Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Neurosci ; 17: 1236815, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37564364

RESUMO

Axons are processes of neurons, up to a metre long, that form the essential biological cables wiring nervous systems. They must survive, often far away from their cell bodies and up to a century in humans. This requires self-sufficient cell biology including structural proteins, organelles, and membrane trafficking, metabolic, signalling, translational, chaperone, and degradation machinery-all maintaining the homeostasis of energy, lipids, proteins, and signalling networks including reactive oxygen species and calcium. Axon maintenance also involves specialised cytoskeleton including the cortical actin-spectrin corset, and bundles of microtubules that provide the highways for motor-driven transport of components and organelles for virtually all the above-mentioned processes. Here, we aim to provide a conceptual overview of key aspects of axon biology and physiology, and the homeostatic networks they form. This homeostasis can be derailed, causing axonopathies through processes of ageing, trauma, poisoning, inflammation or genetic mutations. To illustrate which malfunctions of organelles or cell biological processes can lead to axonopathies, we focus on axonopathy-linked subcellular defects caused by genetic mutations. Based on these descriptions and backed up by our comprehensive data mining of genes linked to neural disorders, we describe the 'dependency cycle of local axon homeostasis' as an integrative model to explain why very different causes can trigger very similar axonopathies, providing new ideas that can drive the quest for strategies able to battle these devastating diseases.

2.
Front Physiol ; 14: 1111244, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37256074

RESUMO

The sensitivity of animals to sensory input must be regulated to ensure that signals are detected and also discriminable. However, how circuits regulate the dynamic range of sensitivity to sensory stimuli is not well understood. A given odor is represented in the insect mushroom bodies (MBs) by sparse combinatorial coding by Kenyon cells (KCs), forming an odor quality representation. To address how intensity of sensory stimuli is processed at the level of the MB input region, the calyx, we characterized a set of novel mushroom body output neurons that respond preferentially to high odor concentrations. We show that a pair of MB calyx output neurons, MBON-a1/2, are postsynaptic in the MB calyx, where they receive extensive synaptic inputs from KC dendrites, the inhibitory feedback neuron APL, and octopaminergic sVUM1 neurons, but relatively few inputs from projection neurons. This pattern is broadly consistent in the third-instar larva as well as in the first instar connectome. MBON-a1/a2 presynaptic terminals innervate a region immediately surrounding the MB medial lobe output region in the ipsilateral and contralateral brain hemispheres. By monitoring calcium activity using jRCamP1b, we find that MBON-a1/a2 responses are odor-concentration dependent, responding only to ethyl acetate (EA) concentrations higher than a 200-fold dilution, in contrast to MB neurons which are more concentration-invariant and respond to EA dilutions as low as 10-4. Optogenetic activation of the calyx-innervating sVUM1 modulatory neurons originating in the SEZ (Subesophageal zone), did not show a detectable effect on MBON-a1/a2 odor responses. Optogenetic activation of MBON-a1/a2 using CsChrimson impaired odor discrimination learning compared to controls. We propose that MBON-a1/a2 form an output channel of the calyx, summing convergent sensory and modulatory input, firing preferentially to high odor concentration, and might affect the activity of downstream MB targets.

3.
J Cell Biol ; 222(6)2023 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-36952540

RESUMO

Neuronal endoplasmic reticulum (ER) appears continuous throughout the cell. Its shape and continuity are influenced by ER-shaping proteins, mutations in which can cause distal axon degeneration in Hereditary Spastic Paraplegia (HSP). We therefore asked how loss of Rtnl1, a Drosophila ortholog of the human HSP gene RTN2 (SPG12), which encodes an ER-shaping protein, affects ER organization and the function of presynaptic terminals. Loss of Rtnl1 depleted ER membrane markers at Drosophila presynaptic motor terminals and appeared to deplete narrow tubular ER while leaving cisternae largely unaffected, thus suggesting little change in resting Ca2+ storage capacity. Nevertheless, these changes were accompanied by major reductions in activity-evoked Ca2+ fluxes in the cytosol, ER lumen, and mitochondria, as well as reduced evoked and spontaneous neurotransmission. We found that reduced STIM-mediated ER-plasma membrane contacts underlie presynaptic Ca2+ defects in Rtnl1 mutants. Our results show the importance of ER architecture in presynaptic physiology and function, which are therefore potential factors in the pathology of HSP.


Assuntos
Cálcio , Proteínas de Drosophila , Drosophila , Retículo Endoplasmático , Proteínas de Membrana , Animais , Humanos , Cálcio/metabolismo , Proteínas de Drosophila/genética , Retículo Endoplasmático/metabolismo , Proteínas de Membrana/genética , Paraplegia Espástica Hereditária/genética , Paraplegia Espástica Hereditária/patologia
4.
Front Physiol ; 14: 1076533, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36776967

RESUMO

As a model organism, Drosophila is uniquely placed to contribute to our understanding of how brains control complex behavior. Not only does it have complex adaptive behaviors, but also a uniquely powerful genetic toolkit, increasingly complete dense connectomic maps of the central nervous system and a rapidly growing set of transcriptomic profiles of cell types. But this also poses a challenge: Given the massive amounts of available data, how are researchers to Find, Access, Integrate and Reuse (FAIR) relevant data in order to develop an integrated anatomical and molecular picture of circuits, inform hypothesis generation, and find reagents for experiments to test these hypotheses? The Virtual Fly Brain (virtualflybrain.org) web application & API provide a solution to this problem, using FAIR principles to integrate 3D images of neurons and brain regions, connectomics, transcriptomics and reagent expression data covering the whole CNS in both larva and adult. Users can search for neurons, neuroanatomy and reagents by name, location, or connectivity, via text search, clicking on 3D images, search-by-image, and queries by type (e.g., dopaminergic neuron) or properties (e.g., synaptic input in the antennal lobe). Returned results include cross-registered 3D images that can be explored in linked 2D and 3D browsers or downloaded under open licenses, and extensive descriptions of cell types and regions curated from the literature. These solutions are potentially extensible to cover similar atlasing and data integration challenges in vertebrates.

6.
Learn Mem ; 28(2): 53-71, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33452115

RESUMO

Discrimination of sensory signals is essential for an organism to form and retrieve memories of relevance in a given behavioral context. Sensory representations are modified dynamically by changes in behavioral state, facilitating context-dependent selection of behavior, through signals carried by noradrenergic input in mammals, or octopamine (OA) in insects. To understand the circuit mechanisms of this signaling, we characterized the function of two OA neurons, sVUM1 neurons, that originate in the subesophageal zone (SEZ) and target the input region of the memory center, the mushroom body (MB) calyx, in larval Drosophila We found that sVUM1 neurons target multiple neurons, including olfactory projection neurons (PNs), the inhibitory neuron APL, and a pair of extrinsic output neurons, but relatively few mushroom body intrinsic neurons, Kenyon cells. PN terminals carried the OA receptor Oamb, a Drosophila α1-adrenergic receptor ortholog. Using an odor discrimination learning paradigm, we showed that optogenetic activation of OA neurons compromised discrimination of similar odors but not learning ability. Our results suggest that sVUM1 neurons modify odor representations via multiple extrinsic inputs at the sensory input area to the MB olfactory learning circuit.


Assuntos
Comportamento Animal/fisiologia , Discriminação Psicológica/fisiologia , Larva/fisiologia , Aprendizagem/fisiologia , Corpos Pedunculados/fisiologia , Neurônios/fisiologia , Octopamina/metabolismo , Percepção Olfatória/fisiologia , Animais , Drosophila , Neurônios/metabolismo , Optogenética
7.
Front Neurosci ; 14: 816, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32903680

RESUMO

Genes for endoplasmic reticulum (ER)-shaping proteins are among the most commonly mutated in hereditary spastic paraplegia (HSP). Mutation of these genes in model organisms can lead to disruption of the ER network. To investigate how the physiological roles of the ER might be affected by such disruption, we developed tools to interrogate its Ca2+ signaling function. We generated GAL4-driven Ca2+ sensors targeted to the ER lumen, to record ER Ca2+ fluxes in identified Drosophila neurons. Using GAL4 lines specific for Type Ib or Type Is larval motor neurons, we compared the responses of different lumenal indicators to electrical stimulation, in axons and presynaptic terminals. The most effective sensor, ER-GCaMP6-210, had a Ca2+ affinity close to the expected ER lumenal concentration. Repetitive nerve stimulation generally showed a transient increase of lumenal Ca2+ in both the axon and presynaptic terminals. Mutants lacking neuronal reticulon and REEP proteins, homologs of human HSP proteins, showed a larger ER lumenal evoked response compared to wild type; we propose mechanisms by which this phenotype could lead to neuronal dysfunction or degeneration. Our lines are useful additions to a Drosophila Ca2+ imaging toolkit, to explore the physiological roles of ER, and its pathophysiological roles in HSP and in axon degeneration more broadly.

8.
Front Neurosci ; 14: 48, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32116502

RESUMO

The physical continuity of axons over long cellular distances poses challenges for their maintenance. One organelle that faces this challenge is endoplasmic reticulum (ER); unlike other intracellular organelles, this forms a physically continuous network throughout the cell, with a single membrane and a single lumen. In axons, ER is mainly smooth, forming a tubular network with occasional sheets or cisternae and low amounts of rough ER. It has many potential roles: lipid biosynthesis, glucose homeostasis, a Ca2+ store, protein export, and contacting and regulating other organelles. This tubular network structure is determined by ER-shaping proteins, mutations in some of which are causative for neurodegenerative disorders such as hereditary spastic paraplegia (HSP). While axonal ER shares many features with the tubular ER network in other contexts, these features must be adapted to the long and narrow dimensions of axons. ER appears to be physically continuous throughout axons, over distances that are enormous on a subcellular scale. It is therefore a potential channel for long-distance or regional communication within neurons, independent of action potentials or physical transport of cargos, but involving its physiological roles such as Ca2+ or organelle homeostasis. Despite its apparent stability, axonal ER is highly dynamic, showing features like anterograde and retrograde transport, potentially reflecting continuous fusion and breakage of the network. Here we discuss the transport processes that must contribute to this dynamic behavior of ER. We also discuss the model that these processes underpin a homeostatic process that ensures both enough ER to maintain continuity of the network and repair breaks in it, but not too much ER that might disrupt local cellular physiology. Finally, we discuss how failure of ER organization in axons could lead to axon degenerative diseases, and how a requirement for ER continuity could make distal axons most susceptible to degeneration in conditions that disrupt ER continuity.

9.
G3 (Bethesda) ; 9(2): 453-462, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30530644

RESUMO

The Drosophila melanogaster larval neuromuscular system is extensively used by researchers to study neuronal cell biology, and Drosophila glutamatergic motor neurons have become a major model system. There are two main Types of glutamatergic motor neurons, Ib and Is, with different structural and physiological properties at synaptic level at the neuromuscular junction. To generate genetic tools to identify and manipulate motor neurons of each Type, we screened for GAL4 driver lines for this purpose. Here we describe GAL4 drivers specific for examples of neurons within each Type, Ib or Is. These drivers showed high expression levels and were expressed in only few motor neurons, making them amenable tools for specific studies of both axonal and synapse biology in identified Type I motor neurons.


Assuntos
Proteínas de Drosophila/genética , Neurônios Motores/metabolismo , Fatores de Transcrição/genética , Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Ácido Glutâmico/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Junção Neuromuscular/metabolismo , Fatores de Transcrição/metabolismo
10.
Elife ; 62017 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-28742022

RESUMO

Axons contain a smooth tubular endoplasmic reticulum (ER) network that is thought to be continuous with ER throughout the neuron; the mechanisms that form this axonal network are unknown. Mutations affecting reticulon or REEP proteins, with intramembrane hairpin domains that model ER membranes, cause an axon degenerative disease, hereditary spastic paraplegia (HSP). We show that Drosophila axons have a dynamic axonal ER network, which these proteins help to model. Loss of HSP hairpin proteins causes ER sheet expansion, partial loss of ER from distal motor axons, and occasional discontinuities in axonal ER. Ultrastructural analysis reveals an extensive ER network in axons, which shows larger and fewer tubules in larvae that lack reticulon and REEP proteins, consistent with loss of membrane curvature. Therefore HSP hairpin-containing proteins are required for shaping and continuity of axonal ER, thus suggesting roles for ER modeling in axon maintenance and function.


Assuntos
Axônios/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Retículo Endoplasmático/metabolismo , Proteínas de Membrana Transportadoras/genética , Paraplegia Espástica Hereditária/genética , Animais , Transporte Axonal , Axônios/ultraestrutura , Modelos Animais de Doenças , Proteínas de Drosophila/deficiência , Drosophila melanogaster/classificação , Drosophila melanogaster/citologia , Drosophila melanogaster/ultraestrutura , Retículo Endoplasmático/ultraestrutura , Expressão Gênica , Humanos , Larva/citologia , Larva/genética , Larva/metabolismo , Larva/ultraestrutura , Proteínas de Membrana Transportadoras/deficiência , Mutação , Filogenia , Isoformas de Proteínas/deficiência , Isoformas de Proteínas/genética , Paraplegia Espástica Hereditária/metabolismo , Paraplegia Espástica Hereditária/patologia
11.
Nat Commun ; 7: 13821, 2016 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-27929117

RESUMO

Aberrant protein aggregation is controlled by various chaperones, including CCT (chaperonin containing TCP-1)/TCP-1/TRiC. Mutated CCT4/5 subunits cause sensory neuropathy and CCT5 expression is decreased in Alzheimer's disease. Here, we show that CCT integrity is essential for autophagosome degradation in cells or Drosophila and this phenomenon is orchestrated by the actin cytoskeleton. When autophagic flux is reduced by compromise of individual CCT subunits, various disease-relevant autophagy substrates accumulate and aggregate. The aggregation of proteins like mutant huntingtin, ATXN3 or p62 after CCT2/5/7 depletion is predominantly autophagy dependent, and does not further increase with CCT knockdown in autophagy-defective cells/organisms, implying surprisingly that the effect of loss-of-CCT activity on mutant ATXN3 or huntingtin oligomerization/aggregation is primarily a consequence of autophagy inhibition rather than loss of physiological anti-aggregation activity for these proteins. Thus, our findings reveal an essential partnership between two key components of the proteostasis network and implicate autophagy defects in diseases with compromised CCT complex activity.


Assuntos
Autofagossomos/metabolismo , Autofagia , Chaperonina com TCP-1/metabolismo , Proteína Huntingtina/metabolismo , Agregação Patológica de Proteínas/metabolismo , Animais , Ataxina-3/metabolismo , Drosophila , Feminino , Células HeLa , Humanos , Lisossomos/metabolismo , Masculino , Camundongos Transgênicos , Proteínas de Ligação a RNA/metabolismo
12.
Nat Neurosci ; 19(10): 1311-20, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27526206

RESUMO

Brain function relies on fast and precisely timed synaptic vesicle (SV) release at active zones (AZs). Efficacy of SV release depends on distance from SV to Ca(2+) channel, but molecular mechanisms controlling this are unknown. Here we found that distances can be defined by targeting two unc-13 (Unc13) isoforms to presynaptic AZ subdomains. Super-resolution and intravital imaging of developing Drosophila melanogaster glutamatergic synapses revealed that the Unc13B isoform was recruited to nascent AZs by the scaffolding proteins Syd-1 and Liprin-α, and Unc13A was positioned by Bruchpilot and Rim-binding protein complexes at maturing AZs. Unc13B localized 120 nm away from Ca(2+) channels, whereas Unc13A localized only 70 nm away and was responsible for docking SVs at this distance. Unc13A(null) mutants suffered from inefficient, delayed and EGTA-supersensitive release. Mathematical modeling suggested that synapses normally operate via two independent release pathways differentially positioned by either isoform. We identified isoform-specific Unc13-AZ scaffold interactions regulating SV-Ca(2+)-channel topology whose developmental tightening optimizes synaptic transmission.


Assuntos
Canais de Cálcio/metabolismo , Proteínas de Transporte/metabolismo , Drosophila melanogaster/metabolismo , Terminações Pré-Sinápticas/metabolismo , Vesículas Sinápticas/metabolismo , Animais , Proteínas de Transporte/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Feminino , Proteínas Ativadoras de GTPase/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Modelos Neurológicos , Mutação , Fosfoproteínas/metabolismo , Isoformas de Proteínas , Proteínas rab3 de Ligação ao GTP/metabolismo
13.
Nat Chem Biol ; 11(5): 347-354, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25848931

RESUMO

Huntington's disease (HD) is a currently incurable neurodegenerative condition caused by an abnormally expanded polyglutamine tract in huntingtin (HTT). We identified new modifiers of mutant HTT toxicity by performing a large-scale 'druggable genome' siRNA screen in human cultured cells, followed by hit validation in Drosophila. We focused on glutaminyl cyclase (QPCT), which had one of the strongest effects on mutant HTT-induced toxicity and aggregation in the cell-based siRNA screen and also rescued these phenotypes in Drosophila. We found that QPCT inhibition induced the levels of the molecular chaperone αB-crystallin and reduced the aggregation of diverse proteins. We generated new QPCT inhibitors using in silico methods followed by in vitro screening, which rescued the HD-related phenotypes in cell, Drosophila and zebrafish HD models. Our data reveal a new HD druggable target affecting mutant HTT aggregation and provide proof of principle for a discovery pipeline from druggable genome screen to drug development.


Assuntos
Aminoaciltransferases/efeitos dos fármacos , Aminoaciltransferases/genética , Doença de Huntington/tratamento farmacológico , Doença de Huntington/genética , RNA Interferente Pequeno , Aminoaciltransferases/antagonistas & inibidores , Animais , Células Cultivadas , Biologia Computacional , Drosophila , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteína Huntingtina , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Peixe-Zebra , Cadeia B de alfa-Cristalina/metabolismo
14.
Nat Commun ; 5: 4998, 2014 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-25241929

RESUMO

Genome-wide association studies have identified several loci associated with Alzheimer's disease (AD), including proteins involved in endocytic trafficking such as PICALM/CALM (phosphatidylinositol binding clathrin assembly protein). It is unclear how these loci may contribute to AD pathology. Here we show that CALM modulates autophagy and alters clearance of tau, a protein which is a known autophagy substrate and which is causatively linked to AD, both in vitro and in vivo. Furthermore, altered CALM expression exacerbates tau-mediated toxicity in zebrafish transgenic models. CALM influences autophagy by regulating the endocytosis of SNAREs, such as VAMP2, VAMP3 and VAMP8, which have diverse effects on different stages of the autophagy pathway, from autophagosome formation to autophagosome degradation. This study suggests that the AD genetic risk factor CALM modulates autophagy, and this may affect disease in a number of ways including modulation of tau turnover.


Assuntos
Autofagia , Proteínas Monoméricas de Montagem de Clatrina/metabolismo , Proteínas tau/metabolismo , Animais , Proteína 12 Relacionada à Autofagia , Linhagem Celular , Drosophila , Endocitose , Feminino , Fibroblastos/metabolismo , Estudo de Associação Genômica Ampla , Células HEK293 , Células HeLa , Humanos , Masculino , Camundongos , Fagossomos , Ligação Proteica , RNA Interferente Pequeno/metabolismo , Fatores de Risco , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Transfecção , Proteína 2 Associada à Membrana da Vesícula/metabolismo , Peixe-Zebra
15.
Artigo em Inglês | MEDLINE | ID: mdl-24782716

RESUMO

Inhibition has a central role in defining the selectivity of the responses of higher order neurons to sensory stimuli. However, the circuit mechanisms of regulation of these responses by inhibitory neurons are still unclear. In Drosophila, the mushroom bodies (MBs) are necessary for olfactory memory, and by implication for the selectivity of learned responses to specific odors. To understand the circuitry of inhibition in the calyx (the input dendritic region) of the MBs, and its relationship with MB excitatory activity, we used the simple anatomy of the Drosophila larval olfactory system to identify any inhibitory inputs that could contribute to the selectivity of MB odor responses. We found that a single neuron accounts for all detectable GABA innervation in the calyx of the MBs, and that this neuron has pre-synaptic terminals in the calyx and post-synaptic branches in the MB lobes (output axonal area). We call this neuron the larval anterior paired lateral (APL) neuron, because of its similarity to the previously described adult APL neuron. Reconstitution of GFP partners (GRASP) suggests that the larval APL makes extensive contacts with the MB intrinsic neurons, Kenyon Cells (KCs), but few contacts with incoming projection neurons (PNs). Using calcium imaging of neuronal activity in live larvae, we show that the larval APL responds to odors, in a manner that requires output from KCs. Our data suggest that the larval APL is the sole GABAergic neuron that innervates the MB input region and carries inhibitory feedback from the MB output region, consistent with a role in modulating the olfactory selectivity of MB neurons.


Assuntos
Neurônios GABAérgicos/fisiologia , Larva/fisiologia , Corpos Pedunculados/fisiologia , Odorantes , Percepção Olfatória/fisiologia , Olfato/fisiologia , Animais , Discriminação Psicológica/fisiologia , Drosophila/fisiologia , Aprendizagem/fisiologia , Memória/fisiologia
16.
CEUR Workshop Proc ; 1265: 85-96, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29724079

RESUMO

A massive effort is underway to map the structure of the Drosophila nervous system and to genetically dissect its function. Virtual Fly Brain (VFB; http://www.virtualflybrain.org) is a popular, OWL-based resource providing neuroinformatics support for this work. It provides: curated descriptions of brain regions and neurons; queries for neurons based on their relationship to gross neuroanatomy; and queries for reagents based on their expression patterns. Query results are enriched by OWL axiomatisation allowing basic mereological reasoning. To keep reasoning fast and scalable, VFB confines expressiveness to the EL profile of OWL. As a result, VFB does not provide queries involving negation, despite there being both demand and sufficient information to support them. Recent developments in reasoning technology may make more expressive queries practical. Here we present design patterns to support queries with negation that are compatible with the mereological reasoning used in VFB.

17.
BMC Med Genomics ; 6: 52, 2013 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-24229347

RESUMO

BACKGROUND: Neurodegenerative diseases (NDs) are characterized by the progressive loss of neurons in the human brain. Although the majority of NDs are sporadic, evidence is accumulating that they have a strong genetic component. Therefore, significant efforts have been made in recent years to not only identify disease-causing genes but also genes that modify the severity of NDs, so-called genetic modifiers. To date there exists no compendium that lists and cross-links genetic modifiers of different NDs. DESCRIPTION: In order to address this need, we present NeuroGeM, the first comprehensive knowledgebase providing integrated information on genetic modifiers of nine different NDs in the model organisms D. melanogaster, C. elegans, and S. cerevisiae. NeuroGeM cross-links curated genetic modifier information from the different NDs and provides details on experimental conditions used for modifier identification, functional annotations, links to homologous proteins and color-coded protein-protein interaction networks to visualize modifier interactions. We demonstrate how this database can be used to generate new understanding through meta-analysis. For instance, we reveal that the Drosophila genes DnaJ-1, thread, Atx2, and mub are generic modifiers that affect multiple if not all NDs. CONCLUSION: As the first compendium of genetic modifiers, NeuroGeM will assist experimental and computational scientists in their search for the pathophysiological mechanisms underlying NDs. http://chibi.ubc.ca/neurogem.


Assuntos
Biologia Computacional/métodos , Bases de Dados Genéticas , Doenças Neurodegenerativas/genética , Animais , Mineração de Dados , Ontologia Genética , Humanos
18.
J Genet Genomics ; 40(6): 297-306, 2013 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-23790629

RESUMO

At least 25 genes, many involved in trafficking, localisation or shaping of membrane organelles, have been identified as causative genes for the neurodegenerative disorder hereditary spastic paraplegia (HSP). One of the most commonly mutated HSP genes, atlastin-1, encodes a dynamin-like GTPase that mediates homotypic fusion of endoplasmic reticulum (ER) membranes. However, the molecular mechanisms of atlastin-1-related membrane fusion and axonopathy remain unclear. To better understand its mode of action, we used affinity purification coupled with mass spectrometry to identify protein interactors of atlastin in Drosophila. Analysis of 72 identified proteins revealed that the atlastin interactome contains many proteins involved in protein processing and transport, in addition to proteins with roles in mRNA binding, metabolism and mitochondrial proteins. The highest confidence interactor from mass spectrometry analysis, the ubiquitin-selective AAA-ATPase valosin-containing protein (VCP), was validated as an atlastin-interacting protein, and VCP and atlastin showed overlapping subcellular distributions. Furthermore, VCP acted as a genetic modifier of atlastin: loss of VCP partially suppressed an eye phenotype caused by atlastin overexpression, whereas overexpression of VCP enhanced this phenotype. These interactions between atlastin and VCP suggest a functional relationship between these two proteins, and point to potential shared mechanisms between HSP and other forms of neurodegeneration.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Drosophila/enzimologia , GTP Fosfo-Hidrolases/metabolismo , Mapeamento de Interação de Proteínas , Paraplegia Espástica Hereditária/enzimologia , Adenosina Trifosfatases/genética , Animais , Modelos Animais de Doenças , Drosophila/genética , Drosophila/crescimento & desenvolvimento , Proteínas de Drosophila/genética , Retículo Endoplasmático/enzimologia , Retículo Endoplasmático/genética , Olho/enzimologia , Olho/crescimento & desenvolvimento , Feminino , GTP Fosfo-Hidrolases/genética , Humanos , Masculino , Ligação Proteica , Paraplegia Espástica Hereditária/genética , Proteína com Valosina
19.
Science ; 336(6083): 925-31, 2012 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-22605779

RESUMO

Color and motion information are thought to be channeled through separate neural pathways, but it remains unclear whether and how these pathways interact to improve motion perception. In insects, such as Drosophila, it has long been believed that motion information is fed exclusively by one spectral class of photoreceptor, so-called R1 to R6 cells; whereas R7 and R8 photoreceptors, which exist in multiple spectral classes, subserve color vision. Here, we report that R7 and R8 also contribute to the motion pathway. By using electrophysiological, optical, and behavioral assays, we found that R7/R8 information converge with and shape the motion pathway output, explaining flies' broadly tuned optomotor behavior by its composite responses. Our results demonstrate that inputs from photoreceptors of different spectral sensitivities improve motion discrimination, increasing robustness of perception.


Assuntos
Drosophila melanogaster/fisiologia , Percepção de Movimento , Células Fotorreceptoras de Invertebrados/fisiologia , Animais , Visão de Cores , Proteínas de Drosophila , Drosophila melanogaster/genética , Voo Animal , Junções Comunicantes/fisiologia , Genes de Insetos , Luz , Modelos Neurológicos , Mutação , Neurônios/fisiologia , Opsinas/metabolismo , Lobo Óptico de Animais não Mamíferos/citologia , Lobo Óptico de Animais não Mamíferos/fisiologia , Técnicas de Patch-Clamp , Células Fotorreceptoras de Invertebrados/ultraestrutura , Transgenes , Raios Ultravioleta , Vias Visuais
20.
Hum Mol Genet ; 21(15): 3356-65, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22543973

RESUMO

Several causative genes for hereditary spastic paraplegia encode proteins with intramembrane hairpin loops that contribute to the curvature of the endoplasmic reticulum (ER), but the relevance of this function to axonal degeneration is not understood. One of these genes is reticulon2. In contrast to mammals, Drosophila has only one widely expressed reticulon orthologue, Rtnl1, and we therefore used Drosophila to test its importance for ER organization and axonal function. Rtnl1 distribution overlapped with that of the ER, but in contrast to the rough ER, was enriched in axons. The loss of Rtnl1 led to the expansion of the rough or sheet ER in larval epidermis and elevated levels of ER stress. It also caused abnormalities specifically within distal portions of longer motor axons and in their presynaptic terminals, including disruption of the smooth ER (SER), the microtubule cytoskeleton and mitochondria. In contrast, proximal axon portions appeared unaffected. Our results provide direct evidence for reticulon function in the organization of the SER in distal longer axons, and support a model in which spastic paraplegia can be caused by impairment of axonal the SER. Our data provide a route to further understanding of both the role of the SER in axons and the pathological consequences of the impairment of this compartment.


Assuntos
Proteínas de Drosophila/genética , Drosophila/metabolismo , Retículo Endoplasmático Liso/metabolismo , Paraplegia Espástica Hereditária/genética , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Animais , Animais Geneticamente Modificados , Axônios/metabolismo , Modelos Animais de Doenças , Proteínas de Drosophila/metabolismo , Paraplegia Espástica Hereditária/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...