Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Invest New Drugs ; 39(3): 829-835, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33415580

RESUMEN

Background Treatment of recurrent, unresectable granulosa cell tumor (GCT) of the ovary can be challenging. Given the rarity of the tumor, alternative therapies have been difficult to evaluate in large prospective clinical trials. Currently, to our knowledge, there are no reports of the use of immune checkpoint inhibitors in GCT patients. Here, we present a case series of GCT patients treated with pembrolizumab who were enrolled in a phase II basket trial in advanced, rare solid tumors (ClinicalTrials.gov: NCT02721732). Cases We identified 5 patients with recurrent GCT (4 adult and 1 juvenile type); they had an extensive history of systemic therapy at study enrollment (range, 3-10), with most regimens resulting in less than 12 months of disease control. Pembrolizumab was administered in these patients, as per trial protocol. Although there were no objective responses according to the irRECIST guidelines, 2 patients with adult-type GCT experienced disease control for ≥ 12 months (565 and 453 days). In one, pembrolizumab represented the longest duration of disease control compared to prior lines of systemic therapy (565 days vs. 13 months). In the other, pembrolizumab was the second longest systemic therapy associated with disease control (453 days vs. 22 months) compared to prior lines of therapy. In this patient, pembrolizumab was discontinued following withdrawal of consent. PD-L1 expression was not observed in any baseline tumor samples. Pembrolizumab was well tolerated, with no grade 3 or 4 treatment-related adverse events. Conclusions Although our results do not support the routine use of pembrolizumab monotherapy in unselected GCT patients, some patients with adult-type GCT may derive a clinical benefit, with a low risk of toxicity. Future studies should investigate the role of immunotherapy and predictors of clinical benefit in this patient population.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Tumor de Células de la Granulosa/tratamiento farmacológico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Adulto , Anciano , Anticuerpos Monoclonales Humanizados/efectos adversos , Proteínas de la Ataxia Telangiectasia Mutada/genética , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Femenino , Proteína Forkhead Box L2/genética , Tumor de Células de la Granulosa/genética , Tumor de Células de la Granulosa/inmunología , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Linfocitos Infiltrantes de Tumor/inmunología , Persona de Mediana Edad , Mutación , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/inmunología , Neurofibromina 1/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Resultado del Tratamiento , Proteína p53 Supresora de Tumor/genética , Adulto Joven
2.
FASEB J ; 35(2): e21231, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33428275

RESUMEN

Tumors of the nervous system including glioblastoma multiforme (GBM) are the most frequent and aggressive form of brain tumors; however, little is known about the impact of the circadian timing system on the formation, growth, and treatment of these tumors. We investigated day/night differences in tumor growth after injection of A530 glioma cells isolated from malignant peripheral nerve sheath tumor (MPNSTs) of NPcis (Trp53+/- ; Nf1+/- ) mice. Synchronized A530 cell cultures expressing typical glial markers were injected at the beginning of the day or night into the sciatic nerve zone of C57BL/6 mice subject to a 12:12 hours light/dark (LD) cycle or after being released to constant darkness (DD). Tumors generated in animals injected early at night in the LD cycle or in DD showed higher growth rates than in animals injected diurnally. No differences were found when animals were injected at the same time with cultures synchronized 12 hours apart. Similar experiments performed with B16 melanoma cells showed higher tumor growth rates in animals injected at the beginning of the night compared to those injected in the daytime. A higher tumor growth rate than that in controls was observed when mice were injected with knocked-down clock gene Bmal1 cells. Finally, when we compared day/night administration of different doses of the proteasome inhibitor Bortezomib (0.5-1.5 mg/kg) in tumor-bearing animals, we found that low-dose chemotherapy displayed higher efficacy when administered at night. Results suggest the existence of a precise temporal control of tumor growth and of drug efficacy in which the host state and susceptibility are critical.


Asunto(s)
Neoplasias Encefálicas/patología , Ritmo Circadiano , Glioblastoma/patología , Fotoperiodo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Factores de Transcripción ARNTL/genética , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Bortezomib/administración & dosificación , Bortezomib/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Esquema de Medicación , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Ratones , Ratones Endogámicos C57BL , Neurofibromina 1/genética , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto/normas
3.
PLoS One ; 15(8): e0237097, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32810864

RESUMEN

Neurofibromatosis type 1 (NF1) is a genetic disorder that affects a range of tissue systems, however the associated muscle weakness and fatigability can have a profound impact on quality of life. Prior studies using the limb-specific Nf1 knockout mouse (Nf1Prx1-/-) revealed an accumulation of intramyocellular lipid (IMCL) that could be rescued by a diet supplemented with L-carnitine and enriched for medium-chain fatty acids (MCFAs). In this study we used the Nf1Prx1-/- mouse to model a range of dietary interventions designed to reduce IMCL accumulation, and analyze using other modalities including in situ muscle physiology and lipid mass spectrometry. Histological IMCL accumulation was significantly reduced by a range of treatments including L-carnitine and high MCFAs alone. A low-fat diet did not affect IMCL, but did provide improvements to muscle strength. Supplementation yielded rapid improvements in IMCL within 4 weeks, but were lost once treatment was discontinued. In situ muscle measurements were highly variable in Nf1Prx1-/- mice, attributable to the severe phenotype present in this model, with fusion of the hips and an overall small hind limb muscle size. Lipidome analysis enabled segregation of the normal and modified chow diets, and fatty acid data suggested increased muscle lipolysis with the intervention. Acylcarnitines were also affected, suggestive of a mitochondrial fatty acid oxidation disorder. These data support the theory that NF1 is a lipid storage disease that can be treated by dietary intervention, and encourages future human trials.


Asunto(s)
Metabolismo de los Lípidos , Fuerza Muscular , Músculo Esquelético/metabolismo , Neurofibromatosis 1/dietoterapia , Animales , Carnitina/administración & dosificación , Carnitina/uso terapéutico , Suplementos Dietéticos , Ácidos Grasos/administración & dosificación , Ácidos Grasos/uso terapéutico , Femenino , Ratones , Músculo Esquelético/fisiopatología , Neurofibromatosis 1/genética , Neurofibromina 1/genética
4.
JAMA Ophthalmol ; 137(12): 1444-1448, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31647501

RESUMEN

Importance: Greater understanding of molecular features of conjunctival melanoma (CM) may improve its clinical management. Objective: To evaluate molecular features of CM and application of this information into clinical care. Design, Setting, and Participants: In a prospective case series of CM with integrative exome and transcriptome analysis, 8 patients at an academic ocular oncology setting were evaluated. The study was conducted from November 2015 to March 2018. Interventions/Exposures: Integrative exome and transcriptome analysis of CMs and clinical management of a patient's care by using this information. Main Outcomes and Measures: Molecular characterization of CM and its potential clinical application. Results: In the 8 patients (4 men) included in analysis, 4 subgroups of CM were observed, including the BRAF V600E mutation in 1 tumor, NRAS Q61R mutation in 3 tumors, NF1 mutations (Q1188X, R440X, or M1215K+ S15fs) in 3 tumors, and triple-wild type (triple-WT) in 1 tumor. The triple-WT case had CCND1 amplification and mutation in the CIC gene (Q1508X). Five tumors, including the triple-WT, also harbored mutations in MAPK genes. In addition to the genes linked to mitogen-activated protein kinase and phosphoinositol 3-kinase pathways, those involved in cell cycle and/or survival, ubiquitin-mediated protein degradation, and chromatin remodeling/epigenetic regulation (ATRX being the most frequently mutated: noted in 5 tumors) may play an important role. Other frequently mutated genes included PREX2 (n = 3), APOB (n = 4), and RYR1/2 (n = 4), although their relevance remains to be determined. The mutation burden ranged from 1.1 to 15.6 mutations per megabase (Mut/Mb) and was 3.3 Mut/Mb or less in 3 tumors and more than 10 Mut/Mb in 2 tumors. A patient with a large tumor and BRAF V600E mutation was treated with combined systemic BRAF (dabrafenib) and MEK (trametinib) inhibitors. After 3 months of therapy, her CM responded substantially and the residual tumor was removed by local surgical excision. Conclusions and Relevance: The NRAS Q61R and NF1 mutations were more common than the BRAF V600E mutation in this series. Although small tumors (where incisional biopsy is not indicated) are treated with surgical excision regardless of mutational profile, in large tumors carrying the BRAF V600E mutation, neoadjuvant therapy with combined systemic BRAF and MEK inhibitors followed by local excision may be used as an alternative to exenteration. Integrative omics analysis of CM may be informative and guide clinical management and treatment in selected cases.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Conjuntiva/genética , Exoma/genética , GTP Fosfohidrolasas/genética , Melanoma/genética , Proteínas de la Membrana/genética , Neurofibromina 1/genética , Proteínas Proto-Oncogénicas B-raf/genética , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Conjuntiva/tratamiento farmacológico , Neoplasias de la Conjuntiva/patología , Análisis Mutacional de ADN , Femenino , Perfilación de la Expresión Génica , Humanos , Imidazoles/uso terapéutico , Masculino , Melanoma/tratamiento farmacológico , Melanoma/patología , Oximas/uso terapéutico , Medicina de Precisión , Estudios Prospectivos , Inhibidores de Proteínas Quinasas/uso terapéutico , Piridonas/uso terapéutico , Pirimidinonas/uso terapéutico
5.
Childs Nerv Syst ; 32(10): 1789-97, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27659822

RESUMEN

Hemispheric low-grade gliomas account for the second most common location in pediatric low-grade gliomas (PLGGs) after the cerebellum. The pathological spectrum includes gangliogliomas, dysembryoplastic neuroepithelial tumors (DNETs), diffuse astrocytomas, pilocytic astrocytomas, and pleomorphic xanthoastrocytomas (PXAs), among others. Clinically, hemispheric PLGGs represent a well-recognized cause of intractable epilepsy in children and adolescents. With an excellent long-term outcome, surgery remains the cornerstone and patients with gross total resection typically do not need any further therapies. The recent literature about hemispheric PLGGs was reviewed to provide an up-to-date overview of the molecular and cell biology of these tumors. Hemispheric PLGGs can harbor multiple alterations involving BRAFV600E, FGFR, NTRK, MYB/MYBL1, IDH, and BRAF-KIAA1549 fusions. However, the clinical significance of most of these alterations is still to be defined. The role of RAS/MAPK mutations and other alterations in hemispheric PLGGs is of interest from diagnostic, prognostic, and therapeutic perspectives. Molecular testing for these tumors should be encouraged, since the findings can have an important impact not only in prognosis but also in therapeutic strategies.


Asunto(s)
Neoplasias Encefálicas/genética , Lateralidad Funcional/genética , Genómica/métodos , Glioma/genética , Adolescente , Quinasa de Linfoma Anaplásico , Niño , Proteínas de Unión al ADN , Femenino , Humanos , Isocitrato Deshidrogenasa/genética , Masculino , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Mutación/genética , Neurofibromina 1/genética , Proteínas Nucleares/genética , Proteínas de Transporte Nucleocitoplasmático/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas de Unión al ARN , Proteínas Tirosina Quinasas Receptoras/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Factores de Transcripción , Proteínas ras/genética
6.
Gastrointest Endosc ; 84(6): 950-958.e3, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27118626

RESUMEN

BACKGROUND AND AIMS: In an era of precision medicine, customized genotyping of GI stromal tumors by screening for driver mutations will become the standard of care. The fidelity of genotype concordance between paired cytology smears and surgical pathology specimens is unknown. In patients with either primary or metastatic sporadic disease, we sought to determine the frequency of KIT and PDGFRA pathogenic alterations within such specimens, imatinib sensitivity, and the concordance of pathogenic alterations between paired specimens. METHODS: DNA obtained from cytology smears from 36 patients, 24 of whom had paired surgical pathology specimens, underwent targeted next-generation sequencing by using a custom panel to evaluate somatic mutations within KIT (exon 2, 9, 10, 11, 13, 14, 15, 17, 18) and PDGFRA (exon 12, 14, 15, 18) genes. Patients with KIT and PDGRFA wild-type genes completed the Qiagen Human Comprehensive Cancer GeneRead DNAseq Targeted Array V2. RESULTS: Genotyping revealed KIT and PDGFRA mutations in 68% and 15% of patients. The wild-type population did not harbor mutations in BRAF, RAS family, SDHB, SETD2, or NF1. Imatinib sensitivity based on the oncogenic kinase mutation prevalence was estimated to be 68%. Mutational concordance between paired cytology and surgical pathology specimens was 96%. CONCLUSIONS: Our data have demonstrated the ability to stratify either primary or metastatic gastrointestinal stromal tumors by mutational subtype using a targeted next-generation sequencing 2 gene mutation panel. We highlight the ability to use cytology specimens obtained via minimally invasive techniques as a surrogate to surgical specimens given the high mutational landscape concordance between paired specimens.


Asunto(s)
ADN de Neoplasias/análisis , Neoplasias Gastrointestinales/genética , Tumores del Estroma Gastrointestinal/genética , Técnicas de Genotipaje , Proteínas Proto-Oncogénicas c-kit/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Antineoplásicos/uso terapéutico , Quimioterapia Adyuvante , Análisis Citogenético , Resistencia a Antineoplásicos/genética , Biopsia por Aspiración con Aguja Fina Guiada por Ultrasonido Endoscópico , Neoplasias Gastrointestinales/patología , Neoplasias Gastrointestinales/terapia , Tumores del Estroma Gastrointestinal/patología , Tumores del Estroma Gastrointestinal/terapia , Secuenciación de Nucleótidos de Alto Rendimiento , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Mesilato de Imatinib/uso terapéutico , Neurofibromina 1/genética , Medicina de Precisión , Proteínas Proto-Oncogénicas B-raf/genética , Radiología Intervencionista , Succinato Deshidrogenasa/genética , Tomografía Computarizada por Rayos X , Proteínas ras/genética
7.
Oncotarget ; 7(9): 10547-56, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26859683

RESUMEN

BACKGROUND: Given the modest responses to everolimus, a mTOR inhibitor, in multiple tumor types, there is a pressing need to identify predictive biomarkers for this drug. Using targeted ultra-deep sequencing, we aimed to explore genomic alterations that confer extreme sensitivity to everolimus. RESULTS: We collected formalin-fixed paraffin-embedded tumor/normal pairs from 39 patients (22 with exceptional clinical benefit, 17 with no clinical benefit) who were treated with everolimus across various tumor types (13 gastric cancers, 15 renal cell carcinomas, 2 thyroid cancers, 2 head and neck cancer, and 7 sarcomas). Ion AmpliSeqTM Comprehensive Cancer Panel was used to identify alterations across all exons of 409 target genes. Tumors were sequenced to a median coverage of 552x. Cancer genomes are characterized by 219 somatic single-nucleotide variants (181 missense, 9 nonsense, 7 splice-site) and 22 frameshift insertions/deletions, with a median of 2.1 mutations per Mb (0 to 12.4 mutations per Mb). Overall, genomic alterations with activating effect on mTOR signaling were identified in 10 of 22 (45%) patients with clinical benefit and these include MTOR, TSC1, TSC2, NF1, PIK3CA and PIK3CG mutations. Recurrently mutated genes in chromatin remodeling genes (BAP1; n = 2, 12%) and receptor tyrosine kinase signaling (FGFR4; n = 2, 12%) were noted only in patients without clinical benefit. CONCLUSIONS: Regardless of different cancer types, mTOR-pathway-activating mutations confer sensitivity to everolimus. Targeted sequencing of mTOR pathway genes facilitates identification of potential candidates for mTOR inhibitors.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos/genética , Everolimus/uso terapéutico , Aparato Lagrimal/patología , Neurofibromina 1/genética , Serina-Treonina Quinasas TOR/genética , Proteína p53 Supresora de Tumor/genética , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Renales/tratamiento farmacológico , Fosfatidilinositol 3-Quinasa Clase I , Fosfatidilinositol 3-Quinasa Clase Ib/genética , Femenino , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Neoplasias Renales/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Mutación/genética , Fosfatidilinositol 3-Quinasas/genética , Polimorfismo de Nucleótido Simple/genética , Sarcoma/tratamiento farmacológico , Neoplasias Gástricas/tratamiento farmacológico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Neoplasias de la Tiroides/tratamiento farmacológico , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética , Adulto Joven
8.
PLoS One ; 10(5): e0124178, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25950952

RESUMEN

Glioblastoma (GBM) is an aggressive, malignant brain tumor typically resulting in death of the patient within one year following diagnosis; and those who survive beyond this point usually present with tumor recurrence within two years (5-year survival is 5%). The genetic heterogeneity of GBM has made the molecular characterization of these tumors an area of great interest and has led to identification of molecular subtypes in GBM. The availability of sequencing platforms that are both fast and economical can further the adoption of tumor sequencing in the clinical environment, potentially leading to identification of clinically actionable genetic targets. In this pilot study, comprised of triplet samples of normal blood, primary tumor, and recurrent tumor samples from three patients; we compared the ability of Illumina whole exome sequencing (ExomeSeq) and the Ion AmpliSeq Comprehensive Cancer Panel (CCP) to identify somatic variants in patient-paired primary and recurrent tumor samples. Thirteen genes were found to harbor variants, the majority of which were exclusive to the ExomeSeq data. Surprisingly, only two variants were identified by both platforms and they were located within the PTCH1 and NF1 genes. Although preliminary in nature, this work highlights major differences in variant identification in data generated from the two platforms. Additional studies with larger samples sizes are needed to further explore the differences between these technologies and to enhance our understanding of the clinical utility of panel based platforms in genomic profiling of brain tumors.


Asunto(s)
Neoplasias Encefálicas/genética , Glioblastoma/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Análisis de Secuencia de ADN/métodos , Adulto , Neoplasias Encefálicas/patología , Exoma , Femenino , Predisposición Genética a la Enfermedad , Variación Genética , Glioblastoma/patología , Humanos , Masculino , Persona de Mediana Edad , Neurofibromina 1/genética , Receptores Patched , Receptor Patched-1 , Proyectos Piloto , Receptores de Superficie Celular/genética
9.
Int J Biol Macromol ; 57: 22-5, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23500436

RESUMEN

It was previously reported that an antitumor polysaccharide (PGPW1) was isolated from the root of Panax ginseng. To extend our study, we investigated here the anti-invasive and metastatic effects of PGPW1 on human gastric cancer cell line HGC-27 and tried to determine its possible mechanism of action. Both scratch wound-healing and Transwell assay identified that PGPW1 dose-dependently inhibited migration and invasiveness of HGC-27 cells. Furthermore, results of western blot showed that protein levels of Twist and AKR1C2 were inhibited by PGPW1, whereas an increase of NF1 was observed. Moreover, down-regulation of Twist expression by PGPW1 blocked epithelial-mesenchymal transition (EMT), characterized by a gain of epithelial cell markers, E-cadherin, and loss of the mesenchymal markers, vimentin and N-cadherin, at protein levels. Collectively, we confirmed that PGPW1 decreased migration and invasion of HGC-27 cells by regulation of Twist, AKR1C2, NF1, E-cadherin, vimentin and N-cadherin expression. In conclusion, PGPW1 may serve as a powerful chemopreventive agent against gastric cancer metastasis.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas Nucleares/biosíntesis , Panax/química , Polisacáridos/farmacología , Neoplasias Gástricas/metabolismo , Proteína 1 Relacionada con Twist/biosíntesis , Antígenos CD/biosíntesis , Antígenos CD/genética , Cadherinas/biosíntesis , Cadherinas/genética , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Hidroxiesteroide Deshidrogenasas/biosíntesis , Hidroxiesteroide Deshidrogenasas/genética , Metástasis de la Neoplasia , Neurofibromina 1/biosíntesis , Neurofibromina 1/genética , Proteínas Nucleares/genética , Polisacáridos/química , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Proteína 1 Relacionada con Twist/genética , Vimentina/biosíntesis , Vimentina/genética
10.
PLoS One ; 7(10): e47854, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23094098

RESUMEN

Neurofibromatosis (NF1) represents the most common single gene cause of learning disabilities. NF1 patients have impairments in frontal lobe based cognitive functions such as attention, working memory, and inhibition. Due to its well-characterized genetic etiology, investigations of NF1 may shed light on neural mechanisms underlying such difficulties in the general population or other patient groups. Prior neuroimaging findings indicate global brain volume increases, consistent with neural over-proliferation. However, little is known about alterations in white matter microstructure in NF1. We performed diffusion tensor imaging (DTI) analyses using tract-based spatial statistics (TBSS) in 14 young adult NF1 patients and 12 healthy controls. We also examined brain volumetric measures in the same subjects. Consistent with prior studies, we found significantly increased overall gray and white matter volume in NF1 patients. Relative to healthy controls, NF1 patients showed widespread reductions in white matter integrity across the entire brain as reflected by decreased fractional anisotropy (FA) and significantly increased absolute diffusion (ADC). When radial and axial diffusion were examined we found pronounced differences in radial diffusion in NF1 patients, indicative of either decreased myelination or increased space between axons. Secondary analyses revealed that FA and radial diffusion effects were of greatest magnitude in the frontal lobe. Such alterations of white matter tracts connecting frontal regions could contribute to the observed cognitive deficits. Furthermore, although the cellular basis of these white matter microstructural alterations remains to be determined, our findings of disproportionately increased radial diffusion against a background of increased white matter volume suggest the novel hypothesis that one potential alteration contributing to increased cortical white matter in NF1 may be looser packing of axons, with or without myelination changes. Further, this indicates that axial and radial diffusivity can uniquely contribute as markers of NF1-associated brain pathology in conjunction with the typically investigated measures.


Asunto(s)
Axones/patología , Lóbulo Frontal/patología , Giro del Cíngulo/patología , Neurofibromatosis 1/patología , Tálamo/patología , Adulto , Anisotropía , Estudios de Casos y Controles , Imagen de Difusión Tensora , Femenino , Humanos , Masculino , Neurofibromina 1/genética , Tamaño de los Órganos
11.
Arch Dermatol Res ; 304(4): 325-31, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22120694

RESUMEN

The neurofibromatosis type 1 (NF1) is characterized by specific cutaneous features (neurofibromas, "café-au-lait" spots of the skin) and alterations of several tissue (nervous, vascular) and bone deformities, such as scoliosis, congenital pseudoarthrosis and bone dysplasia of tibia. Moreover, several studies have shown systemic involvement of bone tissue in NF1 patients, leading to reduced bone mass. The aim of our study was to evaluate some bone mineral metabolism parameters before and after calcium and vitamin D supplementation in NF1 patients. We evaluated in 70 NF1 consecutive patients the mineral metabolism and bone mineral density compared with 40 normal subjects. We showed bone alterations in 35% of patients and the increase of bone formation markers, such as bone isoenzyme of alkaline phosphatase (41.2 ± 15.5 vs. 25.6 ± 8.7 UI; P < 0.05, respectively) and osteocalcin (18.1 ± 5.6 vs. 7.6 ± 1.9 ng/ml; P < 0.05) and reduction of circulating levels of (25OH)-vitamin D (21.8 ± 12.3 ng/ml) with an high percentage of hypovitaminosys D (>60%). Moreover, we revealed a significant reduction of bone mass density at spine (L1-L4) (0.935 ± 0.13 vs. 1.110 ± 0.17 g/cm(2); P < 0.001) and femoral neck side (0.765 ± 0.09 vs. 0.839 ± 0.12 g/cm(2); P < 0.02), with high prevalence of osteopenia (44%) and osteoporosis (18%). After 12 months of calcium (1,200 mg/die) and cholecalciferol (800 UI/die) supplementation, we found a significant increase of (25) OH-vitamin D level (21.8 ± 12.3 vs. 35 ± 13 ng/ml; P < 0.01), without changes in bone mass density. In conclusion, NF1 patients may present a mineral bone involvement, with vitamin D deficiency; calcium and vitamin D supplementation is necessary to restore these bone mineral metabolic alterations.


Asunto(s)
Densidad Ósea , Remodelación Ósea , Huesos/metabolismo , Minerales/metabolismo , Neurofibromatosis 1/metabolismo , Adulto , Densidad Ósea/efectos de los fármacos , Huesos/patología , Calcificación Fisiológica/efectos de los fármacos , Calcio/administración & dosificación , Calcio/deficiencia , Suplementos Dietéticos , Femenino , Humanos , Masculino , Neurofibromatosis 1/patología , Neurofibromina 1/genética , Osteoporosis , Vitamina D/administración & dosificación , Deficiencia de Vitamina D/metabolismo
12.
Hum Mol Genet ; 17(19): 2956-66, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18614544

RESUMEN

To study the role of the neurofibromatosis-1 (NF1) gene in mammalian brain development, we recently generated mice in which Nf1 gene inactivation occurs in neuroglial progenitor cells using the brain lipid binding protein (BLBP) promoter. We found that Nf1(BLBP)CKO mice exhibit significantly reduced body weights and anterior pituitary gland sizes. We further demonstrate that the small anterior pituitary size reflects loss of neurofibromin expression in the hypothalamus, leading to reduced growth hormone releasing hormone, pituitary growth hormone (GH) and liver insulin-like growth factor-1 (IGF1) production. Since neurofibromin both negatively regulates Ras activity and positively modulates cAMP levels, we examined the signaling pathway responsible for these abnormalities. While BLBP-mediated expression of an activated Ras molecule did not recapitulate the body weight and hypothalamic/pituitary defects, treatment of Nf1(BLBP)CKO mice with rolipram to increase cAMP levels resulted in a partial restoration of the body weight phenotype. Furthermore, conditional expression of the Ras regulatory GAP domain of neurofibromin also did not rescue the body weight or Igf1 mRNA defects in Nf1(BLBP)CKO mice. Collectively, these data demonstrate a critical role for neurofibromin in hypothalamic-pituitary axis function and provide further insights into the short stature and GH deficits seen in children with NF1.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Hipotálamo/crecimiento & desarrollo , Neurofibromatosis 1/metabolismo , Neurofibromina 1/metabolismo , Hipófisis/crecimiento & desarrollo , Animales , Peso Corporal , Proteína de Unión a los Ácidos Grasos 7 , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Femenino , Hormona del Crecimiento/metabolismo , Humanos , Hipopituitarismo/genética , Hipopituitarismo/metabolismo , Hipotálamo/química , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neurofibromatosis 1/genética , Neurofibromina 1/química , Neurofibromina 1/genética , Tamaño de los Órganos , Hipófisis/química , Hipófisis/metabolismo , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , Transducción de Señal
13.
Cancer Biol Ther ; 5(3): 305-9, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16418572

RESUMEN

There is increasing evidence that more than 70% of cancers including pancreatic, breast and prostate cancers as well as neurofibromatosis (NF) are highly addicted to abnormal activation of the Ser/Thr kinase PAK1 for their growth. So far FK228 is the most potent among the HDAC (histone deacetylase) inhibitors that block the activation of both PAK1 and another kinase AKT, downstream of PI-3 kinase. However, FK228 is still in clinical trials (phase 2) for a variety of cancers (but not for NF as yet), and not available for most cancer/NF patients. Thus, we have been exploring an alternative which is already in the market, and therefore immediately useful for the treatment of those desperate cancer/NF patients. Here we provide the first evidence that extracts of Chinese/ Japanese peppercorns (Zanthoxyli Fructus) from the plant Zanthoxylum piperitum called "Hua Jiao"/"Sansho", block selectively the key kinase PAK1, leading to the downregulation of cyclin D1. Unlike FK228, these extracts do not inhibit AKT activation at the concentrations that block either cancer growth or PAK1 activation. The Chinese pepper extract selectively inhibits the growth of NF1-deficient malignant peripheral nerve sheath tumor (MPNST) cells, without affecting the growth of normal fibroblasts, and suppresses the growth of NF1-deficient human breast cancer (MDA-MB-231) xenograft in mice. Our data suggest that these peppercorn extracts would be potentially useful for the treatment of PAK1-dependent NF such as MPNST, in addition to a variety of PAK1-dependent cancers including breast cancers.


Asunto(s)
Antimitóticos/farmacología , Ciclina D1/metabolismo , Neurofibromatosis 1/tratamiento farmacológico , Extractos Vegetales/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Zanthoxylum/química , Animales , Antimitóticos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Neoplasias de la Vaina del Nervio/patología , Neurofibromatosis 1/metabolismo , Neurofibromina 1/genética , Fitoterapia , Extractos Vegetales/uso terapéutico , Transducción de Señal , Trasplante Heterólogo , Quinasas p21 Activadas
14.
Oncogene ; 21(12): 1812-22, 2002 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-11896613

RESUMEN

The helix-loop-helix protein Id-1 is a dominant negative regulator of basic helix-loop-helix transcription factors, and plays a key role in the control of breast epithelial cell growth, invasion and differentiation. Previous investigations in our laboratory have shown that Id-1 mRNA was constitutively expressed in highly aggressive and invasive human breast cancer cells in comparison to non-transformed or non-aggressive cancerous cells, and that this loss of regulation is mediated by a 2.2-kb region of the human Id-1 promoter. Here we show that a 31 bp sequence within this 2.2-kb promoter, located 200 bp upstream of the initiation of transcription, is responsible for the constitutive expression of Id-1 in metastatic human breast cancer cells. Using gel shift experiments, we identified a high molecular weight complex present only in non-aggressive breast cancer cells cultured in serum-free medium and which appear to be necessary for proper Id-1 repression. In contrast, nuclear extracts from highly aggressive and metastatic cell lines do not contain this large molecular weight complex. Using DNA affinity precipitation assays (DAPA), we show that this complex contains SP-1, NF-1, Rb and HDAC-1 proteins. On the basis of these findings, we propose a mechanism for the loss of regulation of Id-1 promoter in invasive and metastatic human breast cancer cells.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas de Unión al ADN/genética , Histona Desacetilasas/genética , Neurofibromina 1/genética , Podofilino/análogos & derivados , Regiones Promotoras Genéticas/genética , Proteínas Represoras , Proteína de Retinoblastoma/genética , Factores de Transcripción/genética , Sitios de Unión , Western Blotting , Neoplasias de la Mama/metabolismo , Cartilla de ADN/química , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Proteínas de Unión al ADN/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Secuencias Hélice-Asa-Hélice/genética , Histona Desacetilasa 1 , Histona Desacetilasas/metabolismo , Humanos , Proteína 1 Inhibidora de la Diferenciación , Mutación/genética , Invasividad Neoplásica , Neurofibromina 1/metabolismo , Plásmidos , Podofilino/genética , Podofilino/metabolismo , Podofilotoxina/análogos & derivados , Pruebas de Precipitina , Proteína de Retinoblastoma/metabolismo , Factores de Transcripción/metabolismo , Transfección , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA