Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Neuron ; 109(18): 2864-2883.e8, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34384519

RESUMEN

The molecular and cellular mechanisms underlying complex axon morphogenesis are still poorly understood. We report a novel, evolutionary conserved function for the Drosophila Wnk kinase (dWnk) and its mammalian orthologs, WNK1 and 2, in axon branching. We uncover that dWnk, together with the neuroprotective factor Nmnat, antagonizes the axon-destabilizing factors D-Sarm and Axundead (Axed) during axon branch growth, revealing a developmental function for these proteins. Overexpression of D-Sarm or Axed results in axon branching defects, which can be blocked by overexpression of dWnk or Nmnat. Surprisingly, Wnk kinases are also required for axon maintenance of adult Drosophila and mouse cortical pyramidal neurons. Requirement of Wnk for axon maintenance is independent of its developmental function. Inactivation of dWnk or mouse Wnk1/2 in mature neurons leads to axon degeneration in the adult brain. Therefore, Wnk kinases are novel signaling components that provide a safeguard function in both developing and adult axons.


Asunto(s)
Proteínas del Dominio Armadillo/biosíntesis , Axones/metabolismo , Proteínas del Citoesqueleto/biosíntesis , Proteínas de Drosophila/biosíntesis , Evolución Molecular , Morfogénesis/fisiología , Proteínas Serina-Treonina Quinasas/biosíntesis , Animales , Proteínas del Dominio Armadillo/antagonistas & inhibidores , Proteínas del Dominio Armadillo/genética , Línea Celular Tumoral , Proteínas del Citoesqueleto/antagonistas & inhibidores , Proteínas del Citoesqueleto/genética , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Drosophila melanogaster , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Embarazo , Proteínas Serina-Treonina Quinasas/genética
2.
Dev Cell ; 50(6): 780-792.e7, 2019 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-31447264

RESUMEN

Size trade-offs of visual versus olfactory organs is a pervasive feature of animal evolution. This could result from genetic or functional constraints. We demonstrate that head sensory organ size trade-offs in Drosophila are genetically encoded and arise through differential subdivision of the head primordium into visual versus non-visual fields. We discover that changes in the temporal regulation of the highly conserved eyeless/Pax6 gene expression during development is a conserved mechanism for sensory trade-offs within and between Drosophila species. We identify a natural single nucleotide polymorphism in the cis-regulatory region of eyeless in a binding site of its repressor Cut that is sufficient to alter its temporal regulation and eye size. Because eyeless/Pax6 is a conserved regulator of head sensory placode subdivision, we propose that its temporal regulation is key to define the relative size of head sensory organs.


Asunto(s)
Evolución Biológica , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Regulación del Desarrollo de la Expresión Génica , Órganos de los Sentidos/metabolismo , Animales , Sitios de Unión , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Elementos de Facilitación Genéticos/genética , Ojo/anatomía & histología , Ojo/metabolismo , Femenino , Geografía , Cabeza , Nucleótidos/genética , Tamaño de los Órganos/genética , Polimorfismo de Nucleótido Simple/genética , Factores de Tiempo
3.
J Hematol Oncol ; 11(1): 108, 2018 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-30144809

RESUMEN

BACKGROUND: The CNOT3 protein is a subunit of the CCR4-NOT complex, which is involved in mRNA degradation. We recently identified CNOT3 loss-of-function mutations in patients with T-cell acute lymphoblastic leukemia (T-ALL). METHODS: Here, we use different Drosophila melanogaster eye cancer models to study the potential tumor suppressor function of Not3, the CNOT3 orthologue, and other members of the CCR4-NOT complex. RESULTS: Our data show that knockdown of Not3, the structural components Not1/Not2, and the deadenylases twin/Pop2 all result in increased tumor formation. In addition, overexpression of Not3 could reduce tumor formation. Not3 downregulation has a mild but broad effect on gene expression and leads to increased levels of genes involved in DNA replication and ribosome biogenesis. CycB upregulation also contributes to the Not3 tumor phenotype. Similar findings were obtained in human T-ALL cell lines, pointing out the conserved function of Not3. CONCLUSIONS: Together, our data establish a critical role for Not3 and the entire CCR4-NOT complex as tumor suppressor.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/patogenicidad , Neoplasias del Ojo/genética , Genes Supresores de Tumor/fisiología , Proteínas de Unión al ARN/metabolismo , Ribonucleasas/metabolismo , Animales , Modelos Animales de Enfermedad , Neoplasias del Ojo/metabolismo , Humanos , Unión Proteica
4.
Dev Cell ; 45(1): 53-66.e5, 2018 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-29576424

RESUMEN

The importance of producing the correct numbers of neurons during development is illustrated by both evolutionary enhancement of cognitive capacities in larger brains, and developmental disorders of brain size. In humans, increased neuronal numbers during development is speculated to partly derive from a unique subtype of neural stem cells (NSCs) that undergo a phase of expansion through symmetric self-amplifying divisions before generating neurons. Symmetric amplification also appears to underlie adult neural stem maintenance in the mouse. However, the mechanisms regulating this behavior are unclear. We report the discovery of self-amplifying NSCs in Drosophila and show that they arise by a spatiotemporal conversion of classical self-renewing NSCs. This conversion is regulated by a temporal transition in the expression of proneural transcription factors prior to cell division. We find a causal link between stem cell self-amplification and increased neuronal numbers. We further show that the temporal transcriptional switch controls both stem cell division and subsequent neuronal differentiation.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Drosophila melanogaster/crecimiento & desarrollo , Células-Madre Neurales/citología , Neurogénesis/fisiología , Neuronas/citología , Animales , Recuento de Células , Autorrenovación de las Células , Células Cultivadas , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Femenino , Masculino , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Transducción de Señal , Transcripción Genética
5.
Elife ; 62017 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-28406397

RESUMEN

Animals are characterized by a set of highly conserved developmental regulators. Changes in the cis-regulatory elements of these regulators are thought to constitute the major driver of morphological evolution. However, the role of coding sequence evolution remains unresolved. To address this question, we used the Atonal family of proneural transcription factors as a model. Drosophila atonal coding sequence was endogenously replaced with that of atonal homologues (ATHs) at key phylogenetic positions, non-ATH proneural genes, and the closest homologue to ancestral proneural genes. ATHs and the ancestral-like coding sequences rescued sensory organ fate in atonal mutants, in contrast to non-ATHs. Surprisingly, different ATH factors displayed different levels of proneural activity as reflected by the number and functionality of sense organs. This proneural potency gradient correlated directly with ATH protein stability, including in response to Notch signaling, independently of mRNA levels or codon usage. This establishes a distinct and ancient function for ATHs and demonstrates that coding sequence evolution can underlie quantitative variation in sensory development and function.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas de Drosophila/genética , Drosophila/embriología , Proteínas del Tejido Nervioso/genética , Transcripción Genética , Estructuras Animales/embriología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Morfogénesis , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Recombinación Genética
6.
Front Cell Neurosci ; 11: 416, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29472843

RESUMEN

Injury to the adult central nervous systems (CNS) can result in severe long-term disability because damaged CNS connections fail to regenerate after trauma. Identification of regulators that enhance the intrinsic growth capacity of severed axons is a first step to restore function. Here, we conducted a gain-of-function genetic screen in Drosophila to identify strong inducers of axonal growth after injury. We focus on a novel axis the Down Syndrome Cell Adhesion Molecule (Dscam1), the de-ubiquitinating enzyme Fat Facets (Faf)/Usp9x and the Jun N-Terminal Kinase (JNK) pathway transcription factor Kayak (Kay)/Fos. Genetic and biochemical analyses link these genes in a common signaling pathway whereby Faf stabilizes Dscam1 protein levels, by acting on the 3'-UTR of its mRNA, and Dscam1 acts upstream of the growth-promoting JNK signal. The mammalian homolog of Faf, Usp9x/FAM, shares both the regenerative and Dscam1 stabilizing activities, suggesting a conserved mechanism.

7.
Development ; 143(15): 2760-6, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27385016

RESUMEN

The neurogenin (Ngn) transcription factors control early neurogenesis and neurite outgrowth in mammalian cortex. In contrast to their proneural activity, their function in neurite growth is poorly understood. Drosophila has a single predicted Ngn homolog, Tap, of unknown function. Here we show that Tap is not a proneural protein in Drosophila but is required for proper axonal growth and guidance of neurons of the mushroom body, a neuropile required for associative learning and memory. Genetic and expression analyses suggest that Tap inhibits excessive axonal growth by fine regulation of the levels of the Wnt signaling adaptor protein Dishevelled.


Asunto(s)
Polaridad Celular/fisiología , Proteínas de Drosophila/metabolismo , Neuropéptidos/metabolismo , Factores de Transcripción/metabolismo , Vía de Señalización Wnt/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Orientación del Axón/genética , Orientación del Axón/fisiología , Axones/metabolismo , Polaridad Celular/genética , Drosophila , Proteínas de Drosophila/genética , Cuerpos Pedunculados/metabolismo , Neuropéptidos/genética , Unión Proteica , Factores de Transcripción/genética , Vía de Señalización Wnt/genética
8.
Cell ; 164(3): 460-75, 2016 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-26824657

RESUMEN

Neurogenesis is initiated by the transient expression of the highly conserved proneural proteins, bHLH transcriptional regulators. Here, we discover a conserved post-translational switch governing the duration of proneural protein activity that is required for proper neuronal development. Phosphorylation of a single Serine at the same position in Scute and Atonal proneural proteins governs the transition from active to inactive forms by regulating DNA binding. The equivalent Neurogenin2 Threonine also regulates DNA binding and proneural activity in the developing mammalian neocortex. Using genome editing in Drosophila, we show that Atonal outlives its mRNA but is inactivated by phosphorylation. Inhibiting the phosphorylation of the conserved proneural Serine causes quantitative changes in expression dynamics and target gene expression resulting in neuronal number and fate defects. Strikingly, even a subtle change from Serine to Threonine appears to shift the duration of Atonal activity in vivo, resulting in neuronal fate defects.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/química , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Neurogénesis , Secuencia de Aminoácidos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Drosophila , Proteínas de Drosophila , Ojo/crecimiento & desarrollo , Ojo/ultraestructura , Discos Imaginales/metabolismo , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/metabolismo , Fosforilación , Retina/crecimiento & desarrollo , Alineación de Secuencia
9.
EMBO Mol Med ; 7(4): 423-37, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25693964

RESUMEN

Loss of function of the FMR1 gene leads to fragile X syndrome (FXS), the most common form of intellectual disability. The loss of FMR1 function is usually caused by epigenetic silencing of the FMR1 promoter leading to expansion and subsequent methylation of a CGG repeat in the 5' untranslated region. Very few coding sequence variations have been experimentally characterized and shown to be causal to the disease. Here, we describe a novel FMR1 mutation and reveal an unexpected nuclear export function for the C-terminus of FMRP. We screened a cohort of patients with typical FXS symptoms who tested negative for CGG repeat expansion in the FMR1 locus. In one patient, we identified a guanine insertion in FMR1 exon 15. This mutation alters the open reading frame creating a short novel C-terminal sequence, followed by a stop codon. We find that this novel peptide encodes a functional nuclear localization signal (NLS) targeting the patient FMRP to the nucleolus in human cells. We also reveal an evolutionarily conserved nuclear export function associated with the endogenous C-terminus of FMRP. In vivo analyses in Drosophila demonstrate that a patient-mimetic mutation alters the localization and function of Dfmrp in neurons, leading to neomorphic neuronal phenotypes.


Asunto(s)
Núcleo Celular , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , Síndrome del Cromosoma X Frágil , Mutación , Señales de Localización Nuclear , Expansión de Repetición de Trinucleótido , Animales , Línea Celular Transformada , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/patología , Drosophila melanogaster , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/patología , Humanos , Masculino , Ratones , Señales de Localización Nuclear/genética , Señales de Localización Nuclear/metabolismo , Estructura Terciaria de Proteína , Transporte de Proteínas/genética
10.
PLoS Biol ; 11(5): e1001562, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23690751

RESUMEN

Wnt Planar Cell Polarity (PCP) signaling is a universal regulator of polarity in epithelial cells, but it regulates axon outgrowth in neurons, suggesting the existence of axonal modulators of Wnt-PCP activity. The Amyloid precursor proteins (APPs) are intensely investigated because of their link to Alzheimer's disease (AD). APP's in vivo function in the brain and the mechanisms underlying it remain unclear and controversial. Drosophila possesses a single APP homologue called APP Like, or APPL. APPL is expressed in all neurons throughout development, but has no established function in neuronal development. We therefore investigated the role of Drosophila APPL during brain development. We find that APPL is involved in the development of the Mushroom Body αß neurons and, in particular, is required cell-autonomously for the ß-axons and non-cell autonomously for the α-axons growth. Moreover, we find that APPL is a modulator of the Wnt-PCP pathway required for axonal outgrowth, but not cell polarity. Molecularly, both human APP and fly APPL form complexes with PCP receptors, thus suggesting that APPs are part of the membrane protein complex upstream of PCP signaling. Moreover, we show that APPL regulates PCP pathway activation by modulating the phosphorylation of the Wnt adaptor protein Dishevelled (Dsh) by Abelson kinase (Abl). Taken together our data suggest that APPL is the first example of a modulator of the Wnt-PCP pathway specifically required for axon outgrowth.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Drosophila/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Polaridad Celular , Proteínas Dishevelled , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Células HEK293 , Humanos , Cuerpos Pedunculados/citología , Cuerpos Pedunculados/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo
11.
Elife ; 2: e00337, 2013 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-23471010

RESUMEN

Brain connectivity maps display a delicate balance between individual variation and stereotypy, suggesting the existence of dedicated mechanisms that simultaneously permit and limit individual variation. We show that during the development of the Drosophila central nervous system, mutual inhibition among groups of neighboring postmitotic neurons during development regulates the robustness of axon target choice in a nondeterministic neuronal circuit. Specifically, neighboring postmitotic neurons communicate through Notch signaling during axonal targeting, to ensure balanced alternative axon target choices without a corresponding change in cell fate. Loss of Notch in postmitotic neurons modulates an axon's target choice. However, because neighboring axons respond by choosing the complementary target, the stereotyped connectivity pattern is preserved. In contrast, loss of Notch in clones of neighboring postmitotic neurons results in erroneous coinnervation by multiple axons. Our observations establish mutual inhibition of axonal target choice as a robustness mechanism for brain wiring and unveil a novel cell fate independent function for canonical Notch signaling. DOI:http://dx.doi.org/10.7554/eLife.00337.001.


Asunto(s)
Encéfalo/fisiología , Drosophila/fisiología , Mitosis , Inhibición Neural , Neuronas/fisiología , Vías Visuales/fisiología , Animales , Axones/fisiología , Encéfalo/metabolismo , Línea Celular , Simulación por Computador , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Modelos Biológicos , Neuronas/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal , Factores de Tiempo , Transfección , Vías Visuales/metabolismo , Quinasas p21 Activadas/metabolismo
12.
Nat Genet ; 45(2): 186-90, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23263491

RESUMEN

T-cell acute lymphoblastic leukemia (T-ALL) is caused by the cooperation of multiple oncogenic lesions. We used exome sequencing on 67 T-ALLs to gain insight into the mutational spectrum in these leukemias. We detected protein-altering mutations in 508 genes, with an average of 8.2 mutations in pediatric and 21.0 mutations in adult T-ALL. Using stringent filtering, we predict seven new oncogenic driver genes in T-ALL. We identify CNOT3 as a tumor suppressor mutated in 7 of 89 (7.9%) adult T-ALLs, and its knockdown causes tumors in a sensitized Drosophila melanogaster model. In addition, we identify mutations affecting the ribosomal proteins RPL5 and RPL10 in 12 of 122 (9.8%) pediatric T-ALLs, with recurrent alterations of Arg98 in RPL10. Yeast and lymphoid cells expressing the RPL10 Arg98Ser mutant showed a ribosome biogenesis defect. Our data provide insights into the mutational landscape of pediatric versus adult T-ALL and identify the ribosome as a potential oncogenic factor.


Asunto(s)
Exoma/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Proteínas Ribosómicas/genética , Factores de Transcripción/genética , Animales , Secuencia de Bases , Drosophila melanogaster , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación/genética , Polirribosomas/genética , Interferencia de ARN , Proteína Ribosómica L10 , Saccharomyces cerevisiae , Alineación de Secuencia
13.
Proc Natl Acad Sci U S A ; 107(47): 20553-8, 2010 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-21059961

RESUMEN

In recent years, Drosophila melanogaster has emerged as a powerful model for neuronal circuit development, pathology, and function. A major impediment to these studies has been the lack of a genetically encoded, specific, universal, and phenotypically neutral marker of the somatodendritic compartment. We have developed such a marker and show that it is effective and specific in all neuronal populations tested in the peripheral and central nervous system. The marker, which we name DenMark (Dendritic Marker), is a hybrid protein of the mouse protein ICAM5/Telencephalin and the red fluorescent protein mCherry. We show that DenMark is a powerful tool for revealing novel aspects of the neuroanatomy of developing dendrites, identifying previously unknown dendritic arbors, and elucidating neuronal connectivity.


Asunto(s)
Dendritas/genética , Drosophila melanogaster/genética , Marcadores Genéticos/genética , Proteínas Luminiscentes/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Proteínas Recombinantes de Fusión/metabolismo , Animales , Moléculas de Adhesión Celular/metabolismo , Electrorretinografía , Hipocampo/citología , Inmunohistoquímica , Proteínas Luminiscentes/genética , Glicoproteínas de Membrana/genética , Ratones , Microscopía Confocal , Proteínas del Tejido Nervioso/genética , Proteínas Recombinantes de Fusión/genética , Proteína Fluorescente Roja
14.
PLoS Biol ; 8(7): e1000435, 2010 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-20668662

RESUMEN

A comprehensive systems-level understanding of developmental programs requires the mapping of the underlying gene regulatory networks. While significant progress has been made in mapping a few such networks, almost all gene regulatory networks underlying cell-fate specification remain unknown and their discovery is significantly hampered by the paucity of generalized, in vivo validated tools of target gene and functional enhancer discovery. We combined genetic transcriptome perturbations and comprehensive computational analyses to identify a large cohort of target genes of the proneural and tumor suppressor factor Atonal, which specifies the switch from undifferentiated pluripotent cells to R8 photoreceptor neurons during larval development. Extensive in vivo validations of the predicted targets for the proneural factor Atonal demonstrate a 50% success rate of bona fide targets. Furthermore we show that these enhancers are functionally conserved by cloning orthologous enhancers from Drosophila ananassae and D. virilis in D. melanogaster. Finally, to investigate cis-regulatory cross-talk between Ato and other retinal differentiation transcription factors (TFs), we performed motif analyses and independent target predictions for Eyeless, Senseless, Suppressor of Hairless, Rough, and Glass. Our analyses show that cisTargetX identifies the correct motif from a set of coexpressed genes and accurately predicts target genes of individual TFs. The validated set of novel Ato targets exhibit functional enrichment of signaling molecules and a subset is predicted to be coregulated by other TFs within the retinal gene regulatory network.


Asunto(s)
Drosophila melanogaster/genética , Elementos de Facilitación Genéticos/genética , Perfilación de la Expresión Génica , Genes de Insecto/genética , Genoma/genética , Retina/metabolismo , Sensación/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Sitios de Unión , Secuencia Conservada , Proteínas de Drosophila/metabolismo , Regulación de la Expresión Génica , Redes Reguladoras de Genes/genética , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Mutación/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfoproteínas/metabolismo , Reproducibilidad de los Resultados , Retina/citología , Retina/crecimiento & desarrollo , Factores de Transcripción/metabolismo , Transcripción Genética
15.
Science ; 324(5923): 54, 2009 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-19342580

RESUMEN

Most genes function at multiple stages of metazoan development, in dividing and nondividing cells. Generating mouse conditional knock-outs (cKO), where a gene can be eliminated in a temporally and spatially controlled manner, is a valuable technique because it allows study of gene function at any stage of life. In contrast and despite the development of many other powerful genetic tools, cKO has thus far been lacking in Drosophila. We combined several recent molecular and genetic technical advances in an approach termed integrase-mediated approach for gene knock-out (IMAGO). IMAGO allows the replacement of any genomic sequence, such as a gene, with another desired sequence, including cKO alleles that can be used to create positively marked mutant cells. IMAGO should also be applicable to other genetic model organisms.


Asunto(s)
Drosophila melanogaster/genética , Técnicas de Inactivación de Genes , Mutagénesis , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas de Drosophila , Drosophila melanogaster/citología , Genes de Insecto , Integrasas/metabolismo , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Células Fotorreceptoras de Invertebrados/citología , Células Fotorreceptoras de Invertebrados/fisiología , Recombinación Genética , Órganos de los Sentidos/citología , Órganos de los Sentidos/fisiología
16.
PLoS Genet ; 5(1): e1000351, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19165344

RESUMEN

Genetic screens are powerful methods for the discovery of gene-phenotype associations. However, a systems biology approach to genetics must leverage the massive amount of "omics" data to enhance the power and speed of functional gene discovery in vivo. Thus far, few computational methods for gene function prediction have been rigorously tested for their performance on a genome-wide scale in vivo. In this work, we demonstrate that integrating genome-wide computational gene prioritization with large-scale genetic screening is a powerful tool for functional gene discovery. To discover genes involved in neural development in Drosophila, we extend our strategy for the prioritization of human candidate disease genes to functional prioritization in Drosophila. We then integrate this prioritization strategy with a large-scale genetic screen for interactors of the proneural transcription factor Atonal using genomic deficiencies and mutant and RNAi collections. Using the prioritized genes validated in our genetic screen, we describe a novel genetic interaction network for Atonal. Lastly, we prioritize the whole Drosophila genome and identify candidate gene associations for ten receptor-signaling pathways. This novel database of prioritized pathway candidates, as well as a web application for functional prioritization in Drosophila, called Endeavour-HighFly, and the Atonal network, are publicly available resources. A systems genetics approach that combines the power of computational predictions with in vivo genetic screens strongly enhances the process of gene function and gene-gene association discovery.


Asunto(s)
Biología Computacional/métodos , Drosophila melanogaster/genética , Animales , Bases de Datos Genéticas , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Técnicas Genéticas , Genética , Genoma , Inmunohistoquímica , Modelos Genéticos , Fenotipo , Mapeo de Interacción de Proteínas , Interferencia de ARN , Transducción de Señal
17.
Dev Dyn ; 237(11): 3352-60, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18855900

RESUMEN

BBP proteins constitute a subclass of CUL3 interacting BTB proteins whose in vivo function remains unknown. Here, we show that the Xenopus BBP gene BTBD6 and the single Drosophila homologue of mammalian BBP genes lute are strongly expressed in the developing nervous system. In Xenopus, BTBD6 expression responds positively to proneural and negatively to neurogenic gene overexpression. Knockdown of BTBD6 in Xenopus or loss of Drosophila lute result in embryos with strong defects in late neuronal markers and strongly reduced and disorganized axons while early neural development is unaffected. XBTBD6 knockdown in Xenopus also affects muscle development. Together, these data indicate that BTBD6/lute is required for proper embryogenesis and plays an essential evolutionary conserved role during neuronal development.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Drosophila/inmunología , Desarrollo de Músculos/fisiología , Proteínas Musculares/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/fisiología , Proteínas de Xenopus/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Técnicas de Silenciamiento del Gen , Humanos , Proteínas Musculares/genética , Proteínas del Tejido Nervioso/genética , Sistema Nervioso/citología , Sistema Nervioso/embriología , Homología de Secuencia de Aminoácido , Proteínas de Xenopus/genética , Xenopus laevis
18.
Nucleic Acids Res ; 36(18): e114, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18676454

RESUMEN

Studying gene function in the post-genome era requires methods to localize and inactivate proteins in a standardized fashion in model organisms. While genome-wide gene disruption and over-expression efforts are well on their way to vastly expand the repertoire of Drosophila tools, a complementary method to efficiently and quickly tag proteins expressed under endogenous control does not exist for fruit flies. Here, we describe the development of an efficient procedure to generate protein fusions at either terminus in an endogenous genomic context using recombineering. We demonstrate that the fluorescent protein tagged constructs, expressed under the proper control of regulatory elements, can rescue the respective mutations and enable the detection of proteins in vivo. Furthermore, we also adapted our method for use of the tetracysteine tag that tightly binds the fluorescent membrane-permeable FlAsH ligand. This technology allows us to acutely inactivate any tagged protein expressed under native control using fluorescein-assisted light inactivation and we provide proof of concept by demonstrating that acute loss of clathrin heavy chain function in the fly eye leads to synaptic transmission defects in photoreceptors. Our tagging technology is efficient and versatile, adaptable to any tag desired and paves the way to genome-wide gene tagging in Drosophila.


Asunto(s)
Drosophila melanogaster/genética , Ingeniería de Proteínas/métodos , Proteínas Recombinantes de Fusión/análisis , Animales , Cadenas Pesadas de Clatrina/genética , Drosophila melanogaster/fisiología , Fluoresceínas/química , Colorantes Fluorescentes/química , Vectores Genéticos , Genoma de los Insectos , Genómica/métodos , Compuestos Organometálicos/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Recombinación Genética , Transmisión Sináptica
19.
J Neurosci ; 28(23): 6010-21, 2008 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-18524906

RESUMEN

Drosophila melanogaster is a leading genetic model system in nervous system development and disease research. Using the power of fly genetics in traumatic axonal injury research will significantly speed up the characterization of molecular processes that control axonal regeneration in the CNS. We developed a versatile and physiologically robust preparation for the long-term culture of the whole Drosophila brain. We use this method to develop a novel Drosophila model for CNS axonal injury and regeneration. We first show that, similar to mammalian CNS axons, injured adult wild-type fly CNS axons fail to regenerate, whereas adult-specific enhancement of protein kinase A activity increases the regenerative capacity of lesioned neurons. Combined, these observations suggest conservation of neuronal regeneration mechanisms after injury. We next exploit this model to explore pathways that induce robust regeneration and find that adult-specific activation of c-Jun N-terminal protein kinase signaling is sufficient for de novo CNS axonal regeneration injury, including the growth of new axons past the lesion site and into the normal target area.


Asunto(s)
Axones/patología , Axones/fisiología , Encéfalo/crecimiento & desarrollo , Lesión Axonal Difusa/patología , Lesión Axonal Difusa/fisiopatología , Regeneración Nerviosa/fisiología , Factores de Edad , Animales , Encéfalo/citología , Células Cultivadas , Lesión Axonal Difusa/genética , Modelos Animales de Enfermedad , Drosophila melanogaster/genética , Regeneración Nerviosa/genética , Técnicas de Cultivo de Órganos
20.
Development ; 131(8): 1679-89, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15084454

RESUMEN

How conserved pathways are differentially regulated to produce diverse outcomes is a fundamental question of developmental and evolutionary biology. The conserved process of neural precursor cell (NPC) selection by basic helix-loop-helix (bHLH) proneural transcription factors in the peripheral nervous system (PNS) by atonal related proteins (ARPs) presents an excellent model in which to address this issue. Proneural ARPs belong to two highly related groups: the ATONAL (ATO) group and the NEUROGENIN (NGN) group. We used a cross-species approach to demonstrate that the genetic and molecular mechanisms by which ATO proteins and NGN proteins select NPCs are different. Specifically, ATO group genes efficiently induce neurogenesis in Drosophila but very weakly in Xenopus, while the reverse is true for NGN group proteins. This divergence in proneural activity is encoded by three residues in the basic domain of ATO proteins. In NGN proteins, proneural capacity is encoded by the equivalent three residues in the basic domain and a novel motif in the second Helix (H2) domain. Differential interactions with different types of zinc (Zn)-finger proteins mediate the divergence of ATO and NGN activities: Senseless is required for ATO group activity, whereas MyT1 is required for NGN group function. These data suggest an evolutionary divergence in the mechanisms of NPC selection between protostomes and deuterostomes.


Asunto(s)
Diferenciación Celular/fisiología , Evolución Molecular , Proteínas del Tejido Nervioso/genética , Neuronas/fisiología , Células Madre/fisiología , Animales , Animales Modificados Genéticamente , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Drosophila/genética , Drosophila/fisiología , Proteínas de Drosophila , Proteínas del Tejido Nervioso/fisiología , Xenopus , Proteínas de Xenopus/genética , Proteínas de Xenopus/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA