Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Dev Neurobiol ; 84(3): 217-235, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38837880

RESUMEN

The Pcdhg gene cluster encodes 22 γ-Protocadherin (γ-Pcdh) cell adhesion molecules that critically regulate multiple aspects of neural development, including neuronal survival, dendritic and axonal arborization, and synapse formation and maturation. Each γ-Pcdh isoform has unique protein domains-a homophilically interacting extracellular domain and a juxtamembrane cytoplasmic domain-as well as a C-terminal cytoplasmic domain shared by all isoforms. The extent to which isoform-specific versus shared domains regulate distinct γ-Pcdh functions remains incompletely understood. Our previous in vitro studies identified protein kinase C (PKC) phosphorylation of a serine residue within a shared C-terminal motif as a mechanism through which γ-Pcdh promotion of dendrite arborization via myristoylated alanine-rich C-kinase substrate (MARCKS) is abrogated. Here, we used CRISPR/Cas9 genome editing to generate two new mouse lines expressing only non-phosphorylatable γ-Pcdhs, due either to a serine-to-alanine mutation (PcdhgS/A) or to a 15-amino acid C-terminal deletion resulting from insertion of an early stop codon (PcdhgCTD). Both lines are viable and fertile, and the density and maturation of dendritic spines remain unchanged in both PcdhgS/A and PcdhgCTD cortex. Dendrite arborization of cortical pyramidal neurons, however, is significantly increased in both lines, as are levels of active MARCKS. Intriguingly, despite having significantly reduced levels of γ-Pcdh proteins, the PcdhgCTD mutation yields the strongest phenotype, with even heterozygous mutants exhibiting increased arborization. The present study confirms that phosphorylation of a shared C-terminal motif is a key γ-Pcdh negative regulation point and contributes to a converging understanding of γ-Pcdh family function in which distinct roles are played by both individual isoforms and discrete protein domains.


Asunto(s)
Proteínas Relacionadas con las Cadherinas , Cadherinas , Corteza Cerebral , Dendritas , Proteína Quinasa C , Animales , Corteza Cerebral/metabolismo , Corteza Cerebral/citología , Cadherinas/metabolismo , Cadherinas/genética , Fosforilación/fisiología , Dendritas/metabolismo , Ratones , Proteína Quinasa C/metabolismo , Proteína Quinasa C/genética , Sustrato de la Proteína Quinasa C Rico en Alanina Miristoilada/metabolismo , Sustrato de la Proteína Quinasa C Rico en Alanina Miristoilada/genética , Secuencias de Aminoácidos/fisiología , Ratones Transgénicos
2.
bioRxiv ; 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38328061

RESUMEN

The Pcdhg gene cluster encodes 22 γ-Protocadherin (γ-Pcdh) cell adhesion molecules that critically regulate multiple aspects of neural development, including neuronal survival, dendritic and axonal arborization, and synapse formation and maturation. Each γ-Pcdh isoform has unique protein domains-a homophilically-interacting extracellular domain and a juxtamembrane cytoplasmic domain-as well as a C-terminal cytoplasmic domain shared by all isoforms. The extent to which isoform-specific vs. shared domains regulate distinct γ-Pcdh functions remains incompletely understood. Our previous in vitro studies identified PKC phosphorylation of a serine residue within a shared C-terminal motif as a mechanism through which γ-Pcdh promotion of dendrite arborization via MARCKS is abrogated. Here, we used CRISPR/Cas9 genome editing to generate two new mouse lines expressing only non-phosphorylatable γ-Pcdhs, due either to a serine-to-alanine mutation (PcdhgS/A) or to a 15-amino acid C-terminal deletion resulting from insertion of an early stop codon (PcdhgCTD). Both lines are viable and fertile, and the density and maturation of dendritic spines remains unchanged in both PcdhgS/A and PcdhgCTD cortex. Dendrite arborization of cortical pyramidal neurons, however, is significantly increased in both lines, as are levels of active MARCKS. Intriguingly, despite having significantly reduced levels of γ-Pcdh proteins, the PcdhgCTD mutation yields the strongest phenotype, with even heterozygous mutants exhibiting increased arborization. The present study confirms that phosphorylation of a shared C-terminal motif is a key γ-Pcdh negative regulation point, and contributes to a converging understanding of γ-Pcdh family function in which distinct roles are played by both individual isoforms and discrete protein domains.

3.
Neuron ; 111(11): 1776-1794.e10, 2023 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-37028432

RESUMEN

Light touch sensation begins with activation of low-threshold mechanoreceptor (LTMR) endings in the skin and propagation of their signals to the spinal cord and brainstem. We found that the clustered protocadherin gamma (Pcdhg) gene locus, which encodes 22 cell-surface homophilic binding proteins, is required in somatosensory neurons for normal behavioral reactivity to a range of tactile stimuli. Developmentally, distinct Pcdhg isoforms mediate LTMR synapse formation through neuron-neuron interactions and peripheral axonal branching through neuron-glia interactions. The Pcdhgc3 isoform mediates homophilic interactions between sensory axons and spinal cord neurons to promote synapse formation in vivo and is sufficient to induce postsynaptic specializations in vitro. Moreover, loss of Pcdhgs and somatosensory synaptic inputs to the dorsal horn leads to fewer corticospinal synapses on dorsal horn neurons. These findings reveal essential roles for Pcdhg isoform diversity in somatosensory neuron synapse formation, peripheral axonal branching, and stepwise assembly of central mechanosensory circuitry.


Asunto(s)
Células Receptoras Sensoriales , Médula Espinal , Células Receptoras Sensoriales/fisiología , Médula Espinal/fisiología , Cadherinas/genética , Cadherinas/metabolismo , Sinapsis , Asta Dorsal de la Médula Espinal , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
4.
Elife ; 92020 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-32324136

RESUMEN

Germ cells are vulnerable to stress. Therefore, how organisms protect their future progeny from damage in a fluctuating environment is a fundamental question in biology. We show that in Caenorhabditis elegans, serotonin released by maternal neurons during stress ensures the viability and stress resilience of future offspring. Serotonin acts through a signal transduction pathway conserved between C. elegans and mammalian cells to enable the transcription factor HSF1 to alter chromatin in soon-to-be fertilized germ cells by recruiting the histone chaperone FACT, displacing histones, and initiating protective gene expression. Without serotonin release by maternal neurons, FACT is not recruited by HSF1 in germ cells, transcription occurs but is delayed, and progeny of stressed C. elegans mothers fail to complete development. These studies uncover a novel mechanism by which stress sensing by neurons is coupled to transcription response times of germ cells to protect future offspring.


Asunto(s)
Desarrollo Embrionario/fisiología , Células Germinativas/fisiología , Neuronas/fisiología , Serotonina/fisiología , Estrés Fisiológico/fisiología , Animales , Caenorhabditis elegans/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Proteínas de Unión al ADN/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción del Choque Térmico/fisiología , Proteínas del Grupo de Alta Movilidad/fisiología , Calor , Humanos , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/fisiología , Factores de Elongación Transcripcional/fisiología
5.
PLoS Genet ; 15(12): e1008554, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31877124

RESUMEN

The mammalian Pcdhg gene cluster encodes a family of 22 cell adhesion molecules, the gamma-Protocadherins (γ-Pcdhs), critical for neuronal survival and neural circuit formation. The extent to which isoform diversity-a γ-Pcdh hallmark-is required for their functions remains unclear. We used a CRISPR/Cas9 approach to reduce isoform diversity, targeting each Pcdhg variable exon with pooled sgRNAs to generate an allelic series of 26 mouse lines with 1 to 21 isoforms disrupted via discrete indels at guide sites and/or larger deletions/rearrangements. Analysis of 5 mutant lines indicates that postnatal viability and neuronal survival do not require isoform diversity. Surprisingly, given reports that it might not independently engage in trans-interactions, we find that γC4, encoded by Pcdhgc4, is the only critical isoform. Because the human orthologue is the only PCDHG gene constrained in humans, our results indicate a conserved γC4 function that likely involves distinct molecular mechanisms.


Asunto(s)
Empalme Alternativo , Cadherinas/genética , Mutación , Neuronas/metabolismo , Animales , Sistemas CRISPR-Cas , Proteínas Relacionadas con las Cadherinas , Cadherinas/metabolismo , Desarrollo Embrionario , Exones , Femenino , Humanos , Mutación INDEL , Masculino , Ratones , Familia de Multigenes , Neuronas/citología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Eliminación de Secuencia , Secuenciación Completa del Genoma
6.
Genesis ; 57(5): e23286, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30801883

RESUMEN

Evolutionarily conserved Akirin nuclear proteins interact with chromatin remodeling complexes at gene enhancers and promoters, and have been reported to regulate cell proliferation and differentiation. Of the two mouse Akirin genes, Akirin2 is essential during embryonic development, with known in vivo roles in immune system function and the formation of the cerebral cortex. Here we demonstrate that Akirin2 is critical for mouse myogenesis, a tightly regulated developmental process through which myoblast precursors fuse to form mature skeletal muscle fibers. Loss of Akirin2 in somitic muscle precursor cells via Sim1-Cre-mediated excision of a conditional Akirin2 allele results in neonatal lethality. Mutant embryos exhibit a complete lack of forelimb, intercostal, and diaphragm muscles due to extensive apoptosis and loss of Pax3-positive myoblasts. Severe skeletal defects, including craniofacial abnormalities, disrupted ossification, and rib fusions are also observed, attributable to lack of skeletal muscles as well as patchy Sim1-Cre activity in the embryonic sclerotome. We further show that Akirin2 levels are tightly regulated during muscle cell differentiation in vitro, and that Akirin2 is required for the proper expression of muscle differentiation factors myogenin and myosin heavy chain. Our results implicate Akirin2 as a major regulator of mammalian muscle formation in vivo.


Asunto(s)
Células Musculares/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Proliferación Celular , Embrión de Mamíferos , Desarrollo Embrionario , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Musculares/fisiología , Desarrollo de Músculos/fisiología , Músculo Esquelético/metabolismo , Músculos/metabolismo , Mioblastos/metabolismo , Cadenas Pesadas de Miosina/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Factores de Transcripción/genética
7.
Sci Rep ; 8(1): 12240, 2018 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-30116001

RESUMEN

The regulation of interdigital tissue regression requires the interplay of multiple spatiotemporally-controlled morphogen gradients to ensure proper limb formation and release of individual digits. Disruption to this process can lead to a number of limb abnormalities, including syndactyly. Akirins are highly conserved nuclear proteins that are known to interact with chromatin remodelling machinery at gene enhancers. In mammals, the analogue Akirin2 is essential for embryonic development and critical for a wide variety of roles in immune function, meiosis, myogenesis and brain development. Here we report a critical role for Akirin2 in the regulation of interdigital tissue regression in the mouse limb. Knockout of Akirin2 in limb epithelium leads to a loss of interdigital cell death and an increase in cell proliferation, resulting in retention of the interdigital web and soft-tissue syndactyly. This is associated with perdurance of Fgf8 expression in the ectoderm overlying the interdigital space. Our study supports a mechanism whereby Akirin2 is required for the downregulation of Fgf8 from the apical ectodermal ridge (AER) during limb development, and implies its requirement in signalling between interdigital mesenchymal cells and the AER.


Asunto(s)
Extremidades/crecimiento & desarrollo , Proteínas Represoras/metabolismo , Animales , Proliferación Celular , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Ratones , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Transducción de Señal
8.
Neural Dev ; 11(1): 21, 2016 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-27871306

RESUMEN

BACKGROUND: The proper spatial and temporal regulation of dorsal telencephalic progenitor behavior is a prerequisite for the formation of the highly-organized, six-layered cerebral cortex. Premature differentiation of cells, disruption of cell cycle timing, excessive apoptosis, and/or incorrect neuronal migration signals can have devastating effects, resulting in a number of neurodevelopmental disorders involving microcephaly and/or lissencephaly. Though genes encoding many key players in cortical development have been identified, our understanding remains incomplete. We show that the gene encoding Akirin2, a small nuclear protein, is expressed in the embryonic telencephalon. Converging evidence indicates that Akirin2 acts as a bridge between transcription factors (including Twist and NF-κB proteins) and the BAF (SWI/SNF) chromatin remodeling machinery to regulate patterns of gene expression. Constitutive knockout of Akirin2 is early embryonic lethal in mice, while restricted loss in B cells led to disrupted proliferation and cell survival. METHODS: We generated cortex-restricted Akirin2 knockouts by crossing mice harboring a floxed Akirin2 allele with the Emx1-Cre transgenic line and assessed the resulting embryos using in situ hybridization, EdU labeling, and immunohistochemistry. RESULTS: The vast majority of Akirin2 mutants do not survive past birth, and exhibit extreme microcephaly, with little dorsal telencephalic tissue and no recognizable cortex. This is primarily due to massive cell death of early cortical progenitors, which begins at embryonic day (E)10, shortly after Emx1-Cre is active. Immunostaining and cell cycle analysis using EdU labeling indicate that Akirin2-null progenitors fail to proliferate normally, produce fewer neurons, and undergo extensive apoptosis. All of the neurons that are generated in Akirin2 mutants also undergo apoptosis by E12. In situ hybridization for Wnt3a and Wnt-responsive genes suggest defective formation and/or function of the cortical hem in Akirin2 null mice. Furthermore, the apical ventricular surface becomes disrupted, and Sox2-positive progenitors are found to "spill" into the lateral ventricle. CONCLUSIONS: Our data demonstrate a previously-unsuspected role for Akirin2 in early cortical development and, given its known nuclear roles, suggest that it may act to regulate gene expression patterns critical for early progenitor cell behavior and cortical neuron production.


Asunto(s)
Corteza Cerebral/embriología , Regulación del Desarrollo de la Expresión Génica , Proteínas Represoras/metabolismo , Animales , Apoptosis , Ciclo Celular , Proliferación Celular , Corteza Cerebral/anomalías , Corteza Cerebral/metabolismo , Ratones , Ratones Noqueados , Microcefalia/genética , Células-Madre Neurales/fisiología , Proteínas Represoras/genética , Vía de Señalización Wnt
9.
Reprod Toxicol ; 16(6): 825-39, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12401512

RESUMEN

Neural crest cells (NCCs) exit the dorsal neural tube and migrate to sites where they form diverse tissues. Valproic acid (VPA) is an anticonvulsant drug that induces neural tube and related defects. Altered NCC migration and proliferation have been proposed as mechanisms of teratogenicity. We cultured neural tube segments from chick embryos in 0.75-3.0mM VPA. We used image analysis, proliferation assays, and fluorescence localization to investigate NCCs during VPA exposure. VPA inhibited attachment of explants and the number that produced migrating cells. VPA markedly decreased the proportion of cells migrating individually, promoting migration as epithelial sheets. VPA at 3mM decreased cellular spreading. Area and perimeter change per minute were reduced, but migration velocity was not. VPA at 2mM reduced proliferation 11% and 3mM arrested proliferation. Immunostaining of VPA-exposed explants revealed N-cadherin-positive cell boundaries within sheets, but independent NCCs did not stain. F-actin staining was reduced in independent NCCs. The data support a VPA mechanism involving interference with epithelial-mesenchymal transition.


Asunto(s)
Anticonvulsivantes/toxicidad , Movimiento Celular/efectos de los fármacos , Cresta Neural/efectos de los fármacos , Ácido Valproico/toxicidad , Animales , Bromodesoxiuridina/metabolismo , División Celular/efectos de los fármacos , Embrión de Pollo , Citoesqueleto/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Antagonismo de Drogas , Combinación de Medicamentos , Ácido Fólico/farmacología , Procesamiento de Imagen Asistido por Computador , Uniones Intercelulares/efectos de los fármacos , Microscopía Fluorescente , Microscopía por Video , Cresta Neural/citología , Cresta Neural/metabolismo , Técnicas de Cultivo de Órganos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...