Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Mol Syst Biol ; 16(12): e9819, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33289969

RESUMEN

Alzheimer's disease (AD) is characterized by the appearance of amyloid-ß plaques, neurofibrillary tangles, and inflammation in brain regions involved in memory. Using mass spectrometry, we have quantified the phosphoproteome of the CK-p25, 5XFAD, and Tau P301S mouse models of neurodegeneration. We identified a shared response involving Siglec-F which was upregulated on a subset of reactive microglia. The human paralog Siglec-8 was also upregulated on microglia in AD. Siglec-F and Siglec-8 were upregulated following microglial activation with interferon gamma (IFNγ) in BV-2 cell line and human stem cell-derived microglia models. Siglec-F overexpression activates an endocytic and pyroptotic inflammatory response in BV-2 cells, dependent on its sialic acid substrates and immunoreceptor tyrosine-based inhibition motif (ITIM) phosphorylation sites. Related human Siglecs induced a similar response in BV-2 cells. Collectively, our results point to an important role for mouse Siglec-F and human Siglec-8 in regulating microglial activation during neurodegeneration.


Asunto(s)
Inflamación/patología , Microglía/metabolismo , Degeneración Nerviosa/patología , Fosfoproteínas/metabolismo , Proteómica , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Secuencia de Aminoácidos , Animales , Anticuerpos/metabolismo , Muerte Celular , Línea Celular , Humanos , Inflamación/metabolismo , Interferón gamma/metabolismo , Ratones Transgénicos , Microglía/patología , Degeneración Nerviosa/metabolismo , Péptidos/metabolismo , Fagocitosis , Fosfotirosina/metabolismo , Proteoma/metabolismo , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/química , Transducción de Señal , Regulación hacia Arriba
2.
J Neurosci ; 37(41): 9917-9924, 2017 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-28912154

RESUMEN

Increased p25, a proteolytic fragment of the regulatory subunit p35, is known to induce aberrant activity of cyclin-dependent kinase 5 (Cdk5), which is associated with neurodegenerative disorders, including Alzheimer's disease. Previously, we showed that replacing endogenous p35 with the noncleavable mutant p35 (Δp35) attenuated amyloidosis and improved cognitive function in a familial Alzheimer's disease mouse model. Here, to address the role of p25/Cdk5 in tauopathy, we generated double-transgenic mice by crossing mice overexpressing mutant human tau (P301S) with Δp35KI mice. We observed significant reduction of phosphorylated tau and its seeding activity in the brain of double transgenic mice compared with the P301S mice. Furthermore, synaptic loss and impaired LTP at hippocampal CA3 region of P301S mice were attenuated by blocking p25 generation. To further validate the role of p25/Cdk5 in tauopathy, we used frontotemporal dementia patient-derived induced pluripotent stem cells (iPSCs) carrying the Tau P301L mutation and generated P301L:Δp35KI isogenic iPSC lines using CRISPR/Cas9 genome editing. We created cerebral organoids from the isogenic iPSCs and found that blockade of p25 generation reduced levels of phosphorylated tau and increased expression of synaptophysin. Together, these data demonstrate a crucial role for p25/Cdk5 in mediating tau-associated pathology and suggest that inhibition of this kinase can remedy neurodegenerative processes in the presence of pathogenic tau mutation.SIGNIFICANCE STATEMENT Accumulation of p25 results in aberrant Cdk5 activation and induction of numerous pathological phenotypes, such as neuroinflammation, synaptic loss, Aß accumulation, and tau hyperphosphorylation. However, it was not clear whether p25/Cdk5 activity is necessary for the progression of these pathological changes. We recently developed the Δp35KI transgenic mouse that is deficient in p25 generation and Cdk5 hyperactivation. In this study, we used this mouse model to elucidate the role of p25/Cdk5 in FTD mutant tau-mediated pathology. We also used a frontotemporal dementia patient-derived induced pluripotent stem cell carrying the Tau P301L mutation and generated isogenic lines in which p35 is replaced with noncleavable mutant Δp35. Our data suggest that p25/Cdk5 plays an important role in tauopathy in both mouse and human model systems.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/genética , Demencia Frontotemporal/genética , Fosfotransferasas/genética , Células Madre Pluripotentes , Tauopatías/genética , Animales , Región CA3 Hipocampal/patología , Región CA3 Hipocampal/fisiopatología , Quinasa 5 Dependiente de la Ciclina/antagonistas & inhibidores , Demencia Frontotemporal/prevención & control , Humanos , Potenciación a Largo Plazo/genética , Ratones , Ratones Transgénicos , Fibras Musgosas del Hipocampo/patología , Fosforilación , Fosfotransferasas/antagonistas & inhibidores , Trasplante de Células Madre , Sinapsis/patología , Sinaptofisina/genética , Tauopatías/prevención & control
3.
Nature ; 546(7658): 361-362, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28562586
4.
J Neurosci ; 34(8): 2860-70, 2014 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-24553927

RESUMEN

An increasing number of proteins involved in genome organization have been implicated in neurodevelopmental disorders, highlighting the importance of chromatin architecture in the developing CNS. The CCCTC-binding factor (CTCF) is a zinc finger DNA binding protein involved in higher-order chromatin organization, and mutations in the human CTCF gene cause an intellectual disability syndrome associated with microcephaly. However, information on CTCF function in vivo in the developing brain is lacking. To address this gap, we conditionally inactivated the Ctcf gene at early stages of mouse brain development. Cre-mediated Ctcf deletion in the telencephalon and anterior retina at embryonic day 8.5 triggered upregulation of the p53 effector PUMA (p53 upregulated modulator of apoptosis), resulting in massive apoptosis and profound ablation of telencephalic structures. Inactivation of Ctcf several days later at E11 also resulted in PUMA upregulation and increased apoptotic cell death, and the Ctcf-null forebrain was hypocellular and disorganized at birth. Although deletion of both Ctcf and Puma in the embryonic brain efficiently rescued Ctcf-null progenitor cell apoptosis, it failed to improve neonatal hypocellularity due to decreased proliferative capacity of rescued apical and outer radial glia progenitor cells. This was exacerbated by an independent effect of CTCF loss that resulted in depletion of the progenitor pool due to premature neurogenesis earlier in development. Our findings demonstrate that CTCF activities are required for two distinct events in early cortex formation: first, to correctly regulate the balance between neuroprogenitor cell proliferation and differentiation, and second, for the survival of neuroprogenitor cells, providing new clues regarding the contributions of CTCF in microcephaly/intellectual disability syndrome pathologies.


Asunto(s)
Diferenciación Celular/genética , Diferenciación Celular/fisiología , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Células-Madre Neurales/fisiología , Proteínas Represoras/fisiología , Animales , Antimetabolitos , Apoptosis/genética , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/fisiología , Encéfalo/anomalías , Bromodesoxiuridina , Factor de Unión a CCCTC , Muerte Celular/fisiología , Inmunoprecipitación de Cromatina , Exones/genética , Femenino , Técnica del Anticuerpo Fluorescente , Genes p53/genética , Genes p53/fisiología , Procesamiento de Imagen Asistido por Computador , Ratones , Ratones Noqueados , Nestina/genética , Nestina/fisiología , Embarazo , Cultivo Primario de Células , Reacción en Cadena en Tiempo Real de la Polimerasa , Retina/citología , Retina/fisiología , Telencéfalo/citología , Telencéfalo/fisiología , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/fisiología
5.
Hum Mol Genet ; 22(24): 5015-25, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23892236

RESUMEN

ATR-X syndrome is a rare genetic disorder caused by mutations in the ATRX gene. Affected individuals are cognitively impaired and display a variety of developmental abnormalities, including skeletal deformities. To investigate the function of ATRX during skeletal development, we selectively deleted the gene in the developing forelimb mesenchyme of mice. The absence of ATRX in the limb mesenchyme resulted in shorter digits, or brachydactyly, a defect also observed in a subset of ATR-X patients. This phenotype persisted until adulthood, causing reduced grip strength and altered gait in mutant mice. Examination of the embryonic ATRX-null forelimbs revealed a significant increase in apoptotic cell death, which could explain the reduced digit length. In addition, staining for the DNA damage markers γ-histone 2A family member X (γ-H2AX) and 53BP1 demonstrated a significant increase in the number of cells with DNA damage in the embryonic ATRX-null forepaw. Strikingly, only one large bright DNA damage event was observed per nucleus in proliferating cells. These large γ-H2AX foci were located in close proximity to the nuclear lamina and remained largely unresolved after cell differentiation. In addition, ATRX-depleted forelimb mesenchymal cells did not exhibit hypersensitivity to DNA fork-stalling compounds, suggesting that the nature as well as the response to DNA damage incurred by loss of ATRX in the developing limb fundamentally differs from other tissues. Our data suggest that DNA damage-induced apoptosis is a novel cellular mechanism underlying brachydactyly that might be relevant to additional skeletal syndromes.


Asunto(s)
Braquidactilia/genética , ADN Helicasas/genética , Miembro Anterior/anomalías , Mesodermo/metabolismo , Proteínas Nucleares/genética , Animales , Braquidactilia/metabolismo , Muerte Celular/genética , Condrocitos/metabolismo , ADN Helicasas/deficiencia , ADN Helicasas/metabolismo , Modelos Animales de Enfermedad , Femenino , Miembro Anterior/embriología , Miembro Anterior/fisiopatología , Estudios de Asociación Genética , Histonas/genética , Histonas/metabolismo , Hidroxiurea/farmacología , Esbozos de los Miembros/embriología , Esbozos de los Miembros/metabolismo , Masculino , Mesodermo/efectos de los fármacos , Ratones , Ratones Noqueados , Proteínas Nucleares/deficiencia , Proteínas Nucleares/metabolismo , Fenotipo , Proteína Nuclear Ligada al Cromosoma X
6.
Front Genet ; 14: 1198129, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37323671

RESUMEN

Down syndrome (DS), caused by triplication of chromosome 21, is the most frequent aneuploidy observed in the human population and represents the most common genetic form of intellectual disability and early-onset Alzheimer's disease (AD). Individuals with DS exhibit a wide spectrum of clinical presentation, with a number of organs implicated including the neurological, immune, musculoskeletal, cardiac, and gastrointestinal systems. Decades of DS research have illuminated our understanding of the disorder, however many of the features that limit quality of life and independence of individuals with DS, including intellectual disability and early-onset dementia, remain poorly understood. This lack of knowledge of the cellular and molecular mechanisms leading to neurological features of DS has caused significant roadblocks in developing effective therapeutic strategies to improve quality of life for individuals with DS. Recent technological advances in human stem cell culture methods, genome editing approaches, and single-cell transcriptomics have provided paradigm-shifting insights into complex neurological diseases such as DS. Here, we review novel neurological disease modeling approaches, how they have been used to study DS, and what questions might be addressed in the future using these innovative tools.

7.
Cell Stem Cell ; 29(1): 116-130.e7, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34995493

RESUMEN

Down syndrome (DS) is a genetic disorder driven by the triplication of chromosome 21 (T21) and characterized by a wide range of neurodevelopmental and physical disabilities. Transcriptomic analysis of tissue samples from individuals with DS has revealed that T21 induces a genome-wide transcriptional disruption. However, the consequences of T21 on the nuclear architecture and its interplay with the transcriptome remain unknown. In this study, we find that unlike human induced pluripotent stem cells (iPSCs), iPSC-derived neural progenitor cells (NPCs) exhibit genome-wide "chromosomal introversion," disruption of lamina-associated domains, and global chromatin accessibility changes in response to T21, consistent with the transcriptional and nuclear architecture changes characteristic of senescent cells. Treatment of T21-harboring NPCs with senolytic drugs alleviates the transcriptional, molecular, and cellular dysfunctions associated with DS. Our findings provide a mechanistic link between T21 and global transcriptional disruption and indicate that senescence-associated phenotypes may play a key role in the neurodevelopmental pathogenesis of DS.


Asunto(s)
Síndrome de Down , Células Madre Pluripotentes Inducidas , Células-Madre Neurales , Perfilación de la Expresión Génica , Humanos , Transcriptoma/genética
8.
Nat Commun ; 11(1): 2484, 2020 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-32424276

RESUMEN

DNA damage contributes to brain aging and neurodegenerative diseases. However, the factors stimulating DNA repair to stave off functional decline remain obscure. We show that HDAC1 modulates OGG1-initated 8-oxoguanine (8-oxoG) repair in the brain. HDAC1-deficient mice display age-associated DNA damage accumulation and cognitive impairment. HDAC1 stimulates OGG1, a DNA glycosylase known to remove 8-oxoG lesions that are associated with transcriptional repression. HDAC1 deficiency causes impaired OGG1 activity, 8-oxoG accumulation at the promoters of genes critical for brain function, and transcriptional repression. Moreover, we observe elevated 8-oxoG along with reduced HDAC1 activity and downregulation of a similar gene set in the 5XFAD mouse model of Alzheimer's disease. Notably, pharmacological activation of HDAC1 alleviates the deleterious effects of 8-oxoG in aged wild-type and 5XFAD mice. Our work uncovers important roles for HDAC1 in 8-oxoG repair and highlights the therapeutic potential of HDAC1 activation to counter functional decline in brain aging and neurodegeneration.


Asunto(s)
Envejecimiento/patología , Enfermedad de Alzheimer/patología , Encéfalo/patología , Daño del ADN , ADN Glicosilasas/metabolismo , Histona Desacetilasa 1/metabolismo , Estrés Oxidativo , Acetilación , Envejecimiento/genética , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/fisiopatología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Secuencia de Bases , Benzofenonas/farmacología , Cognición/efectos de los fármacos , Trastornos del Conocimiento/complicaciones , Trastornos del Conocimiento/patología , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Ontología de Genes , Guanina/análogos & derivados , Guanina/metabolismo , Memoria/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Regiones Promotoras Genéticas/genética
9.
Curr Opin Neurobiol ; 43: 48-55, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28024185

RESUMEN

Different aspects of learning, memory, and cognition are regulated by epigenetic mechanisms such as covalent DNA modifications and histone post-translational modifications. More recently, the modulation of chromatin architecture and nuclear organization is emerging as a key factor in dynamic transcriptional regulation of the post-mitotic neuron. For instance, neuronal activity induces relocalization of gene loci to 'transcription factories', and specific enhancer-promoter looping contacts allow for precise transcriptional regulation. Moreover, neuronal activity-dependent DNA double-strand break formation in the promoter of immediate early genes appears to overcome topological constraints on transcription. Together, these findings point to a critical role for genome topology in integrating dynamic environmental signals to define precise spatiotemporal gene expression programs supporting cognitive processes.


Asunto(s)
Cromatina/química , Epigénesis Genética , Genoma , Aprendizaje/fisiología , Memoria/fisiología , Histonas/genética , Humanos
10.
Nat Neurosci ; 19(11): 1477-1488, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27694995

RESUMEN

De novo mutations in CHD8 are strongly associated with autism spectrum disorder, but the basic biology of CHD8 remains poorly understood. Here we report that Chd8 knockdown during cortical development results in defective neural progenitor proliferation and differentiation that ultimately manifests in abnormal neuronal morphology and behaviors in adult mice. Transcriptome analysis revealed that while Chd8 stimulates the transcription of cell cycle genes, it also precludes the induction of neural-specific genes by regulating the expression of PRC2 complex components. Furthermore, knockdown of Chd8 disrupts the expression of key transducers of Wnt signaling, and enhancing Wnt signaling rescues the transcriptional and behavioral deficits caused by Chd8 knockdown. We propose that these roles of Chd8 and the dynamics of Chd8 expression during development help negotiate the fine balance between neural progenitor proliferation and differentiation. Together, these observations provide new insights into the neurodevelopmental role of Chd8.


Asunto(s)
Trastorno del Espectro Autista/genética , Ciclo Celular/genética , Diferenciación Celular/genética , Proteínas de Unión al ADN/genética , Regulación del Desarrollo de la Expresión Génica/genética , Neurogénesis , Transcripción Genética , Vía de Señalización Wnt/genética , Animales , División Celular/genética , Femenino , Ratones , Células-Madre Neurales/metabolismo
11.
Epigenomics ; 7(8): 1365-78, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26646632

RESUMEN

ATRX was identified over 20 years ago as the gene responsible for a rare developmental disorder characterized by α-thalassemia and intellectual disability. Similarities to the sucrose nonfermentable SNF2 type chromatin remodelers initially suggested a role in transcriptional regulation. However, over the last years, our knowledge of the epigenetic activities of ATRX has expanded steadily. Recent exciting discoveries have propelled ATRX into the limelight of chromatin and telomere biology, development and cancer research. This review summarizes recent breakthroughs in understanding ATRX function in heterochromatin structure, genome stability and its frequent dysregulation in a variety of cancers.


Asunto(s)
ADN Helicasas/genética , ADN Helicasas/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Ensamble y Desensamble de Cromatina , Proteínas Co-Represoras , G-Cuádruplex , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Inestabilidad Genómica , Heterocromatina/genética , Heterocromatina/metabolismo , Humanos , Chaperonas Moleculares , Complejos Multiproteicos/metabolismo , Mutación , Neoplasias/diagnóstico , Unión Proteica , Homeostasis del Telómero , Proteína Nuclear Ligada al Cromosoma X
12.
Biol Open ; 3(12): 1158-63, 2014 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-25395668

RESUMEN

Mutations in the alpha-thalassemia mental retardation X-linked (ATRX) gene cause a spectrum of abnormalities including intellectual disability, developmental delay, seizures, and microcephaly. The ATRX protein is highly enriched at heterochromatic repetitive sequences adjacent to the centromere, and ATRX depletion results in chromosome congression, segregation, and cohesion defects. Here, we show that Cre-mediated inactivation of Atrx in the embryonic mouse (Mus musculus) brain results in expansion of cerebral cortical layer VI, and a concurrent thinning of layers II-IV. We observed increased cell cycle exit during early-mid neurogenesis, and a depletion of apical progenitors by late neurogenesis in the Atrx-null neocortex, explaining the disproportionate layering. Premature differentiation was associated with an increased generation of outer radial glia (oRG) and TBR2-expressing basal progenitors, as well as increased generation of early-born post-mitotic projection neurons. Atrx deletion also reduced the fidelity of mitotic spindle orientation in apical progenitors, where mutant cells were often oriented at non-parallel angles of division relative to the ventricular surface. We conclude that ATRX is required for correct lamination of the mouse neocortex by regulating the timing of neuroprogenitor cell differentiation.

13.
J Clin Invest ; 123(5): 2049-63, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23563309

RESUMEN

Human ATRX mutations are associated with cognitive deficits, developmental abnormalities, and cancer. We show that the Atrx-null embryonic mouse brain accumulates replicative damage at telomeres and pericentromeric heterochromatin, which is exacerbated by loss of p53 and linked to ATM activation. ATRX-deficient neuroprogenitors exhibited higher incidence of telomere fusions and increased sensitivity to replication stress-inducing drugs. Treatment of Atrx-null neuroprogenitors with the G-quadruplex (G4) ligand telomestatin increased DNA damage, indicating that ATRX likely aids in the replication of telomeric G4-DNA structures. Unexpectedly, mutant mice displayed reduced growth, shortened life span, lordokyphosis, cataracts, heart enlargement, and hypoglycemia, as well as reduction of mineral bone density, trabecular bone content, and subcutaneous fat. We show that a subset of these defects can be attributed to loss of ATRX in the embryonic anterior pituitary that resulted in low circulating levels of thyroxine and IGF-1. Our findings suggest that loss of ATRX increases DNA damage locally in the forebrain and anterior pituitary and causes tissue attrition and other systemic defects similar to those seen in aging.


Asunto(s)
Encéfalo/metabolismo , ADN Helicasas/genética , ADN Helicasas/fisiología , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Telómero/ultraestructura , Envejecimiento , Animales , Huesos/metabolismo , Trastornos del Conocimiento/genética , Daño del ADN , ADN Helicasas/deficiencia , Femenino , Fibroblastos/citología , G-Cuádruplex , Genotipo , Heterocromatina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ligandos , Longevidad , Masculino , Ratones , Ratones Transgénicos , Microscopía Fluorescente/métodos , Proteínas Nucleares/deficiencia , Oxazoles/farmacología , Fenotipo , Células Madre/citología , Tiroxina/metabolismo , Proteína Nuclear Ligada al Cromosoma X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA