Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Genet ; 16(11): e1009120, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33137117

RESUMO

Animals typically avoid unwanted situations with stereotyped escape behavior. For instance, Drosophila larvae often escape from aversive stimuli to the head, such as mechanical stimuli and blue light irradiation, by backward locomotion. Responses to these aversive stimuli are mediated by a variety of sensory neurons including mechanosensory class III da (C3da) sensory neurons and blue-light responsive class IV da (C4da) sensory neurons and Bolwig's organ (BO). How these distinct sensory pathways evoke backward locomotion at the circuit level is still incompletely understood. Here we show that a pair of cholinergic neurons in the subesophageal zone, designated AMBs, evoke robust backward locomotion upon optogenetic activation. Anatomical and functional analysis shows that AMBs act upstream of MDNs, the command-like neurons for backward locomotion. Further functional analysis indicates that AMBs preferentially convey aversive blue light information from C4da neurons to MDNs to elicit backward locomotion, whereas aversive information from BO converges on MDNs through AMB-independent pathways. We also found that, unlike in adult flies, MDNs are dispensable for the dead end-evoked backward locomotion in larvae. Our findings thus reveal the neural circuits by which two distinct blue light-sensing pathways converge on the command-like neurons to evoke robust backward locomotion, and suggest that distinct but partially redundant neural circuits including the command-like neurons might be utilized to drive backward locomotion in response to different sensory stimuli as well as in adults and larvae.


Assuntos
Neurônios Colinérgicos/fisiologia , Drosophila melanogaster/fisiologia , Reação de Fuga/fisiologia , Células Receptoras Sensoriais/fisiologia , Comportamento Estereotipado/fisiologia , Vias Aferentes/fisiologia , Animais , Animais Geneticamente Modificados , Proteínas de Bactérias/genética , Encéfalo/fisiologia , Channelrhodopsins/genética , Proteínas de Drosophila/genética , Reação de Fuga/efeitos da radiação , Feminino , Larva/fisiologia , Luz , Proteínas Luminescentes/genética , Masculino , Optogenética , Comportamento Estereotipado/efeitos da radiação , Fatores de Transcrição/genética
2.
PLoS Genet ; 16(8): e1008942, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32764744

RESUMO

To remodel functional neuronal connectivity, neurons often alter dendrite arbors through elimination and subsequent regeneration of dendritic branches. However, the intrinsic mechanisms underlying this developmentally programmed dendrite regeneration and whether it shares common machinery with injury-induced regeneration remain largely unknown. Drosophila class IV dendrite arborization (C4da) sensory neurons regenerate adult-specific dendrites after eliminating larval dendrites during metamorphosis. Here we show that the microRNA miR-87 is a critical regulator of dendrite regeneration in Drosophila. miR-87 knockout impairs dendrite regeneration after developmentally-programmed pruning, whereas miR-87 overexpression in C4da neurons leads to precocious initiation of dendrite regeneration. Genetic analyses indicate that the transcriptional repressor Tramtrack69 (Ttk69) is a functional target for miR-87-mediated repression as ttk69 expression is increased in miR-87 knockout neurons and reducing ttk69 expression restores dendrite regeneration to mutants lacking miR-87 function. We further show that miR-87 is required for dendrite regeneration after acute injury in the larval stage, providing a mechanistic link between developmentally programmed and injury-induced dendrite regeneration. These findings thus indicate that miR-87 promotes dendrite regrowth during regeneration at least in part through suppressing Ttk69 in Drosophila sensory neurons and suggest that developmental and injury-induced dendrite regeneration share a common intrinsic mechanism to reactivate dendrite growth.


Assuntos
Proteínas de Drosophila/genética , Metamorfose Biológica/genética , MicroRNAs/genética , Regeneração Nervosa/genética , Proteínas Repressoras/genética , Animais , Dendritos/genética , Dendritos/fisiologia , Drosophila melanogaster/genética , Regulação da Expressão Gênica no Desenvolvimento , Larva/genética , Larva/crescimento & desenvolvimento , Células Receptoras Sensoriais/metabolismo
3.
Genes Dev ; 29(16): 1763-75, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26302791

RESUMO

Sensory neurons with common functions are often nonrandomly arranged and form dendritic territories in stereotypic spatial patterns throughout the nervous system, yet molecular mechanisms of how neurons specify dendritic territories remain largely unknown. In Drosophila larvae, dendrites of class IV sensory (C4da) neurons completely but nonredundantly cover the whole epidermis, and the boundaries of these tiled dendritic fields are specified through repulsive interactions between homotypic dendrites. Here we report that, unlike the larval C4da neurons, adult C4da neurons rely on both dendritic repulsive interactions and external positional cues to delimit the boundaries of their dendritic fields. We identify Wnt5 derived from sternites, the ventral-most part of the adult abdominal epidermis, as the critical determinant for the ventral boundaries. Further genetic data indicate that Wnt5 promotes dendrite termination on the periphery of sternites through the Ryk receptor family kinase Derailed (Drl) and the Rho GTPase guanine nucleotide exchange factor Trio in C4da neurons. Our findings thus uncover the dendritic contact-independent mechanism that is required for dendritic boundary specification and suggest that combinatory actions of the dendritic contact-dependent and -independent mechanisms may ensure appropriate dendritic territories of a given neuron.


Assuntos
Dendritos , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Células Receptoras Sensoriais , Transdução de Sinais , Proteínas Wnt/metabolismo , Animais , Dendritos/genética , Dendritos/metabolismo , Drosophila melanogaster/enzimologia , Drosophila melanogaster/genética , Células Epidérmicas , Epiderme/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/metabolismo
4.
J Biol Chem ; 288(7): 4922-34, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23269685

RESUMO

Type IV P-type ATPases (P4-ATPases) and CDC50 family proteins form a putative phospholipid flippase complex that mediates the translocation of aminophospholipids such as phosphatidylserine (PS) and phosphatidylethanolamine (PE) from the outer to inner leaflets of the plasma membrane. In Chinese hamster ovary (CHO) cells, at least eight members of P4-ATPases were identified, but only a single CDC50 family protein, CDC50A, was expressed. We demonstrated that CDC50A associated with and recruited P4-ATPase ATP8A1 to the plasma membrane. Overexpression of CDC50A induced extensive cell spreading and greatly enhanced cell migration. Depletion of either CDC50A or ATP8A1 caused a severe defect in the formation of membrane ruffles, thereby inhibiting cell migration. Analyses of phospholipid translocation at the plasma membrane revealed that the depletion of CDC50A inhibited the inward translocation of both PS and PE, whereas the depletion of ATP8A1 inhibited the translocation of PE but not that of PS, suggesting that the inward translocation of cell-surface PE is involved in cell migration. This hypothesis was further examined by using a PE-binding peptide and a mutant cell line with defective PE synthesis; either cell-surface immobilization of PE by the PE-binding peptide or reduction in the cell-surface content of PE inhibited the formation of membrane ruffles, causing a severe defect in cell migration. These results indicate that the phospholipid flippase complex of ATP8A1 and CDC50A plays a major role in cell migration and suggest that the flippase-mediated translocation of PE at the plasma membrane is involved in the formation of membrane ruffles to promote cell migration.


Assuntos
Adenosina Trifosfatases/química , Regulação da Expressão Gênica , Proteínas de Membrana/química , Proteínas de Transferência de Fosfolipídeos/química , Fosfolipídeos/química , Alelos , Animais , Células CHO , Membrana Celular/metabolismo , Movimento Celular , Cricetinae , Bicamadas Lipídicas/química , Lipídeos de Membrana/química , Fosfatidiletanolaminas/química , Fosfatidilserinas/química , Transporte Proteico
5.
Proc Natl Acad Sci U S A ; 108(48): 19389-94, 2011 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-22084112

RESUMO

The axonal projection pattern of sensory neurons typically is regulated by environmental signals, but how different sensory afferents can establish distinct projections in the same environment remains largely unknown. Drosophila class IV dendrite arborization (C4da) sensory neurons project subtype-specific axonal branches in the ventral nerve cord, and we show that the Tripartite motif protein, Anomalies in sensory axon patterning (Asap) is a critical determinant of the axonal projection patterns of different C4da neurons. Asap is highly expressed in C4da neurons with both ipsilateral and contralateral axonal projections, but the Asap level is low in neurons that have only ipsilateral projections. Mutations in asap cause a specific loss of contralateral projections, whereas overexpression of Asap induces ectopic contralateral projections in C4da neurons. We also show by biochemical and genetic analysis that Asap regulates Netrin signaling, at least in part by linking the Netrin receptor Frazzled to the downstream effector Pico. In the absence of Asap, the sensory afferent connectivity within the ventral nerve cord is disrupted, resulting in specific larval behavioral deficits. These results indicate that different levels of Asap determine distinct patterns of axonal projections of C4da neurons by modulating Netrin signaling and that the Asap-mediated axonal projection is critical for assembly of a functional sensory circuit.


Assuntos
Axônios/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/fisiologia , Células Receptoras Sensoriais/fisiologia , Transdução de Sinais/fisiologia , Animais , Axônios/fisiologia , Proteínas de Drosophila/genética , Imunoprecipitação , Proteínas do Tecido Nervoso/genética , Receptores de Netrina , Receptores de Superfície Celular/metabolismo , Células Receptoras Sensoriais/citologia , Transdução de Sinais/genética , Técnicas do Sistema de Duplo-Híbrido
6.
EMBO J ; 28(24): 3879-92, 2009 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-19875983

RESUMO

To cover the receptive field completely and non-redundantly, neurons of certain functional groups arrange tiling of their dendrites. In Drosophila class IV dendrite arborization (da) neurons, the NDR family kinase Tricornered (Trc) is required for homotypic repulsion of dendrites that facilitates dendritic tiling. We here report that Sin1, Rictor, and target of rapamycin (TOR), components of the TOR complex 2 (TORC2), are required for dendritic tiling of class IV da neurons. Similar to trc mutants, dendrites of sin1 and rictor mutants show inappropriate overlap of the dendritic fields. TORC2 components physically and genetically interact with Trc, consistent with a shared role in regulating dendritic tiling. Moreover, TORC2 is essential for Trc phosphorylation on a residue that is critical for Trc activity in vivo and in vitro. Remarkably, neuronal expression of a dominant active form of Trc rescues the tiling defects in sin1 and rictor mutants. These findings suggest that TORC2 likely acts together with the Trc signalling pathway to regulate the dendritic tiling of class IV da neurons, and thus uncover the first neuronal function of TORC2 in vivo.


Assuntos
Dendritos/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/metabolismo , Neurônios/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Proteínas de Transporte/metabolismo , Cruzamentos Genéticos , Células HeLa , Humanos , Mutação , Fenótipo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Quinases , Proteína Companheira de mTOR Insensível à Rapamicina , Células Receptoras Sensoriais/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR
7.
Genes Cells ; 17(8): 728-36, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22747682

RESUMO

Phosphatidylserine (PS), a relatively minor constituent in the plasma membrane (PM), participates in various cellular processes such as clearance of apoptotic cells and recruitment of signaling molecules. PS also localizes in the membranes of endocytic organelles, such as recycling endosomes (REs). We recently showed that in REs, PS binds to the pleckstrin homology (PH) domain of evectin-2, thereby regulating retrograde traffic from REs to the Golgi. However, direct evidence that PS has a role in retrograde traffic is lacking. Here, we examined the contribution of PS to endosomal membrane traffic by exploiting a mutant CHO cell line (PSA-3) that is defective in PS synthesis. In PSA-3 cells, the Golgi localization of TGN38, a protein that circulates between the Golgi and the PM through endosomes by retrograde traffic, was abolished, whereas the localizations of other organelle markers remained unchanged. Increasing the cellular PS level by adding ethanolamine to the culture medium restored the Golgi localization of TGN38. Tracking the endocytic fate of cell surface TGN38 that was labeled by anti-TGN38 antibody showed that retrograde transport of TGN38 was impaired at endosomes, not at the PM. These findings provide direct evidence that intracellular PS is required for retrograde traffic through endosomes.


Assuntos
Endocitose , Endossomos/metabolismo , Proteínas de Membrana/metabolismo , Fosfatidilserinas/biossíntese , Animais , Células CHO , Membrana Celular/metabolismo , Cricetinae , Meios de Cultura/metabolismo , Citoplasma/metabolismo , Endossomos/efeitos dos fármacos , Etanolamina/farmacologia , Complexo de Golgi/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Fosfatidilserinas/metabolismo , Transporte Proteico , Ratos , Transfecção
8.
FASEB J ; 26(8): 3230-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22522110

RESUMO

The protein kinase liver kinase B1 (LKB1) regulates cell polarity and intercellular junction stability. Also, LKB1 controls the activity of salt-inducible kinase 1 (SIK1). The role and relevance of SIK1 and its downstream effectors in linking the LKB1 signals within these processes are partially understood. We hypothesize that SIK1 may link LKB1 signals to the maintenance of epithelial junction stability by regulating E-cadherin expression. Results from our studies using a mouse lung alveolar epithelial (MLE-12) cell line or human renal proximal tubule (HK2) cell line transiently or stably lacking the expression of SIK1 (using SIK1 siRNAs or shRNAs), or with its expression abrogated (sik1(+/+) vs. sik1(-/-) mice), indicate that suppression of SIK1 (∼40%) increases the expression of the transcriptional repressors Snail2 (∼12-fold), Zeb1 (∼100%), Zeb2 (∼50%), and TWIST (∼20-fold) by activating cAMP-response element binding protein. The lack of SIK1 and activation of transcriptional repressors decreases the availability of E-cadherin (mRNA and protein expression by ∼100 and 80%, respectively) and the stability of intercellular junctions in epithelia (decreases in transepithelial resistance). Furthermore, LKB1-mediated increases in E-cadherin expression are impaired in cells where SIK1 has been disabled. We conclude that SIK1 is a key regulator of E-cadherin expression, and thereby contributes to the stability of intercellular junctions.


Assuntos
Caderinas/biossíntese , Junções Intercelulares/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Quinases Proteína-Quinases Ativadas por AMP , Animais , Linhagem Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Humanos , Junções Intercelulares/metabolismo , Túbulos Renais Proximais/metabolismo , Camundongos , Fatores de Transcrição da Família Snail , Fatores de Transcrição/biossíntese
9.
Nat Commun ; 14(1): 3987, 2023 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-37443364

RESUMO

Animals must adapt sensory responses to an ever-changing environment for survival. Such sensory modulation is especially critical in a threatening situation, in which animals often promote aversive responses to, among others, visual stimuli. Recently, threatened Drosophila has been shown to exhibit a defensive internal state. Whether and how threatened Drosophila promotes visual aversion, however, remains elusive. Here we report that mechanical threats to Drosophila transiently gate aversion from an otherwise neutral visual object. We further identified the neuropeptide tachykinin, and a single cluster of neurons expressing it ("Tk-GAL42 ∩ Vglut neurons"), that are responsible for gating visual aversion. Calcium imaging analysis revealed that mechanical threats are encoded in Tk-GAL42 ∩ Vglut neurons as elevated activity. Remarkably, we also discovered that a visual object is encoded in Tk-GAL42 ∩ Vglut neurons as θ oscillation, which is causally linked to visual aversion. Our data reveal how a single cluster of neurons adapt organismal sensory response to a threatening situation through a neuropeptide and a combination of rate/temporal coding schemes.


Assuntos
Proteínas de Drosophila , Neuropeptídeos , Animais , Drosophila melanogaster/fisiologia , Drosophila , Neuropeptídeos/genética , Neurônios/fisiologia
10.
Commun Biol ; 6(1): 332, 2023 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-36973364

RESUMO

The daily activity in the brain is typically fine-tuned by the circadian clock in the local neurons as well as by the master circadian clock in the suprachiasmatic nucleus (SCN) of the hypothalamus. In the olfactory response, odor-evoked activity in the piriform cortex (PC) and olfactory behavior retain circadian rhythmicity in the absence of the SCN, yet how the circadian rhythm in the PC is achieved independently of the SCN remains elusive. Here, to define neurons regulating the circadian rhythm of the odor-evoked activity in the PC, we knocked out the clock gene Bmal1 in a host of specific neurons along the olfactory circuit. We discovered that Bmal1 knockout in the PC largely abolishes the circadian rhythm of the odor-evoked activity. We further showed that isolated PC exhibits sustained circadian rhythms of the clock gene Per2 expression. Quantitative PCR analysis revealed that expression patterns of multiple genes involved in neural activity and synaptic transmission exhibit circadian rhythm in the PC in a BMAL1-dependent manner. Our findings indicate that BMAL1 acts intrinsically in the PC to control the circadian rhythm of the odor-evoked activity in the PC, possibly through regulating expression patterns of multiple genes involved in neural activity and transmission.


Assuntos
Relógios Circadianos , Córtex Piriforme , Relógios Circadianos/genética , Odorantes , Fatores de Transcrição ARNTL/genética , Ritmo Circadiano/genética
11.
Science ; 381(6663): 1197-1205, 2023 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-37708280

RESUMO

Inactivation of the ubiquitin ligase Ube3a causes the developmental disorder Angelman syndrome, whereas increased Ube3a dosage is associated with autism spectrum disorders. Despite the enriched localization of Ube3a in the axon terminals including presynapses, little is known about the presynaptic function of Ube3a and mechanisms underlying its presynaptic localization. We show that developmental synapse elimination requires presynaptic Ube3a activity in Drosophila neurons. We further identified the domain of Ube3a that is required for its interaction with the kinesin motor. Angelman syndrome-associated missense mutations in the interaction domain attenuate presynaptic targeting of Ube3a and prevent synapse elimination. Conversely, increased Ube3a activity in presynapses leads to precocious synapse elimination and impairs synaptic transmission. Our findings reveal the physiological role of Ube3a and suggest potential pathogenic mechanisms associated with Ube3a dysregulation.


Assuntos
Síndrome de Angelman , Transtorno do Espectro Autista , Proteínas de Drosophila , Drosophila melanogaster , Transmissão Sináptica , Ubiquitina-Proteína Ligases , Animais , Síndrome de Angelman/enzimologia , Síndrome de Angelman/genética , Transtorno do Espectro Autista/enzimologia , Transtorno do Espectro Autista/genética , Regulação para Baixo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Sinapses/enzimologia , Sinapses/genética
12.
J Biosci Bioeng ; 136(3): 239-245, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37344278

RESUMO

This study describes a co-culture system of human skin equivalents (HSEs) and dorsal root ganglion (DRG) neurons. We prepared spheroids of mouse DRG neurons with or without Schwann cells (SCs). Spheroids comprising DRG neurons and SCs showed longer neurite extensions than those comprising DRG neurons alone. Neurite extension of more than 1 mm was observed from spheroids cultured inside HSEs, whereas neurite extension was primarily observed on the surface of HSEs from spheroids cultured on HSEs. We propose that our model may be a useful tool for studying neurite extension in the human skin.


Assuntos
Neuritos , Neurônios , Humanos , Camundongos , Animais , Técnicas de Cocultura , Neuritos/fisiologia , Células de Schwann , Células Cultivadas
13.
Neuroscience ; 521: 102-109, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37142179

RESUMO

Social buffering is a phenomenon where stress responses are ameliorated by an affiliative conspecific. Our previous findings suggest that the posterior complex of the anterior olfactory nucleus (AOP) is well positioned to participate in the neural mechanisms underlying social buffering. However, the lack of anatomical information prevents us from further estimating the role of the AOP. Here, we obtained anatomical information regarding the AOP in male rats. In Experiment 1 (n = 5), among 4',6-diamidino-2-phenylindole-positive cells in the AOP, the proportion of glutamic acid decarboxylase 67 (GAD67)-positive cells was 13.8% ± 1.2%. In Experiment 2 (n = 5), among the cells that were labeled by a retrograde tracer injected into the basolateral complex of the amygdala (BLA), the proportion of GAD67-positive cells was 18.6% ± 0.8%. In Experiment 3 (n = 5), we demonstrated the existence of cells that were labeled by the retrograde tracer injected into the posterior part of the medial amygdala (MeP), mostly into the ventral part of the MeP. In addition, the proportion of GAD67-positive cells among the tracer-labeled cells was 21.7% ± 1.7%. In Experiment 4 (n = 3), the retrograde tracers were injected into the BLA and MeP, mostly into the ventral part of the MeP. The proportion of double-labeled cells among the tracer-labeled cells was 2.1% ± 1.2%. Taken together, these results suggest that the AOP is predominantly composed of glutamatergic neurons. In addition, the AOP sends mutually independent glutamatergic-predominant projections to the BLA and MeP.


Assuntos
Tonsila do Cerebelo , Córtex Olfatório , Ratos , Masculino , Animais , Tonsila do Cerebelo/fisiologia , Vias Neurais
14.
Nat Commun ; 14(1): 6515, 2023 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-37845214

RESUMO

Although painful stimuli elicit defensive responses including escape behavior for survival, starved animals often prioritize feeding over escape even in a noxious environment. This behavioral priority is typically mediated by suppression of noxious inputs through descending control in the brain, yet underlying molecular and cellular mechanisms are incompletely understood. Here we identify a cluster of GABAergic neurons in Drosophila larval brain, designated as SEZ-localized Descending GABAergic neurons (SDGs), that project descending axons onto the axon terminals of the peripheral nociceptive neurons and prevent presynaptic activity through GABAB receptors. Remarkably, glucose feeding to starved larvae causes sustained activation of SDGs through glucose-sensing neurons and subsequent insulin signaling in SDGs, which attenuates nociception and thereby suppresses escape behavior in response to multiple noxious stimuli. These findings illustrate a neural mechanism by which sugar sensing neurons in the brain engages descending GABAergic neurons in nociceptive gating to achieve hierarchical interaction between feeding and escape behavior.


Assuntos
Drosophila , Açúcares , Animais , Nociceptividade/fisiologia , Larva/fisiologia , Receptores de GABA-B , Encéfalo , Glucose
15.
Commun Biol ; 6(1): 440, 2023 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-37085665

RESUMO

RAB35 is a multifunctional small GTPase that regulates endocytic recycling, cytoskeletal rearrangement, and cytokinesis. However, its physiological functions in mammalian development remain unclear. Here, we generated Rab35-knockout mice and found that RAB35 is essential for early embryogenesis. Interestingly, brain-specific Rab35-knockout mice displayed severe defects in hippocampal lamination owing to impaired distribution of pyramidal neurons, although defects in cerebral cortex formation were not evident. In addition, Rab35-knockout mice exhibited defects in spatial memory and anxiety-related behaviors. Quantitative proteomics indicated that the loss of RAB35 significantly affected the levels of other RAB proteins associated with endocytic trafficking, as well as some neural cell adhesion molecules, such as contactin-2. Collectively, our findings revealed that RAB35 is required for precise neuronal distribution in the developing hippocampus by regulating the expression of cell adhesion molecules, thereby influencing spatial memory.


Assuntos
Hipocampo , Neurônios , Proteínas rab de Ligação ao GTP , Animais , Camundongos , Transporte Biológico , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Mamíferos , Camundongos Knockout , Neurônios/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo
16.
Nature ; 443(7108): 210-3, 2006 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-16906135

RESUMO

Precise patterning of dendritic fields is essential for neuronal circuit formation and function, but how neurons establish and maintain their dendritic fields during development is poorly understood. In Drosophila class IV dendritic arborization neurons, dendritic tiling, which allows for the complete but non-overlapping coverage of the dendritic fields, is established through a 'like-repels-like' behaviour of dendrites mediated by Tricornered (Trc), one of two NDR (nuclear Dbf2-related) family kinases in Drosophila. Here we report that the other NDR family kinase, the tumour suppressor Warts/Lats (Wts), regulates the maintenance of dendrites; in wts mutants, dendrites initially tile the body wall normally, but progressively lose branches at later larval stages, whereas the axon shows no obvious defects. We further provide biochemical and genetic evidence for the tumour suppressor kinase Hippo (Hpo) as an upstream regulator of Wts and Trc for dendrite maintenance and tiling, respectively, thereby revealing important functions of tumour suppressor genes of the Hpo signalling pathway in dendrite morphogenesis.


Assuntos
Dendritos/enzimologia , Dendritos/fisiologia , Proteínas de Drosophila/metabolismo , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Contagem de Células , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Forma Celular , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Drosophila melanogaster/enzimologia , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Larva/citologia , Larva/crescimento & desenvolvimento , Larva/metabolismo , Morfogênese , Mutação/genética , Fosforilação , Fosfotreonina/metabolismo , Ligação Proteica , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases/química , Treonina/metabolismo
17.
Mol Brain ; 15(1): 70, 2022 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-35941689

RESUMO

Adeno-associated virus (AAV) vector is a critical tool for gene delivery through its durable transgene expression and safety profile. Among many serotypes, AAV2-retro is typically utilized for dissecting neural circuits with its retrograde functionality. However, this vector requires a relatively long-term incubation period (over 2 weeks) to obtain enough gene expression levels presumably due to low efficiency in gene transduction. Here, we aimed to enhance transgene expression efficiency of AAV2-retro vectors by substituting multiple tyrosine residues with phenylalanines (YF mutations) in the virus capsid, which is previously reported to improve the transduction efficiency of AAV2-infected cells by evading host cell responses. We found that AAV2-retro with YF mutations (AAV2-retroYF)-mediated transgene expression was significantly enhanced in the primary culture of murine cortical neurons at 1 week after application, comparable to that of the conventional AAV2-retro at 2 week after application. Moreover, transgene expressions in the retrogradely labeled neurons mediated by AAV2-retroYF were significantly increased both in the cortico-cortical circuits and in the subcortical circuits in vivo, while the retrograde functionality of AAV2-retroYF was equally effective as that of AAV2-retro. Our data indicate that YF mutations boost AAV2-retro-mediated retrograde gene transduction in vivo and suggest that the AAV2-retroYF should be useful for efficient targeting of the projection-defined neurons, which is suited to applications for dissecting neural circuits during development as well as future clinical applications.


Assuntos
Capsídeo , Dependovirus , Animais , Dependovirus/genética , Vetores Genéticos , Camundongos , Mutação/genética , Transdução Genética , Tirosina/genética
18.
Neuron ; 54(3): 403-16, 2007 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-17481394

RESUMO

A neuron's dendrites typically do not cross one another. This intrinsic self-avoidance mechanism ensures unambiguous processing of sensory or synaptic inputs. Moreover, some neurons respect the territory of others of the same type, a phenomenon known as tiling. Different types of neurons, however, often have overlapping dendritic fields. We found that Down's syndrome Cell Adhesion Molecule (Dscam) is required for dendritic self-avoidance of all four classes of Drosophila dendritic arborization (da) neurons. However, neighboring mutant class IV da neurons still exhibited tiling, suggesting that self-avoidance and tiling differ in their recognition and repulsion mechanisms. Introducing 1 of the 38,016 Dscam isoforms to da neurons in Dscam mutants was sufficient to significantly restore self-avoidance. Remarkably, expression of a common Dscam isoform in da neurons of different classes prevented their dendrites from sharing the same territory, suggesting that coexistence of dendritic fields of different neuronal classes requires divergent expression of Dscam isoforms.


Assuntos
Dendritos/fisiologia , Proteínas de Drosophila/fisiologia , Neurônios Aferentes/fisiologia , Animais , Animais Geneticamente Modificados , Moléculas de Adesão Celular , Forma Celular/fisiologia , Dendritos/ultraestrutura , Drosophila , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Mutação/fisiologia , Neurônios Aferentes/classificação , Neurônios Aferentes/citologia , Órgãos dos Sentidos/citologia , Coloração e Rotulagem
19.
Dev Growth Differ ; 53(3): 277-86, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21492146

RESUMO

One of the most important features of neuronal function is the capacity to dynamically adapt in response to changes in the environment and neuronal activity. Among cellular elements that show this kind of plasticity are dendrites, the components that receive and process neuronal inputs. Dendrite remodeling occurs during normal development of the nervous system as well as in response to injury or diseases in the adult. In either case, selective stabilization and/or elimination of dendritic branches is likely important to shape dendritic arbors. Here I review examples of the phenomena and consider potential cellular and molecular mechanisms that underlie dendrite remodeling and how they might relate in development and disease.


Assuntos
Dendritos/metabolismo , Doenças do Sistema Nervoso/metabolismo , Sistema Nervoso/metabolismo , Plasticidade Neuronal , Animais , Humanos , Sistema Nervoso/patologia , Sistema Nervoso/fisiopatologia , Doenças do Sistema Nervoso/patologia , Doenças do Sistema Nervoso/fisiopatologia
20.
Neurosci Res ; 167: 54-63, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33309868

RESUMO

Developmental neurite pruning is a process by which neurons selectively eliminate unnecessary processes of axons and/or dendrites without cell death, which shapes the mature wiring of nervous systems. In this sense, developmental neurite pruning requires spatiotemporally precise control of local degradation of cellular components including cytoskeletons and membranes. The Drosophila nervous system undergoes large-scale remodeling, including axon/dendrite pruning, during metamorphosis. In addition to this unique phenomenon in the nervous system, powerful genetic tools make the Drosophila nervous system a sophisticated model to investigate spatiotemporal regulation of neural remodeling. This article reviews recent advances to our understanding of the molecular and cellular mechanisms of developmental axon/dendrite pruning, mainly focusing on studies in Drosophila sensory neurons and mushroom body neurons.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Dendritos/metabolismo , Proteínas de Drosophila/genética , Regulação da Expressão Gênica no Desenvolvimento , Corpos Pedunculados , Plasticidade Neuronal
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA