Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 189
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 140(4): 477-90, 2010 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-20178741

RESUMEN

Current models imply that the FERM domain protein Merlin, encoded by the tumor suppressor NF2, inhibits mitogenic signaling at or near the plasma membrane. Here, we show that the closed, growth-inhibitory form of Merlin accumulates in the nucleus, binds to the E3 ubiquitin ligase CRL4(DCAF1), and suppresses its activity. Depletion of DCAF1 blocks the promitogenic effect of inactivation of Merlin. Conversely, enforced expression of a Merlin-insensitive mutant of DCAF1 counteracts the antimitogenic effect of Merlin. Re-expression of Merlin and silencing of DCAF1 implement a similar, tumor-suppressive program of gene expression. Tumor-derived mutations invariably disrupt Merlin's ability to interact with or inhibit CRL4(DCAF1). Finally, depletion of DCAF1 inhibits the hyperproliferation of Schwannoma cells from NF2 patients and suppresses the oncogenic potential of Merlin-deficient tumor cell lines. We propose that Merlin suppresses tumorigenesis by translocating to the nucleus to inhibit CRL4(DCAF1).


Asunto(s)
Proteínas Portadoras/metabolismo , Genes Supresores de Tumor , Mesotelioma/metabolismo , Neurilemoma/metabolismo , Neurofibromina 2/metabolismo , Transporte Activo de Núcleo Celular , Animales , Proteínas Portadoras/química , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Humanos , Modelos Moleculares , Proteínas Serina-Treonina Quinasas , Ubiquitina-Proteína Ligasas
2.
J Neurooncol ; 167(2): 339-348, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38372904

RESUMEN

PURPOSE: NF2-related schwannomatosis (NF2) is characterized by bilateral vestibular schwannomas (VS) often causing hearing and neurologic deficits, with currently no FDA-approved drug treatment. Pre-clinical studies highlighted the potential of mTORC1 inhibition in delaying schwannoma progression. We conducted a prospective open-label, phase II study of everolimus for progressive VS in NF2 patients and investigated imaging as a potential biomarker predicting effects on growth trajectory. METHODS: The trial enrolled 12 NF2 patients with progressive VS. Participants received oral everolimus daily for 52 weeks. Brain imaging was obtained quarterly. As primary endpoint, radiographic response (RR) was defined as ≥ 20% decrease in target VS volume. Secondary endpoints included other tumors RR, hearing outcomes, drug safety and quality of life (QOL). RESULTS: Eight participants completed the trial and four discontinued the drug early due to significant volumetric VS progression. After 52 weeks of treatment, the median annual VS growth rate decreased from 77.2% at baseline to 29.4%. There was no VS RR and 3 of 8 (37.5%) participants had stable disease. Decreased or unchanged VS volume after 3 months of treatment was predictive of stabilization at 12 months. Seven of eight participants had stable hearing during treatment except one with a decline in word recognition score. Ten of twelve participants reported only minimal changes to their QOL scores. CONCLUSIONS: Volumetric imaging at 3 months can serve as an early biomarker to predict long-term sensitivity to everolimus treatment. Everolimus may represent a safe treatment option to decrease the growth of NF2-related VS in patients who have stable hearing and neurological condition. TRN: NCT01345136 (April 29, 2011).


Asunto(s)
Neurofibromatosis 2 , Neuroma Acústico , Humanos , Biomarcadores , Everolimus , Neurofibromatosis 2/diagnóstico por imagen , Neurofibromatosis 2/tratamiento farmacológico , Neurofibromatosis 2/complicaciones , Neuroma Acústico/diagnóstico por imagen , Neuroma Acústico/tratamiento farmacológico , Neuroma Acústico/etiología , Calidad de Vida , Resultado del Tratamiento
3.
Clin Trials ; 21(1): 51-66, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37937606

RESUMEN

Numerous successful gene-targeted therapies are arising for the treatment of a variety of rare diseases. At the same time, current treatment options for neurofibromatosis 1 and schwannomatosis are limited and do not directly address loss of gene/protein function. In addition, treatments have mostly focused on symptomatic tumors, but have failed to address multisystem involvement in these conditions. Gene-targeted therapies hold promise to address these limitations. However, despite intense interest over decades, multiple preclinical and clinical issues need to be resolved before they become a reality. The optimal approaches to gene-, mRNA-, or protein restoration and to delivery to the appropriate cell types remain elusive. Preclinical models that recapitulate manifestations of neurofibromatosis 1 and schwannomatosis need to be refined. The development of validated assays for measuring neurofibromin and merlin activity in animal and human tissues will be critical for early-stage trials, as will the selection of appropriate patients, based on their individual genotypes and risk/benefit balance. Once the safety of gene-targeted therapy for symptomatic tumors has been established, the possibility of addressing a wide range of symptoms, including non-tumor manifestations, should be explored. As preclinical efforts are underway, it will be essential to educate both clinicians and those affected by neurofibromatosis 1/schwannomatosis about the risks and benefits of gene-targeted therapy for these conditions.


Asunto(s)
Neurilemoma , Neurofibromatosis , Neurofibromatosis 1 , Neurofibromatosis 2 , Neoplasias Cutáneas , Animales , Humanos , Neurofibromatosis 1/genética , Neurofibromatosis 1/terapia , Neurofibromatosis 2/diagnóstico , Neurofibromatosis 2/genética , Neurofibromatosis 2/patología , Neurofibromatosis/genética , Neurofibromatosis/terapia , Neurofibromatosis/diagnóstico , Neurilemoma/genética , Neurilemoma/terapia , Neurilemoma/diagnóstico
4.
Acta Haematol ; 146(5): 408-412, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37231772

RESUMEN

We describe a case of acute myeloid leukemia with NPM1 mutation and disseminated leukemia cutis in a very old patient, who achieved a long-lasting response to the azacitidine/venetoclax combination with molecular complete remission, given the potential value of this rarely observed clinical outcome.


Asunto(s)
Leucemia Mieloide Aguda , Humanos , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Proteínas Nucleares/genética , Azacitidina/uso terapéutico , Mutación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico
5.
Genet Med ; 24(9): 1967-1977, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35674741

RESUMEN

PURPOSE: Neurofibromatosis type 2 (NF2) and schwannomatosis (SWN) are genetically distinct tumor predisposition syndromes with overlapping phenotypes. We sought to update the diagnostic criteria for NF2 and SWN by incorporating recent advances in genetics, ophthalmology, neuropathology, and neuroimaging. METHODS: We used a multistep process, beginning with a Delphi method involving global disease experts and subsequently involving non-neurofibromatosis clinical experts, patients, and foundations/patient advocacy groups. RESULTS: We reached consensus on the minimal clinical and genetic criteria for diagnosing NF2 and SWN. These criteria incorporate mosaic forms of these conditions. In addition, we recommend updated nomenclature for these disorders to emphasize their phenotypic overlap and to minimize misdiagnosis with neurofibromatosis type 1. CONCLUSION: The updated criteria for NF2 and SWN incorporate clinical features and genetic testing, with a focus on using molecular data to differentiate the 2 conditions. It is likely that continued refinement of these new criteria will be necessary as investigators study the diagnostic properties of the revised criteria and identify new genes associated with SWN. In the revised nomenclature, the term "neurofibromatosis 2" has been retired to improve diagnostic specificity.


Asunto(s)
Neurilemoma , Neurofibromatosis , Neurofibromatosis 1 , Neurofibromatosis 2 , Neoplasias Cutáneas , Consenso , Humanos , Neurilemoma/diagnóstico , Neurilemoma/genética , Neurilemoma/patología , Neurofibromatosis/diagnóstico , Neurofibromatosis/genética , Neurofibromatosis 1/genética , Neurofibromatosis 2/diagnóstico , Neurofibromatosis 2/genética , Neoplasias Cutáneas/genética
6.
Int J Cancer ; 147(9): 2564-2577, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32525563

RESUMEN

Merlin is a versatile tumor suppressor protein encoded by the NF2 gene. Several lines of evidence suggest that Merlin exerts its tumor suppressor activity, at least in part, by forming an inhibitory complex with cluster of differentiation 44 (CD44). Consistently, numerous NF2 mutations in cancer patients are predicted to perturb the interaction of Merlin with CD44. We hypothesized that disruption of the Merlin-CD44 complex through loss of Merlin, unleashes putative tumor- or metastasis-promoting functions of CD44. To evaluate the relevance of the Merlin-CD44 interaction in vivo, we compared tumor growth and progression in Cd44-positive and Cd44-negative Nf2-mutant mice. Heterozygous Nf2-mutant mice were prone to developing highly metastatic osteosarcomas. Importantly, while the absence of the Cd44 gene had no effect on the frequency of primary osteosarcoma development, it strongly diminished osteosarcoma metastasis formation in the Nf2-mutant mice. In vitro assays identified transendothelial migration as the most prominent cellular phenotype dependent on CD44. Adhesion to endothelial cells was blocked by interfering with integrin α4ß1 (very late antigen-4, VLA-4) on osteosarcoma cells and CD44 upregulated levels of integrin VLA-4 ß1 subunit. Among other putative functions of CD44, which may contribute to the metastatic behavior, the passage through the endothelial cells also appears to be critical in vivo, as CD44 significantly promoted formation of lung metastasis upon intravenous injection of osteosarcoma cells into immunocompromised mice. Altogether, our results strongly suggest that CD44 plays a metastasis-promoting role in the absence of Merlin.


Asunto(s)
Neoplasias Óseas/genética , Receptores de Hialuranos/metabolismo , Neoplasias Pulmonares/genética , Neurofibromina 2/genética , Osteosarcoma/genética , Animales , Neoplasias Óseas/patología , Huesos/patología , Adhesión Celular/genética , Línea Celular Tumoral/trasplante , Proliferación Celular/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Receptores de Hialuranos/genética , Pulmón/patología , Neoplasias Pulmonares/secundario , Masculino , Ratones , Ratones Noqueados , Osteosarcoma/secundario
7.
Am J Med Genet A ; 179(6): 1098-1106, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30908866

RESUMEN

The neurofibromatoses are inherited, tumor suppressor disorders that are characterized by multiple, benign peripheral nerve sheath tumors and other nervous system tumors. Each disease is associated with a distinct genetic mutation and with a different pathogenesis and clinical course. Neurofibromatosis 1 (NF1) is common and epitomized by multiple neurofibromas with widespread complications. NF2 and schwannomatosis are rare diseases that are typified by multiple schwannomas that are particularly painful in people with schwannomatosis. Since 1985, the Children's Tumor Foundation (formerly the National Neurofibromatosis Foundation) has hosted an international Neurofibromatosis Conference, bringing together international participants who are focused on NF research and clinical care. The 2017 Conference, held in Washington, DC, was among the largest gatherings of NF researchers to date and included presentations from clinicians and basic scientists, highlighting new data regarding the molecular and cellular mechanisms underlying each of these diseases as well as results from clinical studies and clinical trials. This article summarizes the findings presented at the meeting and represents the current state-of-the art for NF research.


Asunto(s)
Neurilemoma/etiología , Neurofibromatosis/etiología , Neurofibromatosis 1/etiología , Neurofibromatosis 2/etiología , Neoplasias Cutáneas/etiología , Animales , Susceptibilidad a Enfermedades , Humanos , Neurilemoma/diagnóstico , Neurilemoma/metabolismo , Neurilemoma/terapia , Neurofibromatosis/diagnóstico , Neurofibromatosis/metabolismo , Neurofibromatosis/terapia , Neurofibromatosis 1/diagnóstico , Neurofibromatosis 1/metabolismo , Neurofibromatosis 1/terapia , Neurofibromatosis 2/diagnóstico , Neurofibromatosis 2/metabolismo , Neurofibromatosis 2/terapia , Neoplasias Cutáneas/diagnóstico , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/terapia
9.
Development ; 142(11): 2026-36, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25953347

RESUMEN

SCHIP1 is a cytoplasmic partner of cortical cytoskeleton ankyrins. The IQCJ-SCHIP1 isoform is a component of axon initial segments and nodes of Ranvier of mature axons in peripheral and central nervous systems, where it associates with membrane complexes comprising cell adhesion molecules. SCHIP1 is also expressed in the mouse developing central nervous system during embryonic stages of active axonogenesis. Here, we identify a new and early role for SCHIP1 during axon development and establishment of the anterior commissure (AC). The AC is composed of axons from the piriform cortex, the anterior olfactory nucleus and the amygdala. Schip1 mutant mice displayed early defects in AC development that might result from impaired axon growth and guidance. In addition, mutant mice presented a reduced thickness of the piriform cortex, which affected projection neurons in layers 2/3 and was likely to result from cell death rather than from impairment of neuron generation or migration. Piriform cortex neurons from E14.5 mutant embryos displayed axon initiation/outgrowth delay and guidance defects in vitro. The sensitivity of growth cones to semaphorin 3F and Eph receptor B2, two repulsive guidance cues crucial for AC development, was increased, providing a possible basis for certain fiber tract alterations. Thus, our results reveal new evidence for the involvement of cortical cytoskeleton-associated proteins in the regulation of axon development and their importance for the formation of neuronal circuits.


Asunto(s)
Comisura Anterior Cerebral/embriología , Comisura Anterior Cerebral/metabolismo , Axones/metabolismo , Proteínas Portadoras/metabolismo , Citoesqueleto/metabolismo , Corteza Piriforme/embriología , Corteza Piriforme/metabolismo , Animales , Muerte Celular , Embrión de Mamíferos/metabolismo , Conos de Crecimiento/metabolismo , Ratones , Ratones Mutantes , Proteínas del Tejido Nervioso/metabolismo , Receptor EphB2/metabolismo
10.
Am J Med Genet A ; 176(5): 1258-1269, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29681099

RESUMEN

Organized and hosted by the Children's Tumor Foundation (CTF), the Neurofibromatosis (NF) conference is the premier annual gathering for clinicians and researchers interested in neurofibromatosis type 1 (NF1), neurofibromatosis type 2 (NF2), and schwannomatosis (SWN). The 2016 edition constituted a blend of clinical and basic aspects of NF research that helped in clarifying different advances in the field. The incorporation of next generation sequencing is changing the way genetic diagnostics is performed for NF and related disorders, providing solutions to problems like genetic heterogeneity, overlapping clinical manifestations, or the presence of mosaicism. The transformation from plexiform neurofibroma (PNF) to malignant peripheral nerve sheath tumor (MPNST) is being clarified, along with new management and treatments for benign and premalignant tumors. Promising new cellular and in vivo models for understanding the musculoskeletal abnormalities in NF1, the development of NF2 or SWN associated schwannomas, and clarifying the cells that give rise to NF1-associated optic pathway glioma were presented. The interaction of neurofibromin and SPRED1 was described comprehensively, providing functional insight that will help in the interpretation of pathogenicity of certain missense variants identified in NF1 and Legius syndrome patients. Novel promising imaging techniques are being developed, as well as new integrative and holistic management models for patients that take into account psychological, social, and biological factors. Importantly, new therapeutic approaches for schwannomas, meningiomas, ependymomas, PNF, and MPNST are being pursued. This report highlights the major advances that were presented at the 2016 CTF NF conference.


Asunto(s)
Neurilemoma/diagnóstico , Neurilemoma/etiología , Neurofibromatosis/diagnóstico , Neurofibromatosis/etiología , Neurofibromatosis 1/diagnóstico , Neurofibromatosis 1/etiología , Neurofibromatosis 2/diagnóstico , Neurofibromatosis 2/etiología , Neoplasias Cutáneas/diagnóstico , Neoplasias Cutáneas/etiología , Animales , Manejo de la Enfermedad , Modelos Animales de Enfermedad , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Técnicas de Diagnóstico Molecular , Neurilemoma/terapia , Neurofibromatosis/terapia , Neurofibromatosis 1/terapia , Neurofibromatosis 2/terapia , Neoplasias Cutáneas/terapia , Investigación Biomédica Traslacional
11.
J Neurooncol ; 137(1): 33-38, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29230670

RESUMEN

In sporadic schwannomas, inactivation of both copies of the NF2 tumor suppressor gene on 22q is common. Constitutional mutations of SMARCB1 are responsible of schwannomatosis, an inherited tumor predisposition syndrome, characterized by the development of multiple schwannomas. We analysed the frequency of copy number changes on chromosome 22 and the mutation of NF2 and SMARCB1 in 26 sporadic schwannomas. We found two spinal schwannomas with an identical somatic missense mutation in SMARCB1 exon 9: p.(Arg377His). Both SMARCB1 mutated schwannomas had LOH of 22q and one of them harbored an inactivating mutation of NF2. The p.(Arg377His) change was not found in a series of 28 vestibular schwannomas. Our data indicate that mutations affecting SMARCB1 play a role in the development or progression of a small subset of spinal schwannomas and that biallelic inactivation of SMARCB1 may cooperate with deficiency of NF2 function in schwannoma tumorigenesis according to the "four-hit/three events" mechanism of tumorigenesis that we demonstrated in schwannomatosis-associated schwannomas.


Asunto(s)
Neurilemoma/genética , Neurofibromina 2/genética , Proteína SMARCB1/genética , Neoplasias de la Columna Vertebral/genética , Adulto , Anciano , Niño , Cromosomas Humanos Par 22/genética , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Mutación , Neuroma Acústico/genética , Adulto Joven
12.
Genes Dev ; 24(16): 1718-30, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20675406

RESUMEN

The molecular signals that control the maintenance and activation of liver stem/progenitor cells are poorly understood, and the role of liver progenitor cells in hepatic tumorigenesis is unclear. We report here that liver-specific deletion of the neurofibromatosis type 2 (Nf2) tumor suppressor gene in the developing or adult mouse specifically yields a dramatic, progressive expansion of progenitor cells throughout the liver without affecting differentiated hepatocytes. All surviving mice eventually developed both cholangiocellular and hepatocellular carcinoma, suggesting that Nf2(-/-) progenitors can be a cell of origin for these tumors. Despite the suggested link between Nf2 and the Hpo/Wts/Yki signaling pathway in Drosophila, and recent studies linking the corresponding Mst/Lats/Yap pathway to mammalian liver tumorigenesis, our molecular studies suggest that Merlin is not a major regulator of YAP in liver progenitors, and that the overproliferation of Nf2(-/-) liver progenitors is instead driven by aberrant epidermal growth factor receptor (EGFR) activity. Indeed, pharmacologic inhibition of EGFR blocks the proliferation of Nf2(-/-) liver progenitors in vitro and in vivo, consistent with recent studies indicating that the Nf2-encoded protein Merlin can control the abundance and signaling of membrane receptors such as EGFR. Together, our findings uncover a critical role for Nf2/Merlin in controlling homeostasis of the liver stem cell niche.


Asunto(s)
Homeostasis/fisiología , Neoplasias Hepáticas/fisiopatología , Hígado/fisiopatología , Neurofibromina 2/genética , Neurofibromina 2/metabolismo , Células Madre , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/fisiopatología , Proteínas de Ciclo Celular , Proliferación Celular , Células Cultivadas , Colangiocarcinoma/genética , Colangiocarcinoma/fisiopatología , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Femenino , Hepatomegalia/genética , Hepatomegalia/fisiopatología , Homeostasis/genética , Hígado/citología , Neoplasias Hepáticas/genética , Masculino , Ratones , Neurofibromatosis 2/genética , Fosfoproteínas/metabolismo , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo , Proteínas Señalizadoras YAP
13.
Am J Med Genet A ; 173(6): 1714-1721, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28436162

RESUMEN

The Annual Children's Tumor Foundation International Neurofibromatosis Meeting is the premier venue for connecting discovery, translational and clinical scientists who are focused on neurofibromatosis types 1 and 2 (NF1 and NF2) and schwannomatosis (SWN). The meeting also features rare tumors such as glioma, meningioma, sarcoma, and neuroblastoma that occur both within these syndromes and spontaneously; associated with somatic mutations in NF1, NF2, and SWN. The meeting addresses both state of the field for current clinical care as well as emerging preclinical models fueling discovery of new therapeutic targets and discovery science initiatives investigating mechanisms of tumorigenesis. Importantly, this conference is a forum for presenting work in progress and bringing together all stakeholders in the scientific community. A highlight of the conference was the involvement of scientists from the pharmaceutical industry who presented growing efforts for rare disease therapeutic development in general and specifically, in pediatric patients with rare tumor syndromes. Another highlight was the focus on new investigators who presented new data about biomarker discovery, tumor pathogenesis, and diagnostic tools for NF1, NF2, and SWN. This report summarizes the themes of the meeting and a synthesis of the scientific discoveries presented at the conference in order to make the larger research community aware of progress in the neurofibromatoses.


Asunto(s)
Neurilemoma/terapia , Neurofibromatosis/terapia , Neurofibromatosis 1/terapia , Neurofibromatosis 2/terapia , Neoplasias Cutáneas/terapia , Niño , Humanos , Neurilemoma/genética , Neurofibromatosis/genética , Neurofibromatosis 1/genética , Neurofibromatosis 2/genética , Pediatría/tendencias , Neoplasias Cutáneas/genética
14.
J Neurooncol ; 135(1): 47-56, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28735458

RESUMEN

Neurofibromatosis type 2 (NF2), a neurogenetic condition manifest by peripheral nerve sheath tumors (PNST) throughout the neuroaxis for which there are no approved therapies. In vitro and in vivo studies presented here examine agents targeting signaling pathways, angiogenesis, and DNA repair mechanisms. In vitro dose response assays demonstrated potent activity of lapatinib and nilotinib against the mouse schwannoma SC4 (Nf2 -/-) cell line. We then examined the efficacy of everolimus, nilotinib, lapatinib, bevacizumab and radiation (RT) as mono- and combination therapies in flank and sciatic nerve in vivo NF2-PNST models. Data were analyzed using generalized linear models, two sample T-tests and paired T-tests, and linear regression models. SC4(Nf2 -/-) cells implanted in the flank or sciatic nerve showed similar rates of growth (p = 0.9748). Lapatinib, nilotinib and RT significantly reduced tumor growth rate versus controls in the in vivo flank model (p = 0.0025, 0.0062, and 0.009, respectively) whereas bevacizumab and everolimus did not. The best performers were tested in the in vivo sciatic nerve model of NF2 associated PNST, where chemoradiation outperformed nilotinib or lapatinib as single agents (nilotinib vs. nilotinib + RT, p = 0.0001; lapatinib versus lapatinib + RT, p < 0.0001) with no observed toxicity. There was no re-growth of tumors even 14 days after treatment was stopped. The combination of either lapatinib or nilotinib with RT resulted in greater delays in tumor growth rate than any modality alone. This data suggest that concurrent low dose RT and targeted therapy may have a role in addressing progressive PNST in patients with NF2.


Asunto(s)
Antineoplásicos/farmacología , Neurilemoma/terapia , Neurofibromatosis 2/terapia , Neoplasias del Sistema Nervioso Periférico/terapia , Pirimidinas/farmacología , Quinazolinas/farmacología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Bevacizumab/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Quimioradioterapia , Everolimus/farmacología , Lapatinib , Ratones , Ratones Desnudos , Ratones Transgénicos , Modelos Estadísticos , Trasplante de Neoplasias , Neurilemoma/patología , Neurofibromatosis 2/patología , Neoplasias del Sistema Nervioso Periférico/patología , Nervio Ciático , Factores de Tiempo
15.
Dev Biol ; 398(1): 97-109, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25433207

RESUMEN

Molecular mechanisms governing the maintenance and proliferation of dorsal root ganglia (DRG) progenitors are largely unknown. Here we reveal that the Hippo pathway regulates the expansion of DRG progenitors and glia during mammalian DRG development. The key effectors of this pathway, transcriptional coactivators Yap and Taz, are expressed in DRG progenitors and glia during DRG development but are at least partially inhibited from activating transcription. Aberrant YAP activation leads to overexpansion of DRG progenitor and glial populations. We further show that the Neurofibromatosis 2 (Nf2) tumor suppressor inhibits Yap during DRG development. Loss of Nf2 leads to similar phenotypes as does YAP hyperactivation, and deleting Yap suppresses these phenotypes. Our study demonstrates that Nf2-Yap signaling plays important roles in controlling the expansion of DRG progenitors and glia during DRG development.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Ganglios Espinales/embriología , Regulación del Desarrollo de la Expresión Génica , Neurofibromina 2/metabolismo , Neuroglía/citología , Fosfoproteínas/metabolismo , Transducción de Señal , Células Madre/citología , Aciltransferasas , Animales , Proteínas de Ciclo Celular , Movimiento Celular , Eliminación de Gen , Perfilación de la Expresión Génica , Genes de la Neurofibromatosis 2 , Genes Supresores de Tumor , Vía de Señalización Hippo , Hibridación in Situ , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación , Neuroglía/metabolismo , Fenotipo , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Tiempo , Factores de Transcripción/metabolismo , Proteínas Señalizadoras YAP
16.
Development ; 140(16): 3323-34, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23863479

RESUMEN

Brain development requires a precise balance between expansion of the neural progenitor pool and the production of postmitotic neurons and glia. Disruption of this equilibrium results in a myriad of structural abnormalities and disorders of the nervous system. The molecular mechanism that restricts neural progenitor expansion is poorly understood. Here we show that the tumor suppressor neurofibromatosis 2 (Nf2; merlin) limits the expansion of neural progenitor cells (NPCs) in the mammalian dorsal telencephalon. Nf2 is localized at the apical region of NPCs. In the absence of Nf2, NPCs of the cortical hem, hippocampal primordium and neocortical primordium overexpand, while production of Cajal-Retzius cells and hippocampal neurons decreases, resulting in severe malformation of the hippocampus in adult mice. We further show that Nf2 functions by inhibiting the Yap/Taz transcriptional coactivators, probably through a mechanism that is distinct from the canonical Hippo pathway. Overexpressing human YAP in NPCs causes a hippocampal malformation phenotype that closely resembles that of Nf2 mutants and, importantly, deleting Yap in the Nf2 mutant background largely restores hippocampal development. Our studies uncover Nf2 as an important inhibitor of neural progenitor expansion and establish Yap/Taz as key downstream effectors of Nf2 during brain development.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células-Madre Neurales/metabolismo , Neurofibromina 2/metabolismo , Fosfoproteínas/metabolismo , Factores de Transcripción/metabolismo , Aciltransferasas , Proteínas Adaptadoras Transductoras de Señales/genética , Agenesia del Cuerpo Calloso/metabolismo , Agenesia del Cuerpo Calloso/patología , Animales , Tipificación del Cuerpo , Proteínas de Ciclo Celular , Núcleo Celular/metabolismo , Polaridad Celular , Femenino , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Hipocampo/embriología , Hipocampo/metabolismo , Hipocampo/patología , Ratones , Ratones Noqueados , Células-Madre Neurales/patología , Neurofibromina 2/genética , Fenotipo , Fosfoproteínas/genética , Embarazo , Factores de Transcripción/genética , Activación Transcripcional , Proteínas Señalizadoras YAP
17.
J Neurooncol ; 129(1): 47-56, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27177628

RESUMEN

Vestibular schwannoma is a benign neoplasm arising from the Schwann cell sheath of the auditory-vestibular nerve. It most commonly affects both sides in the genetic condition Neurofibromatosis type 2, causing progressive high frequency sensorineural hearing loss. Here, we describe a microsurgical technique and stereotactic coordinates for schwannoma cell grafting in the vestibular nerve region that recapitulates local tumor growth in the cerebellopontine angle and inner auditory canal with resulting hearing loss. Tumor growth was monitored by bioluminescence and MRI in vivo imaging, and hearing assessed by auditory brainstem responses. These techniques, by potentially enabling orthotopic grafting of a variety of cell lines will allow studies on the pathogenesis of tumor-related hearing loss and preclinical drug evaluation, including hearing endpoints, for NF2-related and sporadic schwannomas.


Asunto(s)
Modelos Animales de Enfermedad , Pérdida Auditiva/fisiopatología , Neuroma Acústico/fisiopatología , Trasplante Homólogo , Animales , Línea Celular Tumoral , Potenciales Evocados Auditivos del Tronco Encefálico , Pérdida Auditiva/etiología , Ratones , Ratones Endogámicos BALB C , Neurofibromatosis 2/genética , Neuroma Acústico/complicaciones , Nervio Vestibulococlear/cirugía
18.
Development ; 139(10): 1724-33, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22461560

RESUMEN

The WNT pathway plays multiple roles in neural development and is crucial for establishment of the embryonic cerebellum. In addition, WNT pathway mutations are associated with medulloblastoma, the most common malignant brain tumor in children. However, the cell types within the cerebellum that are responsive to WNT signaling remain unknown. Here we investigate the effects of canonical WNT signaling on two important classes of progenitors in the developing cerebellum: multipotent neural stem cells (NSCs) and granule neuron precursors (GNPs). We show that WNT pathway activation in vitro promotes proliferation of NSCs but not GNPs. Moreover, mice that express activated ß-catenin in the cerebellar ventricular zone exhibit increased proliferation of NSCs in that region, whereas expression of the same protein in GNPs impairs proliferation. Although ß-catenin-expressing NSCs proliferate they do not undergo prolonged expansion or neoplastic growth; rather, WNT signaling markedly interferes with their capacity for self-renewal and differentiation. At a molecular level, mutant NSCs exhibit increased expression of c-Myc, which might account for their transient proliferation, but also express high levels of bone morphogenetic proteins and the cyclin-dependent kinase inhibitor p21, which might contribute to their altered self-renewal and differentiation. These studies suggest that the WNT pathway is a potent regulator of cerebellar stem cell growth and differentiation.


Asunto(s)
Cerebelo/citología , Cerebelo/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Vía de Señalización Wnt/fisiología , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proliferación Celular , Células Cultivadas , Cerebelo/embriología , Citometría de Flujo , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Vía de Señalización Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
19.
J Neurooncol ; 122(2): 313-20, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25567352

RESUMEN

Neurofibromatosis type 2 (NF2) is a genetic disorder with bilateral vestibular schwannomas (VS) as the most frequent manifestation. Merlin, the NF2 tumor suppressor, was identified as a negative regulator of mammalian target of rapamycin complex 1. Pre-clinical data in mice showed that mTORC1 inhibition delayed growth of NF2-schwannomas. We conducted a prospective single-institution open-label phase II study to evaluate the effects of everolimus in ten NF2 patients with progressive VS. Drug activity was monitored every 3 months. Everolimus was administered orally for 12 months and, if the decrease in tumor volume was >20 % from baseline, treatment was continued for 12 additional months. Other patients stopped when completed 12 months of everolimus but were allowed to resume treatment when VS volume was >20 % during 1 year follow-up. Nine patients were evaluable. Safety was evaluated using CTCAE 3.0 criteria. After 12 months of everolimus, no reduction in volume ≥20 % was observed. Four patients had progressive disease, and five patients had stable disease with a median annual growth rate decreasing from 67 %/year before treatment to 0.5 %/year during treatment. In these patients, tumor growth resumed within 3-6 months after treatment discontinuation. Everolimus was then reintroduced and VS decreased by a median 6.8 % at 24 months. Time to tumor progression increased threefold from 4.2 months before treatment to > 12 months. Hearing was stable under treatment. The safety of everolimus was manageable. Although the primary endpoint was not reached, further studies are required to confirm the potential for stabilization of everolimus.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de los Nervios Craneales/tratamiento farmacológico , Everolimus/uso terapéutico , Neurilemoma/tratamiento farmacológico , Neurofibromatosis 2/tratamiento farmacológico , Enfermedades del Nervio Vestibulococlear/tratamiento farmacológico , Adolescente , Adulto , Antineoplásicos/efectos adversos , Biomarcadores de Tumor/metabolismo , Neoplasias de los Nervios Craneales/patología , Neoplasias de los Nervios Craneales/fisiopatología , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Everolimus/efectos adversos , Femenino , Estudios de Seguimiento , Humanos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/metabolismo , Neurilemoma/patología , Neurilemoma/fisiopatología , Neurofibromatosis 2/patología , Neurofibromatosis 2/fisiopatología , Estudios Prospectivos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Resultado del Tratamiento , Carga Tumoral , Enfermedades del Nervio Vestibulococlear/patología , Enfermedades del Nervio Vestibulococlear/fisiopatología , Adulto Joven
20.
Support Care Cancer ; 23(1): 5-7, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25341550

RESUMEN

Although major therapeutic advances have led to improved survival for many hematologic malignancies in recent years, survival remains poor for some disease subtypes and a substantial proportion of patients are ultimately destined to die from their disease and/or related complications. Despite this, there is evidence that patients are not always referred to palliative/home care services as often as those with other cancers, although this situation may be improving in some areas. More research is needed, however, to explore reasons for this and identify whether patients may consequently have unmet needs that impact on their quality of life at this time.


Asunto(s)
Necesidades y Demandas de Servicios de Salud , Neoplasias Hematológicas/mortalidad , Neoplasias Hematológicas/terapia , Servicios de Atención de Salud a Domicilio , Cuidados Paliativos al Final de la Vida , Hematología , Humanos , Cuidados Paliativos , Calidad de Vida , Enfermo Terminal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA