Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2023 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-37790503

RESUMEN

Proper brain function requires the assembly and function of diverse populations of neurons and glia. Single cell gene expression studies have mostly focused on characterization of neuronal cell diversity; however, recent studies have revealed substantial diversity of glial cells, particularly astrocytes. To better understand glial cell types and their roles in neurobiology, we built a new suite of adeno-associated viral (AAV)-based genetic tools to enable genetic access to astrocytes and oligodendrocytes. These oligodendrocyte and astrocyte enhancer-AAVs are highly specific (usually > 95% cell type specificity) with variable expression levels, and our astrocyte enhancer-AAVs show multiple distinct expression patterns reflecting the spatial distribution of astrocyte cell types. To provide the best glial-specific functional tools, several enhancer-AAVs were: optimized for higher expression levels, shown to be functional and specific in rat and macaque, shown to maintain specific activity in epilepsy where traditional promoters changed activity, and used to drive functional transgenes in astrocytes including Cre recombinase and acetylcholine-responsive sensor iAChSnFR. The astrocyte-specific iAChSnFR revealed a clear reward-dependent acetylcholine response in astrocytes of the nucleus accumbens during reinforcement learning. Together, this collection of glial enhancer-AAVs will enable characterization of astrocyte and oligodendrocyte populations and their roles across species, disease states, and behavioral epochs.

2.
Nat Nanotechnol ; 18(10): 1241-1251, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37430038

RESUMEN

Crossing the blood-brain barrier in primates is a major obstacle for gene delivery to the brain. Adeno-associated viruses (AAVs) promise robust, non-invasive gene delivery from the bloodstream to the brain. However, unlike in rodents, few neurotropic AAVs efficiently cross the blood-brain barrier in non-human primates. Here we report on AAV.CAP-Mac, an engineered variant identified by screening in adult marmosets and newborn macaques, which has improved delivery efficiency in the brains of multiple non-human primate species: marmoset, rhesus macaque and green monkey. CAP-Mac is neuron biased in infant Old World primates, exhibits broad tropism in adult rhesus macaques and is vasculature biased in adult marmosets. We demonstrate applications of a single, intravenous dose of CAP-Mac to deliver functional GCaMP for ex vivo calcium imaging across multiple brain areas, or a cocktail of fluorescent reporters for Brainbow-like labelling throughout the macaque brain, circumventing the need for germline manipulations in Old World primates. As such, CAP-Mac is shown to have potential for non-invasive systemic gene transfer in the brains of non-human primates.


Asunto(s)
Encéfalo , Callithrix , Humanos , Animales , Recién Nacido , Chlorocebus aethiops , Macaca mulatta/genética , Callithrix/genética , Encéfalo/fisiología , Técnicas de Transferencia de Gen , Neuronas , Vectores Genéticos/genética
3.
Nat Commun ; 14(1): 3345, 2023 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-37291094

RESUMEN

Delivering genes to and across the brain vasculature efficiently and specifically across species remains a critical challenge for addressing neurological diseases. We have evolved adeno-associated virus (AAV9) capsids into vectors that transduce brain endothelial cells specifically and efficiently following systemic administration in wild-type mice with diverse genetic backgrounds, and in rats. These AAVs also exhibit superior transduction of the CNS across non-human primates (marmosets and rhesus macaques), and in ex vivo human brain slices, although the endothelial tropism is not conserved across species. The capsid modifications translate from AAV9 to other serotypes such as AAV1 and AAV-DJ, enabling serotype switching for sequential AAV administration in mice. We demonstrate that the endothelial-specific mouse capsids can be used to genetically engineer the blood-brain barrier by transforming the mouse brain vasculature into a functional biofactory. We apply this approach to Hevin knockout mice, where AAV-X1-mediated ectopic expression of the synaptogenic protein Sparcl1/Hevin in brain endothelial cells rescued synaptic deficits.


Asunto(s)
Células Endoteliales , Roedores , Ratones , Ratas , Animales , Células Endoteliales/metabolismo , Roedores/genética , Macaca mulatta/genética , Encéfalo/metabolismo , Tropismo/genética , Ratones Noqueados , Dependovirus/metabolismo , Vectores Genéticos/genética , Transducción Genética , Proteínas de Unión al Calcio/metabolismo , Proteínas de la Matriz Extracelular/genética
4.
Res Sq ; 2023 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-36789432

RESUMEN

Adeno-associated viruses (AAVs) promise robust gene delivery to the brain through non-invasive, intravenous delivery. However, unlike in rodents, few neurotropic AAVs efficiently cross the blood-brain barrier in non-human primates (NHPs). Here we describe AAV.CAP-Mac, an engineered variant identified by screening in adult marmosets and newborn macaques with improved efficiency in the brain of multiple NHP species: marmoset, rhesus macaque, and green monkey. CAP-Mac is neuron-biased in infant Old World primates, exhibits broad tropism in adult rhesus macaques, and is vasculature-biased in adult marmosets. We demonstrate applications of a single, intravenous dose of CAP-Mac to deliver (1) functional GCaMP for ex vivo calcium imaging across multiple brain areas, and (2) a cocktail of fluorescent reporters for Brainbow-like labeling throughout the macaque brain, circumventing the need for germline manipulations in Old World primates. Given its capabilities for systemic gene transfer in NHPs, CAP-Mac promises to help unlock non-invasive access to the brain.

5.
bioRxiv ; 2023 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-36711773

RESUMEN

Delivering genes to and across the brain vasculature efficiently and specifically across species remains a critical challenge for addressing neurological diseases. We have evolved adeno-associated virus (AAV9) capsids into vectors that transduce brain endothelial cells specifically and efficiently following systemic administration in wild-type mice with diverse genetic backgrounds and rats. These AAVs also exhibit superior transduction of the CNS across non-human primates (marmosets and rhesus macaques), and ex vivo human brain slices although the endothelial tropism is not conserved across species. The capsid modifications translate from AAV9 to other serotypes such as AAV1 and AAV-DJ, enabling serotype switching for sequential AAV administration in mice. We demonstrate that the endothelial specific mouse capsids can be used to genetically engineer the blood-brain barrier by transforming the mouse brain vasculature into a functional biofactory. Vasculature-secreted Hevin (a synaptogenic protein) rescued synaptic deficits in a mouse model.

6.
bioRxiv ; 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38168178

RESUMEN

Dravet syndrome (DS) is a devastating developmental epileptic encephalopathy marked by treatment-resistant seizures, developmental delay, intellectual disability, motor deficits, and a 10-20% rate of premature death. Most DS patients harbor loss-of-function mutations in one copy of SCN1A , which has been associated with inhibitory neuron dysfunction. Here we developed an interneuron-targeting AAV human SCN1A gene replacement therapy using cell class-specific enhancers. We generated a split-intein fusion form of SCN1A to circumvent AAV packaging limitations and deliver SCN1A via a dual vector approach using cell class-specific enhancers. These constructs produced full-length Na V 1.1 protein and functional sodium channels in HEK293 cells and in brain cells in vivo . After packaging these vectors into enhancer-AAVs and administering to mice, immunohistochemical analyses showed telencephalic GABAergic interneuron-specific and dose-dependent transgene biodistribution. These vectors conferred strong dose-dependent protection against postnatal mortality and seizures in two DS mouse models carrying independent loss-of-function alleles of Scn1a, at two independent research sites, supporting the robustness of this approach. No mortality or toxicity was observed in wild-type mice injected with single vectors expressing either the N-terminal or C-terminal halves of SCN1A , or the dual vector system targeting interneurons. In contrast, nonselective neuronal targeting of SCN1A conferred less rescue against mortality and presented substantial preweaning lethality. These findings demonstrate proof-of-concept that interneuron-specific AAV-mediated SCN1A gene replacement is sufficient for significant rescue in DS mouse models and suggest it could be an effective therapeutic approach for patients with DS.

7.
Neuron ; 109(9): 1449-1464.e13, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33789083

RESUMEN

Rapid cell type identification by new genomic single-cell analysis methods has not been met with efficient experimental access to these cell types. To facilitate access to specific neural populations in mouse cortex, we collected chromatin accessibility data from individual cells and identified enhancers specific for cell subclasses and types. When cloned into recombinant adeno-associated viruses (AAVs) and delivered to the brain, these enhancers drive transgene expression in specific cortical cell subclasses. We extensively characterized several enhancer AAVs to show that they label different projection neuron subclasses as well as a homologous neuron subclass in human cortical slices. We also show how coupling enhancer viruses expressing recombinases to a newly generated transgenic mouse, Ai213, enables strong labeling of three different neuronal classes/subclasses in the brain of a single transgenic animal. This approach combines unprecedented flexibility with specificity for investigation of cell types in the mouse brain and beyond.


Asunto(s)
Encéfalo/citología , Neuronas/clasificación , Neuronas/citología , Análisis de la Célula Individual/métodos , Animales , Conjuntos de Datos como Asunto , Dependovirus , Humanos , Ratones , Ratones Transgénicos
8.
Cell Rep ; 34(13): 108754, 2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33789096

RESUMEN

Viral genetic tools that target specific brain cell types could transform basic neuroscience and targeted gene therapy. Here, we use comparative open chromatin analysis to identify thousands of human-neocortical-subclass-specific putative enhancers from across the genome to control gene expression in adeno-associated virus (AAV) vectors. The cellular specificity of reporter expression from enhancer-AAVs is established by molecular profiling after systemic AAV delivery in mouse. Over 30% of enhancer-AAVs produce specific expression in the targeted subclass, including both excitatory and inhibitory subclasses. We present a collection of Parvalbumin (PVALB) enhancer-AAVs that show highly enriched expression not only in cortical PVALB cells but also in some subcortical PVALB populations. Five vectors maintain PVALB-enriched expression in primate neocortex. These results demonstrate how genome-wide open chromatin data mining and cross-species AAV validation can be used to create the next generation of non-species-restricted viral genetic tools.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Neocórtex/metabolismo , Animales , Cromatina/genética , Cromatina/metabolismo , Bases de Datos Genéticas , Dependovirus/genética , Enfermedad/genética , Epigénesis Genética , Vectores Genéticos/metabolismo , Genoma , Humanos , Ratones , Neuronas/metabolismo , Parvalbúminas/metabolismo , Primates , Especificidad de la Especie
9.
Elife ; 62017 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-28394253

RESUMEN

The maintenance of excitatory and inhibitory balance in the brain is essential for its function. Here we find that the developmental axon guidance receptor Roundabout 2 (Robo2) is critical for the maintenance of inhibitory synapses in the adult ventral tegmental area (VTA), a brain region important for the production of the neurotransmitter dopamine. Following selective genetic inactivation of Robo2 in the adult VTA of mice, reduced inhibitory control results in altered neural activity patterns, enhanced phasic dopamine release, behavioral hyperactivity, associative learning deficits, and a paradoxical inversion of psychostimulant responses. These behavioral phenotypes could be phenocopied by selective inactivation of synaptic transmission from local GABAergic neurons of the VTA, demonstrating an important function for Robo2 in regulating the excitatory and inhibitory balance of the adult brain.


Asunto(s)
Dopamina/metabolismo , Receptores Inmunológicos/metabolismo , Transmisión Sináptica , Área Tegmental Ventral/fisiología , Animales , Conducta Animal , Femenino , Técnicas de Inactivación de Genes , Masculino , Ratones Endogámicos C57BL , Ácido gamma-Aminobutírico/metabolismo
10.
J Biol Chem ; 290(22): 14107-19, 2015 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-25882844

RESUMEN

Anchoring proteins direct protein kinases and phosphoprotein phosphatases toward selected substrates to control the efficacy, context, and duration of neuronal phosphorylation events. The A-kinase anchoring protein AKAP79/150 interacts with protein kinase A (PKA), protein kinase C (PKC), and protein phosphatase 2B (calcineurin) to modulate second messenger signaling events. In a mass spectrometry-based screen for additional AKAP79/150 binding partners, we have identified the Roundabout axonal guidance receptor Robo2 and its ligands Slit2 and Slit3. Biochemical and cellular approaches confirm that a linear sequence located in the cytoplasmic tail of Robo2 (residues 991-1070) interfaces directly with sites on the anchoring protein. Parallel studies show that AKAP79/150 interacts with the Robo3 receptor in a similar manner. Immunofluorescent staining detects overlapping expression patterns for murine AKAP150, Robo2, and Robo3 in a variety of brain regions, including hippocampal region CA1 and the islands of Calleja. In vitro kinase assays, peptide spot array mapping, and proximity ligation assay staining approaches establish that human AKAP79-anchored PKC selectively phosphorylates the Robo3.1 receptor subtype on serine 1330. These findings imply that anchored PKC locally modulates the phosphorylation status of Robo3.1 in brain regions governing learning and memory and reward.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Proteína Quinasa C/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Encéfalo/metabolismo , Citoplasma/metabolismo , Silenciador del Gen , Glutatión Transferasa/metabolismo , Células HEK293 , Hipocampo/metabolismo , Humanos , Ligandos , Sustancias Macromoleculares , Espectrometría de Masas , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Fosforilación , Mapeo de Interacción de Proteínas , Isoformas de Proteínas , ARN Interferente Pequeño/metabolismo , Receptores de Superficie Celular , Transducción de Señal
11.
Learn Mem ; 21(11): 575-9, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25320348

RESUMEN

Dopamine is broadly implicated in fear-related processes, yet we know very little about signaling dynamics in these neurons during active fear conditioning. We describe the direct imaging of calcium signals of dopamine neurons during Pavlovian fear conditioning using fiber-optic confocal microscopy coupled with the genetically encoded calcium indicator GCaMP3. We observed calcium transients in a subset of dopamine neurons to an unconditioned fear stimulus on the first day of Pavlovian fear conditioning. On the second day, calcium transients occurred in response to conditioned and unconditioned stimuli. These results demonstrate plasticity in dopamine neuron calcium signals and the occurrence of activity-dependent processes in these neurons during fear conditioning.


Asunto(s)
Señalización del Calcio , Neuronas Dopaminérgicas/metabolismo , Miedo/fisiología , Plasticidad Neuronal , Área Tegmental Ventral/metabolismo , Animales , Condicionamiento Clásico/fisiología , Electrochoque , Ratones
12.
J Neurosci ; 33(20): 8640-9, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23678109

RESUMEN

The dopamine D1 receptor (D1R) facilitates reward acquisition and its alteration leads to profound learning deficits. However, its minimal functional circuit requirement is unknown. Using conditional reconstruction of functional D1R signaling in D1R knock-out mice, we define distinct requirements of D1R in subregions of the nucleus accumbens (NAc) for specific dimensions of reward. We demonstrate that D1R expression in the core region of the NAc (NAc(Core)), but not the shell (NAc(Shell)), enhances selectively a unique form of pavlovian conditioned approach and mediates D1R-dependent cocaine sensitization. However, D1R expression in either the NAc(Core) or the NAc(Shell) improves instrumental responding for reward. In contrast, neither NAc(Core) nor NAc(Shell) D1R is sufficient to promote motivation to work for reward in a progressive ratio task or for motor learning. These results highlight dissociated circuit requirements of D1R for dopamine-dependent behaviors.


Asunto(s)
Condicionamiento Clásico/fisiología , Condicionamiento Operante/fisiología , Núcleo Accumbens/fisiología , Receptores de Dopamina D1/metabolismo , Recompensa , Transducción de Señal/genética , Análisis de Varianza , Animales , Benzazepinas/administración & dosificación , Cocaína/administración & dosificación , Condicionamiento Clásico/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Agonistas de Dopamina/administración & dosificación , Inhibidores de Captación de Dopamina/administración & dosificación , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Locomoción/efectos de los fármacos , Locomoción/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Núcleo Accumbens/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores de Dopamina D1/genética , Prueba de Desempeño de Rotación con Aceleración Constante , Transducción de Señal/efectos de los fármacos
13.
Curr Protoc Neurosci ; 65: 4.35.1-20, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-25429312

RESUMEN

The mammalian brain contains tremendous structural and genetic complexity that is vital for its function. The elucidation of gene expression profiles in the brain, coupled with the development of large-scale connectivity maps and emerging viral vector-based approaches for target-selective gene manipulation, now allow for detailed dissection of gene-circuit interfaces. This protocol details how to perform combinatorial viral injections to manipulate gene expression in subsets of neurons interconnecting two brain regions. This method utilizes stereotaxic injection of a retrograde transducing CAV2-Cre virus into one brain region, combined with injection of a locally transducing Cre-dependent AAV virus into another brain region. This technique is widely applicable to the genetic dissection of neural circuitry, as it enables selective expression of candidate genes, dominant-negatives, fluorescent reporters, or genetic tools within heterogeneous populations of neurons based upon their projection targets.


Asunto(s)
Adenovirus Caninos , Encéfalo/fisiología , Dependovirus , Expresión Génica/genética , Técnicas Genéticas , Vectores Genéticos , Animales , Ratones , Modelos Animales , Neuronas/fisiología , Transgenes
14.
Neuron ; 58(3): 325-32, 2008 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-18466743

RESUMEN

Alternative splicing provides a means to increase the complexity of gene function in numerous biological processes, including nervous system wiring. Navigating axons switch responses from attraction to repulsion at intermediate targets, allowing them to grow to each intermediate target and then to move on. The mechanisms underlying this switch remain poorly characterized. We previously showed that the Slit receptor Robo3 is required for spinal commissural axons to enter and cross the midline intermediate target. We report here the existence of two functionally antagonistic isoforms of Robo3 with distinct carboxy termini arising from alternative splicing. Robo3.1 is deployed on the precrossing and crossing portions of commissural axons and allows midline crossing by silencing Slit repulsion. Robo3.2 becomes expressed on the postcrossing portion and blocks midline recrossing, favoring Slit repulsion. The tight spatial regulation of opponent splice variants helps ensure high-fidelity transition of axonal responses from attraction to repulsion at the midline.


Asunto(s)
Empalme Alternativo/fisiología , Axones/fisiología , Encéfalo/embriología , Conos de Crecimiento/fisiología , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Animales , Encéfalo/citología , Encéfalo/fisiología , Células COS , Embrión de Pollo , Pollos , Chlorocebus aethiops , Técnicas de Cultivo de Embriones , Regulación del Desarrollo de la Expresión Génica/fisiología , Isomerismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Ratones , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Fenotipo , Precursores del ARN/genética , Receptores de Superficie Celular
15.
Neuron ; 57(4): 501-10, 2008 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-18304480

RESUMEN

Commissural axons are attracted to the midline intermediate target by chemoattractants, but upon crossing the midline they switch off responsiveness to attractants and switch on responsiveness to midline repellents, which expel the axons from the midline and enable them to move on. Here we show that midline exit also requires the stimulation of axon outgrowth by Stem Cell Factor (SCF, also known as Steel Factor). SCF is expressed by midline floor plate cells, and its receptor Kit, a receptor tyrosine kinase, is expressed by commissural axons. In Steel and Kit mutant mice, the majority of commissural axons line up transiently at the contralateral edge of the floor plate, showing a delay in midline exit. In vitro, SCF selectively promotes outgrowth of postcrossing, but not precrossing, commissural axons. Our findings identify SCF as a guidance cue in the CNS, and provide evidence that exiting intermediate targets requires activation of outgrowth-promoting mechanisms.


Asunto(s)
Axones/fisiología , Placa Neural/embriología , Placa Neural/metabolismo , Factor de Células Madre/biosíntesis , Animales , Ratones , Ratones Mutantes , Placa Neural/crecimiento & desarrollo , Proteínas Proto-Oncogénicas c-kit/biosíntesis , Proteínas Proto-Oncogénicas c-kit/genética , Factor de Células Madre/genética
16.
Dev Neurobiol ; 68(1): 83-100, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17948241

RESUMEN

The division of the mammalian forebrain into distinct left and right hemispheres represents a critical step in neural development. Several signaling molecules including sonic hedgehog (SHH), fibroblast growth factor 8 (FGF8), and bone morphogenetic proteins (BMPs) have been implicated in dorsal midline development, and prior work suggests that the organizing centers from which these proteins are secreted mutually regulate one another during development. To explore the role of the ventral organizing center in the formation of two hemispheres, we assessed dorsal midline development in Shh mutant embryos and in wildtype embryos treated with the SHH signaling inhibitor HhAntag. Collectively, our findings demonstrate that SHH signaling plays an important role in maintaining the normal expression patterns of Fgf8 and Bmp4 in the developing forebrain. We further show that FGF8 can induce the expression of Zic2, which is normally expressed at the midline and is required in vivo for hemispheric cleavage, suggesting that FGF signaling may stimulate dorsal midline development by inducing Zic2 expression.


Asunto(s)
Embrión de Mamíferos/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas Hedgehog/fisiología , Prosencéfalo/embriología , Prosencéfalo/metabolismo , Transducción de Señal/fisiología , Animales , Tipificación del Cuerpo/genética , Proteína Morfogenética Ósea 4 , Proteínas Morfogenéticas Óseas/metabolismo , Bromodesoxiuridina/metabolismo , Relación Dosis-Respuesta a Droga , Inducción Embrionaria/fisiología , Inhibidores Enzimáticos/farmacología , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Proteínas Hedgehog/antagonistas & inhibidores , Proteínas Hedgehog/genética , Holoprosencefalia/genética , Etiquetado Corte-Fin in Situ/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Técnicas de Cultivo de Órganos , Factores de Transcripción/metabolismo
17.
PLoS One ; 2(6): e575, 2007 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-17593974

RESUMEN

BACKGROUND: Heparan sulfate proteoglycans (HSPGs) use highly sulfated polysaccharide side-chains to interact with several key growth factors and morphogens, thereby regulating their accessibility and biological activity. Various sulfotransferases and sulfatases with differing specificities control the pattern of HSPG sulfation, which is functionally critical. Among these enzymes in the mouse are two secreted 6-O-endosulfatases, Sulf1 and Sulf2, which modify HSPGs in the extracellular matrix and on the cell surface. The roles of Sulf1 and Sulf2 during normal development are not well understood. METHODS/RESULTS: To investigate the importance of Sulf1 and Sulf2 for embryonic development, we generated mice genetically deficient in these genes and assessed the phenotypes of the resulting secreted sulfatase-deficient mice. Surprisingly, despite the established crucial role of HSPG interactions during development, neither Sulf1- nor Sulf2-deficient mice showed significant developmental flaws. In contrast, mice deficient in both Sulf1and Sulf2 exhibited highly penetrant neonatal lethality. Loss of viability was associated with multiple, although subtle, developmental defects, including skeletal and renal abnormalities. CONCLUSIONS: These results show that Sulf1 and Sulf2 play overlapping yet critical roles in mouse development and are redundant and essential for neonatal survival.


Asunto(s)
Animales Recién Nacidos/crecimiento & desarrollo , Genes Letales/fisiología , Proteoglicanos de Heparán Sulfato/metabolismo , Sulfatasas/fisiología , Sulfotransferasas/fisiología , Animales , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Hibridación in Situ , Riñón/anomalías , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/anomalías , Embarazo , Transducción de Señal , Tasa de Supervivencia
18.
Science ; 310(5745): 106-10, 2005 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-16210539

RESUMEN

Inhibitory molecules associated with myelin and the glial scar limit axon regeneration in the adult central nervous system (CNS), but the underlying signaling mechanisms of regeneration inhibition are not fully understood. Here, we show that suppressing the kinase function of the epidermal growth factor receptor (EGFR) blocks the activities of both myelin inhibitors and chondroitin sulfate proteoglycans in inhibiting neurite outgrowth. In addition, regeneration inhibitors trigger the phosphorylation of EGFR in a calcium-dependent manner. Local administration of EGFR inhibitors promotes significant regeneration of injured optic nerve fibers, pointing to a promising therapeutic avenue for enhancing axon regeneration after CNS injury.


Asunto(s)
Axones/fisiología , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Proteínas de la Mielina/metabolismo , Regeneración Nerviosa , Animales , Axones/efectos de los fármacos , Calcio/metabolismo , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Clorhidrato de Erlotinib , Proteínas Ligadas a GPI , Humanos , Ratones , Proteínas de la Mielina/farmacología , Compresión Nerviosa , Regeneración Nerviosa/efectos de los fármacos , Neuritas/efectos de los fármacos , Neuritas/fisiología , Proteínas Nogo , Receptor Nogo 1 , Nervio Óptico/efectos de los fármacos , Nervio Óptico/fisiología , Traumatismos del Nervio Óptico/tratamiento farmacológico , Fosforilación , Quinazolinas/farmacología , Receptores de Superficie Celular/metabolismo , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/fisiología , Transducción de Señal/efectos de los fármacos , Tirfostinos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA