Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 33
2.
Curr Opin Neurobiol ; 80: 102698, 2023 06.
Article En | MEDLINE | ID: mdl-36893490

The cerebral cortex is compartmentalized into multiple regions, including the newly evolved neocortex and evolutionarily older paleocortex and archicortex. These broad cortical regions can be further subdivided into different functional domains, each with its own unique cytoarchitecture and distinct set of input and output projections to perform specific functions. While many excitatory projection neurons show region-specific gene expression profiles, the cells are derived from the seemingly uniform progenitors in the dorsal telencephalon. Much progress has been made in defining the genetic mechanisms involved in generating the morphological and functional diversity of the central nervous system. In this review, we summarize the current knowledge of mouse corticogenesis and discuss key events involved in cortical patterning during early developmental stages.


Cerebral Cortex , Neocortex , Mice , Animals , Gene Expression Regulation, Developmental/genetics
3.
Cell Death Dis ; 14(1): 52, 2023 01 21.
Article En | MEDLINE | ID: mdl-36681682

Patients with mutations of WDR4, a substrate adaptor of the CUL4 E3 ligase complex, develop cerebellar atrophy and gait phenotypes. However, the underlying mechanisms remain unexplored. Here, we identify a crucial role of Wdr4 in cerebellar development. Wdr4 deficiency in granule neuron progenitors (GNPs) not only reduces foliation and the sizes of external and internal granular layers but also compromises Purkinje neuron organization and the size of the molecular layer, leading to locomotion defects. Mechanistically, Wdr4 supports the proliferation of GNPs by preventing their cell cycle exit. This effect is mediated by Wdr4-induced ubiquitination and degradation of Arhgap17, thereby activating Rac1 to facilitate cell cycle progression. Disease-associated Wdr4 variants, however, cannot provide GNP cell cycle maintenance. Our study identifies Wdr4 as a previously unappreciated participant in cerebellar development and locomotion, providing potential insights into treatment strategies for diseases with WDR4 mutations, such as primordial dwarfism and Galloway-Mowat syndrome.


Microcephaly , Neurogenesis , Humans , Neurogenesis/physiology , Neurons/metabolism , Purkinje Cells/metabolism , Microcephaly/genetics , Locomotion , Cerebellum , GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism
4.
Cereb Cortex ; 33(5): 1693-1707, 2023 02 20.
Article En | MEDLINE | ID: mdl-35512682

Establishing neuronal circuits requires interactions between pre- and postsynaptic neurons. While presynaptic neurons were shown to play instructive roles for the postsynaptic neurons, how postsynaptic neurons provide feedback to regulate the presynaptic neuronal development remains elusive. To elucidate the mechanisms for circuit formation, we study the development of barrel cortex (the primary sensory cortex, S1), whose development is instructed by presynaptic thalamocortical axons (TCAs). In the first postnatal weeks, TCA terminals arborize in layer (L) 4 to fill in the barrel center, but it is unclear how TCA development is regulated. Here, we reported that the deletion of Lhx2 specifically in the cortical neurons in the conditional knockout (cKO) leads to TCA arborization defects, which is accompanied with deficits in sensory-evoked and spontaneous cortical activities and impaired lesion-induced plasticity following early whisker follicle ablation. Reintroducing Lhx2 back in L4 neurons in cKO ameliorated TCA arborization and plasticity defects. By manipulating L4 neuronal activity, we further demonstrated that Lhx2 induces TCA arborization via an activity-dependent mechanism. Additionally, we identified the extracellular signaling protein Sema7a as an activity-dependent downstream target of Lhx2 in regulating TCA branching. Thus, we discovered a bottom-up feedback mechanism for the L4 neurons to regulate TCA development.


Neurons , Thalamus , Feedback , Thalamus/physiology , Neurons/physiology , Axons/physiology , Signal Transduction , Somatosensory Cortex/physiology
5.
J Neurosci ; 42(22): 4435-4448, 2022 06 01.
Article En | MEDLINE | ID: mdl-35501157

The whiskers of rodents are a key sensory organ that provides critical tactile information for animal navigation and object exploration throughout life. Previous work has explored the developmental sensory-driven activation of the primary sensory cortex processing whisker information (wS1), also called barrel cortex. This body of work has shown that the barrel cortex is already activated by sensory stimuli during the first postnatal week. However, it is currently unknown when over the course of development these stimuli begin being processed by higher-order cortical areas, such as secondary whisker somatosensory area (wS2). Here we investigate the developmental engagement of wS2 by whisker stimuli and the emergence of corticocortical communication from wS1 to wS2. Using in vivo wide-field imaging and multielectrode recordings in control and conditional KO mice of either sex with thalamocortical innervation defects, we find that wS1 and wS2 are able to process bottom-up information coming from the thalamus from birth. We also identify that it is only at the end of the first postnatal week that wS1 begins to provide functional excitation into wS2, switching to more inhibitory actions after the second postnatal week. Therefore, we have uncovered a developmental window when information transfer between wS1 and wS2 reaches mature function.SIGNIFICANCE STATEMENT At the end of the first postnatal week, the primary whisker somatosensory area starts providing excitatory input to the secondary whisker somatosensory area 2. This excitatory drive weakens during the second postnatal week and switches to inhibition in the adult.


Somatosensory Cortex , Vibrissae , Animals , Mice , Somatosensory Cortex/physiology , Thalamus , Touch/physiology , Vibrissae/innervation
6.
Sci Adv ; 7(27)2021 07.
Article En | MEDLINE | ID: mdl-34215582

Development of cortical regions with precise, sharp, and regular boundaries is essential for physiological function. However, little is known of the mechanisms ensuring these features. Here, we show that determination of the boundary between neocortex and medial entorhinal cortex (MEC), two abutting cortical regions generated from the same progenitor lineage, relies on COUP-TFI (chicken ovalbumin upstream promoter-transcription factor I), a patterning transcription factor with graded expression in cortical progenitors. In contrast with the classical paradigm, we found that increased COUP-TFI expression expands MEC, creating protrusions and disconnected ectopic tissue. We further developed a mathematical model that predicts that neuronal specification and differential cell affinity contribute to the emergence of an instability region and boundary sharpness. Correspondingly, we demonstrated that high expression of COUP-TFI induces MEC cell fate and protocadherin 19 expression. Thus, we conclude that a sharp boundary requires a subtle interplay between patterning transcription factors and differential cell affinity.


Neocortex , COUP Transcription Factor I/metabolism , Cell Adhesion , Entorhinal Cortex , Neocortex/metabolism , Transcription Factors/metabolism
7.
Sci Rep ; 11(1): 8349, 2021 04 16.
Article En | MEDLINE | ID: mdl-33863910

The piriform cortex (PC) is a major cortical processing center for the sense of smell that receives direct inputs from the olfactory bulb. In mice, the PC consists of three neuronal layers, which are populated by cells with distinct developmental origins. One origin of PC neurons is the pool of Dbx1-expressing neural progenitors located in the ventral pallium at the pallial-subpallial boundary. Since the precise mechanisms of PC neuron development are largely unknown, we sought to define the distribution, timing of neurogenesis, morphology and projection patterns of PC neurons from the Dbx1 lineage. We found that Dbx1-lineage neurons are preferentially distributed in layer 2 and enriched in the ventral portion of the PC. Further, Dbx1 neurons are early-born neurons and contribute to most neuronal subtypes in the PC. Our data also revealed an enrichment of Dbx1-lineage neurons in the ventral anterior PC that project to the orbitofrontal cortex. These findings suggest a specific association between the developmental origin of PC neurons and their neuronal properties.


Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Neurogenesis/physiology , Neurons/metabolism , Neurons/physiology , Piriform Cortex/cytology , Piriform Cortex/physiology , Smell , Animals , Gene Expression , Mice, Knockout , Olfactory Bulb/physiology , Stem Cells/metabolism , Stem Cells/physiology
8.
Sci Adv ; 7(7)2021 02.
Article En | MEDLINE | ID: mdl-33579706

The role of protein stabilization in cortical development remains poorly understood. A recessive mutation in the USP11 gene is found in a rare neurodevelopmental disorder with intellectual disability, but its pathogenicity and molecular mechanism are unknown. Here, we show that mouse Usp11 is expressed highly in embryonic cerebral cortex, and Usp11 deficiency impairs layer 6 neuron production, delays late-born neuronal migration, and disturbs cognition and anxiety behaviors. Mechanistically, these functions are mediated by a previously unidentified Usp11 substrate, Sox11. Usp11 ablation compromises Sox11 protein accumulation in the developing cortex, despite the induction of Sox11 mRNA. The disease-associated Usp11 mutant fails to stabilize Sox11 and is unable to support cortical neurogenesis and neuronal migration. Our findings define a critical function of Usp11 in cortical development and highlight the importance of orchestrating protein stabilization mechanisms into transcription regulatory programs for a robust induction of cell fate determinants during early brain development.


Cerebral Cortex , Neurogenesis , Animals , Cell Differentiation , Cell Movement , Cerebral Cortex/metabolism , Mice , Neurons/physiology , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism
10.
Cereb Cortex ; 30(5): 3015-3029, 2020 05 14.
Article En | MEDLINE | ID: mdl-31838488

Establishing a balance between excitation and inhibition is critical for brain functions. However, how inhibitory interneurons (INs) generated in the ventral telencephalon integrate with the excitatory neurons generated in the dorsal telencephalon remains elusive. Previous studies showed that INs migrating tangentially to enter the neocortex (NCx), remain in the migratory stream for days before invading the cortical plate during late corticogenesis. Here we show that in developing mouse cortices, INs in the piriform cortex (PCx; the major olfactory cortex) distribute differently from those in the NCx. We provide evidence that during development INs invade and mature earlier in PCx than in NCx, likely owing to the lack of CXCR4 expression in INs from PCx compared to those in NCx. We analyzed IN distribution patterns in Lhx2 cKO mice, where projection neurons in the lateral NCx are re-fated to generate an ectopic PCx (ePCx). The PCx-specific IN distribution patterns found in ePCx suggest that properties of PCx projection neurons regulate IN distribution. Collectively, our results show that the timing of IN invasion in the developing PCx fundamentally differs from what is known in the NCx. Further, our results suggest that projection neurons instruct the PCx-specific pattern of IN distribution.


Interneurons/physiology , Neocortex/embryology , Neocortex/growth & development , Piriform Cortex/enzymology , Piriform Cortex/growth & development , Age Factors , Animals , Mice , Mice, Knockout , Mice, Transgenic , Neocortex/cytology , Neurogenesis/physiology , Piriform Cortex/cytology
11.
Front Behav Neurosci ; 13: 5, 2019.
Article En | MEDLINE | ID: mdl-30728769

The rodent olfactory bulb (OB) contains two distinct populations of postnatally born interneurons, mainly granule cells (GCs), to support local circuits throughout life. During the early postnatal period (i.e., 2 weeks after birth), GCs are mostly produced locally from progenitor cells in the OB with a proportion of them deriving from proliferating cells in the rostral migratory stream (RMS). Afterward, the replenishment of GCs involves differentiated neuroblasts from the subventricular zone (SVZ) in a process known as adult neurogenesis. Although numerous studies have addressed the role of SVZ-born GCs in olfactory behaviors, the function of GCs produced early postnatally in the OB remains elusive. Our previous study demonstrated that the translational regulator, cytoplasmic polyadenylation element-binding protein 4 (CPEB4), is a survival factor exclusively for neonate-born but not SVZ/adult-derived GCs, so CPEB4-knockout (KO) mice provide unique leverage to study early postnatal-born GC-regulated olfactory functions. CPEB4-KO mice with hypoplastic OBs showed normal olfactory sensitivity and short-term memory, but impaired ability to spontaneously discriminate two odors. Such olfactory dysfunction was recapitulated in specific ablation of Cpeb4 gene in inhibitory interneurons but not in excitatory projection neurons or SVZ-derived interneurons. The continuous supply of GCs from adult neurogenesis eventually restored the OB size but not the discrimination function in 6-month-old KO mice. Hence, in the early postnatal OB, whose function cannot be replaced by adult-born GCs, construct critical circuits for odor discrimination.

12.
Brain Res ; 1705: 1-14, 2019 02 15.
Article En | MEDLINE | ID: mdl-29522720

A hundred years after Lhx2 ortholog apterous was identified as a critical regulator of wing development in Drosophila, LIM-HD gene family members have proved to be versatile and powerful components of the molecular machinery that executes the blueprint of embryogenesis across vertebrate and invertebrate species. Here, we focus on the spatio-temporally varied functions of LIM-homeodomain transcription factor LHX2 in the developing mouse forebrain. Right from its earliest known role in telencephalic and eye field patterning, to the control of the neuron-glia cell fate switch, and the regulation of axon pathfinding and dendritic arborization in late embryonic stages, LHX2 has been identified as a fundamental, temporally dynamic, always necessary, and often sufficient factor in a range of critical developmental phenomena. While Lhx2 mutant phenotypes have been characterized in detail in multiple brain structures, only recently have we advanced in our understanding of the molecular mechanisms by which this factor acts. Common themes emerge from how this multifunctional molecule controls a range of developmental steps in distinct forebrain structures. Examining these shared features, and noting unique aspects of LHX2 function is likely to inform our understanding of how a single factor can bring about a diversity of effects and play central and critical roles across systems and stages. The parallels in LHX2 and APTEROUS functions, and the protein complexes they participate in, offer insights into evolutionary strategies that conserve tool kits and deploy them to play new, yet familiar roles in species separated by hundreds of millions of years.


LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/physiology , Prosencephalon/embryology , Transcription Factors/metabolism , Transcription Factors/physiology , Animals , Biological Evolution , Cell Differentiation , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/genetics , Mice , Neurogenesis/physiology , Neuroglia/metabolism , Neurons/metabolism , Prosencephalon/metabolism , Spatio-Temporal Analysis , Transcription Factors/genetics
13.
Cell Rep ; 18(4): 849-856, 2017 01 24.
Article En | MEDLINE | ID: mdl-28122236

Cortical neurons must be specified and make the correct connections during development. Here, we examine a mechanism initiating neuronal circuit formation in the barrel cortex, a circuit comprising thalamocortical axons (TCAs) and layer 4 (L4) neurons. When Lhx2 is selectively deleted in postmitotic cortical neurons using conditional knockout (cKO) mice, L4 neurons in the barrel cortex are initially specified but fail to form cellular barrels or develop polarized dendrites. In Lhx2 cKO mice, TCAs from the thalamic ventral posterior nucleus reach the barrel cortex but fail to further arborize to form barrels. Several activity-regulated genes and genes involved in regulating barrel formation are downregulated in the Lhx2 cKO somatosensory cortex. Among them, Btbd3, an activity-regulated gene controlling dendritic development, is a direct downstream target of Lhx2. We find that Lhx2 confers neuronal competency for activity-dependent dendritic development in L4 neurons by inducing the expression of Btbd3.


Gene Expression , LIM-Homeodomain Proteins/metabolism , Neurons/metabolism , Somatosensory Cortex/physiology , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Animals , Cell Line, Tumor , Chromatin Immunoprecipitation , Ephrin-A5/genetics , Ephrin-A5/metabolism , Evoked Potentials , In Situ Hybridization , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/pathology , Nuclear Receptor Subfamily 1, Group F, Member 2/genetics , Nuclear Receptor Subfamily 1, Group F, Member 2/metabolism , Patch-Clamp Techniques , Promoter Regions, Genetic , Thalamus/metabolism , Transcription Factors/genetics
14.
J Neurosci ; 36(45): 11394-11401, 2016 11 09.
Article En | MEDLINE | ID: mdl-27911741

This review accompanies a 2016 SFN mini-symposium presenting examples of current studies that address a central question: How do neural stem cells (NSCs) divide in different ways to produce heterogeneous daughter types at the right time and in proper numbers to build a cerebral cortex with the appropriate size and structure? We will focus on four aspects of corticogenesis: cytokinesis events that follow apical mitoses of NSCs; coordinating abscission with delamination from the apical membrane; timing of neurogenesis and its indirect regulation through emergence of intermediate progenitors; and capacity of single NSCs to generate the correct number and laminar fate of cortical neurons. Defects in these mechanisms can cause microcephaly and other brain malformations, and understanding them is critical to designing diagnostic tools and preventive and corrective therapies.


Cerebral Cortex/cytology , Cerebral Cortex/physiology , Cytokinesis/physiology , Neurogenesis/physiology , Neurons/cytology , Neurons/physiology , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Humans , Neuronal Plasticity/physiology
15.
Sci Rep ; 6: 19274, 2016 Jan 20.
Article En | MEDLINE | ID: mdl-26786896

Allen Brain Atlas (ABA) provides a valuable resource of spatial/temporal gene expressions in mammalian brains. Despite rich information extracted from this database, current analyses suffer from several limitations. First, most studies are either gene-centric or region-centric, thus are inadequate to capture the superposition of multiple spatial-temporal patterns. Second, standard tools of expression analysis such as matrix factorization can capture those patterns but do not explicitly incorporate spatial dependency. To overcome those limitations, we proposed a computational method to detect recurrent patterns in the spatial-temporal gene expression data of developing mouse brains. We demonstrated that regional distinction in brain development could be revealed by localized gene expression patterns. The patterns expressed in the forebrain, medullary and pontomedullary, and basal ganglia are enriched with genes involved in forebrain development, locomotory behavior, and dopamine metabolism respectively. In addition, the timing of global gene expression patterns reflects the general trends of molecular events in mouse brain development. Furthermore, we validated functional implications of the inferred patterns by showing genes sharing similar spatial-temporal expression patterns with Lhx2 exhibited differential expression in the embryonic forebrains of Lhx2 mutant mice. These analysis outcomes confirm the utility of recurrent expression patterns in studying brain development.


Brain/metabolism , Gene Expression Profiling , Gene Expression Regulation , Transcriptome , Animals , Cluster Analysis , Computational Biology , Gene Expression Regulation, Developmental , LIM-Homeodomain Proteins/deficiency , Mice , Mice, Knockout , Transcription Factors/deficiency
16.
Elife ; 42015 Dec 24.
Article En | MEDLINE | ID: mdl-26705332

In mammals, the neocortical layout consists of few modality-specific primary sensory areas and a multitude of higher order ones. Abnormal layout of cortical areas may disrupt sensory function and behavior. Developmental genetic mechanisms specify primary areas, but mechanisms influencing higher order area properties are unknown. By exploiting gain-of and loss-of function mouse models of the transcription factor Emx2, we have generated bi-directional changes in primary visual cortex size in vivo and have used it as a model to show a novel and prominent function for genetic mechanisms regulating primary visual area size and also proportionally dictating the sizes of surrounding higher order visual areas. This finding redefines the role for intrinsic genetic mechanisms to concomitantly specify and scale primary and related higher order sensory areas in a linear fashion.


Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Visual Cortex/anatomy & histology , Visual Cortex/physiology , Animals , Mice, Inbred C57BL , Mice, Transgenic , Vision, Ocular
17.
Proc Natl Acad Sci U S A ; 112(39): 12199-204, 2015 Sep 29.
Article En | MEDLINE | ID: mdl-26371318

The timing of cortical neurogenesis has a major effect on the size and organization of the mature cortex. The deletion of the LIM-homeodomain transcription factor Lhx2 in cortical progenitors by Nestin-cre leads to a dramatically smaller cortex. Here we report that Lhx2 regulates the cortex size by maintaining the cortical progenitor proliferation and delaying the initiation of neurogenesis. The loss of Lhx2 in cortical progenitors results in precocious radial glia differentiation and a temporal shift of cortical neurogenesis. We further investigated the underlying mechanisms at play and demonstrated that in the absence of Lhx2, the Wnt/ß-catenin pathway failed to maintain progenitor proliferation. We developed and applied a mathematical model that reveals how precocious neurogenesis affected cortical surface and thickness. Thus, we concluded that Lhx2 is required for ß-catenin function in maintaining cortical progenitor proliferation and controls the timing of cortical neurogenesis.


Cerebral Cortex/embryology , Gene Expression Regulation, Developmental/genetics , LIM-Homeodomain Proteins/metabolism , Models, Neurological , Neurogenesis/genetics , Neurogenesis/physiology , Transcription Factors/metabolism , Animals , Computer Simulation , DNA Primers/genetics , Galactosides , Gene Expression Regulation, Developmental/physiology , HEK293 Cells , Humans , In Situ Hybridization , Indoles , LIM-Homeodomain Proteins/genetics , Luciferases , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Transcription Factors/genetics , Wnt Signaling Pathway/physiology , beta Catenin/metabolism
18.
Proc Natl Acad Sci U S A ; 112(21): 6736-41, 2015 May 26.
Article En | MEDLINE | ID: mdl-25971728

Current knowledge suggests that cortical sensory area identity is controlled by transcription factors (TFs) that specify area features in progenitor cells and subsequently their progeny in a one-step process. However, how neurons acquire and maintain these features is unclear. We have used conditional inactivation restricted to postmitotic cortical neurons in mice to investigate the role of the TF LIM homeobox 2 (Lhx2) in this process and report that in conditional mutant cortices area patterning is normal in progenitors but strongly affected in cortical plate (CP) neurons. We show that Lhx2 controls neocortical area patterning by regulating downstream genetic and epigenetic regulators that drive the acquisition of molecular properties in CP neurons. Our results question a strict hierarchy in which progenitors dominate area identity, suggesting a novel and more comprehensive two-step model of area patterning: In progenitors, patterning TFs prespecify sensory area blueprints. Sequentially, sustained function of alignment TFs, including Lhx2, is essential to maintain and to translate the blueprints into functional sensory area properties in cortical neurons postmitotically. Our results reemphasize critical roles for Lhx2 that acts as one of the terminal selector genes in controlling principal properties of neurons.


LIM-Homeodomain Proteins/physiology , Models, Neurological , Neocortex/growth & development , Neocortex/physiology , Transcription Factors/physiology , Animals , Body Patterning/genetics , Body Patterning/physiology , Epigenesis, Genetic , LIM-Homeodomain Proteins/deficiency , LIM-Homeodomain Proteins/genetics , Mice , Mice, Knockout , Mitosis , Neocortex/cytology , Neural Pathways/cytology , Neural Pathways/growth & development , Neural Pathways/physiology , Neurons/cytology , Neurons/physiology , Transcription Factors/deficiency , Transcription Factors/genetics
19.
Proc Natl Acad Sci U S A ; 111(13): E1240-8, 2014 Apr 01.
Article En | MEDLINE | ID: mdl-24639535

Breast cancer susceptibility gene 1 (BRCA1) is a breast and ovarian cancer tumor suppressor whose loss leads to DNA damage and defective centrosome functions. Despite its tumor suppression functions, BRCA1 is most highly expressed in the embryonic neuroepithelium when the neural progenitors are highly proliferative. To determine its functional significance, we deleted BRCA1 in the developing brain using a neural progenitor-specific driver. The phenotype is characterized by severe agenesis of multiple laminated cerebral structures affecting most notably the neocortex, hippocampus, cerebellum, and olfactory bulbs. Major phenotypes are caused by excess apoptosis, as these could be significantly suppressed by the concomitant deletion of p53. Certain phenotypes attributable to centrosomal and cell polarity functions could not be rescued by p53 deletion. A double KO with the DNA damage sensor kinase ATM was able to rescue BRCA1 loss to a greater extent than p53. Our results suggest distinct apoptotic and centrosomal functions of BRCA1 in neural progenitors, with important implications to understand the sensitivity of the embryonic brain to DNA damage, as well as the developmental regulation of brain size.


BRCA1 Protein/metabolism , Brain/embryology , Brain/metabolism , Animals , Apoptosis , Ataxia Telangiectasia Mutated Proteins/metabolism , Brain/cytology , Cell Polarity , Cell Proliferation , Cell Survival , Cognition/physiology , Gene Deletion , Learning , Mice , Mice, Knockout , Motor Activity/physiology , Nestin/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Phenotype , Signal Transduction , Tumor Suppressor Protein p53/metabolism
20.
Nat Neurosci ; 17(2): 240-7, 2014 Feb.
Article En | MEDLINE | ID: mdl-24441682

The neuron-specific transcription factor T-box brain 1 (TBR1) regulates brain development. Disruptive mutations in the TBR1 gene have been repeatedly identified in patients with autism spectrum disorders (ASDs). Here, we show that Tbr1 haploinsufficiency results in defective axonal projections of amygdalar neurons and the impairment of social interaction, ultrasonic vocalization, associative memory and cognitive flexibility in mice. Loss of a copy of the Tbr1 gene altered the expression of Ntng1, Cntn2 and Cdh8 and reduced both inter- and intra-amygdalar connections. These developmental defects likely impair neuronal activation upon behavioral stimulation, which is indicated by fewer c-FOS-positive neurons and lack of GRIN2B induction in Tbr1(+/-) amygdalae. We also show that upregulation of amygdalar neuronal activity by local infusion of a partial NMDA receptor agonist, d-cycloserine, ameliorates the behavioral defects of Tbr1(+/-) mice. Our study suggests that TBR1 is important in the regulation of amygdalar axonal connections and cognition.


Amygdala/pathology , Axons/pathology , Cognition Disorders/genetics , Cognition Disorders/pathology , DNA-Binding Proteins/deficiency , Animals , Antimetabolites/therapeutic use , Axons/metabolism , Cadherins/metabolism , Cognition Disorders/drug therapy , Contactin 2/metabolism , Cycloserine/therapeutic use , Disease Models, Animal , Exploratory Behavior/physiology , Gene Expression Profiling , Gene Expression Regulation/genetics , MEF2 Transcription Factors/metabolism , Magnetic Resonance Imaging , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/metabolism , Netrins , Oligonucleotide Array Sequence Analysis , Organ Culture Techniques , Proto-Oncogene Proteins c-fos/metabolism , T-Box Domain Proteins
...