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











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 5110, 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38877018

RESUMO

Tyrosine kinase (TK) fusions are frequently found in cancers, either as initiating events or as a mechanism of resistance to targeted therapy. Partner genes and exons in most TK fusions are followed typical recurrent patterns, but the underlying mechanisms and clinical implications of these patterns are poorly understood. By developing Functionally Active Chromosomal Translocation Sequencing (FACTS), we discover that typical TK fusions involving ALK, ROS1, RET and NTRK1 are selected from pools of chromosomal rearrangements by two major determinants: active transcription of the fusion partner genes and protein stability. In contrast, atypical TK fusions that are rarely seen in patients showed reduced protein stability, decreased downstream oncogenic signaling, and were less responsive to inhibition. Consistently, patients with atypical TK fusions were associated with a reduced response to TKI therapies. Our findings highlight the principles of oncogenic TK fusion formation and selection in cancers, with clinical implications for guiding targeted therapy.


Assuntos
Neoplasias , Proteínas de Fusão Oncogênica , Proteínas Tirosina Quinases , Proteínas Proto-Oncogênicas c-ret , Translocação Genética , Humanos , Neoplasias/genética , Neoplasias/tratamento farmacológico , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-ret/genética , Proteínas Proto-Oncogênicas c-ret/metabolismo , Quinase do Linfoma Anaplásico/genética , Quinase do Linfoma Anaplásico/metabolismo , Receptor trkA/genética , Receptor trkA/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais/genética , Linhagem Celular Tumoral
2.
Cell ; 186(16): 3522-3522.e1, 2023 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-37541203

RESUMO

Located in each brain ventricle, choroid plexus (ChP) tissue forms a blood-CSF barrier and produces cerebrospinal fluid (CSF) and other supportive factors. Sheets of ChP epithelial cells enclose a vascularized stroma of mesenchymal, immune, and neuron/glia-like cells. Burgeoning ChP studies are revealing its complex set of functions across the lifespan. To view this SnapShot, open or download the PDF.


Assuntos
Encéfalo , Plexo Corióideo , Barreira Hematoencefálica , Longevidade , Células Epiteliais , Líquido Cefalorraquidiano
3.
Neuron ; 111(10): 1591-1608.e4, 2023 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-36893755

RESUMO

Post-hemorrhagic hydrocephalus (PHH) refers to a life-threatening accumulation of cerebrospinal fluid (CSF) that occurs following intraventricular hemorrhage (IVH). An incomplete understanding of this variably progressive condition has hampered the development of new therapies beyond serial neurosurgical interventions. Here, we show a key role for the bidirectional Na-K-Cl cotransporter, NKCC1, in the choroid plexus (ChP) to mitigate PHH. Mimicking IVH with intraventricular blood led to increased CSF [K+] and triggered cytosolic calcium activity in ChP epithelial cells, which was followed by NKCC1 activation. ChP-targeted adeno-associated viral (AAV)-NKCC1 prevented blood-induced ventriculomegaly and led to persistently increased CSF clearance capacity. These data demonstrate that intraventricular blood triggered a trans-choroidal, NKCC1-dependent CSF clearance mechanism. Inactive, phosphodeficient AAV-NKCC1-NT51 failed to mitigate ventriculomegaly. Excessive CSF [K+] fluctuations correlated with permanent shunting outcome in humans following hemorrhagic stroke, suggesting targeted gene therapy as a potential treatment to mitigate intracranial fluid accumulation following hemorrhage.


Assuntos
Plexo Corióideo , Hidrocefalia , Humanos , Hidrocefalia/terapia , Hemorragia Cerebral/complicações , Hemorragia Cerebral/terapia
4.
STAR Protoc ; 4(1): 101975, 2023 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-36580401

RESUMO

Experimentally targeting mouse choroid plexus (ChP) provides a valuable approach for investigating mechanisms of ChP-cerebrospinal fluid (CSF) biology. Here, we provide a protocol to deliver adeno-associated viral vectors (AAVs) by in utero intracerebroventricular (ICV) injection to ChP epithelial cells. We begin by describing steps for induction anesthesia of the pregnant dam, laparotomy, and in utero ICV injection. We also detail post-surgical care and immunoblot validation. For complete details on the use and execution of this protocol, please refer to Jang et al. (2022).1.


Assuntos
Plexo Corióideo , Células Epiteliais , Camundongos , Animais , Feminino , Gravidez
5.
Neuron ; 110(20): 3288-3301.e8, 2022 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-36070751

RESUMO

For many cancer patients, chemotherapy produces untreatable life-long neurologic effects termed chemotherapy-related cognitive impairment (CRCI). We discovered that the chemotherapy methotrexate (MTX) adversely affects oxidative metabolism of non-cancerous choroid plexus (ChP) cells and the cerebrospinal fluid (CSF). We used a ChP-targeted adeno-associated viral (AAV) vector approach in mice to augment CSF levels of the secreted antioxidant SOD3. AAV-SOD3 gene therapy increased oxidative defense capacity of the CSF and prevented MTX-induced lipid peroxidation in the hippocampus. Furthermore, this gene therapy prevented anxiety and deficits in short-term learning and memory caused by MTX. MTX-induced oxidative damage to cultured human cortical neurons and analyses of CSF samples from MTX-treated lymphoma patients demonstrated that MTX diminishes antioxidant capacity of patient CSF. Collectively, our findings motivate the advancement of ChP- and CSF-targeted anti-oxidative prophylactic measures to relieve CRCI.


Assuntos
Antioxidantes , Neoplasias , Humanos , Animais , Camundongos , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Plexo Corióideo , Metotrexato/toxicidade , Estresse Oxidativo , Hipocampo , Neoplasias/induzido quimicamente
6.
Fluids Barriers CNS ; 19(1): 36, 2022 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-35619113

RESUMO

Choroid plexus (ChP) epithelial cells are crucial for the function of the blood-cerebrospinal fluid barrier (BCSFB) in the developing and mature brain. The ChP is considered the primary source and regulator of CSF, secreting many important factors that nourish the brain. It also performs CSF clearance functions including removing Amyloid beta and potassium. As such, the ChP is a promising target for gene and drug therapy for neurodevelopmental and neurological disorders in the central nervous system (CNS). This review describes the current successful and emerging experimental approaches for targeting ChP epithelial cells. We highlight methodological strategies to specifically target these cells for gain or loss of function in vivo. We cover both genetic models and viral gene delivery systems. Additionally, several lines of reporters to access the ChP epithelia are reviewed. Finally, we discuss exciting new approaches, such as chemical activation and transplantation of engineered ChP epithelial cells. We elaborate on fundamental functions of the ChP in secretion and clearance and outline experimental approaches paving the way to clinical applications.


Assuntos
Peptídeos beta-Amiloides , Plexo Corióideo , Contagem de Células , Células Epiteliais , Epitélio
7.
Cell Death Differ ; 29(8): 1596-1610, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35322202

RESUMO

Multiciliated cells (MCCs) in the brain reside in the ependyma and the choroid plexus (CP) epithelia. The CP secretes cerebrospinal fluid that circulates within the ventricular system, driven by ependymal cilia movement. Tumors of the CP are rare primary brain neoplasms mostly found in children. CP tumors exist in three forms: CP papilloma (CPP), atypical CPP, and CP carcinoma (CPC). Though CPP and atypical CPP are generally benign and can be resolved by surgery, CPC is a particularly aggressive and little understood cancer with a poor survival rate and a tendency for recurrence and metastasis. In contrast to MCCs in the CP epithelia, CPCs in humans are characterized by solitary cilia, frequent TP53 mutations, and disturbances to multiciliogenesis program directed by the GMNC-MCIDAS transcriptional network. GMNC and MCIDAS are early transcriptional regulators of MCC fate differentiation in diverse tissues. Consistently, components of the GMNC-MCIDAS transcriptional program are expressed during CP development and required for multiciliation in the CP, while CPC driven by deletion of Trp53 and Rb1 in mice exhibits multiciliation defects consequent to deficiencies in the GMNC-MCIDAS program. Previous studies revealed that abnormal NOTCH pathway activation leads to CPP. Here we show that combined defects in NOTCH and Sonic Hedgehog signaling in mice generates tumors that are similar to CPC in humans. NOTCH-driven CP tumors are monociliated, and disruption of the NOTCH complex restores multiciliation and decreases tumor growth. NOTCH suppresses multiciliation in tumor cells by inhibiting the expression of GMNC and MCIDAS, while Gmnc-Mcidas overexpression rescues multiciliation defects and suppresses tumor cell proliferation. Taken together, these findings indicate that reactivation of the GMNC-MCIDAS multiciliogenesis program is critical for inhibiting tumorigenesis in the CP, and it may have therapeutic implications for the treatment of CPC.


Assuntos
Carcinoma , Proteínas de Ciclo Celular , Neoplasias do Plexo Corióideo , Proteínas Nucleares , Animais , Carcinoma/genética , Proteínas de Ciclo Celular/genética , Neoplasias do Plexo Corióideo/genética , Neoplasias do Plexo Corióideo/patologia , Proteínas Hedgehog/genética , Humanos , Camundongos , Proteínas Nucleares/genética
8.
Trends Neurosci ; 44(11): 864-875, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34312005

RESUMO

The choroid plexus (ChP), an epithelial bilayer containing a network of mesenchymal, immune, and neuronal cells, forms the blood-cerebrospinal fluid (CSF) barrier (BCSFB). While best recognized for secreting CSF, the ChP is also a hotbed of immune cell activity and can provide circulating peripheral immune cells with passage into the central nervous system (CNS). Here, we review recent studies on ChP immune cells, with a focus on the ontogeny, development, and behaviors of ChP macrophages, the principal resident immune cells of the ChP. We highlight the implications of immune cells for ChP barrier function, CSF cytokines and volume regulation, and their contribution to neurodevelopmental disorders, with possible age-specific features to be elucidated in the future.


Assuntos
Barreira Hematoencefálica , Plexo Corióideo , Barreira Hematoencefálica/fisiologia , Sistema Nervoso Central , Líquido Cefalorraquidiano/fisiologia , Plexo Corióideo/fisiologia , Humanos , Imunidade , Macrófagos
9.
Development ; 148(10)2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-34032267

RESUMO

The choroid plexus (ChP) produces cerebrospinal fluid and forms an essential brain barrier. ChP tissues form in each brain ventricle, each one adopting a distinct shape, but remarkably little is known about the mechanisms underlying ChP development. Here, we show that epithelial WNT5A is crucial for determining fourth ventricle (4V) ChP morphogenesis and size in mouse. Systemic Wnt5a knockout, or forced Wnt5a overexpression beginning at embryonic day 10.5, profoundly reduced ChP size and development. However, Wnt5a expression was enriched in Foxj1-positive epithelial cells of 4V ChP plexus, and its conditional deletion in these cells affected the branched, villous morphology of the 4V ChP. We found that WNT5A was enriched in epithelial cells localized to the distal tips of 4V ChP villi, where WNT5A acted locally to activate non-canonical WNT signaling via ROR1 and ROR2 receptors. During 4V ChP development, MEIS1 bound to the proximal Wnt5a promoter, and gain- and loss-of-function approaches demonstrated that MEIS1 regulated Wnt5a expression. Collectively, our findings demonstrate a dual function of WNT5A in ChP development and identify MEIS transcription factors as upstream regulators of Wnt5a in the 4V ChP epithelium.


Assuntos
Plexo Corióideo/embriologia , Epitélio/metabolismo , Quarto Ventrículo/embriologia , Proteína Meis1/metabolismo , Proteína Wnt-5a/metabolismo , Animais , Encéfalo/embriologia , Sistemas CRISPR-Cas/genética , Linhagem Celular , Células Epiteliais/metabolismo , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas/genética , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Transdução de Sinais/fisiologia , Proteína Wnt-5a/genética
10.
Cell ; 184(11): 3056-3074.e21, 2021 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-33932339

RESUMO

The choroid plexus (ChP) in each brain ventricle produces cerebrospinal fluid (CSF) and forms the blood-CSF barrier. Here, we construct a single-cell and spatial atlas of each ChP in the developing, adult, and aged mouse brain. We delineate diverse cell types, subtypes, cell states, and expression programs in epithelial and mesenchymal cells across ages and ventricles. In the developing ChP, we predict a common progenitor pool for epithelial and neuronal cells, validated by lineage tracing. Epithelial and fibroblast cells show regionalized expression by ventricle, starting at embryonic stages and persisting with age, with a dramatic transcriptional shift with maturation, and a smaller shift in each aged cell type. With aging, epithelial cells upregulate host-defense programs, and resident macrophages upregulate interleukin-1ß (IL-1ß) signaling genes. Our atlas reveals cellular diversity, architecture and signaling across ventricles during development, maturation, and aging of the ChP-brain barrier.


Assuntos
Plexo Corióideo/embriologia , Plexo Corióideo/metabolismo , Fatores Etários , Envelhecimento/fisiologia , Animais , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Encéfalo/fisiologia , Encefalopatias/genética , Encefalopatias/fisiopatologia , Diferenciação Celular/genética , Linhagem da Célula/genética , Plexo Corióideo/fisiologia , Células Epiteliais/metabolismo , Feminino , Masculino , Camundongos/embriologia , Camundongos Endogâmicos C57BL , Transdução de Sinais , Análise de Célula Única
11.
Nat Commun ; 12(1): 447, 2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33469018

RESUMO

Cerebrospinal fluid (CSF) provides vital support for the brain. Abnormal CSF accumulation, such as hydrocephalus, can negatively affect perinatal neurodevelopment. The mechanisms regulating CSF clearance during the postnatal critical period are unclear. Here, we show that CSF K+, accompanied by water, is cleared through the choroid plexus (ChP) during mouse early postnatal development. We report that, at this developmental stage, the ChP showed increased ATP production and increased expression of ATP-dependent K+ transporters, particularly the Na+, K+, Cl-, and water cotransporter NKCC1. Overexpression of NKCC1 in the ChP resulted in increased CSF K+ clearance, increased cerebral compliance, and reduced circulating CSF in the brain without changes in intracranial pressure in mice. Moreover, ChP-specific NKCC1 overexpression in an obstructive hydrocephalus mouse model resulted in reduced ventriculomegaly. Collectively, our results implicate NKCC1 in regulating CSF K+ clearance through the ChP in the critical period during postnatal neurodevelopment in mice.


Assuntos
Líquido Cefalorraquidiano/metabolismo , Plexo Corióideo/patologia , Hidrocefalia/patologia , Membro 2 da Família 12 de Carreador de Soluto/metabolismo , Animais , Animais Recém-Nascidos , Plexo Corióideo/diagnóstico por imagem , Plexo Corióideo/crescimento & desenvolvimento , Plexo Corióideo/metabolismo , Dependovirus/genética , Modelos Animais de Doenças , Embrião de Mamíferos , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Hidrocefalia/congênito , Hidrocefalia/diagnóstico , Hidrocefalia/fisiopatologia , Injeções Intraventriculares , Pressão Intracraniana/fisiologia , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Transgênicos , Membro 2 da Família 12 de Carreador de Soluto/genética
12.
Am J Pathol ; 188(6): 1334-1344, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29545198

RESUMO

Choroid plexus tumors and ciliary body medulloepithelioma are predominantly pediatric neoplasms. Progress in understanding the pathogenesis of these tumors has been hindered by their rarity and lack of models that faithfully recapitulate the disease. Here, we find that endogenous Myc proto-oncogene protein is down-regulated in the forebrain neuroepithelium, whose neural plate border domains give rise to the anterior choroid plexus and ciliary body. To uncover the consequences of persistent Myc expression, MYC expression was forced in multipotent neural precursors (nestin-Cre:Myc), which produced fully penetrant models of choroid plexus carcinoma and ciliary body medulloepithelioma. Nestin-mediated MYC expression in the epithelial cells of choroid plexus leads to the regionalized formation of choroid plexus carcinoma in the posterior domain of the lateral ventricle choroid plexus and the fourth ventricle choroid plexus that is accompanied by loss of multiple cilia, up-regulation of protein biosynthetic machinery, and hydrocephalus. Parallel MYC expression in the ciliary body leads also to up-regulation of protein biosynthetic machinery. Additionally, Myc expression in human choroid plexus tumors increases with aggressiveness of disease. Collectively, our findings expose a select vulnerability of the neuroepithelial lineage to postnatal tumorigenesis and provide a new mouse model for investigating the pathogenesis of these rare pediatric neoplasms.


Assuntos
Carcinogênese/patologia , Neoplasias do Plexo Corióideo/patologia , Corpo Ciliar/patologia , Modelos Animais de Doenças , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Adolescente , Adulto , Animais , Carcinogênese/genética , Carcinogênese/metabolismo , Criança , Pré-Escolar , Neoplasias do Plexo Corióideo/genética , Neoplasias do Plexo Corióideo/metabolismo , Corpo Ciliar/metabolismo , Feminino , Humanos , Lactente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/patologia , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-myc/genética , Adulto Jovem
13.
Epileptic Disord ; 18(S2): 28-37, 2016 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-27582036

RESUMO

We first review the clinical presentation and current therapeutic approaches available for treating Unverricht-Lundborg disease (ULD), a progressive myoclonus epilepsy. Next, we describe the identification of disease causing mutations in the gene encoding cystatin B (CSTB). A Cstb-deficient mouse model, which recapitulates the key features of ULD including myoclonic seizures, ataxia, and neuronal loss, was generated to shed light on the mechanisms contributing to disease pathophysiology. Studies with this model have elucidated the diverse biological roles for Cstb from functioning as a protease inhibitor, to regulating glial activation, oxidative stress, serotonergic neurotransmission, and hyperexcitability. These findings set the stage for future studies that may open avenues to improved therapeutic approaches.


Assuntos
Cistatina B/genética , Síndrome de Unverricht-Lundborg/genética , Animais , Humanos
14.
Nat Cell Biol ; 18(4): 418-30, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26999738

RESUMO

Aberrant Notch signalling has been linked to many cancers including choroid plexus (CP) tumours, a group of rare and predominantly paediatric brain neoplasms. We developed animal models of CP tumours, by inducing sustained expression of Notch1, that recapitulate properties of human CP tumours with aberrant NOTCH signalling. Whole-transcriptome and functional analyses showed that tumour cell proliferation is associated with Sonic Hedgehog (Shh) in the tumour microenvironment. Unlike CP epithelial cells, which have multiple primary cilia, tumour cells possess a solitary primary cilium as a result of Notch-mediated suppression of multiciliate differentiation. A Shh-driven signalling cascade in the primary cilium occurs in tumour cells but not in epithelial cells. Lineage studies show that CP tumours arise from monociliated progenitors in the roof plate characterized by elevated Notch signalling. Abnormal SHH signalling and distinct ciliogenesis are detected in human CP tumours, suggesting the SHH pathway and cilia differentiation as potential therapeutic avenues.


Assuntos
Proliferação de Células/genética , Neoplasias do Plexo Corióideo/genética , Proteínas Hedgehog/genética , Receptor Notch1/genética , Animais , Western Blotting , Plexo Corióideo/metabolismo , Plexo Corióideo/patologia , Plexo Corióideo/ultraestrutura , Neoplasias do Plexo Corióideo/metabolismo , Neoplasias do Plexo Corióideo/patologia , Cílios/metabolismo , Cílios/ultraestrutura , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Proteínas Hedgehog/metabolismo , Humanos , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Análise de Sequência com Séries de Oligonucleotídeos , Receptor Notch1/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Células Tumorais Cultivadas , Microambiente Tumoral/genética
15.
J Neurosci ; 35(12): 4903-16, 2015 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-25810521

RESUMO

A sheet of choroid plexus epithelial cells extends into each cerebral ventricle and secretes signaling factors into the CSF. To evaluate whether differences in the CSF proteome across ventricles arise, in part, from regional differences in choroid plexus gene expression, we defined the transcriptome of lateral ventricle (telencephalic) versus fourth ventricle (hindbrain) choroid plexus. We find that positional identities of mouse, macaque, and human choroid plexi derive from gene expression domains that parallel their axial tissues of origin. We then show that molecular heterogeneity between telencephalic and hindbrain choroid plexi contributes to region-specific, age-dependent protein secretion in vitro. Transcriptome analysis of FACS-purified choroid plexus epithelial cells also predicts their cell-type-specific secretome. Spatial domains with distinct protein expression profiles were observed within each choroid plexus. We propose that regional differences between choroid plexi contribute to dynamic signaling gradients across the mammalian cerebroventricular system.


Assuntos
Líquido Cefalorraquidiano/metabolismo , Plexo Corióideo/metabolismo , Quarto Ventrículo/metabolismo , Ventrículos Laterais/metabolismo , Transcriptoma , Envelhecimento/metabolismo , Animais , Células Epiteliais/metabolismo , Feminino , Humanos , Macaca mulatta , Masculino , Camundongos
16.
Neuron ; 74(1): 41-8, 2012 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-22500628

RESUMO

Hemimegalencephaly (HMG) is a developmental brain disorder characterized by an enlarged, malformed cerebral hemisphere, typically causing epilepsy that requires surgical resection. We studied resected HMG tissue to test whether the condition might reflect somatic mutations affecting genes critical to brain development. We found that two out of eight HMG samples showed trisomy of chromosome 1q, which encompasses many genes, including AKT3, a gene known to regulate brain size. A third case showed a known activating mutation in AKT3 (c.49G→A, creating p.E17K) that was not present in the patient's blood cells. Remarkably, the E17K mutation in AKT3 is exactly paralogous to E17K mutations in AKT1 and AKT2 recently discovered in somatic overgrowth syndromes. We show that AKT3 is the most abundant AKT paralog in the brain during neurogenesis and that phosphorylated AKT is abundant in cortical progenitor cells. Our data suggest that somatic mutations limited to the brain could represent an important cause of complex neurogenetic disease.


Assuntos
Cérebro/anormalidades , Cromossomos Humanos Par 1/genética , Malformações do Desenvolvimento Cortical/genética , Neurogênese/genética , Proteínas Proto-Oncogênicas c-akt/genética , Trissomia/genética , Cérebro/crescimento & desenvolvimento , Cérebro/patologia , Epilepsia/etiologia , Epilepsia/patologia , Epilepsia/cirurgia , Humanos , Malformações do Desenvolvimento Cortical/complicações , Malformações do Desenvolvimento Cortical/patologia
17.
Neuron ; 69(5): 893-905, 2011 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-21382550

RESUMO

Cortical development depends on the active integration of cell-autonomous and extrinsic cues, but the coordination of these processes is poorly understood. Here, we show that the apical complex protein Pals1 and Pten have opposing roles in localizing the Igf1R to the apical, ventricular domain of cerebral cortical progenitor cells. We found that the cerebrospinal fluid (CSF), which contacts this apical domain, has an age-dependent effect on proliferation, much of which is attributable to Igf2, but that CSF contains other signaling activities as well. CSF samples from patients with glioblastoma multiforme show elevated Igf2 and stimulate stem cell proliferation in an Igf2-dependent manner. Together, our findings demonstrate that the apical complex couples intrinsic and extrinsic signaling, enabling progenitors to sense and respond appropriately to diffusible CSF-borne signals distributed widely throughout the brain. The temporal control of CSF composition may have critical relevance to normal development and neuropathological conditions.


Assuntos
Córtex Cerebral/fisiologia , Líquido Cefalorraquidiano/fisiologia , Células-Tronco Neurais/fisiologia , Análise de Variância , Animais , Neoplasias Encefálicas/líquido cefalorraquidiano , Proliferação de Células , Córtex Cerebral/citologia , Glioblastoma/líquido cefalorraquidiano , Humanos , Fator de Crescimento Insulin-Like II/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Células-Tronco Neurais/citologia , Neurônios/metabolismo , Núcleosídeo-Fosfato Quinase/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Receptor IGF Tipo 1/metabolismo , Estatísticas não Paramétricas
18.
J Neurosci ; 29(18): 5910-5, 2009 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-19420257

RESUMO

The progressive myoclonus epilepsies, featuring the triad of myoclonus, seizures, and ataxia, comprise a large group of inherited neurodegenerative diseases that remain poorly understood and refractory to treatment. The Cystatin B gene is mutated in one of the most common forms of progressive myoclonus epilepsy, Unverricht-Lundborg disease (EPM1). Cystatin B knockout in a mouse model of EPM1 triggers progressive degeneration of cerebellar granule neurons. Here, we report impaired redox homeostasis as a key mechanism by which Cystatin B deficiency triggers neurodegeneration. Oxidative stress induces the expression of Cystatin B in cerebellar granule neurons, and EPM1 patient-linked mutation of the Cystatin B gene promoter impairs oxidative stress induction of Cystatin B transcription. Importantly, Cystatin B knockout or knockdown sensitizes cerebellar granule neurons to oxidative stress-induced cell death. The Cystatin B deficiency-induced predisposition to oxidative stress in neurons is mediated by the lysosomal protease Cathepsin B. We uncover evidence of oxidative damage, reflected by depletion of antioxidants and increased lipid peroxidation, in the cerebellum of Cystatin B knock-out mice in vivo. Collectively, our findings define a pathophysiological mechanism in EPM1, whereby Cystatin B deficiency couples oxidative stress to neuronal death and degeneration, and may thus provide the basis for novel treatment approaches for the progressive myoclonus epilepsies.


Assuntos
Cistationina gama-Liase/deficiência , Neurônios/fisiologia , Estresse Oxidativo/genética , Síndrome de Unverricht-Lundborg/fisiopatologia , Análise de Variância , Animais , Animais Recém-Nascidos , Catepsina B , Morte Celular/genética , Células Cultivadas , Cerebelo/patologia , Modelos Animais de Doenças , Progressão da Doença , Galactosídeos/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Ácido Glutâmico/farmacologia , Proteínas de Fluorescência Verde/genética , Peróxido de Hidrogênio/farmacologia , Camundongos , Camundongos Knockout , Neurônios/efeitos dos fármacos , Oxidantes/farmacologia , Oxirredução/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Ratos , Transfecção/métodos , Síndrome de Unverricht-Lundborg/genética , Síndrome de Unverricht-Lundborg/patologia
19.
J Biol Chem ; 284(17): 11285-92, 2009 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-19221179

RESUMO

The protein kinase mammalian Sterile 20-like kinase 1 (MST1) plays a critical role in the regulation of cell death. Recent studies suggest that MST1 mediates oxidative stress-induced neuronal cell death by phosphorylating the transcription factor FOXO3 at serine 207, a site that is conserved in other FOXO family members. Here, we show that MST1-induced phosphorylation of FOXO1 at serine 212, corresponding to serine 207 in FOXO3, disrupts the association of FOXO1 with 14-3-3 proteins. Accordingly, MST1 mediates the nuclear translocation of FOXO1 in primary rat cerebellar granule neurons that are deprived of neuronal activity. We also find a requirement for MST1 in cell death of granule neurons upon withdrawal of growth factors and neuronal activity, and MST1 induces cell death in a FOXO1-dependent manner. Finally, we show that the MST1-regulatory, scaffold protein Nore1 is required for survival factor deprivation induced neuronal death. Collectively, these findings define MST1-FOXO1 signaling as an important link survival factor deprivation-induced neuronal cell death with implications for our understanding of brain development and neurological diseases.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Fator de Crescimento de Hepatócito/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Proteínas Proto-Oncogênicas/metabolismo , Animais , Morte Celular , Linhagem Celular , Cerebelo/metabolismo , Proteína Forkhead Box O1 , Humanos , Modelos Biológicos , Fosforilação , Ratos , Transdução de Sinais
20.
Curr Mol Med ; 6(8): 871-81, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17168738

RESUMO

Oxidative stress is associated with the onset and pathogenesis of several prominent central nervous system disorders. Consequently, there is a pressing need for experimental methods for studying neuronal responses to oxidative stress. A number of techniques for modeling oxidative stress have been developed, including the use of inhibitors of the mitochondrial respiratory chain, depletion of endogenous antioxidants, application of products of lipid peroxidation, use of heavy metals, and models of ischemic brain injury. These experimental approaches can be applied from cell culture to in vivo animal models. Their use has provided insight into the molecular underpinnings of oxidative stress responses in the nervous system, including cell recovery and cell death. Reactive oxygen species contribute to conformational change-induced activation of signaling pathways, inactivation of enzymes through modification of catalytic cysteine residues, and subcellular redistribution of signaling molecules. In this review, we will discuss several methods for inducing oxidative stress in the nervous system and explore newly emerging concepts in oxidative stress signaling.


Assuntos
Sistema Nervoso Central/metabolismo , Modelos Biológicos , Doenças Neurodegenerativas/etiologia , Estresse Oxidativo , Transdução de Sinais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA