Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 19(8): e1010874, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37594984

RESUMEN

In the mammalian cerebral cortex, the hippocampal primordium (Hcp) occupies a discrete position in the dorsal telencephalic neuroepithelium adjacent to the neocortical primordium (Ncp). We examined transcriptomic and chromatin-level features that distinguish the Hcp from the Ncp in the mouse during the early neurogenic period, embryonic day (E)12.5. ATAC-seq revealed that the Hcp was more accessible than the Ncp at this stage. Motif analysis of the differentially accessible loci in these tissues revealed LHX2 as a candidate transcription factor for modulating gene regulatory networks (GRNs). We analyzed LHX2 occupancy profiles and compared these with transcriptomic data from control and Lhx2 mutant Hcp and Ncp at E12.5. Our results revealed that LHX2 directly regulates distinct genes in the Hcp and Ncp within a set of common pathways that control fundamental aspects of development namely pluripotency, axon pathfinding, Wnt, and Hippo signaling. Loss of Lhx2 caused a decrease in accessibility, specifically in hippocampal chromatin, suggesting that this factor may play a unique role in hippocampal development. We identified 14 genes that were preferentially enriched in the Hcp, for which LHX2 regulates both chromatin accessibility and mRNA expression, which have not thus far been examined in hippocampal development. Together, these results provide mechanistic insight into how LHX2 function in the Hcp may contribute to the process by which the hippocampus acquires features distinct from the neocortex.


Asunto(s)
Cromatina , Neocórtex , Animales , Ratones , Hipocampo , Proteínas con Homeodominio LIM , Mamíferos , Factores de Transcripción , Transcriptoma
2.
Development ; 149(21)2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36196585

RESUMEN

The dentate gyrus, a gateway for input to the hippocampal formation, arises from progenitors in the medial telencephalic neuroepithelium adjacent to the cortical hem. Dentate progenitors navigate a complex migratory path guided by two cell populations that arise from the hem, the fimbrial glia and Cajal-Retzius (CR) cells. As the hem expresses multiple Wnt genes, we examined whether ß-catenin, which mediates canonical Wnt signaling and also participates in cell adhesion, is necessary for the development of hem-derived lineages. We report that, in mice, the fimbrial glial scaffold is disorganized and CR cells are mispositioned upon hem-specific disruption of ß-catenin. Consequently, the dentate migratory stream is severely affected, and the dentate gyrus fails to form. Using selective Cre drivers, we further determined that ß-catenin function is required in the fimbrial glial scaffold, but not in the CR cells, for guiding the dentate migration. Our findings highlight a primary requirement for ß-catenin for the organization of the fimbrial scaffold and a secondary role for this factor in dentate gyrus morphogenesis.


Asunto(s)
Giro Dentado , Morfogénesis , beta Catenina , Animales , Ratones , beta Catenina/metabolismo , Giro Dentado/metabolismo , Hipocampo/metabolismo , Morfogénesis/genética , Neuroglía/metabolismo , Neuronas/metabolismo
3.
Development ; 147(20)2020 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-32994168

RESUMEN

The protein co-factor Ldb1 regulates cell fate specification by interacting with LIM-homeodomain (LIM-HD) proteins in a tetrameric complex consisting of an LDB:LDB dimer that bridges two LIM-HD molecules, a mechanism first demonstrated in the Drosophila wing disc. Here, we demonstrate conservation of this interaction in the regulation of mammalian hippocampal development, which is profoundly defective upon loss of either Lhx2 or Ldb1 Electroporation of a chimeric construct that encodes the Lhx2-HD and Ldb1-DD (dimerization domain) in a single transcript cell-autonomously rescues a comprehensive range of hippocampal deficits in the mouse Ldb1 mutant, including the acquisition of field-specific molecular identity and the regulation of the neuron-glia cell fate switch. This demonstrates that the LHX:LDB complex is an evolutionarily conserved molecular regulatory device that controls complex aspects of regional cell identity in the developing brain.


Asunto(s)
Linaje de la Célula , Secuencia Conservada , Proteínas de Unión al ADN/genética , Evolución Molecular , Hipocampo/citología , Proteínas con Dominio LIM/genética , Proteínas con Homeodominio LIM/genética , Factores de Transcripción/genética , Animales , Tipificación del Cuerpo , Proteínas de Unión al ADN/metabolismo , Proteínas con Dominio LIM/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Ratones , Mutación/genética , Neurogénesis , Neuroglía/citología , Neuroglía/metabolismo , Unión Proteica , Factores de Transcripción/metabolismo
4.
J Neurosci ; 41(32): 6822-6835, 2021 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-34193558

RESUMEN

The cortical subplate is critical in regulating the entry of thalamocortical sensory afferents into the cortex. These afferents reach the subplate at embryonic day (E)15.5 in the mouse, but "wait" for several days, entering the cortical plate postnatally. We report that when transcription factor LHX2 is lost in E11.5 cortical progenitors, which give rise to subplate neurons, thalamocortical afferents display premature, exuberant ingrowth into the E15.5 cortex. Embryonic mutant subplate neurons are correctly positioned below the cortical plate, but they display an altered transcriptome and immature electrophysiological properties during the waiting period. The sensory thalamus in these cortex-specific Lhx2 mutants displays atrophy and by postnatal day (P) 7, sensory innervation to the cortex is nearly eliminated leading to a loss of the somatosensory barrels. Strikingly, these phenotypes do not manifest if LHX2 is lost in postmitotic subplate neurons, and the transcriptomic dysregulation in the subplate resulting from postmitotic loss of LHX2 is vastly distinct from that seen when LHX2 is lost in progenitors. These results demonstrate a mechanism operating in subplate progenitors that has profound consequences on the growth of thalamocortical axons into the cortex.SIGNIFICANCE STATEMENT Thalamocortical nerves carry sensory information from the periphery to the cortex. When they first grow into the embryonic cortex, they "wait" at the subplate, a structure critical for the guidance and eventual connectivity of thalamic axons with their cortical targets. How the properties of subplate neurons are regulated is unclear. We report that transcription factor LHX2 is required in the progenitor "mother" cells of the cortical primordium when they are producing their "daughter" subplate neurons, in order for the thalamocortical pathway to wait at the subplate. Without LHX2 function in subplate progenitors, thalamocortical axons grow past the subplate, entering the cortical plate prematurely. This is followed by their eventual attrition and, consequently, a profound loss of sensory innervation of the mature cortex.


Asunto(s)
Encéfalo/embriología , Células-Madre Neurales/citología , Neurogénesis/fisiología , Neuronas Aferentes/citología , Animales , Movimiento Celular/fisiología , Femenino , Proteínas con Homeodominio LIM/metabolismo , Masculino , Ratones , Vías Nerviosas/embriología , Células-Madre Neurales/metabolismo , Neuronas Aferentes/metabolismo , Factores de Transcripción/metabolismo
5.
Development ; 145(1)2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29229772

RESUMEN

During forebrain development, a telencephalic organizer called the cortical hem is crucial for inducing hippocampal fate in adjacent cortical neuroepithelium. How the hem is restricted to its medial position is therefore a fundamental patterning issue. Here, we demonstrate that Foxg1-Lhx2 interactions are crucial for the formation of the hem. Loss of either gene causes a region of the cortical neuroepithelium to transform into hem. We show that FOXG1 regulates Lhx2 expression in the cortical primordium. In the absence of Foxg1, the presence of Lhx2 is sufficient to suppress hem fate, and hippocampal markers appear selectively in Lhx2-expressing regions. FOXG1 also restricts the temporal window in which loss of Lhx2 results in a transformation of cortical primordium into hem. Therefore, Foxg1 and Lhx2 form a genetic hierarchy in the spatiotemporal regulation of cortical hem specification and positioning, and together ensure the normal development of this hippocampal organizer.


Asunto(s)
Factores de Transcripción Forkhead/biosíntesis , Regulación del Desarrollo de la Expresión Génica/fisiología , Hipocampo/embriología , Proteínas con Homeodominio LIM/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Telencéfalo/embriología , Factores de Transcripción/biosíntesis , Animales , Factores de Transcripción Forkhead/genética , Proteínas con Homeodominio LIM/genética , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Factores de Transcripción/genética
6.
J Neurosci ; 37(46): 11245-11254, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29025924

RESUMEN

Regulation of the neuron-glia cell-fate switch is a critical step in the development of the CNS. Previously, we demonstrated that Lhx2 is a necessary and sufficient regulator of this process in the mouse hippocampal primordium, such that Lhx2 overexpression promotes neurogenesis and suppresses gliogenesis, whereas loss of Lhx2 has the opposite effect. We tested a series of transcription factors for their ability to mimic Lhx2 overexpression and suppress baseline gliogenesis, and also to compensate for loss of Lhx2 and suppress the resulting enhanced level of gliogenesis in the hippocampus. Here, we demonstrate a novel function of Dmrt5/Dmrta2 as a neurogenic factor in the developing hippocampus. We show that Dmrt5, as well as known neurogenic factors Neurog2 and Pax6, can each not only mimic Lhx2 overexpression, but also can compensate for loss of Lhx2 to different extents. We further uncover a reciprocal regulatory relationship between Dmrt5 and Lhx2, such that each can compensate for loss of the other. Dmrt5 and Lhx2 also have opposing regulatory control on Pax6 and Neurog2, indicating a complex bidirectionally regulated network that controls the neuron-glia cell-fate switch.SIGNIFICANCE STATEMENT We identify Dmrt5 as a novel regulator of the neuron-glia cell-fate switch in the developing hippocampus. We demonstrate Dmrt5 to be neurogenic, and reciprocally regulated by Lhx2: loss of either factor promotes gliogenesis; overexpression of either factor suppresses gliogenesis and promotes neurogenesis; each can substitute for loss of the other. Furthermore, each factor has opposing effects on established neurogenic genes Neurog2 and Pax6 Dmrt5 is known to suppress their expression, and we show that Lhx2 is required to maintain it. Our study reveals a complex regulatory network with bidirectional control of a fundamental feature of CNS development, the control of the production of neurons versus astroglia in the developing hippocampus.Finally, we confirm that Lhx2 binds a highly conserved putative enhancer of Dmrt5, suggesting an evolutionarily conserved regulatory relationship between these factors. Our findings uncover a complex network that involves Lhx2, Dmrt5, Neurog2, and Pax6, and that ensures the appropriate amount and timing of neurogenesis and gliogenesis in the developing hippocampus.


Asunto(s)
Hipocampo/fisiología , Proteínas con Homeodominio LIM/fisiología , Neurogénesis/fisiología , Neuroglía/fisiología , Neuronas/fisiología , Factores de Transcripción/fisiología , Animales , Secuencia de Bases , Diferenciación Celular/fisiología , Células Cultivadas , Femenino , Hipocampo/citología , Hipocampo/embriología , Masculino , Ratones , Ratones Transgénicos , Embarazo
7.
J Neurosci ; 37(1): 194-203, 2017 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-28053041

RESUMEN

In the developing cerebral cortex, sequential transcriptional programs take neuroepithelial cells from proliferating progenitors to differentiated neurons with unique molecular identities. The regulatory changes that occur in the chromatin of the progenitors are not well understood. During deep layer neurogenesis, we show that transcription factor LHX2 binds to distal regulatory elements of Fezf2 and Sox11, critical determinants of neuron subtype identity in the mouse neocortex. We demonstrate that LHX2 binds to the nucleosome remodeling and histone deacetylase histone remodeling complex subunits LSD1, HDAC2, and RBBP4, which are proximal regulators of the epigenetic state of chromatin. When LHX2 is absent, active histone marks at the Fezf2 and Sox11 loci are increased. Loss of LHX2 produces an increase, and overexpression of LHX2 causes a decrease, in layer 5 Fezf2 and CTIP2-expressing neurons. Our results provide mechanistic insight into how LHX2 acts as a necessary and sufficient regulator of genes that control cortical neuronal subtype identity. SIGNIFICANCE STATEMENT: The functional complexity of the cerebral cortex arises from an array of distinct neuronal subtypes with unique connectivity patterns that are produced from common progenitors. This study reveals that transcription factor LHX2 regulates the numbers of specific cortical output neuron subtypes by controlling the genes that are required to produce them. Loss or increase in LHX2 during neurogenesis is sufficient to increase or decrease, respectively, a particular subcerebrally projecting population. Mechanistically, LHX2 interacts with chromatin modifying protein complexes to edit the chromatin landscape of its targets Fezf2 and Sox11, which regulates their expression and consequently the identities of the neurons produced. Thus, LHX2 is a key component of the control network for producing neurons that will participate in cortical circuitry.


Asunto(s)
Corteza Cerebral/citología , Proteínas de Unión al ADN/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/fisiología , Factores de Transcripción SOXC/metabolismo , Factores de Transcripción/metabolismo , Animales , Corteza Cerebral/diagnóstico por imagen , Cromatina/genética , Epigénesis Genética , Femenino , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Masculino , Ratones , Nucleosomas/metabolismo , Embarazo
8.
Cereb Cortex ; 27(5): 2841-2856, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27178193

RESUMEN

A unique population of cells, called "lot cells," circumscribes the path of the lateral olfactory tract (LOT) in the rodent brain and acts to restrict its position at the lateral margin of the telencephalon. Lot cells were believed to originate in the dorsal pallium (DP). We show that Lhx2 null mice that lack a DP show a significant increase in the number of mGluR1/lot cells in the piriform cortex, indicating a non-DP origin of these cells. Since lot cells present common developmental features with Cajal-Retzius (CR) cells, we analyzed Wnt3a- and Dbx1-reporter mouse lines and found that mGluR1/lot cells are not generated in the cortical hem, ventral pallium, or septum, the best characterized sources of CR cells. Finally, we identified a novel origin for the lot cells by combining in utero electroporation assays and histochemical characterization. We show that mGluR1/lot cells are specifically generated in the lateral thalamic eminence and that they express mitral cell markers, although a minority of them express ΔNp73 instead. We conclude that most mGluR1/lot cells are prospective mitral cells migrating to the accessory olfactory bulb (OB), whereas mGluR1+, ΔNp73+ cells are CR cells that migrate through the LOT to the piriform cortex and the OB.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Receptores de Glutamato Metabotrópico/metabolismo , Células Madre/fisiología , Tálamo/citología , Tálamo/metabolismo , Animales , Movimiento Celular , Células Cultivadas , Embrión de Mamíferos , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/fisiología , Embarazo , Receptores de Glutamato Metabotrópico/genética , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína Tumoral p73/genética , Proteína Tumoral p73/metabolismo
9.
Cell Mol Life Sci ; 73(13): 2467-90, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26994098

RESUMEN

The components of the nervous system are assembled in development by the process of cell migration. Although the principles of cell migration are conserved throughout the brain, different subsystems may predominantly utilize specific migratory mechanisms, or may display unusual features during migration. Examining these subsystems offers not only the potential for insights into the development of the system, but may also help in understanding disorders arising from aberrant cell migration. The olfactory system is an ancient sensory circuit that is essential for the survival and reproduction of a species. The organization of this circuit displays many evolutionarily conserved features in vertebrates, including molecular mechanisms and complex migratory pathways. In this review, we describe the elaborate migrations that populate each component of the olfactory system in rodents and compare them with those described in the well-studied neocortex. Understanding how the components of the olfactory system are assembled will not only shed light on the etiology of olfactory and sexual disorders, but will also offer insights into how conserved migratory mechanisms may have shaped the evolution of the brain.


Asunto(s)
Movimiento Celular , Bulbo Olfatorio/embriología , Corteza Olfatoria/embriología , Vías Olfatorias , Roedores/embriología , Animales , Evolución Biológica , Hipotálamo/citología , Hipotálamo/embriología , Neuronas/citología , Bulbo Olfatorio/citología , Corteza Olfatoria/citología , Prosencéfalo/citología , Prosencéfalo/embriología , Olfato , Órgano Vomeronasal/citología , Órgano Vomeronasal/embriología
10.
Proc Natl Acad Sci U S A ; 110(50): E4913-21, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24262147

RESUMEN

LIM homeodomain transcription factors are critical regulators of early development in multiple systems but have yet to be examined for a role in circuit formation. The LIM homeobox gene Lhx2 is expressed in cortical progenitors during development and also in the superficial layers of the neocortex in maturity. However, analysis of Lhx2 function at later stages of cortical development has been hampered by severe phenotypes associated with early loss of function. We identified a particular Cre-recombinase line that acts in the cortical primordium after its specification is complete, permitting an analysis of Lhx2 function in neocortical lamination, regionalization, and circuit formation by selective elimination of Lhx2 in the dorsal telencephalon. We report a profound disruption of cortical neuroanatomical and molecular features upon loss of Lhx2 in the cortex from embryonic day 11.5. A unique feature of cortical circuitry, the somatosensory barrels, is undetectable, and molecular patterning of cortical regions appears disrupted. Surprisingly, thalamocortical afferents innervate the mutant cortex with apparently normal regional specificity. Electrophysiological recordings reveal a loss of responses evoked by stimulation of individual whiskers, but responses to simultaneous stimulation of multiple whiskers were present, suggesting that thalamic afferents are unable to organize the neurocircuitry for barrel formation because of a cortex-specific requirement of Lhx2. We report that Lhx2 is required for the expression of transcription factor paired box gene 6, axon guidance molecule Ephrin A5, and the receptor NMDA receptor 1. These genes may mediate Lhx2 function in the formation of specialized neurocircuitry necessary for neocortical function.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Proteínas con Homeodominio LIM/metabolismo , Corteza Somatosensorial/embriología , Factores de Transcripción/metabolismo , Animales , Inmunoprecipitación de Cromatina , Efrina-A5/metabolismo , Potenciales Evocados/fisiología , Proteínas del Ojo/metabolismo , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Hibridación in Situ , Integrasas , Proteínas con Homeodominio LIM/deficiencia , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Vías Nerviosas/embriología , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Proteínas Represoras/metabolismo , Corteza Somatosensorial/metabolismo , Factores de Transcripción/deficiencia
11.
Cereb Cortex ; 24(5): 1361-72, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-23307637

RESUMEN

Early brain development is regulated by the coordinated actions of multiple signaling centers at key boundaries between compartments. Three telencephalic midline structures are in a position to play such roles in forebrain patterning: The cortical hem, the septum, and the thalamic eminence at the diencephalic-telencephalic boundary. These structures express unique complements of signaling molecules, and they also produce distinct populations of Cajal-Retzius cells, which are thought to act as "mobile patterning units," migrating tangentially to cover the telencephalic surface. We show that these 3 structures require the transcription factor Lhx2 to delimit their extent. In the absence of Lhx2 function, all 3 structures are greatly expanded, and the Cajal-Retzius cell population is dramatically increased. We propose that the hem, septum, and thalamic eminence together form a "forebrain hem system" that defines and regulates the formation of the telencephalic midline. Disruptions in the forebrain hem system may be implicated in severe brain malformations such as holoprosencephaly. Lhx2 functions as a central regulator of this system's development. Since all components of the forebrain hem system have been identified across several vertebrate species, the mechanisms that regulate them may have played a fundamental role in driving key aspects of forebrain evolution.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Proteínas con Homeodominio LIM/metabolismo , Vías Nerviosas/embriología , Vías Nerviosas/metabolismo , Prosencéfalo/embriología , Prosencéfalo/metabolismo , Factores de Transcripción/metabolismo , Factores de Edad , Animales , Evolución Biológica , Bromodesoxiuridina/metabolismo , Diferenciación Celular , Embrión de Mamíferos , Feto , Humanos , Antígeno Ki-67/metabolismo , Proteínas con Homeodominio LIM/genética , Ratones , Ratones Transgénicos , Modelos Neurológicos , Mutación/genética , Prosencéfalo/citología , Factores de Transcripción/genética
12.
Proc Natl Acad Sci U S A ; 109(46): 18839-44, 2012 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-23112163

RESUMEN

The search for developmental mechanisms driving vertebrate organogenesis has paved the way toward a deeper understanding of birth defects. During embryogenesis, parts of the heart and craniofacial muscles arise from pharyngeal mesoderm (PM) progenitors. Here, we reveal a hierarchical regulatory network of a set of transcription factors expressed in the PM that initiates heart and craniofacial organogenesis. Genetic perturbation of this network in mice resulted in heart and craniofacial muscle defects, revealing robust cross-regulation between its members. We identified Lhx2 as a previously undescribed player during cardiac and pharyngeal muscle development. Lhx2 and Tcf21 genetically interact with Tbx1, the major determinant in the etiology of DiGeorge/velo-cardio-facial/22q11.2 deletion syndrome. Furthermore, knockout of these genes in the mouse recapitulates specific cardiac features of this syndrome. We suggest that PM-derived cardiogenesis and myogenesis are network properties rather than properties specific to individual PM members. These findings shed new light on the developmental underpinnings of congenital defects.


Asunto(s)
Tipificación del Cuerpo/fisiología , Embrión de Mamíferos/embriología , Cabeza/embriología , Corazón/embriología , Mesodermo/embriología , Músculo Esquelético/embriología , Miocardio , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Noqueados
13.
J Neurosci ; 33(16): 6877-84, 2013 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-23595746

RESUMEN

Eye formation is regulated by a complex network of eye field transcription factors (EFTFs), including LIM-homeodomain gene LHX2. We disrupted LHX2 function at different stages during this process using a conditional knock-out strategy in mice. We find that LHX2 function is required in an ongoing fashion to maintain optic identity across multiple stages, from the formation of the optic vesicle to the differentiation of the neuroretina. At each stage, loss of Lhx2 led to upregulation of a set of molecular markers that are normally expressed in the thalamic eminence and in the anterodorsal hypothalamus in a portion of the optic vesicle or retina. Furthermore, the longer LHX2 function was maintained, the further optic morphogenesis progressed. Early loss of function caused profound mispatterning of the entire telencephalic-optic-hypothalamic field, such that the optic vesicle became mispositioned and appeared to arise from the diencephalic-telencephalic boundary. At subsequent stages, loss of Lhx2 did not affect optic vesicle position but caused arrest of optic cup formation. If Lhx2 was selectively disrupted in the neuroretina from E11.5, the neuroretina showed gross dysmorphology along with aberrant expression of markers specific to the thalamic eminence and anterodorsal hypothalamus. Our findings indicate a continual requirement for LHX2 throughout the early stages of optic development, not only to maintain optic identity by suppressing alternative fates but also to mediate multiple steps of optic morphogenesis. These findings provide new insight into the anophthalmic phenotype of the Lhx2 mutant and reveal novel roles for this transcription factor in eye development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Proteínas con Homeodominio LIM/metabolismo , Morfogénesis/genética , Organogénesis/genética , Factores de Transcripción/metabolismo , Vías Visuales/fisiología , Factores de Edad , Animales , Tipificación del Cuerpo/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteínas con Homeodominio LIM/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Proteínas del Tejido Nervioso/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/metabolismo , Proteínas/genética , ARN no Traducido , Proteínas Represoras/metabolismo , Retina/anomalías , Retina/patología , Tamoxifeno/farmacología , Factores de Transcripción/genética , Vías Visuales/embriología
14.
Proc Natl Acad Sci U S A ; 108(27): E265-74, 2011 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-21690374

RESUMEN

The sequential production of neurons and astrocytes from neuroepithelial precursors is a fundamental feature of central nervous system development. We report that LIM-homeodomain (LIM-HD) transcription factor Lhx2 regulates this transition in the developing hippocampus. Disrupting Lhx2 function in the embryonic hippocampus by in utero electroporation and in organotypic slice culture caused the premature production of astrocytes at stages when neurons are normally generated. Lhx2 function is therefore necessary to suppress astrogliogenesis during the neurogenic period. Furthermore, Lhx2 overexpression was sufficient to suppress astrogliogenesis and prolong the neurogenic period. We provide evidence that Lhx2 overexpression can counteract the instructive astrogliogenic effect of Notch activation. Lhx2 overexpression was also able to override and suppress the activation of the GFAP promoter by Nfia, a Notch-regulated transcription factor that is required for gliogenesis. Thus, Lhx2 appears to act as a "brake" on Notch/Nfia-mediated astrogliogenesis. This critical role for Lhx2 is spatially restricted to the hippocampus, because loss of Lhx2 function in the neocortex did not result in premature astrogliogenesis at the expense of neurogenesis. Our results therefore place Lhx2 as a central regulator of the neuron-glia cell fate decision in the hippocampus and reveal a striking regional specificity of this fundamental function within the dorsal telencephalon.


Asunto(s)
Hipocampo/embriología , Proteínas de Homeodominio/fisiología , Neurogénesis/fisiología , Factores de Transcripción/fisiología , Animales , Astrocitos/citología , Astrocitos/fisiología , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Femenino , Hipocampo/citología , Hipocampo/fisiología , Proteínas de Homeodominio/genética , Proteínas con Homeodominio LIM , Ratones , Ratones Noqueados , Ratones Transgénicos , Factores de Transcripción NFI/fisiología , Neocórtex/citología , Neocórtex/embriología , Neocórtex/fisiología , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Neurogénesis/genética , Fenotipo , Embarazo , Receptores Notch/fisiología , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
15.
Oxf Open Neurosci ; 3: kvae001, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38595939

RESUMEN

PRDM16 is a dynamic transcriptional regulator of various stem cell niches, including adipocytic, hematopoietic, cardiac progenitors, and neural stem cells. PRDM16 has been suggested to contribute to 1p36 deletion syndrome, one of the most prevalent subtelomeric microdeletion syndromes. We report a patient with a de novo nonsense mutation in the PRDM16 coding sequence, accompanied by lissencephaly and microcephaly features. Human stem cells were genetically modified to mimic this mutation, generating cortical organoids that exhibited altered cell cycle dynamics. RNA sequencing of cortical organoids at day 32 unveiled changes in cell adhesion and WNT-signaling pathways. ChIP-seq of PRDM16 identified binding sites in postmortem human fetal cortex, indicating the conservation of PRDM16 binding to developmental genes in mice and humans, potentially at enhancer sites. A shared motif between PRDM16 and LHX2 was identified and further examined through comparison with LHX2 ChIP-seq data from mice. These results suggested a collaborative partnership between PRDM16 and LHX2 in regulating a common set of genes and pathways in cortical radial glia cells, possibly via their synergistic involvement in cortical development.

16.
J Neurosci ; 32(13): 4372-85, 2012 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-22457488

RESUMEN

The assembly of neural circuits is dependent upon the generation of specific neuronal subtypes, each subtype displaying unique properties that direct the formation of selective connections with appropriate target cells. Actions of transcription factors in neural progenitors and postmitotic cells are key regulators in this process. LIM-homeodomain transcription factors control crucial aspects of neuronal differentiation, including subtype identity and axon guidance. Nonetheless, their regulation during development is poorly understood and the identity of the downstream molecular effectors of their activity remains largely unknown. Here, we demonstrate that the Lhx2 transcription factor is dynamically regulated in distinct pools of thalamic neurons during the development of thalamocortical connectivity in mice. Indeed, overexpression of Lhx2 provokes defective thalamocortical axon guidance in vivo, while specific conditional deletion of Lhx2 in the thalamus produces topographic defects that alter projections from the medial geniculate nucleus and from the caudal ventrobasal nucleus in particular. Moreover, we demonstrate that Lhx2 influences axon guidance and the topographical sorting of axons by regulating the expression of Robo1 and Robo2 guidance receptors, which are essential for these axons to establish correct connections in the cerebral cortex. Finally, augmenting Robo1 function restores normal axon guidance in Lhx2-overexpressing neurons. By regulating axon guidance receptors, such as Robo1 and Robo2, Lhx2 differentially regulates the axon guidance program of distinct populations of thalamic neurons, thus enabling the establishment of specific neural connections.


Asunto(s)
Axones/fisiología , Corteza Cerebral/fisiología , Proteínas con Homeodominio LIM/fisiología , Proteínas del Tejido Nervioso/biosíntesis , Neurogénesis/fisiología , Receptores Inmunológicos/biosíntesis , Tálamo/fisiología , Factores de Transcripción/fisiología , Animales , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/metabolismo , Embrión de Mamíferos , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Proteínas con Homeodominio LIM/metabolismo , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Ratones Transgénicos , Vías Nerviosas/crecimiento & desarrollo , Vías Nerviosas/metabolismo , Vías Nerviosas/fisiología , Transducción de Señal/fisiología , Tálamo/crecimiento & desarrollo , Tálamo/metabolismo , Factores de Transcripción/metabolismo , Proteínas Roundabout
17.
bioRxiv ; 2023 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-37609127

RESUMEN

PRDM16 is a dynamic transcriptional regulator of various stem cell niches, including adipocytic, hematopoietic, cardiac progenitors, and neural stem cells. PRDM16 has been suggested to contribute to 1p36 deletion syndrome, one of the most prevalent subtelomeric microdeletion syndromes. We report a patient with a de novo nonsense mutation in the PRDM16 coding sequence, accompanied by lissencephaly and microcephaly features. Human stem cells were genetically modified to mimic this mutation, generating cortical organoids that exhibited altered cell cycle dynamics. RNA sequencing of cortical organoids at day 32 unveiled changes in cell adhesion and WNT-signaling pathways. ChIP-seq of PRDM16 identified binding sites in postmortem human fetal cortex, indicating the conservation of PRDM16 binding to developmental genes in mice and humans, potentially at enhancer sites. A shared motif between PRDM16 and LHX2 was identified and further examined through comparison with LHX2 ChIP-seq data from mice. These results suggested a collaborative partnership between PRDM16 and LHX2 in regulating a common set of genes and pathways in cortical radial glia cells, possibly via their synergistic involvement in cortical development.

18.
Mol Neurobiol ; 59(9): 5750-5765, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35781633

RESUMEN

Changes in the transcription factor (TF) expression are critical for brain development, and they may also underlie neurodevelopmental disorders. Indeed, T-box brain1 (Tbr1) is a TF crucial for the formation of neocortical layer VI, and mutations and microdeletions in that gene are associated with malformations in the human cerebral cortex, alterations that accompany autism spectrum disorder (ASD). Interestingly, Tbr1 upregulation has also been related to the occurrence of ASD-like symptoms, although limited studies have addressed the effect of increased Tbr1 levels during neocortical development. Here, we analysed the impact of Tbr1 misexpression in mouse neural progenitor cells (NPCs) at embryonic day 14.5 (E14.5), when they mainly generate neuronal layers II-IV. By E18.5, cells accumulated in the intermediate zone and in the deep cortical layers, whereas they became less abundant in the upper cortical layers. In accordance with this, the proportion of Sox5+ cells in layers V-VI increased, while that of Cux1+ cells in layers II-IV decreased. On postnatal day 7, fewer defects in migration were evident, although a higher proportion of Sox5+ cells were seen in the upper and deep layers. The abnormal neuronal migration could be partially due to the altered multipolar-bipolar neuron morphologies induced by Tbr1 misexpression, which also reduced dendrite growth and branching, and disrupted the corpus callosum. Our results indicate that Tbr1 misexpression in cortical NPCs delays or disrupts neuronal migration, neuronal specification, dendrite development and the formation of the callosal tract. Hence, genetic changes that provoke ectopic Tbr1 upregulation during development could provoke cortical brain malformations.


Asunto(s)
Trastorno del Espectro Autista , Neocórtex , Animales , Trastorno del Espectro Autista/genética , Corteza Cerebral/metabolismo , Humanos , Ratones , Neocórtex/metabolismo , Neurogénesis/genética , Neuronas/metabolismo , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/metabolismo
19.
Nat Commun ; 13(1): 633, 2022 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-35110543

RESUMEN

The choroid plexus secretes cerebrospinal fluid and is critical for the development and function of the brain. In the telencephalon, the choroid plexus epithelium arises from the Wnt- expressing cortical hem. Canonical Wnt signaling pathway molecules such as nuclear ß-CATENIN are expressed in the mouse and human embryonic choroid plexus epithelium indicating that this pathway is active. Point mutations in human ß-CATENIN are known to result in the constitutive activation of canonical Wnt signaling. In a mouse model that recapitulates this perturbation, we report a loss of choroid plexus epithelial identity and an apparent transformation of this tissue to a neuronal identity. Aspects of this phenomenon are recapitulated in human embryonic stem cell derived organoids. The choroid plexus is also disrupted when ß-Catenin is conditionally inactivated. Together, our results indicate that canonical Wnt signaling is required in a precise and regulated manner for normal choroid plexus development in the mammalian brain.


Asunto(s)
Plexo Coroideo/metabolismo , Epitelio/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/fisiología , Animales , Diferenciación Celular , Núcleo Celular/metabolismo , Plexo Coroideo/patología , Femenino , Humanos , Masculino , Ratones , Telencéfalo/metabolismo , Vía de Señalización Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
20.
Front Cell Dev Biol ; 10: 1011109, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36263009

RESUMEN

Early forebrain patterning entails the correct regional designation of the neuroepithelium, and appropriate specification, generation, and distribution of neural cells during brain development. Specific signaling and transcription factors are known to tightly regulate patterning of the dorsal telencephalon to afford proper structural/functional cortical arealization and morphogenesis. Nevertheless, whether and how changes of the chromatin structure link to the transcriptional program(s) that control cortical patterning remains elusive. Here, we report that the BAF chromatin remodeling complex regulates the spatiotemporal patterning of the mouse dorsal telencephalon. To determine whether and how the BAF complex regulates cortical patterning, we conditionally deleted the BAF complex scaffolding subunits BAF155 and BAF170 in the mouse dorsal telencephalic neuroepithelium. Morphological and cellular changes in the BAF mutant forebrain were examined using immunohistochemistry and in situ hybridization. RNA sequencing, Co-immunoprecipitation, and mass spectrometry were used to investigate the molecular basis of BAF complex involvement in forebrain patterning. We found that conditional ablation of BAF complex in the dorsal telencephalon neuroepithelium caused expansion of the cortical hem and medial cortex beyond their developmental boundaries. Consequently, the hippocampal primordium is not specified, the mediolateral cortical patterning is compromised, and the cortical identity is disturbed in the absence of BAF complex. The BAF complex was found to interact with the cortical hem suppressor LHX2. The BAF complex suppresses cortical hem fate to permit proper forebrain patterning. We provide evidence that BAF complex modulates mediolateral cortical patterning possibly by interacting with the transcription factor LHX2 to drive the LHX2-dependent transcriptional program essential for dorsal telencephalon patterning. Our data suggest a putative mechanistic synergy between BAF chromatin remodeling complex and LHX2 in regulating forebrain patterning and ontogeny.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA