Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Microsc ; 284(1): 56-73, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34214188

RESUMEN

A modern day light microscope has evolved from a tool devoted to making primarily empirical observations to what is now a sophisticated , quantitative device that is an integral part of both physical and life science research. Nowadays, microscopes are found in nearly every experimental laboratory. However, despite their prevalent use in capturing and quantifying scientific phenomena, neither a thorough understanding of the principles underlying quantitative imaging techniques nor appropriate knowledge of how to calibrate, operate and maintain microscopes can be taken for granted. This is clearly demonstrated by the well-documented and widespread difficulties that are routinely encountered in evaluating acquired data and reproducing scientific experiments. Indeed, studies have shown that more than 70% of researchers have tried and failed to repeat another scientist's experiments, while more than half have even failed to reproduce their own experiments. One factor behind the reproducibility crisis of experiments published in scientific journals is the frequent underreporting of imaging methods caused by a lack of awareness and/or a lack of knowledge of the applied technique. Whereas quality control procedures for some methods used in biomedical research, such as genomics (e.g. DNA sequencing, RNA-seq) or cytometry, have been introduced (e.g. ENCODE), this issue has not been tackled for optical microscopy instrumentation and images. Although many calibration standards and protocols have been published, there is a lack of awareness and agreement on common standards and guidelines for quality assessment and reproducibility. In April 2020, the QUality Assessment and REProducibility for instruments and images in Light Microscopy (QUAREP-LiMi) initiative was formed. This initiative comprises imaging scientists from academia and industry who share a common interest in achieving a better understanding of the performance and limitations of microscopes and improved quality control (QC) in light microscopy. The ultimate goal of the QUAREP-LiMi initiative is to establish a set of common QC standards, guidelines, metadata models and tools, including detailed protocols, with the ultimate aim of improving reproducible advances in scientific research. This White Paper (1) summarizes the major obstacles identified in the field that motivated the launch of the QUAREP-LiMi initiative; (2) identifies the urgent need to address these obstacles in a grassroots manner, through a community of stakeholders including, researchers, imaging scientists, bioimage analysts, bioimage informatics developers, corporate partners, funding agencies, standards organizations, scientific publishers and observers of such; (3) outlines the current actions of the QUAREP-LiMi initiative and (4) proposes future steps that can be taken to improve the dissemination and acceptance of the proposed guidelines to manage QC. To summarize, the principal goal of the QUAREP-LiMi initiative is to improve the overall quality and reproducibility of light microscope image data by introducing broadly accepted standard practices and accurately captured image data metrics.


Asunto(s)
Microscopía , Estándares de Referencia , Reproducibilidad de los Resultados
3.
Cytometry A ; 97(9): 882-886, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32583531

RESUMEN

Operating shared resource laboratories (SRLs) in times of pandemic is a challenge for research institutions. In a multiuser, high-turnover working space, the transmission of infectious agents is difficult to control. To address this challenge, imaging core facility managers being members of German BioImaging discussed how shared microscopes could be operated with minimal risk of spreading SARS-CoV-2 between users and staff. Here, we describe the resulting guidelines and explain their rationale, with a focus on separating users in space and time, protective face masks, and keeping surfaces virus-free. These recommendations may prove useful for other types of SRLs. © 2020 The Authors. Cytometry Part A published by Wiley Periodicals LLC. on behalf of International Society for Advancement of Cytometry.


Asunto(s)
Betacoronavirus/patogenicidad , Investigación Biomédica/organización & administración , Infecciones por Coronavirus/prevención & control , Control de Infecciones , Laboratorios/organización & administración , Microscopía , Salud Laboral , Pandemias/prevención & control , Neumonía Viral/prevención & control , COVID-19 , Conducta Cooperativa , Infecciones por Coronavirus/transmisión , Infecciones por Coronavirus/virología , Descontaminación , Contaminación de Equipos/prevención & control , Alemania , Humanos , Exposición Profesional/prevención & control , Equipo de Protección Personal , Neumonía Viral/transmisión , Neumonía Viral/virología , Factores Protectores , Investigadores/organización & administración , Medición de Riesgo , Factores de Riesgo , SARS-CoV-2 , Flujo de Trabajo
5.
Hum Mol Genet ; 24(13): 3623-37, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25794683

RESUMEN

The gene mapt codes for the microtubule-associated protein Tau. The R406W amino acid substitution in Tau is associated with frontotemporal dementia with parkinsonism linked to chromosome 17 (FTDP-17) characterized by Tau-positive filamentous inclusions. These filamentous Tau inclusions are present in a group of neurodegenerative diseases known as tauopathies, including Alzheimer's disease (AD). To gain more insights into the pathomechanism of tauopathies, we performed an RNAi-based large-scale screen in Drosophila melanogaster to identify genetic modifiers of Tau[R406W]-induced toxicity. A collection of RNAi lines, putatively silencing more than 7000 genes, was screened for the ability to modify Tau[R406W]-induced toxicity in vivo. This collection covered more than 50% of all protein coding fly genes and more than 90% of all fly genes known to have a human ortholog. Hereby, we identified 62 genes that, when silenced by RNAi, modified Tau-induced toxicity specifically. Among these 62 modifiers were three subunits of the Dynein/Dynactin complex. Analysis on segmental nerves of fly larvae showed that pan neural Tau[R406W] expression and concomitant silencing of Dynein/Dynactin complex members synergistically caused strong pathological changes within the axonal compartment, but only minor changes at synapses. At the larval stage, these alterations did not cause locomotion deficits, but became evident in adult flies. Our data suggest that Tau-induced detrimental effects most likely originate from axonal rather than synaptic dysfunction and that impaired retrograde transport intensifies detrimental effects of Tau in axons. In conclusion, our findings contribute to the elucidation of disease mechanisms in tauopathies like FTDP-17 or AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Proteínas de Drosophila/toxicidad , Drosophila melanogaster/metabolismo , Dineínas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas tau/toxicidad , Enfermedad de Alzheimer/genética , Animales , Axones/metabolismo , Modelos Animales de Enfermedad , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Complejo Dinactina , Dineínas/genética , Femenino , Humanos , Masculino , Proteínas Asociadas a Microtúbulos/genética , Mutación Missense , Transporte de Proteínas , Interferencia de ARN , Proteínas tau/genética , Proteínas tau/metabolismo
6.
J Neurochem ; 137(1): 12-25, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26756400

RESUMEN

Abnormal tau accumulations were observed and documented in post-mortem brains of patients affected by Alzheimer's disease (AD) long before the identification of mutations in the Microtubule-associated protein tau (MAPT) gene, encoding the tau protein, in a different neurodegenerative disease called Frontotemporal dementia and Parkinsonism linked to chromosome 17 (FTDP-17). The discovery of mutations in the MAPT gene associated with FTDP-17 highlighted that dysfunctions in tau alone are sufficient to cause neurodegeneration. Invertebrate models have been diligently utilized in investigating tauopathies, contributing to the understanding of cellular and molecular pathways involved in disease etiology. An important discovery came with the demonstration that over-expression of human tau in Drosophila leads to premature mortality and neuronal dysfunction including neurodegeneration, recapitulating some key neuropathological features of the human disease. The simplicity of handling invertebrate models combined with the availability of a diverse range of experimental resources make these models, in particular Drosophila a powerful invertebrate screening tool. Consequently, several large-scale screens have been performed using Drosophila, to identify modifiers of tau toxicity. The screens have revealed not only common cellular and molecular pathways, but in some instances the same modifier has been independently identified in two or more screens suggesting a possible role for these modifiers in regulating tau toxicity. The purpose of this review is to discuss the genetic modifier screens on tauopathies performed in Drosophila and C. elegans models, and to highlight the common cellular and molecular pathways that have emerged from these studies. Here, we summarize results of tau toxicity screens providing mechanistic insights into pathological alterations in tauopathies. Key pathways or modifiers that have been identified are associated with a broad range of processes including, but not limited to, phosphorylation, cytoskeleton organization, axonal transport, regulation of cellular proteostasis, transcription, RNA metabolism, cell cycle regulation, and apoptosis. We discuss the utility and application of invertebrate models in elucidating the cellular and molecular functions of novel and uncharacterized disease modifiers identified in large-scale screens as well as for investigating the function of genes identified as risk factors in genome-wide association studies from human patients in the post-genomic era. In this review, we combined and summarized several large-scale modifier screens performed in invertebrate models to identify modifiers of tau toxicity. A summary of the screens show that diverse cellular processes are implicated in the modification of tau toxicity. Kinases and phosphatases are the most predominant class of modifiers followed by components required for cellular proteostasis and axonal transport and cytoskeleton elements.


Asunto(s)
Invertebrados/metabolismo , Tauopatías/metabolismo , Animales , Animales Modificados Genéticamente , Apoptosis , Transporte Axonal , Caenorhabditis elegans/metabolismo , Ciclo Celular , Citoesqueleto/metabolismo , Citoesqueleto/ultraestructura , Modelos Animales de Enfermedad , Drosophila melanogaster/metabolismo , Regulación de la Expresión Génica , Humanos , Longevidad/genética , Redes y Vías Metabólicas , Ratones , Ratones Noqueados , Mutación , Degeneración Nerviosa/genética , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/toxicidad , Pez Cebra , Proteínas tau/genética , Proteínas tau/metabolismo , Proteínas tau/toxicidad
7.
J Biol Chem ; 289(27): 19164-79, 2014 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-24825905

RESUMEN

Trans-activation element DNA-binding protein of 43 kDa (TDP-43) characterizes insoluble protein aggregates in distinct subtypes of frontotemporal lobar degeneration and amyotrophic lateral sclerosis. TDP-43 mediates many RNA processing steps within distinct protein complexes. Here we identify novel TDP-43 protein interactors found in a yeast two-hybrid screen using an adult human brain cDNA library. We confirmed the TDP-43 interaction of seven hits by co-immunoprecipitation and assessed their co-localization in HEK293E cells. As pathological TDP-43 is ubiquitinated, we focused on the ubiquitin-conjugating enzyme UBE2E3 and the ubiquitin isopeptidase Y (UBPY). When cells were treated with proteasome inhibitor, ubiquitinated and insoluble TDP-43 species accumulated. All three UBE2E family members could enhance the ubiquitination of TDP-43, whereas catalytically inactive UBE2E3(C145S) was much less efficient. Conversely, silencing of UBE2E3 reduced TDP-43 ubiquitination. We examined 15 of the 48 known disease-associated TDP-43 mutants and found that one was excessively ubiquitinated. This strong TDP-43(K263E) ubiquitination was further enhanced by proteasomal inhibition as well as UBE2E3 expression. Conversely, UBE2E3 silencing and expression of UBPY reduced TDP-43(K263E) ubiquitination. Moreover, wild-type but not active site mutant UBPY reduced ubiquitination of TDP-43 C-terminal fragments and of a nuclear import-impaired mutant. In Drosophila melanogaster, UBPY silencing enhanced neurodegenerative TDP-43 phenotypes and the accumulation of insoluble high molecular weight TDP-43 and ubiquitin species. Thus, UBE2E3 and UBPY participate in the regulation of TDP-43 ubiquitination, solubility, and neurodegeneration.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Endopeptidasas/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitinación , Adulto , Animales , Encéfalo/metabolismo , Drosophila melanogaster/metabolismo , Endopeptidasas/deficiencia , Complejos de Clasificación Endosomal Requeridos para el Transporte/deficiencia , Células HEK293 , Humanos , Neurotoxinas/metabolismo , Transporte de Proteínas , Técnicas del Sistema de Dos Híbridos , Ubiquitina Tiolesterasa/deficiencia
8.
Neurobiol Dis ; 74: 76-88, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25447237

RESUMEN

Inclusions containing Fused in Sarcoma (FUS) are found in familial and sporadic cases of the incurable progressive motor neuron disease amyotrophic lateral sclerosis and in a common form of dementia, frontotemporal dementia. Most disease-associated mutations are located in the C-terminal proline-tyrosine nuclear localization sequence (PY-NLS) of FUS and impair its nuclear import. It has been shown in cell culture that the nuclear import of FUS is mediated by transportin, which binds the PY-NLS and the last arginine/glycine/glycine-rich (RGG) domain of FUS. Methylation of this last RGG domain by protein arginine methyltransferases (PRMTs) weakens transportin binding and therefore impairs nuclear translocation of FUS. To investigate the requirements for the nuclear import of FUS in an in vivo model, we generated different transgenic Drosophila lines expressing human FUS wild type (hFUS wt) and two disease-related variants P525L and R495X, in which the NLS is mutated or completely absent, respectively. To rule out effects caused by heterologous hFUS expression, we analysed the corresponding variants for the Drosophila FUS orthologue Cabeza (Caz wt, P398L, Q349X). Expression of these variants in eyes and motor neurons confirmed the PY-NLS-dependent nuclear localization of FUS/Caz and caused neurodegenerative effects. Surprisingly, FUS/Caz toxicity was correlated to the degree of its nuclear localization in this overexpression model. High levels of nuclear FUS/Caz became insoluble and reduced the endogenous Caz levels, confirming FUS autoregulation in Drosophila. RNAi-mediated knockdown of the two transportin orthologues interfered with the nuclear import of FUS/Caz and also enhanced the eye phenotype. Finally, we screened the Drosophila PRMT proteins (DART1-9) and found that knockdown of Dart1 led to a reduction in methylation of hFUS P525L and aggravated its phenotype. These findings show that the molecular mechanisms controlling the nuclear import of FUS/Caz and FUS autoregulation are conserved between humans and Drosophila. In addition to the well-known neurodegenerative effects of FUS loss-of function, our data suggest toxic potential of overexpressed FUS in the nucleus and of insoluble FUS.


Asunto(s)
Transporte Activo de Núcleo Celular/fisiología , Proteínas de Drosophila/metabolismo , Metiltransferasas/metabolismo , Proteína FUS de Unión a ARN/metabolismo , Proteínas de Unión al ARN/metabolismo , Factor de Transcripción TFIID/metabolismo , Animales , Animales Modificados Genéticamente , Metilación de ADN/fisiología , Modelos Animales de Enfermedad , Proteínas de Drosophila/genética , Drosophila melanogaster , Ojo/metabolismo , Ojo/patología , Técnicas de Silenciamiento del Gen , Humanos , Carioferinas/metabolismo , Metiltransferasas/genética , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Trastornos del Movimiento/patología , Trastornos del Movimiento/fisiopatología , Mutación , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/fisiopatología , Interferencia de ARN , Proteína FUS de Unión a ARN/genética , Proteínas de Unión al ARN/genética , Factor de Transcripción TFIID/genética
9.
PLoS Genet ; 8(11): e1003066, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23209432

RESUMEN

Hereditary spastic paraplegias (HSPs) comprise a group of genetically heterogeneous neurodegenerative disorders characterized by spastic weakness of the lower extremities. We have generated a Drosophila model for HSP type 10 (SPG10), caused by mutations in KIF5A. KIF5A encodes the heavy chain of kinesin-1, a neuronal microtubule motor. Our results imply that SPG10 is not caused by haploinsufficiency but by the loss of endogenous kinesin-1 function due to a selective dominant-negative action of mutant KIF5A on kinesin-1 complexes. We have not found any evidence for an additional, more generalized toxicity of mutant Kinesin heavy chain (Khc) or the affected kinesin-1 complexes. Ectopic expression of Drosophila Khc carrying a human SPG10-associated mutation (N256S) is sufficient to disturb axonal transport and to induce motoneuron disease in Drosophila. Neurofilaments, which have been recently implicated in SPG10 disease manifestation, are absent in arthropods. Impairments in the transport of kinesin-1 cargos different from neurofilaments are thus sufficient to cause HSP-like pathological changes such as axonal swellings, altered structure and function of synapses, behavioral deficits, and increased mortality.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Cinesinas/genética , Paraplejía Espástica Hereditaria/genética , Animales , Transporte Axonal/genética , Transporte Axonal/fisiología , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Ratones , Microtúbulos/genética , Microtúbulos/metabolismo , Mutación , Sinapsis/genética , Sinapsis/patología
10.
EMBO J ; 29(1): 209-21, 2010 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-19910924

RESUMEN

TDP-43 is an RNA/DNA-binding protein implicated in transcriptional repression and mRNA processing. Inclusions of TDP-43 are hallmarks of frontotemporal dementia and amyotrophic lateral sclerosis. Besides aggregation of TDP-43, loss of nuclear localization is observed in disease. To identify relevant targets of TDP-43, we performed expression profiling. Thereby, histone deacetylase 6 (HDAC6) downregulation was discovered on TDP-43 silencing and confirmed at the mRNA and protein level in human embryonic kidney HEK293E and neuronal SH-SY5Y cells. This was accompanied by accumulation of the major HDAC6 substrate, acetyl-tubulin. HDAC6 levels were restored by re-expression of TDP-43, dependent on RNA binding and the C-terminal protein interaction domains. Moreover, TDP-43 bound specifically to HDAC6 mRNA arguing for a direct functional interaction. Importantly, in vivo validation in TDP-43 knockout Drosophila melanogaster confirmed the specific downregulation of HDAC6. HDAC6 is necessary for protein aggregate formation and degradation. Indeed, HDAC6-dependent reduction of cellular aggregate formation and increased cytotoxicity of polyQ-expanded ataxin-3 were found in TDP-43 silenced cells. In conclusion, loss of functional TDP-43 causes HDAC6 downregulation and might thereby contribute to pathogenesis.


Asunto(s)
Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Animales , Secuencia de Bases , Línea Celular , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/genética , Regulación hacia Abajo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Histona Desacetilasa 6 , Humanos , Neuronas/metabolismo , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteinopatías TDP-43/genética , Proteinopatías TDP-43/metabolismo
11.
J Bacteriol ; 194(19): 5218-27, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22821973

RESUMEN

Filamentous cyanobacteria of the order Nostocales display typical properties of multicellular organisms. In response to nitrogen starvation, some vegetative cells differentiate into heterocysts, where fixation of N(2) takes place. Heterocysts provide a micro-oxic compartment to protect nitrogenase from the oxygen produced by the vegetative cells. Differentiation involves fundamental remodeling of the gram-negative cell wall by deposition of a thick envelope and by formation of a neck-like structure at the contact site to the vegetative cells. Cell wall-hydrolyzing enzymes, like cell wall amidases, are involved in peptidoglycan maturation and turnover in unicellular bacteria. Recently, we showed that mutation of the amidase homologue amiC2 gene in Nostoc punctiforme ATCC 29133 distorts filament morphology and function. Here, we present the functional characterization of two amiC paralogues from Anabaena sp. strain PCC 7120. The amiC1 (alr0092) mutant was not able to differentiate heterocysts or to grow diazotrophically, whereas the amiC2 (alr0093) mutant did not show an altered phenotype under standard growth conditions. In agreement, fluorescence recovery after photobleaching (FRAP) studies showed a lack of cell-cell communication only in the AmiC1 mutant. Green fluorescent protein (GFP)-tagged AmiC1 was able to complement the mutant phenotype to wild-type properties. The protein localized in the septal regions of newly dividing cells and at the neck region of differentiating heterocysts. Upon nitrogen step-down, no mature heterocysts were developed in spite of ongoing heterocyst-specific gene expression. These results show the dependence of heterocyst development on amidase function and highlight a pivotal but so far underestimated cellular process, the remodeling of peptidoglycan, for the biology of filamentous cyanobacteria.


Asunto(s)
Amidohidrolasas/metabolismo , Anabaena/enzimología , Anabaena/metabolismo , Pared Celular/enzimología , Interacciones Microbianas/fisiología , Amidohidrolasas/genética , Anabaena/citología , Animales , Citoesqueleto/fisiología , ADN Bacteriano/genética , Regulación Bacteriana de la Expresión Génica , Regulación Enzimológica de la Expresión Génica
12.
Mol Microbiol ; 79(6): 1655-69, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21244533

RESUMEN

Filamentous cyanobacteria of the order Nostocales are primordial multicellular organisms, a property widely considered unique to eukaryotes. Their filaments are composed of hundreds of mutually dependent vegetative cells and regularly spaced N(2)-fixing heterocysts, exchanging metabolites and signalling molecules. Furthermore, they may differentiate specialized spore-like cells and motile filaments. However, the structural basis for cellular communication within the filament remained elusive. Here we present that mutation of a single gene, encoding cell wall amidase AmiC2, completely changes the morphology and abrogates cell differentiation and intercellular communication. Ultrastructural analysis revealed for the first time a contiguous peptidoglycan sacculus with individual cells connected by a single-layered septal cross-wall. The mutant forms irregular clusters of twisted cells connected by aberrant septa. Rapid intercellular molecule exchange takes place in wild-type filaments, but is completely abolished in the mutant, and this blockage obstructs any cell differentiation, indicating a fundamental importance of intercellular communication for cell differentiation in Nostoc. AmiC2-GFP localizes in the cell wall with a focus in the cross walls of dividing cells, implying that AmiC2 processes the newly synthesized septum into a functional cell-cell communication structure during cell division. AmiC2 thus can be considered as a novel morphogene required for cell-cell communication, cellular development and multicellularity.


Asunto(s)
Amidohidrolasas/metabolismo , Proteínas Bacterianas/metabolismo , Nostoc/enzimología , Nostoc/crecimiento & desarrollo , Amidohidrolasas/genética , Proteínas Bacterianas/genética , Pared Celular/enzimología , Pared Celular/genética , Regulación Bacteriana de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Nostoc/genética , Transporte de Proteínas
13.
J Cell Biol ; 177(5): 843-55, 2007 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-17548512

RESUMEN

Synapses can undergo rapid changes in size as well as in their vesicle release function during both plasticity processes and development. This fundamental property of neuronal cells requires the coordinated rearrangement of synaptic membranes and their associated cytoskeleton, yet remarkably little is known of how this coupling is achieved. In a GFP exon-trap screen, we identified Drosophila melanogaster Basigin (Bsg) as an immunoglobulin domain-containing transmembrane protein accumulating at periactive zones of neuromuscular junctions. Bsg is required pre- and postsynaptically to restrict synaptic bouton size, its juxtamembrane cytoplasmic residues being important for that function. Bsg controls different aspects of synaptic structure, including distribution of synaptic vesicles and organization of the presynaptic cortical actin cytoskeleton. Strikingly, bsg function is also required specifically within the presynaptic terminal to inhibit nonsynchronized evoked vesicle release. We thus propose that Bsg is part of a transsynaptic complex regulating synaptic compartmentalization and strength, and coordinating plasma membrane and cortical organization.


Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila melanogaster/metabolismo , Glicoproteínas de Membrana/fisiología , Sinapsis/fisiología , Vesículas Sinápticas/fisiología , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestructura , Secuencia de Aminoácidos , Animales , Adhesión Celular , Compartimento Celular , Regulación hacia Abajo , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Datos de Secuencia Molecular , Mutación , Unión Neuromuscular/metabolismo , Estructura Terciaria de Proteína , Alineación de Secuencia , Sinapsis/metabolismo
14.
Nat Neurosci ; 11(6): 659-66, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18469810

RESUMEN

The subunit composition of postsynaptic non-NMDA-type glutamate receptors (GluRs) determines the function and trafficking of the receptor. Changes in GluR composition have been implicated in the homeostasis of neuronal excitability and synaptic plasticity underlying learning. Here, we imaged GluRs in vivo during the formation of new postsynaptic densities (PSDs) at Drosophila neuromuscular junctions coexpressing GluRIIA and GluRIIB subunits. GluR composition was independently regulated at directly neighboring PSDs on a submicron scale. Immature PSDs typically had large amounts of GluRIIA and small amounts of GluRIIB. During subsequent PSD maturation, however, the GluRIIA/GluRIIB composition changed and became more balanced. Reducing presynaptic glutamate release increased GluRIIA, but decreased GluRIIB incorporation. Moreover, the maturation of GluR composition correlated in a site-specific manner with the level of Bruchpilot, an active zone protein that is essential for mature glutamate release. Thus, we show that an activity-dependent, site-specific control of GluR composition can contribute to match pre- and postsynaptic assembly.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Unión Neuromuscular/metabolismo , Receptores AMPA/fisiología , Animales , Animales Modificados Genéticamente , Simulación por Computador , Drosophila , Proteínas de Drosophila , Potenciales Postsinápticos Excitadores/fisiología , Potenciales Postsinápticos Excitadores/efectos de la radiación , Recuperación de Fluorescencia tras Fotoblanqueo/métodos , Ácido Glutámico/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Modelos Biológicos , Mutación/fisiología , Técnicas de Placa-Clamp , Transporte de Proteínas/fisiología , Receptores AMPA/genética , Factores de Tiempo
15.
Autophagy ; 17(10): 3160-3174, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33404278

RESUMEN

We investigated in larval and adult Drosophila models whether loss of the mitochondrial chaperone Hsc70-5 is sufficient to cause pathological alterations commonly observed in Parkinson disease. At affected larval neuromuscular junctions, no effects on terminal size, bouton size or number, synapse size, or number were observed, suggesting that we studied an early stage of pathogenesis. At this stage, we noted a loss of synaptic vesicle proteins and active zone components, delayed synapse maturation, reduced evoked and spontaneous excitatory junctional potentials, increased synaptic fatigue, and cytoskeleton rearrangements. The adult model displayed ATP depletion, altered body posture, and susceptibility to heat-induced paralysis. Adult phenotypes could be suppressed by knockdown of dj-1ß, Lrrk, DCTN2-p50, DCTN1-p150, Atg1, Atg101, Atg5, Atg7, and Atg12. The knockdown of components of the macroautophagy/autophagy machinery or overexpression of human HSPA9 broadly rescued larval and adult phenotypes, while disease-associated HSPA9 variants did not. Overexpression of Pink1 or promotion of autophagy exacerbated defects.Abbreviations: AEL: after egg laying; AZ: active zone; brp: bruchpilot; Csp: cysteine string protein; dlg: discs large; eEJPs: evoked excitatory junctional potentials; GluR: glutamate receptor; H2O2: hydrogen peroxide; mEJP: miniature excitatory junctional potentials; MT: microtubule; NMJ: neuromuscular junction; PD: Parkinson disease; Pink1: PTEN-induced putative kinase 1; PSD: postsynaptic density; SSR: subsynaptic reticulum; SV: synaptic vesicle; VGlut: vesicular glutamate transporter.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Autofagia/genética , Proteína 7 Relacionada con la Autofagia/metabolismo , Proteínas Relacionadas con la Autofagia/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Peróxido de Hidrógeno , Proteínas del Tejido Nervioso/metabolismo , Unión Neuromuscular/metabolismo , Proteína Desglicasa DJ-1/metabolismo , Proteínas Serina-Treonina Quinasas
16.
Neuron ; 49(6): 833-44, 2006 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-16543132

RESUMEN

Neurotransmitters are released at presynaptic active zones (AZs). In the fly Drosophila, monoclonal antibody (MAB) nc82 specifically labels AZs. We employ nc82 to identify Bruchpilot protein (BRP) as a previously unknown AZ component. BRP shows homology to human AZ protein ELKS/CAST/ERC, which binds RIM1 in a complex with Bassoon and Munc13-1. The C terminus of BRP displays structural similarities to multifunctional cytoskeletal proteins. During development, transcription of the bruchpilot locus (brp) coincides with neuronal differentiation. Panneural reduction of BRP expression by RNAi constructs permits a first functional characterization of this large AZ protein: larvae show reduced evoked but normal spontaneous transmission at neuromuscular junctions. In adults, we observe loss of T bars at active zones, absence of synaptic components in electroretinogram, locomotor inactivity, and unstable flight (hence "bruchpilot"-crash pilot). We propose that BRP is critical for intact AZ structure and normal-evoked neurotransmitter release at chemical synapses of Drosophila.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Drosophila/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Unión Neuromuscular/fisiología , Homología Estructural de Proteína , Proteínas Adaptadoras Transductoras de Señales/química , Animales , Animales Modificados Genéticamente , Conducta Animal , Northern Blotting/métodos , Western Blotting/métodos , Clonación Molecular , Drosophila , Proteínas de Drosophila/genética , Dinaminas/metabolismo , Electroforesis en Gel Bidimensional/métodos , Proteínas Fluorescentes Verdes/biosíntesis , Humanos , Inmunoquímica/métodos , Hibridación in Situ/métodos , Péptidos y Proteínas de Señalización Intracelular/química , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Potenciales de la Membrana/fisiología , Microscopía Electrónica de Transmisión/métodos , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/química , Unión Neuromuscular/ultraestructura , Terminales Presinápticos/metabolismo , ARN Polimerasa I , ARN Mensajero/biosíntesis , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Caminata/fisiología
17.
J Neurosci ; 29(37): 11484-94, 2009 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-19759297

RESUMEN

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


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Glucógeno Sintasa Quinasa 3/metabolismo , Unión Neuromuscular/fisiología , Proteína Fosfatasa 2/metabolismo , Animales , Animales Modificados Genéticamente , Citoesqueleto/metabolismo , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Tomografía con Microscopio Electrónico/métodos , Regulación del Desarrollo de la Expresión Génica/genética , Glucógeno Sintasa Quinasa 3 beta , Microscopía Confocal , Mutación/genética , Unión Neuromuscular/citología , Unión Neuromuscular/ultraestructura , Terminales Presinápticos/metabolismo , Terminales Presinápticos/ultraestructura , Transporte de Proteínas/genética , Receptores de Glutamato/metabolismo , Transmisión Sináptica/genética , Transmisión Sináptica/fisiología
18.
Front Bioeng Biotechnol ; 8: 558880, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33117778

RESUMEN

Various pre-trained deep learning models for the segmentation of bioimages have been made available as developer-to-end-user solutions. They are optimized for ease of use and usually require neither knowledge of machine learning nor coding skills. However, individually testing these tools is tedious and success is uncertain. Here, we present the Open Segmentation Framework (OpSeF), a Python framework for deep learning-based instance segmentation. OpSeF aims at facilitating the collaboration of biomedical users with experienced image analysts. It builds on the analysts' knowledge in Python, machine learning, and workflow design to solve complex analysis tasks at any scale in a reproducible, well-documented way. OpSeF defines standard inputs and outputs, thereby facilitating modular workflow design and interoperability with other software. Users play an important role in problem definition, quality control, and manual refinement of results. OpSeF semi-automates preprocessing, convolutional neural network (CNN)-based segmentation in 2D or 3D, and postprocessing. It facilitates benchmarking of multiple models in parallel. OpSeF streamlines the optimization of parameters for pre- and postprocessing such, that an available model may frequently be used without retraining. Even if sufficiently good results are not achievable with this approach, intermediate results can inform the analysts in the selection of the most promising CNN-architecture in which the biomedical user might invest the effort of manually labeling training data. We provide Jupyter notebooks that document sample workflows based on various image collections. Analysts may find these notebooks useful to illustrate common segmentation challenges, as they prepare the advanced user for gradually taking over some of their tasks and completing their projects independently. The notebooks may also be used to explore the analysis options available within OpSeF in an interactive way and to document and share final workflows. Currently, three mechanistically distinct CNN-based segmentation methods, the U-Net implementation used in Cellprofiler 3.0, StarDist, and Cellpose have been integrated within OpSeF. The addition of new networks requires little; the addition of new models requires no coding skills. Thus, OpSeF might soon become both an interactive model repository, in which pre-trained models might be shared, evaluated, and reused with ease.

19.
Nat Neurosci ; 8(7): 898-905, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16136672

RESUMEN

Insight into how glutamatergic synapses form in vivo is important for understanding developmental and experience-triggered changes of excitatory circuits. Here, we imaged postsynaptic densities (PSDs) expressing a functional, GFP-tagged glutamate receptor subunit (GluR-IIA(GFP)) at neuromuscular junctions of Drosophila melanogaster larvae for several days in vivo. New PSDs, associated with functional and structural presynaptic markers, formed independently of existing synapses and grew continuously until reaching a stable size within hours. Both in vivo photoactivation and photobleaching experiments showed that extrasynaptic receptors derived from diffuse, cell-wide pools preferentially entered growing PSDs. After entering PSDs, receptors were largely immobilized. In comparison, other postsynaptic proteins tested (PSD-95, NCAM and PAK homologs) exchanged faster and with no apparent preference for growing synapses. We show here that new glutamatergic synapses form de novo and not by partitioning processes from existing synapses, suggesting that the site-specific entry of particular glutamate receptor complexes directly controls the assembly of individual PSDs.


Asunto(s)
Receptores AMPA/fisiología , Sinapsis/fisiología , Animales , Drosophila melanogaster , Proteínas Fluorescentes Verdes , Larva , Sustancias Luminiscentes , Proteínas del Tejido Nervioso/metabolismo , Unión Neuromuscular/metabolismo , Receptores AMPA/metabolismo
20.
Sci Rep ; 7: 38172, 2017 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-28344334

RESUMEN

The kinesin-3 family member KIF1A has been shown to be important for experience dependent neuroplasticity. In Drosophila, amorphic mutations in the KIF1A homolog unc-104 disrupt the formation of mature boutons. Disease associated KIF1A mutations have been associated with motor and sensory dysfunctions as well as non-syndromic intellectual disability in humans. A hypomorphic mutation in the forkhead-associated domain of Unc-104, unc-104bris, impairs active zone maturation resulting in an increased fraction of post-synaptic glutamate receptor fields that lack the active zone scaffolding protein Bruchpilot. Here, we show that the unc-104brismutation causes defects in synaptic transmission as manifested by reduced amplitude of both evoked and miniature excitatory junctional potentials. Structural defects observed in the postsynaptic compartment of mutant NMJs include reduced glutamate receptor field size, and altered glutamate receptor composition. In addition, we observed marked loss of postsynaptic scaffolding proteins and reduced complexity of the sub-synaptic reticulum, which could be rescued by pre- but not postsynaptic expression of unc-104. Our results highlight the importance of kinesin-3 based axonal transport in synaptic transmission and provide novel insights into the role of Unc-104 in synapse maturation.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Cinesinas/metabolismo , Densidad Postsináptica , Animales , Drosophila/ultraestructura , Proteínas de Drosophila/genética , Cinesinas/genética , Larva , Mutación , Unión Neuromuscular/metabolismo , Transmisión Sináptica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA