Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Small ; 19(52): e2302280, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37649234

RESUMEN

Glioblastoma (GBM) is the most common primary malignant brain cancer in adults with a dismal prognosis. Temozolomide (TMZ) is the first-in-line chemotherapeutic; however, resistance is frequent and multifactorial. While many molecular and genetic factors have been linked to TMZ resistance, the role of the solid tumor morphology and the tumor microenvironment, particularly the blood-brain barrier (BBB), is unknown. Here, the authors investigate these using a complex in vitro model for GBM and its surrounding BBB. The model recapitulates important clinical features such as a dense tumor core with tumor cells that invade along the perivascular space; and a perfusable BBB with a physiological permeability and morphology that is altered in the presence of a tumor spheroid. It is demonstrated that TMZ sensitivity decreases with increasing cancer cell spatial organization, and that the BBB can contribute to TMZ resistance. Proteomic analysis with next-generation low volume sample workflows of these cultured microtissues revealed potential clinically relevant proteins involved in tumor aggressiveness and TMZ resistance, demonstrating the utility of complex in vitro models for interrogating the tumor microenvironment and therapy validation.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Temozolomida/farmacología , Temozolomida/uso terapéutico , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Barrera Hematoencefálica/metabolismo , Microambiente Tumoral , Proteómica , Resistencia a Antineoplásicos/genética , Línea Celular Tumoral , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Pharmacol Res ; 182: 106308, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35714825

RESUMEN

This review describes recent technological advances applied to glioblastoma (GBM), a brain tumor with dismal prognosis. International consortial efforts suggest the presence of molecular subtypes within histologically identical GBM tumors. This emphasizes that future treatment decisions should no longer be made based solely on morphological analyses, but must now take into consideration such molecular and cellular heterogeneity. The use of single-cell technologies has advanced our understanding and assignation of functional subtypes revealing therapeutic vulnerabilities. Our team has developed stratification approaches in the past few years, and we have been able to identify patient cohorts enriched for various signaling pathways. Importantly, our Glioportal brain tumor resource has been established under the National Neuroscience Institute Tissue Bank in 2021. This resource offers preclinical capability to validate working hypotheses established from patient clinical datasets. This review highlights recent developments with the ultimate goal of assigning functional meaning to molecular subtypes, revealing therapeutic vulnerabilities.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Humanos , Terapia Molecular Dirigida , Medicina de Precisión , Pronóstico
3.
Anal Chem ; 93(4): 2377-2384, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33443405

RESUMEN

Recent genomic studies on the glioblastoma (GBM) subtypes (e.g., mesenchymal, proneural, and classical) pave a way for effective clinical treatments of the recurrent brain tumor. However, identification of the GBM subtype is complicated by the intratumoral heterogeneity that results in coexistence of multiple subtypes within the tissue specimen. Here, we present the use of hyperspectral stimulated Raman scattering (SRS) microscopy for rapid, label-free molecular assessment of GBM intratumoral heterogeneity with submicron resolution. We develop a unique label-free Raman imaging diagnostic platform consisting of the spectral focusing hyperspectral SRS imaging of the large-area GBM tissue specimens, SRS images, and spectrum retrieval using the multivariate curve resolution algorithm and subtype classification based on the quadratic support vector machine model for rapid molecular subtyping of GBMs. Both the stain-free SRS histological images and 2D subtype maps can be obtained within 20-30 min which is superior to the days of the conventional single-cell RNA sequencing. While the SRS histology assesses the demyelination status as a new diagnostic feature, the SRS mapping provides a new insight into intratumoral heterogeneity across GBM tissue specimens. We find that the major proportions of the GBM tissues agree with the diagnostic results of the genomic analysis, but nontrivial portions of the remaining SRS image tiles in the specimens are found to belong to other molecular subtypes, implying the substantial degree of GBM heterogeneity. The rapid SRS imaging diagnostic platform developed has shown the ability of unveiling tumor heterogeneity in GBM tissues accurately, which would promote the improvement of the GBM-targeted therapy in near future.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Técnicas Histológicas , Microscopía Óptica no Lineal/métodos , Neoplasias Encefálicas/diagnóstico por imagen , Glioblastoma/diagnóstico por imagen , Humanos , Sensibilidad y Especificidad
4.
Proc Natl Acad Sci U S A ; 115(22): E5086-E5095, 2018 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-29764999

RESUMEN

Competitive BET bromodomain inhibitors (BBIs) targeting BET proteins (BRD2, BRD3, BRD4, and BRDT) show promising preclinical activities against brain cancers. However, the BET protein-dependent glioblastoma (GBM)-promoting transcriptional network remains elusive. Here, with mechanistic exploration of a next-generation chemical degrader of BET proteins (dBET6), we reveal a profound and consistent impact of BET proteins on E2F1- dependent transcriptional program in both differentiated GBM cells and brain tumor-initiating cells. dBET6 treatment drastically reduces BET protein genomic occupancy, RNA-Pol2 activity, and permissive chromatin marks. Subsequently, dBET6 represses the proliferation, self-renewal, and tumorigenic ability of GBM cells. Moreover, dBET6-induced degradation of BET proteins exerts superior antiproliferation effects compared to conventional BBIs and overcomes both intrinsic and acquired resistance to BBIs in GBM cells. Our study reveals crucial functions of BET proteins and provides the rationale and therapeutic merits of targeted degradation of BET proteins in GBM.


Asunto(s)
Antineoplásicos/farmacología , Factor de Transcripción E2F1 , Glioblastoma , Proteínas Serina-Treonina Quinasas , Proteínas de Unión al ARN , Proteínas de Ciclo Celular , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Factor de Transcripción E2F1/antagonistas & inhibidores , Factor de Transcripción E2F1/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Dominios Proteicos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo
5.
BMC Cancer ; 20(1): 79, 2020 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-32005184

RESUMEN

BACKGROUND: Gliomas consist of a heterogeneous group of tumors. This study aimed to report the incidences of O6-methylguanine-DNA-methyltransferase (MGMT) promoter methylation, 1p19q co-deletion, isocitrate dehydrogenase (IDH) gene mutations, and inactivating mutations of alpha-thalassemia/mental retardation syndrome X-linked (ATRX) in high-grade gliomas in an ethnically diverse population. METHODS: Records of patients who underwent surgery for high-grade gliomas from January 2013 to March 2017 at our institution were obtained. The patients' age, gender, ethnicity, Karnofsky Performance Scale (KPS) score, ability to perform activities of daily living (ADLs), tumor location and biomarkers status were recorded. Data were analyzed using chi-square and Mann-Whitney U tests, Kaplan-Meier estimates and log-rank test. RESULTS: 181 patients were selected (56 with grade III gliomas, 125 with grade IV gliomas). In the grade III group, 55% had MGMT promoter methylation, 41% had 1p19q co-deletion, 35% had IDH1 mutation and none had ATRX loss. In the grade IV group, 30% had MGMT promoter methylation, 2% had 1p19q co-deletion, 15% had IDH1 mutation and 8% had ATRX loss. After adjusting for effects of age, surgery and pre-operative ADL statuses, only MGMT promoter methylation was found to be significantly associated with longer overall survival time in grade III (p = 0.024) and IV patients (p = 0.006). CONCLUSIONS: The incidences of MGMT promoter methylation and IDH1 mutation were found to be comparable to globally reported rates, but those of 1p19q co-deletion and ATRX loss seemed to be lower in our cohort. MGMT promoter methylation was associated with increased overall survival in our cohort and might serve as favorable prognostic factor.


Asunto(s)
Biomarcadores de Tumor/genética , Metilación de ADN , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Glioma/cirugía , Isocitrato Deshidrogenasa/genética , Proteínas Supresoras de Tumor/genética , Proteína Nuclear Ligada al Cromosoma X/genética , Actividades Cotidianas , Adulto , Asia Sudoriental/etnología , Cromosomas Humanos Par 1/genética , Epigénesis Genética , Femenino , Glioma/genética , Glioma/mortalidad , Glioma/patología , Humanos , Incidencia , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Mutación , Clasificación del Tumor , Pronóstico , Regiones Promotoras Genéticas , Estudios Retrospectivos , Eliminación de Secuencia , Análisis de Supervivencia
6.
J Neurooncol ; 141(1): 167-182, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30446902

RESUMEN

PURPOSE: This study aims to identify the neuropsychological tests commonly used for assessment in each neurocognitive domain, and quantify the post-operative changes in neurocognitive function in the immediate post-operation and follow-up. METHODS: With the use of the PubMed, a comprehensive search of the English literature was performed following PRISMA (Preferred Reporting Items for Systematic reviews and Meta-Analyses) guidelines. There were 1021 publications identified for screening. Standardized mean differences (SMD) in neuropsychological task performance were calculated both for immediate post-operation (up to 1 week) and follow-up (up to 6 months). RESULTS: Out of 12 studies which met the inclusion criteria, 11 studies were analyzed in this meta-analysis, with a total of 313 patients (age range 18-82, 50% males) with intracranial gliomas (45% high-grade, 55% low-grade). Complex attention, language and executive function were the most frequently tested neurocognitive domains. Surgery had a positive impact in the domains of complex attention, language, learning and memory tasks in the immediate post-operative period and sustained improvement at follow-up. In contrast, surgery was found to negatively impact performance for executive function in the immediate post-operative period with sustained decline in performance in the long term. CONCLUSIONS: This meta-analysis suggests that surgery for glioma confers a benefit for the domains of complex attention, language, learning and memory, while negatively affecting executive function, in the periods immediately after surgery and at 6 months follow-up. In addition, awake surgery seemed to confer a beneficial effect on neurocognitive functions. Future research should attempt to standardize a battery of neuropsychological tests for patients undergoing surgical resection for glioma, perhaps with a particular focus on executive function.


Asunto(s)
Neoplasias Encefálicas/psicología , Neoplasias Encefálicas/cirugía , Glioma/psicología , Glioma/cirugía , Pruebas Neuropsicológicas , Adulto , Anciano , Neoplasias Encefálicas/diagnóstico , Bases de Datos Factuales , Femenino , Estudios de Seguimiento , Glioma/diagnóstico , Humanos , Masculino , Persona de Mediana Edad , Periodo Posoperatorio , Resultado del Tratamiento
7.
Adv Exp Med Biol ; 951: 47-56, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27837553

RESUMEN

The Cancer Genome Atlas effort has generated significant interest in a new paradigm shift in tumor tissue analysis, patient diagnosis and subsequent treatment decision. Findings have highlighted the limitation of sole reliance on histology, which can be confounded by inter-observer variability. Such studies demonstrate that histologically similar grade IV brain tumors can be divided into four molecular subtypes based on gene expression, with each subtype demonstrating unique genomic aberrations and clinical outcome. These advances indicate that curative therapeutic strategies must now take into account the molecular information in tumor tissue, with the goal of identifying molecularly stratified patients that will most likely to receive treatment benefit from targeted therapy. This in turn spares non-responders from chemotherapeutic side effects and financial costs. In advancing clinical stage drug candidates, the banking of brain tumor tissue necessitates the acquisition of not just tumor tissue with clinical history and robust follow-up, but also high quality molecular information such as somatic mutation, transcriptomic and DNA methylation profiles which have been shown to predict patient survival independent of current clinical indicators. Additionally, the derivation of cell lines from such tumor tissue facilitates the development of clinically relevant patient-derived xenograft mouse models that can prospectively reform the tumor for further studies, yet have retrospective clinical history to associate bench and in vivo findings with clinical data. This represents a core capability of Precision Medicine where the focus is on understanding inter- and intra-tumor heterogeneity so as to best tailor therapies that will result in improved treatment outcomes.


Asunto(s)
Bancos de Muestras Biológicas/estadística & datos numéricos , Neoplasias Encefálicas/terapia , Regulación Neoplásica de la Expresión Génica , Glioblastoma/terapia , Proteínas de Neoplasias/genética , Transcriptoma , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Biología Computacional/métodos , Metilación de ADN , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Ratones , Mutación , Clasificación del Tumor , Proteínas de Neoplasias/metabolismo , Medicina de Precisión , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Acta Neurochir Suppl ; 122: 85-91, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27165883

RESUMEN

Previous work has been demonstrated that tracking features describing the dynamic and time-varying patterns in brain monitoring signals provide additional predictive information beyond that derived from static features based on snapshot measurements. To achieve more accurate predictions of outcomes of patients with traumatic brain injury (TBI), we proposed a statistical framework to extract dynamic features from brain monitoring signals based on the framework of Gaussian processes (GPs). GPs provide an explicit probabilistic, nonparametric Bayesian approach to metric regression problems. This not only provides probabilistic predictions, but also gives the ability to cope with missing data and infer model parameters such as those that control the function's shape, noise level and dynamics of the signal. Through experimental evaluation, we have demonstrated that dynamic features extracted from GPs provide additional predictive information in addition to the features based on the pressure reactivity index (PRx). Significant improvements in patient outcome prediction were achieved by combining GP-based and PRx-based dynamic features. In particular, compared with the a baseline PRx-based model, the combined model achieved over 30 % improvement in prediction accuracy and sensitivity and over 20 % improvement in specificity and the area under the receiver operating characteristic curve.


Asunto(s)
Presión Arterial/fisiología , Lesiones Traumáticas del Encéfalo/fisiopatología , Presión Intracraneal/fisiología , Recuperación de la Función , Teorema de Bayes , Lesiones Traumáticas del Encéfalo/mortalidad , Humanos , Modelos Estadísticos , Monitoreo Fisiológico , Distribución Normal , Estado Vegetativo Persistente/epidemiología , Pronóstico , Curva ROC , Análisis de Regresión
9.
Neuro Oncol ; 26(7): 1247-1261, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38366847

RESUMEN

BACKGROUND: Precision treatment of glioblastoma is increasingly focused on molecular subtyping, with the mesenchymal subtype particularly resistant to temozolomide. Here, we aim to develop a targeted therapy for temozolomide resensitization in the mesenchymal subtype. METHODS: We integrated kinomic profiles and kinase inhibitor screens from patient-derived proneural and mesenchymal glioma-propagating cells and public clinical datasets to identify key protein kinases implicated in temozolomide resistance. RNAseq, apoptosis assays, and comet assays were used to examine the role of p38MAPK signaling and adaptive chemoresistance in mesenchymal cells. The efficacy of dual p38MAPK and MEK/ERK inhibition using ralimetinib (selective orally active p38MAPK inhibitor; phase I/II for glioblastoma) and binimetinib (approved MEK1/2 inhibitor for melanoma; phase II for high-grade glioma) in primary and recurrent mesenchymal tumors was evaluated using an intracranial patient-derived tumor xenograft model, focusing on survival analysis. RESULTS: Our transcriptomic-kinomic integrative analysis revealed p38MAPK as the prime target whose gene signature enables patient stratification based on their molecular subtypes and provides prognostic value. Repurposed p38MAPK inhibitors synergize favorably with temozolomide to promote intracellular retention of temozolomide and exacerbate DNA damage. Mesenchymal cells exhibit adaptive chemoresistance to p38MAPK inhibition through a pH-/calcium-mediated MEK/ERK pathway. Dual p38MAPK and MEK inhibition effectively maintain temozolomide sensitivity in primary and recurrent intracranial mesenchymal glioblastoma xenografts. CONCLUSIONS: Temozolomide resistance in mesenchymal glioblastoma is associated with p38MAPK activation. Adaptive chemoresistance in p38MAPK-resistant cells is mediated by MEK/ERK signaling. Adjuvant therapy with dual p38MAPK and MEK inhibition prolongs temozolomide sensitivity, which can be developed into a precision therapy for the mesenchymal subtype.


Asunto(s)
Neoplasias Encefálicas , Resistencia a Antineoplásicos , Glioblastoma , Temozolomida , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Quinasas p38 Activadas por Mitógenos , Temozolomida/farmacología , Humanos , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Glioblastoma/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Ratones , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Antineoplásicos Alquilantes/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Células Tumorales Cultivadas , Proliferación Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos , Pronóstico
10.
J Neural Eng ; 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38986465

RESUMEN

OBJECTIVE: Micro-electrocorticographic (µECoG) arrays are able to record neural activities from the cortical surface, without the need to penetrate the brain parenchyma. Owing in part to small electrode sizes, previous studies have demonstrated that single-unit spikes could be detected from the cortical surface, and likely from Layer I neurons of the neocortex. Here we tested the ability to use µECoG array to decode, in rats, body position during open field navigation, through isolated single-unit activities. Approach: µECoG arrays were chronically implanted onto primary motor cortex (M1) of Wistar rats, and neural recording was performed in awake, behaving rats in an open-field enclosure. The signals were band-pass filtered between 300 to 3000 Hz. Threshold-crossing spikes were identified and sorted into distinct units based on defined criteria including waveform morphology and refractory period. Body positions were derived from video recordings. We used gradient-boosting machine to predict body position based on previous 100 ms of spike data, and correlation analyses to elucidate the relationship between position and spike patterns. Main results: Single-unit spikes could be extracted during chronic recording from µECoG, and spatial position could be decoded from these spikes with a mean absolute error of prediction of 0.135 and 0.090 in the x- and y- dimensions (of a normalized range from 0 to 1), and Pearson's r of 0.607 and 0.571, respectively. Significance: µECoG can detect single-unit activities that likely arise from superficial neurons in the cortex and is a promising alternative to intracortical arrays, with the added benefit of scalability to cover large cortical surface with minimal incremental risks. More studies should be performed in human related to its use as brain-machine interface.

11.
Nat Commun ; 14(1): 4278, 2023 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-37460561

RESUMEN

Current technologies to subtype glioblastoma (GBM), the most lethal brain tumor, require highly invasive brain biopsies. Here, we develop a dedicated analytical platform to achieve direct and multiplexed profiling of circulating RNAs in extracellular vesicles for blood-based GBM characterization. The technology, termed 'enzyme ZIF-8 complexes for regenerative and catalytic digital detection of RNA' (EZ-READ), leverages an RNA-responsive transducer to regeneratively convert and catalytically enhance signals from rare RNA targets. Each transducer comprises hybrid complexes - protein enzymes encapsulated within metal organic frameworks - to configure strong catalytic activity and robust protection. Upon target RNA hybridization, the transducer activates directly to liberate catalytic complexes, in a target-recyclable manner; when partitioned within a microfluidic device, these complexes can individually catalyze strong chemifluorescence reactions for digital RNA quantification. The EZ-READ platform thus enables programmable and reliable RNA detection, across different-sized RNA subtypes (miRNA and mRNA), directly in sample lysates. When clinically evaluated, the EZ-READ platform established composite signatures for accurate blood-based GBM diagnosis and subtyping.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , MicroARNs , Humanos , MicroARNs/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , ARN Mensajero , Hibridación de Ácido Nucleico , Glioblastoma/genética , Glioblastoma/patología
12.
Neuroendocrinology ; 95(4): 305-16, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22286004

RESUMEN

OBJECTIVE: Gonadotropin-inhibitory hormone (GnIH)-3 is a neuropeptide that plays a major role in the regulation of reproduction and feeding in mammals. MATERIALS AND METHODS: We measured endocrine and behavioural parameters of reproduction in sheep, and sexual behaviour in sheep, mice and cynomolgus monkeys. In addition, GnIH gene expression (in situ hybridization) was examined in ewes, and effects of GnIH-3 on food intake and energy expenditure were measured in various species. GnIH-3 was infused (i.v.) into ewes after an i.m. injection of estradiol benzoate to determine whether the peptide blocks the surge in luteinizing hormone (LH) secretion. RESULTS: GnIH gene expression was reduced in the preovulatory period in ewes. Infusion (i.v.) of GnIH-3 blocked the estrogen-induced LH surge (in ewes). Intracerebroventricular infusion had no effect on female or male sexual behaviour in each of the three species, but increased food intake. There were no effects on energy expenditure in sheep or rats. GnIH increased fos protein (immunohistochemistry) was seen in orexigenic neurons (in sheep and rats), but also in anorexigenic neurons (in sheep). CONCLUSIONS: GnIH-3 reduces reproductive hormone levels and increases food intake in mammals without reducing energy expenditure. There is minimal effect on reproductive behaviour. The dual effect on reproduction and feeding suggests that GnIH-3 provides a molecular switch between these two functions. Blockade of the positive feedback effect of estrogen with parenteral infusion indicates that this peptide may have utility as a blocker of reproductive function in mammals.


Asunto(s)
Conducta Alimentaria/fisiología , Glicoproteínas/fisiología , Hormonas Hipotalámicas/fisiología , Reproducción , Animales , Evaluación Preclínica de Medicamentos , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/genética , Ingestión de Alimentos/fisiología , Conducta Alimentaria/efectos de los fármacos , Femenino , Genes de Cambio/fisiología , Glicoproteínas/genética , Glicoproteínas/farmacología , Hormonas Hipotalámicas/genética , Hormonas Hipotalámicas/farmacología , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos C57BL , Neuropéptidos/genética , Neuropéptidos/farmacología , Neuropéptidos/fisiología , Ratas , Reproducción/efectos de los fármacos , Reproducción/genética , Conducta Sexual Animal/efectos de los fármacos , Conducta Sexual Animal/fisiología , Ovinos
13.
Acta Neurochir Suppl ; 114: 343-6, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22327720

RESUMEN

BACKGROUND: Statins have been shown to reduce mortality and morbidity in ischemic stroke, subarachnoid hemorrhage, and traumatic brain injuries, but their effect on intracerebral hemorrhage (ICH) remains to be determined. This study aimed to investigate the effect of prior statin use on survival following spontaneous primary intracerebral hemorrhage in a multi-ethnic Asian population. SUBJECTS AND METHODS: A study cohort of patients admitted with spontaneous primary ICH was obtained from our database. There were 1,381 patients who met the inclusion criteria. Multivariate logistic regression was used to identify independent predictors and computed odds ratios for 30-day mortality. Kaplan-Meier and Cox proportional hazard survival analyses were used to examine the effect of prior statin use on survival after ICH. RESULTS: Multivariate logistic regression controlling for baseline characteristics and in-hospital interventions, did not demonstrate any effect of prior statin use (p = 0.781) on mortality. Survival analyses also failed to demonstrate any differences in survival after ICH with prior statin use. Similarly subgroup analyses showed no difference. CONCLUSION: No beneficial effect on survival after ICH of prior statin use could be demonstrated in our large multi-ethnic Asian patient cohort.


Asunto(s)
Hemorragia Cerebral , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Anciano , Pueblo Asiatico , Hemorragia Cerebral/epidemiología , Hemorragia Cerebral/mortalidad , Hemorragia Cerebral/prevención & control , Estudios de Cohortes , Diversidad Cultural , Femenino , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Estadísticas no Paramétricas , Análisis de Supervivencia
14.
Acta Neurochir Suppl ; 114: 51-9, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22327664

RESUMEN

BACKGROUND: Despite the wealth of information carried, periodic brain monitoring data are often incomplete with a significant amount of missing values. Incomplete monitoring data are usually discarded to ensure purity of data. However, this approach leads to the loss of statistical power, potentially biased study and a great waste of resources. Thus, we propose to reuse incomplete brain monitoring data by imputing the missing values - a green solution! To support our proposal, we have conducted a feasibility study to investigate the reusability of incomplete brain monitoring data based on the estimated imputation error. MATERIALS AND METHODS: Seventy-seven patients, who underwent invasive monitoring of ICP, MAP, PbtO (2) and brain temperature (BTemp) for more than 24 consecutive hours and were connected to a bedside computerized system, were selected for the study. In the feasibility study, the imputation error is experimentally assessed with simulated missing values and 17 state-of-the-art predictive methods. A framework is developed for neuroclinicians and neurosurgeons to determine the best re-usage strategy and predictive methods based on our feasibility study. RESULTS/CONCLUSION: The monitoring data of MAP and BTemp are more reliable for reuse than ICP and PbtO (2); and, for ICP and PbtO (2) data, a more cautious re-usage strategy should be employed. We also observe that, for the scenarios tested, the lazy learning method, K-STAR, and the tree-based method, M5P, are consistently 2 of the best among the 17 predictive methods investigated in this study.


Asunto(s)
Lesiones Encefálicas/patología , Encéfalo/fisiopatología , Interpretación Estadística de Datos , Presión Intracraneal/fisiología , Monitoreo Fisiológico/métodos , Adulto , Anciano , Sesgo , Presión Sanguínea , Temperatura Corporal/fisiología , Encéfalo/metabolismo , Niño , Estudios de Factibilidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Oxígeno/metabolismo , Estudios Retrospectivos , Máquina de Vectores de Soporte , Adulto Joven
15.
Theranostics ; 11(11): 5127-5142, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33859738

RESUMEN

Hypoxic microenvironment is a hallmark of solid tumors, especially glioblastoma. The strong reliance of glioma-propagating cells (GPCs) on hypoxia-induced survival advantages is potentially exploitable for drug development. Methods: To identify key signaling pathways for hypoxia adaptation by patient-derived GPCs, we performed a kinase inhibitor profiling by screening 188 small molecule inhibitors against 130 different kinases in normoxia and hypoxia. Potential kinase candidates were prioritized for in vitro and in vivo investigations using a ranking algorithm that integrated information from the kinome connectivity network and estimated patients' survival based on expression status. Results: Hypoxic drug screen highlighted extensive modifications of kinomic landscape and a crucial functionality of c-MET-PI3K. c-MET inhibitors diminished phosphorylation of c-MET and PI3K in GPCs subjected to hypoxia, suggesting its role in the hypoxic adaptation of GPCs. Mechanistically, the inhibition of c-MET and PI3K impaired antioxidant defense, leading to oxidative catastrophe and apoptosis. Repurposed c-MET inhibitors PF04217903 and tivantinib exhibited hypoxic-dependent drug synergism with temozolomide, resulting in reduced tumor load and growth of GPC xenografts. Detailed analysis of bulk and single-cell glioblastoma transcriptomes associates the cellular subpopulation over-expressing c-MET with inflamed, hypoxic, metastatic, and stem-like phenotypes. Conclusions: Thus, our "bench to bedside (the use of patient-derived GPCs and xenografts for basic research) and back (validation with independent glioblastoma transcriptome databases)" analysis unravels the novel therapeutic indications of c-MET and PI3K/Akt inhibitors for the treatment of glioblastoma, and potentially other cancers, in the hypoxic tumor microenvironment.


Asunto(s)
Glioma/genética , Hipoxia/genética , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-met/genética , Animales , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioma/tratamiento farmacológico , Humanos , Hipoxia/tratamiento farmacológico , Masculino , Ratones , Fosforilación/efectos de los fármacos , Fosforilación/genética , Pirazinas/farmacología , Pirrolidinonas/farmacología , Quinolinas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Temozolomida/farmacología , Transcriptoma/efectos de los fármacos , Transcriptoma/genética , Triazoles/farmacología
16.
J Crit Care ; 64: 255-261, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34051579

RESUMEN

BACKGROUND/OBJECTIVES: The pharmacokinetics (PK) of drugs is dramatically altered in critical illness. Augmented renal clearance (ARC), a phenomenon characterized by creatinine clearance (CrCl) greater than 130 ml/min/1.73m2, is commonly described in critically ill patients. Levetiracetam, an antiepileptic drug commonly prescribed for seizure prophylaxis in the neurosurgical ICU, undergoes predominant elimination via the kidneys. Hence, we hypothesize that current dosing practice of intravenous (IV) levetiracetam 500 mg twice daily is inadequate for critically ill patients due to enhanced drug elimination. The objectives of our study were to describe the population PK of levetiractam using a nonparametric approach to design an optimal dosing regimen for critically ill neurosurgical patients. METHODS: This was a prospective, observational, population PK study. Serial blood samples were obtained from neurosurgical ICU patients who received at least one dose of IV levetiracetam. We used uHPLC to analyze these samples and Pmetrics™ software to perform PK analysis. RESULTS: Twenty subjects were included, with a median age of 54 years and CrCl of 104 ml/min. A two-compartmental model with linear elimination adequately described the profile of levetiracetam. Mean clearance (CL) was 3.55 L/h and volume of distribution (V) was 18.8 L. No covariates were included in the final model. Monte Carlo simulations showed a low probability of target attainment (PTA, trough at steady state of ≥6 mg/L) with a standard dose of 500 mg twice daily. A dose of at least 1000 mg twice daily was required to achieve 80% PTA. Two subjects, both with subtherapeutic trough levels, developed early onset seizures. CONCLUSION: Our study examined the population PK of levetiracetam in a critically ill neurosurgical population. We found that this population displayed higher clearance and required higher doses to achieve target levels.


Asunto(s)
Anticonvulsivantes , Enfermedad Crítica , Antibacterianos/uso terapéutico , Humanos , Unidades de Cuidados Intensivos , Levetiracetam , Persona de Mediana Edad , Estudios Prospectivos
17.
Dev Cell ; 56(20): 2841-2855.e8, 2021 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-34559979

RESUMEN

Glioblastoma are heterogeneous tumors composed of highly invasive and highly proliferative clones. Heterogeneity in invasiveness could emerge from discrete biophysical properties linked to specific molecular expression. We identified clones of patient-derived glioma propagating cells that were either highly proliferative or highly invasive and compared their cellular architecture, migratory, and biophysical properties. We discovered that invasiveness was linked to cellular fitness. The most invasive cells were stiffer, developed higher mechanical forces on the substrate, and moved stochastically. The mechano-chemical-induced expression of the formin FMN1 conferred invasive strength that was confirmed in patient samples. Moreover, FMN1 expression was also linked to motility in other cancer and normal cell lines, and its ectopic expression increased fitness parameters. Mechanistically, FMN1 acts from the microtubule lattice and promotes a robust mechanical cohesion, leading to highly invasive motility.


Asunto(s)
Movimiento Celular/fisiología , Forminas/metabolismo , Glioblastoma/metabolismo , Invasividad Neoplásica/patología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proteínas Fetales/metabolismo , Glioblastoma/patología , Humanos , Proteínas de Microfilamentos/metabolismo
18.
Sci Adv ; 7(18)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33931443

RESUMEN

Molecular profiling of the most aggressive brain tumor glioblastoma (GBM) on the basis of gene expression, DNA methylation, and genomic variations advances both cancer research and clinical diagnosis. The enhancer architectures and regulatory circuitries governing tumor-intrinsic transcriptional diversity and subtype identity are still elusive. Here, by mapping H3K27ac deposition, we analyze the active regulatory landscapes across 95 GBM biopsies, 12 normal brain tissues, and 38 cell line counterparts. Analyses of differentially regulated enhancers and super-enhancers uncovered previously unrecognized layers of intertumor heterogeneity. Integrative analysis of variant enhancer loci and transcriptome identified topographies of transcriptional enhancers and core regulatory circuitries in four molecular subtypes of primary tumors: AC1-mesenchymal, AC1-classical, AC2-proneural, and AC3-proneural. Moreover, this study reveals core oncogenic dependency on super-enhancer-driven transcriptional factors, long noncoding RNAs, and druggable targets in GBM. Through profiling of transcriptional enhancers, we provide clinically relevant insights into molecular classification, pathogenesis, and therapeutic intervention of GBM.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Cromatina/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/patología , Humanos
19.
Stem Cells ; 27(1): 29-39, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18845764

RESUMEN

Cancer stem cells have been shown to initiate and sustain tumor growth. In many instances, clinical material is limited, compounded by a lack of methods to preserve such cells at convenient time points. Although brain tumor-initiating cells grown in a spheroid manner have been shown to maintain their integrity through serial transplantation in immune-compromised animals, practically, it is not always possible to have access to animals of suitable ages to continuously maintain these cells. We therefore explored vitrification as a cryopreservation technique for brain tumor-initiating cells. Tumor neurospheres were derived from five patients with glioblastoma multiforme (GBM). Cryopreservation in 90% serum and 10% dimethyl sulfoxide yielded greatest viability and could be explored in future studies. Vitrification yielded cells that maintained self-renewal and multipotentiality properties. Karyotypic analyses confirmed the presence of GBM hallmarks. Upon implantation into NOD/SCID mice, our vitrified cells reformed glioma masses that could be serially transplanted. Transcriptome analysis showed that the vitrified and nonvitrified samples in either the stem-like or differentiated states clustered together, providing evidence that vitrification does not change the genotype of frozen cells. Upon induction of differentiation, the transcriptomes of vitrified cells associated with the original primary tumors, indicating that tumor stem-like cells are a genetically distinct population from the differentiated mass, underscoring the importance of working with the relevant tumor-initiating population. Our results demonstrate that vitrification of brain tumor-initiating cells preserves the biological phenotype and genetic profiles of the cells. This should facilitate the establishment of a repository of tumor-initiating cells for subsequent experimental designs.


Asunto(s)
Criopreservación , Glioblastoma/patología , Neuronas/patología , Esferoides Celulares/patología , Antígeno AC133 , Animales , Antígenos CD/metabolismo , Agregación Celular , Diferenciación Celular , Proliferación Celular , Forma de la Célula , Supervivencia Celular , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glicoproteínas/metabolismo , Humanos , Cariotipificación , Ratones , Ratones SCID , Células Madre Multipotentes/patología , Células Madre Neoplásicas/patología , Péptidos/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Br J Neurosurg ; 24(1): 69-74, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20158356

RESUMEN

OBJECTIVE: To assess the utility of pre-operative 3-dimension (3D) visualisation and surgical planning with the Dextroscope in combination with the use of DEX-Ray-a novel augmented reality surgical navigation platform for resection of meningiomas in the falcine, convexity and parasagittal regions. METHODS AND RESULTS: Magnetic resonance imaging (MRI) and magnetic resonance venogram (MRV) images of the patients were reconstructed in 3D using the Dextroscope workstation. Using a variety of available tools, we were able to view the tumour in various surgical angles and appreciate the intricate relationship of the tumour with respect to the surrounding structures and venous anatomy. Critical draining veins both superficial and deep to the tumour were well visualised. By varying the transparency of the overlying scalp and bone we were able to preoperatively determine the ideal size of our scalp flap and bone window for surgical approach. The Dextroscope enabled us to simulate surgical opening and various trajectories of approach while the DexRay virtual reality navigation system enabled the transfer of the Dextroscope 3D planning data into the operating by displaying it in real-time video-augmented mode which further enhanced the appreciation of the tumour's location in 3D space. Four patients underwent total excision of their meningioma while one patient had near total excision with a small residual remnant left behind at the medial third of the superior sagittal sinus. All 5 patients had good neurological recovery post-operatively. CONCLUSION: The use of the Dextroscope for pre-operative surgical planning allows for appreciation of complex anatomical relationships in 3D. This appreciation is further translated for use during surgical navigation utilizing the DEX-Ray platform that provided us with the superior advantage of allowing fast and accurate surgical resection confidently.


Asunto(s)
Imagen por Resonancia Magnética/instrumentación , Neoplasias Meníngeas/cirugía , Meningioma/cirugía , Cirugía Asistida por Computador/métodos , Adulto , Anciano , Mapeo Encefálico , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética/métodos , Masculino , Neoplasias Meníngeas/diagnóstico , Meningioma/diagnóstico , Persona de Mediana Edad , Cuidados Preoperatorios/métodos , Cirugía Asistida por Computador/instrumentación , Interfaz Usuario-Computador
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA