Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 1584, 2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38383565

RESUMEN

Astrocytes, a type of glial cell in the central nervous system (CNS), adopt diverse states in response to injury that are influenced by their location relative to the insult. Here, we describe a platform for spatially resolved, single-cell transcriptomics and proteomics, called tDISCO (tissue-digital microfluidic isolation of single cells for -Omics). We use tDISCO alongside two high-throughput platforms for spatial (Visium) and single-cell transcriptomics (10X Chromium) to examine the heterogeneity of the astrocyte response to a cortical ischemic stroke in male mice. We show that integration of Visium and 10X Chromium datasets infers two astrocyte populations, proximal or distal to the injury site, while tDISCO determines the spatial boundaries and molecular profiles that define these populations. We find that proximal astrocytes show differences in lipid shuttling, with enriched expression of Apoe and Fabp5. Our datasets provide a resource for understanding the roles of astrocytes in stroke and showcase the utility of tDISCO for hypothesis-driven, spatially resolved single-cell experiments.


Asunto(s)
Astrocitos , Accidente Cerebrovascular , Ratones , Animales , Masculino , Astrocitos/metabolismo , Sistema Nervioso Central/metabolismo , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/metabolismo , Perfilación de la Expresión Génica , Cromo/metabolismo
2.
Stem Cell Rev Rep ; 19(4): 983-1000, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36617597

RESUMEN

The mammalian adult brain contains two neural stem and precursor (NPC) niches: the subventricular zone [SVZ] lining the lateral ventricles and the subgranular zone [SGZ] in the hippocampus. From these, SVZ NPCs represent the largest NPC pool. While SGZ NPCs typically only produce neurons and astrocytes, SVZ NPCs produce neurons, astrocytes and oligodendrocytes throughout life. Of particular importance is the generation and replacement of oligodendrocytes, the only myelinating cells of the central nervous system (CNS). SVZ NPCs contribute to myelination by regenerating the parenchymal oligodendrocyte precursor cell (OPC) pool and by differentiating into oligodendrocytes in the developing and demyelinated brain. The neurosphere assay has been widely adopted by the scientific community to facilitate the study of NPCs in vitro. Here, we present a streamlined protocol for culturing postnatal and adult SVZ NPCs and OPCs from primary neurosphere cells. We characterize the purity and differentiation potential as well as provide RNA-sequencing profiles of postnatal SVZ NPCs, postnatal SVZ OPCs and adult SVZ NPCs. We show that primary neurospheres cells generated from postnatal and adult SVZ differentiate into neurons, astrocytes and oligodendrocytes concurrently and at comparable levels. SVZ OPCs are generated by subjecting primary neurosphere cells to OPC growth factors fibroblast growth factor (FGF) and platelet-derived growth factor-AA (PDGF-AA). We further show SVZ OPCs can differentiate into oligodendrocytes in the absence and presence of thyroid hormone T3. Transcriptomic analysis confirmed the identities of each cell population and revealed novel immune and signalling pathways expressed in an age and cell type specific manner.


Asunto(s)
Ventrículos Laterales , Transcriptoma , Ratones , Animales , Transcriptoma/genética , Encéfalo , Neuronas , Diferenciación Celular/genética , Factores de Crecimiento de Fibroblastos , Mamíferos
3.
Cell Rep ; 33(2): 108257, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-33053360

RESUMEN

Here, we ask how neural stem cells (NSCs) transition in the developing neocortex from a rapidly to a slowly proliferating state, a process required to maintain lifelong stem cell pools. We identify LRIG1, known to regulate receptor tyrosine kinase signaling in other cell types, as a negative regulator of cortical NSC proliferation. LRIG1 is expressed in murine cortical NSCs as they start to proliferate more slowly during embryogenesis and then peaks postnatally when they transition to give rise to a portion of adult NSCs. Constitutive or acute loss of Lrig1 in NSCs over this developmental time frame causes stem cell expansion due to increased proliferation. LRIG1 controls NSC proliferation by associating with and negatively regulating the epidermal growth factor receptor (EGFR). These data support a model in which LRIG1 dampens the stem cell response to EGFR ligands within the cortical environment to slow their proliferation as they transition to postnatal adult NSCs.


Asunto(s)
Receptores ErbB/metabolismo , Glicoproteínas de Membrana/metabolismo , Neocórtex/citología , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Transducción de Señal , Animales , Animales Recién Nacidos , Proliferación Celular , Autorrenovación de las Células , Embrión de Mamíferos/citología , Desarrollo Embrionario , Ratones , Ratones Noqueados , Neurogénesis
4.
eNeuro ; 7(3)2020.
Artículo en Inglés | MEDLINE | ID: mdl-32349983

RESUMEN

Peripheral nerves provide a supportive growth environment for developing and regenerating axons and are essential for maintenance and repair of many non-neural tissues. This capacity has largely been ascribed to paracrine factors secreted by nerve-resident Schwann cells. Here, we used single-cell transcriptional profiling to identify ligands made by different injured rodent nerve cell types and have combined this with cell-surface mass spectrometry to computationally model potential paracrine interactions with peripheral neurons. These analyses show that peripheral nerves make many ligands predicted to act on peripheral and CNS neurons, including known and previously uncharacterized ligands. While Schwann cells are an important ligand source within injured nerves, more than half of the predicted ligands are made by nerve-resident mesenchymal cells, including the endoneurial cells most closely associated with peripheral axons. At least three of these mesenchymal ligands, ANGPT1, CCL11, and VEGFC, promote growth when locally applied on sympathetic axons. These data therefore identify an unexpected paracrine role for nerve mesenchymal cells and suggest that multiple cell types contribute to creating a highly pro-growth environment for peripheral axons.


Asunto(s)
Regeneración Nerviosa , Análisis de la Célula Individual , Axones , Ligandos , Nervios Periféricos , Células de Schwann
5.
Dev Cell ; 52(4): 509-524.e9, 2020 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-31902657

RESUMEN

Here, we investigate the origin and nature of blastema cells that regenerate the adult murine digit tip. We show that Pdgfra-expressing mesenchymal cells in uninjured digits establish the regenerative blastema and are essential for regeneration. Single-cell profiling shows that the mesenchymal blastema cells are distinct from both uninjured digit and embryonic limb or digit Pdgfra-positive cells. This unique blastema state is environmentally determined; dermal fibroblasts transplanted into the regenerative, but not non-regenerative, digit express blastema-state genes and contribute to bone regeneration. Moreover, lineage tracing with single-cell profiling indicates that endogenous osteoblasts or osteocytes acquire a blastema mesenchymal transcriptional state and contribute to both dermis and bone regeneration. Thus, mammalian digit tip regeneration occurs via a distinct adult mechanism where the regenerative environment promotes acquisition of a blastema state that enables cells from tissues such as bone to contribute to the regeneration of other mesenchymal tissues such as the dermis.


Asunto(s)
Diferenciación Celular , Extremidades/fisiología , Regulación del Desarrollo de la Expresión Génica , Células Madre Mesenquimatosas/citología , Receptores del Factor de Crecimiento Derivado de Plaquetas/fisiología , Regeneración , Animales , Linaje de la Célula , Células Cultivadas , Extremidades/embriología , Extremidades/lesiones , Femenino , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Análisis de la Célula Individual , Transcriptoma
6.
Cell Stem Cell ; 24(2): 240-256.e9, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30503141

RESUMEN

Peripheral innervation plays an important role in regulating tissue repair and regeneration. Here we provide evidence that injured peripheral nerves provide a reservoir of mesenchymal precursor cells that can directly contribute to murine digit tip regeneration and skin repair. In particular, using single-cell RNA sequencing and lineage tracing, we identify transcriptionally distinct mesenchymal cell populations within the control and injured adult nerve, including neural crest-derived cells in the endoneurium with characteristics of mesenchymal precursor cells. Culture and transplantation studies show that these nerve-derived mesenchymal cells have the potential to differentiate into non-nerve lineages. Moreover, following digit tip amputation, neural crest-derived nerve mesenchymal cells contribute to the regenerative blastema and, ultimately, to the regenerated bone. Similarly, neural crest-derived nerve mesenchymal cells contribute to the dermis during skin wound healing. These findings support a model where peripheral nerves directly contribute mesenchymal precursor cells to promote repair and regeneration of injured mammalian tissues.


Asunto(s)
Células Madre Mesenquimatosas/citología , Regeneración Nerviosa/fisiología , Tejido Nervioso/patología , Cicatrización de Heridas , Animales , Regeneración Ósea , Diferenciación Celular , Linaje de la Célula , Ratones , Cresta Neural/citología , Osteogénesis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células de Schwann/patología , Nervio Ciático/lesiones , Nervio Ciático/patología , Transcripción Genética , Transcriptoma/genética
7.
Neuron ; 97(3): 520-537.e6, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29395907

RESUMEN

The mechanisms instructing genesis of neuronal subtypes from mammalian neural precursors are not well understood. To address this issue, we have characterized the transcriptional landscape of radial glial precursors (RPs) in the embryonic murine cortex. We show that individual RPs express mRNA, but not protein, for transcriptional specifiers of both deep and superficial layer cortical neurons. Some of these mRNAs, including the superficial versus deep layer neuron transcriptional regulators Brn1 and Tle4, are translationally repressed by their association with the RNA-binding protein Pumilio2 (Pum2) and the 4E-T protein. Disruption of these repressive complexes in RPs mid-neurogenesis by knocking down 4E-T or Pum2 causes aberrant co-expression of deep layer neuron specification proteins in newborn superficial layer neurons. Thus, cortical RPs are transcriptionally primed to generate diverse types of neurons, and a Pum2/4E-T complex represses translation of some of these neuronal identity mRNAs to ensure appropriate temporal specification of daughter neurons.


Asunto(s)
Corteza Cerebral/embriología , Células Ependimogliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células-Madre Neurales/metabolismo , Neurogénesis , Animales , Corteza Cerebral/metabolismo , Factor 4E Eucariótico de Iniciación/metabolismo , Femenino , Masculino , Ratones , Proteínas del Tejido Nervioso/metabolismo , Factores del Dominio POU/metabolismo , Cultivo Primario de Células , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/metabolismo , Análisis de Secuencia de ARN
8.
Cell Rep ; 21(13): 3970-3986, 2017 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-29281841

RESUMEN

Adult neural stem cells (NSCs) derive from embryonic precursors, but little is known about how or when this occurs. We have addressed this issue using single-cell RNA sequencing at multiple developmental time points to analyze the embryonic murine cortex, one source of adult forebrain NSCs. We computationally identify all major cortical cell types, including the embryonic radial precursors (RPs) that generate adult NSCs. We define the initial emergence of RPs from neuroepithelial stem cells at E11.5. We show that, by E13.5, RPs express a transcriptional identity that is maintained and reinforced throughout their transition to a non-proliferative state between E15.5 and E17.5. These slowly proliferating late embryonic RPs share a core transcriptional phenotype with quiescent adult forebrain NSCs. Together, these findings support a model wherein cortical RPs maintain a core transcriptional identity from embryogenesis through to adulthood and wherein the transition to a quiescent adult NSC occurs during late neurogenesis.


Asunto(s)
Células Madre Adultas/metabolismo , Perfilación de la Expresión Génica , Células-Madre Neurales/metabolismo , Análisis de la Célula Individual , Animales , Proliferación Celular/genética , Corteza Cerebral/citología , Embrión de Mamíferos/citología , Desarrollo Embrionario/genética , Ratones Endogámicos C57BL , Neurogénesis/genética , Neuronas/citología , Neuronas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcriptoma/genética
9.
Neuron ; 94(3): 500-516.e9, 2017 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-28472653

RESUMEN

During development, newborn interneurons migrate throughout the embryonic brain. Here, we provide evidence that these interneurons act in a paracrine fashion to regulate developmental oligodendrocyte formation. Specifically, we show that medial ganglionic eminence (MGE) interneurons secrete factors that promote genesis of oligodendrocytes from glially biased cortical precursors in culture. Moreover, when MGE interneurons are genetically ablated in vivo prior to their migration, this causes a deficit in cortical oligodendrogenesis. Modeling of the interneuron-precursor paracrine interaction using transcriptome data identifies the cytokine fractalkine as responsible for the pro-oligodendrocyte effect in culture. This paracrine interaction is important in vivo, since knockdown of the fractalkine receptor CX3CR1 in embryonic cortical precursors, or constitutive knockout of CX3CR1, causes decreased numbers of oligodendrocyte progenitor cells (OPCs) and oligodendrocytes in the postnatal cortex. Thus, in addition to their role in regulating neuronal excitability, interneurons act in a paracrine fashion to promote the developmental genesis of oligodendrocytes.


Asunto(s)
Encéfalo/embriología , Quimiocina CX3CL1/metabolismo , Interneuronas/metabolismo , Eminencia Media/citología , Células-Madre Neurales/metabolismo , Neurogénesis , Oligodendroglía/metabolismo , Receptores de Quimiocina/genética , Animales , Receptor 1 de Quimiocinas CX3C , Diferenciación Celular , Movimiento Celular , Corteza Cerebral/citología , Embrión de Mamíferos , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Células-Madre Neurales/citología , Oligodendroglía/citología , Receptores de Quimiocina/metabolismo
10.
PLoS Genet ; 13(4): e1006746, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28453519

RESUMEN

Mid-hindbrain malformations can occur during embryogenesis through a disturbance of transient and localized gene expression patterns within these distinct brain structures. Rho guanine nucleotide exchange factor (ARHGEF) family members are key for controlling the spatiotemporal activation of Rho GTPase, to modulate cytoskeleton dynamics, cell division, and cell migration. We identified, by means of whole exome sequencing, a homozygous frameshift mutation in the ARHGEF2 as a cause of intellectual disability, a midbrain-hindbrain malformation, and mild microcephaly in a consanguineous pedigree of Kurdish-Turkish descent. We show that loss of ARHGEF2 perturbs progenitor cell differentiation and that this is associated with a shift of mitotic spindle plane orientation, putatively favoring more symmetric divisions. The ARHGEF2 mutation leads to reduction in the activation of the RhoA/ROCK/MLC pathway crucial for cell migration. We demonstrate that the human brain malformation is recapitulated in Arhgef2 mutant mice and identify an aberrant migration of distinct components of the precerebellar system as a pathomechanism underlying the midbrain-hindbrain phenotype. Our results highlight the crucial function of ARHGEF2 in human brain development and identify a mutation in ARHGEF2 as novel cause of a neurodevelopmental disorder.


Asunto(s)
Movimiento Celular/genética , Mutación del Sistema de Lectura/genética , Discapacidad Intelectual/genética , Factores de Intercambio de Guanina Nucleótido Rho/genética , Animales , Citoesqueleto/genética , Exoma/genética , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Homocigoto , Humanos , Discapacidad Intelectual/diagnóstico por imagen , Discapacidad Intelectual/patología , Imagen por Resonancia Magnética , Masculino , Mesencéfalo/diagnóstico por imagen , Mesencéfalo/patología , Ratones , Linaje , Rombencéfalo/diagnóstico por imagen , Rombencéfalo/patología , Transducción de Señal , Proteína de Unión al GTP rhoA/genética
11.
Neurogenesis (Austin) ; 4(1): e1286425, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28265587

RESUMEN

The diverse subtypes of neurons that comprise the mammalian cerebral cortex are produced from a single population of cortical neural precursor cells during the period of embryonic neurogenesis. While this process of neurogenesis is tightly controlled at the transcriptional and translational levels, substantial opportunity exists for extrinsic or niche control of the process of neurogenesis. In our recently published work we made use of a combination of computational and biologic approaches to characterize cell-cell communication between cortical neurons and cortical precursor cells and thereby reveal an unexpectedly complex growth factor communication network that accurately predicted new regulators of cortical neurogenesis.

12.
Methods Mol Biol ; 1523: 237-248, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27975253

RESUMEN

Assembly of the microtubule-associated protein tau (tau) into paired helical filaments that ultimately give rise to neurofibrillary tangles (NFTs) makes up one half of the two hallmark pathologies of Alzheimer's disease (AD). Tau has been shown to be modified with O-linked N-acetylglucosamine residues (O-GlcNAc), which is the modification of serine and threonine residues of nucleocytoplasmic proteins with N-acetyl-D-glucosamine (GlcNAc) moieties. Increasing O-GlcNAc in mouse models of tauopathy has been shown to hinder the progression of symptoms in these mice and impair the aggregation of tau into NFTs. In order to study how O-GlcNAc on tau may contribute to the protective effects observed in tauopathy mouse models, it is beneficial to study O-GlcNAc modified tau in vitro. Here we describe a method for producing, purifying and enriching recombinant tau that is O-GlcNAc modified. These methods have enabled the identification of O-GlcNAc modification sites on tau including Ser400. We further describe the detection of Ser400 O-GlcNAc on tau from brain lysates.


Asunto(s)
Acetilglucosamina/química , Acetilglucosamina/metabolismo , Proteínas tau/química , Proteínas tau/metabolismo , Animales , Encéfalo/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Técnicas In Vitro , Ratones , Procesamiento Proteico-Postraduccional , Ratas , Ratas Sprague-Dawley , Tauopatías/genética , Tauopatías/metabolismo
13.
Neuron ; 91(5): 988-1004, 2016 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-27545711

RESUMEN

The neural stem cell decision to self-renew or differentiate is tightly regulated by its microenvironment. Here, we have asked about this microenvironment, focusing on growth factors in the embryonic cortex at a time when it is largely comprised of neural precursor cells (NPCs) and newborn neurons. We show that cortical NPCs secrete factors that promote their maintenance, while cortical neurons secrete factors that promote differentiation. To define factors important for these activities, we used transcriptome profiling to identify ligands produced by NPCs and neurons, cell-surface mass spectrometry to identify receptors on these cells, and computational modeling to integrate these data. The resultant model predicts a complex growth factor environment with multiple autocrine and paracrine interactions. We tested this communication model, focusing on neurogenesis, and identified IFNγ, Neurturin (Nrtn), and glial-derived neurotrophic factor (GDNF) as ligands with unexpected roles in promoting neurogenic differentiation of NPCs in vivo.


Asunto(s)
Corteza Cerebral/crecimiento & desarrollo , Factor Neurotrófico Derivado de la Línea Celular Glial/fisiología , Interferón gamma/fisiología , Modelos Neurológicos , Factores de Crecimiento Nervioso/metabolismo , Neurogénesis/fisiología , Neurturina/fisiología , Animales , Diferenciación Celular/fisiología , Corteza Cerebral/metabolismo , Humanos , Ligandos , Ratones , Células-Madre Neurales/fisiología , Neuronas/metabolismo , Neuronas/fisiología , Cultivo Primario de Células , Transcriptoma/fisiología
14.
Cell Stem Cell ; 19(4): 433-448, 2016 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-27376984

RESUMEN

Adult mammals have lost multi-tissue regenerative capacity, except for the distal digit, which is able to regenerate via mechanisms that remain largely unknown. Here, we show that, after adult mouse distal digit removal, nerve-associated Schwann cell precursors (SCPs) dedifferentiate and secrete growth factors that promote expansion of the blastema and digit regeneration. When SCPs were dysregulated or ablated, mesenchymal precursor proliferation in the blastema was decreased and nail and bone regeneration were impaired. Transplantation of exogenous SCPs rescued these regeneration defects. We found that SCPs secrete factors that promote self-renewal of mesenchymal precursors, and we used transcriptomic and proteomic analysis to define candidate factors. Two of these, oncostatin M (OSM) and platelet-derived growth factor AA (PDGF-AA), are made by SCPs in the regenerating digit and rescued the deficits in regeneration caused by loss of SCPs. As all peripheral tissues contain nerves, these results could have broad implications for mammalian tissue repair and regeneration.


Asunto(s)
Desdiferenciación Celular , Extremidades/fisiología , Mamíferos/fisiología , Células-Madre Neurales/citología , Comunicación Paracrina , Regeneración , Células de Schwann/citología , Envejecimiento/fisiología , Animales , Desdiferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Autorrenovación de las Células/efectos de los fármacos , Desnervación , Extremidades/inervación , Eliminación de Gen , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Mesodermo/citología , Ratones , Ratones Noqueados , Células-Madre Neurales/trasplante , Oncostatina M/farmacología , Comunicación Paracrina/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Ratas , Regeneración/efectos de los fármacos , Factores de Transcripción SOXB1/metabolismo , Células de Schwann/trasplante , Piel/patología , Cicatrización de Heridas/efectos de los fármacos
15.
Stem Cell Reports ; 6(1): 74-84, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26724904

RESUMEN

Here, we asked whether we could identify pharmacological agents that enhance endogenous stem cell function to promote skin repair, focusing on skin-derived precursors (SKPs), a dermal precursor cell population. Libraries of compounds already used in humans were screened for their ability to enhance the self-renewal of human and rodent SKPs. We identified and validated five such compounds, and showed that two of them, alprostadil and trimebutine maleate, enhanced the repair of full thickness skin wounds in middle-aged mice. Moreover, SKPs isolated from drug-treated skin displayed long-term increases in self-renewal when cultured in basal growth medium without drugs. Both alprostadil and trimebutine maleate likely mediated increases in SKP self-renewal by moderate hyperactivation of the MEK-ERK pathway. These findings identify candidates for potential clinical use in human skin repair, and provide support for the idea that pharmacological activation of endogenous tissue precursors represents a viable therapeutic strategy.


Asunto(s)
Preparaciones Farmacéuticas/administración & dosificación , Piel/efectos de los fármacos , Células Madre/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Alprostadil/administración & dosificación , Alprostadil/farmacología , Animales , Animales Recién Nacidos , Western Blotting , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Autorrenovación de las Células/efectos de los fármacos , Autorrenovación de las Células/genética , Células Cultivadas , Medios de Cultivo/química , Medios de Cultivo/farmacología , Perfilación de la Expresión Génica/métodos , Ontología de Genes , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Células 3T3 NIH , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Piel/metabolismo , Piel/fisiopatología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Esferoides Celulares/fisiología , Células Madre/metabolismo , Células Madre/fisiología , Trimebutino/administración & dosificación , Trimebutino/farmacología , Cicatrización de Heridas/genética
16.
J Mol Cell Biol ; 8(1): 2-16, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26031751

RESUMEN

O-glycosylation of the nuclear pore complex (NPC) by O-linked N-acetylglucosamine (O-GlcNAc) is conserved within metazoans. Many nucleoporins (Nups) comprising the NPC are constitutively O-GlcNAcylated, but the functional role of this modification remains enigmatic. We show that loss of O-GlcNAc, induced by either inhibition of O-GlcNAc transferase (OGT) or deletion of the gene encoding OGT, leads to decreased cellular levels of a number of natively O-GlcNAcylated Nups. Loss of O-GlcNAc enables increased ubiquitination of these Nups and their increased proteasomal degradation. The decreased half-life of these deglycosylated Nups manifests in their gradual loss from the NPC and a downstream malfunction of the nuclear pore selective permeability barrier in both dividing and post-mitotic cells. These findings define a critical role of O-GlcNAc modification of the NPC in maintaining its composition and the function of the selectivity filter. The results implicate NPC glycosylation as a regulator of NPC function and reveal the role of conserved glycosylation of the NPC among metazoans.


Asunto(s)
Poro Nuclear/metabolismo , Animales , Línea Celular , Glicosilación , Ratones , Proteínas de Complejo Poro Nuclear/metabolismo , Procesamiento Proteico-Postraduccional/genética , Procesamiento Proteico-Postraduccional/fisiología , Estabilidad Proteica , Ubiquitinación/genética , Ubiquitinación/fisiología
17.
J Biol Chem ; 289(50): 34472-81, 2014 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-25336656

RESUMEN

Regional glucose hypometabolism is a defining feature of Alzheimer disease (AD). One emerging link between glucose hypometabolism and progression of AD is the nutrient-responsive post-translational O-GlcNAcylation of nucleocytoplasmic proteins. O-GlcNAc is abundant in neurons and occurs on both tau and amyloid precursor protein. Increased brain O-GlcNAcylation protects against tau and amyloid-ß peptide toxicity. Decreased O-GlcNAcylation occurs in AD, suggesting that glucose hypometabolism may impair the protective roles of O-GlcNAc within neurons and enable neurodegeneration. Here, we review how O-GlcNAc may link cerebral glucose hypometabolism to progression of AD and summarize data regarding the protective role of O-GlcNAc in AD models.


Asunto(s)
Acetilglucosamina/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Glucosa/metabolismo , Humanos , Neuronas/metabolismo , Proteínas tau/metabolismo
18.
Mol Neurodegener ; 9: 42, 2014 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-25344697

RESUMEN

BACKGROUND: Amyloid plaques and neurofibrillary tangles (NFTs) are the defining pathological hallmarks of Alzheimer's disease (AD). Increasing the quantity of the O-linked N-acetylglucosamine (O-GlcNAc) post-translational modification of nuclear and cytoplasmic proteins slows neurodegeneration and blocks the formation of NFTs in a tauopathy mouse model. It remains unknown, however, if O-GlcNAc can influence the formation of amyloid plaques in the presence of tau pathology. RESULTS: We treated double transgenic TAPP mice, which express both mutant human tau and amyloid precursor protein (APP), with a highly selective orally bioavailable inhibitor of the enzyme responsible for removing O-GlcNAc (OGA) to increase O-GlcNAc in the brain. We find that increased O-GlcNAc levels block cognitive decline in the TAPP mice and this effect parallels decreased ß-amyloid peptide levels and decreased levels of amyloid plaques. CONCLUSIONS: This study indicates that increased O-GlcNAc can influence ß-amyloid pathology in the presence of tau pathology. The findings provide good support for OGA as a promising therapeutic target to alter disease progression in Alzheimer disease.


Asunto(s)
Enfermedad de Alzheimer/enzimología , N-Acetilglucosaminiltransferasas/metabolismo , Placa Amiloide/patología , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Western Blotting , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunohistoquímica , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Transgénicos , Proteínas tau/genética
19.
Chem Soc Rev ; 43(19): 6839-58, 2014 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-24759912

RESUMEN

Alzheimer disease (AD) is a growing problem for aging populations worldwide. Despite significant efforts, no therapeutics are available that stop or slow progression of AD, which has driven interest in the basic causes of AD and the search for new therapeutic strategies. Longitudinal studies have clarified that defects in glucose metabolism occur in patients exhibiting Mild Cognitive Impairment (MCI) and glucose hypometabolism is an early pathological change within AD brain. Further, type 2 diabetes mellitus (T2DM) is a strong risk factor for the development of AD. These findings have stimulated interest in the possibility that disrupted glucose regulated signaling within the brain could contribute to the progression of AD. One such process of interest is the addition of O-linked N-acetylglucosamine (O-GlcNAc) residues onto nuclear and cytoplasmic proteins within mammals. O-GlcNAc is notably abundant within brain and is present on hundreds of proteins including several, such as tau and the amyloid precursor protein, which are involved in the pathophysiology AD. The cellular levels of O-GlcNAc are coupled to nutrient availability through the action of just two enzymes. O-GlcNAc transferase (OGT) is the glycosyltransferase that acts to install O-GlcNAc onto proteins and O-GlcNAcase (OGA) is the glycoside hydrolase that acts to remove O-GlcNAc from proteins. Uridine 5'-diphosphate-N-acetylglucosamine (UDP-GlcNAc) is the donor sugar substrate for OGT and its levels vary with cellular glucose availability because it is generated from glucose through the hexosamine biosynthetic pathway (HBSP). Within the brains of AD patients O-GlcNAc levels have been found to be decreased and aggregates of tau appear to lack O-GlcNAc entirely. Accordingly, glucose hypometabolism within the brain may result in disruption of the normal functions of O-GlcNAc within the brain and thereby contribute to downstream neurodegeneration. While this hypothesis remains largely speculative, recent studies using different mouse models of AD have demonstrated the protective benefit of pharmacologically increased brain O-GlcNAc levels. In this review we summarize the state of knowledge in the area of O-GlcNAc as it pertains to AD while also addressing some of the basic biochemical roles of O-GlcNAc and how these might contribute to protecting against AD and other neurodegenerative diseases.


Asunto(s)
Acetilglucosamina/metabolismo , Enfermedad de Alzheimer/patología , Enfermedades Neurodegenerativas/patología , Acetilglucosamina/química , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Animales , Antígenos de Neoplasias/metabolismo , Histona Acetiltransferasas/antagonistas & inhibidores , Histona Acetiltransferasas/metabolismo , Humanos , Hialuronoglucosaminidasa/antagonistas & inhibidores , Hialuronoglucosaminidasa/metabolismo , N-Acetilglucosaminiltransferasas/antagonistas & inhibidores , N-Acetilglucosaminiltransferasas/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Proteínas tau/química , Proteínas tau/genética , Proteínas tau/metabolismo
20.
J Mol Biol ; 426(8): 1736-52, 2014 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-24444746

RESUMEN

The aggregation of the microtubule-associated protein tau into paired helical filaments to form neurofibrillary tangles constitutes one of the pathological hallmarks of Alzheimer's disease. Tau is post-translationally modified by the addition of N-acetyl-D-glucosamine O-linked to several serine and threonine residues (O-GlcNAc). Previously, increased O-GlcNAcylation of tau has been shown to block the accumulation of tau aggregates within a tauopathy mouse model. Here we show that O-GlcNAc modification of full-length human tau impairs the rate and extent of its heparin-induced aggregation without perturbing its activity toward microtubule polymerization. O-GlcNAcylation, however, does not impact the "global-fold" of tau as measured by a Förster resonance energy transfer assay. Similarly, nuclear magnetic resonance studies demonstrated that O-GlcNAcylation only minimally perturbs the local structural and dynamic features of a tau fragment (residues 353-408) spanning the last microtubule binding repeat to the major GlcNAc-acceptor Ser400. These data indicate that the inhibitory effects of O-GlcNAc on tau aggregation may result from enhanced monomer solubility or the destabilization of fibrils or soluble aggregates, rather than by altering the conformational properties of the monomeric protein. This work further underscores the potential of targeting the O-GlcNAc pathway for potential Alzheimer's disease therapeutics.


Asunto(s)
Proteínas tau/química , Acetilglucosamina/química , Acetilglucosamina/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Transferencia Resonante de Energía de Fluorescencia , Humanos , Ratones , Microtúbulos/química , Microtúbulos/metabolismo , Mutagénesis Sitio-Dirigida , Resonancia Magnética Nuclear Biomolecular , Conformación Proteica , Pliegue de Proteína , Multimerización de Proteína , Procesamiento Proteico-Postraduccional , Estructura Secundaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...