Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
BMJ Case Rep ; 17(1)2024 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-38191218

RESUMEN

The opioid epidemic has become a significant public health crisis worldwide. With the rise in popularity of fentanyl, opioid overdoses continue to rise at unprecedented rates. Unfortunately, young children have become collateral damage in the face of the opioid epidemic. Accidental exposures and ingestions are the leading cause of opioid overdose in this age group and can result in significant acute complications, long-term sequelae and even death. We present the case of a toddler with accidental fentanyl ingestion who experienced seizures and required intubation for respiratory distress. He was found to have notable diffusion restriction cerebellar changes on MRI and ultimately discharged with normal neurological function. Our case adds to the growing literature of the clinical presentation and neuroimaging features associated with opioid toxicity in young children.


Asunto(s)
Sobredosis de Opiáceos , Masculino , Humanos , Preescolar , Neuroimagen , Progresión de la Enfermedad , Analgésicos Opioides , Fentanilo
3.
Mol Ther Oncolytics ; 20: 265-276, 2021 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-33614910

RESUMEN

The glioma-associated family of transcription factors (GLI) have emerged as a promising therapeutic target for a variety of human cancers. In particular, GLI1 plays a central role as a transcriptional regulator for multiple oncogenic signaling pathways, including the hedgehog (Hh) signaling pathway. We undertook a computational screening approach to identify small molecules that directly bind GLI1 for potential development as inhibitors of GLI-mediated transcription. Through these studies, we identified compound 1, which is an 8-hydroxyquinoline, as a high-affinity binder of GLI1. Compound 1 inhibits GLI1-mediated transcriptional activity in several Hh-dependent cellular models, including a primary model of murine medulloblastoma. We also performed a series of computational analyses to define more clearly the mechanism(s) through which 1 inhibits GLI1 function after binding. Our results strongly suggest that binding of 1 to GLI1 does not prevent GLI1/DNA binding nor disrupt the GLI1/DNA complex, but rather, it induces specific conformational changes in the overall complex that prevent proper GLI function. These results highlight the potential of this compound for further development as an anti-cancer agent that targets GLI1.

4.
Cancer Res ; 80(23): 5393-5407, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33046443

RESUMEN

Medulloblastoma is among the most common malignant brain tumors in children. Recent studies have identified at least four subgroups of the disease that differ in terms of molecular characteristics and patient outcomes. Despite this heterogeneity, most patients with medulloblastoma receive similar therapies, including surgery, radiation, and intensive chemotherapy. Although these treatments prolong survival, many patients still die from the disease and survivors suffer severe long-term side effects from therapy. We hypothesize that each patient with medulloblastoma is sensitive to different therapies and that tailoring therapy based on the molecular and cellular characteristics of patients' tumors will improve outcomes. To test this, we assembled a panel of orthotopic patient-derived xenografts (PDX) and subjected them to DNA sequencing, gene expression profiling, and high-throughput drug screening. Analysis of DNA sequencing revealed that most medulloblastomas do not have actionable mutations that point to effective therapies. In contrast, gene expression and drug response data provided valuable information about potential therapies for every tumor. For example, drug screening demonstrated that actinomycin D, which is used for treatment of sarcoma but rarely for medulloblastoma, was active against PDXs representing Group 3 medulloblastoma, the most aggressive form of the disease. Functional analysis of tumor cells was successfully used in a clinical setting to identify more treatment options than sequencing alone. These studies suggest that it should be possible to move away from a one-size-fits-all approach and begin to treat each patient with therapies that are effective against their specific tumor. SIGNIFICANCE: These findings show that high-throughput drug screening identifies therapies for medulloblastoma that cannot be predicted by genomic or transcriptomic analysis.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Cerebelosas/tratamiento farmacológico , Meduloblastoma/tratamiento farmacológico , Medicina de Precisión/métodos , Animales , Línea Celular Tumoral , Neoplasias Cerebelosas/genética , Niño , Dactinomicina/farmacología , Regulación Neoplásica de la Expresión Génica , Ensayos Analíticos de Alto Rendimiento , Humanos , Masculino , Meduloblastoma/genética , Ratones Endogámicos NOD , Mutación , Polimorfismo de Nucleótido Simple , Secuenciación del Exoma , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cell Rep ; 31(12): 107782, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32579914

RESUMEN

Tumor cells are characterized by unlimited proliferation and perturbed differentiation. Using single-cell RNA sequencing, we demonstrate that tumor cells in medulloblastoma (MB) retain their capacity to differentiate in a similar way as their normal originating cells, cerebellar granule neuron precursors. Once they differentiate, MB cells permanently lose their proliferative capacity and tumorigenic potential. Differentiated MB cells highly express NeuroD1, a helix-loop-helix transcription factor, and forced expression of NeuroD1 promotes the differentiation of MB cells. The expression of NeuroD1 in bulk MB cells is repressed by trimethylation of histone 3 lysine-27 (H3K27me3). Inhibition of the histone lysine methyltransferase EZH2 prevents H3K27 trimethylation, resulting in increased NeuroD1 expression and enhanced differentiation in MB cells, which consequently reduces tumor growth. These studies reveal the mechanisms underlying MB cell differentiation and provide rationales to treat MB (potentially other malignancies) by stimulating tumor cell differentiation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Neoplasias Cerebelosas/patología , Meduloblastoma/patología , Proteínas del Tejido Nervioso/metabolismo , Animales , Carcinogénesis/patología , Línea Celular Tumoral , Proliferación Celular , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Proteínas Hedgehog/metabolismo , Histonas/metabolismo , Humanos , Lisina/metabolismo , Metilación , Ratones , Neuronas/metabolismo , Neuronas/patología , Receptor Patched-1/metabolismo , Transducción de Señal , Análisis de la Célula Individual
6.
Nat Neurosci ; 23(7): 842-853, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32424282

RESUMEN

Many immunotherapies act by enhancing the ability of cytotoxic T cells to kill tumor cells. Killing depends on T cell recognition of antigens presented by class I major histocompatibility complex (MHC-I) proteins on tumor cells. In this study, we showed that medulloblastomas lacking the p53 tumor suppressor do not express surface MHC-I and are therefore resistant to immune rejection. Mechanistically, this is because p53 regulates expression of the peptide transporter Tap1 and the aminopeptidase Erap1, which are required for MHC-I trafficking to the cell surface. In vitro, tumor necrosis factor (TNF) or lymphotoxin-ß receptor agonist can rescue expression of Erap1, Tap1 and MHC-I on p53-mutant tumor cells. In vivo, low doses of TNF prolong survival and synergize with immune checkpoint inhibitors to promote tumor rejection. These studies identified p53 as a key regulator of immune evasion and suggest that TNF could be used to enhance sensitivity of tumors to immunotherapy.


Asunto(s)
Neoplasias Cerebelosas/inmunología , Meduloblastoma/inmunología , Escape del Tumor/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Proteína p53 Supresora de Tumor/inmunología , Animales , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/metabolismo , Meduloblastoma/genética , Meduloblastoma/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Factor de Necrosis Tumoral alfa/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
8.
Cancer Res ; 79(9): 2208-2219, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30885981

RESUMEN

Choroid plexus carcinoma (CPC) is a rare brain tumor that occurs most commonly in very young children and has a dismal prognosis despite intensive therapy. Improved outcomes for patients with CPC depend on a deeper understanding of the mechanisms underlying the disease. Here we developed transgenic models of CPCs by activating the Myc oncogene and deleting the Trp53 tumor suppressor gene in murine neural stem cells or progenitors. Murine CPC resembled their human counterparts at a histologic level, and like the hypodiploid subset of human CPC, exhibited multiple whole-chromosome losses, particularly of chromosomes 8, 12, and 19. Analysis of murine and human CPC gene expression profiles and copy number changes revealed altered expression of genes involved in cell cycle, DNA damage response, and cilium function. High-throughput drug screening identified small molecule inhibitors that decreased the viability of CPC. These models will be valuable tools for understanding the biology of choroid plexus tumors and for testing novel approaches to therapy. SIGNIFICANCE: This study describes new mouse models of choroid plexus carcinoma and uses them to investigate the biology and therapeutic responsiveness of this highly malignant pediatric brain tumor.


Asunto(s)
Carcinoma/patología , Neoplasias del Plexo Coroideo/patología , Células-Madre Neurales/patología , Proteínas Proto-Oncogénicas c-myc/fisiología , Bibliotecas de Moléculas Pequeñas/farmacología , Proteína p53 Supresora de Tumor/fisiología , Animales , Antineoplásicos/farmacología , Carcinoma/tratamiento farmacológico , Carcinoma/genética , Neoplasias del Plexo Coroideo/tratamiento farmacológico , Neoplasias del Plexo Coroideo/genética , Ensayos Analíticos de Alto Rendimiento , Ratones , Ratones Noqueados , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Células Tumorales Cultivadas
9.
J Med Chem ; 62(8): 3873-3885, 2019 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-30896941

RESUMEN

The Food and Drug Administration-approved antifungal agent, itraconazole (ITZ), has been increasingly studied for its novel biological properties. In particular, ITZ inhibits the hedgehog (Hh) signaling pathway and has the potential to serve as an anticancer chemotherapeutic against several Hh-dependent malignancies. We have extended our studies on ITZ analogues as Hh pathway inhibitors through the design, synthesis, and evaluation of novel des-triazole ITZ analogues that incorporate modifications to the triazolone/side chain region of the scaffold. Our overall results suggest that the triazolone/side chain region can be replaced with various functionalities (hydrazine carboxamides and meta-substituted amides) resulting in improved potency when compared to ITZ. Our studies also indicate that the stereochemical orientation of the dioxolane ring is important for both potent Hh pathway inhibition and compound stability. Finally, our studies suggest that the ITZ scaffold can be successfully modified in terms of functionality and stereochemistry to further improve its anti-Hh potency and physicochemical properties.


Asunto(s)
Proteínas Hedgehog/antagonistas & inhibidores , Itraconazol/química , Triazoles/química , Animales , Sitios de Unión , Línea Celular , Proliferación Celular , Diseño de Fármacos , Proteínas Hedgehog/metabolismo , Humanos , Itraconazol/metabolismo , Itraconazol/farmacología , Ratones , Simulación de Dinámica Molecular , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
11.
Nat Commun ; 10(1): 332, 2019 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-30659187

RESUMEN

Drugs that modify the epigenome are powerful tools for treating cancer, but these drugs often have pleiotropic effects, and identifying patients who will benefit from them remains a major clinical challenge. Here we show that medulloblastomas driven by the transcription factor Gfi1 are exquisitely dependent on the enzyme lysine demethylase 1 (Kdm1a/Lsd1). We demonstrate that Lsd1 physically associates with Gfi1, and that these proteins cooperate to inhibit genes involved in neuronal commitment and differentiation. We also show that Lsd1 is essential for Gfi1-mediated transformation: Gfi1 proteins that cannot recruit Lsd1 are unable to drive tumorigenesis, and genetic ablation of Lsd1 markedly impairs tumor growth in vivo. Finally, pharmacological inhibitors of Lsd1 potently inhibit growth of Gfi1-driven tumors. These studies provide important insight into the mechanisms by which Gfi1 contributes to tumorigenesis, and identify Lsd1 inhibitors as promising therapeutic agents for Gfi1-driven medulloblastoma.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Neoplasias Cerebelosas/patología , Proteínas de Unión al ADN/metabolismo , Histona Demetilasas/metabolismo , Meduloblastoma/patología , Factores de Transcripción/metabolismo , Animales , Antibióticos Antineoplásicos/uso terapéutico , Proliferación Celular/efectos de los fármacos , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/terapia , Proteínas de Unión al ADN/genética , Doxorrubicina/uso terapéutico , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Histona Demetilasas/genética , Humanos , Meduloblastoma/genética , Meduloblastoma/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Células 3T3 NIH , Trasplante de Neoplasias , Virus Oncogénicos , Retroviridae , Factores de Transcripción/genética
12.
Eur J Med Chem ; 162: 495-506, 2019 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-30471551

RESUMEN

The Hedgehog (Hh) pathway is a developmental pathway with therapeutic potential as a target for a variety of cancers. In recent years, several vitamin D-based compounds have been identified as potent inhibitors of Hh signaling. These analogues contain aromatic phenol A-ring mimics coupled to the CD-ring side chain of vitamin D3 through modified seco-B regions. To continue structure-activity relationship studies on this class of Hh pathway inhibitors, multiple series of vitamin D-based analogues that contain an amine-based seco-B tether and/or incorporate a hydroxyl moiety on C-25 were designed and synthesized. These compounds were evaluated in multiple cell lines for their anti-Hh activity, and we identify analogues 16, 21, 22 as potent vitamin D-based Hh inhibitors (IC50 values of 110-340 nM). We also performed a series of mechanism of action studies in knockout cell lines to further explore whether these analogues inhibit the Hh pathway through a known Hh pathway component or the vitamin D receptor. While the specific cellular target that mediates these effects remains elusive, our studies suggest multiple cellular targets may mediate the anti-Hh activity of this scaffold.


Asunto(s)
Colecalciferol/síntesis química , Proteínas Hedgehog/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular , Colecalciferol/análogos & derivados , Colecalciferol/farmacología , Técnicas de Inactivación de Genes , Humanos , Concentración 50 Inhibidora , Receptores de Calcitriol/metabolismo , Relación Estructura-Actividad , Proteína con Dedos de Zinc GLI1/metabolismo
13.
Eur J Med Chem ; 163: 320-332, 2019 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-30529635

RESUMEN

Inhibition of the hedgehog (Hh) signaling pathway has been validated as a therapeutic strategy to treat basal cell carcinoma and holds potential for several other forms of human cancer. Itraconazole and posaconazole are clinically useful triazole anti-fungals that are being repurposed as anti-cancer agents based on their ability to inhibit the Hh pathway. We have previously demonstrated that removal of the triazole from itraconazole does not affect its ability to inhibit the Hh pathway while abolishing its primary side effect, potent inhibition of Cyp3A4. To develop structure-activity relationships for the related posaconazole scaffold, we synthesized and evaluated a series of des-triazole analogues designed through both ligand- and structure-based methods. These compounds demonstrated improved anti-Hh properties compared to posaconazole and enhanced stability without inhibiting Cyp3A4. In addition, we utilized a series of molecular dynamics and binding energy studies to probe specific interactions between the compounds and their proposed binding site on Smoothened. These studies strongly suggest that the tetrahydrofuran region of the scaffold projects out of the binding site and that π-π interactions between the compound and Smoothened play a key role in stabilizing the bound analogues.


Asunto(s)
Proteínas Hedgehog/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Triazoles/farmacología , Antifúngicos/uso terapéutico , Carcinoma Basocelular/tratamiento farmacológico , Humanos , Simulación de Dinámica Molecular , Unión Proteica , Receptor Smoothened/metabolismo , Relación Estructura-Actividad , Triazoles/química , Triazoles/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...