Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
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
2.
Genome Res ; 29(10): 1659-1672, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31515287

RESUMEN

Induction and reversal of chromatin silencing is critical for successful development, tissue homeostasis, and the derivation of induced pluripotent stem cells (iPSCs). X-Chromosome inactivation (XCI) and reactivation (XCR) in female cells represent chromosome-wide transitions between active and inactive chromatin states. Although XCI has long been studied, providing important insights into gene regulation, the dynamics and mechanisms underlying the reversal of stable chromatin silencing of X-linked genes are much less understood. Here, we use allele-specific transcriptomics to study XCR during mouse iPSC reprogramming in order to elucidate the timing and mechanisms of chromosome-wide reversal of gene silencing. We show that XCR is hierarchical, with subsets of genes reactivating early, late, and very late during reprogramming. Early genes are activated before the onset of late pluripotency genes activation. Early genes are located genomically closer to genes that escape XCI, unlike genes reactivating late. Early genes also show increased pluripotency transcription factor (TF) binding. We also reveal that histone deacetylases (HDACs) restrict XCR in reprogramming intermediates and that the severe hypoacetylation state of the inactive X Chromosome (Xi) persists until late reprogramming stages. Altogether, these results reveal the timing of transcriptional activation of monoallelically repressed genes during iPSC reprogramming, and suggest that allelic activation involves the combined action of chromatin topology, pluripotency TFs, and chromatin regulators. These findings are important for our understanding of gene silencing, maintenance of cell identity, reprogramming, and disease.


Asunto(s)
Reprogramación Celular/genética , Células Madre Pluripotentes Inducidas/citología , ARN Largo no Codificante/genética , Inactivación del Cromosoma X/genética , Animales , Cromatina/genética , Femenino , Silenciador del Gen , Genes Ligados a X/genética , Histona Desacetilasas/genética , Ratones , Activación Transcripcional/genética , Cromosoma X/genética
3.
J Cell Sci ; 132(20)2019 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-31519808

RESUMEN

Reprogramming to induced pluripotency induces the switch of somatic cell identity to induced pluripotent stem cells (iPSCs). However, the mediators and mechanisms of reprogramming remain largely unclear. To elucidate the mediators and mechanisms of reprogramming, we used a siRNA-mediated knockdown approach for selected candidate genes during the conversion of somatic cells into iPSCs. We identified Tox4 as a novel factor that modulates cell fate through an assay that determined the efficiency of iPSC reprogramming. We found that Tox4 is needed early in reprogramming to efficiently generate early reprogramming intermediates, irrespective of the reprogramming conditions used. Tox4 enables proper exogenous reprogramming factor expression, and the closing and opening of putative somatic and pluripotency enhancers early during reprogramming, respectively. We show that the TOX4 protein assembles into a high molecular form. Moreover, Tox4 is also required for the efficient conversion of fibroblasts towards the neuronal fate, suggesting a broader role of Tox4 in modulating cell fate. Our study reveals Tox4 as a novel transcriptional modulator of cell fate that mediates reprogramming from the somatic state to the pluripotent and neuronal fate.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Reprogramación Celular , Fibroblastos/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Células-Madre Neurales/metabolismo , Animales , Línea Celular , Fibroblastos/citología , Proteínas del Grupo de Alta Movilidad/genética , Células Madre Pluripotentes Inducidas/citología , Ratones , Células-Madre Neurales/citología
4.
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
5.
PLoS Biol ; 7(2): e40, 2009 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-19243220

RESUMEN

The acquisition of terminal cell fate and onset of differentiation are instructed by cell type-specific master control genes. Loss of differentiation is frequently observed during cancer progression, but the underlying causes and mechanisms remain poorly understood. We tested the hypothesis that master regulators of differentiation may be key regulators of tumor formation. Using loss- and gain-of-function analyses in Drosophila, we describe a critical anti-oncogenic function for the atonal transcription factor in the fly retina, where atonal instructs tissue differentiation. In the tumor context, atonal acts by regulating cell proliferation and death via the JNK stress response pathway. Combined with evidence that atonal's mammalian homolog, ATOH1, is a tumor suppressor gene, our data support a critical, evolutionarily conserved, function for ato in oncogenesis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Transformación Celular Neoplásica/genética , Drosophila , Neoplasias del Ojo/genética , Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Proteínas del Tejido Nervioso/genética , Animales , Apoptosis/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Tipificación del Cuerpo/genética , Diferenciación Celular/genética , Drosophila/embriología , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Neoplasias del Ojo/metabolismo , Silenciador del Gen , Proteínas del Tejido Nervioso/metabolismo , Organismos Modificados Genéticamente , Retina/citología , Retina/embriología
6.
PLoS Biol ; 7(2): e39, 2009 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-19243219

RESUMEN

Colon cancer accounts for more than 10% of all cancer deaths annually. Our genetic evidence from Drosophila and previous in vitro studies of mammalian Atonal homolog 1 (Atoh1, also called Math1 or Hath1) suggest an anti-oncogenic function for the Atonal group of proneural basic helix-loop-helix transcription factors. We asked whether mouse Atoh1 and human ATOH1 act as tumor suppressor genes in vivo. Genetic knockouts in mouse and molecular analyses in the mouse and in human cancer cell lines support a tumor suppressor function for ATOH1. ATOH1 antagonizes tumor formation and growth by regulating proliferation and apoptosis, likely via activation of the Jun N-terminal kinase signaling pathway. Furthermore, colorectal cancer and Merkel cell carcinoma patients show genetic and epigenetic ATOH1 loss-of-function mutations. Our data indicate that ATOH1 may be an early target for oncogenic mutations in tissues where it instructs cellular differentiation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Carcinoma de Células de Merkel/genética , Neoplasias Colorrectales/genética , Genes Supresores de Tumor/fisiología , Neoplasias Cutáneas/genética , Animales , Apoptosis/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinoma de Células de Merkel/metabolismo , Carcinoma de Células de Merkel/patología , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Análisis Mutacional de ADN , Regulación Neoplásica de la Expresión Génica , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Proteínas Quinasas JNK Activadas por Mitógenos , Masculino , Ratones , Ratones Noqueados , Mutación , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología
7.
Mol Cancer Ther ; 21(4): 568-581, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35149549

RESUMEN

Clinical use of doxorubicin (Dox) is limited by cumulative myelo- and cardiotoxicity. This research focuses on the detailed characterization of PhAc-ALGP-Dox, a targeted tetrapeptide prodrug with a unique dual-step activation mechanism, designed to circumvent Dox-related toxicities and is ready for upcoming clinical investigation. Coupling Dox to a phosphonoacetyl (PhAc)-capped tetrapeptide forms the cell-impermeable, inactive compound, PhAc-ALGP-Dox. After extracellular cleavage by tumor-enriched thimet oligopeptidase-1 (THOP1), a cell-permeable but still biologically inactive dipeptide-conjugate is formed (GP-Dox), which is further processed intracellularly to Dox by fibroblast activation protein-alpha (FAPα) and/or dipeptidyl peptidase-4 (DPP4). In vitro, PhAc-ALGP-Dox is effective in various 2D- and 3D-cancer models, while showing improved safety toward normal epithelium, hematopoietic progenitors, and cardiomyocytes. In vivo, these results translate into a 10-fold higher tolerability and 5-fold greater retention of Dox in the tumor microenvironment compared with the parental drug. PhAc-ALGP-Dox demonstrates 63% to 96% tumor growth inhibition in preclinical models, an 8-fold improvement in efficacy in patient-derived xenograft (PDX) models, and reduced metastatic burden in a murine model of experimental lung metastasis, improving survival by 30%. The current findings highlight the potential clinical benefit of PhAc-ALGP-Dox, a targeted drug-conjugate with broad applicability, favorable tissue biodistribution, significantly improved tolerability, and tumor growth inhibition at primary and metastatic sites in numerous solid tumor models.


Asunto(s)
Antineoplásicos , Neoplasias Pulmonares , Profármacos , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Doxorrubicina/química , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Profármacos/farmacología , Profármacos/uso terapéutico , Índice Terapéutico , Distribución Tisular , Microambiente Tumoral
8.
Genome Biol ; 22(1): 302, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34724962

RESUMEN

BACKGROUND: Precise gene dosage of the X chromosomes is critical for normal development and cellular function. In mice, XX female somatic cells show transcriptional X chromosome upregulation of their single active X chromosome, while the other X chromosome is inactive. Moreover, the inactive X chromosome is reactivated during development in the inner cell mass and in germ cells through X chromosome reactivation, which can be studied in vitro by reprogramming of somatic cells to pluripotency. How chromatin processes and gene regulatory networks evolved to regulate X chromosome dosage in the somatic state and during X chromosome reactivation remains unclear. RESULTS: Using genome-wide approaches, allele-specific ATAC-seq and single-cell RNA-seq, in female embryonic fibroblasts and during reprogramming to pluripotency, we show that chromatin accessibility on the upregulated mammalian active X chromosome is increased compared to autosomes. We further show that increased accessibility on the active X chromosome is erased by reprogramming, accompanied by erasure of transcriptional X chromosome upregulation and the loss of increased transcriptional burst frequency. In addition, we characterize gene regulatory networks during reprogramming and X chromosome reactivation, revealing changes in regulatory states. Our data show that ZFP42/REX1, a pluripotency-associated gene that evolved specifically in placental mammals, targets multiple X-linked genes, suggesting an evolutionary link between ZFP42/REX1, X chromosome reactivation, and pluripotency. CONCLUSIONS: Our data reveal the existence of intrinsic compensatory mechanisms that involve modulation of chromatin accessibility to counteract X-to-Autosome gene dosage imbalances caused by evolutionary or in vitro X chromosome loss and X chromosome inactivation in mammalian cells.


Asunto(s)
Cromatina/metabolismo , Inactivación del Cromosoma X , Alelos , Aneuploidia , Animales , Reprogramación Celular/genética , Redes Reguladoras de Genes , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , RNA-Seq , Análisis de la Célula Individual , Factores de Transcripción/metabolismo , Transcripción Genética , Cromosoma X
9.
Nat Cell Biol ; 23(1): 49-60, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33420491

RESUMEN

Totipotency is the ability of a single cell to give rise to all of the differentiated cell types that build the conceptus, yet how to capture this property in vitro remains incompletely understood. Defining totipotency relies on a variety of assays of variable stringency. Here, we describe criteria to define totipotency. We explain how distinct criteria of increasing stringency can be used to judge totipotency by evaluating candidate totipotent cell types in mice, including early blastomeres and expanded or extended pluripotent stem cells. Our data challenge the notion that expanded or extended pluripotent states harbour increased totipotent potential relative to conventional embryonic stem cells under in vitro and in vivo conditions.


Asunto(s)
Blastómeros/citología , Diferenciación Celular , Linaje de la Célula/genética , Embrión de Mamíferos/citología , Células Madre Embrionarias/citología , Células Madre Pluripotentes/citología , Células Madre Totipotentes/citología , Animales , Blastómeros/metabolismo , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/metabolismo , Femenino , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Masculino , Ratones , Células Madre Pluripotentes/metabolismo , Análisis de la Célula Individual , Células Madre Totipotentes/metabolismo
10.
Biochemistry ; 49(8): 1577-89, 2010 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-20102160

RESUMEN

Neuronal specification is regulated by the activity of transcription factors containing the basic helix-loop-helix motif (bHLH); these regulating proteins include, among others, the neurogenin (Ngn) family, related to the atonal family of genes. Neurogenin 1 (NGN1) is a 237-residue protein that contains a bHLH domain and is involved in neuronal differentiation. In this work, we synthesized the bHLH region of NGN1 (bHLHN) comprising residues 90-150 of the full-length NGN1. The domain is a monomeric natively unfolded protein with a pH-dependent premolten globule conformation, as shown by several spectroscopic techniques (namely, NMR, fluorescence, FTIR, and circular dichroism). The unfolded character of the domain also explains, first, the impossibility of its overexpression in several Escherichia coli strains and, second, its insolubility in aqueous buffers. To the best of our knowledge, this is the first extensive study of the conformational preferences of a bHLH domain under different solution conditions. Upon binding to two DNA E-boxes, the protein forms "fuzzy" complexes (that is, the complexes were not fully folded). The affinities of bHLHN for both DNA boxes were smaller than those of other bHLH domains, which might explain why the protein-DNA complexes were not fully folded.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/química , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , ADN/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Dicroismo Circular , Secuencias Hélice-Asa-Hélice/genética , Secuencias Hélice-Asa-Hélice/fisiología , Humanos , Proteínas del Tejido Nervioso/genética , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Pliegue de Proteína , Espectroscopía Infrarroja por Transformada de Fourier
11.
Stem Cell Reports ; 12(2): 333-350, 2019 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-30639215

RESUMEN

Reprogramming female mouse somatic cells into induced pluripotent stem cells (iPSCs) leads to X-chromosome reactivation. The extent to which increased X-chromosome dosage (X-dosage) in female iPSCs compared with male iPSCs leads to differences in the properties of iPSCs is still unclear. We show that chromatin accessibility in mouse iPSCs is modulated by X-dosage. Specific sets of transcriptional regulator motifs are enriched in chromatin with increased accessibility in XX or XY iPSCs. The transcriptome, growth and pluripotency exit are also modulated by X-dosage in iPSCs. To understand how increased X-dosage modulates the properties of mouse pluripotent stem cells, we used heterozygous deletions of the X-linked gene Dusp9. We show that X-dosage regulates the transcriptome, open chromatin landscape, growth, and pluripotency exit largely independently of global DNA methylation. Our results provide insights into how gene dosage modulates the epigenetic and genetic mechanisms that regulate cell identity.


Asunto(s)
Metilación de ADN/genética , Dosificación de Gen/genética , Células Madre Pluripotentes/fisiología , Cromosoma X/genética , Animales , Línea Celular , Reprogramación Celular/genética , Cromatina/genética , Fosfatasas de Especificidad Dual/genética , Epigénesis Genética/genética , Epigenómica/métodos , Femenino , Células Madre Pluripotentes Inducidas/fisiología , Masculino , Ratones , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/genética , Transcriptoma/genética
12.
Epigenet Insights ; 11: 2516865718802931, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30443643

RESUMEN

How the epigenome of one cell type is remodeled during reprogramming into another unrelated type of cell remains unclear. Overexpression of transcription factors in somatic cells enables the induction of induced pluripotent stem cells (iPSCs). This process entails genome-wide remodeling of DNA methylation, chromatin, and transcription. Recent work suggests that the number of active X chromosomes present in a cell influences remodeling of DNA methylation during somatic cell reprogramming to mouse iPSCs. Female iPSCs with 2 active X chromosomes display global DNA hypomethylation, whereas male XY iPSCs show DNA methylation levels similar to the somatic cells they are derived from. Global DNA methylation erasure in female iPSCs takes place genome-wide and involves repression of DNA methyltransferases. However, on loss of one X chromosome, female iPSCs acquire a DNA methylation landscape resembling that of XY iPSCs. Therefore, it is the X chromosome dosage that dictates global DNA methylation levels in iPSCs. Here, we discuss the evidence that links X chromosome dosage with the regulation of DNA methylation in pluripotent stem cells. We focus on iPSCs reprogramming studies, where X chromosome status is a novel factor impacting our understanding of epigenetic remodeling.

13.
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
14.
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.

15.
Brain Res ; 1121(1): 238-45, 2006 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-17046729

RESUMEN

After our initial discovery of under expression of the GABA(A) receptor delta subunit in a genome wide screening for differentially expressed mRNAs in brain of fragile X mice, a validated model for fragile X mental retardation syndrome, we analyzed expression of the 17 remaining subunits of the GABA(A) receptor using real-time PCR. We confirmed nearly 50% under expression of the delta subunit and found a significant 35%-50% reduction in expression of 7 additional subunit mRNAs, namely alpha(1), alpha(3), and alpha(4), beta(1) and beta(2) and gamma(1) and gamma(2), in fragile X mice compared to wild-type littermates. In concordance with previous results, under expression was found in cortex, but not in cerebellum. Moreover, decreased expression of specific GABA(A) receptor subunits in fragile X syndrome seems to be an evolutionary conserved hallmark since in the fragile X fly (Drosophila melanogaster) model we also found almost 50% under expression of all 3 subunits which make up the invertebrate GABA receptor, namely Grd, Rdl and Lcch3. In addition, we demonstrated a direct correlation between the amount of dFmrp and the expression of the GABA receptor subunits Rdl and Grd. Our results add evidence to previous observations of an altered GABAergic system in fragile X syndrome. Because GABA(A) receptors are the major inhibitory receptors in brain, involved in anxiety, depression, insomnia, learning and memory and epilepsy, processes also disturbed in fragile X patients, the well described GABA(A) receptor pharmacology might open new powerful opportunities for treatment of the behavioral and epileptic phenotype associated with fragile X syndrome.


Asunto(s)
Síndrome del Cromosoma X Frágil/genética , Regulación de la Expresión Génica , Receptores de GABA-A/genética , Animales , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Regulación hacia Abajo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación , ARN Mensajero/genética
16.
Dev Cell ; 39(2): 267-278, 2016 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-27780041

RESUMEN

The axonal wiring molecule Slit and its Round-About (Robo) receptors are conserved regulators of nerve cord patterning. Robo receptors also contribute to wiring brain circuits. Whether molecular mechanisms regulating these signals are modified to fit more complex brain wiring processes is unclear. We investigated the role of Slit and Robo receptors in wiring Drosophila higher-order brain circuits and identified differences in the cellular and molecular mechanisms of Robo/Slit function. First, we find that signaling by Robo receptors in the brain is regulated by the Receptor Protein Tyrosine Phosphatase RPTP69d. RPTP69d increases membrane availability of Robo3 without affecting its phosphorylation state. Second, we detect no midline localization of Slit during brain development. Instead, Slit is enriched in the mushroom body, a neuronal structure covering large areas of the brain. Thus, a divergent molecular mechanism regulates neuronal circuit wiring in the Drosophila brain, partly in response to signals from the mushroom body.


Asunto(s)
Encéfalo/metabolismo , Proteínas de Drosophila/metabolismo , Red Nerviosa/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurópilo/metabolismo , Proteínas Tirosina Fosfatasas Similares a Receptores/metabolismo , Receptores Inmunológicos/metabolismo , Transducción de Señal , Animales , Axones/metabolismo , Membrana Celular/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Epistasis Genética , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Humanos , Larva/metabolismo , Complejos Multiproteicos/metabolismo , Cuerpos Pedunculados/metabolismo , Proteínas del Tejido Nervioso/genética , Fenotipo
17.
Micron ; 35(1-2): 99-100, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15036305

RESUMEN

Glycopeptides were isolated from functional units of two molluscan hemocyanins (Hcs). They were analyzed and localized in the sequences. A comparison with potential N-glycosylation sites of two other molluscan Hcs was made. An immunological cross-reactivity was observed between the beta-Hc and the alpha-macroglobulin of Helix pomotia. ELISA experiments with glycopeptide fractions indicated a competition.


Asunto(s)
Caracoles Helix/química , Hemocianinas/química , Moluscos/química , Animales , Glicosilación , Hemocianinas/inmunología , Subunidades de Proteína
18.
Neural Dev ; 9: 4, 2014 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-24571981

RESUMEN

BACKGROUND: Vertebrates and invertebrates obtain visual motion information by channeling moving visual cues perceived by the retina through specific motion sensitive synaptic relays in the brain. In Drosophila, the series of synaptic relays forming the optic lobe are known as the lamina, medulla, lobula and lobula plate neuropiles. The fly's motion detection output neurons, called the T4 and T5 cells, reside in the lobula plate. Adult optic lobe neurons are derived from larval neural progenitors in two proliferating compartments known as the outer and inner proliferation centers (OPC and IPC). Important insight has been gained into molecular mechanisms involved in the development of the lamina and medulla from the OPC, though less is known about the development of the lobula and lobula plate. RESULTS: Here we show that the proneural gene Atonal is expressed in a subset of IPC progenitors that give rise to the higher order motion detection neurons, T4 and T5, of the lobula plate. We also show that Atonal does not act as a proneural gene in this context. Rather, it is required specifically in IPC neural progenitors to regulate neurite outgrowth in the neuronal progeny. CONCLUSIONS: Our findings reveal that a proneural gene is expressed in progenitors but is required for neurite development of their progeny neurons. This suggests that transcriptional programs initiated specifically in progenitors are necessary for subsequent neuronal morphogenesis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas del Tejido Nervioso/genética , Células-Madre Neurales/metabolismo , Neuritas/metabolismo , Neuronas/metabolismo , Lóbulo Óptico de Animales no Mamíferos/metabolismo , Animales , Proteínas de Drosophila , Drosophila melanogaster , Percepción de Movimiento/fisiología
19.
Elife ; 3: e01699, 2014 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-24755286

RESUMEN

Axonal branching allows a neuron to connect to several targets, increasing neuronal circuit complexity. While axonal branching is well described, the mechanisms that control it remain largely unknown. We find that in the Drosophila CNS branches develop through a process of excessive growth followed by pruning. In vivo high-resolution live imaging of developing brains as well as loss and gain of function experiments show that activation of Epidermal Growth Factor Receptor (EGFR) is necessary for branch dynamics and the final branching pattern. Live imaging also reveals that intrinsic asymmetry in EGFR localization regulates the balance between dynamic and static filopodia. Elimination of signaling asymmetry by either loss or gain of EGFR function results in reduced dynamics leading to excessive branch formation. In summary, we propose that the dynamic process of axon branch development is mediated by differential local distribution of signaling receptors. DOI: http://dx.doi.org/10.7554/eLife.01699.001.


Asunto(s)
Axones/fisiología , Plasticidad Neuronal , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal , Animales , Drosophila , Proteínas de Drosophila/metabolismo , Receptores ErbB/metabolismo , Imagen Óptica , Receptores de Péptidos de Invertebrados/metabolismo
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA