Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Circ Res ; 127(6): 727-743, 2020 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-32552404

RESUMO

RATIONALE: We previously identified somatic activating mutations in the KRAS (Kirsten rat sarcoma viral oncogene homologue) gene in the endothelium of the majority of human sporadic brain arteriovenous malformations; a disorder characterized by direct connections between arteries and veins. However, whether this genetic abnormality alone is sufficient for lesion formation, as well as how active KRAS signaling contributes to arteriovenous malformations, remains unknown. OBJECTIVE: To establish the first in vivo models of somatic KRAS gain of function in the endothelium in both mice and zebrafish to directly observe the phenotypic consequences of constitutive KRAS activity at a cellular level in vivo, and to test potential therapeutic interventions for arteriovenous malformations. METHODS AND RESULTS: Using both postnatal and adult mice, as well as embryonic zebrafish, we demonstrate that endothelial-specific gain of function mutations in Kras (G12D or G12V) are sufficient to induce brain arteriovenous malformations. Active KRAS signaling leads to altered endothelial cell morphogenesis and increased cell size, ectopic sprouting, expanded vessel lumen diameter, and direct connections between arteries and veins. Furthermore, we show that these lesions are not associated with altered endothelial growth dynamics or a lack of proper arteriovenous identity but instead seem to feature exuberant angiogenic signaling. Finally, we demonstrate that KRAS-dependent arteriovenous malformations in zebrafish are refractory to inhibition of the downstream effector PI3K but instead require active MEK (mitogen-activated protein kinase kinase 1) signaling. CONCLUSIONS: We demonstrate that active KRAS expression in the endothelium is sufficient for brain arteriovenous malformations, even in the setting of uninjured adult vasculature. Furthermore, the finding that KRAS-dependent lesions are reversible in zebrafish suggests that MEK inhibition may represent a promising therapeutic treatment for arteriovenous malformation patients. Graphical Abstract: A graphical abstract is available for this article.


Assuntos
Células Endoteliais/enzimologia , Mutação com Ganho de Função , Malformações Arteriovenosas Intracranianas/genética , MAP Quinase Quinase 1/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Animais , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/patologia , Feminino , Predisposição Genética para Doença , Células Endoteliais da Veia Umbilical Humana/enzimologia , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Malformações Arteriovenosas Intracranianas/enzimologia , Malformações Arteriovenosas Intracranianas/patologia , Hemorragias Intracranianas/enzimologia , Hemorragias Intracranianas/genética , Hemorragias Intracranianas/patologia , MAP Quinase Quinase 1/antagonistas & inibidores , Masculino , Camundongos Transgênicos , Permeabilidade , Fenótipo , Fosfatidilinositol 3-Quinase/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase/farmacologia , Transdução de Sinais , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra
2.
Nature ; 538(7624): 253-256, 2016 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-27698417

RESUMO

Atypical food intake is a primary cause of obesity and other eating and metabolic disorders. Insight into the neural control of feeding has previously focused mainly on signalling mechanisms associated with the hypothalamus, the major centre in the brain that regulates body weight homeostasis. However, roles of non-canonical central nervous system signalling mechanisms in regulating feeding behaviour have been largely uncharacterized. Acetylcholine has long been proposed to influence feeding owing in part to the functional similarity between acetylcholine and nicotine, a known appetite suppressant. Nicotine is an exogenous agonist for acetylcholine receptors, suggesting that endogenous cholinergic signalling may play a part in normal physiological regulation of feeding. However, it remains unclear how cholinergic neurons in the brain regulate food intake. Here we report that cholinergic neurons of the mouse basal forebrain potently influence food intake and body weight. Impairment of cholinergic signalling increases food intake and results in severe obesity, whereas enhanced cholinergic signalling decreases food consumption. We found that cholinergic circuits modulate appetite suppression on downstream targets in the hypothalamus. Together our data reveal the cholinergic basal forebrain as a major modulatory centre underlying feeding behaviour.


Assuntos
Regulação do Apetite/fisiologia , Prosencéfalo Basal/citologia , Prosencéfalo Basal/fisiologia , Neurônios Colinérgicos/metabolismo , Comportamento Alimentar/fisiologia , Resposta de Saciedade/fisiologia , Acetilcolina/metabolismo , Animais , Peso Corporal/fisiologia , Morte Celular , Colina O-Acetiltransferase/deficiência , Agonistas Colinérgicos , Neurônios Colinérgicos/patologia , Ingestão de Alimentos/fisiologia , Ingestão de Alimentos/psicologia , Comportamento Alimentar/psicologia , Feminino , Homeostase , Hiperfagia/enzimologia , Hiperfagia/genética , Hiperfagia/patologia , Hipotálamo/citologia , Hipotálamo/fisiologia , Masculino , Camundongos , Camundongos Knockout , Modelos Neurológicos , Nicotina/metabolismo , Obesidade/enzimologia , Obesidade/genética , Obesidade/patologia , Receptores Colinérgicos/metabolismo
3.
N Engl J Med ; 378(3): 250-261, 2018 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-29298116

RESUMO

BACKGROUND: Sporadic arteriovenous malformations of the brain, which are morphologically abnormal connections between arteries and veins in the brain vasculature, are a leading cause of hemorrhagic stroke in young adults and children. The genetic cause of this rare focal disorder is unknown. METHODS: We analyzed tissue and blood samples from patients with arteriovenous malformations of the brain to detect somatic mutations. We performed exome DNA sequencing of tissue samples of arteriovenous malformations of the brain from 26 patients in the main study group and of paired blood samples from 17 of those patients. To confirm our findings, we performed droplet digital polymerase-chain-reaction (PCR) analysis of tissue samples from 39 patients in the main study group (21 with matching blood samples) and from 33 patients in an independent validation group. We interrogated the downstream signaling pathways, changes in gene expression, and cellular phenotype that were induced by activating KRAS mutations, which we had discovered in tissue samples. RESULTS: We detected somatic activating KRAS mutations in tissue samples from 45 of the 72 patients and in none of the 21 paired blood samples. In endothelial cell-enriched cultures derived from arteriovenous malformations of the brain, we detected KRAS mutations and observed that expression of mutant KRAS (KRASG12V) in endothelial cells in vitro induced increased ERK (extracellular signal-regulated kinase) activity, increased expression of genes related to angiogenesis and Notch signaling, and enhanced migratory behavior. These processes were reversed by inhibition of MAPK (mitogen-activated protein kinase)-ERK signaling. CONCLUSIONS: We identified activating KRAS mutations in the majority of tissue samples of arteriovenous malformations of the brain that we analyzed. We propose that these malformations develop as a result of KRAS-induced activation of the MAPK-ERK signaling pathway in brain endothelial cells. (Funded by the Swiss Cancer League and others.).


Assuntos
Malformações Arteriovenosas Intracranianas/genética , Mutação , Proteínas Proto-Oncogênicas p21(ras)/genética , Adulto , Células Cultivadas , Análise Mutacional de DNA , Exoma , Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Malformações Arteriovenosas Intracranianas/etiologia , Malformações Arteriovenosas Intracranianas/patologia , MAP Quinase Quinase Quinases/metabolismo , Sistema de Sinalização das MAP Quinases , Fosforilação , Proteínas Proto-Oncogênicas p21(ras)/metabolismo
4.
Development ; 144(13): 2428-2444, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28536097

RESUMO

The transcriptional pathways activated downstream of vascular endothelial growth factor (VEGF) signaling during angiogenesis remain incompletely characterized. By assessing the signals responsible for induction of the Notch ligand delta-like 4 (DLL4) in endothelial cells, we find that activation of the MAPK/ERK pathway mirrors the rapid and dynamic induction of DLL4 transcription and that this pathway is required for DLL4 expression. Furthermore, VEGF/ERK signaling induces phosphorylation and activation of the ETS transcription factor ERG, a prerequisite for DLL4 induction. Transcription of DLL4 coincides with dynamic ERG-dependent recruitment of the transcriptional co-activator p300. Genome-wide gene expression profiling identified a network of VEGF-responsive and ERG-dependent genes, and ERG chromatin immunoprecipitation (ChIP)-seq revealed the presence of conserved ERG-bound putative enhancer elements near these target genes. Functional experiments performed in vitro and in vivo confirm that this network of genes requires ERK, ERG and p300 activity. Finally, genome-editing and transgenic approaches demonstrate that a highly conserved ERG-bound enhancer located upstream of HLX (which encodes a transcription factor implicated in sprouting angiogenesis) is required for its VEGF-mediated induction. Collectively, these findings elucidate a novel transcriptional pathway contributing to VEGF-dependent angiogenesis.


Assuntos
Proteína p300 Associada a E1A/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Ligação ao Cálcio , Bovinos , Elementos Facilitadores Genéticos/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Íntrons/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Neovascularização Fisiológica/genética , Regulador Transcricional ERG/metabolismo , Peixe-Zebra/embriologia
5.
J Neurosci Res ; 98(2): 312-324, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31630455

RESUMO

Recent advances in three-dimensional (3D) fluorescence microscopy offer the ability to image the entire vascular network in entire organs, or even whole animals. However, these imaging modalities rely on either endogenous fluorescent reporters or involved immunohistochemistry protocols, as well as optical clearing of the tissue and refractive index matching. Conversely, X-ray-based 3D imaging modalities, such as micro CT, can image non-transparent samples, at high resolution, without requiring complicated or expensive immunolabeling and clearing protocols, or fluorescent reporters. Here, we compared two "homemade" barium-based contrast agents to the field standard, lead-containing Microfil, for micro-computed tomography (micro CT) imaging of the adult mouse cerebrovasculature. The perfusion pressure required for uniform vessel filling was significantly lower with the barium-based contrast agents compared to the polymer-based Microfil. Accordingly, the barium agents showed no evidence of vascular distension or rupture, common problems associated with Microfil. Compellingly, perfusion of an aqueous BaCl2 /gelatin mixture yielded equal or superior visualization of the cerebrovasculature by micro CT compared to Microfil. However, phosphate-containing buffers and fixatives were incompatible with BaCl2 due to the formation of unwanted precipitates. X-ray attenuation of the vessels also decreased overtime, as the BaCl2 appeared to gradually diffuse into surrounding tissues. A second, unique formulation composed of BaSO4 microparticles, generated in-house by mixing BaCl2 and MgSO4 , suffered none of these drawbacks. These microparticles, however, were unable to pass small diameter capillary vessels, conveniently labeling only the arterial cerebrovasculature. In summary, we present an affordable, robust, low pressure, non-toxic, and straightforward methodology for 3D visualization of the cerebrovasculature.


Assuntos
Bário , Circulação Cerebrovascular/fisiologia , Imageamento Tridimensional/métodos , Microtomografia por Raio-X/métodos , Animais , Meios de Contraste , Camundongos
7.
Hum Mol Genet ; 20(11): 2091-102, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21378096

RESUMO

Intraneuronal amyloid-ß (Aß) may contribute to extracellular plaque deposition, the characteristic pathology of Alzheimer's disease (AD). The E3-ubiquitin ligase parkin ubiquitinates intracellular proteins and induces mitophagy. We previously demonstrated that parkin reduces Aß levels in lentiviral models of intracellular Aß. Here we used a triple transgenic AD (3xTg-AD) mouse, which over-expresses APP(Swe), Tau(P301L) and harbor the PS1(M146V) knock-in mutation and found that lentiviral parkin ubiquitinated intracellular Aß in vivo, stimulated beclin-dependent molecular cascade of autophagy and facilitated clearance of vesicles containing debris and defective mitochondria. Parkin expression decreased intracellular Aß levels and extracellular plaque deposition. Parkin expression also attenuated caspase activity, prevented mitochondrial dysfunction and oxidative stress and restored neurotransmitter synthesis. Restoration of glutamate synthesis, which was independent of glial-neuronal recycling, depended on mitochondrial activity and led to an increase in γ-amino butyric acid levels. These data indicate that parkin may be used as an alternative strategy to reduce Aß levels and enhance autophagic clearance of Aß-induced defects in AD. Parkin-mediated clearance of ubiquitinated Aß may act in parallel with autophagy to clear molecular debris and defective mitochondria and restore neurotransmitter balance.


Assuntos
Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia , Mitocôndrias/patologia , Ubiquitina-Proteína Ligases/metabolismo , Doença de Alzheimer/metabolismo , Animais , Proteína Beclina-1 , Western Blotting , Caspase 3/metabolismo , Caspase 9/metabolismo , Córtex Cerebral/fisiopatologia , Córtex Cerebral/ultraestrutura , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Técnicas de Introdução de Genes , Vetores Genéticos , Imuno-Histoquímica/métodos , Lentivirus/metabolismo , Espectrometria de Massas , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Mutação , Neurotransmissores/análise , Neurotransmissores/metabolismo , Estresse Oxidativo , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
8.
Mol Cell Neurosci ; 49(1): 44-53, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21945393

RESUMO

Neurodegeneration involves multiple pathogenic proteins, including Tau, Aß, TDP-43 and α-Synuclein, but there is little information how these pathogenic proteins interact. We cloned human wild type 4 repeat Tau (Tau(wt)) and mutant Tau(P301L) into a lentivirus and performed stereotaxic injection into the rat motor cortex to examine Tau modification, neuro-inflammation and changes of other proteins associated with neurodegeneration. Tau(P301L) was associated with more phosphorylation of Tau, including Thr 181 and Ser 262 residues and resulted in more aggregation. Both forms of Tau expression increased glycogen synthase kinase-3 (GSK-3) activity, polo-like kinase-2 (PLK2) levels and decreased protein phosphatase activity, but had no effects on casein kinase-1 (CK1). No changes were observed in glial fibrillary acidic protein (GFAP) staining with either Tau(wt) or Tau(P301L), but both caused microglial changes and higher interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) levels. Tau(wt) and Tau(P301L) increased the levels of endogenous α-Synuclein, but not ß-amyloid precursor protein (ßAPP) or Tar-DNA binding protein (TDP-43). The levels of phosphorylated Ser-129 α-Synuclein (p-Ser129) were also increased with Tau(wt) and Tau(P301L) expressing animals. These data suggest that Tau(wt) and Tau(P301L) alter kinase activities, but they differentially induce inflammation, Tau modification and α-Synuclein phosphorylation. This change of α-Synuclein in Tau gene transfer models suggests that Tau pathology may lead to α-Synuclein modification in neurodegenerative diseases.


Assuntos
Tauopatias/genética , alfa-Sinucleína/metabolismo , Proteínas tau/genética , Animais , Proteínas de Ligação a DNA/metabolismo , Técnicas de Transferência de Genes , Quinase 3 da Glicogênio Sintase/metabolismo , Inflamação/genética , Inflamação/metabolismo , Interleucina-6/metabolismo , Lentivirus/genética , Modelos Animais , Córtex Motor/metabolismo , Fosforilação/fisiologia , Ratos , Ratos Sprague-Dawley , Tauopatias/metabolismo , alfa-Sinucleína/genética , Proteínas tau/metabolismo
9.
Neuro Oncol ; 23(6): 932-944, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33367832

RESUMO

BACKGROUND: Glioblastoma is the most common and aggressive type of primary brain tumor, as most patients succumb to the disease less than two years after diagnosis. Critically, studies demonstrate that glioma recruits surrounding blood vessels, while some work suggests that tumor stem cells themselves directly differentiate into endothelial cells, yet the molecular and cellular dynamics of the endothelium in glioma are poorly characterized. The goal of this study was to establish molecular and morphological benchmarks for tumor associated vessels (TAVs) and tumor derived endothelial cells (TDECs) during glioblastoma progression. METHODS: Using In-Utero Electroporation and CRISPR/Cas9 genome engineering to generate a native, immunocompetent mouse model of glioma, we characterized vascular-tumor dynamics in three dimensions during tumor progression. We employed bulk and single-cell RNA-Sequencing to elucidate the relationship between TAVs and TDECs. We confirmed our findings in a patient derived orthotopic xenograft (PDOX) model. RESULTS: Using a mouse model of glioma, we identified progressive alteration of vessel function and morphogenesis over time. We also showed in our mouse model that TDECs are a rare subpopulation that contributes to vessels within the tumor, albeit to a limited degree. Furthermore, transcriptional profiling demonstrates that both TAVs and TDECs are molecularly distinct, and both populations feature extensive molecular heterogeneity. Finally, the distinct molecular signatures of these heterogeneous populations are also present in human glioma. CONCLUSIONS: Our findings show extensive endothelial heterogeneity within the tumor and tumor microenvironment and provide insights into the diverse cellular and molecular mechanisms that drive glioma vascularization and angiogenesis during tumorigenesis.


Assuntos
Neoplasias Encefálicas , Glioma , Neoplasias Encefálicas/genética , Células Endoteliais , Endotélio , Glioma/genética , Humanos , Neovascularização Patológica , Microambiente Tumoral
10.
Biol Open ; 7(3)2018 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-29437553

RESUMO

Canonical Notch signaling requires the presence of a membrane bound ligand and a corresponding transmembrane Notch receptor. Receptor engagement induces multiple proteolytic cleavage events culminating in the nuclear accumulation of the Notch intracellular domain and its binding to a transcriptional co-factor to mediate gene expression. Notch signaling networks are essential regulators of vascular patterning and angiogenesis, as well as myriad other biological processes. Delta-like 4 (Dll4) encodes the earliest Notch ligand detected in arterial cells, and is enriched in sprouting endothelial tip cells. Dll4 expression has often been inferred by proxy using a lacZ knockin reporter allele. This is problematic, as a single copy of Dll4 is haploinsufficient. Additionally, Notch activity regulates Dll4 transcription, making it unclear whether these reporter lines accurately reflect Dll4 expression. Accordingly, precisely defining Dll4 expression is essential for determining its role in development and disease. To address these limitations, we generated a novel BAC transgenic allele with a nuclear-localized ß-galactosidase reporter (Dll4-BAC-nlacZ). Through a comparative analysis, we show the BAC line overcomes previous issues of haploinsufficiency, it recapitulates Dll4 expression in vivo, and allows superior visualization and imaging. As such, this novel Dll4 reporter is an important addition to the growing Notch toolkit.

11.
Brain Struct Funct ; 221(1): 1-20, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25224546

RESUMO

Neuropeptides play important physiological functions during distinct behaviors such as arousal, learning, memory, and reproduction. However, the role of local, extrahypothalamic neuropeptide signaling in shaping synapse formation and neuronal plasticity in the brain is not well understood. Here, we characterize the spatiotemporal expression profile of the neuropeptide corticotropin-releasing hormone (CRH) and its receptor CRHR1 in the mouse OB throughout development. We found that CRH-expressing interneurons are present in the external plexiform layer, that its cognate receptor is expressed by granule cells, and show that both CRH and CRHR1 expression enriches in the postnatal period when olfaction becomes important towards olfactory-related behaviors. Further, we provide electrophysiological evidence that CRHR1-expressing granule cells functionally respond to CRH ligand, and that the physiological circuitry of CRHR1 knockout mice is abnormal, leading to impaired olfactory behaviors. Together, these data suggest a physiologically relevant role for local CRH signaling towards shaping the neuronal circuitry within the mouse OB.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Interneurônios/fisiologia , Bulbo Olfatório/fisiologia , Percepção Olfatória/fisiologia , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Animais , Discriminação Psicológica/fisiologia , Potenciais Pós-Sinápticos Excitadores , Feminino , Interneurônios/metabolismo , Memória de Curto Prazo/fisiologia , Camundongos , Camundongos Knockout , Odorantes , Bulbo Olfatório/crescimento & desenvolvimento , Bulbo Olfatório/metabolismo , Receptores de Hormônio Liberador da Corticotropina/genética
12.
Front Neural Circuits ; 8: 124, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25368555

RESUMO

The evolution of genetically targeted tools has begun to allow us to dissect anatomically and functionally heterogeneous interneurons, and to probe circuit function from synapses to behavior. Over the last decade, these tools have been used widely to visualize neurons in a cell type-specific manner, and engage them to activate and inactivate with exquisite precision. In this process, we have expanded our understanding of interneuron diversity, their functional connectivity, and how selective inhibitory circuits contribute to behavior. Here we discuss the relative assets of genetically encoded fluorescent proteins (FPs), viral tracing methods, optogenetics, chemical genetics, and biosensors in the study of inhibitory interneurons and their respective circuits.


Assuntos
Encéfalo/anatomia & histologia , Interneurônios/fisiologia , Rede Nervosa/metabolismo , Inibição Neural/fisiologia , Animais , Imunofluorescência/métodos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Inibição Neural/genética , Optogenética , Transdução Genética
13.
Elife ; 3: e01481, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24473077

RESUMO

Channelrhodopsin-2 (ChR2) has quickly gained popularity as a powerful tool for eliciting genetically targeted neuronal activation. However, little has been reported on the response kinetics of optogenetic stimulation across different neuronal subtypes. With excess stimulation, neurons can be driven into depolarization block, a state where they cease to fire action potentials. Herein, we demonstrate that light-induced depolarization block in neurons expressing ChR2 poses experimental challenges for stable activation of specific cell types and may confound interpretation of experiments when 'activated' neurons are in fact being functionally silenced. We show both ex vivo and in vivo that certain neuronal subtypes targeted for ChR2 expression become increasingly susceptible to depolarization block as the duration of light pulses are increased. We find that interneuron populations have a greater susceptibility to this effect than principal excitatory neurons, which are more resistant to light-induced depolarization block. Our results highlight the need to empirically determine the photo-response properties of targeted neurons when using ChR2, particularly in studies designed to elicit complex circuit responses in vivo where neuronal activity will not be recorded simultaneous to light stimulation. DOI: http://dx.doi.org/10.7554/eLife.01481.001.


Assuntos
Encéfalo/efeitos da radiação , Interneurônios/efeitos da radiação , Luz , Inibição Neural/efeitos da radiação , Potenciais de Ação/efeitos da radiação , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Channelrhodopsins , Técnicas In Vitro , Interneurônios/metabolismo , Cinética , Camundongos Transgênicos , Optogenética , Estimulação Luminosa
14.
Dev Cell ; 30(6): 645-59, 2014 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-25199688

RESUMO

Neural activity either enhances or impairs de novo synaptogenesis and circuit integration of neurons, but how this activity is mechanistically relayed in the adult brain is largely unknown. Neuropeptide-expressing interneurons are widespread throughout the brain and are key candidates for conveying neural activity downstream via neuromodulatory pathways that are distinct from classical neurotransmission. With the goal of identifying signaling mechanisms that underlie neuronal circuit integration in the adult brain, we have virally traced local corticotropin-releasing hormone (CRH)-expressing inhibitory interneurons with extensive presynaptic inputs onto new neurons that are continuously integrated into the adult rodent olfactory bulb. Local CRH signaling onto adult-born neurons promotes and/or stabilizes chemical synapses in the olfactory bulb, revealing a neuromodulatory mechanism for continued circuit plasticity, synapse formation, and integration of new neurons in the adult brain.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Interneurônios/fisiologia , Neurogênese , Sinapses/fisiologia , Animais , Hormônio Liberador da Corticotropina/genética , Interneurônios/citologia , Interneurônios/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Plasticidade Neuronal , Bulbo Olfatório/citologia , Bulbo Olfatório/crescimento & desenvolvimento , Bulbo Olfatório/metabolismo , Sinapses/metabolismo
15.
Exp Neurol ; 235(1): 297-305, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22402344

RESUMO

The transactivation DNA-binding protein (TDP-43) pathology is associated with fronto-temporal lobar dementia (FTLD) with ubiquitinated inclusions and some cases of Alzheimer's disease (AD). Proteolytic fragments of ß-amyloid precursor protein (ßAPP) are detected in AD as well as the cerebrospinal fluid (CSF) from FTLD and Amyotrophic Lateral Sclerosis (ALS) patients, suggesting alteration in APP processing. Because of the overlap in TDP-43 pathology between FTLD and AD, we sought to determine whether there is a relationship between TDP-43 and APP metabolism. We generated gene transfer models using lentiviral delivery of human TDP-43 and Aß(1-42) into the rat primary motor cortex and examined their role 2 weeks post-injection. Expression of TDP-43 and/or Aß(1-42) increase pro-inflammatory markers, including Interleukin (IL)-6, tumor necrosis factor (TNF-α), glial neurofibrillary proteins (GFAP) and ionized calcium binding adaptor molecule 1 (IBA-1). Lentiviral Aß(1-42) up-regulates endogenous TDP-43 and promotes its phosphorylation, aggregation and cleavage into 35 kDa fragments. Inversely, lentiviral TDP-43 expression increases the levels and activity of ß-secretase (BACE), accelerating production of APP C-terminal fragments (C99) and Aß(1-40). Here we show that TDP-43 up-regulates APP metabolism and suggest a mechanistic link between TDP-43 and BACE.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Proteínas de Ligação a DNA/metabolismo , Encefalite/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Ácido Aspártico Endopeptidases/metabolismo , Encéfalo/patologia , Proteínas de Ligação a DNA/genética , Encefalite/patologia , Técnicas de Transferência de Genes , Interleucina-6/metabolismo , Lentivirus/genética , Masculino , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/metabolismo
16.
J Neuroimmunol ; 238(1-2): 1-11, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21820744

RESUMO

Inflammation is secondary to protein accumulation in neurodegenerative diseases, including Alzheimer's, Parkinson's and Amyotrophic Lateral Sclerosis. Emerging evidence indicate sustained inflammatory responses, involving microglia and astrocytes in animal models of neurodegeneration. It is unknown whether inflammation is beneficial or detrimental to disease progression and how inflammatory responses are induced within the CNS. Persistence of an inflammatory stimulus or failure to resolve sustained inflammation can result in pathology, thus, mechanisms that counteract inflammation are indispensable. Here we review studies on inflammation mediated by innate and adaptive immunity in the early stages of neurodegeneration and highlight important areas for future investigation.


Assuntos
Sistema Nervoso Central/metabolismo , Inflamação/etiologia , Doenças Neurodegenerativas/complicações , Doenças Neurodegenerativas/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Humanos , Inflamação/metabolismo , Doenças Neurodegenerativas/classificação , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo
17.
Brain Res ; 1386: 191-9, 2011 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-21376022

RESUMO

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease associated with loss of motor neurons in the brain and spinal cord. ALS is occasionally diagnosed with frontotemporal lobar dementia with ubiquitin-positive inclusions (FTLD-U). Alzheimer's disease (AD) is the most common type of age-associated dementia. Abnormal levels of aggregated Tar-DNA binding protein-43 (TDP-43) are detected in the majority of patients with ALS, FTLD and AD. We observed a significant increase (200%) in the levels of TDP-43 in cortical autopsies of late stage AD patients. Lentiviral expression of Aß(1-42) in the rat motor cortex led to an increase in TDP-43 pathology, including up-regulation of the mature ~44kDa protein, identical to the pathological changes seen in AD. Furthermore, expression of Aß(1-42) was associated with TDP-43 phosphorylation and accumulation in the cytosol. Clearance of Aß with parkin prevented TDP-43 pathology. TDP-43 modifications were also observed in 3xTransgenic AD (3xTg-AD) compared to wild type mice, but these changes were attenuated in parkin-injected hippocampi, even in the presence of Tau pathology, suggesting that TDP-43 pathology is triggered by Aß, independent of Tau. Increased levels of casein kinase (CK1 and CK2), which are associated with TDP-43 phosphorylation, were also observed in Aß(1-42) expressing brains. These data indicate an overlap in TDP-43 pathology between AD and ALS-FTLD and suggest that Aß triggers modifications of TDP-43.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/toxicidade , Esclerose Lateral Amiotrófica/metabolismo , Proteínas de Ligação a DNA/biossíntese , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Esclerose Lateral Amiotrófica/patologia , Animais , Proteínas de Ligação a DNA/fisiologia , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Córtex Motor/metabolismo , Córtex Motor/patologia , Ratos , Ratos Sprague-Dawley , Regulação para Cima/genética
18.
Cell Stress Chaperones ; 16(6): 607-19, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21761186

RESUMO

FKBP65 is an endoplasmic reticulum (ER)-localized chaperone and rotamase, with cargo proteins that include tropoelastin and collagen. In humans, mutations in FKBP65 have recently been shown to cause a form of osteogenesis imperfecta (OI), a brittle bone disease resulting from deficient secretion of mature type I collagen. In this work, we describe the rapid proteolysis of FKBP65 in response to ER stress signals that activate the release of ER Ca(2+) stores. A large-scale screen for stress-induced cellular changes revealed FKBP65 proteins to decrease within 6-12 h of stress activation. Inhibiting IP(3)R-mediated ER Ca(2+) release blocked this response. No other ER-localized chaperone and folding mediators assessed in the study displayed this phenomenon, indicating that this rapid proteolysis of folding mediator is distinctive. Imaging and cellular fractionation confirmed the localization of FKBP65 (72 kDa glycoprotein) to the ER of untreated cells, a rapid decrease in protein levels following ER stress, and the corresponding appearance of a 30-kDa fragment in the cytosol. Inhibition of the proteasome during ER stress revealed an accumulation of FKBP65 in the cytosol, consistent with retrotranslocation and a proteasome-based proteolysis. To assess the role of Ca(2+)-binding EF-hand domains in FKBP65 stability, a recombinant FKBP65-GFP construct was engineered to ablate Ca(2+) binding at each of two EF-hand domains. Cells transfected with the wild-type construct displayed ER localization of the FKBP65-GFP protein and a proteasome-dependent proteolysis in response to ER stress. Recombinant FKBP65-GFP carrying a defect in the EF1 Ca(2+)-binding domain displayed diminished protein in the ER when compared to wild-type FKBP65-GFP. Proteasome inhibition restored mutant protein to levels similar to that of the wild-type FKBP65-GFP. A similar mutation in EF2 did not confer FKBP65 proteolysis. This work supports a model in which stress-induced changes in ER Ca(2+) stores induce the rapid proteolysis of FKBP65, a chaperone and folding mediator of collagen and tropoelastin. The destruction of this protein may identify a cellular strategy for replacement of protein folding machinery following ER stress. The implications for stress-induced changes in the handling of aggregate-prone proteins in the ER-Golgi secretory pathway are discussed. This work was supported by grants from the National Institutes of Health (R15GM065139) and the National Science Foundation (DBI-0452587).


Assuntos
Motivos EF Hand , Estresse do Retículo Endoplasmático , Proteínas de Ligação a Tacrolimo/genética , Proteínas de Ligação a Tacrolimo/metabolismo , Animais , Sítios de Ligação , Cálcio/metabolismo , Linhagem Celular , Cricetinae , Humanos , Camundongos , Mutagênese Sítio-Dirigida , Mutação , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Ratos , Proteínas de Ligação a Tacrolimo/análise , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA