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










Base de dados
Intervalo de ano de publicação
1.
Cell Rep Methods ; 3(11): 100646, 2023 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-37989082

RESUMO

In this issue of Cell Reports Methods, Jin et al. report the generation and validation of a rabies variant, RVΔL, for projection-based neuronal labeling. RVΔL shows little toxicity in vivo and has an improved growth advantage over another variant, RVΔGL, making it a useful tool for a wide variety of systems neuroscience-based studies.


Assuntos
Vírus da Raiva , Raiva , Humanos , Vírus da Raiva/genética , Vetores Genéticos
2.
bioRxiv ; 2023 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-37693480

RESUMO

The medial prefrontal cortex (mPFC) plays a key role in learning, mood and decision making, including in how individuals respond to threats 1-6 . mPFC undergoes a uniquely protracted development, with changes in synapse density, cortical thickness, long-range connectivity, and neuronal encoding properties continuing into early adulthood 7-21 . Models suggest that before adulthood, the slow-developing mPFC cannot adequately regulate activity in faster-developing subcortical centers 22,23 . They propose that during development, the enhanced influence of subcortical systems underlies distinctive behavioural strategies of juveniles and adolescents and that increasing mPFC control over subcortical structures eventually allows adult behaviours to emerge. Yet it has remained unclear how a progressive strengthening of top-down control can lead to nonlinear changes in behaviour as individuals mature 24,25 . To address this discrepancy, here we monitored and manipulated activity in the developing brain as animals responded to threats, establishing direct causal links between frontolimbic circuit activity and the behavioural strategies of juvenile, adolescent and adult mice. Rather than a linear strengthening of mPFC synaptic connectivity progressively regulating behaviour, we uncovered multiple developmental switches in the behavioural roles of mPFC circuits targeting the basolateral amygdala (BLA) and nucleus accumbens (NAc). We show these changes are accompanied by axonal pruning coinciding with functional strengthening of synaptic connectivity in the mPFC-BLA and mPFC-NAc pathways, which mature at different rates. Our results reveal how developing mPFC circuits pass through distinct architectures that may make them optimally adapted to the demands of age-specific challenges.

3.
bioRxiv ; 2023 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-36798174

RESUMO

As the discovery of cellular diversity in the brain accelerates, so does the need for functional tools that target cells based on multiple features, such as gene expression and projection target. By selectively driving recombinase expression in a feature-specific manner, one can utilize intersectional strategies to conditionally promote payload expression only where multiple features overlap. We developed Conditional Viral Expression by Ribozyme Guided Degradation (ConVERGD), a single-construct intersectional targeting strategy that combines a self-cleaving ribozyme with traditional FLEx switches. ConVERGD offers benefits over existing platforms, such as expanded intersectionality, the ability to accommodate larger and more complex payloads, and a vector design that is easily modified to better facilitate rapid toolkit expansion. To demonstrate its utility for interrogating neural circuitry, we employed ConVERGD to target an unexplored subpopulation of norepinephrine (NE)-producing neurons within the rodent locus coeruleus (LC) identified via single-cell transcriptomic profiling to co-express the stress-related endogenous opioid gene prodynorphin (Pdyn). These studies showcase ConVERGD as a versatile tool for targeting diverse cell types and reveal Pdyn-expressing NE+ LC neurons as a small neuronal subpopulation capable of driving anxiogenic behavioral responses in rodents.

4.
Acta Physiol (Oxf) ; 237(3): e13896, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36251565

RESUMO

AIM: Physiological functions in mammals show circadian oscillations, synchronized by daily cycles of light and temperature. Central and peripheral clocks participate in this regulation. Since the ion channel TRPM8 is a critical cold sensor, we investigated its role in circadian function. METHODS: We used TRPM8 reporter mouse lines and TRPM8-deficient mice. mRNA levels were determined by in situ hybridization or RT-qPCR and protein levels by immunofluorescence. A telemetry system was used to measure core body temperature (Tc). RESULTS: TRPM8 is expressed in the retina, specifically in cholinergic amacrine interneurons and in a subset of melanopsin-positive ganglion cells which project to the central pacemaker, the suprachiasmatic nucleus (SCN) of the hypothalamus. TRPM8-positive fibres were also found innervating choroid and ciliary body vasculature, with a putative function in intraocular temperature, as shown in TRPM8-deficient mice. Interestingly, Trpm8-/- animals displayed increased expression of the clock gene Per2 and vasopressin (AVP) in the SCN, suggesting a regulatory role of TRPM8 on the central oscillator. Since SCN AVP neurons control body temperature, we studied Tc in driven and free-running conditions. TRPM8-deficiency increased the amplitude of Tc oscillations and, under dim constant light, induced a greater phase delay and instability of Tc rhythmicity. Finally, TRPM8-positive fibres innervate peripheral organs, like liver and white adipose tissue. Notably, Trpm8-/- mice displayed a dysregulated expression of Per2 mRNA in these metabolic tissues. CONCLUSION: Our findings support a function of TRPM8 as a temperature sensor involved in the regulation of central and peripheral clocks and the circadian control of Tc.


Assuntos
Ritmo Circadiano , Canais de Cátion TRPM , Camundongos , Animais , Ritmo Circadiano/fisiologia , Temperatura Corporal/fisiologia , Núcleo Supraquiasmático/metabolismo , Canais Iônicos/metabolismo , Mamíferos , RNA Mensageiro/metabolismo , Canais de Cátion TRPM/metabolismo
5.
Elife ; 112022 12 30.
Artigo em Inglês | MEDLINE | ID: mdl-36583530

RESUMO

Breathing, and the motor circuits that control it, is essential for life. At the core of respiratory circuits are Dbx1-derived interneurons, which generate the rhythm and pattern of breathing, and phrenic motor neurons (MNs), which provide the final motor output that drives diaphragm muscle contractions during inspiration. Despite their critical function, the principles that dictate how respiratory circuits assemble are unknown. Here, we show that coordinated activity of a type I cadherin (N-cadherin) and type II cadherins (Cadherin-6, -9, and -10) is required in both MNs and Dbx1-derived neurons to generate robust respiratory motor output. Both MN- and Dbx1-specific cadherin inactivation in mice during a critical developmental window results in perinatal lethality due to respiratory failure and a striking reduction in phrenic MN bursting activity. This combinatorial cadherin code is required to establish phrenic MN cell body and dendritic topography; surprisingly, however, cell body position appears to be dispensable for the targeting of phrenic MNs by descending respiratory inputs. Our findings demonstrate that type I and II cadherins function cooperatively throughout the respiratory circuit to generate a robust breathing output and reveal novel strategies that drive the assembly of motor circuits.


The neural circuits which control breathing are established in the womb, ready to switch on with the first gulp of air. Defects in the way that this network is assembled can result in conditions such as sudden infant death syndrome. This process, however, remains poorly understood; in particular, it is still unclear exactly how the two main types of nerve cells which form respiratory circuits start to 'talk' to each other. Known as Dbx1-derived interneurons and phrenic motor neurons, these cell populations reside in different parts of the body and perform distinct roles. The interneurons, which are present in the brainstem, act as a pacemaker to set the rhythm of respiration; the motor neurons reside in the spinal cord, connecting the interneurons with the muscles which allow the lungs to fill with air. Vagnozzi et al. aimed to identify how phrenic motor neurons connect to and relay signals from other neurons involved in breathing to the diaphragm muscle. To do so, the team focused on cadherins, a group of proteins which allow cells to attach to one another. Studded through the membrane, these molecules are also often involved in forming connections from one cell to another that allow them to communicate. Newborn mice in which phrenic motor neurons lacked a specific combination of cadherins experienced respiratory failure, showing that these proteins were needed for breathing circuits to develop normally. Electrical activity recorded from these cells showed that phrenic motor neurons lacking cadherins could not receive the signals required to activate the breathing muscles. Microscopy imaging also revealed that the loss of cadherins shifted the position of the phrenic motor neurons within the spinal cord; however, this change did not seem to affect the connections these cells could establish. The ability to breathe is compromised in many incurable human diseases such as muscular dystrophies and amyotrophic lateral sclerosis. It may be possible to alleviate some of these symptoms by integrating phrenic motor neurons created in the laboratory into existing circuits. Studies which aim to decipher how the respiratory network is established, such as the one conducted by Vagnozzi et al., are essential in this effort.


Assuntos
Neurônios Motores , Respiração , Animais , Camundongos , Neurônios Motores/fisiologia , Interneurônios/fisiologia , Taxa Respiratória , Caderinas , Nervo Frênico , Proteínas de Homeodomínio/metabolismo
6.
Cell Rep ; 35(12): 109274, 2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34161767

RESUMO

Mosaic analysis with double markers (MADM) offers one approach to visualize and concomitantly manipulate genetically defined cells in mice with single-cell resolution. MADM applications include the analysis of lineage, single-cell morphology and physiology, genomic imprinting phenotypes, and dissection of cell-autonomous gene functions in vivo in health and disease. Yet, MADM can only be applied to <25% of all mouse genes on select chromosomes to date. To overcome this limitation, we generate transgenic mice with knocked-in MADM cassettes near the centromeres of all 19 autosomes and validate their use across organs. With this resource, >96% of the entire mouse genome can now be subjected to single-cell genetic mosaic analysis. Beyond a proof of principle, we apply our MADM library to systematically trace sister chromatid segregation in distinct mitotic cell lineages. We find striking chromosome-specific biases in segregation patterns, reflecting a putative mechanism for the asymmetric segregation of genetic determinants in somatic stem cell division.


Assuntos
Biblioteca Gênica , Genoma , Mosaicismo , Análise de Célula Única , Polipose Adenomatosa do Colo/metabolismo , Células-Tronco Adultas/metabolismo , Animais , Cromátides/genética , Segregação de Cromossomos , Cromossomos de Mamíferos/genética , Modelos Animais de Doenças , Marcadores Genéticos , Impressão Genômica , Fígado/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mitose , Modelos Biológicos , Neoplasias/genética , Neoplasias/patologia , Fenótipo , Recombinação Genética/genética , Nicho de Células-Tronco , Dissomia Uniparental
7.
Cell Rep ; 31(11): 107779, 2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32553161

RESUMO

Emotions are distinct patterns of behavioral and physiological responses triggered by stimuli that induce different brain states. Elucidating the circuits is difficult because of challenges in interrogating emotional brain states and their complex outputs. Here, we leverage the recent discovery in mice of a neural circuit for sighing, a simple, quantifiable output of various emotions. We show that mouse confinement triggers sighing, and this "claustrophobic" sighing, but not accompanying tachypnea, requires the same medullary neuromedin B (Nmb)-expressing neurons as physiological sighing. Retrograde tracing from the Nmb neurons identified 12 forebrain centers providing presynaptic input, including hypocretin (Hcrt)-expressing lateral hypothalamic neurons. Confinement activates Hcrt neurons, and optogenetic activation induces sighing and tachypnea whereas pharmacologic inhibition suppresses both responses. The effect on sighing is mediated by HCRT directly on Nmbneurons. We propose that this HCRT-NMB neuropeptide relay circuit mediates claustrophobic sighing and that activated Hcrt neurons are a claustrophobia brain state that directly controls claustrophobic outputs.


Assuntos
Encéfalo/metabolismo , Vias Neurais/metabolismo , Neurônios/metabolismo , Orexinas/metabolismo , Transtornos Fóbicos/metabolismo , Animais , Comportamento Animal , Região Hipotalâmica Lateral/metabolismo , Camundongos , Neuropeptídeos/metabolismo , Optogenética/métodos
8.
J Neurosci ; 39(42): 8239-8249, 2019 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-31619493

RESUMO

The locus coeruleus (LC) is a seemingly singular and compact neuromodulatory nucleus that is a prominent component of disparate theories of brain function due to its broad noradrenergic projections throughout the CNS. As a diffuse neuromodulatory system, noradrenaline affects learning and decision making, control of sleep and wakefulness, sensory salience including pain, and the physiology of correlated forebrain activity (ensembles and networks) and brain hemodynamic responses. However, our understanding of the LC is undergoing a dramatic shift due to the application of state-of-the-art methods that reveal a nucleus of many modules that provide targeted neuromodulation. Here, we review the evidence supporting a modular LC based on multiple levels of observation (developmental, genetic, molecular, anatomical, and neurophysiological). We suggest that the concept of the LC as a singular nucleus and, alongside it, the role of the LC in diverse theories of brain function must be reconsidered.


Assuntos
Neurônios Adrenérgicos/fisiologia , Função Executiva/fisiologia , Locus Cerúleo/fisiologia , Neurônios/fisiologia , Animais , Humanos , Rede Nervosa/fisiologia , Vias Neurais/fisiologia , Dor/fisiopatologia , Sono/fisiologia , Vigília/fisiologia
9.
Science ; 355(6332): 1411-1415, 2017 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-28360327

RESUMO

Slow, controlled breathing has been used for centuries to promote mental calming, and it is used clinically to suppress excessive arousal such as panic attacks. However, the physiological and neural basis of the relationship between breathing and higher-order brain activity is unknown. We found a neuronal subpopulation in the mouse preBötzinger complex (preBötC), the primary breathing rhythm generator, which regulates the balance between calm and arousal behaviors. Conditional, bilateral genetic ablation of the ~175 Cdh9/Dbx1 double-positive preBötC neurons in adult mice left breathing intact but increased calm behaviors and decreased time in aroused states. These neurons project to, synapse on, and positively regulate noradrenergic neurons in the locus coeruleus, a brain center implicated in attention, arousal, and panic that projects throughout the brain.


Assuntos
Nível de Alerta/fisiologia , Locus Cerúleo/fisiologia , Neurônios/fisiologia , Respiração , Animais , Nível de Alerta/genética , Caderinas/genética , Proteínas de Homeodomínio/genética , Locus Cerúleo/citologia , Camundongos , Camundongos Mutantes , Transtorno de Pânico/genética , Transtorno de Pânico/fisiopatologia , Respiração/genética
10.
Neuron ; 92(2): 392-406, 2016 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-27693255

RESUMO

Haploinsufficiency of Retinoic Acid Induced 1 (RAI1) causes Smith-Magenis syndrome (SMS), which is associated with diverse neurodevelopmental and behavioral symptoms as well as obesity. RAI1 encodes a nuclear protein but little is known about its molecular function or the cell types responsible for SMS symptoms. Using genetically engineered mice, we found that Rai1 preferentially occupies DNA regions near active promoters and promotes the expression of a group of genes involved in circuit assembly and neuronal communication. Behavioral analyses demonstrated that pan-neural loss of Rai1 causes deficits in motor function, learning, and food intake. These SMS-like phenotypes are produced by loss of Rai1 function in distinct neuronal types: Rai1 loss in inhibitory neurons or subcortical glutamatergic neurons causes learning deficits, while Rai1 loss in Sim1+ or SF1+ cells causes obesity. By integrating molecular and organismal analyses, our study suggests potential therapeutic avenues for a complex neurodevelopmental disorder.


Assuntos
Comportamento Animal , Regulação da Expressão Gênica/genética , Neurônios/metabolismo , Obesidade/genética , Síndrome de Smith-Magenis/genética , Transativadores/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Variações do Número de Cópias de DNA , Ingestão de Alimentos/genética , Técnicas de Introdução de Genes , Ácido Glutâmico/metabolismo , Haploinsuficiência , Aprendizagem , Camundongos , Camundongos Knockout , Inibição Neural , Fenótipo , Fatores de Processamento de RNA/metabolismo , Proteínas Repressoras/metabolismo
11.
Nat Commun ; 7: 12142, 2016 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-27374071

RESUMO

The precise annotation and accurate identification of neural structures are prerequisites for studying mammalian brain function. The orientation of neurons and neural circuits is usually determined by mapping brain images to coarse axial-sampling planar reference atlases. However, individual differences at the cellular level likely lead to position errors and an inability to orient neural projections at single-cell resolution. Here, we present a high-throughput precision imaging method that can acquire a co-localized brain-wide data set of both fluorescent-labelled neurons and counterstained cell bodies at a voxel size of 0.32 × 0.32 × 2.0 µm in 3 days for a single mouse brain. We acquire mouse whole-brain imaging data sets of multiple types of neurons and projections with anatomical annotation at single-neuron resolution. The results show that the simultaneous acquisition of labelled neural structures and cytoarchitecture reference in the same brain greatly facilitates precise tracing of long-range projections and accurate locating of nuclei.


Assuntos
Encéfalo/diagnóstico por imagem , Conectoma/métodos , Imageamento Tridimensional/métodos , Vias Neurais/diagnóstico por imagem , Neurônios/ultraestrutura , Animais , Encéfalo/citologia , Cor , Estudos de Viabilidade , Processamento de Imagem Assistida por Computador , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia/métodos , Modelos Animais , Análise de Célula Única/métodos , Tomografia/métodos
12.
Curr Biol ; 25(21): R1051-R1056, 2015 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-26528750

RESUMO

The release of the neurotransmitter norepinephrine throughout the mammalian brain is important for modulating attention, arousal, and cognition during many behaviors. Furthermore, disruption of norepinephrine-mediated signaling is strongly associated with several psychiatric and neurodegenerative disorders in humans, emphasizing the clinical importance of this system. Most of the norepinephrine released in the brain is supplied by a very small, bilateral nucleus in the brainstem called the locus coeruleus. The goal of this minireview is to emphasize the complexity of the locus coeruleus beyond its primary definition as a norepinephrine-producing nucleus. Several recent studies utilizing innovative technologies highlight how the locus coeruleus-norepinephrine system can now be targeted with increased accuracy and resolution, in order to better understand its role in modulating diverse behaviors.


Assuntos
Locus Cerúleo/fisiologia , Norepinefrina/fisiologia , Animais , Atenção , Encéfalo/metabolismo , Encéfalo/fisiologia , Tronco Encefálico/metabolismo , Cognição , Humanos , Locus Cerúleo/metabolismo , Norepinefrina/metabolismo
13.
Nature ; 524(7563): 88-92, 2015 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-26131933

RESUMO

Deciphering how neural circuits are anatomically organized with regard to input and output is instrumental in understanding how the brain processes information. For example, locus coeruleus noradrenaline (also known as norepinephrine) (LC-NE) neurons receive input from and send output to broad regions of the brain and spinal cord, and regulate diverse functions including arousal, attention, mood and sensory gating. However, it is unclear how LC-NE neurons divide up their brain-wide projection patterns and whether different LC-NE neurons receive differential input. Here we developed a set of viral-genetic tools to quantitatively analyse the input-output relationship of neural circuits, and applied these tools to dissect the LC-NE circuit in mice. Rabies-virus-based input mapping indicated that LC-NE neurons receive convergent synaptic input from many regions previously identified as sending axons to the locus coeruleus, as well as from newly identified presynaptic partners, including cerebellar Purkinje cells. The 'tracing the relationship between input and output' method (or TRIO method) enables trans-synaptic input tracing from specific subsets of neurons based on their projection and cell type. We found that LC-NE neurons projecting to diverse output regions receive mostly similar input. Projection-based viral labelling revealed that LC-NE neurons projecting to one output region also project to all brain regions we examined. Thus, the LC-NE circuit overall integrates information from, and broadcasts to, many brain regions, consistent with its primary role in regulating brain states. At the same time, we uncovered several levels of specificity in certain LC-NE sub-circuits. These tools for mapping output architecture and input-output relationship are applicable to other neuronal circuits and organisms. More broadly, our viral-genetic approaches provide an efficient intersectional means to target neuronal populations based on cell type and projection pattern.


Assuntos
Encéfalo/citologia , Encéfalo/metabolismo , Técnicas de Rastreamento Neuroanatômico/métodos , Neurônios/metabolismo , Neurônios/virologia , Norepinefrina/metabolismo , Vírus da Raiva/fisiologia , Animais , Axônios/fisiologia , Axônios/virologia , Encéfalo/virologia , Feminino , Locus Cerúleo/citologia , Locus Cerúleo/metabolismo , Locus Cerúleo/virologia , Masculino , Camundongos , Vias Neurais , Projetos Piloto , Células de Purkinje/fisiologia , Células de Purkinje/virologia , Ratos , Ratos Wistar , Reprodutibilidade dos Testes , Sinapses/metabolismo , Sinapses/virologia
14.
J Neurosci ; 34(50): 16637-49, 2014 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-25505317

RESUMO

The trafficking of AMPA receptors (AMPARs) to and from synapses is crucial for synaptic plasticity. Previous work has demonstrated that AMPARs undergo activity-dependent ubiquitination by the E3 ubiquitin ligase Nedd4-1, which promotes their internalization and degradation in lysosomes. Here, we define the molecular mechanisms involved in ubiquitination and deubiquitination of AMPARs. We report that Nedd4-1 is rapidly redistributed to dendritic spines in response to AMPAR activation and not in response to NMDA receptor (NMDAR) activation in cultured rat neurons. In contrast, NMDAR activation directly antagonizes Nedd4-1 function by promoting the deubiquitination of AMPARs. We show that NMDAR activation causes the rapid dephosphorylation and activation of the deubiquitinating enzyme (DUB) USP8. Surface AMPAR levels and synaptic strength are inversely regulated by Nedd4-1 and USP8. Strikingly, we show that homeostatic downscaling of synaptic strength is accompanied by an increase and decrease in Nedd4-1 and USP8 protein levels, respectively. Furthermore, we show that Nedd4-1 is required for homeostatic loss of surface AMPARs and downscaling of synaptic strength. This study provides the first mechanistic evidence for rapid and opposing activity-dependent control of a ubiquitin ligase and DUB at mammalian CNS synapses. We propose that the dynamic regulation of these opposing forces is critical in maintaining synapses and scaling them during homeostatic plasticity.


Assuntos
Endopeptidases/fisiologia , Complexos Endossomais de Distribuição Requeridos para Transporte/fisiologia , Sinapses/fisiologia , Ubiquitina Tiolesterase/fisiologia , Ubiquitina-Proteína Ligases/fisiologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Ubiquitina-Proteína Ligases Nedd4 , Transporte Proteico/fisiologia , Ratos
15.
Mol Cell Neurosci ; 49(3): 387-93, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21884797

RESUMO

Extracellular signaling between cells is often transduced via receptors that reside at the cell membrane. In neurons this receptor-mediated signaling can promote a variety of cellular events such as differentiation, axon outgrowth and guidance, and synaptic development and function. Endocytic membrane trafficking of receptors ensures that the strength and duration of an extracellular signal is properly regulated. The covalent modification of membrane proteins by ubiquitin is a key biological mechanism controlling receptor internalization and endocytic sorting to recycling and degradative pathways in many cell types. In this review we highlight recent findings regarding the ubiquitin-dependent trafficking and turnover of receptors in neurons and the implications for neuronal development and function.


Assuntos
Endocitose/fisiologia , Proteínas de Membrana/metabolismo , Neurônios/metabolismo , Transporte Proteico/fisiologia , Ubiquitina/metabolismo , Axônios/metabolismo , Humanos , Transdução de Sinais/fisiologia , Sinapses/metabolismo
16.
J Neurosci ; 30(49): 16718-29, 2010 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-21148011

RESUMO

The accurate trafficking of AMPA receptors (AMPARs) to and from the synapse is a critical component of learning and memory in the brain, whereas dysfunction of AMPAR trafficking is hypothesized to be an underlying mechanism of Alzheimer's disease. Previous work has shown that ubiquitination of integral membrane proteins is a common posttranslational modification used to mediate endocytosis and endocytic sorting of surface proteins in eukaryotic cells. Here we report that mammalian AMPARs become ubiquitinated in response to their activation. Using a mutant of GluA1 that is unable to be ubiquitinated at lysines on its C-terminus, we demonstrate that ubiquitination is required for internalization of surface AMPARs and their trafficking to the lysosome in response to the AMPAR agonist AMPA but not for internalization of AMPARs in response to the NMDA receptor agonist NMDA. Through overexpression or RNA interference-mediated knockdown, we identify that a specific E3 ligase, Nedd4-1 (neural-precursor cell-expressed developmentally downregulated gene 4-1), is necessary for this process. Finally, we show that ubiquitination of GluA1 by Nedd4-1 becomes more prevalent as neurons mature. Together, these data show that ubiquitination of GluA1-containing AMPARs by Nedd4-1 mediates their endocytosis and trafficking to the lysosome. Furthermore, these results provide insight into how hippocampal neurons regulate AMPAR trafficking and degradation with high specificity in response to differing neuronal signaling cues and suggest that changes to this pathway may occur as neurons mature.


Assuntos
Endocitose/fisiologia , Neurônios/metabolismo , Receptores de AMPA/metabolismo , Ubiquitinação/fisiologia , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Análise de Variância , Animais , Animais Recém-Nascidos , Células Cultivadas , Endocitose/efeitos dos fármacos , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Agonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Proteínas de Fluorescência Verde/genética , Hipocampo/citologia , Humanos , Imunoprecipitação/métodos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Microscopia Confocal/métodos , N-Metilaspartato/farmacologia , Ubiquitina-Proteína Ligases Nedd4 , Neurônios/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , Interferência de RNA/fisiologia , Ratos , Transfecção/métodos , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação/efeitos dos fármacos , Valina/análogos & derivados , Valina/farmacologia , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia
17.
J Biol Chem ; 284(39): 26655-65, 2009 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-19638347

RESUMO

Protein degradation via the ubiquitin proteasome system has been shown to regulate changes in synaptic strength that underlie multiple forms of synaptic plasticity. It is plausible, therefore, that the ubiquitin proteasome system is itself regulated by synaptic activity. By utilizing live-cell imaging strategies we report the rapid and dynamic regulation of the proteasome in hippocampal neurons by synaptic activity. We find that the blockade of action potentials (APs) with tetrodotoxin inhibited the activity of the proteasome, whereas the up-regulation of APs with bicuculline dramatically increased the activity of the proteasome. In addition, the regulation of the proteasome is dependent upon external calcium entry in part through N-methyl-D-aspartate receptors and L-type voltage-gated calcium channels and requires the activity of calcium/calmodulin-dependent protein kinase II (CaMKII). Using in vitro and in vivo assays we find that CaMKII stimulates proteasome activity and directly phosphorylates Rpt6, a subunit of the 19 S (PA700) subcomplex of the 26 S proteasome. Our data provide a novel mechanism whereby CaMKII may regulate the proteasome in neurons to facilitate remodeling of synaptic connections through protein degradation.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Neurônios/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Cálcio/metabolismo , Cálcio/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Linhagem Celular , Células Cultivadas , Dendritos/efeitos dos fármacos , Dendritos/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/citologia , Humanos , Imunoprecipitação , Microscopia Confocal , Mutação , Neurônios/citologia , Neurônios/fisiologia , Fosforilação , Ratos , Bloqueadores dos Canais de Sódio/farmacologia , Tetrodotoxina/farmacologia , Transfecção , Ubiquitina/metabolismo
18.
BMC Pharmacol ; 7: 16, 2007 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-18096057

RESUMO

BACKGROUND: In addition to the regulation of blood pressure, alpha2- and beta-adrenoceptor (AR) subtypes play an important role in the modulation of noradrenergic neurotransmission in the human CNS and PNS. Several studies suggest that the alpha2-AR responsiveness in cells and tissues after chronic epinephrine (EPI) or norepinephrine (NE) exposure may vary, depending on the beta-AR activity present there. Recently, we reported that in BE(2)-C human neuroblastoma cells (endogenously expressing alpha2A- and beta2-AR), chronic EPI treatment (300 nM) produced a dramatic beta-adrenoceptor-dependent desensitization of the alpha2A-AR response. The aim of this study is to determine if stable addition of a beta2-AR to a second neuroblastoma cell line (SH-SY5Y), that normally expresses only alpha2A-ARs that are not sensitive to 300 nM EPI exposure, would suddenly render alpha2A-ARs in that cell line sensitive to treatment with the same EPI concentration. METHODS: These studies employed RT-PCR, receptor binding and inhibition of cAMP accumulation to confirm alpha2-AR subtype expression. Stable clones of SH-SY5Y cells transfected to stably express functional beta2-ARs (SHbeta2AR4) were selected to compare sensitivity of alpha2-AR to EPI in the presence or absence of beta2-ARs. RESULTS: A series of molecular, biochemical and pharmacological studies indicated that the difference between the cell lines could not be attributed to alpha2-AR heterogeneity. We now report that after transfection of functional beta2-AR into SH-SY5Y cells (SHbeta2AR4), chronic treatment with modest levels of EPI desensitizes the alpha2A-AR. This effect results from a beta2-AR dependent down-regulation of native alpha2A-ARs by EPI accompanied by enhanced translocation of GRK2 and GRK3 to the membrane (required for GRK-mediated phosphorylation of agonist-occupied receptors). CONCLUSION: This study further supports the hypothesis that the presence of the beta-AR renders the alpha2A-AR more susceptible to desensitization with physiological levels of EPI.


Assuntos
Epinefrina/farmacologia , Receptores Adrenérgicos alfa 2/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Agonistas de Receptores Adrenérgicos alfa 2 , Agonistas alfa-Adrenérgicos/farmacologia , Antagonistas de Receptores Adrenérgicos beta 2 , Tartarato de Brimonidina , Linhagem Celular Tumoral , Membrana Celular/metabolismo , AMP Cíclico/antagonistas & inibidores , AMP Cíclico/metabolismo , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Quinase 3 de Receptor Acoplado a Proteína G/metabolismo , Humanos , Norepinefrina/farmacologia , Propranolol/farmacologia , Quinoxalinas/farmacologia , RNA Mensageiro/metabolismo , Receptores Adrenérgicos alfa 2/genética , Receptores Adrenérgicos beta 2/genética , Transfecção
19.
Mol Ther ; 5(4): 455-62, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11945073

RESUMO

The anti-mitotic drugs colchicine and paclitaxel increase transfection efficiency of cationic liposomes. Using combined lipid-mediated transfection with anti-mitotic agents for gene therapy of cancer has been limited due to the likely development of multi-drug resistance (MDR). We treated human cancer cell lines and normal liver cells with glucocorticoids in combination with the antimitotics paclitaxel or colchicine before transient, cationic lipid-mediated transfection. Colchicine and paclitaxel each enhanced transgene expression in several cell lines. Moreover, glucocorticoid, combined with paclitaxel or colchicine, significantly increased reporter gene expression above that seen in cells treated with each drug alone. P-glycoprotein (PGP), a drug exporter encoded by ABCB1, exports both paclitaxel and colchicine. To determine the influence of PGP in colchicine- or paclitaxel-mediated enhancement of transgene expression, cells were treated with a histone deacetylase inhibitor, trichostatin A (TSA), known to induce ABCB1 expression, before treatment with colchicine or paclitaxel. TSA significantly reduced colchicine-mediated increases in reporter gene expression. Addition of glucocorticoid to colchicine pretreatment significantly attenuated TSA-mediated inhibition of colchicine-induced increases in transgene expression. TSA accelerated and glucocorticoid blocked export of rhodamine 123, a molecule known to be exported by PGP. The glucocorticoid/paclitaxel combination also increased reporter gene expression in BE(2)C cells, which constitutively express high levels of PGP. Thus, the degree of enhancement of transgene expression mediated by these anti-mitotics seems to be dependent on PGP activity. Glucocorticoids augment colchicine- or paclitaxel-mediated enhancement of transgene expression most likely by reducing drug egress through PGP.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/farmacologia , Transgenes , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Budesonida/farmacologia , Colchicina/farmacologia , Interações Medicamentosas , Resistência a Múltiplos Medicamentos/genética , Genes Reporter , Terapia Genética , Humanos , Ácidos Hidroxâmicos/farmacologia , Lipossomos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Paclitaxel/farmacologia , Transfecção , Células Tumorais Cultivadas , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...