Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2022 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-36299421

RESUMO

The coronavirus disease 2019 (COVID19) continues to spread despite global vaccination efforts (1). This, alongside the rapid emergence of vaccine resistant variants, creates a need for orthogonal therapeutic strategies targeting more conserved facets of severe acute respiratory syndrome coronavirus (SARS-CoV-2) (2-4). One conserved feature of all coronaviruses is their ability to undergo discontinuous transcription wherein individual open reading frames fuse with the 5'-UTR leader sequence during negative-strand RNA synthesis (5). As such all viral protein coding genes use the same 5'-UTR for translation (6). Using in vitro reporter assays, we demonstrate that the SARS-CoV-2 5'-UTR efficiently initiates protein translation despite its predicted structural complexity. Through a combination of bioinformatic and biochemical assays, we demonstrate that a single METTL3-dependent m6A methylation event in SARS-CoV-2 5'-UTR regulates the rate of translation initiation. We show that m6A likely exerts this effect by destabilizing secondary structure in the 5'-UTR, thereby facilitating access to the ribosomal pre-initiation complex. This discovery opens new avenues for novel therapeutic strategies aimed at controlling the ability of SARS-CoV-2 to replicate in host cells.

2.
Nucleic Acids Res ; 50(17): 9748-9764, 2022 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-36029115

RESUMO

Retrograde bone morphogenetic protein (BMP) signaling at the Drosophila neuromuscular junction (NMJ) has served as a paradigm to study TGF-ß-dependent synaptic function and maturation. Yet, how retrograde BMP signaling transcriptionally regulates these functions remains unresolved. Here, we uncover a gene network, enriched for neurotransmission-related genes, that is controlled by retrograde BMP signaling in motor neurons through two Smad-binding cis-regulatory motifs, the BMP-activating (BMP-AE) and silencer (BMP-SE) elements. Unpredictably, both motifs mediate direct gene activation, with no involvement of the BMP derepression pathway regulators Schnurri and Brinker. Genome editing of candidate BMP-SE and BMP-AE within the locus of the active zone gene bruchpilot, and a novel Ly6 gene witty, demonstrated the role of these motifs in upregulating genes required for the maturation of pre- and post-synaptic NMJ compartments. Our findings uncover how Smad-dependent transcriptional mechanisms specific to motor neurons directly orchestrate a gene network required for synaptic maturation by retrograde BMP signaling.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Drosophila , Drosophila/metabolismo , Redes Reguladoras de Genes , Junção Neuromuscular/metabolismo , Animais , Animais Geneticamente Modificados , Drosophila/genética , Proteínas de Drosophila/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
3.
Proc Natl Acad Sci U S A ; 119(34): e2110097119, 2022 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-35969789

RESUMO

While the role of barrier function in establishing a protective, nutrient-rich, and ionically balanced environment for neurons has been appreciated for some time, little is known about how signaling cues originating in barrier-forming cells participate in maintaining barrier function and influence synaptic activity. We have identified Delta/Notch signaling in subperineurial glia (SPG), a crucial glial type for Drosophila motor axon ensheathment and the blood-brain barrier, to be essential for controlling the expression of matrix metalloproteinase 1 (Mmp1), a major regulator of the extracellular matrix (ECM). Our genetic analysis indicates that Delta/Notch signaling in SPG exerts an inhibitory control on Mmp1 expression. In the absence of this inhibition, abnormally enhanced Mmp1 activity disrupts septate junctions and glial ensheathment of peripheral motor nerves, compromising neurotransmitter release at the neuromuscular junction (NMJ). Temporally controlled and cell type-specific transgenic analysis shows that Delta/Notch signaling inhibits transcription of Mmp1 by inhibiting c-Jun N-terminal kinase (JNK) signaling in SPG. Our results provide a mechanistic insight into the regulation of neuronal health and function via glial-initiated signaling and open a framework for understanding the complex relationship between ECM regulation and the maintenance of barrier function.


Assuntos
Proteínas de Drosophila , Metaloproteinase 1 da Matriz , Neuroglia , Transmissão Sináptica , Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Matriz Extracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Metaloproteinase 1 da Matriz/metabolismo , Proteínas de Membrana/metabolismo , Neuroglia/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais
4.
Cell Rep ; 39(10): 110911, 2022 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-35675781

RESUMO

Genetic perturbances in translational regulation result in defects in cerebellar motor learning; however, little is known about the role of translational mechanisms in the regulation of cerebellar plasticity. We show that genetic removal of 4E-BP, a translational suppressor and target of mammalian target of rapamycin complex 1, results in a striking change in cerebellar synaptic plasticity. We find that cerebellar long-term depression (LTD) at parallel fiber-Purkinje cell synapses is converted to long-term potentiation in 4E-BP knockout mice. Biochemical and pharmacological experiments suggest that increased phosphatase activity largely accounts for the defects in LTD. Our results point to a model in which translational regulation through the action of 4E-BP plays a critical role in establishing the appropriate kinase/phosphatase balance required for normal synaptic plasticity in the cerebellum.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Ciclo Celular , Potenciação de Longa Duração , Depressão Sináptica de Longo Prazo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas de Ciclo Celular/genética , Cerebelo/fisiologia , Potenciação de Longa Duração/fisiologia , Depressão Sináptica de Longo Prazo/fisiologia , Mamíferos , Camundongos , Plasticidade Neuronal/fisiologia , Monoéster Fosfórico Hidrolases , Células de Purkinje/fisiologia , Sinapses/fisiologia
5.
Cell Rep ; 26(7): 1774-1786.e4, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30759389

RESUMO

Pathogenic mutations in leucine-rich repeat kinase 2 (LRRK2) induce an age-dependent loss of dopaminergic (DA) neurons. We have identified Furin 1, a pro-protein convertase, as a translational target of LRRK2 in DA neurons. Transgenic knockdown of Furin1 or its substrate the bone morphogenic protein (BMP) ligand glass bottom boat (Gbb) protects against LRRK2-induced loss of DA neurons. LRRK2 enhances the accumulation of phosphorylated Mad (pMad) in the nuclei of glial cells in the vicinity of DA neurons but not in DA neurons. Consistently, exposure to paraquat enhances Furin 1 levels in DA neurons and induces BMP signaling in glia. In support of a neuron-glial signaling model, knocking down BMP pathway members only in glia, but not in neurons, can protect against paraquat toxicity. We propose that a neuron-glial BMP-signaling cascade is critical for mediating age-dependent neurodegeneration in two models of Parkinson's disease, thus opening avenues for future therapeutic interventions.


Assuntos
Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Doenças Neurodegenerativas/genética , Neuroglia/metabolismo , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Transdução de Sinais
6.
Cell Rep ; 23(1): 11-22, 2018 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-29617653

RESUMO

Throughout the developing nervous system, considerable synaptic re-organization takes place as postsynaptic neurons extend dendrites and incoming axons refine their synapses, strengthening some and eliminating others. It is well accepted that these processes rely on synaptic activity; however, the mechanisms that lead to this developmental reorganization are not fully understood. Here, we explore the regulation of cap-dependent translation, a mechanism known to play a role in synaptic growth and plasticity. Using sympathetic ganglia in α3 nicotinic acetylcholine receptor (nAChR)-knockout (KO) mice, we establish that electrophysiologically silent synapses between preganglionic axons and postsynaptic sympathetic neurons do not refine, and the growth of dendrites and the targeting of synapses on postsynaptic neurons are impaired. Remarkably, genetically removing 4E-BP, a suppressor of cap-dependent translation, from these α3 nAChR-KO mice largely restores these features. We conclude that synaptic connections can re-organize and refine without postsynaptic activity during post-natal development when 4E-BP-regulated cap-dependent translation is enhanced.


Assuntos
Proteínas de Transporte/genética , Fosfoproteínas/genética , Sinapses/metabolismo , Potenciais Sinápticos , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Ciclo Celular , Fatores de Iniciação em Eucariotos , Gânglios Simpáticos/citologia , Gânglios Simpáticos/metabolismo , Gânglios Simpáticos/fisiologia , Camundongos , Receptores Nicotínicos/genética , Sinapses/fisiologia
7.
PLoS Genet ; 14(1): e1007184, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29373576

RESUMO

Retrograde signaling is essential for neuronal growth, function and survival; however, we know little about how signaling endosomes might be directed from synaptic terminals onto retrograde axonal pathways. We have identified Khc-73, a plus-end directed microtubule motor protein, as a regulator of sorting of endosomes in Drosophila larval motor neurons. The number of synaptic boutons and the amount of neurotransmitter release at the Khc-73 mutant larval neuromuscular junction (NMJ) are normal, but we find a significant decrease in the number of presynaptic release sites. This defect in Khc-73 mutant larvae can be genetically enhanced by a partial genetic loss of Bone Morphogenic Protein (BMP) signaling or suppressed by activation of BMP signaling in motoneurons. Consistently, activation of BMP signaling that normally enhances the accumulation of phosphorylated form of BMP transcription factor Mad in the nuclei, can be suppressed by genetic removal of Khc-73. Using a number of assays including live imaging in larval motor neurons, we show that loss of Khc-73 curbs the ability of retrograde-bound endosomes to leave the synaptic area and join the retrograde axonal pathway. Our findings identify Khc-73 as a regulator of endosomal traffic at the synapse and modulator of retrograde BMP signaling in motoneurons.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Drosophila/fisiologia , Endossomos/metabolismo , Cinesinas/fisiologia , Junção Neuromuscular/metabolismo , Animais , Animais Geneticamente Modificados , Proteínas de Drosophila/genética , Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Cinesinas/genética , Neurônios Motores/metabolismo , Terminações Pré-Sinápticas/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Sinapses/metabolismo
8.
Neuron ; 92(6): 1204-1212, 2016 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-27916456

RESUMO

While beneficial effects of fasting on organismal function and health are well appreciated, we know little about the molecular details of how fasting influences synaptic function and plasticity. Our genetic and electrophysiological experiments demonstrate that acute fasting blocks retrograde synaptic enhancement that is normally triggered as a result of reduction in postsynaptic receptor function at the Drosophila larval neuromuscular junction (NMJ). This negative regulation critically depends on transcriptional enhancement of eukaryotic initiation factor 4E binding protein (4E-BP) under the control of the transcription factor Forkhead box O (Foxo). Furthermore, our findings indicate that postsynaptic 4E-BP exerts a constitutive negative input, which is counteracted by a positive regulatory input from the Target of Rapamycin (TOR). This combinatorial retrograde signaling plays a key role in regulating synaptic strength. Our results provide a mechanistic insight into how cellular stress and nutritional scarcity could acutely influence synaptic homeostasis and functional stability in neural circuits.


Assuntos
Proteínas de Drosophila/genética , Jejum/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Músculo Esquelético/metabolismo , Junção Neuromuscular/metabolismo , Fatores de Iniciação de Peptídeos/genética , Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Mutação , Plasticidade Neuronal/genética , Fatores de Iniciação de Peptídeos/metabolismo , Biossíntese de Proteínas , Receptores Ionotrópicos de Glutamato/genética , Proteínas Quinases S6 Ribossômicas/genética , Transmissão Sináptica , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
9.
Nat Commun ; 7: 12188, 2016 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-27432119

RESUMO

Parkinson's disease gene leucine-rich repeat kinase 2 (LRRK2) has been implicated in a number of processes including the regulation of mitochondrial function, autophagy and endocytic dynamics; nevertheless, we know little about its potential role in the regulation of synaptic plasticity. Here we demonstrate that postsynaptic knockdown of the fly homologue of LRRK2 thwarts retrograde, homeostatic synaptic compensation at the larval neuromuscular junction. Conversely, postsynaptic overexpression of either the fly or human LRRK2 transgene induces a retrograde enhancement of presynaptic neurotransmitter release by increasing the size of the release ready pool of vesicles. We show that LRRK2 promotes cap-dependent translation and identify Furin 1 as its translational target, which is required for the synaptic function of LRRK2. As the regulation of synaptic homeostasis plays a fundamental role in ensuring normal and stable synaptic function, our findings suggest that aberrant function of LRRK2 may lead to destabilization of neural circuits.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Junção Neuromuscular/metabolismo , Sinapses/metabolismo , Animais , Furina/metabolismo , Células HEK293 , Homeostase , Humanos , Larva/metabolismo , Proteínas Mutantes/metabolismo , Mutação/genética , Neurotransmissores/metabolismo , Biossíntese de Proteínas , Capuzes de RNA/metabolismo , Transdução de Sinais , Transmissão Sináptica , Vesículas Sinápticas/metabolismo
10.
Neuron ; 74(1): 166-78, 2012 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-22500638

RESUMO

Homeostatic mechanisms operate to stabilize synaptic function; however, we know little about how they are regulated. Exploiting Drosophila genetics, we have uncovered a critical role for the target of rapamycin (TOR) in the regulation of synaptic homeostasis at the Drosophila larval neuromuscular junction. Loss of postsynaptic TOR disrupts a retrograde compensatory enhancement in neurotransmitter release that is normally triggered by a reduction in postsynaptic glutamate receptor activity. Moreover, postsynaptic overexpression of TOR or a phosphomimetic form of S6 ribosomal protein kinase, a common target of TOR, can trigger a strong retrograde increase in neurotransmitter release. Interestingly, heterozygosity for eIF4E, a critical component of the cap-binding protein complex, blocks the retrograde signal in all these cases. Our findings suggest that cap-dependent translation under the control of TOR plays a critical role in establishing the activity dependent homeostatic response at the NMJ.


Assuntos
Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica/fisiologia , Homeostase/fisiologia , Junção Neuromuscular/metabolismo , Proteínas Quinases/metabolismo , Transmissão Sináptica/fisiologia , Animais , Drosophila , Proteínas de Drosophila/genética , Fator de Iniciação 4E em Eucariotos/genética , Fator de Iniciação 4E em Eucariotos/fisiologia , Exocitose/fisiologia , Larva/metabolismo , Neurônios Motores/metabolismo , Mutação , Proteínas Quinases/genética , Transporte Proteico/fisiologia , Receptores de Glutamato/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR
11.
PLoS Genet ; 8(2): e1002515, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22347817

RESUMO

miR-124 is conserved in sequence and neuronal expression across the animal kingdom and is predicted to have hundreds of mRNA targets. Diverse defects in neural development and function were reported from miR-124 antisense studies in vertebrates, but a nematode knockout of mir-124 surprisingly lacked detectable phenotypes. To provide genetic insight from Drosophila, we deleted its single mir-124 locus and found that it is dispensable for gross aspects of neural specification and differentiation. On the other hand, we detected a variety of mutant phenotypes that were rescuable by a mir-124 genomic transgene, including short lifespan, increased dendrite variation, impaired larval locomotion, and aberrant synaptic release at the NMJ. These phenotypes reflect extensive requirements of miR-124 even under optimal culture conditions. Comparison of the transcriptomes of cells from wild-type and mir-124 mutant animals, purified on the basis of mir-124 promoter activity, revealed broad upregulation of direct miR-124 targets. However, in contrast to the proposed mutual exclusion model for miR-124 function, its functional targets were relatively highly expressed in miR-124-expressing cells and were not enriched in genes annotated with epidermal expression. A notable aspect of the direct miR-124 network was coordinate targeting of five positive components in the retrograde BMP signaling pathway, whose activation in neurons increases synaptic release at the NMJ, similar to mir-124 mutants. Derepression of the direct miR-124 target network also had many secondary effects, including over-activity of other post-transcriptional repressors and a net incomplete transition from a neuroblast to a neuronal gene expression signature. Altogether, these studies demonstrate complex consequences of miR-124 loss on neural gene expression and neurophysiology.


Assuntos
Drosophila melanogaster/genética , MicroRNAs/genética , Mutação/genética , Neurogênese/genética , Junção Neuromuscular/genética , Animais , Diferenciação Celular/genética , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Locomoção/genética , Junção Neuromuscular/fisiologia , Fenótipo , Células Receptoras Sensoriais/patologia , Transdução de Sinais/genética , Sinapses/patologia , Transcriptoma/genética
12.
Neuron ; 68(5): 879-93, 2010 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-21145002

RESUMO

Emerging data implicate microRNAs (miRNAs) in the regulation of synaptic structure and function, but we know little about their role in the regulation of neurotransmission in presynaptic neurons. Here, we demonstrate that the miR-310-313 cluster is required for normal synaptic transmission at the Drosophila larval neuromuscular junction. Loss of miR-310-313 cluster leads to a significant enhancement of neurotransmitter release, which can be rescued with temporally restricted expression of mir-310-313 in larval presynaptic neurons. Kinesin family member, Khc-73 is a functional target for miR-310-313 as its expression is increased in mir-310-313 mutants and reducing it restores normal synaptic function. Cluster mutants show an increase in the active zone protein Bruchpilot accompanied by an increase in electron dense T bars. Finally, we show that repression of Khc-73 by miR-310-313 cluster influences the establishment of normal synaptic homeostasis. Our findings establish a role for miRNAs in the regulation of neurotransmitter release.


Assuntos
Proteínas de Drosophila/genética , Drosophila/genética , MicroRNAs/genética , Junção Neuromuscular/genética , Transmissão Sináptica/genética , Animais , Drosophila/crescimento & desenvolvimento , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Cinesinas/metabolismo , Larva/genética , Larva/metabolismo , Neurônios Motores/metabolismo , Família Multigênica/genética , Junção Neuromuscular/metabolismo , Transmissão Sináptica/fisiologia
13.
Neuron ; 66(4): 536-49, 2010 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-20510858

RESUMO

Retrograde signaling is essential for coordinating the growth of synaptic structures; however, it is not clear how it can lead to modulation of cytoskeletal dynamics and structural changes at presynaptic terminals. We show that loss of retrograde bone morphogenic protein (BMP) signaling at the Drosophila larval neuromuscular junction (NMJ) leads to a significant reduction in levels of Rac GEF Trio and a diminution of transcription at the trio locus. We further find that Trio is required in motor neurons for normal structural growth. Finally, we show that transgenic expression of Trio in motor neurons can partially restore NMJ defects in larvae mutant for BMP signaling. Based on our findings, we propose a model in which a retrograde BMP signal from the muscle modulates GTPase activity through transcriptional regulation of Rac GEF trio, thereby regulating the homeostasis of synaptic growth at the NMJ.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Proteínas de Drosophila/biossíntese , Fatores de Troca do Nucleotídeo Guanina/biossíntese , Neurônios Motores/fisiologia , Junção Neuromuscular/fisiologia , Fosfoproteínas/biossíntese , Proteínas Serina-Treonina Quinases/biossíntese , Sinapses/fisiologia , Animais , Linhagem Celular , Drosophila , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Transdução de Sinais/fisiologia , Sinapses/ultraestrutura
14.
Neuron ; 41(6): 891-905, 2004 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-15046722

RESUMO

Highwire (Hiw), a putative RING finger E3 ubiquitin ligase, negatively regulates synaptic growth at the neuromuscular junction (NMJ) in Drosophila. hiw mutants have dramatically larger synaptic size and increased numbers of synaptic boutons. Here we show that Hiw binds to the Smad protein Medea (Med). Med is part of a presynaptic bone morphogenetic protein (BMP) signaling cascade consisting of three receptor subunits, Wit, Tkv, and Sax, in addition to the Smad transcription factor Mad. When compared to wild-type, mutants of BMP signaling components have smaller NMJ size, reduced neurotransmitter release, and aberrant synaptic ultrastructure. BMP signaling mutants suppress the excessive synaptic growth in hiw mutants. Activation of BMP signaling, which in wild-type does not cause additional growth, in hiw mutants does lead to further synaptic expansion. These results reveal a balance between positive BMP signaling and negative regulation by Highwire, governing the growth of neuromuscular synapses.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crescimento & desenvolvimento , Proteínas do Tecido Nervoso/metabolismo , Junção Neuromuscular/crescimento & desenvolvimento , Animais , Proteínas Morfogenéticas Ósseas/genética , Tamanho Celular/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Drosophila melanogaster/ultraestrutura , Microscopia Eletrônica , Neurônios Motores/citologia , Neurônios Motores/metabolismo , Mutação/genética , Proteínas do Tecido Nervoso/genética , Junção Neuromuscular/metabolismo , Junção Neuromuscular/ultraestrutura , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/patologia , Terminações Pré-Sinápticas/ultraestrutura , Ligação Proteica/genética , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais/genética , Proteína Smad4 , Transmissão Sináptica/genética , Transativadores/genética , Transativadores/metabolismo
15.
Neuron ; 39(2): 241-54, 2003 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-12873382

RESUMO

We show that the BMP ortholog Gbb can signal by a retrograde mechanism to regulate synapse growth of the Drosophila neuromuscular junction (NMJ). gbb mutants have a reduced NMJ synapse size, decreased neurotransmitter release, and aberrant presynaptic ultrastructure. These defects are similar to those we observe in mutants of BMP receptors and Smad transcription factors. However, whereas these BMP receptors and signaling components are required in the presynaptic motoneuron, Gbb expression is required in large part in postsynaptic muscles; gbb expression in muscle rescues key aspects of the gbb mutant phenotype. Consistent with this notion, we find that blocking retrograde axonal transport by overexpression of dominant-negative p150/Glued in neurons inhibits BMP signaling in motoneurons. These experiments reveal that a muscle-derived BMP retrograde signal participates in coordinating neuromuscular synapse development and growth.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Mutação , Ácido Micofenólico/análogos & derivados , Junção Neuromuscular/crescimento & desenvolvimento , Sinapses/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Nucleotídeos de Adenina , Animais , Células Cultivadas/metabolismo , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/crescimento & desenvolvimento , Sistema Nervoso Central/metabolismo , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Complexo Dinactina , Dineínas/metabolismo , Eletrofisiologia , Potenciais Evocados/genética , Genes Dominantes/genética , Imuno-Histoquímica/métodos , Hibridização In Situ/métodos , Larva/genética , Larva/crescimento & desenvolvimento , Larva/metabolismo , Larva/ultraestrutura , Microscopia Eletrônica , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios Motores/metabolismo , Músculos/metabolismo , Mutação/genética , Ácido Micofenólico/metabolismo , Junção Neuromuscular/embriologia , Junção Neuromuscular/metabolismo , Proteínas/genética , Proteínas/metabolismo , RNA/biossíntese , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/fisiologia , Sinapses/metabolismo , Sinapses/ultraestrutura , Transfecção , Fator de Crescimento Transformador beta/genética , Asas de Animais/embriologia , Asas de Animais/crescimento & desenvolvimento
16.
Neuron ; 39(2): 255-67, 2003 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-12873383

RESUMO

Retrograde signaling plays an important role in synaptic homeostasis, growth, and plasticity. A retrograde signal at the neuromuscular junction (NMJ) of Drosophila controls the homeostasis of neurotransmitter release. Here, we show that this retrograde signal is regulated by the postsynaptic activity of Ca2+/calmodulin-dependent protein kinase II (CaMKII). Reducing CaMKII activity in muscles enhances the signal and increases neurotransmitter release, while constitutive activation of CaMKII in muscles inhibits the signal and decreases neurotransmitter release. Postsynaptic inhibition of CaMKII increases the number of presynaptic, vesicle-associated T bars at the active zones. Consistently, we show that glutamate receptor mutants also have a higher number of T bars; this increase is suppressed by postsynaptic activation of CaMKII. Furthermore, we demonstrate that presynaptic BMP receptor wishful thinking is required for the retrograde signal to function. Our results indicate that CaMKII plays a key role in the retrograde control of homeostasis of synaptic transmission at the NMJ of Drosophila.


Assuntos
Proteínas de Ligação ao Cálcio , Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Junção Neuromuscular/enzimologia , Junção Neuromuscular/fisiologia , Proteínas de Saccharomyces cerevisiae , Transmissão Sináptica/fisiologia , Animais , Animais Geneticamente Modificados , Cálcio/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Relação Dose-Resposta a Droga , Drosophila melanogaster , Eletrofisiologia , Potenciais Pós-Sinápticos Excitadores/genética , Potenciais Pós-Sinápticos Excitadores/fisiologia , Regulação Enzimológica da Expressão Gênica , Genes de Insetos , Imuno-Histoquímica , Manosiltransferases/metabolismo , Manosiltransferases/fisiologia , Glicoproteínas de Membrana/metabolismo , Microscopia Eletrônica , Músculos/metabolismo , Músculos/fisiologia , Mutagênese , Proteínas do Tecido Nervoso/metabolismo , Junção Neuromuscular/ultraestrutura , Neurônios/metabolismo , Neurônios/fisiologia , Neurotransmissores , Fragmentos de Peptídeos/fisiologia , Terminações Pré-Sinápticas/enzimologia , Terminações Pré-Sinápticas/ultraestrutura , Teoria Quântica , Receptores de AMPA/genética , Receptores de AMPA/fisiologia , Sinaptotagminas
17.
Neuron ; 33(4): 545-58, 2002 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-11856529

RESUMO

We conducted a large-scale screen for Drosophila mutants that have structural abnormalities of the larval neuromuscular junction (NMJ). We recovered mutations in wishful thinking (wit), a gene that positively regulates synaptic growth. wit encodes a BMP type II receptor. In wit mutant larvae, the size of the NMJs is greatly reduced relative to the size of the muscles. wit NMJs have reduced evoked excitatory junctional potentials, decreased levels of the synaptic cell adhesion molecule Fasciclin II, and synaptic membrane detachment at active zones. Wit is expressed by a subset of neurons, including motoneurons. The NMJ phenotype is specifically rescued by transgenic expression of Wit only in motoneurons. Thus, Wit appears to function as a presynaptic receptor that regulates synaptic size at the Drosophila NMJ.


Assuntos
Padronização Corporal/genética , Sistema Nervoso Central/anormalidades , Proteínas de Drosophila/genética , Drosophila melanogaster/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Mutação/genética , Junção Neuromuscular/anormalidades , Proteínas Serina-Treonina Quinases/genética , Animais , Animais Geneticamente Modificados/anormalidades , Animais Geneticamente Modificados/crescimento & desenvolvimento , Animais Geneticamente Modificados/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo II , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Adesão Celular/genética , Sistema Nervoso Central/crescimento & desenvolvimento , Sistema Nervoso Central/ultraestrutura , Regulação para Baixo/genética , Proteínas de Drosophila/isolamento & purificação , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/ultraestrutura , Venenos Elapídicos/metabolismo , Feminino , Testes Genéticos , Masculino , Dados de Sequência Molecular , Junção Neuromuscular/crescimento & desenvolvimento , Junção Neuromuscular/ultraestrutura , Plasticidade Neuronal/genética , Neurotransmissores/genética , Neurotransmissores/metabolismo , Proteínas Serina-Treonina Quinases/isolamento & purificação , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico , Transdução de Sinais/genética , Membranas Sinápticas/genética , Membranas Sinápticas/metabolismo , Membranas Sinápticas/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...