Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Cell ; 149(6): 1327-38, 2012 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-22682252

RESUMEN

The Drosophila fruitless (fru) gene encodes a set of putative transcription factors that promote male sexual behavior by controlling the development of sexually dimorphic neuronal circuitry. However, the mechanism whereby fru establishes the sexual fate of neurons remains enigmatic. Here, we show that Fru forms a complex with the transcriptional cofactor Bonus (Bon), which, in turn, recruits either of two chromatin regulators, Histone deacetylase 1 (HDAC1), which masculinizes individual sexually dimorphic neurons, or Heterochromatin protein 1a (HP1a), which demasculinizes them. Manipulations of HDAC1 or HP1a expression change the proportion of male-typical neurons and female-typical neurons rather than producing neurons with intersexual characteristics, indicating that on a single neuron level, this sexual switch operates in an all-or-none manner.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Histona Desacetilasa 1/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Caracteres Sexuales , Factores de Transcripción/metabolismo , Animales , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/genética , Drosophila/genética , Proteínas de Drosophila/genética , Femenino , Histona Desacetilasa 1/genética , Masculino , Conducta Sexual Animal , Transcripción Genética
2.
J Neurosci ; 43(22): 3989-4004, 2023 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-37117011

RESUMEN

The presentation of nicotinic acetylcholine receptors (nAChRs) on synaptic membranes is crucial for generating cholinergic circuits, some of which are associated with memory function and neurodegenerative disorders. Although the physiology and structure of nAChR, a cation channel comprising five subunits, have been extensively studied, little is known about how the receptor levels in interneuronal synapses are determined and which nAChR subunits participate in the regulatory process in cooperation with synaptic cleft matrices and intracellular proteins. By a genetic screen of Drosophila, we identified mutations in the nAChR subunit Dα5 gene as suppressors that restored the mutant phenotypes of hig, which encodes a secretory matrix protein localized to cholinergic synaptic clefts in the brain. Only the loss of function of Dα5 among the 10 nAChR subunits suppressed hig mutant phenotypes in both male and female flies. Dα5 behaved as a lethal factor when Hig was defective; loss of Dα5 in hig mutants rescued lethality, upregulating Dα6 synaptic levels. By contrast, levels of Dα5, Dα6, and Dα7 subunits were all reduced in hig mutants. These three subunits have distinct properties for interaction with Hig or trafficking, as confirmed by chimeric subunit experiments. Notably, the chimeric Dα5 protein, which has the extracellular sequences that display no positive interaction with Hig, exhibited abnormal distribution and lethality even in the presence of Hig. We propose that the sequestering subunit Dα5 functions by reducing synaptic levels of nAChR through internalization, and this process is blocked by Hig, which tethers Dα5 to the synaptic cleft matrix.SIGNIFICANCE STATEMENT Because the cholinergic synapse is one of the major synapses that generate various brain functions, numerous studies have sought to reveal the physiology and structure of the nicotinic acetylcholine receptor (nAChR). However, little is known about how synaptic levels of nAChR are controlled and which nAChR subunits participate in the regulatory process in cooperation with synaptic cleft matrices. By a genetic screen of Drosophila, we identified mutations in the nAChR subunit Dα5 gene as suppressors that restored the mutant phenotypes of hig, which encodes a secretory matrix protein localized to cholinergic synaptic clefts. Our data indicate that Dα5 functions in reducing synaptic levels of nAChR, and this process is blocked by Hig, which tethers Dα5 to the synaptic cleft matrix.


Asunto(s)
Proteínas de Drosophila , Receptores Nicotínicos , Animales , Femenino , Masculino , Colinérgicos , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Receptores Nicotínicos/metabolismo , Transmisión Sináptica
3.
J Neurosci ; 36(2): 590-606, 2016 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-26758847

RESUMEN

The synaptic cleft is the space through which neurotransmitters convey neural information between two synaptic terminals. This space is presumably filled with extracellular matrix molecules involved in synaptic function or differentiation. However, little is known about the identities of the matrix components, and it remains unclear how these molecules organize the matrix in synaptic clefts. In this study, we identified Hasp, a Drosophila secretory protein containing CCP and WAP domains. Molecular genetic analysis revealed that Hasp diffuses extracellularly and is predominantly captured at synaptic clefts of cholinergic synapses. Furthermore, Hasp regulates levels of DLG and the nAChR subunits Dα6 and Dα7 at postsynaptic terminals. Hasp is required for trapping of another matrix protein, Hig, which is also secreted and diffused in the brain, at synaptic clefts of cholinergic synapses; however, Hig is dispensable for localization of Hasp at synaptic clefts. In addition, in the brains of triple mutants for the nAChR subunits Dα5, Dα6, and Dα7, the level of Hig, but not Hasp, was markedly reduced in synaptic regions, indicating that these nAChR subunits are required to anchor Hig to synaptic clefts. High-resolution microscopy revealed that Hasp and Hig exhibit segregated distribution within individual synaptic clefts, reflecting their differing roles in synaptogenesis. These data provide insight into how Hasp and Hig construct the synaptic cleft matrix and regulate the differentiation of cholinergic synapses, and also illuminate a previously unidentified architecture within synaptic clefts. SIGNIFICANCE STATEMENT: The synapse has been extensively studied because it is essential for neurotransmission. By contrast, the space between the synaptic terminals, the synaptic cleft, is still an undeveloped research area despite its ubiquity in synapses. In fruit fly brains, we obtained evidence that the matrix protein Hasp and the previously identified Hig, both of which are secreted extracellularly, localize predominantly to synaptic clefts of cholinergic synapses, and modulate the levels of nAChR subunits on postsynaptic membranes. However, Hasp and Hig play differential roles in matrix formation and exhibit segregated distribution within synaptic clefts. These results reveal the molecular mechanisms of synaptic matrix construction and illuminate a molecular architecture within synaptic clefts previously unrevealed in any animal species.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/genética , Neuronas/ultraestructura , Proteínas Serina-Treonina Quinasas/metabolismo , Sinapsis/metabolismo , Sinapsis/ultraestructura , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Encéfalo/citología , Drosophila , Proteínas de Drosophila/genética , Regulación de la Expresión Génica/genética , Masculino , Proteínas del Tejido Nervioso/genética , Neurogénesis/fisiología , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Receptores Colinérgicos/genética , Receptores Colinérgicos/metabolismo , Proteínas de Transporte Vesicular de Glutamato/genética , Proteínas de Transporte Vesicular de Glutamato/metabolismo
4.
J Neurosci ; 34(42): 13872-7, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25319684

RESUMEN

The synaptic cleft, a crucial space involved in neurotransmission, is filled with extracellular matrix that serves as a scaffold for synaptic differentiation. However, little is known about the proteins present in the matrix and their functions in synaptogenesis, especially in the CNS. Here, we report that Hikaru genki (Hig), a secreted protein with an Ig motif and complement control protein domains, localizes specifically to the synaptic clefts of cholinergic synapses in the Drosophila CNS. The data indicate that this specific localization is achieved by capture of secreted Hig in synaptic clefts, even when it is ectopically expressed in glia. In the absence of Hig, the cytoskeletal scaffold protein DLG accumulated abnormally in cholinergic postsynapses, and the synaptic distribution of acetylcholine receptor (AchR) subunits Dα6 and Dα7 significantly decreased. hig mutant flies consistently exhibited resistance to the AchR agonist spinosad, which causes lethality by specifically activating the Dα6 subunit, suggesting that loss of Hig compromises the cholinergic synaptic activity mediated by Dα6. These results indicate that Hig is a specific component of the synaptic cleft matrix of cholinergic synapses and regulates their postsynaptic organization in the CNS.


Asunto(s)
Encéfalo/fisiología , Neuronas Colinérgicas/fisiología , Proteínas de Drosophila/fisiología , Proteínas del Tejido Nervioso/fisiología , Densidad Postsináptica/fisiología , Sinapsis/fisiología , Animales , Animales Modificados Genéticamente , Química Encefálica/fisiología , Neuronas Colinérgicas/química , Drosophila , Proteínas de Drosophila/análisis , Masculino , Proteínas del Tejido Nervioso/análisis , Densidad Postsináptica/química , Sinapsis/química
5.
Nat Neurosci ; 10(2): 153-60, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17220884

RESUMEN

An essential feature of the organization and function of the vertebrate and insect olfactory systems is the generation of a variety of olfactory receptor neurons (ORNs) that have different specificities in regard to both odorant receptor expression and axonal targeting. Yet the underlying mechanisms that generate this neuronal diversity remain elusive. Here we demonstrate that the Notch signal is involved in the diversification of ORNs in Drosophila melanogaster. A systematic clonal analysis showed that a cluster of ORNs housed in each sensillum were differentiated into two classes, depending on the level of Notch activity in their sibling precursors. Notably, ORNs of different classes segregated their axonal projections into distinct domains in the antennal lobes. In addition, both the odorant receptor expression and the axonal targeting of ORNs were specified according to their Notch-mediated identities. Thus, Notch signaling contributes to the diversification of ORNs, thereby regulating multiple developmental events that establish the olfactory map in Drosophila.


Asunto(s)
Diferenciación Celular/genética , Linaje de la Célula/genética , Drosophila melanogaster/crecimiento & desarrollo , Vías Olfatorias/crecimiento & desarrollo , Neuronas Receptoras Olfatorias/crecimiento & desarrollo , Receptores Notch/metabolismo , Animales , Encéfalo/citología , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Conos de Crecimiento/metabolismo , Conos de Crecimiento/ultraestructura , Hormonas Juveniles/genética , Hormonas Juveniles/metabolismo , Mutación/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Vías Olfatorias/citología , Vías Olfatorias/metabolismo , Neuronas Receptoras Olfatorias/citología , Neuronas Receptoras Olfatorias/metabolismo , Receptores Notch/genética , Receptores Odorantes/genética , Receptores Odorantes/metabolismo , Transducción de Señal/genética
6.
J Neurosci ; 29(15): 4972-80, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19369566

RESUMEN

In Drosophila, odor information received by olfactory receptor neurons (ORNs) is processed by glomeruli, which are organized in a stereotypic manner in the antennal lobe (AL). This glomerular organization is regulated by Wnt5 signaling. In the embryonic CNS, Wnt5 signaling is transduced by the Drl receptor, a member of the Ryk family. During development of the olfactory system, however, it is antagonized by Drl. Here, we identify Drl-2 as a receptor mediating Wnt5 signaling. Drl is found in the neurites of brain cells in the AL and specific glia, whereas Drl-2 is predominantly found in subsets of growing ORN axons. A drl-2 mutation produces only mild deficits in glomerular patterning, but when it is combined with a drl mutation, the phenotype is exacerbated and more closely resembles the Wnt5 phenotype. Wnt5 overexpression in ORNs induces aberrant glomeruli positioning. This phenotype is ameliorated in the drl-2 mutant background, indicating that Drl-2 mediates Wnt5 signaling. In contrast, forced expression of Drl-2 in the glia of drl mutants rescues the glomerular phenotype caused by the loss of antagonistic Drl function. Therefore, Drl-2 can also antagonize Wnt5 signaling. Additionally, our genetic data suggest that Drl localized to developing glomeruli mediates Wnt5 signaling. Thus, these two members of the Ryk family are capable of carrying out a similar molecular function, but they can play opposing roles in Wnt5 signaling, depending on the type of cells in which they are expressed. These molecules work cooperatively to establish the olfactory circuitry in Drosophila.


Asunto(s)
Proteínas de Drosophila/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Vías Olfatorias/crecimiento & desarrollo , Proteínas Proto-Oncogénicas/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Transducción de Señal/fisiología , Olfato/fisiología , Proteínas Wnt/fisiología , Animales , Animales Modificados Genéticamente , Drosophila , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/genética , Mutación , Neuroglía/metabolismo , Neuroglía/fisiología , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Proteínas Tirosina Quinasas Receptoras/genética , Transducción de Señal/genética , Proteínas Wnt/biosíntesis , Proteínas Wnt/genética
8.
Neurosci Res ; 69(2): 87-92, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21093502

RESUMEN

Neurotransmitter receptors play central roles in basic neurotransmission and synaptic plasticity. Recent studies have revealed that some transmembrane and extracellular proteins bind to neurotransmitter receptors, forming protein complexes that are required for proper synaptic localization or gating of core receptor molecules. Consequently, the components of these complexes contribute to long-term potentiation, a process that is critical for learning and memory. Here, we review factors that regulate neurotransmitter receptors, with a focus on proteins containing CUB (complement C1r/C1s, Uegf, Bmp1) or CCP (complement control protein) domains, which are frequently found in complement system proteins. Proteins that contain these domains are structurally distinct from TARPs (transmembrane AMPA receptor regulatory proteins), and may constitute new protein families that modulate either the localization or function of neurotransmitter receptors. In addition, other CCP domain-containing proteins participate in dendritic patterning and/or synaptic differentiation, although current evidence has not identified any direct activities on neurotransmitter receptors. Some of these proteins are involved in pathologic conditions such as epileptic seizure and mental retardation. Together, these lines of information have shown that CUB and CCP domain-containing proteins contribute to a wide variety of neuronal events that ultimately establish neural circuits.


Asunto(s)
Diferenciación Celular , Plasticidad Neuronal/fisiología , Receptores de Neurotransmisores/metabolismo , Sinapsis/fisiología , Transmisión Sináptica/fisiología , Animales , Enzimas Activadoras de Complemento/química , Enzimas Activadoras de Complemento/metabolismo , Humanos
10.
J Neurosci Res ; 83(8): 1461-70, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16652343

RESUMEN

Microglia perform both neuroprotective and neurotoxic functions in the brain, with this depending on their state of activation and their release of mediators. Upon P2X(7) receptor stimulation, for example, microglia release small amounts of TNF, which protect neurons, whereas LPS causes massive TNF release leading to neuroinflammation. Here we report that, in rat primary cultured microglia, nicotine enhances P2X(7) receptor-mediated TNF release, whilst suppressing LPS-induced TNF release but without affecting TNF mRNA expression via activation of alpha7 nicotinic acetylcholine receptors (alpha7 nAChRs). In microglia, nicotine elicited a transient increase in intracellular Ca(2+) levels, which was abolished by specific blockers of alpha7 nAChRs. However, this response was independent of extracellular Ca(2+) and blocked by U73122, an inhibitor of phospholipase C (PLC), and xestospongin C, a blocker of the IP(3) receptor. Repeated experiments showed that currents were not detected in nicotine-stimulated microglia. Moreover, nicotine modulation of LPS-induced TNF release was also blocked by xestospongin C. Upon LPS stimulation, inhibition of TNF release by nicotine was associated with the suppression of JNK and p38 MAP kinase activation, which regulate the post-transcriptional steps of TNF synthesis. In contrast, nicotine did not alter any MAP kinase activation, but enhanced Ca(2+) response in P2X(7) receptor-activated microglia. In conclusion, microglial alpha7 nAChRs might drive a signaling process involving the activation of PLC and Ca(2+) release from intracellular Ca(2+) stores, rather than function as conventional ion channels. This novel alpha7 nAChR signal may be involved in the nicotine modification of microglia activation towards a neuroprotective role by suppressing the inflammatory state and strengthening the protective function.


Asunto(s)
Encefalitis/metabolismo , Gliosis/metabolismo , Inositol 1,4,5-Trifosfato/metabolismo , Microglía/metabolismo , Receptores Nicotínicos/metabolismo , Fosfolipasas de Tipo C/metabolismo , Animales , Animales Recién Nacidos , Canales de Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Técnicas de Cultivo de Célula , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Citoprotección/efectos de los fármacos , Citoprotección/fisiología , Encefalitis/fisiopatología , Inhibidores Enzimáticos/farmacología , Gliosis/fisiopatología , Receptores de Inositol 1,4,5-Trifosfato , Lipopolisacáridos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Microglía/efectos de los fármacos , Nicotina/farmacología , Ratas , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Nicotínicos/efectos de los fármacos , Receptores Purinérgicos P2/efectos de los fármacos , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2X7 , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Fosfolipasas de Tipo C/antagonistas & inhibidores , Receptor Nicotínico de Acetilcolina alfa 7
11.
Dev Biol ; 281(1): 121-33, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15848394

RESUMEN

The nucleolus dynamically alters its shape through the assembly and disassembly of a variety of nucleolar components in proliferating cells. While the nucleolus is known to function in vital cellular events, little is known about how its components are correctly assembled. Through the analysis of a Drosophila mutant that exhibits a reduced number of mushroom body (MB) neurons in the brain, we reveal that the slender lobes (sle) gene encodes a novel nuclear protein that affects nucleolar organization during development. In sle mutant neuroblasts, the nucleolus was packed more tightly, forming a dense sphere, and the nucleolar proteins fibrillarin and Nop60B were abnormally distributed in the interphase nucleolus. Moreover, another nucleolar marker, Aj1 antigen, was localized to the center of the nucleolus in a manner complementary to the Nop60B distribution, and also formed a large aggregate in the cytoplasm. While developmental defects were limited to a few tissues in sle mutants, including MBs and nurse cells, the altered organization of the nucleolar components were evident in most developing tissues. Therefore, we conclude that Sle is a general factor of nuclear architecture in Drosophila that is required for the correct organization of the nucleolus during development.


Asunto(s)
Nucléolo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Cuerpos Pedunculados/crecimiento & desarrollo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Ciclo Celular , Nucléolo Celular/ultraestructura , Proliferación Celular , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Inmunohistoquímica , Cuerpos Pedunculados/citología , Mutación , Fenotipo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA