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.
Dev Neurobiol ; 84(2): 74-92, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38509451

RESUMEN

The organization of neurons into distinct layers, known as lamination, is a common feature of the nervous system. This process, which arises from the direct coupling of neurogenesis and neuronal migration, plays a crucial role in the development of the cerebellum, a structure exhibiting a distinct folding cytoarchitecture with cells arranged in discrete layers. Disruptions to neuronal migration can lead to various neurodevelopmental disorders, highlighting the significance of understanding the molecular regulation of lamination. We report a role Mllt11/Af1q/Tcf7c (myeloid/lymphoid or mixed-lineage leukemia; translocated to chromosome 11/All1 fused gene from chromosome 1q, also known as Mllt11 transcriptional cofactor 7; henceforth referred to Mllt11) in the migration of cerebellar granule cells (GCs). We now show that Mllt11 plays a role in both the tangential and radial migration of GCs. Loss of Mllt11 led to an accumulation of GC precursors in the rhombic lip region and a reduction in the number of GCs successfully populating developing folia. Consequently, this results in smaller folia and an overall reduction in cerebellar size. Furthermore, analysis of the anchoring centers reveals disruptions in the perinatal folia cytoarchitecture, including alterations in the Bergmann glia fiber orientation and reduced infolding of the Purkinje cell plate. Lastly, we demonstrate that Mllt11 interacts with non-muscle myosin IIB (NMIIB) and Mllt11 loss-reduced NMIIB expression. We propose that the dysregulation of NMIIB underlies altered GC migratory behavior. Taken together, the findings reported herein demonstrate a role for Mllt11 in regulating neuronal migration within the developing cerebellum, which is necessary for its proper neuroanatomical organization.


Asunto(s)
Cerebelo , Estructuras Embrionarias , Metencéfalo/embriología , Neuronas , Embarazo , Femenino , Humanos , Neuronas/metabolismo , Neuroglía/metabolismo , Movimiento Celular/fisiología
2.
Dev Dyn ; 252(2): 305-319, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36131367

RESUMEN

BACKGROUND: The vertebrate retina is an organized laminar structure comprised of distinct cell types populating three nuclear layers. During development, each retinal cell type follows a stereotypical temporal order of genesis, differentiation, and migration, giving rise to its stratified organization. Once born, the precise positioning of cells along the apico-basal (radial) axis of the retina is critical for subsequent connections to form, relying on highly orchestrated migratory processes. While these processes are critical for visual function to arise, the regulators of cellular migration and retinal lamination remain largely unexplored. RESULTS: We report a role for a microtubule-interacting protein, Mllt11 (myeloid/lymphoid or mixed-lineage leukemia; translocated to chromosome 11/All1 fused gene from chromosome 1q) in mammalian retinal cell migration during retinogenesis. We show that Mllt11 loss-of-function in mouse retinal neuroblasts affected the migration of ganglion and amacrine cells into the ganglion cell layer and led to their aberrant accumulation in the inner nuclear and plexiform layers. CONCLUSIONS: We demonstrate a role for Mllt11 in neuroblast migration and formation of the ganglion cell layer of the retina.


Asunto(s)
Células Amacrinas , Retina , Animales , Ratones , Células Amacrinas/metabolismo , Factores de Transcripción/metabolismo , Diferenciación Celular , Movimiento Celular , Proteínas del Citoesqueleto , Mamíferos
3.
J Neurosci ; 42(19): 3931-3948, 2022 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-35379703

RESUMEN

The formation of connections within the mammalian neocortex is highly regulated by both extracellular guidance mechanisms and intrinsic gene expression programs. There are two types of cortical projection neurons (CPNs): those that project locally and interhemispherically and those that project to subcerebral structures such as the thalamus, hindbrain, and spinal cord. The regulation of cortical projection morphologies is not yet fully understood at the molecular level. Here, we report a role for Mllt11 (Myeloid/lymphoid or mixed-lineage leukemia; translocated to chromosome 11/All1 Fused Gene From Chromosome 1q) in the migration and neurite outgrowth of callosal projection neurons during mouse brain formation. We show that Mllt11 expression is exclusive to developing neurons and is enriched in the developing cortical plate (CP) during the formation of the superficial cortical layers. In cultured primary cortical neurons, Mllt11 is detected in varicosities and growth cones as well as the soma. Using conditional loss-of-function and gain-of-function analysis we show that Mllt11 is required for neuritogenesis and proper migration of upper layer CPNs. Loss of Mllt11 in the superficial cortex of male and female neonates leads to a severe reduction in fibers crossing the corpus callosum (CC), a progressive loss in the maintenance of upper layer projection neuron gene expression, and reduced complexity of dendritic arborization. Proteomic analysis revealed that Mllt11 associates with stabilized microtubules, and Mllt11 loss affected microtubule staining in callosal axons. Taken together, our findings support a role for Mllt11 in promoting the formation of mature upper-layer neuron morphologies and connectivity in the cerebral cortex.SIGNIFICANCE STATEMENT The regulation of cortical projection neuron (CPN) morphologies is an area of active investigation since the time of Cajal. Yet the molecular mechanisms of how the complex dendritic and axonal morphologies of projection neurons are formed remains incompletely understood. Although conditional mutagenesis analysis in the mouse, coupled with overexpression assays in the developing fetal brain, we show that a novel protein called Mllt11 is sufficient and necessary to regulate the dendritic and axonal characteristics of callosal projection neurons in the developing mammalian neocortex. Furthermore, we show that Mllt11 interacts with microtubules, likely accounting for its role in neuritogenesis.


Asunto(s)
Corteza Cerebral , Neocórtex , Proyección Neuronal , Proteínas Proto-Oncogénicas , Animales , Axones/fisiología , Corteza Cerebral/citología , Corteza Cerebral/fisiología , Cuerpo Calloso/fisiología , Femenino , Masculino , Ratones , Neocórtex/metabolismo , Vías Nerviosas/fisiología , Neuronas/fisiología , Proteómica , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/fisiología
4.
Genesis ; 60(1-2): e23464, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34897971

RESUMEN

Brain acid soluble protein 1 (BASP1; previously NAP22 or CAP23) is an actin-associating protein that is highly expressed in the nervous system throughout development. While its roles at the neuromuscular junction and in certain non-neuronal tissues have been previously characterized, its function in the early neural tube is unclear. Using in ovo electroporation in the chicken (Gallus gallus) embryonic neural tube, we show that BASP1 overexpression resulted in the appearance of ectopic neural progenitor cells within the marginal zone of the neural tube. BASP1 knockdown did not affect the position of neural progenitors but did alter the complexity of axons developing from differentiated neurons. This suggests a role for BASP1 in regulating the apical polarity of progenitor cells and axon trajectories from developing neurons.


Asunto(s)
Células-Madre Neurales , Tubo Neural , Actinas/genética , Actinas/metabolismo , Citoesqueleto/metabolismo , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Células-Madre Neurales/metabolismo , Neuronas
5.
Open Biol ; 11(5): 210042, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33947245

RESUMEN

The dorsal medial region of the developing mammalian telencephalon plays a central role in the patterning of the adjacent brain regions. This review describes the development of this specialized region of the vertebrate brain, called the cortical hem, and the formation of the various cells and structures it gives rise to, including the choroid plexus, Cajal-Retzius cells and the hippocampus. We highlight the ontogenic processes that create these different forebrain derivatives from their shared embryonic origin and discuss the key signalling pathways and molecules that influence the patterning of the cortical hem. These include BMP, Wnt, FGF and Shh signalling pathways acting with Homeobox factors to carve the medial telencephalon into district progenitor regions, which in turn give rise to the choroid plexus, dentate gyrus and hippocampus. We then link the formation of the lateral ventricle choroid plexus with embryonic and postnatal neurogenesis in the hippocampus.


Asunto(s)
Plexo Coroideo/embriología , Giro Dentado/embriología , Proteínas Hedgehog/metabolismo , Neurogénesis , Transducción de Señal , Animales , Humanos
6.
Neuroscience ; 458: 229-243, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33460728

RESUMEN

Crucial to an animal's movement through their environment and to the maintenance of their homeostatic physiology is the integration of sensory information. This is achieved by axons communicating from organs, muscle spindles and skin that connect to the sensory ganglia composing the peripheral nervous system (PNS), enabling organisms to collect an ever-constant flow of sensations and relay it to the spinal cord. The sensory system carries a wide spectrum of sensory modalities - from sharp pain to cool refreshing touch - traveling from the periphery to the spinal cord via the dorsal root ganglia (DRG). This review covers the origins and development of the DRG and the cells that populate it, and focuses on how sensory connectivity to the spinal cord is achieved by the diverse developmental and molecular processes that control axon guidance in the trunk sensory system. We also describe convergences and differences in sensory neuron formation among different vertebrate species to gain insight into underlying developmental mechanisms.


Asunto(s)
Orientación del Axón , Ganglios Espinales , Animales , Axones , Células Receptoras Sensoriales , Médula Espinal , Vertebrados
7.
Development ; 147(19)2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32907847

RESUMEN

Pattern formation is influenced by transcriptional regulation as well as by morphogenetic mechanisms that shape organ primordia, although factors that link these processes remain under-appreciated. Here we show that, apart from their established transcriptional roles in pattern formation, IRX3/5 help to shape the limb bud primordium by promoting the separation and intercalation of dividing mesodermal cells. Surprisingly, IRX3/5 are required for appropriate cell cycle progression and chromatid segregation during mitosis, possibly in a nontranscriptional manner. IRX3/5 associate with, promote the abundance of, and share overlapping functions with co-regulators of cell division such as the cohesin subunits SMC1, SMC3, NIPBL and CUX1. The findings imply that IRX3/5 coordinate early limb bud morphogenesis with skeletal pattern formation.


Asunto(s)
Cromátides/metabolismo , Proteínas de Homeodominio/metabolismo , Esbozos de los Miembros/embriología , Esbozos de los Miembros/metabolismo , Factores de Transcripción/metabolismo , Animales , Western Blotting , Segregación Cromosómica/genética , Segregación Cromosómica/fisiología , Femenino , Técnica del Anticuerpo Fluorescente , Células HEK293 , Proteínas de Homeodominio/genética , Humanos , Inmunoprecipitación , Espectrometría de Masas , Ratones , Mitosis/genética , Mitosis/fisiología , Embarazo , RNA-Seq , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción/genética
8.
Mech Dev ; 160: 103577, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31634536

RESUMEN

The formation of the vertebrate nervous system depends on the complex interplay of morphogen signaling pathways and cell cycle progression to establish distinct cell fates. The Sonic hedgehog (Shh) signaling pathway is well understood to promote ventral cell fates in the developing spinal cord. A key regulator of Shh signaling is its receptor Patched1 (Ptch1). However, because the Ptch1 null mutation is lethal early in mouse embryogenesis, its role in controlling cell cycle progression, neurogenesis, and axon guidance in the developing spinal cord is not fully understood. An allele of Ptch1 called Wiggable (Ptch1Wig), which was previously shown to enhance Shh signaling, was used to test its ability to regulate neurogenesis and proliferation in the developing spinal cord. Ptch1Wig/Wig mutants displayed enhanced ventral proneural gene activation, and aberrant proliferation of the neural tube and floor plate cells, the latter normally being a quiescent population. The expression of the cell cycle regulators p27Kip1 and p57Kip2 were expanded in Ptch1Wig/Wig mutant spinal cords, as was the number of mitotic and S-phase nuclei, suggesting enhanced cell cycle progression. However, Ptch1Wig/Wig mutants also showed enhanced apoptosis in the ventral embryonic spinal cord, which resulted in thinner spinal cords at later embryonic stages. Commissural axons largely failed to cross the floor plate of Ptch1Wig/Wig mutant embryos, suggesting enhanced Shh signaling in these mutants led to a dorsal expansion of the chemoattraction front. These findings are consistent with a role of Ptch1 in regulating neurogenesis and proliferation of neural progenitors, and in restricting the influence of Shh signaling in commissural axon guidance to the floor plate.


Asunto(s)
Orientación del Axón , Diferenciación Celular , Embrión de Mamíferos/citología , Proteínas Hedgehog/metabolismo , Neurogénesis , Receptor Patched-1/metabolismo , Médula Espinal/citología , Médula Espinal/embriología , Animales , Ciclo Celular , Proliferación Celular , Ratones , Mutación/genética , Tubo Neural/embriología , Tubo Neural/metabolismo , Células Neuroepiteliales/citología , Células Neuroepiteliales/metabolismo
9.
Dev Dyn ; 248(7): 506-513, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31131952

RESUMEN

The cerebellum coordinates vestibular input into the hindbrain to control balance and movement, and its anatomical complexity is increasingly viewed as a high-throughput processing center for sensory and cognitive functions. Cerebellum development however is relatively simple, and arises from a specialized structure in the anterior hindbrain called the rhombic lip, which along with the ventricular zone of the rostral-most dorsal hindbrain region, give rise to the distinct cell types that constitute the cerebellum. Granule cells, being the most numerous cell types, arise from the rhombic lip and form a dense and distinct layer of the cerebellar cortex. In this short review, we describe the various strategies used by amniotes and anamniotes to generate and diversify granule cell types during cerebellar development.


Asunto(s)
Cerebelo/citología , Animales , Diferenciación Celular , Cerebelo/crecimiento & desarrollo , Humanos , Neocórtex/citología , Rombencéfalo
10.
Dev Dyn ; 247(1): 170-184, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28891097

RESUMEN

BACKGROUND: Vertebrate spinal cord development requires Sonic Hedgehog (Shh) signaling from the floorplate and notochord, where it is thought to act in concentration dependent manner to pattern distinct cell identities along the ventral-to-dorsal axis. While in vitro experiments demonstrate naïve neural tissues are sensitive to small changes in Shh levels, genetic studies illustrate that some degree of ventral patterning can occur despite significant perturbations in Shh signaling. Consequently, the mechanistic relationship between Shh morphogen levels and acquisition of distinct cell identities remains unclear. RESULTS: We addressed this using Hedgehog acetyltransferase (HhatCreface ) and Wiggable mouse mutants. Hhat encodes a palmitoylase required for the secretion of Hedgehog proteins and formation of the Shh gradient. In its absence, the spinal cord develops without floorplate cells and V3 interneurons. Wiggable is an allele of the Shh receptor Patched1 (Ptch1Wig ) that is unable to inhibit Shh signal transduction, resulting in expanded ventral progenitor domains. Surprisingly, HhatCreface/Creface ; Ptch1Wig/Wig double mutants displayed fully restored ventral patterning despite an absence of Shh secretion from the floorplate. CONCLUSIONS: The full range of neuronal progenitor types can be generated in the absence of a Shh gradient provided pathway repression is dampened, illustrating the complexity of morphogen dynamics in vertebrate patterning. Developmental Dynamics 247:170-184, 2018. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Tipificación del Cuerpo/genética , Proteínas Hedgehog/metabolismo , Tubo Neural/embriología , Transducción de Señal/fisiología , Médula Espinal/embriología , Aciltransferasas/genética , Aciltransferasas/metabolismo , Animales , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/genética , Ratones Transgénicos , Tubo Neural/metabolismo , Receptor Patched-1/genética , Receptor Patched-1/metabolismo , Médula Espinal/metabolismo
11.
Cold Spring Harb Protoc ; 2017(6): pdb.prot094011, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28572189

RESUMEN

Sectioning with a vibrating microtome (vibratome) is a valuable procedure for generating thick sections that can be used for immunohistochemistry and in situ hybridization. It is particularly useful for revealing histological and 3D detail in tissues and embryos that have been subjected to various whole-mount histological procedures such as ß-galactosidase and alkaline phosphatase staining, and fluorescent and DAB (diaminobenzidine) immunostaining. Vibratome sectioning does not involve any harsh organic solvents and is therefore suited for processing specimens stained with fluorescent antibodies or dyes. In addition, live tissue can be sectioned for subsequent culture, manipulation, and live imaging. Very few materials are required for vibratome sectioning, and it is a relatively straightforward procedure that is quickly mastered. This protocol describes basic vibratome sectioning that can be used for any staining procedure typically used in histology.


Asunto(s)
Microtomía/métodos , Animales , Técnicas Histológicas/métodos , Humanos , Coloración y Etiquetado/métodos
12.
Dev Dyn ; 245(8): 881-96, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27198720

RESUMEN

BACKGROUND: The rhombic lip (RL), a germinal zone in the developing hindbrain, gives rise to all of the excitatory neurons of the cerebellum. It is presently unclear what factors distinguish between RL progenitor pools and play a role in differentiating the multiple cell types that arise from this region. The transcription factor Cux2 has been shown to play important roles in proliferation and differentiation of distinct neuronal populations during embryogenesis, but its role in cerebellar fate restriction is unknown. RESULTS: Through expression analysis and genetic fate mapping studies we show that Cux2 is expressed in the RL of the fetal brain and is restricted to a pool of cerebellar granule cell precursors and unipolar brush cells. This restriction was remarkably specific because regardless of the timing of Cux2 reporter gene activation in the RL, only granule cell layer derivatives were labeled. However, the overexpression of Cux2 in naïve hindbrain tissue was insufficient to force progenitor cells to adopt a granule cell fate. CONCLUSIONS: Our results suggest that Cux2 delineates the pool of cerebellar granule cell layer progenitors from other RL and ventricular zone derivatives, and plays a role in fate restricting, but not differentiating, this population. Developmental Dynamics 245:881-896, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Neuronas/citología , Neuronas/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proliferación Celular/genética , Proliferación Celular/fisiología , Cerebelo/citología , Cerebelo/metabolismo , Embrión de Pollo , Proteínas de Homeodominio/genética , Ratones , Rombencéfalo/citología , Rombencéfalo/metabolismo , Células Madre/citología , Células Madre/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
13.
J Biol Chem ; 290(37): 22520-31, 2015 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-26221032

RESUMEN

CUX1 and CUX2 proteins are characterized by the presence of three highly similar regions called Cut repeats 1, 2, and 3. Although CUX1 is ubiquitously expressed, CUX2 plays an important role in the specification of neuronal cells and continues to be expressed in postmitotic neurons. Cut repeats from the CUX1 protein were recently shown to stimulate 8-oxoguanine DNA glycosylase 1 (OGG1), an enzyme that removes oxidized purines from DNA and introduces a single strand break through its apurinic/apyrimidinic lyase activity to initiate base excision repair. Here, we investigated whether CUX2 plays a similar role in the repair of oxidative DNA damage. Cux2 knockdown in embryonic cortical neurons increased levels of oxidative DNA damage. In vitro, Cut repeats from CUX2 increased the binding of OGG1 to 7,8-dihydro-8-oxoguanine-containing DNA and stimulated both the glycosylase and apurinic/apyrimidinic lyase activities of OGG1. Genetic inactivation in mouse embryo fibroblasts or CUX2 knockdown in HCC38 cells delayed DNA repair and increased DNA damage. Conversely, ectopic expression of Cut repeats from CUX2 accelerated DNA repair and reduced levels of oxidative DNA damage. These results demonstrate that CUX2 functions as an accessory factor that stimulates the repair of oxidative DNA damage. Neurons produce a high level of reactive oxygen species because of their dependence on aerobic oxidation of glucose as their source of energy. Our results suggest that the persistent expression of CUX2 in postmitotic neurons contributes to the maintenance of genome integrity through its stimulation of oxidative DNA damage repair.


Asunto(s)
Corteza Cerebral/metabolismo , Daño del ADN , Reparación del ADN , Proteínas de Homeodominio/metabolismo , Neuronas/metabolismo , Animales , Línea Celular , Corteza Cerebral/citología , ADN Glicosilasas/genética , ADN Glicosilasas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Ratones , Neuronas/citología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oxidación-Reducción , Ratas , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
14.
PLoS One ; 10(3): e0120821, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25799573

RESUMEN

Cranial nerves govern sensory and motor information exchange between the brain and tissues of the head and neck. The cranial nerves are derived from two specialized populations of cells, cranial neural crest cells and ectodermal placode cells. Defects in either cell type can result in cranial nerve developmental defects. Although several signaling pathways are known to regulate cranial nerve formation our understanding of how intercellular signaling between neural crest cells and placode cells is coordinated during cranial ganglia morphogenesis is poorly understood. Sonic Hedgehog (Shh) signaling is one key pathway that regulates multiple aspects of craniofacial development, but whether it co-ordinates cranial neural crest cell and placodal cell interactions during cranial ganglia formation remains unclear. In this study we examined a new Patched1 (Ptch1) loss-of-function mouse mutant and characterized the role of Ptch1 in regulating Shh signaling during cranial ganglia development. Ptch1(Wig/ Wig) mutants exhibit elevated Shh signaling in concert with disorganization of the trigeminal and facial nerves. Importantly, we discovered that enhanced Shh signaling suppressed canonical Wnt signaling in the cranial nerve region. This critically affected the survival and migration of cranial neural crest cells and the development of placodal cells as well as the integration between neural crest and placodes. Collectively, our findings highlight a novel and critical role for Shh signaling in cranial nerve development via the cross regulation of canonical Wnt signaling.


Asunto(s)
Nervios Craneales/embriología , Proteínas Hedgehog/metabolismo , Vía de Señalización Wnt , Animales , Muerte Celular , Movimiento Celular , Ectodermo/citología , Nervio Facial/embriología , Ratones , Cresta Neural/citología , Receptores Patched , Receptor Patched-1 , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Eliminación de Secuencia , Nervio Trigémino/embriología
15.
Hippocampus ; 25(2): 253-67, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25252086

RESUMEN

The hippocampus arises from the medial region of the subventricular (SVZ) within the telencephalon. It is one of two regions in the postnatal brain that harbors neural progenitors (NPs) capable of giving rise to new neurons. Neurogenesis in the hippocampus is restricted to the subgranular zone (SGZ) of the dentate gyrus (DG) where it contributes to the generation of granule cell layer (gcl) neurons. It is thought that SGZ progenitors are heterogeneous, differing in their morphology, expression profiles, and developmental potential, however it is currently unknown whether they display differences in their developmental origins and cell fate-restriction in the DG. Here we demonstrate that Cux2 is a marker for SGZ progenitors and nascent granule cell neurons in the perinatal brain. Cux2 was expressed in the presumptive hippocampal forming region of the embryonic forebrain from E14.5 onwards. At fetal stages, Cux2 was expressed in early-forming Prox1(+) granule cell neurons as well as the SVZ of the DG germinal matrix. In the postnatal brain, Cux2 was expressed in several types of progenitors in the SGZ of the DG, including Nestin/Sox2 double-positive radial glia, Sox2(+) cells that lacked a radial glial process, DCX(+) neuroblasts, and Calretinin-expressing nascent neurons. Another domain characterized by a low level of Cux2 expression emerged in Calbindin(+) neurons of the developing DG blades. We used Cux2-Cre mice in genetic fate-mapping studies and showed almost exclusive labeling of Calbindin-positive gcl neurons, but not in any progenitor cell types or astroglia. This suggests that Cux2(+) progenitors directly differentiate into gcl neurons and do not self-renew. Interestingly, developmental profiling of cell fate revealed an outside-in formation of gcl neurons in the DG, likely reflecting the activity of Cux2 in the germinative matrices during DG formation and maturation. However, DG morphogenesis proceeded largely normally in hypomorphic Cux2 mutants lacking Cux2 expression. Taken together we conclude that Cux2 expression reflects hippocampal neurogenesis and identifies non-self-renewing NPs in the SGZ.


Asunto(s)
Hipocampo/fisiología , Proteínas de Homeodominio/metabolismo , Células-Madre Neurales/fisiología , Nicho de Células Madre/fisiología , Animales , Calbindinas/metabolismo , Proteína Doblecortina , Hipocampo/citología , Hipocampo/crecimiento & desarrollo , Proteínas de Homeodominio/genética , Inmunohistoquímica , Ratones Endogámicos C57BL , Ratones Transgénicos , Nestina/metabolismo , Células-Madre Neurales/citología , Neurogénesis/fisiología , Neuroglía/citología , Neuroglía/fisiología , Neuronas/citología , Neuronas/fisiología , Factores de Transcripción SOXB1/metabolismo
16.
Gene Expr Patterns ; 15(2): 80-7, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24839873

RESUMEN

Myeloid/lymphoid or mixed-lineage leukemia; translocated to chromosome 11 or ALL1 fused from chromosome 1q (MLLT11/AF1q) is a highly conserved 90 amino acid protein that functions in hematopoietic differentiation. Its translocation to the Trithorax locus has been implicated in malignancies of the hematopoietic system. However, the spatio-temporal profile of MLLT11 expression during embryonic development has not been characterized. Here we show that MLLT11 has a remarkably specific expression pattern in the developing central and peripheral nervous system. We find high levels of MLLT11 transcript and protein expression in the developing marginal zone of the cortex and spinal cord. MLLT11 co-localized with Tbr2 in the developing subplate region of the cortex and expanded to encompass the cortical plate at late fetal stages. Expression in the peripheral nervous system initiated at E9.5 in the facio-acoustic cranial ganglia and elaborated to identify all the cranio-facial and dorsal root ganglia by E10.5. We also observed expression in the eye and gastrointestinal tract, where MLLT11 transcripts localized to Tuj1-positive inner retinal layer and autonomic neurons, respectively. Altogether these results show that MLLT11 is a pan-neuronal marker, suggesting a role in neural differentiation in the central nervous system and neural crest-cell derived peripheral ganglia.


Asunto(s)
Corteza Cerebral/embriología , Regulación del Desarrollo de la Expresión Génica , Neuronas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Sistema Nervioso Autónomo/embriología , Linaje de la Célula , Sistema Nervioso Central/embriología , Ganglios Espinales/embriología , Perfilación de la Expresión Génica , Inmunohistoquímica , Hibridación in Situ , Ratones , Neurogénesis , Sistema Nervioso Periférico/embriología , Médula Espinal/embriología
17.
J Clin Invest ; 124(4): 1660-71, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24590292

RESUMEN

Cleft lip, which results from impaired facial process growth and fusion, is one of the most common craniofacial birth defects. Many genes are known to be involved in the etiology of this disorder; however, our understanding of cleft lip pathogenesis remains incomplete. In the present study, we uncovered a role for sonic hedgehog (SHH) signaling during lip fusion. Mice carrying compound mutations in hedgehog acyltransferase (Hhat) and patched1 (Ptch1) exhibited perturbations in the SHH gradient during frontonasal development, which led to hypoplastic nasal process outgrowth, epithelial seam persistence, and cleft lip. Further investigation revealed that enhanced SHH signaling restricts canonical WNT signaling in the lambdoidal region by promoting expression of genes encoding WNT inhibitors. Moreover, reduction of canonical WNT signaling perturbed p63/interferon regulatory factor 6 (p63/IRF6) signaling, resulting in increased proliferation and decreased cell death, which was followed by persistence of the epithelial seam and cleft lip. Consistent with our results, mutations in genes that disrupt SHH and WNT signaling have been identified in both mice and humans with cleft lip. Collectively, our data illustrate that altered SHH signaling contributes to the etiology and pathogenesis of cleft lip through antagonistic interactions with other gene regulatory networks, including the canonical WNT and p63/IRF6 signaling pathways.


Asunto(s)
Labio Leporino/etiología , Proteínas Hedgehog/metabolismo , Proteínas Wnt/metabolismo , Aciltransferasas/genética , Aciltransferasas/metabolismo , Animales , Muerte Celular , Proliferación Celular , Labio Leporino/genética , Labio Leporino/metabolismo , Femenino , Redes Reguladoras de Genes , Proteínas Hedgehog/genética , Humanos , Factores Reguladores del Interferón/metabolismo , Masculino , Ratones , Ratones Mutantes , Ratones Transgénicos , Mutación , Receptores Patched , Receptor Patched-1 , Fosfoproteínas/metabolismo , Embarazo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Proteínas Wnt/antagonistas & inhibidores , Proteínas Wnt/genética
18.
Dev Biol ; 388(1): 35-47, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24512687

RESUMEN

Signaling pathways and transcription factors are crucial regulators of vertebrate neurogenesis, exerting their function in a spatial and temporal manner. Despite recent advances in our understanding of the molecular regulation of embryonic neurogenesis, little is known regarding how different signaling pathways interact to tightly regulate this process during the development of neuroepithelia. To address this, we have investigated the events lying upstream and downstream of a key neurogenic factor, the Cut-like homeodomain transcription factor-2 (Cux2), during embryonic neurogenesis in chick and mouse. By using the olfactory epithelium as a model for neurogenesis we have analyzed mouse embryos deficient in Cux2, as well as chick embryos exposed to Cux2 silencing (si) RNA or a Cux2 over-expression construct. We provide evidence that enhanced BMP activity increases Cux2 expression and suppresses olfactory neurogenesis in the chick olfactory epithelium. In addition, our results show that up-regulation of Cux2, either BMP-induced or ectopically over-expressed, reduce Delta1 expression and suppress proliferation. Interestingly, the loss of Cux2 activity, using mutant mice or siRNA in chick, also diminishes neurogenesis, Notch activity and cell proliferation in the olfactory epithelium. Our results suggest that controlled low levels of Cux2 activity are necessary for proper Notch signaling, maintenance of the proliferative pool and ongoing neurogenesis in the olfactory epithelium. Thus, we demonstrate a novel conserved mechanism in vertebrates in which levels of Cux2 activity play an important role for ongoing neurogenesis in the olfactory epithelium.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/fisiología , Neurogénesis/fisiología , Mucosa Olfatoria/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Linaje de la Célula , Proliferación Celular , Embrión de Pollo , Silenciador del Gen , Inmunohistoquímica , Hibridación in Situ , Ratones , Neuronas/metabolismo , ARN Interferente Pequeño/metabolismo , Receptores Notch/metabolismo
19.
PLoS Genet ; 8(10): e1002927, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23055936

RESUMEN

Holoprosencephaly (HPE) is a failure of the forebrain to bifurcate and is the most common structural malformation of the embryonic brain. Mutations in SHH underlie most familial (17%) cases of HPE; and, consistent with this, Shh is expressed in midline embryonic cells and tissues and their derivatives that are affected in HPE. It has long been recognized that a graded series of facial anomalies occurs within the clinical spectrum of HPE, as HPE is often found in patients together with other malformations such as acrania, anencephaly, and agnathia. However, it is not known if these phenotypes arise through a common etiology and pathogenesis. Here we demonstrate for the first time using mouse models that Hedgehog acyltransferase (Hhat) loss-of-function leads to holoprosencephaly together with acrania and agnathia, which mimics the severe condition observed in humans. Hhat is required for post-translational palmitoylation of Hedgehog (Hh) proteins; and, in the absence of Hhat, Hh secretion from producing cells is diminished. We show through downregulation of the Hh receptor Ptch1 that loss of Hhat perturbs long-range Hh signaling, which in turn disrupts Fgf, Bmp and Erk signaling. Collectively, this leads to abnormal patterning and extensive apoptosis within the craniofacial primordial, together with defects in cartilage and bone differentiation. Therefore our work shows that Hhat loss-of-function underscrores HPE; but more importantly it provides a mechanism for the co-occurrence of acrania, holoprosencephaly, and agnathia. Future genetic studies should include HHAT as a potential candidate in the etiology and pathogenesis of HPE and its associated disorders.


Asunto(s)
Aciltransferasas/genética , Proteínas Hedgehog/metabolismo , Holoprosencefalia/genética , Holoprosencefalia/metabolismo , Anomalías Maxilomandibulares/genética , Anomalías Maxilomandibulares/metabolismo , Mutación , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo , Transducción de Señal , Aciltransferasas/metabolismo , Animales , Apoptosis/genética , Expresión Génica , Holoprosencefalia/embriología , Anomalías Maxilomandibulares/embriología , Ratones , Ratones Transgénicos , Cresta Neural/embriología , Cresta Neural/metabolismo , Defectos del Tubo Neural/embriología , Receptores Patched , Receptor Patched-1 , Fenotipo , Receptores de Superficie Celular/metabolismo
20.
Front Physiol ; 2: 16, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21516249

RESUMEN

Embryonic cortical neural stem cells are self-renewing progenitors that can differentiate into neurons and glia. We generated neurospheres from the developing cerebral cortex using a mouse genetic model that allows for lineage selection and found that the self-renewing neural stem cells are restricted to Sox2 expressing cells. Under normal conditions, embryonic cortical neurospheres are heterogeneous with regard to Sox2 expression and contain astrocytes, neural stem cells, and neural progenitor cells sufficiently plastic to give rise to neural crest cells when transplanted into the hindbrain of E1.5 chick and E8 mouse embryos. However, when neurospheres are maintained under lineage selection, such that all cells express Sox2, neural stem cells maintain their Pax6(+) cortical radial glia identity and exhibit a more restricted fate in vitro and after transplantation. These data demonstrate that Sox2 preserves the cortical identity and regulates the plasticity of self-renewing Pax6(+) radial glia cells.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...