Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
eNeuro ; 11(6)2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38777611

RESUMO

Homeostatic plasticity stabilizes firing rates of neurons, but the pressure to restore low activity rates can significantly alter synaptic and cellular properties. Most previous studies of homeostatic readjustment to complete activity silencing in rodent forebrain have examined changes after 2 d of deprivation, but it is known that longer periods of deprivation can produce adverse effects. To better understand the mechanisms underlying these effects and to address how presynaptic as well as postsynaptic compartments change during homeostatic plasticity, we subjected mouse cortical slice cultures to a more severe 5 d deprivation paradigm. We developed and validated a computational framework to measure the number and morphology of presynaptic and postsynaptic compartments from super-resolution light microscopy images of dense cortical tissue. Using these tools, combined with electrophysiological miniature excitatory postsynaptic current measurements, and synaptic imaging at the electron microscopy level, we assessed the functional and morphological results of prolonged deprivation. Excitatory synapses were strengthened both presynaptically and postsynaptically. Surprisingly, we also observed a decrement in the density of excitatory synapses, both as measured from colocalized staining of pre- and postsynaptic proteins in tissue and from the number of dendritic spines. Overall, our results suggest that cortical networks deprived of activity progressively move toward a smaller population of stronger synapses.


Assuntos
Potenciais Pós-Sinápticos Excitadores , Neocórtex , Plasticidade Neuronal , Sinapses , Animais , Plasticidade Neuronal/fisiologia , Sinapses/fisiologia , Neocórtex/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Camundongos Endogâmicos C57BL , Privação Sensorial/fisiologia , Masculino , Camundongos , Feminino , Espinhas Dendríticas/fisiologia
2.
bioRxiv ; 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38659950

RESUMO

Voltage imaging enables high-throughput investigation of neuronal activity, yet its utility is often constrained by a low signal-to-noise ratio (SNR). Conventional denoising algorithms, such as those based on matrix factorization, impose limiting assumptions about the noise process and the spatiotemporal structure of the signal. While deep learning based denoising techniques offer greater adaptability, existing approaches fail to fully exploit the fast temporal dynamics and unique short- and long-range dependencies within voltage imaging datasets. Here, we introduce CellMincer, a novel self-supervised deep learning method designed specifically for denoising voltage imaging datasets. CellMincer operates on the principle of masking and predicting sparse sets of pixels across short temporal windows and conditions the denoiser on precomputed spatiotemporal auto-correlations to effectively model long-range dependencies without the need for large temporal denoising contexts. We develop and utilize a physics-based simulation framework to generate realistic datasets for rigorous hyperparameter optimization and ablation studies, highlighting the key role of conditioning the denoiser on precomputed spatiotemporal auto-correlations to achieve 3-fold further reduction in noise. Comprehensive benchmarking on both simulated and real voltage imaging datasets, including those with paired patch-clamp electrophysiology (EP) as ground truth, demonstrates CellMincer's state-of-the-art performance. It achieves substantial noise reduction across the entire frequency spectrum, enhanced detection of subthreshold events, and superior cross-correlation with ground-truth EP recordings. Finally, we demonstrate how CellMincer's addition to a typical voltage imaging data analysis workflow improves neuronal segmentation, peak detection, and ultimately leads to significantly enhanced separation of functional phenotypes.

3.
bioRxiv ; 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-38076932

RESUMO

Pooled optical screens have enabled the study of cellular interactions, morphology, or dynamics at massive scale, but have not yet leveraged the power of highly-plexed single-cell resolved transcriptomic readouts to inform molecular pathways. Here, we present Perturb-FISH, which bridges these approaches by combining imaging spatial transcriptomics with parallel optical detection of in situ amplified guide RNAs. We show that Perturb-FISH recovers intracellular effects that are consistent with Perturb-seq results in a screen of lipopolysaccharide response in cultured monocytes, and uncover new intercellular and density-dependent regulation of the innate immune response. We further pair Perturb-FISH with a functional readout in a screen of autism spectrum disorder risk genes, showing common calcium activity phenotypes in induced pluripotent stem cell derived astrocytes and their associated genetic interactions and dysregulated molecular pathways. Perturb-FISH is thus a generally applicable method for studying the genetic and molecular associations of spatial and functional biology at single-cell resolution.

4.
iScience ; 26(7): 106995, 2023 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-37534135

RESUMO

Emerging evidence of species divergent features of astrocytes coupled with the relative inaccessibility of human brain tissue underscore the utility of human pluripotent stem cell (hPSC) technologies for the generation and study of human astrocytes. However, existing approaches for hPSC-astrocyte generation are typically lengthy or require intermediate purification steps. Here, we establish a rapid and highly scalable method for generating functional human induced astrocytes (hiAs). These hiAs express canonical astrocyte markers, respond to pro-inflammatory stimuli, exhibit ATP-induced calcium transients and support neuronal network development. Moreover, single-cell transcriptomic analyses reveal the generation of highly reproducible cell populations across individual donors, mostly resembling human fetal astrocytes. Finally, hiAs generated from a trisomy 21 disease model identify expected alterations in cell-cell adhesion and synaptic signaling, supporting their utility for disease modeling applications. Thus, hiAs provide a valuable and practical resource for the study of basic human astrocyte function and dysfunction in disease.

5.
Elife ; 122023 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-36749029

RESUMO

Healthy neuronal networks rely on homeostatic plasticity to maintain stable firing rates despite changing synaptic drive. These mechanisms, however, can themselves be destabilizing if activated inappropriately or excessively. For example, prolonged activity deprivation can lead to rebound hyperactivity and seizures. While many forms of homeostasis have been described, whether and how the magnitude of homeostatic plasticity is constrained remains unknown. Here, we uncover negative regulation of cortical network homeostasis by the PARbZIP family of transcription factors. In cortical slice cultures made from knockout mice lacking all three of these factors, the network response to prolonged activity withdrawal measured with calcium imaging is much stronger, while baseline activity is unchanged. Whole-cell recordings reveal an exaggerated increase in the frequency of miniature excitatory synaptic currents reflecting enhanced upregulation of recurrent excitatory synaptic transmission. Genetic analyses reveal that two of the factors, Hlf and Tef, are critical for constraining plasticity and for preventing life-threatening seizures. These data indicate that transcriptional activation is not only required for many forms of homeostatic plasticity but is also involved in restraint of the response to activity deprivation.


The human brain is made up of billions of nerve cells called neurons which receive and send signals to one another. To avoid being over- or under-stimulated, neurons can adjust the strength of the inputs they receive by altering how connected they are to other nerve cells. This process, known as homeostatic plasticity, is thought to be necessary for normal brain activity as it helps keep the outgoing signals of neurons relatively constant. However, homeostatic plasticity can lead to seizures if it becomes too strong and overcompensates for weak input signals. Regulating this process is therefore central to brain health, but scientists do not understand if or how it is controlled. To address this, Valakh et al. analyzed the genes activated in neurons lacking incoming signals to find proteins that regulate homeostatic plasticity. This revealed a class of molecules called transcription factors (which switch genes on or off) that constrain the process. In brain samples from mice without these regulatory proteins, neurons received twice as much input, leading to an increase in brain activity resembling that observed during seizures. Valakh et al. confirmed this finding using live mice, which developed seizures in the absence of these transcription factors. These findings suggest that this type of regulation to keep homeostatic plasticity from becoming too strong may be important. This could be especially vital as the brain develops, when the strength of connections between neurons changes rapidly. The discovery of the transcription factors involved provides a potential target for activating or restraining homeostatic plasticity. This control could help researchers better understand how the process stabilizes brain signaling.


Assuntos
Neocórtex , Plasticidade Neuronal , Camundongos , Animais , Plasticidade Neuronal/fisiologia , Transmissão Sináptica/fisiologia , Homeostase/fisiologia , Camundongos Knockout , Convulsões/genética , Sinapses/fisiologia , Mamíferos
6.
Cell Rep ; 42(1): 111988, 2023 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-36640364

RESUMO

The maturation of neurons and the development of synapses, although emblematic of neurons, also relies on interactions with astrocytes and other glia. Here, to study the role of glia-neuron interactions, we analyze the transcriptomes of human pluripotent stem cell (hPSC)-derived neurons, from 80 human donors, that were cultured with or without contact with glial cells. We find that the presence of astrocytes enhances synaptic gene-expression programs in neurons when in physical contact with astrocytes. These changes in neurons correlate with increased expression, in the cocultured glia, of genes that encode synaptic cell adhesion molecules. Both the neuronal and astrocyte gene-expression programs are enriched for genes associated with schizophrenia risk. Our results suggest that astrocyte-expressed genes with synaptic functions are associated with stronger expression of synaptic genetic programs in neurons, and they suggest a potential role for astrocyte-neuron interactions in schizophrenia.


Assuntos
Astrócitos , Esquizofrenia , Humanos , Astrócitos/metabolismo , Adesão Celular/genética , Esquizofrenia/genética , Esquizofrenia/metabolismo , Neurônios/metabolismo , Neuroglia , Sinapses/fisiologia
7.
Nat Commun ; 13(1): 3690, 2022 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-35760976

RESUMO

It is unclear how the 22q11.2 deletion predisposes to psychiatric disease. To study this, we generated induced pluripotent stem cells from deletion carriers and controls and utilized CRISPR/Cas9 to introduce the heterozygous deletion into a control cell line. Here, we show that upon differentiation into neural progenitor cells, the deletion acted in trans to alter the abundance of transcripts associated with risk for neurodevelopmental disorders including autism. In excitatory neurons, altered transcripts encoded presynaptic factors and were associated with genetic risk for schizophrenia, including common and rare variants. To understand how the deletion contributed to these changes, we defined the minimal protein-protein interaction network that best explains gene expression alterations. We found that many genes in 22q11.2 interact in presynaptic, proteasome, and JUN/FOS transcriptional pathways. Our findings suggest that the 22q11.2 deletion impacts genes that may converge with psychiatric risk loci to influence disease manifestation in each deletion carrier.


Assuntos
Síndrome de DiGeorge , Células-Tronco Pluripotentes Induzidas , Esquizofrenia , Linhagem Celular , Síndrome de DiGeorge/genética , Humanos , Neurônios , RNA , Esquizofrenia/genética
8.
Methods Mol Biol ; 2143: 159-168, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32524479

RESUMO

Axonal damage can cause a loss of neural control of target peripheral muscles and other organs. The hallmark of complete recovery from severe axonal injury is a successful return of function. To assay the degree of functional loss or recovery from injury, a measurement of electrical communication at the nerve-target junction can be used. Drosophila larval neuromuscular junction (NMJ) provides a genetically tractable and easily accessible model to measure the electrophysiological properties of the synapse. To study the functional consequences of injuries to the peripheral nerve, we describe the procedure to measure the spontaneous and evoked response to neurotransmitter release at the NMJ.


Assuntos
Axônios/fisiologia , Drosophila melanogaster/fisiologia , Eletrofisiologia/métodos , Potenciais Evocados/fisiologia , Junção Neuromuscular/fisiologia , Neurotransmissores/metabolismo , Técnicas de Patch-Clamp/métodos , Potenciais Sinápticos/fisiologia , Transmissão Sináptica/fisiologia , Animais , Dissecação/métodos , Drosophila melanogaster/crescimento & desenvolvimento , Larva , Recuperação de Função Fisiológica
9.
10.
Nat Neurosci ; 22(11): 1925-1935, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31527803

RESUMO

The thalamus is the central communication hub of the forebrain and provides the cerebral cortex with inputs from sensory organs, subcortical systems and the cortex itself. Multiple thalamic regions send convergent information to each cortical region, but the organizational logic of thalamic projections has remained elusive. Through comprehensive transcriptional analyses of retrogradely labeled thalamic neurons in adult mice, we identify three major profiles of thalamic pathways. These profiles exist along a continuum that is repeated across all major projection systems, such as those for vision, motor control and cognition. The largest component of gene expression variation in the mouse thalamus is topographically organized, with features conserved in humans. Transcriptional differences between these thalamic neuronal identities are tied to cellular features that are critical for function, such as axonal morphology and membrane properties. Molecular profiling therefore reveals covariation in the properties of thalamic pathways serving all major input modalities and output targets, thus establishing a molecular framework for understanding the thalamus.


Assuntos
Córtex Cerebral/anatomia & histologia , Tálamo/anatomia & histologia , Potenciais de Ação , Animais , Atlas como Assunto , Córtex Cerebral/metabolismo , Córtex Cerebral/fisiologia , Humanos , Camundongos , Camundongos Transgênicos , Vias Neurais/anatomia & histologia , Vias Neurais/metabolismo , Vias Neurais/fisiologia , Tálamo/metabolismo , Tálamo/fisiologia , Transcriptoma
11.
Neuron ; 87(4): 684-98, 2015 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-26291155

RESUMO

Autism spectrum disorders (ASDs) and related neurological disorders are associated with mutations in many genes affecting the ratio between neuronal excitation and inhibition. However, understanding the impact of these mutations on network activity is complicated by the plasticity of these networks, making it difficult in many cases to separate initial deficits from homeostatic compensation. Here we explore the contrasting evidence for primary defects in inhibition or excitation in ASDs and attempt to integrate the findings in terms of the brain's ability to maintain functional homeostasis.


Assuntos
Encéfalo/fisiopatologia , Transtornos Globais do Desenvolvimento Infantil/fisiopatologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Homeostase/fisiologia , Rede Nervosa/fisiopatologia , Inibição Neural/fisiologia , Animais , Encéfalo/metabolismo , Transtornos Globais do Desenvolvimento Infantil/metabolismo , Humanos , Rede Nervosa/metabolismo , Plasticidade Neuronal/fisiologia , Transmissão Sináptica/fisiologia
12.
Neurobiol Dis ; 77: 13-25, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25726747

RESUMO

Nerve injury can lead to axonal regeneration, axonal degeneration, and/or neuronal cell death. Remarkably, the MAP3K dual leucine zipper kinase, DLK, promotes each of these responses, suggesting that DLK is a sensor of axon injury. In Drosophila, mutations in proteins that stabilize the actin and microtubule cytoskeletons activate the DLK pathway, suggesting that DLK may be activated by cytoskeletal disruption. Here we test this model in mammalian sensory neurons. We find that pharmacological agents designed to disrupt either the actin or microtubule cytoskeleton activate the DLK pathway, and that activation is independent of calcium influx or induction of the axon degeneration program. Moreover, activation of the DLK pathway by targeting the cytoskeleton induces a pro-regenerative state, enhancing axon regeneration in response to a subsequent injury in a process akin to preconditioning. This highlights the potential utility of activating the DLK pathway as a method to improve axon regeneration. Moreover, DLK is required for these responses to cytoskeletal perturbations, suggesting that DLK functions as a key neuronal sensor of cytoskeletal damage.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/deficiência , MAP Quinase Quinase 4/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Regeneração Nervosa/efeitos dos fármacos , Células Receptoras Sensoriais/metabolismo , Animais , Proteínas de Ligação ao Cálcio , Células Cultivadas , Quelantes/farmacologia , Citocalasina D/farmacologia , Citoesqueleto/metabolismo , Embrião de Mamíferos , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Feminino , Gânglios Espinais/citologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , MAP Quinase Quinase 4/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Masculino , Camundongos , Camundongos Transgênicos , Regeneração Nervosa/fisiologia , Nocodazol/farmacologia , Células Receptoras Sensoriais/efeitos dos fármacos , Fatores de Tempo
13.
J Neurosci ; 34(25): 8398-410, 2014 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-24948796

RESUMO

The Wallenda (Wnd)/dual leucine zipper kinase (DLK)-Jnk pathway is an evolutionarily conserved MAPK signaling pathway that functions during neuronal development and following axonal injury. Improper pathway activation causes defects in axonal guidance and synaptic growth, whereas loss-of-function mutations in pathway components impairs axonal regeneration and degeneration after injury. Regulation of this pathway is in part through the E3 ubiquitin ligase Highwire (Hiw), which targets Wnd/DLK for degradation to limit MAPK signaling. To explore mechanisms controlling Wnd/DLK signaling, we performed a large-scale genetic screen in Drosophila to identify negative regulators of the pathway. Here we describe the identification and characterization of SkpA, a core component of SCF E3 ubiquitin ligases. Mutants in SkpA display synaptic overgrowth and an increase in Jnk signaling, similar to hiw mutants. The combination of hypomorphic alleles of SkpA and hiw leads to enhanced synaptic growth. Mutants in the Wnd-Jnk pathway suppress the overgrowth of SkpA mutants demonstrating that the synaptic overgrowth is due to increased Jnk signaling. These findings support the model that SkpA and the E3 ligase Hiw function as part of an SCF-like complex that attenuates Wnd/DLK signaling. In addition, SkpA, like Hiw, is required for synaptic and axonal responses to injury. Synapses in SkpA mutants are more stable following genetic or traumatic axonal injury, and axon loss is delayed in SkpA mutants after nerve crush. As in highwire mutants, this axonal protection requires Nmnat. Hence, SkpA is a novel negative regulator of the Wnd-Jnk pathway that functions with Hiw to regulate both synaptic development and axonal maintenance.


Assuntos
Axônios/metabolismo , Proteínas de Drosophila/fisiologia , Degeneração Neural/metabolismo , Terminações Pré-Sinápticas/metabolismo , Proteínas Ligases SKP Culina F-Box/fisiologia , Sinapses/metabolismo , Animais , Animais Geneticamente Modificados , Axônios/patologia , Drosophila melanogaster , Feminino , Masculino , Mutação/genética , Degeneração Neural/genética , Degeneração Neural/patologia , Terminações Pré-Sinápticas/patologia , Sinapses/genética , Sinapses/patologia
14.
Elife ; 3: e01849, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24642409

RESUMO

Neurons regulate ionic fluxes across their plasma membrane to maintain their excitable properties under varying environmental conditions. However, the mechanisms that regulate ion channels abundance remain poorly understood. Here we show that pickpocket 29 (ppk29), a gene that encodes a Drosophila degenerin/epithelial sodium channel (DEG/ENaC), regulates neuronal excitability via a protein-independent mechanism. We demonstrate that the mRNA 3'UTR of ppk29 affects neuronal firing rates and associated heat-induced seizures by acting as a natural antisense transcript (NAT) that regulates the neuronal mRNA levels of seizure (sei), the Drosophila homolog of the human Ether-à-go-go Related Gene (hERG) potassium channel. We find that the regulatory impact of ppk29 mRNA on sei is independent of the sodium channel it encodes. Thus, our studies reveal a novel mRNA dependent mechanism for the regulation of neuronal excitability that is independent of protein-coding capacity. DOI: http://dx.doi.org/10.7554/eLife.01849.001.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Canais de Potássio Éter-A-Go-Go/metabolismo , Resposta ao Choque Térmico , Canais Iônicos/metabolismo , Neurônios/metabolismo , RNA Antissenso/metabolismo , Regiões 3' não Traduzidas , Potenciais de Ação , Animais , Animais Geneticamente Modificados , Comportamento Animal , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Canais de Potássio Éter-A-Go-Go/genética , Regulação da Expressão Gênica , Genótipo , Canais Iônicos/genética , Locomoção , Mutação , Fenótipo , Interferência de RNA , RNA Antissenso/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Fatores de Tempo , Transcrição Gênica
15.
J Neurosci ; 33(45): 17863-73, 2013 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-24198375

RESUMO

The MAPKKK dual leucine zipper-containing kinase (DLK, Wallenda in Drosophila) is an evolutionarily conserved component of the axonal injury response pathway. After nerve injury, DLK promotes degeneration of distal axons and regeneration of proximal axons. This dual role in coordinating degeneration and regeneration suggests that DLK may be a sensor of axon injury, and so understanding how DLK is activated is important. Two mechanisms are known to activate DLK. First, increasing the levels of DLK via overexpression or loss of the PHR ubiquitin ligases that target DLK activate DLK signaling. Second, in Caenorhabditis elegans, a calcium-dependent mechanism, can activate DLK. Here we describe a new mechanism that activates DLK in Drosophila: loss of the spectraplakin short stop (shot). In a genetic screen for mutants with defective neuromuscular junction development, we identify a hypomorphic allele of shot that displays synaptic terminal overgrowth and a precocious regenerative response to nerve injury. We demonstrate that both phenotypes are the result of overactivation of the DLK signaling pathway. We further show that, unlike mutations in the PHR ligase Highwire, loss of function of shot activates DLK without a concomitant increase in the levels of DLK. As a spectraplakin, Shot binds to both actin and microtubules and promotes cytoskeletal stability. The DLK pathway is also activated by downregulation of the TCP1 chaperonin complex, whose normal function is to promote cytoskeletal stability. These findings support the model that DLK is activated by cytoskeletal instability, which is a shared feature of both spectraplakin mutants and injured axons.


Assuntos
Proteínas de Drosophila/genética , Drosophila/genética , MAP Quinase Quinase Quinases/genética , Proteínas dos Microfilamentos/genética , Mutação , Proteínas do Tecido Nervoso/genética , Alelos , Animais , Citoesqueleto/genética , Citoesqueleto/metabolismo , Regulação para Baixo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fenótipo
16.
J Neurosci ; 32(47): 16586-96, 2012 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-23175814

RESUMO

Synaptic communication requires the controlled release of synaptic vesicles from presynaptic axon terminals. Release efficacy is regulated by the many proteins that comprise the presynaptic release apparatus, including Ca(2+) channels and proteins that influence Ca(2+) channel accumulation at release sites. Here we identify Drosophila RIM (Rab3 interacting molecule) and demonstrate that it localizes to active zones at the larval neuromuscular junction. In Drosophila RIM mutants, there is a large decrease in evoked synaptic transmission because of a significant reduction in both the clustering of Ca(2+) channels and the size of the readily releasable pool of synaptic vesicles at active zones. Hence, RIM plays an evolutionarily conserved role in regulating synaptic calcium channel localization and readily releasable pool size. Because RIM has traditionally been studied as an effector of Rab3 function, we investigate whether RIM is involved in the newly identified function of Rab3 in the distribution of presynaptic release machinery components across release sites. Bruchpilot (Brp), an essential component of the active zone cytomatrix T bar, is unaffected by RIM disruption, indicating that Brp localization and distribution across active zones does not require wild-type RIM. In addition, larvae containing mutations in both RIM and rab3 have reduced Ca(2+) channel levels and a Brp distribution that is very similar to that of the rab3 single mutant, indicating that RIM functions to regulate Ca(2+) channel accumulation but is not a Rab3 effector for release machinery distribution across release sites.


Assuntos
Canais de Cálcio/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila/fisiologia , Junção Neuromuscular/metabolismo , Proteínas rab3 de Ligação ao GTP/genética , Proteínas rab3 de Ligação ao GTP/metabolismo , Animais , Clonagem Molecular , Análise Mutacional de DNA , DNA Complementar/biossíntese , DNA Complementar/genética , Proteínas de Drosophila/fisiologia , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Larva , Microscopia Confocal , Microscopia Eletrônica , Técnicas de Patch-Clamp , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/ultraestrutura , Proteínas rab3 de Ligação ao GTP/fisiologia
17.
Dev Biol ; 366(2): 163-71, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22542760

RESUMO

Neuronal circuit development and function require proper synapse formation and maintenance. Genetic screens are one powerful method to identify the mechanisms shaping synaptic development and stability. However, genes with essential roles in non-neural tissues may be missed in traditional loss-of-function screens. In an effort to circumvent this limitation, we used neuron-specific RNAi knock down in Drosophila and assayed the formation, growth, and maintenance of the neuromuscular junction (NMJ). We examined 1970 Drosophila genes, each of which has a conserved ortholog in mammalian genomes. Knock down of 158 genes in post-mitotic neurons led to abnormalities in the neuromuscular system, including misapposition of active zone components opposite postsynaptic glutamate receptors, synaptic terminal overgrowth and undergrowth, abnormal accumulation of synaptic material within the axon, and retraction of synaptic terminals from their postsynaptic targets. Bioinformatics analysis demonstrates that genes with overlapping annotated function are enriched within the hits for each phenotype, suggesting that the shared biological function is important for that aspect of synaptic development. For example, genes for proteasome subunits and mitotic spindle organizers are enriched among the genes whose knock down leads to defects in synaptic apposition and NMJ stability. Such genes play essential roles in all cells, however the use of tissue- and temporally-restricted RNAi indicates that the proteasome and mitotic spindle organizers participate in discrete aspects of synaptic development. In addition to identifying functional classes of genes shaping synaptic development, this screen also identifies candidate genes whose role at the synapse can be validated by traditional loss-of-function analysis. We present one such example, the dynein-interacting protein NudE, and demonstrate that it is required for proper axonal transport and synaptic maintenance. Thus, this screen has identified both functional classes of genes as well as individual candidate genes that are critical for synaptic development and will be a useful resource for subsequent mechanistic analysis of synapse formation and maintenance.


Assuntos
Drosophila/genética , Genes Controladores do Desenvolvimento , Interferência de RNA , Sinapses/fisiologia , Animais , Drosophila/embriologia , Drosophila/fisiologia , Técnicas de Silenciamento de Genes , Receptores de Glutamato/fisiologia , Transmissão Sináptica/fisiologia
18.
Nat Neurosci ; 14(10): 1267-75, 2011 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-21874015

RESUMO

The evolutionarily conserved Highwire (Hiw)/Drosophila Fsn E3 ubiquitin ligase complex is required for normal synaptic morphology during development and axonal regeneration after injury. However, little is known about the molecular mechanisms that regulate the Hiw E3 ligase complex. Using tandem affinity purification techniques, we identified Drosophila Rae1 as a previously unknown component of the Hiw/Fsn complex. Loss of Rae1 function in neurons results in morphological defects at the neuromuscular junction that are similar to those seen in hiw mutants. We found that Rae1 physically and genetically interacts with Hiw and restrains synaptic terminal growth by regulating the MAP kinase kinase kinase Wallenda. Moreover, we found that the Rae1 is both necessary and sufficient to promote Hiw protein abundance, and it does so by binding to Hiw and protecting Hiw from autophagy-mediated degradation. These results describe a previously unknown mechanism that selectively controls Hiw protein abundance during synaptic development.


Assuntos
Ciclo Celular/genética , Proteínas de Drosophila/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/fisiologia , Proteínas Associadas à Matriz Nuclear/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Animais , Animais Geneticamente Modificados , Autofagia/genética , Cromatografia Líquida/métodos , Drosophila , Proteínas de Drosophila/genética , Regulação da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Larva , Mutação/genética , Proteínas do Tecido Nervoso/genética , Junção Neuromuscular/genética , Junção Neuromuscular/metabolismo , Proteínas Associadas à Matriz Nuclear/genética , Proteínas de Transporte Nucleocitoplasmático/genética , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/fisiologia , Ligação Proteica/genética , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Espectrometria de Massas em Tandem/métodos , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
19.
Bull Math Biol ; 73(6): 1271-91, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20640526

RESUMO

DNA replication in Escherichia coli is initiated by DnaA binding to oriC, the replication origin. During the process of assembly of the replication factory, the DnaA is released back into the cytoplasm, where it is competent to reinitiate replication. Premature reinitiation is prevented by binding SeqA to newly formed GATC sites near the replication origin. Resolution of the resulting SeqA cluster is one aspect of timing for reinitiation. A Markov model accounting for the competition between SeqA binding and methylation for one or several GATC sites relates the timing to reaction rates, and consequently to the concentrations of SeqA and methylase. A model is proposed for segregation, the motion of the two daughter DNAs into opposite poles of the cell before septation. This model assumes that the binding of SeqA and its subsequent clustering results in loops from both daughter nucleoids attached to the SeqA cluster at the GATC sites. As desequestration occurs, the cluster is divided in two, one associated with each daughter. As the loops of DNA uncoil, the two subclusters migrate apart due to the Brownian ratchet effect of the DNA loop.


Assuntos
Proteínas da Membrana Bacteriana Externa/fisiologia , Replicação do DNA/fisiologia , Proteínas de Ligação a DNA/fisiologia , Proteínas de Escherichia coli/fisiologia , Escherichia coli/fisiologia , Modelos Biológicos , Origem de Replicação/fisiologia , Divisão Celular/fisiologia , Escherichia coli/citologia , Cadeias de Markov , Processos Estocásticos
20.
J Neurosci ; 29(37): 11484-94, 2009 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-19759297

RESUMO

The synapse is composed of an active zone apposed to a postsynaptic cluster of neurotransmitter receptors. Each Drosophila neuromuscular junction comprises hundreds of such individual release sites apposed to clusters of glutamate receptors. Here, we show that protein phosphatase 2A (PP2A) is required for the development of structurally normal active zones opposite glutamate receptors. When PP2A is inhibited presynaptically, many glutamate receptor clusters are unapposed to Bruchpilot (Brp), an active zone protein required for normal transmitter release. These unapposed receptors are not due to presynaptic retraction of synaptic boutons, since other presynaptic components are still apposed to the entire postsynaptic specialization. Instead, these data suggest that Brp localization is regulated at the level of individual release sites. Live imaging of glutamate receptors demonstrates that this disruption to active zone development is accompanied by abnormal postsynaptic development, with decreased formation of glutamate receptor clusters. Remarkably, inhibition of the serine-threonine kinase GSK-3beta completely suppresses the active zone defect, as well as other synaptic morphology phenotypes associated with inhibition of PP2A. These data suggest that PP2A and GSK-3beta function antagonistically to control active zone development, providing a potential mechanism for regulating synaptic efficacy at a single release site.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Quinase 3 da Glicogênio Sintase/metabolismo , Junção Neuromuscular/fisiologia , Proteína Fosfatase 2/metabolismo , Animais , Animais Geneticamente Modificados , Citoesqueleto/metabolismo , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Tomografia com Microscopia Eletrônica/métodos , Regulação da Expressão Gênica no Desenvolvimento/genética , Glicogênio Sintase Quinase 3 beta , Microscopia Confocal , Mutação/genética , Junção Neuromuscular/citologia , Junção Neuromuscular/ultraestrutura , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/ultraestrutura , Transporte Proteico/genética , Receptores de Glutamato/metabolismo , Transmissão Sináptica/genética , Transmissão Sináptica/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...