Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
EMBO J ; 37(7)2018 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-29459438

RESUMEN

Death receptor 6 (DR6) is an orphan member of the TNF receptor superfamily and controls cell death and differentiation in a cell-autonomous manner in different cell types. Here, we report an additional non-cell-autonomous function for DR6 in the peripheral nervous system (PNS). DR6-knockout (DR6 KO) mice showed precocious myelination in the PNS Using an in vitro myelination assay, we demonstrate that neuronal DR6 acts in trans on Schwann cells (SCs) and reduces SC proliferation and myelination independently of its cytoplasmic death domain. Mechanistically, DR6 was found to be cleaved in neurons by "a disintegrin and metalloprotease 10" (ADAM10), releasing the soluble DR6 ectodomain (sDR6). Notably, in the in vitro myelination assay, sDR6 was sufficient to rescue the DR6 KO phenotype. Thus, in addition to the cell-autonomous receptor function of full-length DR6, the proteolytically released sDR6 can unexpectedly also act as a paracrine signaling factor in the PNS in a non-cell-autonomous manner during SC proliferation and myelination. This new mode of DR6 signaling will be relevant in future attempts to target DR6 in disease settings.


Asunto(s)
Proteína ADAM10/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Proliferación Celular , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Células de Schwann/metabolismo , Animales , Muerte Celular , Línea Celular , Citoplasma/metabolismo , Dominio de Muerte , Desintegrinas/metabolismo , Femenino , Células HEK293 , Humanos , Hibridomas , Masculino , Metaloproteasas/metabolismo , Ratones , Ratones Noqueados , Vaina de Mielina/metabolismo , Comunicación Paracrina , Fenotipo , Receptores del Factor de Necrosis Tumoral/genética , Células de Schwann/ultraestructura , Especificidad por Sustrato
2.
EMBO Rep ; 21(3): e48512, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-31919978

RESUMEN

Regulation of axon guidance and pruning of inappropriate synapses by class 3 semaphorins are key to the development of neural circuits. Collapsin response mediator protein 2 (CRMP2) has been shown to regulate axon guidance by mediating semaphorin 3A (Sema3A) signaling; however, nothing is known about its role in synapse pruning. Here, using newly generated crmp2-/- mice we demonstrate that CRMP2 has a moderate effect on Sema3A-dependent axon guidance in vivo, and its deficiency leads to a mild defect in axon guidance in peripheral nerves and the corpus callosum. Surprisingly, crmp2-/- mice display prominent defects in stereotyped axon pruning in hippocampus and visual cortex and altered dendritic spine remodeling, which is consistent with impaired Sema3F signaling and with models of autism spectrum disorder (ASD). We demonstrate that CRMP2 mediates Sema3F signaling in primary neurons and that crmp2-/- mice display ASD-related social behavior changes in the early postnatal period as well as in adults. Together, we demonstrate that CRMP2 mediates Sema3F-dependent synapse pruning and its dysfunction shares histological and behavioral features of ASD.


Asunto(s)
Trastorno del Espectro Autista , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de la Membrana/fisiología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Semaforinas , Animales , Espinas Dendríticas , Ratones , Ratones Noqueados , Plasticidad Neuronal , Neuronas , Transducción de Señal
3.
Acta Neuropathol ; 140(2): 121-142, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32562018

RESUMEN

Expansion of a (G4C2)n repeat in C9orf72 causes amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), but the link of the five repeat-encoded dipeptide repeat (DPR) proteins to neuroinflammation, TDP-43 pathology, and neurodegeneration is unclear. Poly-PR is most toxic in vitro, but poly-GA is far more abundant in patients. To directly compare these in vivo, we created congenic poly-GA and poly-PR mice. 40% of poly-PR mice were affected with ataxia and seizures, requiring euthanasia by 6 weeks of age. The remaining poly-PR mice were asymptomatic at 14 months of age, likely due to an 80% reduction of the transgene mRNA in this subgroup. In contrast, all poly-GA mice showed selective neuron loss, inflammation, as well as muscle denervation and wasting requiring euthanasia before 7 weeks of age. In-depth analysis of peripheral organs and blood samples suggests that peripheral organ failure does not drive these phenotypes. Although transgene mRNA levels were similar between poly-GA and affected poly-PR mice, poly-GA aggregated far more abundantly than poly-PR in the CNS and was also found in skeletal muscle. In addition, TDP-43 and other disease-linked RNA-binding proteins co-aggregated in rare nuclear inclusions in the hippocampus and frontal cortex only in poly-GA mice. Transcriptome analysis revealed activation of an interferon-responsive pro-inflammatory microglial signature in end-stage poly-GA but not poly-PR mice. This signature was also found in all ALS patients and enriched in C9orf72 cases. In summary, our rigorous comparison of poly-GA and poly-PR toxicity in vivo indicates that poly-GA, but not poly-PR at the same mRNA expression level, promotes interferon responses in C9orf72 disease and contributes to TDP-43 abnormalities and neuron loss selectively in disease-relevant regions.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Proteína C9orf72/genética , Interferones/biosíntesis , Degeneración Nerviosa/patología , Esclerosis Amiotrófica Lateral/inmunología , Esclerosis Amiotrófica Lateral/patología , Animales , Expansión de las Repeticiones de ADN/genética , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Degeneración Nerviosa/genética , Degeneración Nerviosa/inmunología , Neuronas/patología
5.
Development ; 141(1): 28-38, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24284204

RESUMEN

Meis homeodomain transcription factors control cell proliferation, cell fate specification and differentiation in development and disease. Previous studies have largely focused on Meis contribution to the development of non-neuronal tissues. By contrast, Meis function in the brain is not well understood. Here, we provide evidence for a dual role of the Meis family protein Meis2 in adult olfactory bulb (OB) neurogenesis. Meis2 is strongly expressed in neuroblasts of the subventricular zone (SVZ) and rostral migratory stream (RMS) and in some of the OB interneurons that are continuously replaced during adult life. Targeted manipulations with retroviral vectors expressing function-blocking forms or with small interfering RNAs demonstrated that Meis activity is cell-autonomously required for the acquisition of a general neuronal fate by SVZ-derived progenitors in vivo and in vitro. Additionally, Meis2 activity in the RMS is important for the generation of dopaminergic periglomerular neurons in the OB. Chromatin immunoprecipitation identified doublecortin and tyrosine hydroxylase as direct Meis targets in newly generated neurons and the OB, respectively. Furthermore, biochemical analyses revealed a previously unrecognized complex of Meis2 with Pax6 and Dlx2, two transcription factors involved in OB neurogenesis. The full pro-neurogenic activity of Pax6 in SVZ derived neural stem and progenitor cells requires the presence of Meis. Collectively, these results show that Meis2 cooperates with Pax6 in generic neurogenesis and dopaminergic fate specification in the adult SVZ-OB system.


Asunto(s)
Neuronas Dopaminérgicas/citología , Proteínas del Ojo/metabolismo , Proteínas de Homeodominio/metabolismo , Neurogénesis/fisiología , Bulbo Olfatorio/embriología , Factores de Transcripción Paired Box/metabolismo , Proteínas Represoras/metabolismo , Animales , Secuencia de Bases , Proliferación Celular , Neuronas Dopaminérgicas/metabolismo , Proteínas de Dominio Doblecortina , Proteínas de Homeodominio/genética , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Datos de Secuencia Molecular , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Neuropéptidos/metabolismo , Bulbo Olfatorio/citología , Bulbo Olfatorio/crecimiento & desarrollo , Factor de Transcripción PAX6 , Interferencia de ARN , ARN Interferente Pequeño/genética , Factores de Transcripción/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
6.
Proc Natl Acad Sci U S A ; 109(11): 4296-301, 2012 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-22371592

RESUMEN

Axonal transport deficits have been reported in many neurodegenerative conditions and are widely assumed to be an immediate causative step of axon and synapse loss. By imaging changes in axonal morphology and organelle transport over time in several animal models of amyotrophic lateral sclerosis (ALS), we now find that deficits in axonal transport of organelles (mitochondria, endosomes) and axon degeneration can evolve independently. This conclusion rests on the following results: (i) Axons can survive despite long-lasting transport deficits: In the SOD(G93A) model of ALS, transport deficits are detected soon after birth, months before the onset of axon degeneration. (ii) Transport deficits are not necessary for axon degeneration: In the SOD(G85R) model of ALS, motor axons degenerate, but transport is unaffected. (iii) Axon transport deficits are not sufficient to cause immediate degeneration: In mice that overexpress wild-type superoxide dismutase-1 (SOD(WT)), axons show chronic transport deficits, but survive. These data suggest that disturbances of organelle transport are not a necessary step in the emergence of motor neuron degeneration.


Asunto(s)
Esclerosis Amiotrófica Lateral/complicaciones , Esclerosis Amiotrófica Lateral/patología , Transporte Axonal , Degeneración Nerviosa/complicaciones , Degeneración Nerviosa/patología , Esclerosis Amiotrófica Lateral/enzimología , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Superóxido Dismutasa/metabolismo
7.
Development ; 138(6): 1057-68, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21343361

RESUMEN

Little is known about the intrinsic specification of adult neural stem cells (NSCs) and to what extent they depend on their local niche. To observe adult NSC division and lineage progression independent of their niche, we isolated cells from the adult mouse subependymal zone (SEZ) and cultured them at low density without growth factors. We demonstrate here that SEZ cells in this culture system are primarily neurogenic and that adult NSCs progress through stereotypic lineage trees consisting of asymmetric stem cell divisions, symmetric transit-amplifying divisions and final symmetric neurogenic divisions. Stem cells, identified by their astro/radial glial identity and their slow-dividing nature, were observed to generate asymmetrically and fast-dividing cells that maintained an astro/radial glia identity. These, in turn, gave rise to symmetrically and fast-dividing cells that lost glial hallmarks, but had not yet acquired neuronal features. The number of amplifying divisions was limited to a maximum of five in this system. Moreover, we found that cell growth correlated with the number of subsequent divisions of SEZ cells, with slow-dividing astro/radial glia exhibiting the most substantial growth prior to division. The fact that in the absence both of exogenously supplied growth factors and of signals provided by the local niche neurogenic lineage progression takes place in such stereotypic fashion, suggests that lineage progression is, to a significant degree, cell intrinsic or pre-programmed at the beginning of the lineage.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/fisiología , División Celular/fisiología , Linaje de la Célula/fisiología , Rastreo Celular/métodos , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Células Madre Adultas/efectos de los fármacos , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Astrocitos/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , División Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Péptidos y Proteínas de Señalización Intercelular/farmacología , Ratones , Sistema Nervioso/citología , Células-Madre Neurales/efectos de los fármacos , Factores de Tiempo
8.
Stem Cell Reports ; 18(12): 2418-2433, 2023 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-37995703

RESUMEN

Although adult subependymal zone (SEZ) neural stem cells mostly generate GABAergic interneurons, a small progenitor population expresses the proneural gene Neurog2 and produces glutamatergic neurons. Here, we determined whether Neurog2 could respecify SEZ neural stem cells and their progeny toward a glutamatergic fate. Retrovirus-mediated expression of Neurog2 induced the glutamatergic lineage markers TBR2 and TBR1 in cultured SEZ progenitors, which differentiated into functional glutamatergic neurons. Likewise, Neurog2-transduced SEZ progenitors acquired glutamatergic neuron hallmarks in vivo. Intriguingly, they failed to migrate toward the olfactory bulb and instead differentiated within the SEZ or the adjacent striatum, where they received connections from local neurons, as indicated by rabies virus-mediated monosynaptic tracing. In contrast, lentivirus-mediated expression of Neurog2 failed to reprogram early SEZ neurons, which maintained GABAergic identity and migrated to the olfactory bulb. Our data show that NEUROG2 can program SEZ progenitors toward a glutamatergic identity but fails to reprogram their neuronal progeny.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Células-Madre Neurales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neuronas/metabolismo , Células-Madre Neurales/metabolismo , Diferenciación Celular , Bulbo Olfatorio/metabolismo , Neurogénesis/fisiología
9.
Dev Cell ; 58(23): 2641-2651.e6, 2023 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-37890489

RESUMEN

Choroid plexuses (ChPs) produce cerebrospinal fluid and sense non-cell-autonomous stimuli to control the homeostasis of the central nervous system. They are mainly composed of epithelial multiciliated cells, whose development and function are still controversial. We have thus characterized the stepwise order of mammalian ChP epithelia cilia formation using a combination of super-resolution-microscopy approaches and mouse genetics. We show that ChP ciliated cells are built embryonically on a treadmill of spatiotemporally regulated events, starting with atypical centriole amplification and ending with the construction of nodal-like 9+0 cilia, characterized by both primary and motile features. ChP cilia undergo axoneme resorption at early postnatal stages through a microtubule destabilization process controlled by the microtubule-severing enzyme spastin and mitigated by polyglutamylation levels. Notably, this phenotype is preserved in humans, suggesting a conserved ciliary resorption mechanism in mammals.


Asunto(s)
Axonema , Cilios , Humanos , Ratones , Animales , Cilios/fisiología , Células Epiteliales/fisiología , Epitelio , Coroides , Mamíferos
10.
STAR Protoc ; 3(1): 101081, 2022 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-35059654

RESUMEN

This step-by-step protocol provides a fast and easy technique to label and/or genetically manipulate neural cells, achieved by intraventricular injection of viral vectors into neonatal mice under ultrasound guidance. Successful injection of adeno-associated viral vectors (AAV) induces neural transduction as fast as 3 days post injection (dpi) in both the central and peripheral nervous systems. Virally driven expression persists until early adulthood. The same setup enables injection of other viral vectors as well as intramuscular injection. For complete details on the use and execution of this protocol, please refer to Wang et al. (2021) and Brill et al. (2016).


Asunto(s)
Dependovirus , Vectores Genéticos , Animales , Dependovirus/genética , Inyecciones , Inyecciones Intraventriculares , Ratones , Neuronas/metabolismo
11.
Life Sci Alliance ; 5(11)2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35777956

RESUMEN

Ubiquilin-2 (UBQLN2) is a ubiquitin-binding protein that shuttles ubiquitinated proteins to proteasomal and autophagic degradation. UBQLN2 mutations are genetically linked to the neurodegenerative disorders amyotrophic lateral sclerosis and frontotemporal dementia (ALS/FTD). However, it remains elusive how UBQLN2 mutations cause ALS/FTD. Here, we systematically examined proteomic and transcriptomic changes in patient-derived lymphoblasts and CRISPR/Cas9-engineered HeLa cells carrying ALS/FTD UBQLN2 mutations. This analysis revealed a strong up-regulation of the microtubule-associated protein 1B (MAP1B) which was also observed in UBQLN2 knockout cells and primary rodent neurons depleted of UBQLN2, suggesting that a UBQLN2 loss-of-function mechanism is responsible for the elevated MAP1B levels. Consistent with MAP1B's role in microtubule binding, we detected an increase in total and acetylated tubulin. Furthermore, we uncovered that UBQLN2 mutations result in decreased phosphorylation of MAP1B and of the ALS/FTD-linked fused in sarcoma (FUS) protein at S439 which is critical for regulating FUS-RNA binding and MAP1B protein abundance. Together, our findings point to a deregulated UBQLN2-FUS-MAP1B axis that may link protein homeostasis, RNA metabolism, and cytoskeleton dynamics, three molecular pathomechanisms of ALS/FTD.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Esclerosis Amiotrófica Lateral , Proteínas Relacionadas con la Autofagia , Demencia Frontotemporal , Proteínas Asociadas a Microtúbulos , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Demencia Frontotemporal/genética , Demencia Frontotemporal/metabolismo , Células HeLa , Humanos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteómica , ARN/genética , ARN/metabolismo , Proteína FUS de Unión a ARN/genética , Proteína FUS de Unión a ARN/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
12.
Front Cell Dev Biol ; 10: 965382, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36393849

RESUMEN

Aging is a complex process characterized by several molecular and cellular imbalances. The composition and stability of the neuronal cytoskeleton is essential for the maintenance of homeostasis, especially in long neurites. Using human skin biopsies containing sensory axons from a cohort of healthy individuals, we investigate alterations in cytoskeletal content and sensory axon caliber during aging via quantitative immunostainings. Cytoskeletal components show an increase with aging in both sexes, while elevation in axon diameter is only evident in males. Transcriptomic data from aging males illustrate various patterns in gene expression during aging. Together, the data suggest gender-specific changes during aging in peripheral sensory axons, possibly influencing cytoskeletal functionality and axonal caliber. These changes may cumulatively increase susceptibility of aged individuals to neurodegenerative diseases.

13.
J Cell Biol ; 220(4)2021 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-33538762

RESUMEN

Neuronal remodeling and myelination are two fundamental processes during neurodevelopment. How they influence each other remains largely unknown, even though their coordinated execution is critical for circuit function and often disrupted in neuropsychiatric disorders. It is unclear whether myelination stabilizes axon branches during remodeling or whether ongoing remodeling delays myelination. By modulating synaptic transmission, cytoskeletal dynamics, and axonal transport in mouse motor axons, we show that local axon remodeling delays myelination onset and node formation. Conversely, glial differentiation does not determine the outcome of axon remodeling. Delayed myelination is not due to a limited supply of structural components of the axon-glial unit but rather is triggered by increased transport of signaling factors that initiate myelination, such as neuregulin. Further, transport of promyelinating signals is regulated via local cytoskeletal maturation related to activity-dependent competition. Our study reveals an axon branch-specific fine-tuning mechanism that locally coordinates axon remodeling and myelination.


Asunto(s)
Axones , Neuronas Motoras/metabolismo , Vaina de Mielina/metabolismo , Animales , Ratones , Ratones Transgénicos , Transmisión Sináptica
14.
J Neurosci ; 29(13): 4172-88, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19339612

RESUMEN

Adult neuronal precursors retain the remarkable capacity to migrate long distances from the posterior (subventricular zone) to the most anterior [olfactory bulb (OB)] parts of the brain. The knowledge about the mechanisms that keep neuronal precursors in the migratory stream and organize this long-distance migration is incomplete. Here we show that blood vessels precisely outline the migratory stream for new neurons in the adult mammalian forebrain. Real-time video imaging of cell migration in the acute slices demonstrate that neuronal precursors are retained in the migratory stream and guided into the OB by blood vessels that serve as a physical substrate for migrating neuroblasts. Our data suggest that endothelial cells of blood vessels synthesize brain-derived neurotrophic factor (BDNF) that fosters neuronal migration via p75NTR expressed on neuroblasts. Interestingly, GABA released from neuroblasts induces Ca(2+)-dependent insertion of high-affinity TrkB receptors on the plasma membrane of astrocytes that trap extracellular BDNF. We hypothesize that this renders BDNF unavailable for p75NTR-expressing migrating cells and leads to their entrance into the stationary period. Our findings provide new insights into the functional organization of substrates that facilitate the long-distance journey of adult neuronal precursors.


Asunto(s)
Células Madre Adultas/fisiología , Vasos Sanguíneos/fisiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Movimiento Celular/fisiología , Neuronas/fisiología , Prosencéfalo/fisiología , Transducción de Señal/fisiología , Animales , Astrocitos , Bicuculina/farmacología , Compuestos de Boro/farmacología , Factor Neurotrófico Derivado del Encéfalo/genética , Bromodesoxiuridina/metabolismo , Calcio/metabolismo , Movimiento Celular/genética , Células Cultivadas , Células Endoteliales/fisiología , Transportador 1 de Aminoácidos Excitadores/genética , Citometría de Flujo/métodos , Antagonistas del GABA/farmacología , Expresión Génica/fisiología , Proteína Ácida Fibrilar de la Glía/metabolismo , Glutamato Descarboxilasa/deficiencia , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía por Video/métodos , Neuronas/efectos de los fármacos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Prosencéfalo/citología , Transporte de Proteínas/efectos de los fármacos , ARN Interferente Pequeño/farmacología , Receptor trkB/metabolismo , Receptores de Factor de Crecimiento Nervioso/deficiencia , Transducción de Señal/genética , Técnicas de Cultivo de Tejidos , Ácido gamma-Aminobutírico/farmacología
15.
J Neurosci ; 28(2): 434-46, 2008 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-18184786

RESUMEN

In the mammalian brain, neurogenesis continues only in few regions of the forebrain. The molecular signals governing neurogenesis in these unique neurogenic niches, however, are still ill defined. Here, we show that bone morphogenic protein (BMP)-mediated signaling is active in adult neural stem cells and is crucial to initiate the neurogenic lineage in the adult mouse subependymal zone. Conditional deletion of Smad4 in adult neural stem cells severely impairs neurogenesis, and this is phenocopied by infusion of Noggin, an extracellular antagonist of BMP. Smad4 deletion in stem, but not progenitor cells, as well as Noggin infusion lead to an increased number of Olig2-expressing progeny that migrate to the corpus callosum and differentiate into oligodendrocytes. Transplantation experiments further verified the cell-autonomous nature of this phenotype. Thus, BMP-mediated signaling via Smad4 is required to initiate neurogenesis from adult neural stem cells and suppress the alternative fate of oligodendrogliogenesis.


Asunto(s)
Células Madre Adultas/fisiología , Proteínas Morfogenéticas Óseas/metabolismo , Proliferación Celular , Transducción de Señal/fisiología , Proteína Smad4/fisiología , Sistema de Transporte de Aminoácidos X-AG/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Bromodesoxiuridina/metabolismo , Proteínas Portadoras/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Trasplante de Células/métodos , Antagonistas de Estrógenos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/metabolismo , Factor de Transcripción 2 de los Oligodendrocitos , Transducción de Señal/efectos de los fármacos , Proteína Smad4/deficiencia , Tamoxifeno/farmacología , Factores de Transcripción/metabolismo
16.
J Neurosci ; 28(25): 6439-52, 2008 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-18562615

RESUMEN

Distinct olfactory bulb (OB) interneurons are thought to become specified depending on from which of the different subregions lining the lateral ventricle wall they originate, but the role of region-specific transcription factors (TFs) in the generation of OB interneurons diversity is still poorly understood. Despite the crucial roles of the Dlx family of TFs for patterning and neurogenesis in the ventral telencephalon during embryonic development, their role in adult neurogenesis has not yet been addressed. Here we show that in the adult brain, Dlx 1 and Dlx2 are expressed in progenitors of the lateral but not the dorsal subependymal zone (SEZ), thus exhibiting a striking regional specificity. Using retroviral vectors to examine the function of Dlx2 in a cell-autonomous manner, we demonstrate that this TF is necessary for neurogenesis of virtually all OB interneurons arising from the lateral SEZ. Beyond its function in generic neurogenesis, Dlx2 also plays a crucial role in neuronal subtype specification in the OB, promoting specification of adult-born periglomerular neurons (PGNs) toward a dopaminergic fate. Strikingly, Dlx2 requires interaction with Pax6, because Pax6 deletion blocks Dlx2-mediated PGN specification. Thus, Dlx2 wields a dual function by first instructing generic neurogenesis from adult precursors and subsequently specifying PGN subtypes in conjunction with Pax6.


Asunto(s)
Linaje de la Célula/genética , Proteínas del Ojo/genética , Proteínas de Homeodominio/genética , Neuronas/fisiología , Bulbo Olfatorio/metabolismo , Factores de Transcripción Paired Box/genética , Proteínas Represoras/genética , Factores de Transcripción/genética , Transcripción Genética/fisiología , Factores de Edad , Animales , Diferenciación Celular/genética , Células Cultivadas , Proteínas del Ojo/fisiología , Femenino , Proteínas de Homeodominio/fisiología , Humanos , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Bulbo Olfatorio/citología , Bulbo Olfatorio/crecimiento & desarrollo , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/fisiología , Embarazo , Proteínas Represoras/fisiología , Factores de Transcripción/fisiología
17.
Sci Rep ; 9(1): 15940, 2019 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-31685876

RESUMEN

Microtubule severing regulates cytoskeletal rearrangement underlying various cellular functions. Katanin, a heterodimer, consisting of catalytic (p60) and regulatory (p80) subunits severs dynamic microtubules to modulate several stages of cell division. The role of p60 katanin in the mammalian brain with respect to embryonic and adult neurogenesis is poorly understood. Here, we generated a Katna1 knockout mouse and found that consistent with a critical role of katanin in mitosis, constitutive homozygous Katna1 depletion is lethal. Katanin p60 haploinsufficiency induced an accumulation of neuronal progenitors in the subventricular zone during corticogenesis, and impaired their proliferation in the adult hippocampus dentate gyrus (DG) subgranular zone. This did not compromise DG plasticity or spatial and contextual learning and memory tasks employed in our study, consistent with the interpretation that adult neurogenesis may be associated with selective forms of hippocampal-dependent cognitive processes. Our data identify a critical role for the microtubule-severing protein katanin p60 in regulating neuronal progenitor proliferation in vivo during embryonic development and adult neurogenesis.


Asunto(s)
Diferenciación Celular , Katanina/genética , Microtúbulos/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis , Factores de Edad , Alelos , Animales , Diferenciación Celular/genética , Proliferación Celular , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Giro Dentado/embriología , Giro Dentado/metabolismo , Marcación de Gen , Haploinsuficiencia , Katanina/metabolismo , Aprendizaje , Memoria , Ratones , Ratones Noqueados , Neurogénesis/genética , Neuronas/citología , Neuronas/metabolismo , Organogénesis , Fenotipo
19.
Neuron ; 92(4): 845-856, 2016 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-27773584

RESUMEN

Developmental axon remodeling is characterized by the selective removal of branches from axon arbors. The mechanisms that underlie such branch loss are largely unknown. Additionally, how neuronal resources are specifically assigned to the branches of remodeling arbors is not understood. Here we show that axon branch loss at the developing mouse neuromuscular junction is mediated by branch-specific microtubule severing, which results in local disassembly of the microtubule cytoskeleton and loss of axonal transport in branches that will subsequently dismantle. Accordingly, pharmacological microtubule stabilization delays neuromuscular synapse elimination. This branch-specific disassembly of the cytoskeleton appears to be mediated by the microtubule-severing enzyme spastin, which is dysfunctional in some forms of upper motor neuron disease. Our results demonstrate a physiological role for a neurodegeneration-associated modulator of the cytoskeleton, reveal unexpected cell biology of branch-specific axon plasticity and underscore the mechanistic similarities of axon loss in development and disease.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Transporte Axonal , Microtúbulos/metabolismo , Unión Neuromuscular/metabolismo , Plasticidad Neuronal , Adenosina Trifosfatasas/genética , Animales , Citoesqueleto/metabolismo , Ratones , Ratones Noqueados , Enfermedad de la Neurona Motora/metabolismo , Espastina
20.
Neuron ; 82(3): 635-44, 2014 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-24811382

RESUMEN

In central mammalian neurons, activation of metabotropic glutamate receptor type1 (mGluR1) evokes a complex synaptic response consisting of IP3 receptor-dependent Ca(2+) release from internal Ca(2+) stores and a slow depolarizing potential involving TRPC3 channels. It is largely unclear how mGluR1 is linked to its downstream effectors. Here, we explored the role of stromal interaction molecule 1 (STIM1) in regulating neuronal Ca(2+) signaling and mGluR1-dependent synaptic transmission. By analyzing mouse cerebellar Purkinje neurons, we demonstrate that STIM1 is an essential regulator of the Ca(2+) level in neuronal endoplasmic reticulum Ca(2+) stores. Both mGluR1-dependent synaptic potentials and IP3 receptor-dependent Ca(2+) signals are strongly attenuated in the absence of STIM1. Furthermore, the Purkinje neuron-specific deletion of Stim1 causes impairments in cerebellar motor behavior. Together, our results demonstrate that in the mammalian nervous system STIM1 is a key regulator of intracellular Ca(2+) signaling, metabotropic glutamate receptor-dependent synaptic transmission, and motor coordination.


Asunto(s)
Señalización del Calcio/fisiología , Cerebelo/fisiología , Glicoproteínas de Membrana/fisiología , Actividad Motora/fisiología , Receptores de Glutamato Metabotrópico/fisiología , Transmisión Sináptica/fisiología , Animales , Canales de Calcio , Cerebelo/citología , Potenciales Postsinápticos Excitadores/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Neuronas/fisiología , Técnicas de Cultivo de Órganos , Molécula de Interacción Estromal 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA