Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Development ; 150(16)2023 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-37522516

RESUMEN

During embryonic development, tissue-specific transcription factors and chromatin remodelers function together to ensure gradual, coordinated differentiation of multiple lineages. Here, we define this regulatory interplay in the developing retinal pigmented epithelium (RPE), a neuroectodermal lineage essential for the development, function and maintenance of the adjacent retina. We present a high-resolution spatial transcriptomic atlas of the developing mouse RPE and the adjacent ocular mesenchyme obtained by geographical position sequencing (Geo-seq) of a single developmental stage of the eye that encompasses young and more mature ocular progenitors. These transcriptomic data, available online, reveal the key transcription factors and their gene regulatory networks during RPE and ocular mesenchyme differentiation. Moreover, conditional inactivation followed by Geo-seq revealed that this differentiation program is dependent on the activity of SWI/SNF complexes, shown here to control the expression and activity of RPE transcription factors and, at the same time, inhibit neural progenitor and cell proliferation genes. The findings reveal the roles of the SWI/SNF complexes in controlling the intersection between RPE and neural cell fates and the coupling of cell-cycle exit and differentiation.


Asunto(s)
Epitelio Pigmentado de la Retina , Factores de Transcripción , Femenino , Embarazo , Ratones , Animales , Diferenciación Celular/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Proliferación Celular/genética , Epitelio/metabolismo
2.
Development ; 148(3)2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33462115

RESUMEN

Fine-tuned gene expression is crucial for neurodevelopment. The gene expression program is tightly controlled at different levels, including RNA decay. N6-methyladenosine (m6A) methylation-mediated degradation of RNA is essential for brain development. However, m6A methylation impacts not only RNA stability, but also other RNA metabolism processes. How RNA decay contributes to brain development is largely unknown. Here, we show that Exosc10, a RNA exonuclease subunit of the RNA exosome complex, is indispensable for forebrain development. We report that cortical cells undergo overt apoptosis, culminating in cortical agenesis upon conditional deletion of Exosc10 in mouse cortex. Mechanistically, Exosc10 directly binds and degrades transcripts of the P53 signaling-related genes, such as Aen and Bbc3. Overall, our findings suggest a crucial role for Exosc10 in suppressing the P53 pathway, in which the rapid turnover of the apoptosis effectors Aen and Bbc3 mRNAs is essential for cell survival and normal cortical histogenesis.


Asunto(s)
Supervivencia Celular/fisiología , Exosomas/genética , Exosomas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Prosencéfalo/crecimiento & desarrollo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis , Biología Computacional , Exorribonucleasas/genética , Complejo Multienzimático de Ribonucleasas del Exosoma/genética , Complejo Multienzimático de Ribonucleasas del Exosoma/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Prosencéfalo/patología , ARN/metabolismo , Estabilidad del ARN , Transducción de Señal/genética , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor
3.
PLoS Genet ; 17(3): e1009446, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33750945

RESUMEN

The BAF complex plays an important role in the development of a wide range of tissues by modulating gene expression programs at the chromatin level. However, its role in neural crest development has remained unclear. To determine the role of the BAF complex, we deleted BAF155/BAF170, the core subunits required for the assembly, stability, and functions of the BAF complex in neural crest cells (NCCs). Neural crest-specific deletion of BAF155/BAF170 leads to embryonic lethality due to a wide range of developmental defects including craniofacial, pharyngeal arch artery, and OFT defects. RNAseq and transcription factor enrichment analysis revealed that the BAF complex modulates the expression of multiple signaling pathway genes including Hippo and Notch, essential for the migration, proliferation, and differentiation of the NCCs. Furthermore, we demonstrated that the BAF complex is essential for the Brg1-Yap-Tead-dependent transcription of target genes in NCCs. Together, our results demonstrate an important role of the BAF complex in modulating the gene regulatory network essential for neural crest development.


Asunto(s)
Ensamble y Desensamble de Cromatina , Proteínas de Unión al ADN/genética , Regulación del Desarrollo de la Expresión Génica , Cresta Neural/embriología , Cresta Neural/metabolismo , Neurogénesis/genética , Animales , Diferenciación Celular/genética , Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Desarrollo Embrionario/genética , Eliminación de Gen , Redes Reguladoras de Genes , Genes Reporteros , Ratones , Ratones Transgénicos , Especificidad de Órganos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética
4.
PLoS Genet ; 12(9): e1006274, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27611684

RESUMEN

Neurogenesis is a key developmental event through which neurons are generated from neural stem/progenitor cells. Chromatin remodeling BAF (mSWI/SNF) complexes have been reported to play essential roles in the neurogenesis of the central nervous system. However, whether BAF complexes are required for neuron generation in the olfactory system is unknown. Here, we identified onscBAF and ornBAF complexes, which are specifically present in olfactory neural stem cells (oNSCs) and olfactory receptor neurons (ORNs), respectively. We demonstrated that BAF155 subunit is highly expressed in both oNSCs and ORNs, whereas high expression of BAF170 subunit is observed only in ORNs. We report that conditional deletion of BAF155, a core subunit in both onscBAF and ornBAF complexes, causes impaired proliferation of oNSCs as well as defective maturation and axonogenesis of ORNs in the developing olfactory epithelium (OE), while the high expression of BAF170 is important for maturation of ORNs. Interestingly, in the absence of BAF complexes in BAF155/BAF170 double-conditional knockout mice (dcKO), OE is not specified. Mechanistically, BAF complex is required for normal activation of Pax6-dependent transcriptional activity in stem cells/progenitors of the OE. Our findings unveil a novel mechanism mediated by the mSWI/SNF complex in OE neurogenesis and development.


Asunto(s)
Proteínas Cromosómicas no Histona/genética , Neurogénesis , Mucosa Olfatoria/metabolismo , Factores de Transcripción/genética , Animales , Células Cultivadas , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN , Eliminación de Gen , Ratones , Ratones Endogámicos C57BL , Mucosa Olfatoria/citología , Mucosa Olfatoria/embriología , Neuronas Receptoras Olfatorias/citología , Neuronas Receptoras Olfatorias/metabolismo , Factores de Transcripción/metabolismo
6.
Cell Mol Life Sci ; 71(17): 3199-218, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24614969

RESUMEN

The mammalian neocortex is a sheet of cells covering the cerebrum that provides the structural basis for the perception of sensory inputs, motor output responses, cognitive function, and mental capacity of primates. Recent discoveries promote the concept that increased cortical surface size and thickness in phylogenetically advanced species is a result of an increased generation of neurons, a process that underlies higher cognitive and intellectual performance in higher primates and humans. Here, we review some of the advances in the field, focusing on the diversity of neocortical progenitors in different species and the cellular mechanisms of neurogenesis. We discuss recent views on intrinsic and extrinsic molecular determinants, including the role of epigenetic chromatin modifiers and microRNA, in the control of neuronal output in developing cortex and in the establishment of normal cortical architecture.


Asunto(s)
Neocórtex/crecimiento & desarrollo , Animales , División Celular , Polaridad Celular , Ensamble y Desensamble de Cromatina , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Mamíferos/crecimiento & desarrollo , MicroARNs/fisiología , Neocórtex/anatomía & histología , Proteínas del Tejido Nervioso/fisiología , Células-Madre Neurales/citología , Neurogénesis , Neuronas/citología , Tamaño de los Órganos , Procesamiento Postranscripcional del ARN , Transducción de Señal , Especificidad de la Especie
7.
Cell Tissue Res ; 356(3): 575-84, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24496512

RESUMEN

ATP-dependent BAF chromatin remodeling complexes play an essential role in the maintenance of the gene expression program by regulating the structure of chromatin. There is increasing evidence that BAF complexes based on the alternative ATPase subunits, Brg1 and Brm, control the differentiation of neural stem cells (NSCs) to generate distinct neural cell types and modulate trans-differentiation between cell types. The BAF complexes have dedicated functions at different stages of neural differentiation that appear to arise by combinatorial assembly of their subunits. Furthermore, the differentiation of NSCs is regulated by the tight interactions between the BAF chromatin remodeling complex and the transcriptional machinery. Here, we review recent insights into the functional interaction between BAF complexes and various transcription factors (TFs) in neural differentiation and cellular reprogramming.


Asunto(s)
Diferenciación Celular/fisiología , Ensamble y Desensamble de Cromatina/fisiología , Proteínas de Unión al ADN/metabolismo , Complejos Multiproteicos/metabolismo , Células-Madre Neurales/metabolismo , Proteínas Nucleares/metabolismo , Animales , ADN Helicasas/genética , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/genética , Humanos , Complejos Multiproteicos/genética , Células-Madre Neurales/citología , Proteínas Nucleares/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Biology (Basel) ; 13(2)2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38392287

RESUMEN

Enrichment of basal progenitors (BPs) in the developing neocortex is a central driver of cortical enlargement. The transcription factor Pax6 is known as an essential regulator in generation of BPs. H3 lysine 9 acetylation (H3K9ac) has emerged as a crucial epigenetic mechanism that activates the gene expression program required for BP pool amplification. In this current work, we applied immunohistochemistry, RNA sequencing, chromatin immunoprecipitation and sequencing, and the yeast two-hybrid assay to reveal that the BP-genic effect of H3 acetylation is dependent on Pax6 functionality in the developing mouse cortex. In the presence of Pax6, increased H3 acetylation caused BP pool expansion, leading to enhanced neurogenesis, which evoked expansion and quasi-convolution of the mouse neocortex. Interestingly, H3 acetylation activation exacerbates the BP depletion and corticogenesis reduction effect of Pax6 ablation in cortex-specific Pax6 mutants. Furthermore, we found that H3K9 acetyltransferase KAT2A/GCN5 interacts with Pax6 and potentiates Pax6-dependent transcriptional activity. This explains a genome-wide lack of H3K9ac, especially in the promoter regions of BP-genic genes, in the Pax6 mutant cortex. Together, these findings reveal a mechanistic coupling of H3 acetylation and Pax6 in orchestrating BP production and cortical expansion through the promotion of a BP gene expression program during cortical development.

9.
Front Neurosci ; 16: 824802, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35281509

RESUMEN

Cortical development is dependent on key processes that can influence apical progenitor cell division and progeny. Pivotal among such critical cellular processes is the intricate mechanism of cell delamination. This indispensable cell detachment process mainly entails the loss of apical anchorage, and subsequent migration of the mitotic derivatives of the highly polarized apical cortical progenitors. Such apical progenitor derivatives are responsible for the majority of cortical neurogenesis. Many factors, including transcriptional and epigenetic/chromatin regulators, are known to tightly control cell attachment and delamination tendency in the cortical neurepithelium. Activity of these molecular regulators principally coordinate morphogenetic cues to engender remodeling or disassembly of tethering cellular components and external cell adhesion molecules leading to exit of differentiating cells in the ventricular zone. Improper cell delamination is known to frequently impair progenitor cell fate commitment and neuronal migration, which can cause aberrant cortical cell number and organization known to be detrimental to the structure and function of the cerebral cortex. Indeed, some neurodevelopmental abnormalities, including Heterotopia, Schizophrenia, Hydrocephalus, Microcephaly, and Chudley-McCullough syndrome have been associated with cell attachment dysregulation in the developing mammalian cortex. This review sheds light on the concept of cell delamination, mechanistic (transcriptional and epigenetic regulation) nuances involved, and its importance for corticogenesis. Various neurodevelopmental disorders with defective (too much or too little) cell delamination as a notable etiological underpinning are also discussed.

10.
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.

11.
Front Cell Dev Biol ; 9: 656849, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34095121

RESUMEN

Dynamic modification of RNA affords proximal regulation of gene expression triggered by non-genomic or environmental changes. One such epitranscriptomic alteration in RNA metabolism is the installation of a methyl group on adenosine [N6-methyladenosine (m6A)] known to be the most prevalent modified state of messenger RNA (mRNA) in the mammalian cell. The methylation machinery responsible for the dynamic deposition and recognition of m6A on mRNA is composed of subunits that play specific roles, including reading, writing, and erasing of m6A marks on mRNA to influence gene expression. As a result, peculiar cellular perturbations have been linked to dysregulation of components of the mRNA methylation machinery or its cofactors. It is increasingly clear that neural tissues/cells, especially in the brain, make the most of m6A modification in maintaining normal morphology and function. Neurons in particular display dynamic distribution of m6A marks during development and in adulthood. Interestingly, such dynamic m6A patterns are responsive to external cues and experience. Specific disturbances in the neural m6A landscape lead to anomalous phenotypes, including aberrant stem/progenitor cell proliferation and differentiation, defective cell fate choices, and abnormal synaptogenesis. Such m6A-linked neural perturbations may singularly or together have implications for syndromic or non-syndromic neurological diseases, given that most RNAs in the brain are enriched with m6A tags. Here, we review the current perspectives on the m6A machinery and function, its role in brain development and possible association with brain disorders, and the prospects of applying the clustered regularly interspaced short palindromic repeats (CRISPR)-dCas13b system to obviate m6A-related neurological anomalies.

12.
STAR Protoc ; 2(2): 100503, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-33997817

RESUMEN

Identification of peptides mediating protein-protein interaction (PPI) is crucial for understanding the function of interlinked proteins in cellular processes and amino acid-associated diseases. Traditional PPI assays are laborious, involving the generation of many truncated proteins. SPOT peptide assay allows high-throughput detection of domains essential for PPI by synthesizing several hundred peptides on a cellulose membrane. Here, we present a rapid SPOT peptide protocol for identifying the binding motifs, which mediate interaction between the chromatin remodeling factors BAF155/BAF170 and the epigenetic factor Kdm6b. For complete details on the use and execution of this protocol, please refer to Narayanan et al. (2015).


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Biblioteca de Péptidos , Mapeo de Interacción de Proteínas , Proteómica , Factores de Transcripción/metabolismo , Proteínas de Unión al ADN/química , Células HeLa , Humanos , Histona Demetilasas con Dominio de Jumonji/química , Dominios y Motivos de Interacción de Proteínas , Factores de Transcripción/química
13.
STAR Protoc ; 2(1): 100318, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33554149

RESUMEN

Cell sorting can be used to purify cell populations for cell type-specific molecular probing. Fluorescence-activated cell sorting (FACS) coupled with high-throughput sequencing affords molecular signature identification for specific cell types. FACS has many challenges that limit comprehensive cell purification from the brain, leading to incomplete molecular characterization. Here, we present the intranuclear immunostaining-based FACS protocol with several modified steps, which allows optimized nuclei/cell sorting from mouse or human embryonic cortical tissue for distinct downstream molecular investigation of basal intermediate progenitors.


Asunto(s)
Separación Celular/métodos , Citometría de Flujo/métodos , Inmunoensayo/métodos , Animales , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones
14.
Front Mol Neurosci ; 14: 687581, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34220450

RESUMEN

Radial neuronal migration is a key neurodevelopmental event indispensable for proper cortical laminar organization. Cortical neurons mainly use glial fiber guides, cell adhesion dynamics, and cytoskeletal remodeling, among other discrete processes, to radially trek from their birthplace to final layer positions. Dysregulated radial migration can engender cortical mis-lamination, leading to neurodevelopmental disorders. Epigenetic factors, including chromatin remodelers have emerged as formidable regulators of corticogenesis. Notably, the chromatin remodeler BAF complex has been shown to regulate several aspects of cortical histogenesis. Nonetheless, our understanding of how BAF complex regulates neuronal migration is limited. Here, we report that BAF complex is required for neuron migration during cortical development. Ablation of BAF complex in the developing mouse cortex caused alteration in the cortical gene expression program, leading to loss of radial migration-related factors critical for proper cortical layer formation. Of note, BAF complex inactivation in cortex caused defective neuronal polarization resulting in diminished multipolar-to-bipolar transition and eventual disruption of radial migration of cortical neurons. The abnormal radial migration and cortical mis-lamination can be partly rescued by downregulating WNT signaling hyperactivity in the BAF complex mutant cortex. By implication, the BAF complex modulates WNT signaling to establish the gene expression program required for glial fiber-dependent neuronal migration, and cortical lamination. Overall, BAF complex has been identified to be crucial for cortical morphogenesis through instructing multiple aspects of radial neuronal migration in a WNT signaling-dependent manner.

15.
Stem Cell Reports ; 16(4): 968-984, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33798452

RESUMEN

Intermediate progenitor cells (IPCs) are neocortical neuronal precursors. Although IPCs play crucial roles in corticogenesis, their molecular features remain largely unknown. In this study, we aimed to characterize the molecular profile of IPCs. We isolated TBR2-positive (+) IPCs and TBR2-negative (-) cell populations in the developing mouse cortex. Comparative genome-wide gene expression analysis of TBR2+ IPCs versus TBR2- cells revealed differences in key factors involved in chromatid segregation, cell-cycle regulation, transcriptional regulation, and cell signaling. Notably, mutation of many IPC genes in human has led to intellectual disability and caused a wide range of cortical malformations, including microcephaly and agenesis of corpus callosum. Loss-of-function experiments in cortex-specific mutants of Esco2, one of the novel IPC genes, demonstrate its critical role in IPC maintenance, and substantiate the identification of a central genetic determinant of IPC biogenesis. Our data provide novel molecular characteristics of IPCs in the developing mouse cortex.


Asunto(s)
Acetiltransferasas/metabolismo , Corteza Cerebral/citología , Corteza Cerebral/embriología , Perfilación de la Expresión Génica , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Acetiltransferasas/genética , Animales , Apoptosis/genética , Cromátides/metabolismo , Segregación Cromosómica/genética , Regulación de la Expresión Génica , Humanos , Ratones , Mitosis/genética , Mutación/genética , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Transducción de Señal
16.
Neuroscience ; 463: 303-316, 2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-33774122

RESUMEN

Cortical morphogenesis entails several neurobiological events, including proliferation and differentiation of progenitors, migration of neuroblasts, and neuronal maturation leading to functional neural circuitry. These neurodevelopmental processes are delicately regulated by many factors. Endosomal SNAREs have emerged as formidable modulators of neuronal growth, aside their well-known function in membrane/vesicular trafficking. However, our understanding of their influence on cortex formation is limited. Here, we report that the SNAREs Vti1a and Vti1b (Vti1a/1b) are critical for proper cortical development. Following null mutation of Vti1a/1b in mouse, the late-embryonic mutant cortex appeared dysgenic, and the cortical progenitors therein were depleted beyond normal. Notably, cortical layer 5 (L5) is distinctively disorganized in the absence of Vti1a/1b. The latter defect, coupled with an overt apoptosis of Ctip2-expressing L5 neurons, likely contributed to the substantial loss of corticospinal and callosal projections in the Vti1a/1b-deficient mouse brain. These findings suggest that Vti1a/1b serve key neurodevelopmental functions during cortical histogenesis, which when mechanistically elucidated, can lend clarity to how endosomal SNAREs regulate brain development, or how their dysfunction may have implications for neurological disorders.


Asunto(s)
Células-Madre Neurales , Neuronas , Animales , Diferenciación Celular , Corteza Cerebral/metabolismo , Ratones , Células-Madre Neurales/metabolismo , Neurogénesis , Neuronas/metabolismo , Proteínas Qb-SNARE/metabolismo , Proteínas SNARE/metabolismo
17.
Front Cell Dev Biol ; 9: 619538, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34336815

RESUMEN

Oligodendrocytes are responsible for axon myelination in the brain and spinal cord. Generation of oligodendrocytes entails highly regulated multistage neurodevelopmental events, including proliferation, differentiation and maturation. The chromatin remodeling BAF (mSWI/SNF) complex is a notable regulator of neural development. In our previous studies, we determined the indispensability of the BAF complex scaffolding subunits BAF155 and BAF170 for neurogenesis, whereas their role in gliogenesis is unknown. Here, we show that the expression of BAF155 and BAF170 is essential for the genesis of oligodendrocytes during brain development. We report that the ablation of BAF155 and BAF170 in the dorsal telencephalic (dTel) neural progenitors or in oligodendrocyte-producing progenitors in the ventral telencephalon (vTel) in double-conditional knockout (dcKO) mouse mutants, perturbed the process of oligodendrogenesis. Molecular marker and cell cycle analyses revealed impairment of oligodendrocyte precursor specification and proliferation, as well as overt depletion of oligodendrocytes pool in dcKO mutants. Our findings unveil a central role of BAF155 and BAF170 in oligodendrogenesis, and thus substantiate the involvement of the BAF complex in the production of oligodendrocytes in the forebrain.

18.
Sci Adv ; 7(38): eabc6792, 2021 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-34524839

RESUMEN

Increase in the size of human neocortex­acquired in evolution­accounts for the unique cognitive capacity of humans. This expansion reflects the evolutionarily enhanced proliferative ability of basal progenitors (BPs), including the basal radial glia and basal intermediate progenitors (bIPs) in mammalian cortex, which may have been acquired through epigenetic alterations in BPs. However, how the epigenome in BPs differs across species is not known. Here, we report that histone H3 acetylation is a key epigenetic regulation in bIP amplification and cortical expansion. Through epigenetic profiling of sorted bIPs, we show that histone H3 lysine 9 acetylation (H3K9ac) is low in murine bIPs and high in human bIPs. Elevated H3K9ac preferentially increases bIP proliferation, increasing the size and folding of the normally smooth mouse neocortex. H3K9ac drives bIP amplification by increasing expression of the evolutionarily regulated gene, Trnp1, in developing cortex. Our findings demonstrate a previously unknown mechanism that controls cortical architecture.

19.
J Neurosci ; 29(26): 8335-49, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19571125

RESUMEN

The transcription factor Pax6 has been implicated in neocortical neurogenesis in vertebrates, including humans. Analyses of the role of Pax6 in layer formation and cognitive abilities have been hampered by perinatal lethality of Pax6 mutants. Here, we generated viable mutants exhibiting timed, restricted inactivation of Pax6 during early and late cortical neurogenesis using Emx1-Cre and hGFAP-Cre lines, respectively. The disruption of Pax6 at the onset of neurogenesis using Emx1-Cre line resulted in premature cell cycle exit of early progenitors, increase of early born neuronal subsets located in the marginal zone and lower layers, and a nearly complete absence of upper layer neurons, especially in the rostral cortex. Furthermore, progenitors, which accumulated in the enlarged germinal neuroepithelium at the pallial/subpallial border in the Pax6 mutants, produced an excess of oligodendrocytes. The inactivation of Pax6 after generation of the lower neuronal layers using hGFAP-Cre line did not affect specification or numbers of late-born neurons, indicating that the severe reduction of upper layer neurons in Pax6 deficiency is mostly attributable to a depletion of the progenitor pool, available for late neurogenesis. We further show that Pax6(fl/fl);Emx1-Cre mutants exhibited deficiencies in sensorimotor information integration, and both hippocampus-dependent short-term and neocortex-dependent long-term memory recall. Because a majority of the morphological and behavior disabilities of the Pax6 mutant mice parallel abnormalities reported for aniridia patients, a condition caused by PAX6 haploinsufficiency, the Pax6 conditional mutant mice generated here represent a valuable genetic tool to understand how the developmental cortical disruption can lead to a human behavior abnormality.


Asunto(s)
Corteza Cerebral/anomalías , Corteza Cerebral/patología , Anomalías Congénitas/genética , Trastornos Mentales/genética , Factores de Transcripción Paired Box/deficiencia , Análisis de Varianza , Animales , Animales Recién Nacidos , Conducta Animal/fisiología , Bromodesoxiuridina/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Ciclo Celular/genética , Diferenciación Celular/genética , Movimiento Celular/genética , Corteza Cerebral/embriología , Corteza Cerebral/crecimiento & desarrollo , Condicionamiento Clásico/fisiología , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos , Proteínas de la Matriz Extracelular/metabolismo , Proteínas del Ojo , Miedo/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas de Homeodominio/genética , Indoles , Antígeno Ki-67/metabolismo , Trastornos de la Memoria/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/genética , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/genética , Neuronas/metabolismo , Factor de Transcripción PAX6 , Umbral del Dolor/fisiología , Desempeño Psicomotor/fisiología , Tiempo de Reacción/genética , Proteína Reelina , Proteínas Represoras , Serina Endopeptidasas/metabolismo , Factores de Transcripción/genética
20.
J Neurosci ; 28(35): 8724-34, 2008 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-18753373

RESUMEN

Transcription factor Pax6 exerts a prominent rostrolateral(high) to caudomedial(low) expression gradient in the cortical progenitors and have been implicated in regulation of area identity in the mammalian cortex. Herein, we analyzed the role of Pax6 in molecular arealization and development of thalamocortical connections in the juvenile cortex-specific conditional Pax6 knock-out mice (Pax6cKO). Using a set of molecular markers of positional identity (Id2, Cadherin6, COUP-TF1, RZRbeta, and EphA7), we show that, in the juvenile Pax6cKO, the relative size of caudal cortical areas (putative visual and somatosensory) are mildly enlarged, whereas the rostral domain (putative motor) is severely reduced. Despite the rostral shift of graded expression of areal markers, the distribution of area-specific thalamocortical and corticofugal projections appear normal in the Pax6cKO. This indicates that change of the size of cortical areas is not accompanied by a change in cortical identity. We show furthermore that, despite a severe depletion of supragranular cortical layers and accumulation of cells along the pallial-subpallial boundary, thalamocortical fibers establish a periphery-related pattern of the somatosensory cortex in normal position in Pax6cKO. Our findings indicate that Pax6 expression gradients in cortical progenitors do not directly impart thalamocortical or corticofugal areal identity.


Asunto(s)
Corteza Cerebral/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica/genética , Morfogénesis/genética , Factores de Transcripción Paired Box/deficiencia , Tálamo/crecimiento & desarrollo , Animales , Animales Recién Nacidos , Factor de Transcripción COUP I/metabolismo , Cadherinas/metabolismo , Corteza Cerebral/metabolismo , Proteínas del Ojo , Proteínas de Homeodominio/genética , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Vías Nerviosas/metabolismo , Miembro 2 del Grupo F de la Subfamilia 1 de Receptores Nucleares , Factor de Transcripción PAX6 , Receptor EphA7/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Represoras , Tálamo/metabolismo , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA