Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Acad Med ; 99(5): 477-481, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38266214

RESUMO

ABSTRACT: Artificial intelligence (AI) methods, especially machine learning and natural language processing, are increasingly affecting health professions education (HPE), including the medical school application and selection processes, assessment, and scholarship production. The rise of large language models over the past 18 months, such as ChatGPT, has raised questions about how best to incorporate these methods into HPE. The lack of training in AI among most HPE faculty and scholars poses an important challenge in facilitating such discussions. In this commentary, the authors provide a primer on the AI methods most often used in the practice and scholarship of HPE, discuss the most pressing challenges and opportunities these tools afford, and underscore that these methods should be understood as part of the larger set of statistical tools available.Despite their ability to process huge amounts of data and their high performance completing some tasks, AI methods are only as good as the data on which they are trained. Of particular importance is that these models can perpetuate the biases that are present in those training datasets, and they can be applied in a biased manner by human users. A minimum set of expectations for the application of AI methods in HPE practice and scholarship is discussed in this commentary, including the interpretability of the models developed and the transparency needed into the use and characteristics of such methods.The rise of AI methods is affecting multiple aspects of HPE including raising questions about how best to incorporate these models into HPE practice and scholarship. In this commentary, we provide a primer on the AI methods most often used in HPE and discuss the most pressing challenges and opportunities these tools afford.


Assuntos
Inteligência Artificial , Ocupações em Saúde , Humanos , Ocupações em Saúde/educação , Bolsas de Estudo/métodos , Processamento de Linguagem Natural , Aprendizado de Máquina , Educação Médica/métodos
3.
BMC Med Educ ; 21(1): 108, 2021 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-33596875

RESUMO

The need for subspecialty-trained neurologists is growing in parallel with increasing disease burden. However, despite the immense burden of neurological diseases, like headache and neurodegenerative disorders, recruitment into these subspecialties remains insufficient in the United States. In this manuscript, a group of educators from the American Academy of Neurology's A.B. Baker Section on Neurological Education sought to review and discuss the current landscape of neurology fellowships in the United States, the factors driving fellowship recruitment and the educational barriers. Moreover, suggestions to potentially improve recruitment for under-selected fellowships, which can contribute towards an alignment between neurological education and neurological needs, and future educational scenarios are discussed.


Assuntos
Bolsas de Estudo , Neurologia , Humanos , Estados Unidos
4.
Clin Case Rep ; 8(2): 363-368, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32128190

RESUMO

Hydrocephalus is rare in sarcoidosis, especially as the presenting symptom. Neurosarcoidosis as a cause of unexplained communicating hydrocephalus should be considered in cases of abnormal cerebrospinal fluid (CSF) and negative infectious and tumoral studies.

6.
J Neurosci ; 36(23): 6213-24, 2016 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-27277800

RESUMO

UNLABELLED: Voltage-gated sodium channel (VGSC) ß subunits signal through multiple pathways on multiple time scales. In addition to modulating sodium and potassium currents, ß subunits play nonconducting roles as cell adhesion molecules, which allow them to function in cell-cell communication, neuronal migration, neurite outgrowth, neuronal pathfinding, and axonal fasciculation. Mutations in SCN1B, encoding VGSC ß1 and ß1B, are associated with epilepsy. Autosomal-dominant SCN1B-C121W, the first epilepsy-associated VGSC mutation identified, results in genetic epilepsy with febrile seizures plus (GEFS+). This mutation has been shown to disrupt both the sodium-current-modulatory and cell-adhesive functions of ß1 subunits expressed in heterologous systems. The goal of this study was to compare mice heterozygous for Scn1b-C121W (Scn1b(+/W)) with mice heterozygous for the Scn1b-null allele (Scn1b(+/-)) to determine whether the C121W mutation results in loss-of-function in vivo We found that Scn1b(+/W) mice were more susceptible than Scn1b(+/-) and Scn1b(+/+) mice to hyperthermia-induced convulsions, a model of pediatric febrile seizures. ß1-C121W subunits are expressed at the neuronal cell surface in vivo However, despite this, ß1-C121W polypeptides are incompletely glycosylated and do not associate with VGSC α subunits in the brain. ß1-C121W subcellular localization is restricted to neuronal cell bodies and is not detected at axon initial segments in the cortex or cerebellum or at optic nerve nodes of Ranvier of Scn1b(W/W) mice. These data, together with our previous results showing that ß1-C121W cannot participate in trans-homophilic cell adhesion, lead to the hypothesis that SCN1B-C121W confers a deleterious gain-of-function in human GEFS+ patients. SIGNIFICANCE STATEMENT: The mechanisms underlying genetic epilepsy syndromes are poorly understood. Closing this gap in knowledge is essential to the development of new medicines to treat epilepsy. We have used mouse models to understand the mechanism of a mutation in the sodium channel gene SCN1B linked to genetic epilepsy with febrile seizures plus. We report that sodium channel ß1 subunit proteins encoded by this mutant gene are expressed at the surface of neuronal cell bodies; however, they do not associate with the ion channel complex nor are they transported to areas of the axon that are critical for proper neuronal firing. We conclude that this disease-causing mutation is not simply a loss-of-function, but instead results in a deleterious gain-of-function in the brain.


Assuntos
Epilepsia/genética , Neurônios/fisiologia , Polimorfismo de Nucleotídeo Único/genética , Subunidade beta-1 do Canal de Sódio Disparado por Voltagem/genética , Subunidade beta-1 do Canal de Sódio Disparado por Voltagem/metabolismo , Animais , Animais Recém-Nascidos , Biotinilação , Células Cultivadas , Córtex Cerebral/citologia , Cisteína/genética , Modelos Animais de Doenças , Epilepsia/etiologia , Epilepsia/patologia , Febre/complicações , Regulação da Expressão Gênica no Desenvolvimento/genética , Imunoprecipitação , Camundongos , Camundongos Transgênicos , Estatísticas não Paramétricas , Triptofano/genética
7.
Epilepsy Curr ; 14(5): 286-8, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25346642
8.
Epilepsy Curr ; 14(2): 97-8, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24872791
9.
Ann Neurol ; 74(1): 128-39, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23821540

RESUMO

OBJECTIVE: Neuronal channelopathies cause brain disorders, including epilepsy, migraine, and ataxia. Despite the development of mouse models, pathophysiological mechanisms for these disorders remain uncertain. One particularly devastating channelopathy is Dravet syndrome (DS), a severe childhood epilepsy typically caused by de novo dominant mutations in the SCN1A gene encoding the voltage-gated sodium channel Na(v) 1.1. Heterologous expression of mutant channels suggests loss of function, raising the quandary of how loss of sodium channels underlying action potentials produces hyperexcitability. Mouse model studies suggest that decreased Na(v) 1.1 function in interneurons causes disinhibition. We aim to determine how mutant SCN1A affects human neurons using the induced pluripotent stem cell (iPSC) method to generate patient-specific neurons. METHODS: Here we derive forebrain-like pyramidal- and bipolar-shaped neurons from 2 DS subjects and 3 human controls by iPSC reprogramming of fibroblasts. DS and control iPSC-derived neurons are compared using whole-cell patch clamp recordings. Sodium current density and intrinsic neuronal excitability are examined. RESULTS: Neural progenitors from DS and human control iPSCs display a forebrain identity and differentiate into bipolar- and pyramidal-shaped neurons. DS patient-derived neurons show increased sodium currents in both bipolar- and pyramidal-shaped neurons. Consistent with increased sodium currents, both types of patient-derived neurons show spontaneous bursting and other evidence of hyperexcitability. Sodium channel transcripts are not elevated, consistent with a post-translational mechanism. INTERPRETATION: These data demonstrate that epilepsy patient-specific iPSC-derived neurons are useful for modeling epileptic-like hyperactivity. Our findings reveal a previously unrecognized cell-autonomous epilepsy mechanism potentially underlying DS, and offer a platform for screening new antiepileptic therapies.


Assuntos
Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/patologia , Mutação/genética , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Neurônios/fisiologia , Diferenciação Celular , Células Cultivadas , Criança , Feminino , Fibroblastos/fisiologia , Humanos , Potenciais Pós-Sinápticos Inibidores/genética , Masculino , Potenciais da Membrana , Técnicas de Patch-Clamp
10.
J Neurosci ; 31(41): 14577-91, 2011 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-21994374

RESUMO

Scn1b-null mice have a severe neurological and cardiac phenotype. Human mutations in SCN1B result in epilepsy and cardiac arrhythmia. SCN1B is expressed as two developmentally regulated splice variants, ß1 and ß1B, that are each expressed in brain and heart in rodents and humans. Here, we studied the structure and function of ß1B and investigated a novel human SCN1B epilepsy-related mutation (p.G257R) unique to ß1B. We show that wild-type ß1B is not a transmembrane protein, but a soluble protein expressed predominantly during embryonic development that promotes neurite outgrowth. Association of ß1B with voltage-gated Na+ channels Na(v)1.1 or Na(v)1.3 is not detectable by immunoprecipitation and ß1B does not affect Na(v)1.3 cell surface expression as measured by [(3)H]saxitoxin binding. However, ß1B coexpression results in subtle alteration of Na(v)1.3 currents in transfected cells, suggesting that ß1B may modulate Na+ current in brain. Similar to the previously characterized p.R125C mutation, p.G257R results in intracellular retention of ß1B, generating a functional null allele. In contrast, two other SCN1B mutations associated with epilepsy, p.C121W and p.R85H, are expressed at the cell surface. We propose that ß1B p.G257R may contribute to epilepsy through a mechanism that includes intracellular retention resulting in aberrant neuronal pathfinding.


Assuntos
Epilepsia/genética , Mutação/genética , Canais de Sódio/genética , Canais de Sódio/metabolismo , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Arginina/genética , Biotinilação/métodos , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Cerebelo , Cricetinae , Cricetulus , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Genótipo , Glicina/genética , Humanos , Imunoprecipitação/métodos , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Canal de Sódio Disparado por Voltagem NAV1.3 , Neuritos/metabolismo , Neurônios/citologia , Neurônios/fisiologia , Técnicas de Patch-Clamp , Isoformas de Proteínas/genética , RNA Mensageiro/metabolismo , Saxitoxina/farmacocinética , Canais de Sódio/deficiência , Estatísticas não Paramétricas , Transfecção/métodos , Trítio/farmacocinética , Subunidade beta-1 do Canal de Sódio Disparado por Voltagem
11.
Neurosci Lett ; 486(2): 53-9, 2010 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-20600605

RESUMO

Voltage-gated Na+ channel (VGSC) ß Subunits are not "auxiliary." These multi-functional molecules not only modulate Na+ current (I(Na)), but also function as cell adhesion molecules (CAMs)-playing roles in aggregation, migration, invasion, neurite outgrowth, and axonal fasciculation. ß subunits are integral members of VGSC signaling complexes at nodes of Ranvier, axon initial segments, and cardiac intercalated disks, regulating action potential propagation through critical intermolecular and cell-cell communication events. At least in vitro, many ß subunit cell adhesive functions occur both in the presence and absence of pore-forming VGSC α subunits, and in vivo ß subunits are expressed in excitable as well as non-excitable cells, thus ß subunits may play important functional roles on their own, in the absence of α subunits. VGSC ß1 subunits are essential for life and appear to be especially important during brain development. Mutations in ß subunit genes result in a variety of human neurological and cardiovascular diseases. Moreover, some cancer cells exhibit alterations in ß subunit expression during metastasis. In short, these proteins, originally thought of as merely accessory to α subunits, are critical players in their own right in human health and disease. Here we discuss the role of VGSC ß subunits in the nervous system.


Assuntos
Moléculas de Adesão Celular/fisiologia , Sistema Nervoso/metabolismo , Canais de Sódio/fisiologia , Animais , Axônios/fisiologia , Moléculas de Adesão Celular/genética , Movimento Celular , Epilepsia/genética , Epilepsia/metabolismo , Humanos , Ativação do Canal Iônico , Transtornos Mentais/genética , Transtornos Mentais/metabolismo , Mutação , Degeneração Neural/genética , Degeneração Neural/metabolismo , Sistema Nervoso/embriologia , Sistema Nervoso/crescimento & desenvolvimento , Neuritos/fisiologia , Subunidades Proteicas/genética , Subunidades Proteicas/fisiologia , Canais de Sódio/genética
12.
Circ Res ; 105(6): 523-6, 2009 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-19661460

RESUMO

RATIONALE: Plakophilin-2 (PKP2) is an essential component of the cardiac desmosome. Recent data show that it interacts with other molecules of the intercalated disc. Separate studies show preferential localization of the voltage-gated sodium channel (Na(V)1.5) to this region. OBJECTIVE: To establish the association of PKP2 with sodium channels and its role on action potential propagation. METHODS AND RESULTS: Biochemical, patch clamp, and optical mapping experiments demonstrate that PKP2 associates with Na(V)1.5, and that knockdown of PKP2 expression alters the properties of the sodium current, and the velocity of action potential propagation in cultured cardiomyocytes. CONCLUSIONS: These results emphasize the importance of intermolecular interactions between proteins relevant to mechanical junctions, and those involved in electric synchrony. Possible relevance to the pathogenesis of arrhythmogenic right ventricular cardiomyopathy is discussed.


Assuntos
Potenciais de Ação , Desmossomos/metabolismo , Miócitos Cardíacos/metabolismo , Placofilinas/metabolismo , Canais de Sódio/metabolismo , Animais , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/patologia , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Desmossomos/patologia , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Miócitos Cardíacos/patologia , Canal de Sódio Disparado por Voltagem NAV1.5 , Ratos , Ratos Sprague-Dawley , Disfunção Ventricular/metabolismo , Disfunção Ventricular/patologia
13.
J Neurosci ; 29(34): 10764-78, 2009 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-19710327

RESUMO

Dravet syndrome (also called severe myoclonic epilepsy of infancy) is one of the most severe forms of childhood epilepsy. Most patients have heterozygous mutations in SCN1A, encoding voltage-gated sodium channel Na(v)1.1 alpha subunits. Sodium channels are modulated by beta1 subunits, encoded by SCN1B, a gene also linked to epilepsy. Here we report the first patient with Dravet syndrome associated with a recessive mutation in SCN1B (p.R125C). Biochemical characterization of p.R125C in a heterologous system demonstrated little to no cell surface expression despite normal total cellular expression. This occurred regardless of coexpression of Na(v)1.1 alpha subunits. Because the patient was homozygous for the mutation, these data suggest a functional SCN1B null phenotype. To understand the consequences of the lack of beta1 cell surface expression in vivo, hippocampal slice recordings were performed in Scn1b(-/-) versus Scn1b(+/+) mice. Scn1b(-/-) CA3 neurons fired evoked action potentials with a significantly higher peak voltage and significantly greater amplitude compared with wild type. However, in contrast to the Scn1a(+/-) model of Dravet syndrome, we found no measurable differences in sodium current density in acutely dissociated CA3 hippocampal neurons. Whereas Scn1b(-/-) mice seize spontaneously, the seizure susceptibility of Scn1b(+/-) mice was similar to wild type, suggesting that, like the parents of this patient, one functional SCN1B allele is sufficient for normal control of electrical excitability. We conclude that SCN1B p.R125C is an autosomal recessive cause of Dravet syndrome through functional gene inactivation.


Assuntos
Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/fisiopatologia , Polimorfismo de Nucleotídeo Único/genética , Canais de Sódio/genética , Animais , Arginina/genética , Biofísica , Linhagem Celular Transformada , Cisteína/genética , Análise Mutacional de DNA , Modelos Animais de Doenças , Estimulação Elétrica , Epilepsias Mioclônicas/mortalidade , Feminino , Proteínas de Fluorescência Verde/genética , Hipocampo/patologia , Humanos , Técnicas In Vitro , Lactente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Moleculares , Canal de Sódio Disparado por Voltagem NAV1.1 , Proteínas do Tecido Nervoso/deficiência , Oócitos , Canais de Sódio/deficiência , Temperatura , Transfecção , Gêmeos , Subunidade beta-1 do Canal de Sódio Disparado por Voltagem , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA