1
|
Ürel-Demir G, Başer B, Göçmen R, Şimşek-Kiper PÖ, Utine GE, Haliloğlu G. Many Faces of Diencephalic-Mesencephalic Junction Dysplasia Syndrome with GSX2 and PCDH12 Variants. Mol Syndromol 2024; 15:275-283. [PMID: 39119454 PMCID: PMC11305698 DOI: 10.1159/000537831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/11/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction Diencephalic-mesencephalic junction dysplasia syndrome is a rare neurogenetic disorder reported to be caused by variants in several genes. Phenotypic presentation is characterized by clinical findings including developmental delay, hypotonia, spasticity, and dyskinetic movements in combination with distinctive imaging features on brain magnetic resonance imaging (MRI). Methods Whole exome sequencing was conducted to unveil the molecular etiology of patients presenting with neurological manifestations from two unrelated families. Results To the best of our knowledge, here we report the third family affected with diencephalic-mesencephalic junction dysplasia caused by a novel variant in GSX2 and two siblings with a PCDH12 variant exhibiting a less severe phenotype. The siblings with a PCDH12 variant were positioned at the milder end of the phenotypic spectrum. Although both exhibited a clinical phenotype resembling cerebral palsy, one showed partial fusion of the hypothalamus and mesencephalon, whereas MRI was unremarkable in the other. Biallelic GSX2 variants have been implicated in basal ganglia agenesis, and similarly, our patients had basal ganglia hypoplasia along with hypothalamic-mesencephalic fusion. Conclusion Identifying variants associated with the syndrome in different genes will contribute to genotype-phenotype correlation.
Collapse
Affiliation(s)
- Gizem Ürel-Demir
- Division of Pediatric Genetics, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Burak Başer
- Department of Medical Genetics, Mersin City Training and Research Hospital, Mersin, Turkey
| | - Rahşan Göçmen
- Department of Radiology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Pelin Özlem Şimşek-Kiper
- Division of Pediatric Genetics, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Gülen Eda Utine
- Division of Pediatric Genetics, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Göknur Haliloğlu
- Division of Pediatric Neurology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
2
|
Wang N, Wan R, Tang K. Transcriptional regulation in the development and dysfunction of neocortical projection neurons. Neural Regen Res 2024; 19:246-254. [PMID: 37488873 PMCID: PMC10503610 DOI: 10.4103/1673-5374.379039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 04/10/2023] [Accepted: 05/17/2023] [Indexed: 07/26/2023] Open
Abstract
Glutamatergic projection neurons generate sophisticated excitatory circuits to integrate and transmit information among different cortical areas, and between the neocortex and other regions of the brain and spinal cord. Appropriate development of cortical projection neurons is regulated by certain essential events such as neural fate determination, proliferation, specification, differentiation, migration, survival, axonogenesis, and synaptogenesis. These processes are precisely regulated in a tempo-spatial manner by intrinsic factors, extrinsic signals, and neural activities. The generation of correct subtypes and precise connections of projection neurons is imperative not only to support the basic cortical functions (such as sensory information integration, motor coordination, and cognition) but also to prevent the onset and progression of neurodevelopmental disorders (such as intellectual disability, autism spectrum disorders, anxiety, and depression). This review mainly focuses on the recent progress of transcriptional regulations on the development and diversity of neocortical projection neurons and the clinical relevance of the failure of transcriptional modulations.
Collapse
Affiliation(s)
- Ningxin Wang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, Guangdong Province, China
| | - Rong Wan
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, Guangdong Province, China
| | - Ke Tang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, Guangdong Province, China
| |
Collapse
|
3
|
Sokpor G, Kerimoglu C, Ulmke PA, Pham L, Nguyen HD, Brand-Saberi B, Staiger JF, Fischer A, Nguyen HP, Tuoc T. H3 Acetylation-Induced Basal Progenitor Generation and Neocortex Expansion Depends on the Transcription Factor Pax6. BIOLOGY 2024; 13:68. [PMID: 38392287 PMCID: PMC10886678 DOI: 10.3390/biology13020068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 02/24/2024]
Abstract
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.
Collapse
Affiliation(s)
- Godwin Sokpor
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
- Lincoln Medical School, University of Lincoln, Lincoln LN6 7TS, UK
| | - Cemil Kerimoglu
- German Center for Neurodegenerative Diseases, 37077 Goettingen, Germany
| | | | - Linh Pham
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Hoang Duy Nguyen
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany
| | - Jochen F Staiger
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
| | - Andre Fischer
- German Center for Neurodegenerative Diseases, 37077 Goettingen, Germany
| | - Huu Phuc Nguyen
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Tran Tuoc
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| |
Collapse
|
4
|
Manuel M, Tan KB, Kozic Z, Molinek M, Marcos TS, Razak MFA, Dobolyi D, Dobie R, Henderson BEP, Henderson NC, Chan WK, Daw MI, Mason JO, Price DJ. Pax6 limits the competence of developing cerebral cortical cells to respond to inductive intercellular signals. PLoS Biol 2022; 20:e3001563. [PMID: 36067211 PMCID: PMC9481180 DOI: 10.1371/journal.pbio.3001563] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 09/16/2022] [Accepted: 07/08/2022] [Indexed: 12/13/2022] Open
Abstract
The development of stable specialized cell types in multicellular organisms relies on mechanisms controlling inductive intercellular signals and the competence of cells to respond to such signals. In developing cerebral cortex, progenitors generate only glutamatergic excitatory neurons despite being exposed to signals with the potential to initiate the production of other neuronal types, suggesting that their competence is limited. Here, we tested the hypothesis that this limitation is due to their expression of transcription factor Pax6. We used bulk and single-cell RNAseq to show that conditional cortex-specific Pax6 deletion from the onset of cortical neurogenesis allowed some progenitors to generate abnormal lineages resembling those normally found outside the cortex. Analysis of selected gene expression showed that the changes occurred in specific spatiotemporal patterns. We then compared the responses of control and Pax6-deleted cortical cells to in vivo and in vitro manipulations of extracellular signals. We found that Pax6 loss increased cortical progenitors' competence to generate inappropriate lineages in response to extracellular factors normally present in developing cortex, including the morphogens Shh and Bmp4. Regional variation in the levels of these factors could explain spatiotemporal patterns of fate change following Pax6 deletion in vivo. We propose that Pax6's main role in developing cortical cells is to minimize the risk of their development being derailed by the potential side effects of morphogens engaged contemporaneously in other essential functions.
Collapse
Affiliation(s)
- Martine Manuel
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Kai Boon Tan
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Zrinko Kozic
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Michael Molinek
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Tiago Sena Marcos
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Maizatul Fazilah Abd Razak
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Dániel Dobolyi
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Ross Dobie
- Centre for Inflammation Research, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Beth E. P. Henderson
- Centre for Inflammation Research, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Neil C. Henderson
- Centre for Inflammation Research, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Wai Kit Chan
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Michael I. Daw
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, United Kingdom
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, Zhejiang, People’s Republic of China
| | - John O. Mason
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - David J. Price
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
5
|
Tocco C, Bertacchi M, Studer M. Structural and Functional Aspects of the Neurodevelopmental Gene NR2F1: From Animal Models to Human Pathology. Front Mol Neurosci 2022; 14:767965. [PMID: 34975398 PMCID: PMC8715095 DOI: 10.3389/fnmol.2021.767965] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/25/2021] [Indexed: 01/28/2023] Open
Abstract
The assembly and maturation of the mammalian brain result from an intricate cascade of highly coordinated developmental events, such as cell proliferation, migration, and differentiation. Any impairment of this delicate multi-factorial process can lead to complex neurodevelopmental diseases, sharing common pathogenic mechanisms and molecular pathways resulting in multiple clinical signs. A recently described monogenic neurodevelopmental syndrome named Bosch-Boonstra-Schaaf Optic Atrophy Syndrome (BBSOAS) is caused by NR2F1 haploinsufficiency. The NR2F1 gene, coding for a transcriptional regulator belonging to the steroid/thyroid hormone receptor superfamily, is known to play key roles in several brain developmental processes, from proliferation and differentiation of neural progenitors to migration and identity acquisition of neocortical neurons. In a clinical context, the disruption of these cellular processes could underlie the pathogenesis of several symptoms affecting BBSOAS patients, such as intellectual disability, visual impairment, epilepsy, and autistic traits. In this review, we will introduce NR2F1 protein structure, molecular functioning, and expression profile in the developing mouse brain. Then, we will focus on Nr2f1 several functions during cortical development, from neocortical area and cell-type specification to maturation of network activity, hippocampal development governing learning behaviors, assembly of the visual system, and finally establishment of cortico-spinal descending tracts regulating motor execution. Whenever possible, we will link experimental findings in animal or cellular models to corresponding features of the human pathology. Finally, we will highlight some of the unresolved questions on the diverse functions played by Nr2f1 during brain development, in order to propose future research directions. All in all, we believe that understanding BBSOAS mechanisms will contribute to further unveiling pathophysiological mechanisms shared by several neurodevelopmental disorders and eventually lead to effective treatments.
Collapse
Affiliation(s)
- Chiara Tocco
- Université Côte d'Azur, CNRS, Inserm, iBV, Nice, France
| | | | | |
Collapse
|
6
|
D'Souza L, Channakkar AS, Muralidharan B. Chromatin remodelling complexes in cerebral cortex development and neurodevelopmental disorders. Neurochem Int 2021; 147:105055. [PMID: 33964373 PMCID: PMC7611358 DOI: 10.1016/j.neuint.2021.105055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 04/11/2021] [Accepted: 04/24/2021] [Indexed: 12/19/2022]
Abstract
The diverse number of neurons in the cerebral cortex are generated during development by neural stem cells lining the ventricle, and they continue maturing postnatally. Dynamic chromatin regulation in these neural stem cells is a fundamental determinant of the emerging property of the functional neural network, and the chromatin remodellers are critical determinants of this process. Chromatin remodellers participate in several steps of this process from proliferation, differentiation, migration leading to complex network formation which forms the basis of higher-order functions of cognition and behaviour. Here we review the role of these ATP-dependent chromatin remodellers in cortical development in health and disease and highlight several key mouse mutants of the subunits of the complexes which have revealed how the remodelling mechanisms control the cortical stem cell chromatin landscape for expression of stage-specific transcripts. Consistent with their role in cortical development, several putative risk variants in the subunits of the remodelling complexes have been identified as the underlying causes of several neurodevelopmental disorders. A basic understanding of the detailed molecular mechanism of their action is key to understating how mutations in the same networks lead to disease pathologies and perhaps pave the way for therapeutic development for these complex multifactorial disorders.
Collapse
Affiliation(s)
- Leora D'Souza
- Brain Development and Disease Mechanisms, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore Life Science Cluster, Bangalore, India
| | - Asha S Channakkar
- Brain Development and Disease Mechanisms, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore Life Science Cluster, Bangalore, India
| | - Bhavana Muralidharan
- Brain Development and Disease Mechanisms, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore Life Science Cluster, Bangalore, India.
| |
Collapse
|
7
|
Kapourani CA, Argelaguet R, Sanguinetti G, Vallejos CA. scMET: Bayesian modeling of DNA methylation heterogeneity at single-cell resolution. Genome Biol 2021; 22:114. [PMID: 33879195 PMCID: PMC8056718 DOI: 10.1186/s13059-021-02329-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 03/25/2021] [Indexed: 02/06/2023] Open
Abstract
High-throughput single-cell measurements of DNA methylomes can quantify methylation heterogeneity and uncover its role in gene regulation. However, technical limitations and sparse coverage can preclude this task. scMET is a hierarchical Bayesian model which overcomes sparsity, sharing information across cells and genomic features to robustly quantify genuine biological heterogeneity. scMET can identify highly variable features that drive epigenetic heterogeneity, and perform differential methylation and variability analyses. We illustrate how scMET facilitates the characterization of epigenetically distinct cell populations and how it enables the formulation of novel hypotheses on the epigenetic regulation of gene expression. scMET is available at https://github.com/andreaskapou/scMET .
Collapse
Affiliation(s)
- Chantriolnt-Andreas Kapourani
- MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
- School of Informatics, University of Edinburgh, Edinburgh, UK
| | | | - Guido Sanguinetti
- School of Informatics, University of Edinburgh, Edinburgh, UK.
- SISSA, International School of Advanced Studies, Trieste, Italy.
| | - Catalina A Vallejos
- MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK.
- The Alan Turing Institute, London, UK.
| |
Collapse
|
8
|
Ortolano NA, Romero-Morales AI, Rasmussen ML, Bodnya C, Kline LA, Joshi P, Connelly JP, Rose KL, Pruett-Miller SM, Gama V. A proteomics approach for the identification of cullin-9 (CUL9) related signaling pathways in induced pluripotent stem cell models. PLoS One 2021; 16:e0248000. [PMID: 33705438 PMCID: PMC7951927 DOI: 10.1371/journal.pone.0248000] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 02/17/2021] [Indexed: 12/13/2022] Open
Abstract
CUL9 is a non-canonical and poorly characterized member of the largest family of E3 ubiquitin ligases known as the Cullin RING ligases (CRLs). Most CRLs play a critical role in developmental processes, however, the role of CUL9 in neuronal development remains elusive. We determined that deletion or depletion of CUL9 protein causes aberrant formation of neural rosettes, an in vitro model of early neuralization. In this study, we applied mass spectrometric approaches in human pluripotent stem cells (hPSCs) and neural progenitor cells (hNPCs) to identify CUL9 related signaling pathways that may contribute to this phenotype. Through LC-MS/MS analysis of immunoprecipitated endogenous CUL9, we identified several subunits of the APC/C, a major cell cycle regulator, as potential CUL9 interacting proteins. Knockdown of the APC/C adapter protein FZR1 resulted in a significant increase in CUL9 protein levels, however, CUL9 does not appear to affect protein abundance of APC/C subunits and adapters or alter cell cycle progression. Quantitative proteomic analysis of CUL9 KO hPSCs and hNPCs identified protein networks related to metabolic, ubiquitin degradation, and transcriptional regulation pathways that are disrupted by CUL9 deletion in both hPSCs. No significant changes in oxygen consumption rates or ATP production were detected in either cell type. The results of our study build on current evidence that CUL9 may have unique functions in different cell types and that compensatory mechanisms may contribute to the difficulty of identifying CUL9 substrates.
Collapse
Affiliation(s)
- Natalya A. Ortolano
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Alejandra I. Romero-Morales
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Megan L. Rasmussen
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Caroline Bodnya
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Leigh A. Kline
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Piyush Joshi
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Jon P. Connelly
- Department of Cell & Molecular Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- Center for Advanced Genome Engineering, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Kristie L. Rose
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt MSRC Proteomics Core, Nashville, Tennessee, United States of America
| | - Shondra M. Pruett-Miller
- Department of Cell & Molecular Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- Center for Advanced Genome Engineering, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Vivian Gama
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Center for Stem Cell Biology, Nashville, Tennessee, United States of America
- Vanderbilt Brain Institute, Nashville, Tennessee, United States of America
| |
Collapse
|
9
|
Markert F, Müller L, Badstübner-Meeske K, Storch A. Early Chronic Intermittent Maternal Hyperoxygenation Impairs Cortical Development by Inhibition of Pax6-Positive Apical Progenitor Cell Proliferation. J Neuropathol Exp Neurol 2021; 79:1223-1232. [PMID: 32929481 DOI: 10.1093/jnen/nlaa072] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 06/24/2020] [Indexed: 11/12/2022] Open
Abstract
Maternal hyperoxygenation is a feasible, noninvasive method to treat fetal diseases, such as heart hypoplasia, but effects of maternal hyperoxygenation on the developing brain remain poorly understood. Previous studies showed that short-term maternal hyperoxygenation during midneurogenic phase (E14-E16) but not in earlier development (E10-E12) increases oxygen tension and enhances neurogenesis in the developing mouse cortex. We investigated effects of early chronic maternal hyperoxygenation (CMH) as a potential clinical treatment. Pregnant C57BL/6J mice were housed in a chamber at 75% atmospheric oxygen and the brains of E16 fetuses were analyzed using immunohistochemistry. The mitosis marker phH3 showed a significant reduction of proliferation in the dorsolateral cortices of CMH-treated E16 fetuses. Numbers of Tbr2-positive intermediate progenitor cells were unaffected whereas numbers of Pax6-positive apical progenitor cells were significantly reduced in CMH-treated mice. This resulted in altered cortical plate development with fewer Satb2-positive upper layer neurons but more Tbr1-positive neurons corresponding to the deeper layer 6. Thus, maternal hyperoxygenation affects the developing cortex depending on timing and length of applied oxygen. Early CMH causes a severe reduction of neuroprogenitor proliferation likely affecting cortical development. Further studies are needed to investigate the mechanisms underlying these findings and to assess the clinical and neurodevelopmental outcomes of the pups.
Collapse
Affiliation(s)
| | | | | | - Alexander Storch
- Department of Neurology, University of Rostock.,German Center for Neurodegenerative Diseases (DZNE) Rostock, Rostock, Germany
| |
Collapse
|
10
|
Grant MK, Bobilev AM, Branch A, Lauderdale JD. Structural and functional consequences of PAX6 mutations in the brain: Implications for aniridia. Brain Res 2021; 1756:147283. [PMID: 33515537 DOI: 10.1016/j.brainres.2021.147283] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 12/15/2020] [Accepted: 01/05/2021] [Indexed: 12/27/2022]
Abstract
The paired-box 6 (PAX6) gene encodes a highly conserved transcription factor essential for the proper development of the eye and brain. Heterozygous loss-of-function mutations in PAX6 are causal for a condition known as aniridia in humans and the Small eye phenotype in mice. Aniridia is characterized by iris hypoplasia and other ocular abnormalities, but recent evidence of neuroanatomical, sensory, and cognitive impairments in this population has emerged, indicating brain-related phenotypes as a prevalent feature of the disorder. Determining the neurophysiological origins of brain-related phenotypes in this disorder presents a substantial challenge, as the majority of extra-ocular traits in aniridia demonstrate a high degree of heterogeneity. Here, we summarize and integrate findings from human and rodent model studies, which have focused on neuroanatomical and functional consequences of PAX6 mutations. We highlight novel findings from PAX6 central nervous system studies in adult mammals, and integrate these findings into what we know about PAX6's role in development of the central nervous system. This review presents the current literature in the field in order to inform clinical application, discusses what is needed in future studies, and highlights PAX6 as a lens through which to understand genetic disorders affecting the human nervous system.
Collapse
Affiliation(s)
- Madison K Grant
- Department of Cellular Biology, The University of Georgia, Athens, GA 30602, USA.
| | - Anastasia M Bobilev
- Neuroscience Division of the Biomedical and Health Sciences Institute, The University of Georgia, Athens, GA 30602, USA; Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Audrey Branch
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD 21218, USA.
| | - James D Lauderdale
- Department of Cellular Biology, The University of Georgia, Athens, GA 30602, USA; Neuroscience Division of the Biomedical and Health Sciences Institute, The University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
11
|
Abstract
The mammalian cerebral cortex is the pinnacle of brain evolution, reaching its maximum complexity in terms of neuron number, diversity and functional circuitry. The emergence of this outstanding complexity begins during embryonic development, when a limited number of neural stem and progenitor cells manage to generate myriads of neurons in the appropriate numbers, types and proportions, in a process called neurogenesis. Here we review the current knowledge on the regulation of cortical neurogenesis, beginning with a description of the types of progenitor cells and their lineage relationships. This is followed by a review of the determinants of neuron fate, the molecular and genetic regulatory mechanisms, and considerations on the evolution of cortical neurogenesis in vertebrates leading to humans. We finish with an overview on how dysregulation of neurogenesis is a leading cause of human brain malformations and functional disabilities.
Collapse
Affiliation(s)
- Ana Villalba
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, Spain
| | - Magdalena Götz
- Institute for Stem Cell Research, Helmholtz Zentrum München & Biomedical Center, Ludwig-Maximilians Universitaet, Planegg-Martinsried, Germany
| | - Víctor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, Spain.
| |
Collapse
|
12
|
Cole TB, Chang YC, Dao K, Daza R, Hevner R, Costa LG. Developmental exposure to diesel exhaust upregulates transcription factor expression, decreases hippocampal neurogenesis, and alters cortical lamina organization: relevance to neurodevelopmental disorders. J Neurodev Disord 2020; 12:41. [PMID: 33327933 PMCID: PMC7745370 DOI: 10.1186/s11689-020-09340-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 11/13/2020] [Indexed: 12/20/2022] Open
Abstract
Background Exposure to traffic-related air pollution (TRAP) during development and/or in adulthood has been associated in many human studies with both neurodevelopmental and neurodegenerative diseases, such as autism spectrum disorder (ASD) and Alzheimer’s disease (AD) or Parkinson’s disease (PD). Methods In the present study, C57BL/6 J mice were exposed to environmentally relevant levels (250+/−50 μg/m3) of diesel exhaust (DE) or filtered air (FA) during development (E0 to PND21). The expression of several transcription factors relevant for CNS development was assessed on PND3. To address possible mechanistic underpinnings of previously observed behavioral effects of DE exposure, adult neurogenesis in the hippocampus and laminar organization of neurons in the somatosensory cortex were analyzed on PND60. Results were analyzed separately for male and female mice. Results Developmental DE exposure caused a male-specific upregulation of Pax6, Tbr1, Tbr2, Sp1, and Creb1 on PND3. In contrast, in both males and females, Tbr2+ intermediate progenitor cells in the PND60 hippocampal dentate gyrus were decreased, as an indication of reduced adult neurogenesis. In the somatosensory region of the cerebral cortex, laminar distribution of Trb1, calbindin, and parvalbumin (but not of Ctip2 or Cux1) was altered by developmental DE exposure. Conclusions These results provide additional evidence to previous findings indicating the ability of developmental DE exposure to cause biochemical/molecular and behavioral alterations that may be involved in neurodevelopmental disorders such as ASD.
Collapse
Affiliation(s)
- Toby B Cole
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA. .,Center on Human Development and Disability, University of Washington, Seattle, WA, USA.
| | - Yu-Chi Chang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA.,Gradient Corporation, Seattle, WA, USA
| | - Khoi Dao
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Ray Daza
- Department of Pathology, University of California at San Diego, San Diego, CA, USA.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Robert Hevner
- Department of Pathology, University of California at San Diego, San Diego, CA, USA.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA.,Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Lucio G Costa
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA.,Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
13
|
Single-cell RNA-seq analysis revealed long-lasting adverse effects of tamoxifen on neurogenesis in prenatal and adult brains. Proc Natl Acad Sci U S A 2020; 117:19578-19589. [PMID: 32727894 DOI: 10.1073/pnas.1918883117] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The CreER/LoxP system is widely accepted to track neural lineages and study gene functions upon tamoxifen (TAM) administration. We have observed that prenatal TAM treatment caused high rates of delayed delivery and fetal mortality. This substance could produce undesired results, leading to data misinterpretation. Here, we report that administration of TAM during early stages of cortical neurogenesis promoted precocious neural differentiation, while it inhibited neural progenitor cell (NPC) proliferation. The TAM-induced inhibition of NPC proliferation led to deficits in cortical neurogenesis, dendritic morphogenesis, synaptic formation, and cortical patterning in neonatal and postnatal offspring. Mechanistically, by employing single-cell RNA-sequencing (scRNA-seq) analysis combined with in vivo and in vitro assays, we show TAM could exert these drastic effects mainly through dysregulating the Wnt-Dmrta2 signaling pathway. In adult mice, administration of TAM significantly attenuated NPC proliferation in both the subventricular zone and the dentate gyrus. This study revealed the cellular and molecular mechanisms for the adverse effects of TAM on corticogenesis, suggesting that care must be taken when using the TAM-induced CreER/LoxP system for neural lineage tracing and genetic manipulation studies in both embryonic and adult brains.
Collapse
|
14
|
Bertacchi M, Romano AL, Loubat A, Tran Mau-Them F, Willems M, Faivre L, Khau van Kien P, Perrin L, Devillard F, Sorlin A, Kuentz P, Philippe C, Garde A, Neri F, Di Giaimo R, Oliviero S, Cappello S, D'Incerti L, Frassoni C, Studer M. NR2F1 regulates regional progenitor dynamics in the mouse neocortex and cortical gyrification in BBSOAS patients. EMBO J 2020; 39:e104163. [PMID: 32484994 PMCID: PMC7327499 DOI: 10.15252/embj.2019104163] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/01/2020] [Accepted: 04/15/2020] [Indexed: 12/12/2022] Open
Abstract
The relationships between impaired cortical development and consequent malformations in neurodevelopmental disorders, as well as the genes implicated in these processes, are not fully elucidated to date. In this study, we report six novel cases of patients affected by BBSOAS (Boonstra‐Bosch‐Schaff optic atrophy syndrome), a newly emerging rare neurodevelopmental disorder, caused by loss‐of‐function mutations of the transcriptional regulator NR2F1. Young patients with NR2F1 haploinsufficiency display mild to moderate intellectual disability and show reproducible polymicrogyria‐like brain malformations in the parietal and occipital cortex. Using a recently established BBSOAS mouse model, we found that Nr2f1 regionally controls long‐term self‐renewal of neural progenitor cells via modulation of cell cycle genes and key cortical development master genes, such as Pax6. In the human fetal cortex, distinct NR2F1 expression levels encompass gyri and sulci and correlate with local degrees of neurogenic activity. In addition, reduced NR2F1 levels in cerebral organoids affect neurogenesis and PAX6 expression. We propose NR2F1 as an area‐specific regulator of mouse and human brain morphology and a novel causative gene of abnormal gyrification.
Collapse
Affiliation(s)
- Michele Bertacchi
- Université Côte d'Azur, CNRS, Inserm, iBV, Paris, France.,Clinical and Experimental Epileptology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | | | - Agnès Loubat
- Université Côte d'Azur, CNRS, Inserm, iBV, Paris, France
| | - Frederic Tran Mau-Them
- UMR1231 GAD, Inserm - Université Bourgogne-Franche Comté, Dijon, France.,Unité Fonctionnelle Innovation en Diagnostic Génomique des Maladies Rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Marjolaine Willems
- Hôpital Arnaud de Villeneuve, Service de Génétique Médicale, CHU de Montpellier, Montpellier, France
| | - Laurence Faivre
- UMR1231 GAD, Inserm - Université Bourgogne-Franche Comté, Dijon, France.,Unité Fonctionnelle Innovation en Diagnostic Génomique des Maladies Rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France.,Centre de Référence maladies rares « Anomalies du développement et syndromes malformatifs », Centre de Génétique, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Philippe Khau van Kien
- Hôpital Carémeau, UF de Génétique Médicale et Cytogénétique, Centre de Compétences Anomalies du Développement et Syndromes Malformatifs, CHU de Nîmes, Nîmes, France
| | - Laurence Perrin
- Unité Fonctionnelle de Génétique Clinique, Hôpital Robert Debré, Paris, France
| | - Françoise Devillard
- Département de Génétique et Procréation, Hôpital Couple-Enfant, CHU de Grenoble, Grenoble, France
| | - Arthur Sorlin
- UMR1231 GAD, Inserm - Université Bourgogne-Franche Comté, Dijon, France.,Unité Fonctionnelle Innovation en Diagnostic Génomique des Maladies Rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France.,Centre de Référence maladies rares « Anomalies du développement et syndromes malformatifs », Centre de Génétique, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France.,Centre de référence maladies rares « Déficiences intellectuelles de causes rares », Centre de Génétique, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Paul Kuentz
- UMR1231 GAD, Inserm - Université Bourgogne-Franche Comté, Dijon, France.,Génétique Biologique, PCBio, Centre Hospitalier Universitaire de Besançon, Besançon, France
| | - Christophe Philippe
- UMR1231 GAD, Inserm - Université Bourgogne-Franche Comté, Dijon, France.,Unité Fonctionnelle Innovation en Diagnostic Génomique des Maladies Rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Aurore Garde
- Unité Fonctionnelle Innovation en Diagnostic Génomique des Maladies Rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France.,Centre de Référence maladies rares « Anomalies du développement et syndromes malformatifs », Centre de Génétique, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Francesco Neri
- Epigenetics Unit, Italian Institute for Genomic Medicine, University of Torino, Torino, Italy.,Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Rossella Di Giaimo
- Department of Biology, University of Naples Federico II, Napoli, Italy.,Max Planck Institute of Psychiatry, München, Germany
| | - Salvatore Oliviero
- Epigenetics Unit, Italian Institute for Genomic Medicine, University of Torino, Torino, Italy
| | | | - Ludovico D'Incerti
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Carolina Frassoni
- Clinical and Experimental Epileptology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Michèle Studer
- Université Côte d'Azur, CNRS, Inserm, iBV, Paris, France
| |
Collapse
|
15
|
Grant MK, Bobilev AM, Rasys AM, Branson Byers J, Schriever HC, Hekmatyar K, Lauderdale JD. Global and age-related neuroanatomical abnormalities in a Pax6-deficient mouse model of aniridia suggests a role for Pax6 in adult structural neuroplasticity. Brain Res 2020; 1732:146698. [PMID: 32014531 PMCID: PMC10712278 DOI: 10.1016/j.brainres.2020.146698] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/14/2020] [Accepted: 01/30/2020] [Indexed: 12/29/2022]
Abstract
PAX6 encodes a highly conserved transcription factor necessary for normal development of the eyes and central nervous system. Heterozygous loss-of-function mutations in PAX6 cause the disorder aniridia in humans and the Small eye trait in mice. Aniridia is a congenital and progressive disorder known for ocular phenotypes; however, recently, consequences of PAX6 haploinsufficiency in the brains of aniridia patients have been identified. These findings span structural and functional abnormalities, including deficits in cognitive and sensory processing. Furthermore, some of these abnormalities are accelerated as aniridia patients age. Although some functional abnormalities may be explained by structural changes, variability of results remain, and the effects of PAX6 heterozygous loss-of-function mutations on neuroanatomy, particularly with regard to aging, have yet to be resolved. Our study used high-resolution magnetic resonance imaging (MRI) and histology to investigate structural consequences of such mutations in the adult brain of our aniridia mouse model, Small eye Neuherberg allele (Pax6SeyNeu/+), at two adult age groups. Using both MRI and histology enables a direct comparison with human studies, while providing higher resolution for detection of more subtle changes. We show volumetric changes in major brain regions of the the Pax6SeyNeu/+ mouse compared to wild-type including genotype- and age-related olfactory bulb differences, age-related cerebellum differences, and genotype-related eye differences. We also show alterations in thickness of major interhemispheric commissures, particularly those anteriorly located within the brain including the optic chiasm, corpus callosum, and anterior commissure. Together, these genotype and age related changes to brain volumes and structures suggest a global decrease in adult brain structural plasticity in our Pax6SeyNeu/+ mice.
Collapse
Affiliation(s)
- Madison K Grant
- Department of Cellular Biology, University of Georgia, 250B Coverdell Center, 500 D.W. Brooks Drive, Athens, GA 30602, United States.
| | - Anastasia M Bobilev
- Department of Psychiatry, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, United States; Neuroscience Division of the Biomedical and Health Sciences Institute, The University of Georgia, Athens, GA 30602, United States.
| | - Ashley M Rasys
- Department of Cellular Biology, University of Georgia, 250B Coverdell Center, 500 D.W. Brooks Drive, Athens, GA 30602, United States.
| | - J Branson Byers
- Department of Cellular Biology, University of Georgia, 250B Coverdell Center, 500 D.W. Brooks Drive, Athens, GA 30602, United States.
| | - Hannah C Schriever
- Department of Cellular Biology, University of Georgia, 250B Coverdell Center, 500 D.W. Brooks Drive, Athens, GA 30602, United States.
| | - Khan Hekmatyar
- Bio-imaging Research Center, University of Georgia, Coverdell Center, 500 D.W. Brooks Drive, Athens, GA 30602, United States.
| | - James D Lauderdale
- Department of Cellular Biology, University of Georgia, 250B Coverdell Center, 500 D.W. Brooks Drive, Athens, GA 30602, United States; Neuroscience Division of the Biomedical and Health Sciences Institute, The University of Georgia, Athens, GA 30602, United States.
| |
Collapse
|
16
|
De Mori R, Severino M, Mancardi MM, Anello D, Tardivo S, Biagini T, Capra V, Casella A, Cereda C, Copeland BR, Gagliardi S, Gamucci A, Ginevrino M, Illi B, Lorefice E, Musaev D, Stanley V, Micalizzi A, Gleeson JG, Mazza T, Rossi A, Valente EM. Agenesis of the putamen and globus pallidus caused by recessive mutations in the homeobox gene GSX2. Brain 2019; 142:2965-2978. [PMID: 31412107 PMCID: PMC6776115 DOI: 10.1093/brain/awz247] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 06/06/2019] [Accepted: 06/18/2019] [Indexed: 12/31/2022] Open
Abstract
Basal ganglia are subcortical grey nuclei that play essential roles in controlling voluntary movements, cognition and emotion. While basal ganglia dysfunction is observed in many neurodegenerative or metabolic disorders, congenital malformations are rare. In particular, dysplastic basal ganglia are part of the malformative spectrum of tubulinopathies and X-linked lissencephaly with abnormal genitalia, but neurodevelopmental syndromes characterized by basal ganglia agenesis are not known to date. We ascertained two unrelated children (both female) presenting with spastic tetraparesis, severe generalized dystonia and intellectual impairment, sharing a unique brain malformation characterized by agenesis of putamina and globi pallidi, dysgenesis of the caudate nuclei, olfactory bulbs hypoplasia, and anomaly of the diencephalic-mesencephalic junction with abnormal corticospinal tract course. Whole-exome sequencing identified two novel homozygous variants, c.26C>A; p.(S9*) and c.752A>G; p.(Q251R) in the GSX2 gene, a member of the family of homeobox transcription factors, which are key regulators of embryonic development. GSX2 is highly expressed in neural progenitors of the lateral and median ganglionic eminences, two protrusions of the ventral telencephalon from which the basal ganglia and olfactory tubercles originate, where it promotes neurogenesis while negatively regulating oligodendrogenesis. The truncating variant resulted in complete loss of protein expression, while the missense variant affected a highly conserved residue of the homeobox domain, was consistently predicted as pathogenic by bioinformatic tools, resulted in reduced protein expression and caused impaired structural stability of the homeobox domain and weaker interaction with DNA according to molecular dynamic simulations. Moreover, the nuclear localization of the mutant protein in transfected cells was significantly reduced compared to the wild-type protein. Expression studies on both patients' fibroblasts demonstrated reduced expression of GSX2 itself, likely due to altered transcriptional self-regulation, as well as significant expression changes of related genes such as ASCL1 and PAX6. Whole transcriptome analysis revealed a global deregulation in genes implicated in apoptosis and immunity, two broad pathways known to be involved in brain development. This is the first report of the clinical phenotype and molecular basis associated to basal ganglia agenesis in humans.
Collapse
Affiliation(s)
- Roberta De Mori
- Neurogenetics Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | | | | | - Danila Anello
- Neurogenetics Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Silvia Tardivo
- Neurogenetics Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Tommaso Biagini
- IRCCS Casa Sollievo della Sofferenza, Laboratory of Bioinformatics, San Giovanni Rotondo (FG), Italy
| | - Valeria Capra
- Neurosurgery Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | - Cristina Cereda
- Genomic and Postgenomic Lab, IRCCS Mondino Foundation, Pavia, Italy
| | - Brett R Copeland
- Laboratory for Pediatric Brain Diseases, Rady Children’s Institute for Genomic Medicine, University of California San Diego, Howard Hughes Medical Institute, La Jolla (CA), USA
| | - Stella Gagliardi
- Genomic and Postgenomic Lab, IRCCS Mondino Foundation, Pavia, Italy
| | - Alessandra Gamucci
- Child Neuropsychiatry Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Monia Ginevrino
- Neurogenetics Unit, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Barbara Illi
- Institute of Molecular Biology and Pathology, National Research Council, Rome, Italy
| | - Elisa Lorefice
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Damir Musaev
- Laboratory for Pediatric Brain Diseases, Rady Children’s Institute for Genomic Medicine, University of California San Diego, Howard Hughes Medical Institute, La Jolla (CA), USA
| | - Valentina Stanley
- Laboratory for Pediatric Brain Diseases, Rady Children’s Institute for Genomic Medicine, University of California San Diego, Howard Hughes Medical Institute, La Jolla (CA), USA
| | - Alessia Micalizzi
- Laboratory of Medical Genetics, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Joseph G Gleeson
- Laboratory for Pediatric Brain Diseases, Rady Children’s Institute for Genomic Medicine, University of California San Diego, Howard Hughes Medical Institute, La Jolla (CA), USA
| | - Tommaso Mazza
- IRCCS Casa Sollievo della Sofferenza, Laboratory of Bioinformatics, San Giovanni Rotondo (FG), Italy
| | - Andrea Rossi
- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Enza Maria Valente
- Neurogenetics Unit, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| |
Collapse
|
17
|
Dorà E, Price DJ, Mason JO. Loss of Pax6 Causes Regional Changes in Dll1 Expression in Developing Cerebral Cortex. Front Cell Neurosci 2019; 13:78. [PMID: 30894800 PMCID: PMC6414449 DOI: 10.3389/fncel.2019.00078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/15/2019] [Indexed: 11/28/2022] Open
Abstract
The transcription factor Pax6 controls multiple aspects of forebrain development. Conditional deletion of Pax6 in embryonic mouse cortex causes increased proliferation of cortical progenitor cells and a concomitant decrease in neural differentiation. Notch signaling regulates the balance between proliferation and differentiation of cortical progenitor cells, suggesting a possible connection between Pax6 and Notch signaling. We investigated how expression of the Notch ligand delta-like 1 (Dll1) is altered by loss of Pax6. Acute cortex-specific deletion of Pax6 resulted in a widespread decrease in the density of Dll1+ cells at embryonic days 12.5 and 13.5 (E12.5 and E13.5). In constitutive loss-of-function mutants, decreases in the densities of Dll1+ cells were more limited both spatially and temporally. Controlled over-expression of Pax6 had no detectable effect on Dll1 expression. The proneural transcription factor Neurog2 is a target of Pax6 that can activate Dll1 expression and we found clear co-expression of Neurog2 and Dll1 in radial glial progenitors, suggesting that Pax6’s effect on Dll1 could be mediated through Neurog2. However, we found no change in Dll1+ cells in Neurog2−/− cortex suggesting either that Neurog2 is not directly involved, or that its loss of function in embryonic cortex can be compensated for.
Collapse
Affiliation(s)
- Elena Dorà
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - David J Price
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - John O Mason
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
18
|
Quintana-Urzainqui I, Kozić Z, Mitra S, Tian T, Manuel M, Mason JO, Price DJ. Tissue-Specific Actions of Pax6 on Proliferation and Differentiation Balance in Developing Forebrain Are Foxg1 Dependent. iScience 2018; 10:171-191. [PMID: 30529950 PMCID: PMC6287089 DOI: 10.1016/j.isci.2018.11.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/02/2018] [Accepted: 11/16/2018] [Indexed: 12/12/2022] Open
Abstract
Differences in the growth and maturation of diverse forebrain tissues depend on region-specific transcriptional regulation. Individual transcription factors act simultaneously in multiple regions that develop very differently, raising questions about the extent to which their actions vary regionally. We found that the transcription factor Pax6 affects the transcriptomes and the balance between proliferation and differentiation in opposite directions in the diencephalon versus cerebral cortex. We tested several possible mechanisms to explain Pax6's tissue-specific actions and found that the presence of the transcription factor Foxg1 in the cortex but not in the diencephalon was most influential. We found that Foxg1 is responsible for many of the differences in cell cycle gene expression between the diencephalon and cortex and, in cortex lacking Foxg1, Pax6's action on the balance of proliferation versus differentiation becomes diencephalon like. Our findings reveal a mechanism for generating regional forebrain diversity in which one transcription factor completely reverses the actions of another.
Collapse
Affiliation(s)
- Idoia Quintana-Urzainqui
- Simons Initiative for the Developing Brain, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK.
| | - Zrinko Kozić
- Simons Initiative for the Developing Brain, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Soham Mitra
- Simons Initiative for the Developing Brain, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Tian Tian
- Simons Initiative for the Developing Brain, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Martine Manuel
- Simons Initiative for the Developing Brain, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - John O Mason
- Simons Initiative for the Developing Brain, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - David J Price
- Simons Initiative for the Developing Brain, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| |
Collapse
|
19
|
Mi D, Manuel M, Huang YT, Mason JO, Price DJ. Pax6 Lengthens G1 Phase and Decreases Oscillating Cdk6 Levels in Murine Embryonic Cortical Progenitors. Front Cell Neurosci 2018; 12:419. [PMID: 30498434 PMCID: PMC6249377 DOI: 10.3389/fncel.2018.00419] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 10/26/2018] [Indexed: 12/23/2022] Open
Abstract
Pax6 is a key regulator of the rates of progenitor cell division in cerebral corticogenesis. Previous work has suggested that this action is mediated at least in part by regulation of the cell cycle gene Cdk6, which acts largely on the transition from G1 to S phase. We began the present study by investigating whether, in addition to Cdk6, other Pax6-regulated cell cycle genes are likely to be primary mediators of Pax6’s actions on cortical progenitor cell cycles. Following acute cortex-specific deletion of Pax6, Cdk6 showed changes in expression a day earlier than any other Pax6-regulated cell cycle gene suggesting that it is the primary mediator of Pax6’s actions. We then used flow cytometry to show that progenitors lacking Pax6 have a shortened G1 phase and that their Cdk6 levels are increased in all phases. We found that Cdk6 levels oscillate during the cell cycle, increasing from G1 to M phase. We propose a model in which loss of Pax6 shortens G1 phase by raising overall Cdk6 levels, thereby shortening the time taken for Cdk6 levels to cross a threshold triggering transition to S phase.
Collapse
Affiliation(s)
- Da Mi
- Biomedical Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Martine Manuel
- Biomedical Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Yu-Ting Huang
- Biomedical Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - John O Mason
- Biomedical Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - David J Price
- Biomedical Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
20
|
Jha SC, Xia K, Schmitt JE, Ahn M, Girault JB, Murphy VA, Li G, Wang L, Shen D, Zou F, Zhu H, Styner M, Knickmeyer RC, Gilmore JH. Genetic influences on neonatal cortical thickness and surface area. Hum Brain Mapp 2018; 39:4998-5013. [PMID: 30144223 DOI: 10.1002/hbm.24340] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 07/18/2018] [Accepted: 07/20/2018] [Indexed: 01/07/2023] Open
Abstract
Genetic and environmental influences on cortical thickness (CT) and surface area (SA) are thought to vary in a complex and dynamic way across the lifespan. It has been established that CT and SA are genetically distinct in older children, adolescents, and adults, and that heritability varies across cortical regions. Very little, however, is known about how genetic and environmental factors influence infant CT and SA. Using structural MRI, we performed the first assessment of genetic and environmental influences on normal variation of SA and CT in 360 twin neonates. We observed strong and significant additive genetic influences on total SA (a2 = 0.78) and small and nonsignificant genetic influences on average CT (a2 = 0.29). Moreover, we found significant genetic overlap (genetic correlation = 0.65) between these global cortical measures. Regionally, there were minimal genetic influences across the cortex for both CT and SA measures and no distinct patterns of genetic regionalization. Overall, outcomes from this study suggest a dynamic relationship between CT and SA during the neonatal period and provide novel insights into how genetic influences shape cortical structure during early development.
Collapse
Affiliation(s)
- Shaili C Jha
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina
| | - Kai Xia
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina
| | - James Eric Schmitt
- Brain Behavior Laboratory, Departments of Radiology and Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mihye Ahn
- Department of Mathematics and Statistics, University of Nevada, Reno, Nevada
| | - Jessica B Girault
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina
| | - Veronica A Murphy
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina.,Curriculum in Neuroscience, University of North Carolina, Chapel Hill, North Carolina
| | - Gang Li
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, North Carolina
| | - Li Wang
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, North Carolina
| | - Dinggang Shen
- Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, North Carolina
| | - Fei Zou
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina
| | - Hongtu Zhu
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina.,Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Martin Styner
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina.,Department of Computer Science, University of North Carolina, Chapel Hill, North Carolina
| | - Rebecca C Knickmeyer
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina
| | - John H Gilmore
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
21
|
Ruiz-Reig N, Andres B, Lamonerie T, Theil T, Fairén A, Studer M. The caudo-ventral pallium is a novel pallial domain expressing Gdf10 and generating Ebf3-positive neurons of the medial amygdala. Brain Struct Funct 2018; 223:3279-3295. [PMID: 29869132 DOI: 10.1007/s00429-018-1687-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 05/18/2018] [Indexed: 12/16/2022]
Abstract
In rodents, the medial nucleus of the amygdala receives direct inputs from the accessory olfactory bulbs and is mainly implicated in pheromone-mediated reproductive and defensive behaviors. The principal neurons of the medial amygdala are GABAergic neurons generated principally in the caudo-ventral medial ganglionic eminence and preoptic area. Beside GABAergic neurons, the medial amygdala also contains glutamatergic Otp-expressing neurons cells generated in the lateral hypothalamic neuroepithelium and a non-well characterized Pax6-positive population. In the present work, we describe a novel glutamatergic Ebf3-expressing neuronal subpopulation distributed within the periphery of the postero-ventral medial amygdala. These neurons are generated in a pallial domain characterized by high expression of Gdf10. This territory is topologically the most caudal tier of the ventral pallium and accordingly, we named it Caudo-Ventral Pallium (CVP). In the absence of Pax6, the CVP is disrupted and Ebf3-expressing neurons fail to be generated. Overall, this work proposes a novel model of the neuronal composition of the medial amygdala and unravels for the first time a new novel pallial subpopulation originating from the CVP and expressing the transcription factor Ebf3.
Collapse
Affiliation(s)
- Nuria Ruiz-Reig
- Université Côte d'Azur (UCA), CNRS, Inserm, Institut de Biologie Valrose (iBV), 06108, Nice, France.
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), 03550, San Juan de Alicante, Spain.
| | - Belen Andres
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), 03550, San Juan de Alicante, Spain
| | - Thomas Lamonerie
- Université Côte d'Azur (UCA), CNRS, Inserm, Institut de Biologie Valrose (iBV), 06108, Nice, France
| | - Thomas Theil
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Alfonso Fairén
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), 03550, San Juan de Alicante, Spain
- , Palau 11, 03550, San Juan de Alicante, Spain
| | - Michèle Studer
- Université Côte d'Azur (UCA), CNRS, Inserm, Institut de Biologie Valrose (iBV), 06108, Nice, France.
| |
Collapse
|
22
|
Chromatin Remodeling BAF155 Subunit Regulates the Genesis of Basal Progenitors in Developing Cortex. iScience 2018; 4:109-126. [PMID: 30240734 PMCID: PMC6147019 DOI: 10.1016/j.isci.2018.05.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/22/2018] [Accepted: 05/18/2018] [Indexed: 11/21/2022] Open
Abstract
The abundance of basal progenitors (BPs), basal radial glia progenitors (bRGs) and basal intermediate progenitors (bIPs), in primate brain has been correlated to the high degree of cortical folding. Here we examined the role of BAF155, a subunit of the chromatin remodeling BAF complex, in generation of cortical progenitor heterogeneity. The conditional deletion of BAF155 led to diminished bIP pool and increased number of bRGs, due to delamination of apical RGs. We found that BAF155 is required for normal activity of neurogenic transcription factor PAX6, thus controlling the expression of genes that are involved in bIP specification, cell-cell interaction, and establishment of adherens junction. In a PAX6-dependent manner, BAF155 regulates the expression of the CDC42 effector protein CEP4, thereby controlling progenitor delamination. Furthermore, BAF155-dependent chromatin remodeling seems to exert a specific role in the genesis of BPs through the regulation of human RG-specific genes (such as Foxn4) that possibly acquired evolutionary significance.
Collapse
|
23
|
Drosophila Pax6 promotes development of the entire eye-antennal disc, thereby ensuring proper adult head formation. Proc Natl Acad Sci U S A 2018; 114:5846-5853. [PMID: 28584125 DOI: 10.1073/pnas.1610614114] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Paired box 6 (Pax6) is considered to be the master control gene for eye development in all seeing animals studied so far. In vertebrates, it is required not only for lens/retina formation but also for the development of the CNS, olfactory system, and pancreas. Although Pax6 plays important roles in cell differentiation, proliferation, and patterning during the development of these systems, the underlying mechanism remains poorly understood. In the fruit fly, Drosophila melanogaster, Pax6 also functions in a range of tissues, including the eye and brain. In this report, we describe the function of Pax6 in Drosophila eye-antennal disc development. Previous studies have suggested that the two fly Pax6 genes, eyeless (ey) and twin of eyeless (toy), initiate eye specification, whereas eyegone (eyg) and the Notch (N) pathway independently regulate cell proliferation. Here, we show that Pax6 controls eye progenitor cell survival and proliferation through the activation of teashirt (tsh) and eyg, thereby indicating that Pax6 initiates both eye specification and proliferation. Although simultaneous loss of ey and toy during early eye-antennal disc development disrupts the development of all head structures derived from the eye-antennal disc, overexpression of N or tsh in the absence of Pax6 rescues only antennal and head epidermis development. Furthermore, overexpression of tsh induces a homeotic transformation of the fly head into thoracic structures. Taking these data together, we demonstrate that Pax6 promotes development of the entire eye-antennal disc and that the retinal determination network works to repress alternative tissue fates, which ensures proper development of adult head structures.
Collapse
|
24
|
Sokpor G, Castro-Hernandez R, Rosenbusch J, Staiger JF, Tuoc T. ATP-Dependent Chromatin Remodeling During Cortical Neurogenesis. Front Neurosci 2018; 12:226. [PMID: 29686607 PMCID: PMC5900035 DOI: 10.3389/fnins.2018.00226] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/22/2018] [Indexed: 12/20/2022] Open
Abstract
The generation of individual neurons (neurogenesis) during cortical development occurs in discrete steps that are subtly regulated and orchestrated to ensure normal histogenesis and function of the cortex. Notably, various gene expression programs are known to critically drive many facets of neurogenesis with a high level of specificity during brain development. Typically, precise regulation of gene expression patterns ensures that key events like proliferation and differentiation of neural progenitors, specification of neuronal subtypes, as well as migration and maturation of neurons in the developing cortex occur properly. ATP-dependent chromatin remodeling complexes regulate gene expression through utilization of energy from ATP hydrolysis to reorganize chromatin structure. These chromatin remodeling complexes are characteristically multimeric, with some capable of adopting functionally distinct conformations via subunit reconstitution to perform specific roles in major aspects of cortical neurogenesis. In this review, we highlight the functions of such chromatin remodelers during cortical development. We also bring together various proposed mechanisms by which ATP-dependent chromatin remodelers function individually or in concert, to specifically modulate vital steps in cortical neurogenesis.
Collapse
Affiliation(s)
- Godwin Sokpor
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
| | - Ricardo Castro-Hernandez
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
| | - Joachim Rosenbusch
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
| | - Jochen F Staiger
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany.,DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Goettingen, Germany
| | - Tran Tuoc
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany.,DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Goettingen, Germany
| |
Collapse
|
25
|
Kim KC, Choi CS, Gonzales ELT, Mabunga DFN, Lee SH, Jeon SJ, Hwangbo R, Hong M, Ryu JH, Han SH, Bahn GH, Shin CY. Valproic Acid Induces Telomerase Reverse Transcriptase Expression during Cortical Development. Exp Neurobiol 2017; 26:252-265. [PMID: 29093634 PMCID: PMC5661058 DOI: 10.5607/en.2017.26.5.252] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 09/07/2017] [Accepted: 09/24/2017] [Indexed: 01/11/2023] Open
Abstract
The valproic acid (VPA)-induced animal model is one of the most widely utilized environmental risk factor models of autism. Autism spectrum disorder (ASD) remains an insurmountable challenge among neurodevelopmental disorders due to its heterogeneity, unresolved pathological pathways and lack of treatment. We previously reported that VPA-exposed rats and cultured rat primary neurons have increased Pax6 expression during post-midterm embryonic development which led to the sequential upregulation of glutamatergic neuronal markers. In this study, we provide experimental evidence that telomerase reverse transcriptase (TERT), a protein component of ribonucleoproteins complex of telomerase, is involved in the abnormal components caused by VPA in addition to Pax6 and its downstream signals. In embryonic rat brains and cultured rat primary neural progenitor cells (NPCs), VPA induced the increased expression of TERT as revealed by Western blot, RT-PCR, and immunostainings. The HDAC inhibitor property of VPA is responsible for the TERT upregulation. Chromatin immunoprecipitation revealed that VPA increased the histone acetylation but blocked the HDAC1 binding to both Pax6 and Tert genes. Interestingly, the VPA-induced TERT overexpression resulted to sequential upregulations of glutamatergic markers such as Ngn2 and NeuroD1, and inter-synaptic markers such as PSD-95, α-CaMKII, vGluT1 and synaptophysin. Transfection of Tert siRNA reversed the effects of VPA in cultured NPCs confirming the direct involvement of TERT in the expression of those markers. This study suggests the involvement of TERT in the VPA-induced autistic phenotypes and has important implications for the role of TERT as a modulator of balanced neuronal development and transmission in the brain.
Collapse
Affiliation(s)
- Ki Chan Kim
- School of Medicine and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, KU Open Innovation Center, Konkuk University, Seoul 05029, Korea
| | - Chang Soon Choi
- School of Medicine and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, KU Open Innovation Center, Konkuk University, Seoul 05029, Korea
| | - Edson Luck T Gonzales
- School of Medicine and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, KU Open Innovation Center, Konkuk University, Seoul 05029, Korea
| | - Darine Froy N Mabunga
- School of Medicine and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, KU Open Innovation Center, Konkuk University, Seoul 05029, Korea
| | - Sung Hoon Lee
- College of Pharmacy, Chung-Ang Univeristy, Seoul 06974, Korea
| | - Se Jin Jeon
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Ram Hwangbo
- Department of Psychiatry, Kyung Hee University Hospital, Seoul 02447, Korea
| | - Minha Hong
- Department of Psychiatry, Seonam University, College of Medicine, Myongji Hospital, Goyang 10475, Korea
| | - Jong Hoon Ryu
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Korea
| | - Seol-Heui Han
- School of Medicine and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, KU Open Innovation Center, Konkuk University, Seoul 05029, Korea
| | - Geon Ho Bahn
- Department of Neuropsychiatry, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Chan Young Shin
- School of Medicine and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, KU Open Innovation Center, Konkuk University, Seoul 05029, Korea
| |
Collapse
|
26
|
Reis AH, Moreno MM, Maia LA, Oliveira FP, Santos AS, Abreu JG. Cholesterol-rich membrane microdomains modulate Wnt/β-catenin morphogen gradient during Xenopus development. Mech Dev 2016; 142:30-39. [PMID: 27687541 DOI: 10.1016/j.mod.2016.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 08/19/2016] [Accepted: 09/22/2016] [Indexed: 11/30/2022]
Abstract
Wnt/β-catenin has been described as crucial for dorsal-ventral and antero-posterior patterning, playing multiple roles at different stages of development. Cholesterol-rich membrane microdomains (CRMMs), cholesterol- and sphingolipid-enriched domains of the plasma membrane, are known as platforms for signaling pathways. Although we have demonstrated the importance of the CRMMs for head development, how they participate in prechordal plate formation and embryo axis patterning remains an open question. Moreover, the participation of the CRMMs in the Wnt/β-catenin signaling pathway activity in vivo is unclear, particularly during embryonic development. In this study, we demonstrated that CRMMs disruption by methyl-beta-cyclodextrin (MβCD) potentiates the activation of the Wnt/β-catenin signaling pathway in vitro and in vivo during embryonic development, causing head defects by expanding the Wnt expression domain. Furthermore, we also found that the action of CRMMs depends on the microenvironmental context because it also works in conjunction with dkk1, when dkk1 is overexpressed. Thus, we propose CRMMs as a further mechanism of prechordal plate protection against the Wnt signals secreted by posterolateral cells, complementing the action of secreted antagonists.
Collapse
Affiliation(s)
- Alice H Reis
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Marcela M Moreno
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Lorena A Maia
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Fernanda P Oliveira
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Andressa S Santos
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - José Garcia Abreu
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil.
| |
Collapse
|
27
|
Yogarajah M, Matarin M, Vollmar C, Thompson PJ, Duncan JS, Symms M, Moore AT, Liu J, Thom M, van Heyningen V, Sisodiya SM. PAX6, brain structure and function in human adults: advanced MRI in aniridia. Ann Clin Transl Neurol 2016; 3:314-30. [PMID: 27231702 PMCID: PMC4863745 DOI: 10.1002/acn3.297] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 01/27/2016] [Indexed: 01/13/2023] Open
Abstract
Objective PAX6 is a pleiotropic transcription factor essential for the development of several tissues including the eyes, central nervous system, and some endocrine glands. Recently it has also been shown to be important for the maintenance and functioning of corneal and pancreatic tissues in adults. We hypothesized that PAX6 is important for the maintenance of brain integrity in humans, and that adult heterozygotes may have abnormalities of cortical patterning analogous to those found in mouse models. Methods We used advanced magnetic resonance imaging techniques, including surface‐based morphometry and region‐of‐interest analysis in adult humans heterozygously mutated for PAX6 mutations (n = 19 subjects and n = 21 controls). Using immunohistochemistry, we also studied PAX6 expression in the adult brain tissue of healthy subjects (n = 4) and patients with epilepsy (n = 42), some of whom had focal injuries due to intracranial electrode track placement (n = 17). Results There were significant reductions in frontoparietal cortical area after correcting for age and intracranial volume. A greater decline in thickness of the frontoparietal cortex with age, in subjects with PAX6 mutations compared to controls, correlated with age‐corrected, accelerated decline in working memory. These results also demonstrate genotypic effects: those subjects with the most severe genotypes have the most widespread differences compared with controls. We also demonstrated significant increases in PAX6‐expressing cells in response to acute injury in the adult human brain. Interpretation These findings suggest a role for PAX6 in the maintenance and consequent functioning of the adult brain, homologous to that found in other tissues. This has significant implications for the understanding and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Mahinda Yogarajah
- Department of Clinical and Experimental Epilepsy UCL Institute of Neurology National Hospital for Neurology and Neurosurgery London WC1N 3BG United Kingdom; Present address: St George's University Hospitals NHS Foundation Trust London United Kingdom
| | - Mar Matarin
- Department of Clinical and Experimental Epilepsy UCL Institute of Neurology National Hospital for Neurology and Neurosurgery London WC1N 3BG United Kingdom
| | - Christian Vollmar
- Department of Clinical and Experimental Epilepsy UCL Institute of Neurology National Hospital for Neurology and Neurosurgery London WC1N 3BG United Kingdom
| | - Pamela J Thompson
- Department of Clinical and Experimental Epilepsy UCL Institute of Neurology National Hospital for Neurology and Neurosurgery London WC1N 3BG United Kingdom
| | - John S Duncan
- Department of Clinical and Experimental Epilepsy UCL Institute of Neurology National Hospital for Neurology and Neurosurgery London WC1N 3BG United Kingdom
| | - Mark Symms
- Department of Clinical and Experimental Epilepsy UCL Institute of Neurology National Hospital for Neurology and Neurosurgery London WC1N 3BG United Kingdom
| | - Anthony T Moore
- UCL Institute of Ophthalmology and Moorfields Eye Hospital London United Kingdom; Present address: Department of Ophthalmology University of California San Francisco California
| | - Joan Liu
- Division of Neuropathology UCL Institute of Neurology National Hospital for Neurology and Neurosurgery London United Kingdom
| | - Maria Thom
- Division of Neuropathology UCL Institute of Neurology National Hospital for Neurology and Neurosurgery London United Kingdom
| | - Veronica van Heyningen
- MRC Human Genetics UnitI GMM University of Edinburgh Crewe Road Edinburgh EH4 2XU United Kingdom; Present address: UCL Institute of Ophthalmology London United Kingdom
| | - Sanjay M Sisodiya
- Department of Clinical and Experimental Epilepsy UCL Institute of Neurology National Hospital for Neurology and Neurosurgery London WC1N 3BG United Kingdom; Epilepsy Society Chalfont-St-Peter Bucks SL9 0RJ United Kingdom
| |
Collapse
|
28
|
Rennert RC, Schäfer R, Bliss T, Januszyk M, Sorkin M, Achrol AS, Rodrigues M, Maan ZN, Kluba T, Steinberg GK, Gurtner GC. High-Resolution Microfluidic Single-Cell Transcriptional Profiling Reveals Clinically Relevant Subtypes among Human Stem Cell Populations Commonly Utilized in Cell-Based Therapies. Front Neurol 2016; 7:41. [PMID: 27047447 PMCID: PMC4801858 DOI: 10.3389/fneur.2016.00041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 03/10/2016] [Indexed: 12/21/2022] Open
Abstract
Stem cell therapies can promote neural repair and regeneration, yet controversy regarding optimal cell source and mechanism of action has slowed clinical translation, potentially due to undefined cellular heterogeneity. Single-cell resolution is needed to identify clinically relevant subpopulations with the highest therapeutic relevance. We combine single-cell microfluidic analysis with advanced computational modeling to study for the first time two common sources for cell-based therapies, human NSCs and MSCs. This methodology has the potential to logically inform cell source decisions for any clinical application.
Collapse
Affiliation(s)
- Robert C Rennert
- Department of Surgery, Stanford University School of Medicine , Stanford, CA , USA
| | - Richard Schäfer
- Department of Neurosurgery, Stanford University School of Medicine , Stanford, CA , USA
| | - Tonya Bliss
- Department of Neurosurgery, Stanford University School of Medicine , Stanford, CA , USA
| | - Michael Januszyk
- Department of Surgery, Stanford University School of Medicine , Stanford, CA , USA
| | - Michael Sorkin
- Department of Surgery, Stanford University School of Medicine , Stanford, CA , USA
| | - Achal S Achrol
- Department of Neurosurgery, Stanford University School of Medicine , Stanford, CA , USA
| | - Melanie Rodrigues
- Department of Surgery, Stanford University School of Medicine , Stanford, CA , USA
| | - Zeshaan N Maan
- Department of Surgery, Stanford University School of Medicine , Stanford, CA , USA
| | - Torsten Kluba
- Department of Orthopedics, University Hospital Tübingen , Tübingen , Germany
| | - Gary K Steinberg
- Department of Neurosurgery, Stanford University School of Medicine , Stanford, CA , USA
| | - Geoffrey C Gurtner
- Department of Surgery, Stanford University School of Medicine , Stanford, CA , USA
| |
Collapse
|
29
|
Bery A, Mérot Y, Rétaux S. Genes expressed in mouse cortical progenitors are enriched in Pax, Lhx, and Sox transcription factor putative binding sites. Brain Res 2015; 1633:37-51. [PMID: 26721689 DOI: 10.1016/j.brainres.2015.12.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 11/25/2015] [Accepted: 12/14/2015] [Indexed: 11/19/2022]
Abstract
Considerable progress has been made in the understanding of molecular and cellular mechanisms controlling the development of the mammalian cortex. The proliferative and neurogenic properties of cortical progenitors located in the ventricular germinal zone start being understood. Little is known however on the cis-regulatory control that finely tunes gene expression in these progenitors. Here, we undertook an in silico-based approach to address this question, followed by some functional validation. Using the Eurexpress database, we established a list of 30 genes specifically expressed in the cortical germinal zone, we selected mouse/human conserved non-coding elements (CNEs) around these genes and we performed motif-enrichment search in these CNEs. We found an over-representation of motifs corresponding to binding sites for Pax, Sox, and Lhx transcription factors, often found as pairs and located within 100bp windows. A small subset of CNEs (n=7) was tested for enhancer activity, by ex-vivo and in utero electroporation assays. Two showed strong enhancer activity in the germinal zone progenitors. Mutagenesis experiments on a selected CNE showed the functional importance of the Pax, Sox, and Lhx TFBS for conferring enhancer activity to the CNE. Overall, from a cis-regulatory viewpoint, our data suggest an input from Pax, Sox and Lhx transcription factors to orchestrate corticogenesis. These results are discussed with regards to the known functional roles of Pax6, Sox2 and Lhx2 in cortical development.
Collapse
Affiliation(s)
- Amandine Bery
- DECA Group, Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, Université Paris-Sud, CNRS, UMR 9197, 91198 Gif-sur-Yvette, France.
| | - Yohann Mérot
- DECA Group, Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, Université Paris-Sud, CNRS, UMR 9197, 91198 Gif-sur-Yvette, France
| | - Sylvie Rétaux
- DECA Group, Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, Université Paris-Sud, CNRS, UMR 9197, 91198 Gif-sur-Yvette, France.
| |
Collapse
|
30
|
Suzuki T, Takayama R, Sato M. eyeless/Pax6 controls the production of glial cells in the visual center of Drosophila melanogaster. Dev Biol 2015; 409:343-53. [PMID: 26670857 DOI: 10.1016/j.ydbio.2015.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 11/11/2015] [Accepted: 12/04/2015] [Indexed: 10/22/2022]
Abstract
Pax6 is known as a neurogenic factor in the development of the central nervous system and regulates proliferation of neuronal progenitor cells and promotes neuronal differentiation. In addition to neurogenesis, Pax6 is also involved in the specification and maturation of glial cells. Here, we show that Eyeless (Ey), Drosophila homolog of Pax6, regulates the production of glial cells in the brain. In the developing fly visual center, the production of neurons and glial cells are controlled by the temporal transcription factors that are sequentially expressed in neuroblasts (NBs). Among them, NBs of the last temporal window produce astrocyte-like glial cells. Ey is strongly expressed in the middle aged NBs, whose temporal window is earlier compared with glia producing older NBs. Weak Ey expression is also detected in the glia producing NBs. Our results suggest that Ey expression in the middle aged NBs indirectly control gliogenesis from the oldest NBs by regulating other temporal transcription factors. Additionally, weak Ey expression in the NBs of last temporal window may directly control gliogenesis. Ey is also expressed in neurons produced from the NBs of Ey-positive temporal window. Interestingly, neuron-specific overexpression of Ey causes significant increase in glial cells suggesting that neuronal expression of Ey may also contribute to gliogenesis. Thus, Pax6-dependent regulation of astrocyte-like glial development is conserved throughout the animal kingdom.
Collapse
Affiliation(s)
- Takumi Suzuki
- Laboratory of Developmental Neurobiology, Brain/Liver Interface Medicine Research Center, Kanazawa University, 13-1 Takaramachi Kanazawa-shi, Ishikawa 920-8640, Japan
| | - Rie Takayama
- Laboratory of Developmental Neurobiology, Brain/Liver Interface Medicine Research Center, Kanazawa University, 13-1 Takaramachi Kanazawa-shi, Ishikawa 920-8640, Japan; CREST, JST, 4-1-8 Honcho Kawaguchi, Saitama 332-0012, Japan
| | - Makoto Sato
- Laboratory of Developmental Neurobiology, Brain/Liver Interface Medicine Research Center, Kanazawa University, 13-1 Takaramachi Kanazawa-shi, Ishikawa 920-8640, Japan; Graduate School of Medical Sciences, Kanazawa University, 13-1 Takaramachi Kanazawa-shi, Ishikawa 920-8640, Japan; CREST, JST, 4-1-8 Honcho Kawaguchi, Saitama 332-0012, Japan.
| |
Collapse
|
31
|
O'Shea KS, McInnis MG. Neurodevelopmental origins of bipolar disorder: iPSC models. Mol Cell Neurosci 2015; 73:63-83. [PMID: 26608002 DOI: 10.1016/j.mcn.2015.11.006] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Revised: 10/14/2015] [Accepted: 11/18/2015] [Indexed: 12/22/2022] Open
Abstract
Bipolar disorder (BP) is a chronic neuropsychiatric condition characterized by pathological fluctuations in mood from mania to depression. Adoption, twin and family studies have consistently identified a significant hereditary component to BP, yet there is no clear genetic event or consistent neuropathology. BP has been suggested to have a developmental origin, although this hypothesis has been difficult to test since there are no viable neurons or glial cells to analyze, and research has relied largely on postmortem brain, behavioral and imaging studies, or has examined proxy tissues including saliva, olfactory epithelium and blood cells. Neurodevelopmental factors, particularly pathways related to nervous system development, cell migration, extracellular matrix, H3K4 methylation, and calcium signaling have been identified in large gene expression and GWAS studies as altered in BP. Recent advances in stem cell biology, particularly the ability to reprogram adult somatic tissues to a pluripotent state, now make it possible to interrogate these pathways in viable cell models. A number of induced pluripotent stem cell (iPSC) lines from BP patient and healthy control (C) individuals have been derived in several laboratories, and their ability to form cortical neurons examined. Early studies suggest differences in activity, calcium signaling, blocks to neuronal differentiation, and changes in neuronal, and possibly glial, lineage specification. Initial observations suggest that differentiation of BP patient-derived neurons to dorsal telencephalic derivatives may be impaired, possibly due to alterations in WNT, Hedgehog or Nodal pathway signaling. These investigations strongly support a developmental contribution to BP and identify novel pathways, mechanisms and opportunities for improved treatments.
Collapse
Affiliation(s)
- K Sue O'Shea
- Department of Cell and Developmental Biology, University of Michigan, 3051 BSRB, 109 Zina Pitcher PL, Ann Arbor, MI 48109-2200, United States; Department of Psychiatry, University of Michigan, 4250 Plymouth Rd, Ann Arbor, MI 48109-5765, United States.
| | - Melvin G McInnis
- Department of Psychiatry, University of Michigan, 4250 Plymouth Rd, Ann Arbor, MI 48109-5765, United States
| |
Collapse
|
32
|
Ypsilanti AR, Rubenstein JLR. Transcriptional and epigenetic mechanisms of early cortical development: An examination of how Pax6 coordinates cortical development. J Comp Neurol 2015; 524:609-29. [PMID: 26304102 DOI: 10.1002/cne.23866] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 07/14/2015] [Accepted: 07/17/2015] [Indexed: 12/26/2022]
Abstract
The development of the cortex is an elaborate process that integrates a plethora of finely tuned molecular processes ranging from carefully regulated gradients of transcription factors, dynamic changes in the chromatin landscape, or formation of protein complexes to elicit and regulate transcription. Combined with cellular processes such as cell type specification, proliferation, differentiation, and migration, all of these developmental processes result in the establishment of an adult mammalian cortex with its typical lamination and regional patterning. By examining in-depth the role of one transcription factor, Pax6, on the regulation of cortical development, its integration in the regulation of chromatin state, and its regulation by cis-regulatory elements, we aim to demonstrate the importance of integrating each level of regulation in our understanding of cortical development.
Collapse
Affiliation(s)
- Athéna R Ypsilanti
- Department of Psychiatry, Neuroscience Program, and the Nina Ireland Laboratory of Developmental Neurobiology, University of California, San Francisco, San Francisco, California
| | - John L R Rubenstein
- Department of Psychiatry, Neuroscience Program, and the Nina Ireland Laboratory of Developmental Neurobiology, University of California, San Francisco, San Francisco, California
| |
Collapse
|
33
|
Wong FK, Fei JF, Mora-Bermúdez F, Taverna E, Haffner C, Fu J, Anastassiadis K, Stewart AF, Huttner WB. Sustained Pax6 Expression Generates Primate-like Basal Radial Glia in Developing Mouse Neocortex. PLoS Biol 2015; 13:e1002217. [PMID: 26252244 PMCID: PMC4529158 DOI: 10.1371/journal.pbio.1002217] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 06/30/2015] [Indexed: 11/21/2022] Open
Abstract
The evolutionary expansion of the neocortex in mammals has been linked to enlargement of the subventricular zone (SVZ) and increased proliferative capacity of basal progenitors (BPs), notably basal radial glia (bRG). The transcription factor Pax6 is known to be highly expressed in primate, but not mouse, BPs. Here, we demonstrate that sustaining Pax6 expression selectively in BP-genic apical radial glia (aRG) and their BP progeny of embryonic mouse neocortex suffices to induce primate-like progenitor behaviour. Specifically, we conditionally expressed Pax6 by in utero electroporation using a novel, Tis21–CreERT2 mouse line. This expression altered aRG cleavage plane orientation to promote bRG generation, increased cell-cycle re-entry of BPs, and ultimately increased upper-layer neuron production. Upper-layer neuron production was also increased in double-transgenic mouse embryos with sustained Pax6 expression in the neurogenic lineage. Strikingly, increased BPs existed not only in the SVZ but also in the intermediate zone of the neocortex of these double-transgenic mouse embryos. In mutant mouse embryos lacking functional Pax6, the proportion of bRG among BPs was reduced. Our data identify specific Pax6 effects in BPs and imply that sustaining this Pax6 function in BPs could be a key aspect of SVZ enlargement and, consequently, the evolutionary expansion of the neocortex. "Humanizing" the expression of the transcription factor Pax6 in cortical progenitors in the developing mouse brain is sufficient to endow these progenitors with a primate-like proliferative capacity. During development, neural progenitors generate all cells that make up the mammalian brain. Differences in brain size among the various mammalian species are attributed to differences in the abundance and proliferative capacity of a specific class of neural progenitors called basal progenitors. Among these, a specific progenitor type called basal radial glia is thought to have played an important role during evolution in the expansion of the neocortex, the part of the brain associated with higher cognitive functions like conscious thought and language. In the neocortex, the expression of the transcription factor Pax6 in basal progenitors is low in rodents, but high in primates, including humans. In this study, we aimed to mimic the elevated expression pattern of Pax6 seen in humans in basal progenitors of the embryonic mouse neocortex. To this end, we generated a novel, transgenic mouse line that allows sustained expression of the Pax6 gene in basal progenitors. This elevated expression resulted in an increase in the generation of basal radial glia, in the proliferative capacity of basal progenitors, and, ultimately, in the number of neurons produced. Our findings demonstrate that altering the expression of a single transcription factor from a mouse to a human-like pattern suffices to induce a primate-like proliferative behaviour in neural progenitors, which is thought to underlie the evolutionary expansion of the neocortex.
Collapse
Affiliation(s)
- Fong Kuan Wong
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Ji-Feng Fei
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | - Elena Taverna
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Christiane Haffner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Jun Fu
- Biotechnology Center of the Technische Universität Dresden, Dresden, Germany
| | | | - A. Francis Stewart
- Biotechnology Center of the Technische Universität Dresden, Dresden, Germany
| | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- * E-mail:
| |
Collapse
|
34
|
Curto GG, Gard C, Ribes V. Structures and properties of PAX linked regulatory networks architecting and pacing the emergence of neuronal diversity. Semin Cell Dev Biol 2015; 44:75-86. [DOI: 10.1016/j.semcdb.2015.09.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 09/07/2015] [Accepted: 09/16/2015] [Indexed: 12/13/2022]
|
35
|
Tylkowski MA, Yang K, Hoyer-Fender S, Stoykova A. Pax6 controls centriole maturation in cortical progenitors through Odf2. Cell Mol Life Sci 2015; 72:1795-809. [PMID: 25352170 PMCID: PMC11114037 DOI: 10.1007/s00018-014-1766-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 10/14/2014] [Accepted: 10/17/2014] [Indexed: 12/11/2022]
Abstract
Cortical glutamatergic neurons are generated by radial glial cells (RGCs), specified by the expression of transcription factor (TF) Pax6, in the germinative zones of the dorsal telencephalon. Here, we demonstrate that Pax6 regulates the structural assembly of the interphase centrosomes. In the cortex of the Pax6-deficient Small eye (Sey/Sey) mutant, we find a defect of the appendages of the mother centrioles, indicating incomplete centrosome maturation. Consequently, RGCs fail to generate primary cilia, and instead of staying in the germinative zone for renewal, RGCs detach from the ventricular surface thus affecting the interkinetic nuclear migration and they exit prematurely from mitosis. Mechanistically, we show that TF Pax6 directly regulates the activity of the Odf2 gene encoding for the appendage-specific protein Odf2 with a role for the assembly of mother centriole. Our findings demonstrate a molecular mechanism that explains important characteristics of the centrosome disassembly and malfunctioning in developing cortex lacking Pax6.
Collapse
Affiliation(s)
- Marco A. Tylkowski
- Research Group of Molecular Developmental Neurobiology, Department Molecular Cell Biology, Max-Planck Institute for Biophysical Chemistry, Am Faßberg 11, 37077 Göttingen, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), 37075 Göttingen, Germany
| | - Kefei Yang
- Johann-Friedrich-Blumenbach-Institute of Zoology and Anthropology, Developmental Biology, GZMB, Ernst-Caspari-Haus, Georg-August-Universität Göttingen, Justus-von-Liebig-Weg 11, Göttingen, Germany
| | - Sigrid Hoyer-Fender
- Johann-Friedrich-Blumenbach-Institute of Zoology and Anthropology, Developmental Biology, GZMB, Ernst-Caspari-Haus, Georg-August-Universität Göttingen, Justus-von-Liebig-Weg 11, Göttingen, Germany
| | - Anastassia Stoykova
- Research Group of Molecular Developmental Neurobiology, Department Molecular Cell Biology, Max-Planck Institute for Biophysical Chemistry, Am Faßberg 11, 37077 Göttingen, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), 37075 Göttingen, Germany
| |
Collapse
|
36
|
Manuel MN, Mi D, Mason JO, Price DJ. Regulation of cerebral cortical neurogenesis by the Pax6 transcription factor. Front Cell Neurosci 2015; 9:70. [PMID: 25805971 PMCID: PMC4354436 DOI: 10.3389/fncel.2015.00070] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 02/18/2015] [Indexed: 12/19/2022] Open
Abstract
Understanding brain development remains a major challenge at the heart of understanding what makes us human. The neocortex, in evolutionary terms the newest part of the cerebral cortex, is the seat of higher cognitive functions. Its normal development requires the production, positioning, and appropriate interconnection of very large numbers of both excitatory and inhibitory neurons. Pax6 is one of a relatively small group of transcription factors that exert high-level control of cortical development, and whose mutation or deletion from developing embryos causes major brain defects and a wide range of neurodevelopmental disorders. Pax6 is very highly conserved between primate and non-primate species, is expressed in a gradient throughout the developing cortex and is essential for normal corticogenesis. Our understanding of Pax6’s functions and the cellular processes that it regulates during mammalian cortical development has significantly advanced in the last decade, owing to the combined application of genetic and biochemical analyses. Here, we review the functional importance of Pax6 in regulating cortical progenitor proliferation, neurogenesis, and formation of cortical layers and highlight important differences between rodents and primates. We also review the pathological effects of PAX6 mutations in human neurodevelopmental disorders. We discuss some aspects of Pax6’s molecular actions including its own complex transcriptional regulation, the distinct molecular functions of its splice variants and some of Pax6’s known direct targets which mediate its actions during cortical development.
Collapse
Affiliation(s)
- Martine N Manuel
- Centre for Integrative Physiology, The University of Edinburgh, Edinburgh UK
| | - Da Mi
- Centre for Integrative Physiology, The University of Edinburgh, Edinburgh UK
| | - John O Mason
- Centre for Integrative Physiology, The University of Edinburgh, Edinburgh UK
| | - David J Price
- Centre for Integrative Physiology, The University of Edinburgh, Edinburgh UK
| |
Collapse
|
37
|
Curto GG, Nieto-Estévez V, Hurtado-Chong A, Valero J, Gómez C, Alonso JR, Weruaga E, Vicario-Abejón C. Pax6 is essential for the maintenance and multi-lineage differentiation of neural stem cells, and for neuronal incorporation into the adult olfactory bulb. Stem Cells Dev 2014; 23:2813-30. [PMID: 25117830 DOI: 10.1089/scd.2014.0058] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The paired type homeobox 6 (Pax6) transcription factor (TF) regulates multiple aspects of neural stem cell (NSC) and neuron development in the embryonic central nervous system. However, less is known about the role of Pax6 in the maintenance and differentiation of adult NSCs and in adult neurogenesis. Using the +/Sey(Dey) mouse, we have analyzed how Pax6 heterozygosis influences the self-renewal and proliferation of adult olfactory bulb stem cells (aOBSCs). In addition, we assessed its influence on neural differentiation, neuronal incorporation, and cell death in the adult OB, both in vivo and in vitro. Our results indicate that the Pax6 mutation alters Nestin(+)-cell proliferation in vivo, as well as self-renewal, proliferation, and survival of aOBSCs in vitro although a subpopulation of +/Sey(Dey) progenitors is able to expand partially similar to wild-type progenitors. This mutation also impairs aOBSC differentiation into neurons and oligodendrocytes, whereas it increases cell death while preserving astrocyte survival and differentiation. Furthermore, Pax6 heterozygosis causes a reduction in the variety of neurochemical interneuron subtypes generated from aOBSCs in vitro and in the incorporation of newly generated neurons into the OB in vivo. Our findings support an important role of Pax6 in the maintenance of aOBSCs by regulating cell death, self-renewal, and cell fate, as well as in neuronal incorporation into the adult OB. They also suggest that deregulation of the cell cycle machinery and TF expression in aOBSCs which are deficient in Pax6 may be at the origin of the phenotypes observed in this adult NSC population.
Collapse
Affiliation(s)
- Gloria G Curto
- 1 Instituto de Neurociencias de Castilla y León (INCyL), Universidad de Salamanca , Salamanca, Spain
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Tuoc TC, Pavlakis E, Tylkowski MA, Stoykova A. Control of cerebral size and thickness. Cell Mol Life Sci 2014; 71:3199-218. [PMID: 24614969 PMCID: PMC11113230 DOI: 10.1007/s00018-014-1590-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 02/10/2014] [Accepted: 02/13/2014] [Indexed: 11/24/2022]
Abstract
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.
Collapse
Affiliation(s)
- Tran Cong Tuoc
- Institute of Neuroanatomy, Universitätsmedizin Göttingen, Kreuzbergring 40, 37075, Göttingen, Germany,
| | | | | | | |
Collapse
|
39
|
Díaz-Alonso J, Aguado T, de Salas-Quiroga A, Ortega Z, Guzmán M, Galve-Roperh I. CB1 Cannabinoid Receptor-Dependent Activation of mTORC1/Pax6 Signaling Drives Tbr2 Expression and Basal Progenitor Expansion in the Developing Mouse Cortex. Cereb Cortex 2014; 25:2395-408. [PMID: 24610119 DOI: 10.1093/cercor/bhu039] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The CB1 cannabinoid receptor regulates cortical progenitor proliferation during embryonic development, but the molecular mechanism of this action remains unknown. Here, we report that CB1-deficient mouse embryos show premature cell cycle exit, decreased Pax6- and Tbr2-positive cell number, and reduced mammalian target of rapamycin complex 1 (mTORC1) activation in the ventricular and subventricular cortical zones. Pharmacological stimulation of the CB1 receptor in cortical slices and progenitor cell cultures activated the mTORC1 pathway and increased the number of Pax6- and Tbr2-expressing cells. Likewise, acute CB1 knockdown in utero reduced mTORC1 activation and cannabinoid-induced Tbr2-positive cell generation. Luciferase reporter and chromatin immunoprecipitation assays revealed that the CB1 receptor drives Tbr2 expression downstream of Pax6 induction in an mTORC1-dependent manner. Altogether, our results demonstrate that the CB1 receptor tunes dorsal telencephalic progenitor proliferation by sustaining the transcriptional activity of the Pax6-Tbr2 axis via the mTORC1 pathway, and suggest that alterations of CB1 receptor signaling, by producing the missexpression of progenitor identity determinants may contribute to neurodevelopmental alterations.
Collapse
Affiliation(s)
- Javier Díaz-Alonso
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Instituto Universitario de Investigaciones Neuroquímicas (IUIN), Department of Biochemistry and Molecular Biology I, Complutense University, Madrid 28040, Spain
| | - Tania Aguado
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Instituto Universitario de Investigaciones Neuroquímicas (IUIN), Department of Biochemistry and Molecular Biology I, Complutense University, Madrid 28040, Spain
| | - Adán de Salas-Quiroga
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Instituto Universitario de Investigaciones Neuroquímicas (IUIN), Department of Biochemistry and Molecular Biology I, Complutense University, Madrid 28040, Spain
| | - Zaira Ortega
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Instituto Universitario de Investigaciones Neuroquímicas (IUIN), Department of Biochemistry and Molecular Biology I, Complutense University, Madrid 28040, Spain
| | - Manuel Guzmán
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Instituto Universitario de Investigaciones Neuroquímicas (IUIN), Department of Biochemistry and Molecular Biology I, Complutense University, Madrid 28040, Spain
| | - Ismael Galve-Roperh
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Instituto Universitario de Investigaciones Neuroquímicas (IUIN), Department of Biochemistry and Molecular Biology I, Complutense University, Madrid 28040, Spain
| |
Collapse
|
40
|
Aboitiz F, Zamorano F. Neural progenitors, patterning and ecology in neocortical origins. Front Neuroanat 2013; 7:38. [PMID: 24273496 PMCID: PMC3824149 DOI: 10.3389/fnana.2013.00038] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 10/21/2013] [Indexed: 01/13/2023] Open
Abstract
The anatomical organization of the mammalian neocortex stands out among vertebrates for its laminar and columnar arrangement, featuring vertically oriented, excitatory pyramidal neurons. The evolutionary origin of this structure is discussed here in relation to the brain organization of other amniotes, i.e., the sauropsids (reptiles and birds). Specifically, we address the developmental modifications that had to take place to generate the neocortex, and to what extent these modifications were shared by other amniote lineages or can be considered unique to mammals. In this article, we propose a hypothesis that combines the control of proliferation in neural progenitor pools with the specification of regional morphogenetic gradients, yielding different anatomical results by virtue of the differential modulation of these processes in each lineage. Thus, there is a highly conserved genetic and developmental battery that becomes modulated in different directions according to specific selective pressures. In the case of early mammals, ecological conditions like nocturnal habits and reproductive strategies are considered to have played a key role in the selection of the particular brain patterning mechanisms that led to the origin of the neocortex.
Collapse
Affiliation(s)
- Francisco Aboitiz
- Departamento de Psiquiatría, Facultad de Medicina y Centro Interdisciplinario de Neurociencia, Pontificia Universidad Católica de Chile Santiago, Chile
| | | |
Collapse
|
41
|
Mi D, Huang YT, Kleinjan DA, Mason JO, Price DJ. Identification of genomic regions regulating Pax6 expression in embryonic forebrain using YAC reporter transgenic mouse lines. PLoS One 2013; 8:e80208. [PMID: 24223221 PMCID: PMC3819282 DOI: 10.1371/journal.pone.0080208] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 09/28/2013] [Indexed: 11/28/2022] Open
Abstract
The transcription factor Pax6 is a crucial regulator of eye and central nervous system development. Both the spatiotemporal patterns and the precise levels of Pax6 expression are subject to tight control, mediated by an extensive set of cis-regulatory elements. Previous studies have shown that a YAC reporter transgene containing 420Kb of genomic DNA spanning the human PAX6 locus drives expression of a tau-tagged GFP reporter in mice in a pattern that closely resembles that of endogenous Pax6. Here we have closely compared the pattern of tau-GFP reporter expression at the cellular level in the forebrains and eyes of transgenic mice carrying either complete or truncated versions of the YAC reporter transgene with endogenous Pax6 expression and found several areas where expression of tau-GFP and Pax6 diverge. Some discrepancies are due to differences between the intracellular localization or perdurance of tau-GFP and Pax6 proteins, while others are likely to be a consequence of transcriptional differences. We show that cis-regulatory elements that lie outside the 420kb fragment of PAX6 are required for correct expression around the pallial-subpallial boundary, in the amygdala and the prethalamus. Further, we found that the YAC reporter transgene effectively labels cells that contribute to the lateral cortical stream, including cells that arise from the pallium and subpallium, and therefore represents a useful tool for studying lateral cortical stream migration.
Collapse
Affiliation(s)
- Da Mi
- Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- * E-mail: (DM); (DP)
| | - Yu-Ting Huang
- Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Dirk A. Kleinjan
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - John O. Mason
- Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - David J. Price
- Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- * E-mail: (DM); (DP)
| |
Collapse
|
42
|
Abstract
Animal data show that cortical development is initially patterned by genetic gradients largely along three orthogonal axes. We previously reported differences in genetic influences on cortical surface area along an anterior-posterior axis using neuroimaging data of adult human twins. Here, we demonstrate differences in genetic influences on cortical thickness along a dorsal-ventral axis in the same cohort. The phenomenon of orthogonal gradations in cortical organization evident in different structural and functional properties may originate from genetic gradients. Another emerging theme of cortical patterning is that patterns of genetic influences recapitulate the spatial topography of the cortex within hemispheres. The genetic patterning of both cortical thickness and surface area corresponds to cortical functional specializations. Intriguingly, in contrast to broad similarities in genetic patterning, two sets of analyses distinguish cortical thickness and surface area genetically. First, genetic contributions to cortical thickness and surface area are largely distinct; there is very little genetic correlation (i.e., shared genetic influences) between them. Second, organizing principles among genetically defined regions differ between thickness and surface area. Examining the structure of the genetic similarity matrix among clusters revealed that, whereas surface area clusters showed great genetic proximity with clusters from the same lobe, thickness clusters appear to have close genetic relatedness with clusters that have similar maturational timing. The discrepancies are in line with evidence that the two traits follow different mechanisms in neurodevelopment. Our findings highlight the complexity of genetic influences on cortical morphology and provide a glimpse into emerging principles of genetic organization of the cortex.
Collapse
|
43
|
Pax6-Dependent Cortical Glutamatergic Neuronal Differentiation Regulates Autism-Like Behavior in Prenatally Valproic Acid-Exposed Rat Offspring. Mol Neurobiol 2013; 49:512-28. [DOI: 10.1007/s12035-013-8535-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 08/15/2013] [Indexed: 12/19/2022]
|
44
|
Tuoc TC, Narayanan R, Stoykova A. BAF chromatin remodeling complex: cortical size regulation and beyond. Cell Cycle 2013; 12:2953-9. [PMID: 23974113 DOI: 10.4161/cc.25999] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The multi-subunit chromatin remodeling BAF complex controls different developmental processes. Using cortex-specific conditional knockout and overexpression mouse models, we have recently reported that BAF170, a subunit of the vertebrate BAF chromatin remodeling complex, interacts with transcription factor (TF) Pax6 to control cortical size and volume. The mechanistic basis includes suppression of the expression of Pax6 target genes, which are required for genesis of cortical intermediate progenitors (IPs) and specification of late neuronal subtype identity. In addition, we showed that a dynamic competition between BAF170 and BAF155 subunits within the BAF complex during progression of neurogenesis is a primary event in modulating the size of the mammalian cortex. Here, we present additional insights into the interaction between the BAF complex and TF Pax6 in the genesis of IPs of the developing cortex. Furthermore, we show that such competition between BAF170 and BAF155 is involved as well in the determination of the size of the embryonic body. Our results add new insights into a cell-intrinsic mechanism, mediated by the chromatin remodeling BAF complex that controls vertebrate body shape and size.
Collapse
Affiliation(s)
- Tran Cong Tuoc
- Institute of Neuroanatomy; Universitätsmedizin Göttingen; Göttingen, Germany
| | | | | |
Collapse
|
45
|
Drury-Stewart D, Song M, Mohamad O, Guo Y, Gu X, Chen D, Wei L. Highly efficient differentiation of neural precursors from human embryonic stem cells and benefits of transplantation after ischemic stroke in mice. Stem Cell Res Ther 2013; 4:93. [PMID: 23928330 PMCID: PMC3854684 DOI: 10.1186/scrt292] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 07/26/2013] [Indexed: 02/07/2023] Open
Abstract
Introduction Ischemic stroke is a leading cause of death and disability, but treatment options are severely limited. Cell therapy offers an attractive strategy for regenerating lost tissues and enhancing the endogenous healing process. In this study, we investigated the use of human embryonic stem cell-derived neural precursors as a cell therapy in a murine stroke model. Methods Neural precursors were derived from human embryonic stem cells by using a fully adherent SMAD inhibition protocol employing small molecules. The efficiency of neural induction and the ability of these cells to further differentiate into neurons were assessed by using immunocytochemistry. Whole-cell patch-clamp recording was used to demonstrate the electrophysiological activity of human embryonic stem cell-derived neurons. Neural precursors were transplanted into the core and penumbra regions of a focal ischemic stroke in the barrel cortex of mice. Animals received injections of bromodeoxyuridine to track regeneration. Neural differentiation of the transplanted cells and regenerative markers were measured by using immunohistochemistry. The adhesive removal test was used to determine functional improvement after stroke and intervention. Results After 11 days of neural induction by using the small-molecule protocol, over 95% of human embryonic stem-derived cells expressed at least one neural marker. Further in vitro differentiation yielded cells that stained for mature neuronal markers and exhibited high-amplitude, repetitive action potentials in response to depolarization. Neuronal differentiation also occurred after transplantation into the ischemic cortex. A greater level of bromodeoxyuridine co-localization with neurons was observed in the penumbra region of animals receiving cell transplantation. Transplantation also improved sensory recovery in transplant animals over that in control animals. Conclusions Human embryonic stem cell-derived neural precursors derived by using a highly efficient small-molecule SMAD inhibition protocol can differentiate into electrophysiologically functional neurons in vitro. These cells also differentiate into neurons in vivo, enhance regenerative activities, and improve sensory recovery after ischemic stroke.
Collapse
|
46
|
Pax6 exerts regional control of cortical progenitor proliferation via direct repression of Cdk6 and hypophosphorylation of pRb. Neuron 2013; 78:269-84. [PMID: 23622063 PMCID: PMC3898967 DOI: 10.1016/j.neuron.2013.02.012] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2013] [Indexed: 11/22/2022]
Abstract
The mechanisms by which early spatiotemporal expression patterns of transcription factors such as Pax6 regulate cortical progenitors in a region-specific manner are poorly understood. Pax6 is expressed in a gradient across the developing cortex and is essential for normal corticogenesis. We found that constitutive or conditional loss of Pax6 increases cortical progenitor proliferation by amounts that vary regionally with normal Pax6 levels. We compared the gene expression profiles of equivalent Pax6-expressing progenitors isolated from Pax6+/+ and Pax6−/− cortices and identified many negatively regulated cell-cycle genes, including Cyclins and Cdks. Biochemical assays indicated that Pax6 directly represses Cdk6 expression. Cyclin/Cdk repression inhibits retinoblastoma protein (pRb) phosphorylation, thereby limiting the transcription of genes that directly promote the mechanics of the cell cycle, and we found that Pax6 inhibits pRb phosphorylation and represses genes involved in DNA replication. Our results indicate that Pax6’s modulation of cortical progenitor cell cycles is regional and direct. Pax6 loss increases cortical progenitor proliferation by region-specific amounts The size of this effect correlates directly with normal Pax6 expression levels Expression of many key cell-cycle regulators is increased in the absence of Pax6 Pax6 directly represses Cdk6 expression and controls pRb phosphorylation
Collapse
|
47
|
Tuoc TC, Boretius S, Sansom SN, Pitulescu ME, Frahm J, Livesey FJ, Stoykova A. Chromatin regulation by BAF170 controls cerebral cortical size and thickness. Dev Cell 2013; 25:256-69. [PMID: 23643363 DOI: 10.1016/j.devcel.2013.04.005] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 02/21/2013] [Accepted: 04/07/2013] [Indexed: 11/26/2022]
Abstract
Increased cortical size is essential to the enhanced intellectual capacity of primates during mammalian evolution. The mechanisms that control cortical size are largely unknown. Here, we show that mammalian BAF170, a subunit of the chromatin remodeling complex mSWI/SNF, is an intrinsic factor that controls cortical size. We find that conditional deletion of BAF170 promotes indirect neurogenesis by increasing the pool of intermediate progenitors (IPs) and results in an enlarged cortex, whereas cortex-specific BAF170 overexpression results in the opposite phenotype. Mechanistically, BAF170 competes with BAF155 subunit in the BAF complex, affecting euchromatin structure and thereby modulating the binding efficiency of the Pax6/REST-corepressor complex to Pax6 target genes that regulate the generation of IPs and late cortical progenitors. Our findings reveal a molecular mechanism mediated by the mSWI/SNF chromatin-remodeling complex that controls cortical architecture.
Collapse
Affiliation(s)
- Tran Cong Tuoc
- Research Group of Molecular Developmental Neurobiology, Department of Molecular Cell Biology, Max-Planck-Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| | | | | | | | | | | | | |
Collapse
|
48
|
Hubbard KS, Gut IM, Lyman ME, McNutt PM. Longitudinal RNA sequencing of the deep transcriptome during neurogenesis of cortical glutamatergic neurons from murine ESCs. F1000Res 2013; 2:35. [PMID: 24358889 PMCID: PMC3829120 DOI: 10.12688/f1000research.2-35.v1] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/30/2013] [Indexed: 12/23/2022] Open
Abstract
Using paired-end RNA sequencing, we have quantified the deep transcriptional changes that occur during differentiation of murine embryonic stem cells into a highly enriched population of glutamatergic cortical neurons. These data provide a detailed and nuanced account of longitudinal changes in the transcriptome during neurogenesis and neuronal maturation, starting from mouse embryonic stem cells and progressing through neuroepithelial stem cell induction, radial glial cell formation, neurogenesis, neuronal maturation and cortical patterning. Understanding the transcriptional mechanisms underlying the differentiation of stem cells into mature, glutamatergic neurons of cortical identity has myriad applications, including the elucidation of mechanisms of cortical patterning; identification of neurogenic processes; modeling of disease states; detailing of the host cell response to neurotoxic stimuli; and determination of potential therapeutic targets. In future work we anticipate correlating changes in longitudinal gene expression to other cell parameters, including neuronal function as well as characterizations of the proteome and metabolome. In this data article, we describe the methods used to produce the data and present the raw sequence read data in FASTQ files, sequencing run statistics and a summary flatfile of raw counts for 22,164 genes across 31 samples, representing 3-5 biological replicates at each timepoint. We propose that this data will be a valuable contribution to diverse research efforts in bioinformatics, stem cell research and developmental neuroscience studies.
Collapse
Affiliation(s)
- Kyle S Hubbard
- United States Army, Medical Research Institute of Chemical Defense, MD, 21010, USA
| | - Ian M Gut
- United States Army, Medical Research Institute of Chemical Defense, MD, 21010, USA
| | - Megan E Lyman
- United States Army, Medical Research Institute of Chemical Defense, MD, 21010, USA
| | - Patrick M McNutt
- United States Army, Medical Research Institute of Chemical Defense, MD, 21010, USA
| |
Collapse
|
49
|
Xu H, Yang Y, Tang X, Zhao M, Liang F, Xu P, Hou B, Xing Y, Bao X, Fan X. Bergmann glia function in granule cell migration during cerebellum development. Mol Neurobiol 2013; 47:833-44. [PMID: 23329344 DOI: 10.1007/s12035-013-8405-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 01/07/2013] [Indexed: 12/17/2022]
Abstract
Granule cell migration influences the laminar structure of the cerebellum and thereby affects cerebellum function. Bergmann glia are derived from radial glial cells and aid in granule cell radial migration by providing a scaffold for migration and by mediating interactions between Bergmann glia and granule cells. In this review, we summarize Bergmann glia characteristics and the mechanisms underlying the effect of Bergmann glia on the radial migration of granule neurons in the cerebellum. Furthermore, we will focus our discussion on the important factors involved in glia-mediated radial migration so that we may elucidate the possible mechanistic pathways used by Bergmann glia to influence granule cell migration during cerebellum development.
Collapse
Affiliation(s)
- Haiwei Xu
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University, Chongqing 400038, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Xu H, Yang Y, Tang X, Zhao M, Liang F, Xu P, Hou B, Xing Y, Bao X, Fan X. Bergmann glia function in granule cell migration during cerebellum development. Mol Neurobiol 2013. [PMID: 23329344 DOI: 10.1007/s12035‐013‐8405‐y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Granule cell migration influences the laminar structure of the cerebellum and thereby affects cerebellum function. Bergmann glia are derived from radial glial cells and aid in granule cell radial migration by providing a scaffold for migration and by mediating interactions between Bergmann glia and granule cells. In this review, we summarize Bergmann glia characteristics and the mechanisms underlying the effect of Bergmann glia on the radial migration of granule neurons in the cerebellum. Furthermore, we will focus our discussion on the important factors involved in glia-mediated radial migration so that we may elucidate the possible mechanistic pathways used by Bergmann glia to influence granule cell migration during cerebellum development.
Collapse
Affiliation(s)
- Haiwei Xu
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University, Chongqing 400038, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|