Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
1.
Differentiation ; 137: 100781, 2024.
Article En | MEDLINE | ID: mdl-38631141

Pax6 is a critical transcription factor involved in the development of the central nervous system. However, in humans, mutations in Pax6 predominantly result in iris deficiency rather than neurological phenotypes. This may be attributed to the distinct functions of Pax6 isoforms, Pax6a and Pax6b. In this study, we investigated the spatial and temporal expression patterns of Pax6 isoforms during different stages of mouse eye development. We observed a strong correlation between Pax6a expression and the neuroretina gene Sox2, while Pax6b showed a high correlation with iris-component genes, including the mesenchymal gene Foxc1. During early patterning from E10.5, Pax6b was expressed in the hinge of the optic cup and neighboring mesenchymal cells, whereas Pax6a was absent in these regions. At E14.5, both Pax6a and Pax6b were expressed in the future iris and ciliary body, coinciding with the integration of mesenchymal cells and Mitf-positive cells in the outer region. From E18.5, Pax6 isoforms exhibited distinct expression patterns as lineage genes became more restricted. To further validate these findings, we utilized ESC-derived eye organoids, which recapitulated the temporal and spatial expression patterns of lineage genes and Pax6 isoforms. Additionally, we found that the spatial expression patterns of Foxc1 and Mitf were impaired in Pax6b-mutant ESC-derived eye organoids. This in vitro eye organoids model suggested the involvement of Pax6b-positive local mesodermal cells in iris development. These results provide valuable insights into the regulatory roles of Pax6 isoforms during iris and neuroretina development and highlight the potential of ESC-derived eye organoids as a tool for studying normal and pathological eye development.


Eye , Gene Expression Regulation, Developmental , Organoids , PAX6 Transcription Factor , Protein Isoforms , PAX6 Transcription Factor/genetics , PAX6 Transcription Factor/metabolism , Animals , Mice , Organoids/metabolism , Organoids/growth & development , Organoids/cytology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Eye/growth & development , Eye/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Organogenesis/genetics
2.
Acta Neuropathol ; 147(1): 13, 2024 01 09.
Article En | MEDLINE | ID: mdl-38194050

The development of the cerebral cortex involves a series of dynamic events, including cell proliferation and migration, which rely on the motor protein dynein and its regulators NDE1 and NDEL1. While the loss of function in NDE1 leads to microcephaly-related malformations of cortical development (MCDs), NDEL1 variants have not been detected in MCD patients. Here, we identified two patients with pachygyria, with or without subcortical band heterotopia (SBH), carrying the same de novo somatic mosaic NDEL1 variant, p.Arg105Pro (p.R105P). Through single-cell RNA sequencing and spatial transcriptomic analysis, we observed complementary expression of Nde1/NDE1 and Ndel1/NDEL1 in neural progenitors and post-mitotic neurons, respectively. Ndel1 knockdown by in utero electroporation resulted in impaired neuronal migration, a phenotype that could not be rescued by p.R105P. Remarkably, p.R105P expression alone strongly disrupted neuronal migration, increased the length of the leading process, and impaired nucleus-centrosome coupling, suggesting a failure in nucleokinesis. Mechanistically, p.R105P disrupted NDEL1 binding to the dynein regulator LIS1. This study identifies the first lissencephaly-associated NDEL1 variant and sheds light on the distinct roles of NDE1 and NDEL1 in nucleokinesis and MCD pathogenesis.


Lissencephaly , Humans , Lissencephaly/genetics , Cell Movement/genetics , Cell Proliferation , Cerebral Cortex , Dyneins/genetics , Carrier Proteins , Microtubule-Associated Proteins/genetics
3.
Development ; 151(2)2024 Jan 15.
Article En | MEDLINE | ID: mdl-38149472

Lissencephaly is a neurodevelopmental disorder characterized by a loss of brain surface convolutions caused by genetic variants that disrupt neuronal migration. However, the genetic origins of the disorder remain unidentified in nearly one-fifth of people with lissencephaly. Using whole-exome sequencing, we identified a de novo BAIAP2 variant, p.Arg29Trp, in an individual with lissencephaly with a posterior more severe than anterior (P>A) gradient, implicating BAIAP2 as a potential lissencephaly gene. Spatial transcriptome analysis in the developing mouse cortex revealed that Baiap2 is expressed in the cortical plate and intermediate zone in an anterior low to posterior high gradient. We next used in utero electroporation to explore the effects of the Baiap2 variant in the developing mouse cortex. We found that Baiap2 knockdown caused abnormalities in neuronal migration, morphogenesis and differentiation. Expression of the p.Arg29Trp variant failed to rescue the migration defect, suggesting a loss-of-function effect. Mechanistically, the variant interfered with the ability of BAIAP2 to localize to the cell membrane. These results suggest that the functions of BAIAP2 in the cytoskeleton, cell morphogenesis and migration are important for cortical development and for the pathogenesis of lissencephaly in humans.


Lissencephaly , Animals , Humans , Mice , Brain/metabolism , Cell Movement/genetics , Cytoskeleton/metabolism , Lissencephaly/genetics , Lissencephaly/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism
4.
Front Neurosci ; 16: 844410, 2022.
Article En | MEDLINE | ID: mdl-35422684

Expansion of the neocortex is thought to pave the way toward acquisition of higher cognitive functions in mammals. The highly conserved Notch signaling pathway plays a crucial role in this process by regulating the size of the cortical progenitor pool, in part by controlling the balance between self-renewal and differentiation. In this review, we introduce the components of Notch signaling pathway as well as the different mode of molecular mechanisms, including trans- and cis-regulatory processes. We focused on the recent findings with regard to the expression pattern and levels in regulating neocortical formation in mammals and its interactions with other known signaling pathways, including Slit-Robo signaling and Shh signaling. Finally, we review the functions of Notch signaling pathway in different species as well as other developmental process, mainly somitogenesis, to discuss how modifications to the Notch signaling pathway can drive the evolution of the neocortex.

5.
IUBMB Life ; 74(8): 812-825, 2022 08.
Article En | MEDLINE | ID: mdl-35102668

Recent advances in induced pluripotent stem cell (iPSC) technology have allowed researchers to generate neurodegenerative disease-specific iPSCs and use the cells to derive a variety of relevant cell populations for laboratory modeling and drug testing. Nevertheless, these efforts have faced challenges related to immaturity and lack of complex developmental niches in the derived cell populations, limiting the utility of these in vitro models of neurodegenerative disease. Such limitations may be overcome by using human iPSC technology to generate three-dimensional (3D) brain organoids, which better recapitulate in vivo tissue architecture than traditional neuronal cultures to provide more complex and representative disease models and drug testing systems. In this review, we focus on the application of pluripotent stem cell-derived central nervous system (CNS) organoids to model neurodegenerative diseases. We first summarize recent progress in generating and characterizing various CNS organoids from pluripotent stem cells. We then review the application of CNS organoids for modeling several different human neurodegenerative diseases. We also describe several novel pathological mechanisms and drugs that were studied using patient iPSC-derived CNS organoids. Finally, we discuss remaining challenges and emerging opportunities for the use of 3D brain organoids for in vitro modeling of CNS development and neurodegeneration.


Induced Pluripotent Stem Cells , Neurodegenerative Diseases , Central Nervous System , Humans , Induced Pluripotent Stem Cells/physiology , Neurodegenerative Diseases/pathology , Neurons/pathology , Organoids/pathology , Organoids/physiology
6.
Front Cell Neurosci ; 14: 35, 2020.
Article En | MEDLINE | ID: mdl-32158381

Forkhead Box G1 (FOXG1) is a member of the Forkhead family of genes with non-redundant roles in brain development, where alteration of this gene's expression significantly affects the formation and function of the mammalian cerebral cortex. FOXG1 haploinsufficiency in humans is associated with prominent differences in brain size and impaired intellectual development noticeable in early childhood, while homozygous mutations are typically fatal. As such, FOXG1 has been implicated in a wide spectrum of congenital brain disorders, including the congenital variant of Rett syndrome, infantile spasms, microcephaly, autism spectrum disorder (ASD) and schizophrenia. Recent technological advances have yielded greater insight into phenotypic variations observed in FOXG1 syndrome, molecular mechanisms underlying pathogenesis of the disease, and multifaceted roles of FOXG1 expression. In this review, we explore the emerging mechanisms of FOXG1 in a range of transcriptional to posttranscriptional events in order to evolve our current view of how a single transcription factor governs the assembly of an elaborate cortical circuit responsible for higher cognitive functions and neurological disorders.

7.
Nat Commun ; 10(1): 3581, 2019 08 08.
Article En | MEDLINE | ID: mdl-31395862

The bimodal requisite for a genetic program and external stimuli is a key feature of sensory circuit formation. However, the contribution of cell-intrinsic codes to directing sensory-specific circuits remains unknown. Here, we identify the earliest molecular program that preselects projection neuron types in the sensory neocortex. Mechanistically, Foxg1 binds to an H3K4me1-enriched enhancer site to repress COUP-TFI, where ectopic acquisition of Foxg1 in layer 4 cells transforms local projection neurons to callosal projection neurons with pyramidal morphologies. Removal of Foxg1 in long-range projection neurons, in turn, derepresses COUP-TFI and activates a layer 4 neuron-specific program. The earliest segregation of projection subtypes is achieved through repression of Foxg1 in layer 4 precursors by early growth response genes, the major targets of the transforming growth factor-ß signaling pathway. These findings describe the earliest cortex-intrinsic program that restricts neuronal connectivity in sensory circuits, a fundamental step towards the acquisition of mammalian perceptual behavior.


Gene Expression Regulation/physiology , Neocortex/physiology , Nerve Net/physiology , Neurons/metabolism , Perception/physiology , Animals , COUP Transcription Factor I/metabolism , Early Growth Response Transcription Factors/metabolism , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Male , Mice , Mice, Knockout , Neocortex/cytology , Nerve Net/cytology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism
8.
Methods Mol Biol ; 1650: 319-334, 2017.
Article En | MEDLINE | ID: mdl-28809032

The detection of specific RNA molecules in embryonic tissues has wide research applications including studying gene expression dynamics in brain development and evolution. Recent advances in sequencing technologies have introduced new animal models to explore the molecular principles underlying the assembly and diversification of brain circuits between different amniote species. Here, we provide a step-by-step protocol for a versatile in situ hybridization method that is immediately applicable to a range of amniote embryos including zebra finch and Madagascar ground gecko, two new model organisms that have rapidly emerged for comparative brain studies over recent years. The sensitive detection of transcripts from low to high abundance expression range using the same platform enables direct comparison of gene of interest among different amniotes, providing high-resolution spatiotemporal information of gene expression to dissect the molecular principles underlying brain evolution.


Amnion/metabolism , Brain/metabolism , Finches/genetics , Gene Expression Regulation, Developmental , In Situ Hybridization/methods , Lizards/genetics , Amnion/embryology , Animals , Brain/embryology , Madagascar
9.
Stem Cell Reports ; 8(1): 54-68, 2017 01 10.
Article En | MEDLINE | ID: mdl-27940274

Early human embryonic stem cell (hESC)-derived neural populations consist of various embryonic neural progenitors (ENPs) with broad neural developmental propensity. Here, we sought to directly convert human somatic cells into ENP-like phenotypes using hESC-ENP-enriched neural transcription factors (TFs). We demonstrated that induced ENP could be efficiently converted from human fibroblasts using two TF combinations. The iENPs exhibit cellular and molecular characteristics resembling hESC-ENPs and can give rise to astrocytes, oligodendrocytes, and functional neuronal subtypes of the central and peripheral nervous system. Nevertheless, our analyses further revealed that these two iENP populations differ in terms of their proliferation ability and neuronal propensity. Finally, we demonstrated that the iENPs can be induced from fibroblasts from patients with Huntington's disease and Alzheimer's disease, and the diseased iENPs and their neuronal derivatives recapitulated the hallmark pathological features of the diseases. Collectively, our results point toward a promising strategy for generating iENPs from somatic cells for disease modeling and future clinical intervention.


Cell Transdifferentiation/genetics , Cellular Reprogramming/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Transcription Factors/genetics , Animals , Brain/metabolism , Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression , Humans , Neurons , Rats , Transcription Factors/metabolism
10.
Cell Reprogram ; 16(6): 467-76, 2014 Dec.
Article En | MEDLINE | ID: mdl-25364979

Induced pluripotent stem cells (iPSCs) are reprogrammed from somatic cells via ectopic gene expression and, similarly to embryonic stem cells (ESCs), possess powerful abilities to self-renew and differentiate into cells of various lineages. However, the neural differentiation potency of iPSCs remains unknown. In this study, we demonstrated the neural differentiation ability of iPSCs compared with ESCs using an retinoic acid (RA) induction system. The neural differentiation efficiency of iPSCs was obviously lower than that of ESCs. Retinoic acid receptor-α (RARα) was critical in the RA-induced neural differentiation of iPSCs, and the effect of RARα was confirmed by applying a specific RARα antagonist ER50891 to ESCs. These findings indicate that iPSCs do not possess the complete properties that ESCs have.


Cell Differentiation/drug effects , Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , Neurons/cytology , Receptors, Retinoic Acid/metabolism , Animals , Cells, Cultured , Embryonic Stem Cells/cytology , Mice , Receptors, Retinoic Acid/antagonists & inhibitors , Retinoic Acid Receptor alpha , Signal Transduction , Tretinoin/pharmacology
11.
Stem Cell Reports ; 2(2): 189-204, 2014 Feb 11.
Article En | MEDLINE | ID: mdl-24527393

The mechanisms of transcriptional regulation underlying human primordial germ cell (PGC) differentiation are largely unknown. The transcriptional repressor Prdm1/Blimp-1 is known to play a critical role in controlling germ cell specification in mice. Here, we show that PRDM1 is expressed in developing human gonads and contributes to the determination of germline versus neural fate in early development. We show that knockdown of PRDM1 in human embryonic stem cells (hESCs) impairs germline potential and upregulates neural genes. Conversely, ectopic expression of PRDM1 in hESCs promotes the generation of cells that exhibit phenotypic and transcriptomic features of early PGCs. Furthermore, PRDM1 suppresses transcription of SOX2. Overexpression of SOX2 in hESCs under conditions favoring germline differentiation skews cell fate from the germline to the neural lineage. Collectively, our results demonstrate that PRDM1 serves as a molecular switch to modulate the divergence of neural or germline fates through repression of SOX2 during human development.


Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Germ Cells/cytology , Germ Cells/metabolism , Repressor Proteins/metabolism , SOXB1 Transcription Factors/genetics , Bone Morphogenetic Protein 4/metabolism , Cell Differentiation/genetics , Fetus/embryology , Fetus/metabolism , Gonads/embryology , Gonads/metabolism , Humans , Models, Biological , Positive Regulatory Domain I-Binding Factor 1 , Repressor Proteins/genetics , Wnt3A Protein/metabolism
12.
Proc Natl Acad Sci U S A ; 110(50): E4913-21, 2013 Dec 10.
Article En | MEDLINE | ID: mdl-24262147

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.


Gene Expression Regulation/physiology , LIM-Homeodomain Proteins/metabolism , Somatosensory Cortex/embryology , Transcription Factors/metabolism , Animals , Chromatin Immunoprecipitation , Ephrin-A5/metabolism , Evoked Potentials/physiology , Eye Proteins/metabolism , Homeodomain Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , Integrases , LIM-Homeodomain Proteins/deficiency , Mice , Mice, Transgenic , Microscopy, Fluorescence , Neural Pathways/embryology , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Repressor Proteins/metabolism , Somatosensory Cortex/metabolism , Transcription Factors/deficiency
13.
Nucleic Acids Res ; 41(16): 7753-70, 2013 Sep.
Article En | MEDLINE | ID: mdl-23804753

The LIM homeobox 2 transcription factor Lhx2 is known to control crucial aspects of neural development in various species. However, its function in human neural development is still elusive. Here, we demonstrate that LHX2 plays a critical role in human neural differentiation, using human embryonic stem cells (hESCs) as a model. In hESC-derived neural progenitors (hESC-NPs), LHX2 was found to be expressed before PAX6, and co-expressed with early neural markers. Conditional ectopic expression of LHX2 promoted neural differentiation, whereas disruption of LHX2 expression in hESCs significantly impaired neural differentiation. Furthermore, we have demonstrated that LHX2 regulates neural differentiation at two levels: first, it promotes expression of PAX6 by binding to its active enhancers, and second, it attenuates BMP and WNT signaling by promoting expression of the BMP and WNT antagonist Cerberus 1 gene (CER1), to inhibit non-neural differentiation. These findings indicate that LHX2 regulates the transcription of downstream intrinsic and extrinsic molecules that are essential for early neural differentiation in human.


Embryonic Stem Cells/metabolism , Gene Expression Regulation , LIM-Homeodomain Proteins/metabolism , Neural Stem Cells/metabolism , Neurogenesis/genetics , Transcription Factors/metabolism , Transcription, Genetic , Cells, Cultured , Cytokines/biosynthesis , Cytokines/genetics , Embryonic Stem Cells/cytology , Enhancer Elements, Genetic , Eye Proteins/biosynthesis , Eye Proteins/genetics , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , LIM-Homeodomain Proteins/antagonists & inhibitors , LIM-Homeodomain Proteins/genetics , Neural Stem Cells/cytology , PAX6 Transcription Factor , Paired Box Transcription Factors/biosynthesis , Paired Box Transcription Factors/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
14.
Stem Cells ; 28(9): 1510-7, 2010 Sep.
Article En | MEDLINE | ID: mdl-20629177

The zinc finger Krüppel-like transcription factor 4 (KLF4) has been implicated in cancer formation and stem cell regulation. However, the function of KLF4 in tumorigenesis and stem cell regulation are poorly understood due to limited knowledge of its targets in these cells. In this study, we have revealed a surprising link between KLF4 and regulation of telomerase that offers important insight into how KLF4 contributes to cancer formation and stem cell regulation. KLF4 sufficiently activated expression of the human telomerase catalytic subunit, human telomerase reverse transcriptase (hTERT), in telomerase-low alternative lengthening of telomeres (ALT), and fibroblast cells, while downregulation of KLF4 reduced its expression in cancerous and stem cells, which normally exhibits high expression. Furthermore, KLF4-dependent induction of hTERT was mediated by a KLF4 binding site in the proximal promoter region of hTERT. In human embryonic stem cells, expression of hTERT replaced KLF4 function to maintain their self-renewal. Therefore, our findings demonstrate that hTERT is one of the major targets of KLF4 in cancer and stem cells to maintain long-term proliferation potential.


Carcinoma, Squamous Cell/enzymology , Embryonic Stem Cells/enzymology , Kruppel-Like Transcription Factors/metabolism , Telomerase/metabolism , Animals , Binding Sites , Carcinoma, Squamous Cell/pathology , Cell Line , Cell Proliferation , Enzyme Activation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Mice , Promoter Regions, Genetic , RNA Interference , Telomerase/genetics , Transcriptional Activation , Transfection , Two-Hybrid System Techniques
15.
Stem Cells Dev ; 19(6): 763-71, 2010 Jun.
Article En | MEDLINE | ID: mdl-19740021

Permanent and reversible genetic modifications are important approaches to study gene function in different cell types. They are also important for stem cell researchers to explore and test the therapeutic potential of stem cells. The piggyBac transposon from insects is a rising nonviral system that efficiently mutagenizes and mediates gene transfer into the mammalian genome. It is also characterized by its precise excision, leaving no trace sequence behind so that the genomic integrity of the mutated cell can be restored. Here, we use an optimized piggyBac transposon system to mediate gene transfer and expression of a bifunctional fluorescent reporter in human embryonic stem (ES) cells. We provide molecular evidence for transposase-mediated piggyBac integration events and functional evidence for successful expression of a transferred fluorescent protein genes in human ES cells and their in vitro differentiated derivatives. We also demonstrate that the integrated piggyBac transposon can be removed and an undisrupted insertion site can be restored, which implies potential applications for its use in gene therapy and genetics studies.


DNA Transposable Elements/genetics , Embryonic Stem Cells/metabolism , Gene Transfer Techniques , Animals , Base Sequence , Cell Differentiation/genetics , Cell Line , Cell Shape , Chickens , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Humans , Molecular Sequence Data , Transgenes/genetics , Transposases/metabolism
...