Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
J Neurosci ; 41(6): 1174-1190, 2021 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-33303681

RESUMO

The BAD-BAX-caspase-3 cascade is a canonical apoptosis pathway. Macroautophagy ("autophagy" hereinafter) is a process by which organelles and aggregated proteins are delivered to lysosomes for degradation. Here, we report a new function of the BAD-BAX-caspase-3 cascade and autophagy in the control of synaptic vesicle pools. We found that, in hippocampal neurons of male mice, the BAD-BAX-caspase-3 pathway regulates autophagy, which in turn limits the size of synaptic vesicle pools and influences the kinetics of activity-induced depletion and recovery of synaptic vesicle pools. Moreover, the caspase-autophagy pathway is engaged by fear conditioning to facilitate associative fear learning and memory. This work identifies a new mechanism for controlling synaptic vesicle pools, and a novel, nonapoptotic, presynaptic function of the BAD-BAX-caspase-3 cascade.SIGNIFICANCE STATEMENT Despite the importance of synaptic vesicles for neurons, little is known about how the size of synaptic vesicle pools is maintained under basal conditions and regulated by neural activity. This study identifies a new mechanism for the control of synaptic vesicle pools, and a new, nonapoptotic function of the BAD-BAX-caspase-3 pathway in presynaptic terminals. Additionally, it indicates that autophagy is not only a homeostatic mechanism to maintain the integrity of cells and tissues, but also a process engaged by neural activity to regulate synaptic vesicle pools for optimal synaptic responses, learning, and memory.


Assuntos
Autofagia/fisiologia , Caspase 3/deficiência , Transdução de Sinais/fisiologia , Vesículas Sinápticas/metabolismo , Proteína X Associada a bcl-2/deficiência , Proteína de Morte Celular Associada a bcl/deficiência , Animais , Caspase 3/genética , Células Cultivadas , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Knockout , Imagem Molecular/métodos , Técnicas de Cultura de Órgãos , Vesículas Sinápticas/genética , Vesículas Sinápticas/ultraestrutura , Proteína X Associada a bcl-2/genética , Proteína de Morte Celular Associada a bcl/genética
2.
Neurobiol Dis ; 174: 105887, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36209950

RESUMO

We have previously reported that the single transmembrane protein Dipeptidyl Peptidase Like 6 (DPP6) impacts neuronal and synaptic development. DPP6-KO mice are impaired in hippocampal-dependent learning and memory and exhibit smaller brain size. Recently, we have described novel structures in hippocampal area CA1 in aging mice, apparently derived from degenerating presynaptic terminals, that are significantly more prevalent in DPP6-KO mice compared to WT mice of the same age and that these structures were observed earlier in development in DPP6-KO mice. These novel structures appear as clusters of large puncta that colocalize NeuN, synaptophysin, and chromogranin A, and also partially label for MAP2, amyloid ß, APP, α-synuclein, and phosphorylated tau, with synapsin-1 and VGluT1 labeling on their periphery. In this current study, using immunofluorescence and electron microscopy, we confirm that both APP and amyloid ß are prevalent in these structures; and we show with immunofluorescence the presence of similar structures in humans with Alzheimer's disease. Here we also found evidence that aging DPP6-KO mutants show additional changes related to Alzheimer's disease. We used in vivo MRI to show reduced size of the DPP6-KO brain and hippocampus. Aging DPP6-KO hippocampi contained fewer total neurons and greater neuron death and had diagnostic biomarkers of Alzheimer's disease present including accumulation of amyloid ß and APP and increase in expression of hyper-phosphorylated tau. The amyloid ß and phosphorylated tau pathologies were associated with neuroinflammation characterized by increases in microglia and astrocytes. And levels of proinflammatory or anti-inflammatory cytokines increased in aging DPP6-KO mice. We finally show that aging DPP6-KO mice display circadian dysfunction, a common symptom of Alzheimer's disease. Together these results indicate that aging DPP6-KO mice show symptoms of enhanced neurodegeneration reminiscent of dementia associated with a novel structure resulting from synapse loss and neuronal death. This study continues our laboratory's work in discerning the function of DPP6 and here provides compelling evidence of a direct role of DPP6 in Alzheimer's disease.


Assuntos
Doença de Alzheimer , Humanos , Camundongos , Animais , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Envelhecimento/patologia , Hipocampo/metabolismo , Sinapses/metabolismo , Camundongos Transgênicos , Proteínas tau/genética , Proteínas tau/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Dipeptidil Peptidases e Tripeptidil Peptidases/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Canais de Potássio/metabolismo
3.
FASEB J ; 35(1): e21092, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33378124

RESUMO

Myosin 18Aα is a myosin 2-like protein containing unique N- and C-terminal protein interaction domains that co-assembles with myosin 2. One protein known to bind to myosin 18Aα is ß-Pix, a guanine nucleotide exchange factor (GEF) for Rac1 and Cdc42 that has been shown to promote dendritic spine maturation by activating the assembly of actin and myosin filaments in spines. Here, we show that myosin 18A⍺ concentrates in the spines of cerebellar Purkinje neurons via co-assembly with myosin 2 and through an actin binding site in its N-terminal extension. miRNA-mediated knockdown of myosin 18A⍺ results in a significant defect in spine maturation that is rescued by an RNAi-immune version of myosin 18A⍺. Importantly, ß-Pix co-localizes with myosin 18A⍺ in spines, and its spine localization is lost upon myosin 18A⍺ knockdown or when its myosin 18A⍺ binding site is deleted. Finally, we show that the spines of myosin 18A⍺ knockdown Purkinje neurons contain significantly less F-actin and myosin 2. Together, these data argue that mixed filaments of myosin 2 and myosin 18A⍺ form a complex with ß-Pix in Purkinje neuron spines that promotes spine maturation by enhancing the assembly of actin and myosin filaments downstream of ß-Pix's GEF activity.


Assuntos
Espinhas Dendríticas/metabolismo , Miosinas/metabolismo , Células de Purkinje/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Animais , Espinhas Dendríticas/genética , Deleção de Genes , Camundongos , Miosina Tipo II/genética , Miosina Tipo II/metabolismo , Miosinas/genética , Fatores de Troca de Nucleotídeo Guanina Rho/genética
4.
Mol Psychiatry ; 26(9): 4633-4651, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33589740

RESUMO

Mitochondria are cellular ATP generators. They are dynamic structures undergoing fission and fusion. While much is known about the mitochondrial fission machinery, the mechanism of initiating fission and the significance of fission to neurophysiology are largely unclear. Gamma oscillations are synchronized neural activities that impose a great energy challenge to synapses. The cellular mechanism of fueling gamma oscillations has yet to be defined. Here, we show that dysbindin-1, a protein decreased in the brain of individuals with schizophrenia, is required for neural activity-induced fission by promoting Drp1 oligomerization. This process is engaged by gamma-frequency activities and in turn, supports gamma oscillations. Gamma oscillations and novel object recognition are impaired in dysbindin-1 null mice. These defects can be ameliorated by increasing mitochondrial fission. These findings identify a molecular mechanism for activity-induced mitochondrial fission, a role of mitochondrial fission in gamma oscillations, and mitochondrial fission as a potential target for improving cognitive functions.


Assuntos
Mitocôndrias , Dinâmica Mitocondrial , Animais , Dinaminas , Disbindina , Camundongos , Camundongos Knockout , Proteínas Mitocondriais , Sinapses
5.
Biophys J ; 120(11): 2085-2101, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33812847

RESUMO

Neural function depends on continual synthesis and targeted trafficking of intracellular components, including ion channel proteins. Many kinds of ion channels are trafficked over long distances to specific cellular compartments. This raises the question of whether cargo is directed with high specificity during transit or whether cargo is distributed widely and sequestered at specific sites. We addressed this question by experimentally measuring transport and expression densities of Kv4.2, a voltage-gated transient potassium channel that exhibits a specific dendritic expression that increases with distance from the soma and little or no functional expression in axons. In over 500 h of quantitative live imaging, we found substantially higher densities of actively transported Kv4.2 subunits in axons as opposed to dendrites. This paradoxical relationship between functional expression and traffic density supports a model-commonly known as the sushi belt model-in which trafficking specificity is relatively low and active sequestration occurs in compartments where cargo is expressed. In further support of this model, we find that kinetics of active transport differs qualitatively between axons and dendrites, with axons exhibiting strong superdiffusivity, whereas dendritic transport resembles a weakly directed random walk, promoting mixing and opportunity for sequestration. Finally, we use our data to constrain a compartmental reaction-diffusion model that can recapitulate the known Kv4.2 density profile. Together, our results show how nontrivial expression patterns can be maintained over long distances with a relatively simple trafficking mechanism and how the hallmarks of a global trafficking mechanism can be revealed in the kinetics and density of cargo.


Assuntos
Dendritos , Canais de Potássio Shal , Axônios/metabolismo , Transporte Biológico Ativo , Dendritos/metabolismo , Neurônios/metabolismo , Transporte Proteico , Canais de Potássio Shal/metabolismo
6.
Hum Mol Genet ; 28(9): 1530-1547, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30602030

RESUMO

Epilepsy, deafness, onychodystrophy, osteodystrophy and intellectual disability are associated with a spectrum of mutations of human TBC1D24. The mechanisms underlying TBC1D24-associated disorders and the functions of TBC1D24 are not well understood. Using CRISPR-Cas9 genome editing, we engineered a mouse with a premature translation stop codon equivalent to human S324Tfs*3, a recessive mutation of TBC1D24 associated with early infantile epileptic encephalopathy (EIEE). Homozygous S324Tfs*3 mice have normal auditory and vestibular functions but show an abrupt onset of spontaneous seizures at postnatal day 15 recapitulating human EIEE. The S324Tfs*3 variant is located in an alternatively spliced micro-exon encoding six perfectly conserved amino acids incorporated postnatally into TBC1D24 protein due to a micro-exon utilization switch. During embryonic and early postnatal development, S324Tfs*3 homozygotes produce predominantly the shorter wild-type TBC1D24 protein isoform that omits the micro-exon. S324Tfs*3 homozygotes show an abrupt onset of seizures at P15 that correlates with a developmental switch to utilization of the micro-exon. A mouse deficient for alternative splice factor SRRM3 impairs incorporation of the Tbc1d24 micro-exon. Wild-type Tbc1d24 mRNA is abundantly expressed in the hippocampus using RNAscope in situ hybridization. Immunogold electron microscopy using a TBC1D24-specific antibody revealed that TBC1D24 is associated with clathrin-coated vesicles and synapses of hippocampal neurons, suggesting a crucial role of TBC1D24 in vesicle trafficking important for neuronal signal transmission. This is the first characterization of a mouse model of human TBC1D24-associated EIEE that can now be used to screen for antiepileptogenic drugs ameliorating TBCID24 seizure disorders.


Assuntos
Proteínas Ativadoras de GTPase/genética , Estudos de Associação Genética , Predisposição Genética para Doença , Mutação , Fenótipo , Espasmos Infantis/diagnóstico , Espasmos Infantis/genética , Alelos , Animais , Biomarcadores , Encéfalo/metabolismo , Análise Mutacional de DNA , Proteínas Ativadoras de GTPase/metabolismo , Expressão Gênica , Loci Gênicos , Humanos , Masculino , Camundongos , Neurônios/metabolismo , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
7.
J Biol Chem ; 294(30): 11498-11512, 2019 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-31177092

RESUMO

Neurolastin is a dynamin family GTPase that also contains a RING domain and exhibits both GTPase and E3 ligase activities. It is specifically expressed in the brain and is important for synaptic transmission, as neurolastin knockout animals have fewer dendritic spines and exhibit a reduction in functional synapses. Our initial study of neurolastin revealed that it is membrane-associated and partially co-localizes with endosomes. Using various biochemical and cell-culture approaches, we now show that neurolastin also localizes to mitochondria in HeLa cells, cultured neurons, and brain tissue. We found that the mitochondrial localization of neurolastin depends upon an N-terminal mitochondrial targeting sequence and that neurolastin is imported into the mitochondrial intermembrane space. Although neurolastin was only partially mitochondrially localized at steady state, it displayed increased translocation to mitochondria in response to neuronal stress and mitochondrial fragmentation. Interestingly, inactivation or deletion of neurolastin's RING domain also increased its mitochondrial localization. Using EM, we observed that neurolastin knockout animals have smaller but more numerous mitochondria in cerebellar Purkinje neurons, indicating that neurolastin regulates mitochondrial morphology. We conclude that the brain-specific dynamin GTPase neurolastin exhibits stress-responsive localization to mitochondria and is required for proper mitochondrial morphology.


Assuntos
Dinaminas/metabolismo , Mitocôndrias/metabolismo , Células de Purkinje/metabolismo , Animais , Células Cultivadas , Dinaminas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/enzimologia , Mutação , Transporte Proteico
8.
Glia ; 67(1): 78-90, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30306657

RESUMO

Of the seven P2X receptor subtypes, P2X4 receptor (P2X4R) is widely distributed in the central nervous system, including in neurons, astrocytes, and microglia. Accumulating evidence supports roles for P2X4R in the central nervous system, including regulating cell excitability, synaptic transmission, and neuropathic pain. However, little information is available about the distribution and function of P2X4R in the peripheral nervous system. In this study, we find that P2X4R is mainly localized in the lysosomes of Schwann cells in the peripheral nervous system. In cultured Schwann cells, TNF-a not only enhances the synthesis of P2X4R protein but also promotes P2X4R trafficking to the surface of Schwann cells. TNF-a-induced BDNF secretion in Schwann cells is P2X4R dependent. in vivo experiments reveal that expression of P2X4R in Schwann cells of injured nerves is strikingly upregulated following nerve crush injury. Moreover, overexpression of P2X4R in Schwann cells by genetic manipulation promotes motor and sensory functional recovery and accelerates nerve remyelination via BDNF release following nerve injury. Our results suggest that enhancement of P2X4R expression in Schwann cells after nerve injury may be an effective approach to facilitate the regrowth and remyelination of injured nerves.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Traumatismos dos Nervos Periféricos/metabolismo , Receptores Purinérgicos P2X4/biossíntese , Recuperação de Função Fisiológica/fisiologia , Remielinização/fisiologia , Células de Schwann/metabolismo , Animais , Animais Recém-Nascidos , Fator Neurotrófico Derivado do Encéfalo/agonistas , Células Cultivadas , Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Traumatismos dos Nervos Periféricos/patologia , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P2X4/genética , Recuperação de Função Fisiológica/efeitos dos fármacos , Remielinização/efeitos dos fármacos , Células de Schwann/efeitos dos fármacos , Células de Schwann/patologia , Fator de Necrose Tumoral alfa/toxicidade
9.
J Neurosci ; 37(26): 6299-6313, 2017 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-28546313

RESUMO

In sensory hair cells of auditory and vestibular organs, the ribbon synapse is required for the precise encoding of a wide range of complex stimuli. Hair cells have a unique presynaptic structure, the synaptic ribbon, which organizes both synaptic vesicles and calcium channels at the active zone. Previous work has shown that hair-cell ribbon size is correlated with differences in postsynaptic activity. However, additional variability in postsynapse size presents a challenge to determining the specific role of ribbon size in sensory encoding. To selectively assess the impact of ribbon size on synapse function, we examined hair cells in transgenic zebrafish that have enlarged ribbons, without postsynaptic alterations. Morphologically, we found that enlarged ribbons had more associated vesicles and reduced presynaptic calcium-channel clustering. Functionally, hair cells with enlarged ribbons had larger global and ribbon-localized calcium currents. Afferent neuron recordings revealed that hair cells with enlarged ribbons resulted in reduced spontaneous spike rates. Additionally, despite larger presynaptic calcium signals, we observed fewer evoked spikes with longer latencies from stimulus onset. Together, our work indicates that hair-cell ribbon size influences the spontaneous spiking and the precise encoding of stimulus onset in afferent neurons.SIGNIFICANCE STATEMENT Numerous studies support that hair-cell ribbon size corresponds with functional sensitivity differences in afferent neurons and, in the case of inner hair cells of the cochlea, vulnerability to damage from noise trauma. Yet it is unclear whether ribbon size directly influences sensory encoding. Our study reveals that ribbon enlargement results in increased ribbon-localized calcium signals, yet reduces afferent spontaneous activity and disrupts the timing of stimulus onset, a distinct aspect of auditory and vestibular encoding. These observations suggest that varying ribbon size alone can influence sensory encoding, and give further insight into how hair cells transduce signals that cover a wide dynamic range of stimuli.


Assuntos
Potenciais de Ação/fisiologia , Sinalização do Cálcio/fisiologia , Mecanorreceptores/citologia , Mecanorreceptores/fisiologia , Tempo de Reação/fisiologia , Peixe-Zebra/fisiologia , Animais , Animais Geneticamente Modificados , Tamanho Celular , Sistema da Linha Lateral/citologia , Sistema da Linha Lateral/fisiologia , Inibição Neural/fisiologia , Peixe-Zebra/anatomia & histologia
10.
Development ; 142(3): 555-66, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25605782

RESUMO

Primary cilia have been implicated in the generation of planar cell polarity (PCP). However, variations in the severity of polarity defects in different cilia mutants, coupled with recent demonstrations of non-cilia-related actions of some cilia genes, make it difficult to determine the basis of these polarity defects. To address this issue, we evaluated PCP defects in cochlea from a selection of mice with mutations in cilia-related genes. Results indicated notable PCP defects, including mis-oriented hair cell stereociliary bundles, in Bbs8 and Ift20 single mutants that are more severe than in other cilia gene knockouts. In addition, deletion of either Bbs8 or Ift20 results in disruptions in asymmetric accumulation of the core PCP molecule Vangl2 in cochlear cells, suggesting a role for Bbs8 and/or Ift20, possibly upstream of core PCP asymmetry. Consistent with this, co-immunoprecipitation experiments indicate direct interactions of Bbs8 and Ift20 with Vangl2. We observed localization of Bbs and Ift proteins to filamentous actin as well as microtubules. This could implicate these molecules in selective trafficking of membrane proteins upstream of cytoskeletal reorganization, and identifies new roles for cilia-related proteins in cochlear PCP.


Assuntos
Proteínas de Transporte/metabolismo , Polaridade Celular/fisiologia , Cílios/genética , Cóclea/embriologia , Proteínas Associadas aos Microtúbulos/metabolismo , Animais , Cílios/fisiologia , Cílios/ultraestrutura , Cóclea/ultraestrutura , Proteínas do Citoesqueleto , Células Ciliadas Auditivas/patologia , Imuno-Histoquímica , Imunoprecipitação , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Proteínas do Tecido Nervoso
11.
J Neurosci ; 35(49): 16126-41, 2015 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-26658865

RESUMO

The presence of Sonic Hedgehog (Shh) and its signaling components in the neurons of the hippocampus raises a question about what role the Shh signaling pathway may play in these neurons. We show here that activation of the Shh signaling pathway stimulates axon elongation in rat hippocampal neurons. This Shh-induced effect depends on the pathway transducer Smoothened (Smo) and the transcription factor Gli1. The axon itself does not respond directly to Shh; instead, the Shh signal transduction originates from the somatodendritic region of the neurons and occurs in neurons with and without detectable primary cilia. Upon Shh stimulation, Smo localization to dendrites increases significantly. Shh pathway activation results in increased levels of profilin1 (Pfn1), an actin-binding protein. Mutations in Pfn1's actin-binding sites or reduction of Pfn1 eliminate the Shh-induced axon elongation. These findings indicate that Shh can regulate axon growth, which may be critical for development of hippocampal neurons. SIGNIFICANCE STATEMENT: Although numerous signaling mechanisms have been identified that act directly on axons to regulate their outgrowth, it is not known whether signals transduced in dendrites may also affect axon outgrowth. We describe here a transcellular signaling pathway in embryonic hippocampal neurons in which activation of Sonic Hedgehog (Shh) receptors in dendrites stimulates axon growth. The pathway involves the dendritic-membrane-associated Shh signal transducer Smoothened (Smo) and the transcription factor Gli, which induces the expression of the gene encoding the actin-binding protein profilin 1. Our findings suggest scenarios in which stimulation of Shh in dendrites results in accelerated outgrowth of the axon, which therefore reaches its presumptive postsynaptic target cell more quickly. By this mechanism, Shh may play critical roles in the development of hippocampal neuronal circuits.


Assuntos
Axônios/fisiologia , Dendritos/metabolismo , Proteínas Hedgehog/metabolismo , Hipocampo/citologia , Neurônios/citologia , Transdução de Sinais/fisiologia , Animais , Axônios/ultraestrutura , Células Cultivadas , Dendritos/efeitos dos fármacos , Dendritos/ultraestrutura , Embrião de Mamíferos , Regulação da Expressão Gênica/genética , Proteínas Hedgehog/genética , Proteínas Hedgehog/farmacologia , Humanos , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Neurônios/efeitos dos fármacos , Profilinas/genética , Profilinas/metabolismo , Transporte Proteico/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/genética , Receptor Smoothened , Fatores de Tempo , Proteína GLI1 em Dedos de Zinco
12.
J Biol Chem ; 290(30): 18379-90, 2015 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-26045554

RESUMO

NMDA receptors (NMDARs) comprise a subclass of neurotransmitter receptors whose surface expression is regulated at multiple levels, including processing in the endoplasmic reticulum (ER), intracellular trafficking via the Golgi apparatus, internalization, recycling, and degradation. With respect to early processing, NMDARs are regulated by the availability of GluN subunits within the ER, the presence of ER retention and export signals, and posttranslational modifications, including phosphorylation and palmitoylation. However, the role of N-glycosylation, one of the most common posttranslational modifications, in regulating NMDAR processing has not been studied in detail. Using biochemistry, confocal and electron microscopy, and electrophysiology in conjunction with a lentivirus-based molecular replacement strategy, we found that NMDARs are released from the ER only when two asparagine residues in the GluN1 subunit (Asn-203 and Asn-368) are N-glycosylated. Although the GluN2A and GluN2B subunits are also N-glycosylated, their N-glycosylation sites do not appear to be essential for surface delivery of NMDARs. Furthermore, we found that removing N-glycans from native NMDARs altered the receptor affinity for glutamate. Our results suggest a novel mechanism by which neurons ensure that postsynaptic membranes contain sufficient numbers of functional NMDARs.


Assuntos
N-Metilaspartato/metabolismo , Neurônios/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transmissão Sináptica , Animais , Células COS , Chlorocebus aethiops , Retículo Endoplasmático/metabolismo , Glicosilação , Complexo de Golgi/metabolismo , Células HEK293 , Humanos , N-Metilaspartato/química , Neurônios/química , Polissacarídeos/metabolismo , Ratos , Receptores de N-Metil-D-Aspartato/química , Sinapses/metabolismo
13.
J Neurosci ; 34(15): 5261-72, 2014 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-24719104

RESUMO

Acetylcholine is a neuromodulatory transmitter that controls synaptic plasticity and sensory processing in many brain regions. The dorsal cochlear nucleus (DCN) is an auditory brainstem nucleus that integrates auditory signals from the cochlea with multisensory inputs from several brainstem nuclei and receives prominent cholinergic projections. In the auditory periphery, cholinergic modulation serves a neuroprotective function, reducing cochlear output under high sound levels. However, the role of cholinergic signaling in the DCN is less understood. Here we examine postsynaptic mechanisms of cholinergic modulation at glutamatergic synapses formed by parallel fiber axons onto cartwheel cells (CWCs) in the apical DCN circuit from mouse brainstem slice using calcium (Ca) imaging combined with two-photon laser glutamate uncaging onto CWC spines. Activation of muscarinic acetylcholine receptors (mAChRs) significantly increased the amplitude of both uncaging-evoked EPSPs (uEPSPs) and spine Ca transients. Our results demonstrate that mAChRs in CWC spines act by suppressing large-conductance calcium-activated potassium (BK) channels, and this effect is mediated through the cAMP/protein kinase A signaling pathway. Blocking BK channels relieves voltage-dependent magnesium block of NMDA receptors, thereby enhancing uEPSPs and spine Ca transients. Finally, we demonstrate that mAChR activation inhibits L-type Ca channels and thus may contribute to the suppression of BK channels by mAChRs. In summary, we demonstrate a novel role for BK channels in regulating glutamatergic transmission and show that this mechanism is under modulatory control of mAChRs.


Assuntos
Cálcio/metabolismo , Neurônios Colinérgicos/metabolismo , Núcleo Coclear/fisiologia , Espinhas Dendríticas/metabolismo , Potenciais Pós-Sinápticos Excitadores , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Animais , Canais de Cálcio Tipo L/metabolismo , Sinalização do Cálcio , Neurônios Colinérgicos/fisiologia , Núcleo Coclear/citologia , Núcleo Coclear/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Espinhas Dendríticas/fisiologia , Ácido Glutâmico/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos CBA , Bloqueadores dos Canais de Potássio/farmacologia , Receptores Muscarínicos/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/metabolismo , Sinapses/fisiologia
14.
J Cell Sci ; 125(Pt 18): 4207-13, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22641692

RESUMO

Sonic hedgehog (Shh) signaling is essential to the patterning of the embryonic neural tube, but its presence and function in the postmitotic differentiated neurons in the brain remain largely uncharacterized. We recently showed that Shh and its signaling components, Patched and Smoothened, are expressed in postnatal and adult hippocampal neurons. We have now examined whether Shh signaling has a function in these neurons. Using cultured hippocampal neurons as a model system, we found that presynaptic terminals become significantly larger in response to the application of Shh. Ultrastructural examination confirmed the enlarged presynaptic profiles and also revealed variable increases in the size of synaptic vesicles, with a resulting loss of uniformity. Furthermore, electrophysiological analyses showed significant increases in the frequency, but not the amplitude, of spontaneous miniature excitatory postsynaptic currents (mEPSCs) in response to Shh, providing functional evidence of the selective role of Shh in presynaptic terminals. Thus, we conclude that Shh signaling regulates the structure and functional properties of presynaptic terminals of hippocampal neurons.


Assuntos
Proteínas Hedgehog/metabolismo , Hipocampo/citologia , Terminações Pré-Sinápticas/fisiologia , Terminações Pré-Sinápticas/ultraestrutura , Animais , Células HEK293 , Humanos , Neurotransmissores/metabolismo , Tamanho do Órgão , Terminações Pré-Sinápticas/metabolismo , Ratos , Transdução de Sinais , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/ultraestrutura
15.
Autophagy ; : 1-22, 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38899385

RESUMO

In neurons, macroautophagy/autophagy is a frequent and critical process. In the axon, autophagy begins in the axon terminal, where most nascent autophagosomes form. After formation, autophagosomes must initiate transport to exit the axon terminal and move toward the cell body via retrograde transport. During retrograde transport these autophagosomes mature through repetitive fusion events. Complete lysosomal cargo degradation occurs largely in the cell body. The precipitating events to stimulate retrograde autophagosome transport have been debated but their importance is clear: disrupting neuronal autophagy or autophagosome transport is detrimental to neuronal health and function. We have identified the HOPS complex as essential for early autophagosome maturation and consequent initiation of retrograde transport from the axon terminal. In yeast and mammalian cells, HOPS controls fusion between autophagosomes and late endosomes with lysosomes. Using zebrafish strains with loss-of-function mutations in vps18 and vps41, core components of the HOPS complex, we found that disruption of HOPS eliminates autophagosome maturation and disrupts retrograde autophagosome transport initiation from the axon terminal. We confirmed this phenotype was due to loss of HOPS complex formation using an endogenous deletion of the HOPS binding domain in Vps18. Finally, using pharmacological inhibition of lysosomal proteases, we show that initiation of autophagosome retrograde transport requires autophagosome maturation. Together, our data demonstrate that HOPS-mediated fusion events are critical for retrograde autophagosome transport initiation through promoting autophagosome maturation. This reveals critical roles for the HOPS complex in neuronal autophagy which deepens our understanding of the cellular pathology of HOPS-complex linked neurodegenerative diseases.Abbreviations: CORVET: Class C core vacuole/endosome tethering; gRNA: guide RNA; HOPS: homotypic fusion and protein sorting; pLL: posterior lateral line; Vps18: VPS18 core subunit of CORVET and HOPS complexes; Vps41: VPS41 subunit of HOPS complex.

16.
J Biol Chem ; 287(7): 4470-84, 2012 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-22174418

RESUMO

Synaptic adhesion-like molecules (SALMs) are a family of cell adhesion molecules involved in neurite outgrowth and synapse formation. Of the five family members, only SALM1, -2, and -3 contain a cytoplasmic C-terminal PDZ-binding motif. We have found that SALM1 is unique among the SALMs because deletion of its PDZ-binding motif (SALM1ΔPDZ) blocks its surface expression in heterologous cells. When expressed in hippocampal neurons, SALM1ΔPDZ had decreased surface expression in dendrites and the cell soma but not in axons, suggesting that the PDZ-binding domain may influence cellular trafficking of SALMs to specific neuronal locations. Endoglycosidase H digestion assays indicated that SALM1ΔPDZ is retained in the endoplasmic reticulum (ER) in heterologous cells. However, when the entire C-terminal tail of SALM1 was deleted, SALM1 was detected on the cell surface. Using serial deletions, we identified a region of SALM1 that contains a putative dileucine ER retention motif, which is not present in the other SALMs. Mutation of this DXXXLL motif allowed SALM1 to leave the ER and enhanced its surface expression in heterologous cells and neurons. An increase in the number of protrusions at the dendrites and cell body was observed when this SALM1 mutant was expressed in hippocampal neurons. With electron microscopy, these protrusions appeared to be irregular, enlarged spines and filopodia. Thus, enrichment of SALM1 on the cell surface affects dendritic arborization, and intracellular motifs regulate its dendritic versus axonal localization.


Assuntos
Axônios/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Dendritos/metabolismo , Hipocampo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Axônios/ultraestrutura , Moléculas de Adesão Celular Neuronais/genética , Dendritos/ultraestrutura , Células HeLa , Hipocampo/citologia , Humanos , Proteínas do Tecido Nervoso/genética , Domínios PDZ , Transporte Proteico/fisiologia , Deleção de Sequência
17.
J Neurosci ; 31(25): 9289-97, 2011 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-21697378

RESUMO

NMDA receptors (NMDARs) are critical to the development of the nervous system, although their roles at axonal growth cones are unclear. We examined NMDAR localization and function at axonal growth cones of young hippocampal neurons. Our immunocytochemical data showed that native and transfected NMDAR subunits are expressed in axons and growth cones of young (days in vitro 3-6) hippocampal rat neurons. Moreover, immunogold electron microscopy showed that NR1 is expressed in growth cones of postnatal day 2 rat hippocampus. Local application of NMDAR agonists to growth cones of voltage-clamped neurons evoked inward currents that were blocked by bath application of an NMDAR antagonist (dl-APV), indicating that these NMDARs are functional. In addition, calcium imaging experiments indicated that NMDARs present in growth cones mediate calcium influx. Calcium transients in growth cones persisted despite pharmacological blockade of voltage-sensitive calcium channels and depletion of intracellular calcium stores. Our findings reveal the presence of functional NMDARs in axons and growth cones of young neurons, suggesting a role for these receptors in axonal guidance and synapse formation during neuronal development.


Assuntos
Cones de Crescimento/fisiologia , Hipocampo/citologia , Hipocampo/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Axônios/fisiologia , Crescimento Celular , Células Cultivadas , Ratos
18.
Nat Cell Biol ; 7(12): 1179-90, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16299499

RESUMO

Appropriate trafficking and targeting of glutamate receptors (GluRs) to the postsynaptic density is crucial for synaptic function. We show that mPins (mammalian homologue of Drosophila melanogaster partner of inscuteable) interacts with SAP102 and PSD-95 (two PDZ proteins present in neurons), and functions in the formation of the NMDAR-MAGUK (N-methyl-D-aspartate receptor-membrane-associated guanylate kinase) complex. mPins enhances trafficking of SAP102 and NMDARs to the plasma membrane in neurons. Expression of dominant-negative constructs and short-interfering RNA (siRNA)-mediated knockdown of mPins decreases SAP102 in dendrites and modifies surface expression of NMDARs. mPins changes the number and morphology of dendritic spines and these effects depend on its Galphai interaction domain, thus implicating G-protein signalling in the regulation of postsynaptic structure and trafficking of GluRs.


Assuntos
Proteínas de Transporte/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Neuropeptídeos/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Proteínas de Ciclo Celular , Membrana Celular , Dendritos/química , Proteína 4 Homóloga a Disks-Large , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Guanilato Quinases/metabolismo , Imunoprecipitação , Camundongos , Neurônios , Transporte Proteico , Ratos , Transfecção , Técnicas do Sistema de Duplo-Híbrido
19.
Cerebellum ; 11(4): 972-81, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22477363

RESUMO

The Sonic hedgehog (Shh) signaling pathway carries out a wide range of biological functions such as patterning of the embryonic neural tube and expansion of cerebellar granule cell precursors. We previously have found that the Shh signaling receptors, Patched1 (Ptch1) and Smoothened (Smo), are expressed in hippocampal neurons of developing and adult rats, suggesting the continued presence of Shh signaling in postmitotic, differentiated neurons. Here, we report that Ptch1 and Smo are present in the processes and growth cones of immature neurons in the developing cerebellum, and that, in the mature cerebellum, Ptch1 and Smo are expressed by several types of neurons including Purkinje cells, granule cells, and interneurons. Within these neurons, Ptch1 and Smo are predominantly localized in the postsynaptic side of the synapses, a distribution pattern similar to that found in hippocampal neurons. Our findings provide morphological evidence that Shh signaling events are not confined to neuronal precursors and are likely to have ongoing roles within the postmitotic neurons of the developing and adult cerebellum.


Assuntos
Proteínas Hedgehog/metabolismo , Neurônios/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sinapses/ultraestrutura , Animais , Linhagem Celular , Sistema Nervoso Central/metabolismo , Cerebelo/metabolismo , Espaço Intracelular/metabolismo , Masculino , Camundongos , Receptores Patched , Receptor Patched-1 , Células de Purkinje/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Superfície Celular/deficiência , Transdução de Sinais/fisiologia , Receptor Smoothened
20.
STAR Protoc ; 3(1): 101154, 2022 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-35169716

RESUMO

Electron microscopy (EM) is considered the gold standard for studying macroautophagy and mitophagy, essential cellular processes for brain health. Here, we present a protocol using EM to analyze autophagosomes and mitophagosomes in the mouse amygdala. We describe the preparation of brain sections, followed by staining and EM imaging. We then detail the steps to identify and analyze autophagosome-like and mitophagosome-like structures. This protocol can be easily adapted to analyze autophagosomes and mitophagosomes in other mouse brain regions. For complete details on the use and execution of this protocol, please refer to Duan et al. (2021).


Assuntos
Autofagossomos , Mitofagia , Animais , Encéfalo/diagnóstico por imagem , Camundongos , Microscopia Eletrônica , Coloração e Rotulagem
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa