Estudios de Cohortes , Proteínas de Unión al ADN/metabolismo , Femenino , Dosificación de Gen , Humanos , Masculino , Proteínas de Neoplasias/metabolismo , Feocromocitoma/etiología , Feocromocitoma/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Análisis de Secuencia de Proteína , Transcriptoma
3.
J Neurosurg Case Lessons ; 7(9)2024 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-38408351

RESUMEN

BACKGROUND: 5-Aminolevulinic acid (5-ALA) fluorescence-guided surgery is a well-established technique for resecting high-grade gliomas. However, its application in meningiomas, especially those previously treated with radiation therapy, remains under investigation. OBSERVATIONS: A 48-year-old female with recurrent anaplastic meningioma, World Health Organization grade 3, underwent a right-sided craniotomy using off-label 5-ALA as a surgical adjunct. The patient had previously undergone brachytherapy seed implantation (20 × cesium 131) for tumor management. During the surgery, a large fluorescent tumor mass adjacent to the brachytherapy-treated area was resected, and the prior brachytherapy seeds were removed. Interestingly, the surrounding brain tissue in the irradiated area showed robust 5-ALA fluorescence. Pathological examination confirmed that the fluorescent brain tissue was nonneoplastic and associated with lymphocyte and macrophage infiltration. LESSONS: This case report presents unique 5-ALA fluorescence in nonneoplastic tissue following brachytherapy, which was found during the resection of recurrent anaplastic meningioma. This phenomenon may reflect an intricate interplay among radiation therapy, immune cells, the tumor microenvironment, and 5-ALA metabolism. Given that false-positive findings in fluorescence-guided surgery can lead to unnecessary tissue resection and increased surgical morbidity, further research is warranted to elucidate the mechanisms underlying this phenomenon and its implications for meningioma surgery.

4.
J Neurosurg ; 141(1): 72-78, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38277657

RESUMEN

OBJECTIVE: Foramen magnum (FM) meningiomas pose significant surgical challenges and have high morbidity and mortality rates. This study aimed to investigate the distribution of clinically actionable mutations in FM meningiomas and identify clinical characteristics associated with specific mutational profiles. METHODS: The authors conducted targeted next-generation sequencing of 62 FM meningiomas from three international institutions, covering all relevant meningioma genes (AKT1, KLF4, NF2, POLR2A, PIK3CA, SMO, TERT promoter, and TRAF7). Patients with a radiation-induced meningioma or neurofibromatosis type 2 (NF2) were excluded from the study. Additionally, patient and tumor characteristics, including age, sex, radiological features, and tumor location, were retrospectively collected and evaluated. RESULTS: The study cohort consisted of 46 female and 16 male patients. Clinically significant driver mutations were detected in 58 patients (93.5%). The most commonly observed alteration was TRAF7 mutations (26, 41.9%), followed by AKT1E17K mutations (19, 30.6%). Both mutations were significantly associated with an anterolateral tumor location relative to the brainstem (p = 0.0078). NF2 mutations were present in 11 cases (17.7%) and were associated with posterior tumor location, in contrast to tumors with TRAF7 and AKT1E17K mutations. Other common mutations in FM meningiomas included POLR2A mutations (8, 12.9%; 6 POLR2AQ403K and 2 POLR2AH439_L440del), KLF4K409Q mutations (7, 11.3%), and PIK3CA mutations (4, 6.5%; 2 PIK3CAH1047R and 2 PIK3CAE545K). POLR2A and KLF4 mutations exclusively occurred in female patients and showed no significant association with specific tumor locations. All tumors harboring AKT1E17K and POLR2A mutations displayed meningothelial histology. Ten tumors exhibited intratumoral calcification, which was significantly more frequent in NF2-mutant compared with AKT1-mutant FM meningiomas (p = 0.047). CONCLUSIONS: These findings provide important insights into the molecular genetics and clinicopathological characteristics of FM meningiomas. The identification of specific genetic alterations associated with tumor location, volume, calcification, histology, and sex at diagnosis may have implications for personalized treatment strategies in the future.


Asunto(s)
Foramen Magno , Factor 4 Similar a Kruppel , Neoplasias Meníngeas , Meningioma , Mutación , Neurofibromina 2 , Humanos , Meningioma/genética , Meningioma/patología , Masculino , Femenino , Persona de Mediana Edad , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología , Neoplasias Meníngeas/diagnóstico por imagen , Adulto , Anciano , Estudios Retrospectivos , Neurofibromina 2/genética , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/genética , Proteínas Proto-Oncogénicas c-akt/genética , ARN Polimerasa III/genética , Fosfatidilinositol 3-Quinasa Clase I/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Factores de Transcripción de Tipo Kruppel/genética , Receptor Smoothened/genética , Análisis Mutacional de ADN , Adulto Joven , Telomerasa
5.
Nat Commun ; 14(1): 6279, 2023 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-37805627

RESUMEN

Hedgehog signaling mediates embryologic development of the central nervous system and other tissues and is frequently hijacked by neoplasia to facilitate uncontrolled cellular proliferation. Meningiomas, the most common primary brain tumor, exhibit Hedgehog signaling activation in 6.5% of cases, triggered by recurrent mutations in pathway mediators such as SMO. In this study, we find 35.6% of meningiomas that lack previously known drivers acquired various types of somatic structural variations affecting chromosomes 2q35 and 7q36.3. These cases exhibit ectopic expression of Hedgehog ligands, IHH and SHH, respectively, resulting in Hedgehog signaling activation. Recurrent tandem duplications involving IHH permit de novo chromatin interactions between super-enhancers within DIRC3 and a locus containing IHH. Our work expands the landscape of meningioma molecular drivers and demonstrates enhancer hijacking of Hedgehog ligands as a route to activate this pathway  in neoplasia.


Asunto(s)
Neoplasias Meníngeas , Meningioma , Humanos , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Meningioma/genética , Ligandos , Transducción de Señal , Neoplasias Meníngeas/genética
6.
Dev Cell ; 57(14): 1776-1788.e8, 2022 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-35809564

RESUMEN

A multitude of cellular processes involve biomolecular condensates, which has led to the suggestion that diverse pathogenic mutations may dysregulate condensates. Although proof-of-concept studies have identified specific mutations that cause condensate dysregulation, the full scope of the pathological genetic variation that affects condensates is not yet known. Here, we comprehensively map pathogenic mutations to condensate-promoting protein features in putative condensate-forming proteins and find over 36,000 pathogenic mutations that plausibly contribute to condensate dysregulation in over 1,200 Mendelian diseases and 550 cancers. This resource captures mutations presently known to dysregulate condensates, and experimental tests confirm that additional pathological mutations do indeed affect condensate properties in cells. These findings suggest that condensate dysregulation may be a pervasive pathogenic mechanism underlying a broad spectrum of human diseases, provide a strategy to identify proteins and mutations involved in pathologically altered condensates, and serve as a foundation for mechanistic insights into disease and therapeutic hypotheses.


Asunto(s)
Proteínas , Humanos , Mutación/genética
8.
Science ; 368(6497): 1386-1392, 2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-32554597

RESUMEN

The nucleus contains diverse phase-separated condensates that compartmentalize and concentrate biomolecules with distinct physicochemical properties. Here, we investigated whether condensates concentrate small-molecule cancer therapeutics such that their pharmacodynamic properties are altered. We found that antineoplastic drugs become concentrated in specific protein condensates in vitro and that this occurs through physicochemical properties independent of the drug target. This behavior was also observed in tumor cells, where drug partitioning influenced drug activity. Altering the properties of the condensate was found to affect the concentration and activity of drugs. These results suggest that selective partitioning and concentration of small molecules within condensates contributes to drug pharmacodynamics and that further understanding of this phenomenon may facilitate advances in disease therapy.


Asunto(s)
Antineoplásicos/farmacología , Núcleo Celular/metabolismo , Resistencia a Antineoplásicos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Antineoplásicos/uso terapéutico , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Subunidad 1 del Complejo Mediador/genética , Subunidad 1 del Complejo Mediador/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleofosmina , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Factores de Empalme Serina-Arginina/genética , Factores de Empalme Serina-Arginina/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
Neurosurg Clin N Am ; 30(1): 95-101, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30470409

RESUMEN

For malignant gliomas, the survival benefit of new combination therapies after surgical resection is measured in weeks to months. In contrast, optimizing treatment for low-grade gliomas can potentially provide additional years of life. The relatively indolent but not benign clinical course provides the opportunity for clinicians and scientists to focus not only on the duration of survival, but also to maximize quality of life. Ideal management of low-grade gliomas among the most important yet paradoxically most neglected subjects in neuro-oncology. This article examines the molecular underpinnings of these tumors and evaluates the role of extensive surgery in maximizing outcomes.


Asunto(s)
Neoplasias Encefálicas/cirugía , Glioma/cirugía , Procedimientos Neuroquirúrgicos , Resultado del Tratamiento , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Glioma/tratamiento farmacológico , Glioma/patología , Humanos , Clasificación del Tumor , Calidad de Vida
10.
J Neurosurg ; : 1-10, 2019 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-31653806

RESUMEN

OBJECTIVE: Recent large-cohort sequencing studies have investigated the genomic landscape of meningiomas, identifying somatic coding alterations in NF2, SMARCB1, SMARCE1, TRAF7, KLF4, POLR2A, BAP1, and members of the PI3K and Hedgehog signaling pathways. Initial associations between clinical features and genomic subgroups have been described, including location, grade, and histology. However, further investigation using an expanded collection of samples is needed to confirm previous findings, as well as elucidate relationships not evident in smaller discovery cohorts. METHODS: Targeted sequencing of established meningioma driver genes was performed on a multiinstitution cohort of 3016 meningiomas for classification into mutually exclusive subgroups. Relevant clinical information was collected for all available cases and correlated with genomic subgroup. Nominal variables were analyzed using Fisher's exact tests, while ordinal and continuous variables were assessed using Kruskal-Wallis and 1-way ANOVA tests, respectively. Machine-learning approaches were used to predict genomic subgroup based on noninvasive clinical features. RESULTS: Genomic subgroups were strongly associated with tumor locations, including correlation of HH tumors with midline location, and non-NF2 tumors in anterior skull base regions. NF2 meningiomas were significantly enriched in male patients, while KLF4 and POLR2A mutations were associated with female sex. Among histologies, the results confirmed previously identified relationships, and observed enrichment of microcystic features among "mutation unknown" samples. Additionally, KLF4-mutant meningiomas were associated with larger peritumoral brain edema, while SMARCB1 cases exhibited elevated Ki-67 index. Machine-learning methods revealed that observable, noninvasive patient features were largely predictive of each tumor's underlying driver mutation. CONCLUSIONS: Using a rigorous and comprehensive approach, this study expands previously described correlations between genomic drivers and clinical features, enhancing our understanding of meningioma pathogenesis, and laying further groundwork for the use of targeted therapies. Importantly, the authors found that noninvasive patient variables exhibited a moderate predictive value of underlying genomic subgroup, which could improve with additional training data. With continued development, this framework may enable selection of appropriate precision medications without the need for invasive sampling procedures.

12.
Genome Med ; 9(1): 12, 2017 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-28153049

RESUMEN

BACKGROUND: Glioblastoma multiforme (GBM) constitutes nearly half of all malignant brain tumors and has a median survival of 15 months. The standard treatment for these lesions includes maximal resection, radiotherapy, and chemotherapy; however, individual tumors display immense variability in their response to these approaches. Genomic techniques such as whole-exome sequencing (WES) provide an opportunity to understand the molecular basis of this variability. METHODS: Here, we report WES-guided treatment of a patient with a primary GBM and two subsequent recurrences, demonstrating the dynamic nature of treatment-induced molecular changes and their implications for clinical decision-making. We also analyze the Yale-Glioma cohort, composed of 110 whole exome- or whole genome-sequenced tumor-normal pairs, to assess the frequency of genomic events found in the presented case. RESULTS: Our longitudinal analysis revealed how the genomic profile evolved under the pressure of therapy. Specifically targeted approaches eradicated treatment-sensitive clones while enriching for resistant ones, generated due to chromothripsis, which we show to be a frequent event in GBMs based on our extended analysis of 110 gliomas in the Yale-Glioma cohort. Despite chromothripsis and the later acquired mismatch-repair deficiency, genomics-guided personalized treatment extended survival to over 5 years. Interestingly, the case displayed a favorable response to immune checkpoint inhibition after acquiring mismatch repair deficiency. CONCLUSIONS: Our study demonstrates the importance of longitudinal genomic profiling to adjust to the dynamic nature of treatment-induced molecular changes to improve the outcomes of precision therapies.


Asunto(s)
Aberraciones Cromosómicas , Genómica , Glioblastoma/terapia , Recurrencia Local de Neoplasia , Medicina de Precisión , Antineoplásicos/uso terapéutico , Terapia Combinada , Reparación de la Incompatibilidad de ADN , Análisis Mutacional de ADN , ADN de Neoplasias , Progresión de la Enfermedad , Exoma , Femenino , Cirugía General , Genoma Humano , Glioblastoma/genética , Glioblastoma/patología , Humanos , Inmunoterapia , Estudios Longitudinales , Persona de Mediana Edad , Mutación , Radioterapia , Resultado del Tratamiento
13.
Nat Commun ; 8: 14433, 2017 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-28195122

RESUMEN

Meningiomas are mostly benign brain tumours, with a potential for becoming atypical or malignant. On the basis of comprehensive genomic, transcriptomic and epigenomic analyses, we compared benign meningiomas to atypical ones. Here, we show that the majority of primary (de novo) atypical meningiomas display loss of NF2, which co-occurs either with genomic instability or recurrent SMARCB1 mutations. These tumours harbour increased H3K27me3 signal and a hypermethylated phenotype, mainly occupying the polycomb repressive complex 2 (PRC2) binding sites in human embryonic stem cells, thereby phenocopying a more primitive cellular state. Consistent with this observation, atypical meningiomas exhibit upregulation of EZH2, the catalytic subunit of the PRC2 complex, as well as the E2F2 and FOXM1 transcriptional networks. Importantly, these primary atypical meningiomas do not harbour TERT promoter mutations, which have been reported in atypical tumours that progressed from benign ones. Our results establish the genomic landscape of primary atypical meningiomas and potential therapeutic targets.


Asunto(s)
Redes Reguladoras de Genes/genética , Redes Reguladoras de Genes/fisiología , Genoma , Genómica/métodos , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/metabolismo , Meningioma/genética , Meningioma/metabolismo , Sitios de Unión , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Transformación Celular Neoplásica/genética , Inestabilidad Cromosómica , Análisis por Conglomerados , Metilación de ADN , Factor de Transcripción E2F2/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Epigenómica/métodos , Exoma/genética , Proteína Forkhead Box M1/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Genes de la Neurofibromatosis 2 , Técnicas de Genotipaje , Células Madre Embrionarias Humanas/metabolismo , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Técnicas de Sonda Molecular , Mutación , Fenotipo , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Proteína SMARCB1/genética , Análisis de Secuencia , Transducción de Señal/genética , Transcriptoma
14.
Nat Genet ; 48(1): 59-66, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26618343

RESUMEN

Gliomas represent approximately 30% of all central nervous system tumors and 80% of malignant brain tumors. To understand the molecular mechanisms underlying the malignant progression of low-grade gliomas with mutations in IDH1 (encoding isocitrate dehydrogenase 1), we studied paired tumor samples from 41 patients, comparing higher-grade, progressed samples to their lower-grade counterparts. Integrated genomic analyses, including whole-exome sequencing and copy number, gene expression and DNA methylation profiling, demonstrated nonlinear clonal expansion of the original tumors and identified oncogenic pathways driving progression. These include activation of the MYC and RTK-RAS-PI3K pathways and upregulation of the FOXM1- and E2F2-mediated cell cycle transitions, as well as epigenetic silencing of developmental transcription factor genes bound by Polycomb repressive complex 2 in human embryonic stem cells. Our results not only provide mechanistic insight into the genetic and epigenetic mechanisms driving glioma progression but also identify inhibition of the bromodomain and extraterminal (BET) family as a potential therapeutic approach.


Asunto(s)
Neoplasias del Sistema Nervioso Central/genética , Glioma/genética , Isocitrato Deshidrogenasa/genética , Mutación , Neoplasias del Sistema Nervioso Central/patología , Metilación de ADN , Células Madre Embrionarias/metabolismo , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Dosificación de Gen , Regulación Neoplásica de la Expresión Génica , Genes myc , Glioma/patología , Humanos , Isocitrato Deshidrogenasa/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo
15.
Nat Genet ; 48(10): 1253-9, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27548314

RESUMEN

RNA polymerase II mediates the transcription of all protein-coding genes in eukaryotic cells, a process that is fundamental to life. Genomic mutations altering this enzyme have not previously been linked to any pathology in humans, which is a testament to its indispensable role in cell biology. On the basis of a combination of next-generation genomic analyses of 775 meningiomas, we report that recurrent somatic p.Gln403Lys or p.Leu438_His439del mutations in POLR2A, which encodes the catalytic subunit of RNA polymerase II (ref. 1), hijack this essential enzyme and drive neoplasia. POLR2A mutant tumors show dysregulation of key meningeal identity genes, including WNT6 and ZIC1/ZIC4. In addition to mutations in POLR2A, NF2, SMARCB1, TRAF7, KLF4, AKT1, PIK3CA, and SMO, we also report somatic mutations in AKT3, PIK3R1, PRKAR1A, and SUFU in meningiomas. Our results identify a role for essential transcriptional machinery in driving tumorigenesis and define mutually exclusive meningioma subgroups with distinct clinical and pathological features.


Asunto(s)
Neoplasias Meníngeas/genética , Meningioma/genética , Mutación , ARN Polimerasa II/genética , Dominio Catalítico/genética , Cromosomas Humanos Par 22 , Estudios de Cohortes , Análisis Mutacional de ADN , Elementos de Facilitación Genéticos , Exoma , Regulación Neoplásica de la Expresión Génica , Genotipo , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Neoplasias Meníngeas/clasificación , Meningioma/clasificación , Neurofibromina 2/genética , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/genética
16.
J Neurosurg ; : 1-2, 2020 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-32330899
17.
Science ; 339(6123): 1077-80, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23348505

RESUMEN

We report genomic analysis of 300 meningiomas, the most common primary brain tumors, leading to the discovery of mutations in TRAF7, a proapoptotic E3 ubiquitin ligase, in nearly one-fourth of all meningiomas. Mutations in TRAF7 commonly occurred with a recurrent mutation (K409Q) in KLF4, a transcription factor known for its role in inducing pluripotency, or with AKT1(E17K), a mutation known to activate the PI3K pathway. SMO mutations, which activate Hedgehog signaling, were identified in ~5% of non-NF2 mutant meningiomas. These non-NF2 meningiomas were clinically distinctive-nearly always benign, with chromosomal stability, and originating from the medial skull base. In contrast, meningiomas with mutant NF2 and/or chromosome 22 loss were more likely to be atypical, showing genomic instability, and localizing to the cerebral and cerebellar hemispheres. Collectively, these findings identify distinct meningioma subtypes, suggesting avenues for targeted therapeutics.


Asunto(s)
Neoplasias Encefálicas/genética , Factores de Transcripción de Tipo Kruppel/genética , Neoplasias Meníngeas/genética , Meningioma/genética , Proteínas Proto-Oncogénicas c-akt/genética , Receptores Acoplados a Proteínas G/genética , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/genética , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Encefálicas/clasificación , Neoplasias Encefálicas/patología , Cromosomas Humanos Par 22/genética , Análisis Mutacional de ADN , Femenino , Genes de la Neurofibromatosis 2 , Inestabilidad Genómica , Genómica , Humanos , Factor 4 Similar a Kruppel , Masculino , Neoplasias Meníngeas/clasificación , Neoplasias Meníngeas/patología , Meningioma/clasificación , Meningioma/patología , Persona de Mediana Edad , Mutación , Clasificación del Tumor , Receptor Smoothened
18.
Thyroid ; 22(4): 400-6, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22181336

RESUMEN

BACKGROUND: Well-differentiated thyroid cancer arising in struma ovarii is rare. The optimal management of this entity remains undefined. Unilateral cystectomy, unilateral salpingo-oophorectomy (USO), or total abdominal hysterectomy with bilateral salpingo-oophorectomy (TAH/BSO), in addition to total thyroidectomy and radioactive iodine (RAI) ablation, have been employed by various groups. We hypothesized that in patients with thyroid cancer arising within struma ovarii, pelvic surgery alone would be sufficient, provided there is no evidence of gross extra-ovarian extension. METHODS: We review a series of four patients from a single institution and 53 cases from the literature, comparing the extent of treatment and outcomes. Our literature review focused on low-risk patients with struma ovarii confined to the ovary, without evidence of gross extra-ovarian spread or distant metastases. Cumulative recurrence rate was determined by using the Kaplan-Meier method. RESULTS: We report the treatment of four patients with well-differentiated thyroid cancer arising within struma ovarii. Patients underwent USO, BSO, or TAH/BSO. One patient underwent prophylactic total thyroidectomy in anticipation of RAI treatment, and was found to have a synchronous papillary thyroid carcinoma. All patients clinically remain without evidence of disease at a median follow-up of 9 (range 0.8-13) years. Treatment strategies in 53 cases from a review of the literature varied. The pooled cumulative recurrence rate of 57 cases with struma ovarii confined to the ovary was 7.5% at 25 years. CONCLUSIONS: Thyroid cancer arising in struma ovarii is rare. Controversy exists regarding the extent of pelvic resection and management of the thyroid gland. In our series of four patients, all patients are alive without evidence of disease, and the 25-year recurrence rate of 57 cases was low (7.5%), despite a variety of approaches to surgical resection and adjuvant treatment. Extensive pelvic surgery and prophylactic total thyroidectomy to facilitate RAI therapy may be reserved for patients with gross extra-ovarian extension or distant metastases.


Asunto(s)
Carcinoma Papilar Folicular/secundario , Carcinoma Papilar Folicular/cirugía , Estruma Ovárico/patología , Estruma Ovárico/cirugía , Neoplasias de la Tiroides/secundario , Neoplasias de la Tiroides/cirugía , Adulto , Carcinoma Papilar Folicular/patología , Femenino , Humanos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Quistes Ováricos/patología , Ovariectomía , Pelvis/cirugía , Análisis de Supervivencia , Neoplasias de la Tiroides/patología , Tiroidectomía , Resultado del Tratamiento
19.
Development ; 135(23): 3923-33, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18987029

RESUMEN

Alternative splicing is the primary mechanism by which a limited number of protein-coding genes can generate proteome diversity. We have investigated the role of the alternative-splicing factor Sfrs1, an arginine/serine-rich (SR) protein family member, during mouse retinal development. Loss of Sfrs1 function during embryonic retinal development had a profound effect, leading to a small retina at birth. In addition, the retina underwent further degeneration in the postnatal period. Loss of Sfrs1 function resulted in the death of retinal neurons that were born during early to mid-embryonic development. Ganglion cells, cone photoreceptors, horizontal cells and amacrine cells were produced and initiated differentiation. However, these neurons subsequently underwent cell death through apoptosis. By contrast, Sfrs1 was not required for the survival of the neurons generated later, including later-born amacrine cells, rod photoreceptors, bipolar cells and Müller glia. Our results highlight the requirement of Sfrs1-mediated alternative splicing for the survival of retinal neurons, with sensitivity defined by the window of time in which the neuron was generated.


Asunto(s)
Empalme Alternativo/genética , Proteínas Nucleares/genética , Proteínas de Unión al ARN/genética , Neuronas Retinianas/citología , Neuronas Retinianas/metabolismo , Animales , Animales Recién Nacidos , Muerte Celular , Proliferación Celular , Supervivencia Celular , Desarrollo Embrionario/genética , Anomalías del Ojo/embriología , Anomalías del Ojo/patología , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Noqueados , Mitosis , Modelos Biológicos , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/metabolismo , Neuronas Retinianas/ultraestructura , Factores de Empalme Serina-Arginina , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA