Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Genome Res ; 31(7): 1159-1173, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34088716

RESUMEN

Regulatory interactions mediated by transcription factors (TFs) make up complex networks that control cellular behavior. Fully understanding these gene regulatory networks (GRNs) offers greater insight into the consequences of disease-causing perturbations than can be achieved by studying single TF binding events in isolation. Chromosomal translocations of the lysine methyltransferase 2A (KMT2A) gene produce KMT2A fusion proteins such as KMT2A-AFF1 (previously MLL-AF4), causing poor prognosis acute lymphoblastic leukemias (ALLs) that sometimes relapse as acute myeloid leukemias (AMLs). KMT2A-AFF1 drives leukemogenesis through direct binding and inducing the aberrant overexpression of key genes, such as the anti-apoptotic factor BCL2 and the proto-oncogene MYC However, studying direct binding alone does not incorporate possible network-generated regulatory outputs, including the indirect induction of gene repression. To better understand the KMT2A-AFF1-driven regulatory landscape, we integrated ChIP-seq, patient RNA-seq, and CRISPR essentiality screens to generate a model GRN. This GRN identified several key transcription factors such as RUNX1 that regulate target genes downstream of KMT2A-AFF1 using feed-forward loop (FFL) and cascade motifs. A core set of nodes are present in both ALL and AML, and CRISPR screening revealed several factors that help mediate response to the drug venetoclax. Using our GRN, we then identified a KMT2A-AFF1:RUNX1 cascade that represses CASP9, as well as KMT2A-AFF1-driven FFLs that regulate BCL2 and MYC through combinatorial TF activity. This illustrates how our GRN can be used to better connect KMT2A-AFF1 behavior to downstream pathways that contribute to leukemogenesis, and potentially predict shifts in gene expression that mediate drug response.

2.
Proc Natl Acad Sci U S A ; 113(32): E4688-97, 2016 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-27444016

RESUMEN

In Parkinson's disease, misfolded α-synuclein accumulates, often in a ubiquitinated form, in neuronal inclusions termed Lewy bodies. An important outstanding question is whether ubiquitination in Lewy bodies is directly relevant to α-synuclein trafficking or turnover and Parkinson's pathogenesis. By comparative analysis in human postmortem brains, we found that ubiquitin immunoreactivity in Lewy bodies is largely due to K63-linked ubiquitin chains and markedly reduced in the substantia nigra compared with the neocortex. The ubiquitin staining in cells with Lewy bodies inversely correlated with the content and pathological localization of the deubiquitinase Usp8. Usp8 interacted and partly colocalized with α-synuclein in endosomal membranes and, both in cells and after purification, it deubiquitinated K63-linked chains on α-synuclein. Knockdown of Usp8 in the Drosophila eye reduced α-synuclein levels and α-synuclein-induced eye toxicity. Accordingly, in human cells, Usp8 knockdown increased the lysosomal degradation of α-synuclein. In the dopaminergic neurons of the Drosophila model, unlike knockdown of other deubiquitinases, Usp8 protected from α-synuclein-induced locomotor deficits and cell loss. These findings strongly suggest that removal of K63-linked ubiquitin chains on α-synuclein by Usp8 is a critical mechanism that reduces its lysosomal degradation in dopaminergic neurons and may contribute to α-synuclein accumulation in Lewy body disease.


Asunto(s)
Endopeptidasas/fisiología , Complejos de Clasificación Endosomal Requeridos para el Transporte/fisiología , Enfermedad por Cuerpos de Lewy/metabolismo , Ubiquitina Tiolesterasa/fisiología , Ubiquitinación , alfa-Sinucleína/metabolismo , Animales , Neuronas Dopaminérgicas/metabolismo , Drosophila , Humanos , Cuerpos de Lewy/metabolismo , Lisosomas/metabolismo , Masculino , Ubiquitina/análisis , alfa-Sinucleína/análisis , alfa-Sinucleína/toxicidad
3.
J Cell Sci ; 129(7): 1477-89, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26906422

RESUMEN

Epithelial bicellular and tricellular junctions are essential for establishing and maintaining permeability barriers. Tricellular junctions are formed by the convergence of three bicellular junctions at the corners of neighbouring epithelia. Gliotactin, a member of the Neuroligin family, is located at theDrosophilatricellular junction, and is crucial for the formation of tricellular and septate junctions, as well as permeability barrier function. Gliotactin protein levels are tightly controlled by phosphorylation at tyrosine residues and endocytosis. Blocking endocytosis or overexpressing Gliotactin results in the spread of Gliotactin from the tricellular junction, resulting in apoptosis, delamination and migration of epithelial cells. We show that Gliotactin levels are also regulated at the mRNA level by micro (mi)RNA-mediated degradation and that miRNAs are targeted to a short region in the 3'UTR that includes a conserved miR-184 target site. miR-184 also targets a suite of septate junction proteins, including NrxIV, coracle and Mcr. miR-184 expression is triggered when Gliotactin is overexpressed, leading to activation of the BMP signalling pathway. Gliotactin specifically interferes with Dad, an inhibitory SMAD, leading to activation of the Tkv type-I receptor and activation of Mad to elevate the biogenesis and expression of miR-184.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Proteínas de la Membrana/metabolismo , MicroARNs/biosíntesis , Proteínas del Tejido Nervioso/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Apoptosis/fisiología , Movimiento Celular/fisiología , Citocinas/metabolismo , Proteínas de Drosophila/antagonistas & inhibidores , Endocitosis/fisiología , Activación Enzimática , Proteínas de la Membrana/genética , MicroARNs/genética , Proteínas del Tejido Nervioso/genética , Serpinas/metabolismo , Transducción de Señal/genética , Uniones Estrechas/fisiología
4.
PLoS Genet ; 11(5): e1005194, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25993106

RESUMEN

RNA interference-related silencing mechanisms concern very diverse and distinct biological processes, from gene regulation (via the microRNA pathway) to defense against molecular parasites (through the small interfering RNA and the Piwi-interacting RNA pathways). Small non-coding RNAs serve as specificity factors that guide effector proteins to ribonucleic acid targets via base-pairing interactions, to achieve transcriptional or post-transcriptional regulation. Because of the small sequence complementarity required for microRNA-dependent post-transcriptional regulation, thousands of microRNA (miRNA) putative targets have been annotated in Drosophila. In Drosophila somatic ovarian cells, genomic parasites, such as transposable elements (TEs), are transcriptionally repressed by chromatin changes induced by Piwi-interacting RNAs (piRNAs) that prevent them from invading the germinal genome. Here we show, for the first time, that a functional miRNA pathway is required for the piRNA-mediated transcriptional silencing of TEs in this tissue. Global miRNA depletion, caused by tissue- and stage-specific knock down of drosha (involved in miRNA biogenesis), AGO1 or gawky (both responsible for miRNA activity), resulted in loss of TE-derived piRNAs and chromatin-mediated transcriptional de-silencing of TEs. This specific TE de-repression was also observed upon individual titration (by expression of the complementary miRNA sponge) of two miRNAs (miR-14 and miR-34) as well as in a miR-14 loss-of-function mutant background. Interestingly, the miRNA defects differentially affected TE- and 3' UTR-derived piRNAs. To our knowledge, this is the first indication of possible differences in the biogenesis or stability of TE- and 3' UTR-derived piRNAs. This work is one of the examples of detectable phenotypes caused by loss of individual miRNAs in Drosophila and the first genetic evidence that miRNAs have a role in the maintenance of genome stability via piRNA-mediated TE repression.


Asunto(s)
Elementos Transponibles de ADN , Proteínas de Drosophila/metabolismo , Drosophila/genética , MicroARNs/metabolismo , Folículo Ovárico/metabolismo , Interferencia de ARN , Animales , Drosophila/metabolismo , Proteínas de Drosophila/genética , Femenino , Regulación de la Expresión Génica , Silenciador del Gen , MicroARNs/genética , Folículo Ovárico/citología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
5.
Genes Dev ; 24(7): 625-35, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20360381

RESUMEN

Post-transcriptional regulatory mechanisms have emerged as a critical component underlying the diversification and spatiotemporal control of the proteome during the establishment of precise neuronal connectivity. These mechanisms have been shown to be important for virtually all stages of assembling a neural network, from neurite guidance, branching, and growth to synapse morphogenesis and function. From the moment a gene is transcribed, it undergoes a series of post-transcriptional regulatory modifications in the nucleus and cytoplasm until its final deployment as a functional protein. Initially, a message is subjected to extensive structural regulation through alternative splicing, which is capable of greatly expanding the protein repertoire by generating, in some cases, thousands of functionally distinct isoforms from a single gene locus. Then, RNA packaging into neuronal transport granules and recognition by RNA-binding proteins and/or microRNAs is capable of restricting protein synthesis to selective locations and under specific input conditions. This ability of the post-transcriptional apparatus to expand the informational content of a cell and control the deployment of proteins in both spatial and temporal dimensions is a feature well adapted for the extreme morphological properties of neural cells. In this review, we describe recent advances in understanding how post-transcriptional regulatory mechanisms refine the proteomic complexity required for the assembly of intricate and specific neural networks.


Asunto(s)
Neurogénesis/fisiología , Neuronas/metabolismo , Procesamiento Postranscripcional del ARN/genética , Empalme Alternativo , Animales , Humanos , MicroARNs/metabolismo , Red Nerviosa/embriología , Neurogénesis/genética , Neuronas/citología , Biosíntesis de Proteínas/fisiología , Transporte de ARN/fisiología , Sinapsis/metabolismo
6.
Development ; 139(15): 2821-31, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22745315

RESUMEN

microRNAs (miRNAs) are endogenous short RNAs that mediate vast networks of post-transcriptional gene regulation. Although computational searches and experimental profiling provide evidence for hundreds of functional targets for individual miRNAs, such data rarely provide clear insight into the phenotypic consequences of manipulating miRNAs in vivo. We describe a genome-wide collection of 165 Drosophila miRNA transgenes and find that a majority induced specific developmental defects, including phenocopies of mutants in myriad cell-signaling and patterning genes. Such connections allowed us to validate several likely targets for miRNA-induced phenotypes. Importantly, few of these phenotypes could be predicted from computationally predicted target lists, thus highlighting the value of whole-animal readouts of miRNA activities. Finally, we provide an example of the relevance of these data to miRNA loss-of-function conditions. Whereas misexpression of several K box miRNAs inhibited Notch pathway activity, reciprocal genetic interaction tests with miRNA sponges demonstrated endogenous roles of the K box miRNA family in restricting Notch signaling. In summary, we provide extensive evidence that misexpression of individual miRNAs often induces specific mutant phenotypes that can guide their functional study. By extension, these data suggest that the deregulation of individual miRNAs in other animals may frequently yield relatively specific phenotypes during disease conditions.


Asunto(s)
Drosophila melanogaster/genética , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , MicroARNs/genética , MicroARNs/metabolismo , Animales , Bases de Datos Genéticas , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Femenino , Perfilación de la Expresión Génica , Genoma , Masculino , Modelos Biológicos , Fenotipo , Receptores Notch/metabolismo , Transducción de Señal , Transgenes , Alas de Animales/fisiología
7.
Nat Cell Biol ; 9(2): 139-48, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17187063

RESUMEN

Hyperphosphorylated forms of the microtubule-associated protein (MAP) tau accumulate in Alzheimer's disease and related tauopathies and are thought to have an important role in neurodegeneration. However, the mechanisms through which phosphorylated tau induces neurodegeneration have remained elusive. Here, we show that tau-induced neurodegeneration is associated with accumulation of filamentous actin (F-actin) and the formation of actin-rich rods in Drosophila and mouse models of tauopathy. Importantly, modulating F-actin levels genetically leads to dramatic modification of tau-induced neurodegeneration. The ability of tau to interact with F-actin in vivo and in vitro provides a molecular mechanism for the observed phenotypes. Finally, we show that the Alzheimer's disease-linked human beta-amyloid protein (Abeta) synergistically enhances the ability of wild-type tau to promote alterations in the actin cytoskeleton and neurodegeneration. These findings raise the possibility that a direct interaction between tau and actin may be a critical mediator of tau-induced neurotoxicity in Alzheimer's disease and related disorders.


Asunto(s)
Actinas/metabolismo , Enfermedad de Alzheimer/metabolismo , Degeneración Nerviosa/metabolismo , Neuronas/metabolismo , Proteínas tau/metabolismo , Actinas/efectos de los fármacos , Péptidos beta-Amiloides/metabolismo , Animales , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Citoesqueleto/patología , Modelos Animales de Enfermedad , Drosophila , Humanos , Inmunohistoquímica , Ratones , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/patología , Neuronas/efectos de los fármacos , Neuronas/patología , Fenotipo , Proteínas tau/farmacología
8.
Nat Commun ; 15(1): 3173, 2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38609390

RESUMEN

Semaphorin-3A (SEMA3A) functions as a chemorepulsive signal during development and can affect T cells by altering their filamentous actin (F-actin) cytoskeleton. The exact extent of these effects on tumour-specific T cells are not completely understood. Here we demonstrate that Neuropilin-1 (NRP1) and Plexin-A1 and Plexin-A4 are upregulated on stimulated CD8+ T cells, allowing tumour-derived SEMA3A to inhibit T cell migration and assembly of the immunological synapse. Deletion of NRP1 in both CD4+ and CD8+ T cells enhance CD8+ T-cell infiltration into tumours and restricted tumour growth in animal models. Conversely, over-expression of SEMA3A inhibit CD8+ T-cell infiltration. We further show that SEMA3A affects CD8+ T cell F-actin, leading to inhibition of immune synapse formation and motility. Examining a clear cell renal cell carcinoma patient cohort, we find that SEMA3A expression is associated with reduced survival, and that T-cells appear trapped in SEMA3A rich regions. Our study establishes SEMA3A as an inhibitor of effector CD8+ T cell tumour infiltration, suggesting that blocking NRP1 could improve T cell function in tumours.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Animales , Humanos , Actinas , Linfocitos T CD8-positivos , Citoesqueleto , Semaforina-3A/genética
9.
Nature ; 448(7151): 362-5, 2007 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-17637670

RESUMEN

Although directed migration is a feature of both individual cells and cell groups, guided migration has been studied most extensively for single cells in simple environments. Collective guidance of cell groups remains poorly understood, despite its relevance for development and metastasis. Neural crest cells and neuronal precursors migrate as loosely organized streams of individual cells, whereas cells of the fish lateral line, Drosophila tracheal tubes and border-cell clusters migrate as more coherent groups. Here we use Drosophila border cells to examine how collective guidance is performed. We report that border cells migrate in two phases using distinct mechanisms. Genetic analysis combined with live imaging shows that polarized cell behaviour is critical for the initial phase of migration, whereas dynamic collective behaviour dominates later. PDGF- and VEGF-related receptor and epidermal growth factor receptor act in both phases, but use different effector pathways in each. The myoblast city (Mbc, also known as DOCK180) and engulfment and cell motility (ELMO, also known as Ced-12) pathway is required for the early phase, in which guidance depends on subcellular localization of signalling within a leading cell. During the later phase, mitogen-activated protein kinase and phospholipase Cgamma are used redundantly, and we find that the cluster makes use of the difference in signal levels between cells to guide migration. Thus, information processing at the multicellular level is used to guide collective behaviour of a cell group.


Asunto(s)
Movimiento Celular , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Proteínas del Citoesqueleto , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Genes Esenciales/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mutación/genética , Fosfolipasa C gamma/metabolismo , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Receptores de Péptidos de Invertebrados/genética , Receptores de Péptidos de Invertebrados/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas Adaptadoras de la Señalización Shc , Proteínas de Unión al GTP rac/metabolismo , Quinasas raf/genética , Quinasas raf/metabolismo
10.
PLoS Genet ; 6(7): e1001026, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20664788

RESUMEN

Expansion of the lysosomal system, including cathepsin D upregulation, is an early and prominent finding in Alzheimer's disease brain. Cell culture studies, however, have provided differing perspectives on the lysosomal connection to Alzheimer's disease, including both protective and detrimental influences. We sought to clarify and molecularly define the connection in vivo in a genetically tractable model organism. Cathepsin D is upregulated with age in a Drosophila model of Alzheimer's disease and related tauopathies. Genetic analysis reveals that cathepsin D plays a neuroprotective role because genetic ablation of cathepsin D markedly potentiates tau-induced neurotoxicity. Further, generation of a C-terminally truncated form of tau found in Alzheimer's disease patients is significantly increased in the absence of cathepsin D. We show that truncated tau has markedly increased neurotoxicity, while solubility of truncated tau is decreased. Importantly, the toxicity of truncated tau is not affected by removal of cathepsin D, providing genetic evidence that modulation of neurotoxicity by cathepsin D is mediated through C-terminal cleavage of tau. We demonstrate that removing cathepsin D in adult postmitotic neurons leads to aberrant lysosomal expansion and caspase activation in vivo, suggesting a mechanism for C-terminal truncation of tau. We also demonstrate that both cathepsin D knockout mice and cathepsin D-deficient sheep show abnormal C-terminal truncation of tau and accompanying caspase activation. Thus, caspase cleavage of tau may be a molecular mechanism through which lysosomal dysfunction and neurodegeneration are causally linked in Alzheimer's disease.


Asunto(s)
Catepsina D/fisiología , Lisosomas/patología , Síndromes de Neurotoxicidad/etiología , Proteínas tau/metabolismo , Enfermedad de Alzheimer , Animales , Caspasas/metabolismo , Drosophila , Lisosomas/metabolismo , Ratones , Ratones Noqueados , Neuronas/patología , Ovinos
11.
Methods Mol Biol ; 2630: 243-264, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36689187

RESUMEN

Posttranscriptional silencing by microRNAs (miRNAs) is a critical constituent of eukaryotic gene regulation. miRNAs are short (~22 nt) noncoding RNAs capable of specifically targeting the miRNA-induced silencing complex (miRISC) to transcripts bearing a complementary miRNA response element (MRE). Although recent methodological advances have greatly improved our understanding of miRNA biogenesis and the mechanisms by which miRNAs repress their cognate targets, exploring the physiological relevance of direct miRNA-target interactions in vivo has remained an outstanding challenge. Here we describe the experimental protocol underlying a novel approach, which allows direct in situ interrogation of specific miRNA-MRE interactions by CRISPR/Cas9-mediated genome engineering (Bassett G et al., Nat Commun 5, 4640, 2014). In this instance, the CRISPR/Cas9 system is first used to catalyze homology-directed replacement of candidate MREs with molecular barcodes at endogenous loci. Subsequently, the effect of MRE mutation on transcript abundance (i.e., MRE activity) can be rapidly evaluated by routine quantitative PCR. This strategy enables functional investigation of a putative miRNA-target pair in a pool of transiently transfected cells, obviating the need for generation of clonal cell lines or transgenic animals. This protocol can be implemented in any cell line in less than 2 weeks and can readily be scaled up for multiplex studies. To facilitate the conceptual workflow underlying this strategy, we also describe a genome-wide resource for automated design and computational evaluation of CRISPR/Cas9 guide RNAs targeting all predicted MREs in various species (miR-CRISPR).


Asunto(s)
MicroARNs , Animales , MicroARNs/genética , Sistemas CRISPR-Cas , Línea Celular , Genoma , Elementos de Respuesta
12.
Neuron ; 57(3): 339-44, 2008 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-18255027

RESUMEN

The formation of the nervous system during embryonic development is controlled by a complex network of signaling pathways which ensure proper migration and targeting of neuronal projections. Likewise, the function of the adult nervous system relies on complex dynamic interactions between the presynaptic and postsynaptic terminals. Here, we review recent advances in understanding the molecular pathways underlying these seemingly distinct processes. These studies reveal that the conserved E3 ubiquitin ligase PHR (PAM, highwire Rpm-1) controls a regulatory protein degradation pathway essential both for axonal targeting during embryonic development as well as for the proper formation and function of neuron muscular junctions (NMJ).


Asunto(s)
Conos de Crecimiento/fisiología , Neuronas/fisiología , Sinapsis/fisiología , Ubiquitina-Proteína Ligasas/fisiología , Animales , Drosophila , Modelos Biológicos
13.
Nat Methods ; 6(12): 897-903, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19915559

RESUMEN

MicroRNAs are important regulators of gene expression, yet the functional outputs of most microRNA-target interactions remain elusive. Here we introduce the Drosophila melanogaster microRNA sponge (miR-SP) as a powerful transgenic technology to dissect the function of every microRNA with precise spatiotemporal resolution. miR-SPs can be used to characterize tissue-specific microRNA loss-of-function phenotypes, define the spatial regulation of their effectors and uncover interactions between microRNAs and other genes. Using themiR-SP system, we identified an essential role of the conserved microRNA miR-8, in neuromuscular junction formation. Tissue-specific silencing revealed that postsynaptic activity of miR-8 is important for normal neuromuscular junction morphogenesis. Given that miR-SPs rely on a bipartite modular expression system, they could be used to elucidate the endogenous function of microRNAs in any species in which conditional expression can be achieved.


Asunto(s)
Drosophila melanogaster/genética , MicroARNs/genética , Animales , Animales Modificados Genéticamente , MicroARNs/antagonistas & inhibidores
14.
CRISPR J ; 5(5): 642-659, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36206027

RESUMEN

CRISPR-Cas9 has emerged as a major genome manipulation tool. As Cas9 can cause off-target effects, several methods for controlling the expression of CRISPR systems were developed. Recent studies have shown that CRISPR activity could be controlled by sensing expression levels of endogenous transcripts. This is particularly interesting, as endogenous RNAs could harbor important information about the cell type, disease state, and environmental challenges cells are facing. Single-guide RNA (sgRNA) engineering played a major role in the development of RNA-responsive CRISPR systems. Following further optimizations, RNA-responsive sgRNAs could enable the development of novel therapeutic and research applications. This review introduces engineering strategies that could be employed to modify Streptococcus pyogenes sgRNAs with a focus on recent advances made toward the development of RNA-responsive sgRNAs. Future directions and potential applications of these technologies are also discussed.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , ARN Pequeño no Traducido , Sistemas CRISPR-Cas/genética , Edición Génica/métodos , ARN , Streptococcus pyogenes/genética , ARN Pequeño no Traducido/genética
15.
Nat Cell Biol ; 4(9): 715-9, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12198500

RESUMEN

The migration of border cells during Drosophila melanogaster oogenesis is a simple and powerful system for studying invasive cell migration in vivo. Border cells are somatic cells that delaminate from the follicular epithelium of an egg chamber and invade the germ line cluster. They migrate between the nurse cells to reach the oocyte, using DE-cadherin for adhesion to the substratum. Border cells take approximately 6 h to migrate a distance of 100 microm. The migration is guided by EGFR (epidermal growth factor receptor) and PVR (platelet-derived growth factor (PDGF)/vascular endothelial growth factor (VEGF) receptor). Here, we show that a single long cellular extension (LCE), several cell diameters in length, is formed at the initiation of migration. The LCE may function as a 'pathfinder' in response to guidance cues. LCE growth requires directional guidance signals and specific adhesion to the substratum. Interference with actin-myosin interactions allows continued LCE growth while preventing translocation of the cell bodies. We discuss similarities between LCEs and axons and the use of LCE-like structures as a general mechanism for initiating invasive migration in vivo.


Asunto(s)
Movimiento Celular/fisiología , Drosophila melanogaster/citología , Actinas/fisiología , Animales , Animales Modificados Genéticamente , Cadherinas/fisiología , Proteínas de Drosophila/fisiología , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/fisiología , Proteínas del Huevo/fisiología , Receptores ErbB/fisiología , Femenino , Genes de Insecto , Proteínas Fluorescentes Verdes , Operón Lac , Proteínas Luminiscentes/genética , Mutación , Oogénesis/fisiología , Transducción de Señal
16.
Methods Mol Biol ; 2162: 89-114, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32926380

RESUMEN

Transfer RNA (tRNA) and their associated production and processing machinery can be coopted as a versatile tool for the production of guide RNAs (gRNAs) for Cas9-based genome engineering. Using different tRNA variants enables the production of gRNAs at a variety of steady state levels. Furthermore, engineered tRNAs can be used to process gRNAs from Pol-II transcripts, thus enabling spatial/temporal control of gRNA expression. Here we describe the design, cloning, and testing of tRNA scaffolds for both Pol-III-driven expression of different levels of gRNAs, and for processing gRNAs from Pol-II transcripts.


Asunto(s)
Biología Molecular/métodos , Regiones Promotoras Genéticas/genética , Procesamiento Postranscripcional del ARN/genética , ARN de Transferencia/genética , Sistemas CRISPR-Cas/genética , Edición Génica , ARN Polimerasa II/genética , ARN Guía de Kinetoplastida/genética
17.
Methods Mol Biol ; 2162: 153-184, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32926382

RESUMEN

The type-II CRISPR-Cas9 system has been repurposed to create synthetic programmable transcriptional regulators (CRISPR-TRs). Subsequent modifications of the system now allow for spatiotemporal control of CRISPR-mediated gene activation and repression. Among these solutions, the development of inducible spacer-blocking hairpin guide RNAs (iSBH-sgRNAs) provide an easy to implement and versatile way to condition the activation of most CRISPR-TRs on the presence of a user defined inducer. In this chapter, I cover the know-how relating to the design and synthesis of iSBH-sgRNAs, as well as the implementation in mammalian cells of inducible CRISPR-TR strategies based on this technology.


Asunto(s)
Sistemas CRISPR-Cas/genética , Edición Génica/métodos , Activación Transcripcional/genética , Animales , Regulación de la Expresión Génica/genética , Humanos , ARN Guía de Kinetoplastida/genética
18.
J Clin Invest ; 117(1): 236-45, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17173140

RESUMEN

Markers of oxidative damage have been detected in brain tissue from patients with Alzheimer disease (AD) and other neurodegenerative disorders. These findings implicate oxidative injury in the neurodegenerative process, but whether oxidative stress is a cause or a consequence of neurotoxicity remains unclear. We used a Drosophila model of human tauopathies to investigate the role of oxidative stress in neurodegeneration. Genetic and pharmacological manipulation of antioxidant defense mechanisms significantly modified neurodegeneration in our model, suggesting that oxidative stress plays a causal role in neurotoxicity. We demonstrate that the JNK signaling pathway is activated in our model, which is in agreement with previous findings in AD tissue. Furthermore, we show that the extent of JNK activation correlates with the degree of tau-induced neurodegeneration. Finally, our findings suggest that oxidative stress acts not to promote tau phosphorylation, but to enhance tau-induced cell cycle activation. In summary, our study identifies oxidative stress as a causal factor in tau-induced neurodegeneration in Drosophila.


Asunto(s)
Degeneración Nerviosa/patología , Estrés Oxidativo/fisiología , Tauopatías/patología , Vitamina E/farmacología , Proteínas tau/toxicidad , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Animales , Modelos Animales de Enfermedad , Drosophila , Humanos , Degeneración Nerviosa/prevención & control , Paraquat/toxicidad , Tauopatías/fisiopatología
19.
Mol Biol Cell ; 18(12): 5060-8, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17928404

RESUMEN

Tau is a microtubule binding protein implicated in a number of human neurodegenerative disorders, including Alzheimer's disease. Phosphorylation of serine-proline/threonine-proline sites, targeted by proline-directed kinases, coincides temporally with neurodegeneration in the human diseases. Recently, we demonstrated that this unique group of serines and threonines has a critical role in controlling tau toxicity in a Drosophila model of tauopathy. Here, we use a combination of genetic and biochemical approaches to examine these sites individually and to determine which of them is primarily responsible for controlling tau neurotoxicity. Despite the importance placed on individual phosphoepitopes and their contributions to disease pathogenesis, our results indicate that no single phosphorylation residue plays a dominant role in controlling tau toxicity. These findings suggest that serine-proline/threonine-proline sites cooperate to mediate neurodegeneration in vivo.


Asunto(s)
Neuronas/metabolismo , Proteínas tau/metabolismo , Aminoácidos/genética , Aminoácidos/metabolismo , Animales , Animales Modificados Genéticamente , Ciclo Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Electroforesis en Gel de Poliacrilamida , Mutación/genética , Neuronas/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Quinasas , Transducción de Señal , Serina-Treonina Quinasas TOR , Proteínas tau/genética
20.
Nat Commun ; 11(1): 1092, 2020 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-32107390

RESUMEN

Micro(mi)RNA-based post-transcriptional regulatory mechanisms have been broadly implicated in the assembly and modulation of synaptic connections required to shape neural circuits, however, relatively few specific miRNAs have been identified that control synapse formation. Using a conditional transgenic toolkit for competitive inhibition of miRNA function in Drosophila, we performed an unbiased screen for novel regulators of synapse morphogenesis at the larval neuromuscular junction (NMJ). From a set of ten new validated regulators of NMJ growth, we discovered that miR-34 mutants display synaptic phenotypes and cell type-specific functions suggesting distinct downstream mechanisms in the presynaptic and postsynaptic compartments. A search for conserved downstream targets for miR-34 identified the junctional receptor CNTNAP4/Neurexin-IV (Nrx-IV) and the membrane cytoskeletal effector Adducin/Hu-li tai shao (Hts) as proteins whose synaptic expression is restricted by miR-34. Manipulation of miR-34, Nrx-IV or Hts-M function in motor neurons or muscle supports a model where presynaptic miR-34 inhibits Nrx-IV to influence active zone formation, whereas, postsynaptic miR-34 inhibits Hts to regulate the initiation of bouton formation from presynaptic terminals.


Asunto(s)
Proteínas de Unión a Calmodulina/genética , Moléculas de Adhesión Celular Neuronal/genética , Proteínas de Drosophila/genética , Regulación del Desarrollo de la Expresión Génica , MicroARNs/metabolismo , Terminales Presinápticos/fisiología , Animales , Animales Modificados Genéticamente , Proteínas de Unión a Calmodulina/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Larva/crecimiento & desarrollo , Morfogénesis/genética , Mutación , Unión Neuromuscular/citología , Unión Neuromuscular/crecimiento & desarrollo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA