Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Cell ; 155(2): 269-71, 2013 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-24120127

RESUMEN

Environmental and cellular cues pattern dendritic growth and direct dendrites to their targets. However, little is known about the signals regulating interactions with the surrounding substrate. Dong et al. and Salzberg et al. now identify a tripartite ligand-receptor complex that conveys cues from the substrate necessary for the patterning of complex dendrites in C. elegans.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Dendritas/metabolismo , Proteínas de la Membrana/metabolismo , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Neurogénesis , Neuronas/metabolismo , Animales
2.
Nature ; 604(7907): 714-722, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35444284

RESUMEN

Dementia in Alzheimer's disease progresses alongside neurodegeneration1-4, but the specific events that cause neuronal dysfunction and death remain poorly understood. During normal ageing, neurons progressively accumulate somatic mutations5 at rates similar to those of dividing cells6,7 which suggests that genetic factors, environmental exposures or disease states might influence this accumulation5. Here we analysed single-cell whole-genome sequencing data from 319 neurons from the prefrontal cortex and hippocampus of individuals with Alzheimer's disease and neurotypical control individuals. We found that somatic DNA alterations increase in individuals with Alzheimer's disease, with distinct molecular patterns. Normal neurons accumulate mutations primarily in an age-related pattern (signature A), which closely resembles 'clock-like' mutational signatures that have been previously described in healthy and cancerous cells6-10. In neurons affected by Alzheimer's disease, additional DNA alterations are driven by distinct processes (signature C) that highlight C>A and other specific nucleotide changes. These changes potentially implicate nucleotide oxidation4,11, which we show is increased in Alzheimer's-disease-affected neurons in situ. Expressed genes exhibit signature-specific damage, and mutations show a transcriptional strand bias, which suggests that transcription-coupled nucleotide excision repair has a role in the generation of mutations. The alterations in Alzheimer's disease affect coding exons and are predicted to create dysfunctional genetic knockout cells and proteostatic stress. Our results suggest that known pathogenic mechanisms in Alzheimer's disease may lead to genomic damage to neurons that can progressively impair function. The aberrant accumulation of DNA alterations in neurodegeneration provides insight into the cascade of molecular and cellular events that occurs in the development of Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer , Neuronas , Envejecimiento , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , ADN , Exones , Genómica , Hipocampo/citología , Humanos , Tasa de Mutación , Neuronas/patología , Nucleótidos , Corteza Prefrontal/citología , Secuenciación Completa del Genoma
3.
Nature ; 453(7197): 935-9, 2008 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-18432193

RESUMEN

The cellular machinery promoting phagocytosis of corpses of apoptotic cells is well conserved from worms to mammals. An important component is the Caenorhabditis elegans engulfment receptor CED-1 (ref. 1) and its Drosophila orthologue, Draper. The CED-1/Draper signalling pathway is also essential for the phagocytosis of other types of 'modified self' including necrotic cells, developmentally pruned axons and dendrites, and axons undergoing Wallerian degeneration. Here we show that Drosophila Shark, a non-receptor tyrosine kinase similar to mammalian Syk and Zap-70, binds Draper through an immunoreceptor tyrosine-based activation motif (ITAM) in the Draper intracellular domain. We show that Shark activity is essential for Draper-mediated signalling events in vivo, including the recruitment of glial membranes to severed axons and the phagocytosis of axonal debris and neuronal cell corpses by glia. We also show that the Src family kinase (SFK) Src42A can markedly increase Draper phosphorylation and is essential for glial phagocytic activity. We propose that ligand-dependent Draper receptor activation initiates the Src42A-dependent tyrosine phosphorylation of Draper, the association of Shark and the activation of the Draper pathway. These Draper-Src42A-Shark interactions are strikingly similar to mammalian immunoreceptor-SFK-Syk signalling events in mammalian myeloid and lymphoid cells. Thus, Draper seems to be an ancient immunoreceptor with an extracellular domain tuned to modified self, and an intracellular domain promoting phagocytosis through an ITAM-domain-SFK-Syk-mediated signalling cascade.


Asunto(s)
Proteínas de Drosophila/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Neuroglía/citología , Fagocitosis , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Transducción de Señal , Secuencias de Aminoácidos , Animales , Axones/metabolismo , Axones/patología , Línea Celular , Membrana Celular/metabolismo , Sistema Nervioso Central , Proteínas de Drosophila/química , Proteínas de la Membrana/química , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Quinasa Syk , Técnicas del Sistema de Dos Híbridos
4.
bioRxiv ; 2023 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-37986960

RESUMEN

Aging brings dysregulation of various processes across organs and tissues, often stemming from stochastic damage to individual cells over time. Here, we used a combination of single-nucleus RNA-sequencing and single-cell whole-genome sequencing to identify transcriptomic and genomic changes in the prefrontal cortex of the human brain across life span, from infancy to centenarian. We identified infant-specific cell clusters enriched for the expression of neurodevelopmental genes, and a common down-regulation of cell-essential homeostatic genes that function in ribosomes, transport, and metabolism during aging across cell types. Conversely, expression of neuron-specific genes generally remains stable throughout life. We observed a decrease in specific DNA repair genes in aging, including genes implicated in generating brain somatic mutations as indicated by mutation signature analysis. Furthermore, we detected gene-length-specific somatic mutation rates that shape the transcriptomic landscape of the aged human brain. These findings elucidate critical aspects of human brain aging, shedding light on transcriptomic and genomics dynamics.

5.
Front Aging ; 3: 991460, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36313183

RESUMEN

Maintaining genomic integrity in post-mitotic neurons in the human brain is paramount because these cells must survive for an individual's entire lifespan. Due to life-long synaptic plasticity and electrochemical transmission between cells, the brain engages in an exceptionally high level of mitochondrial metabolic activity. This activity results in the generation of reactive oxygen species with 8-oxo-7,8-dihydroguanine (8-oxoG) being one of the most prevalent oxidation products in the cell. 8-oxoG is important for the maintenance and transfer of genetic information into proper gene expression: a low basal level of 8-oxoG plays an important role in epigenetic modulation of neurodevelopment and synaptic plasticity, while a dysregulated increase in 8-oxoG damages the genome leading to somatic mutations and transcription errors. The slow yet persistent accumulation of DNA damage in the background of increasing cellular 8-oxoG is associated with normal aging as well as neurological disorders such as Alzheimer's disease and Parkinson's disease. This review explores the current understanding of how 8-oxoG plays a role in brain function and genomic instability, highlighting new methods being used to advance pathological hallmarks that differentiate normal healthy aging and neurodegenerative disease.

6.
J Cell Biol ; 216(3): 595-605, 2017 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-28223318

RESUMEN

Fragile X syndrome, the most common known monogenic cause of autism, results from the loss of FMR1, a conserved, ubiquitously expressed RNA-binding protein. Recent evidence suggests that Fragile X syndrome and other types of autism are associated with immune system defects. We found that Drosophila melanogaster Fmr1 mutants exhibit increased sensitivity to bacterial infection and decreased phagocytosis of bacteria by systemic immune cells. Using tissue-specific RNAi-mediated knockdown, we showed that Fmr1 plays a cell-autonomous role in the phagocytosis of bacteria. Fmr1 mutants also exhibit delays in two processes that require phagocytosis by glial cells, the immune cells in the brain: neuronal clearance after injury in adults and the development of the mushroom body, a brain structure required for learning and memory. Delayed neuronal clearance is associated with reduced recruitment of activated glia to the site of injury. These results suggest a previously unrecognized role for Fmr1 in regulating the activation of phagocytic immune cells both in the body and the brain.


Asunto(s)
Drosophila melanogaster/inmunología , Síndrome del Cromosoma X Frágil/inmunología , Inmunidad Innata/inmunología , Fagocitosis/inmunología , Animales , Encéfalo/inmunología , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Proteínas de Drosophila/inmunología , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/metabolismo , Aprendizaje/fisiología , Masculino , Memoria/fisiología , Cuerpos Pedunculados/inmunología , Cuerpos Pedunculados/metabolismo , Neuroglía/inmunología , Neuroglía/metabolismo , Neuronas/inmunología , Neuronas/metabolismo , Interferencia de ARN/inmunología , Proteínas de Unión al ARN/inmunología , Proteínas de Unión al ARN/metabolismo
7.
Nat Neurosci ; 15(7): 979-87, 2012 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-22706267

RESUMEN

Glial cells efficiently recognize and clear cellular debris after nervous system injury to maintain brain homeostasis, but pathways governing glial responses to neural injury remain poorly defined. We identify the Drosophila melanogaster guanine nucleotide exchange factor complex Crk/Mbc/dCed-12 and the small GTPase Rac1 as modulators of glial clearance of axonal debris. We found that Crk/Mbc/dCed-12 and Rac1 functioned in a non-redundant fashion with the Draper transmembrane receptor pathway: loss of either pathway fully suppressed clearance of axonal debris. Draper signaling was required early during glial responses, promoting glial activation, which included increased Draper and dCed-6 expression and extension of glial membranes to degenerating axons. In contrast, the Crk/Mbc/dCed-12 complex functioned at later phases, promoting glial phagocytosis of axonal debris. Our work identifies new components of the glial engulfment machinery and shows that glial activation, phagocytosis of axonal debris and termination of responses to injury are genetically separable events mediated by distinct signaling pathways.


Asunto(s)
Axones , Neuroglía/metabolismo , Neuronas Receptoras Olfatorias/metabolismo , Fagocitosis , Animales , Axones/metabolismo , Axones/patología , Axones/fisiología , Axotomía , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Drosophila melanogaster/fisiología , Técnicas de Inactivación de Genes , Vías Nerviosas/metabolismo , Vías Nerviosas/patología , Vías Nerviosas/fisiología , Neuroglía/patología , Neuroglía/fisiología , Neuronas Receptoras Olfatorias/patología , Fagocitosis/genética
8.
Science ; 337(6093): 481-4, 2012 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-22678360

RESUMEN

Axonal and synaptic degeneration is a hallmark of peripheral neuropathy, brain injury, and neurodegenerative disease. Axonal degeneration has been proposed to be mediated by an active autodestruction program, akin to apoptotic cell death; however, loss-of-function mutations capable of potently blocking axon self-destruction have not been described. Here, we show that loss of the Drosophila Toll receptor adaptor dSarm (sterile α/Armadillo/Toll-Interleukin receptor homology domain protein) cell-autonomously suppresses Wallerian degeneration for weeks after axotomy. Severed mouse Sarm1 null axons exhibit remarkable long-term survival both in vivo and in vitro, indicating that Sarm1 prodegenerative signaling is conserved in mammals. Our results provide direct evidence that axons actively promote their own destruction after injury and identify dSarm/Sarm1 as a member of an ancient axon death signaling pathway.


Asunto(s)
Proteínas del Dominio Armadillo/genética , Proteínas del Dominio Armadillo/fisiología , Axones/fisiología , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/fisiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Neuronas/fisiología , Degeneración Walleriana , Animales , Animales Modificados Genéticamente , Apoptosis , Proteínas del Dominio Armadillo/análisis , Axones/ultraestructura , Axotomía , Supervivencia Celular , Células Cultivadas , Proteínas del Citoesqueleto/análisis , Desnervación , Drosophila/embriología , Drosophila/genética , Drosophila/fisiología , Proteínas de Drosophila/análisis , Ratones , Mutación , Nervio Ciático/lesiones , Nervio Ciático/fisiología , Transducción de Señal , Ganglio Cervical Superior/citología , Técnicas de Cultivo de Tejidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA