251
|
Neurog2 and Ascl1 together regulate a postmitotic derepression circuit to govern laminar fate specification in the murine neocortex. Proc Natl Acad Sci U S A 2017; 114:E4934-E4943. [PMID: 28584103 DOI: 10.1073/pnas.1701495114] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
A derepression mode of cell-fate specification involving the transcriptional repressors Tbr1, Fezf2, Satb2, and Ctip2 operates in neocortical projection neurons to specify six layer identities in sequence. Less well understood is how laminar fate transitions are regulated in cortical progenitors. The proneural genes Neurog2 and Ascl1 cooperate in progenitors to control the temporal switch from neurogenesis to gliogenesis. Here we asked whether these proneural genes also regulate laminar fate transitions. Several defects were observed in the derepression circuit in Neurog2-/-;Ascl1-/- mutants: an inability to repress expression of Tbr1 (a deep layer VI marker) during upper-layer neurogenesis, a loss of Fezf2+/Ctip2+ layer V neurons, and precocious differentiation of normally late-born, Satb2+ layer II-IV neurons. Conversely, in stable gain-of-function transgenics, Neurog2 promoted differentiative divisions and extended the period of Tbr1+/Ctip2+ deep-layer neurogenesis while reducing Satb2+ upper-layer neurogenesis. Similarly, acute misexpression of Neurog2 in early cortical progenitors promoted Tbr1 expression, whereas both Neurog2 and Ascl1 induced Ctip2. However, Neurog2 was unable to influence the derepression circuit when misexpressed in late cortical progenitors, and Ascl1 repressed only Satb2. Nevertheless, neurons derived from late misexpression of Neurog2 and, to a lesser extent, Ascl1, extended aberrant subcortical axon projections characteristic of early-born neurons. Finally, Neurog2 and Ascl1 altered the expression of Ikaros and Foxg1, known temporal regulators. Proneural genes thus act in a context-dependent fashion as early determinants, promoting deep-layer neurogenesis in early cortical progenitors via input into the derepression circuit while also influencing other temporal regulators.
Collapse
|
252
|
Powell SK, Gregory J, Akbarian S, Brennand KJ. Application of CRISPR/Cas9 to the study of brain development and neuropsychiatric disease. Mol Cell Neurosci 2017; 82:157-166. [PMID: 28549865 DOI: 10.1016/j.mcn.2017.05.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 05/22/2017] [Indexed: 12/18/2022] Open
Abstract
CRISPR/Cas9 technology has transformed our ability to manipulate the genome and epigenome, from efficient genomic editing to targeted localization of effectors to specific loci. Through the manipulation of DNA- and histone-modifying enzyme activities, activation or repression of gene expression, and targeting of transcriptional regulators, the role of gene-regulatory and epigenetic pathways in basic biology and disease processes can be directly queried. Here, we discuss emerging CRISPR-based methodologies, with specific consideration of neurobiological applications of human induced pluripotent stem cell (hiPSC)-based models.
Collapse
Affiliation(s)
- S K Powell
- Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - J Gregory
- Instructional Technology Group, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - S Akbarian
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - K J Brennand
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States.
| |
Collapse
|
253
|
Li Y, You QL, Zhang SR, Huang WY, Zou WJ, Jie W, Li SJ, Liu JH, Lv CY, Cong J, Hu YY, Gao TM, Li JM. Satb2 Ablation Impairs Hippocampus-Based Long-Term Spatial Memory and Short-Term Working Memory and Immediate Early Genes (IEGs)-Mediated Hippocampal Synaptic Plasticity. Mol Neurobiol 2017:10.1007/s12035-017-0531-5. [PMID: 28421537 DOI: 10.1007/s12035-017-0531-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 04/06/2017] [Indexed: 10/19/2022]
Abstract
Special AT-rich sequence-binding protein 2 (Satb2) is a protein binding to the matrix attachment regions of DNA and important for gene regulation. Patients with SATB2 mutation usually suffer moderate to severe mental retardation. However, the mechanisms for the defects of intellectual activities in patients with SATB2 mutation are largely unclear. Here we established the heterozygous Satb2 mutant mice and Satb2 conditional knockout mice to mimic the patients with SATB2 mutation and figured out the role of Satb2 in mental activities. We found that the spatial memory and working memory were significantly damaged in the heterozygous Satb2 mutant mice, early postnatal Satb2-deficient mice (CaMKIIα-Cre+Satb2fl/fl mice), and adult Satb2 ablation mice (Satb2fl/fl mice injected with CaMKIIα-Cre virus). Functionally, late phase long-term potentiation (L-LTP) in these Satb2 mutant mice was greatly impaired. Morphologically, in CA1 neurons of CaMKIIα-Cre+Satb2fl/fl mice, we found decreased spine density of the basal dendrites and less branches of apical dendrites that extended into lacunar molecular layer. Mechanistically, expression levels of immediate early genes (IEGs) including Fos, FosB, and Egr1 were significantly decreased after Satb2 deletion. And, Satb2 could regulate expression of FosB by binding to the promoter of FosB directly. In general, our study uncovers that Satb2 plays an important role in spatial memory and working memory by regulating IEGs-mediated hippocampal synaptic plasticity.
Collapse
Affiliation(s)
- Ying Li
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Guangzhou, 510120, People's Republic of China
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
- Department of Pathology, Chancheng Central Hospital, Foshan, 528031, People's Republic of China
| | - Qiang-Long You
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Sheng-Rong Zhang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Wei-Yuan Huang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Wen-Jun Zou
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Wei Jie
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Shu-Ji Li
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Ji-Hong Liu
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Chuang-Ye Lv
- College of Clinical Medical, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Jin Cong
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Yu-Ying Hu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Tian-Ming Gao
- State Key Laboratory of Organ Failure Research, Key Laboratory of Psychiatric Disorders of Guangdong Province, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| | - Jian-Ming Li
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Guangzhou, 510120, People's Republic of China.
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China.
- Department of Pathology, Soochow University Medical School, Suzhou, 215123, People's Republic of China.
| |
Collapse
|
254
|
Barford K, Yap CC, Dwyer ND, Winckler B. The related neuronal endosomal proteins NEEP21 (Nsg1) and P19 (Nsg2) have divergent expression profiles in vivo. J Comp Neurol 2017; 525:1861-1878. [PMID: 28299779 DOI: 10.1002/cne.24168] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 10/28/2016] [Accepted: 11/18/2016] [Indexed: 01/23/2023]
Abstract
Endosomal maturation and transport constitutes a complex trafficking system present in all cell types. Neurons have adapted their endosomal system to meet their unique and complex needs. These adaptations include repurposing existing proteins to diversify endocytosis and trafficking, as well as preferential expression of certain regulators more highly in neurons than other cell types. These neuronal regulators include the family of Neuron-Specific Gene family members (Nsg), NEEP21 (Nsg1), and P19 (Nsg2). NEEP21/Nsg1 plays a role in the trafficking of multiple receptors, including the cell adhesion molecule L1/NgCAM, the neurotransmitter receptor GluA2, and β-APP. Recently, we showed that NEEP2/Nsg1 and P19/Nsg2 are not expressed in all neuronal cell types in vitro. However, it is not known where and when NEEP21/Nsg1 and P19/Nsg2 are expressed in vivo, and whether both proteins are always coexpressed. Here, we show that NEEP21/Nsg1 and P19/Nsg2 are present in both overlapping and distinct cell populations in the hippocampus, neocortex, and cerebellum during development. NEEP21/Nsg1 and P19/Nsg2 levels are highest during embryonic development, and expression persists in the juvenile mouse brain. In particular, a subset of layer V cortical neurons retains relatively high expression of both NEEP21/Nsg1 and P19/Nsg2 at postnatal day 16 as well as in the CA1-3 regions of the hippocampus. In the cerebellum, NEEP21/Nsg1 expression becomes largely restricted to Purkinje neurons in adulthood whereas P19/Nsg2 expression strikingly disappears from the cerebellum with age. This divergent and restricted expression likely reflects differential needs for this class of trafficking regulators in different neurons during different stages of maturation.
Collapse
Affiliation(s)
- Kelly Barford
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| | - Chan Choo Yap
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| | - Noelle D Dwyer
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| | - Bettina Winckler
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
255
|
Disruptions in asymmetric centrosome inheritance and WDR62-Aurora kinase B interactions in primary microcephaly. Sci Rep 2017; 7:43708. [PMID: 28272472 PMCID: PMC5341122 DOI: 10.1038/srep43708] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 01/26/2017] [Indexed: 12/11/2022] Open
Abstract
Recessive mutations in WD repeat domain 62 (WDR62) cause microcephaly and a wide spectrum of severe brain malformations. Disruption of the mouse ortholog results in microcephaly underlain by reduced proliferation of neocortical progenitors during late neurogenesis, abnormalities in asymmetric centrosome inheritance leading to neuronal migration delays, and altered neuronal differentiation. Spindle pole localization of WDR62 and mitotic progression are defective in patient-derived fibroblasts, which, similar to mouse neocortical progenitors, transiently arrest at prometaphase. Expression of WDR62 is closely correlated with components of the chromosome passenger complex (CPC), a key regulator of mitosis. Wild type WDR62, but not disease-associated mutant forms, interacts with the CPC core enzyme Aurora kinase B and staining of CPC components at centromeres is altered in patient-derived fibroblasts. Our findings demonstrate critical and diverse functions of WDR62 in neocortical development and provide insight into the mechanisms by which its disruption leads to a plethora of structural abnormalities.
Collapse
|
256
|
Haushalter C, Schuhbaur B, Dollé P, Rhinn M. Meningeal retinoic acid contributes to neocortical lamination and radial migration during mouse brain development. Biol Open 2017; 6:148-160. [PMID: 28011626 PMCID: PMC5312094 DOI: 10.1242/bio.021063] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Retinoic acid (RA) is a diffusible molecule involved in early forebrain patterning. Its later production in the meninges by the retinaldehyde dehydrogenase RALDH2 coincides with the time of cortical neuron generation. A function of RA in this process has not been adressed directly as Raldh2−/− mouse mutants are embryonic lethal. Here, we used a conditional genetic strategy to inactivate Raldh2 just prior to onset of its expression in the developing meninges. This inactivation does not affect the formation of the cortical progenitor populations, their rate of division, or timing of differentiation. However, migration of late-born cortical neurons is delayed, with neurons stalling in the intermediate zone and exhibiting an abnormal multipolar morphology. This suggests that RA controls the multipolar-to-bipolar transition that occurs in the intermediate zone and allows neurons to start locomotion in the cortical plate. Our work also shows a role for RA in cortical lamination, as deep layers are expanded and a subset of layer IV neurons are not formed in the Raldh2-ablated mutants. These data demonstrate that meninges are a source of extrinsic signals important for cortical development. Summary: Involvement of the signalling molecule retinoic acid in neurogenesis of the developing cerebral cortex is shown through targeted deletion of its synthesizing enzyme.
Collapse
Affiliation(s)
- Carole Haushalter
- Development and Stem Cells Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67404, France.,Centre National de la Recherche Scientifique, UMR 7104, Illkirch 67404, France.,Institut National de la Santé et de la Recherche Médicale, U 964, Illkirch 67404, France.,Université de Strasbourg, Illkirch 67404, France
| | - Brigitte Schuhbaur
- Development and Stem Cells Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67404, France.,Centre National de la Recherche Scientifique, UMR 7104, Illkirch 67404, France.,Institut National de la Santé et de la Recherche Médicale, U 964, Illkirch 67404, France.,Université de Strasbourg, Illkirch 67404, France
| | - Pascal Dollé
- Development and Stem Cells Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67404, France .,Centre National de la Recherche Scientifique, UMR 7104, Illkirch 67404, France.,Institut National de la Santé et de la Recherche Médicale, U 964, Illkirch 67404, France.,Université de Strasbourg, Illkirch 67404, France
| | - Muriel Rhinn
- Development and Stem Cells Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch 67404, France .,Centre National de la Recherche Scientifique, UMR 7104, Illkirch 67404, France.,Institut National de la Santé et de la Recherche Médicale, U 964, Illkirch 67404, France.,Université de Strasbourg, Illkirch 67404, France
| |
Collapse
|
257
|
Cellular transformation of human mammary epithelial cells by SATB2. Stem Cell Res 2017; 19:139-147. [PMID: 28167342 DOI: 10.1016/j.scr.2017.01.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 01/04/2017] [Accepted: 01/30/2017] [Indexed: 12/19/2022] Open
Abstract
Breast tumors are heterogeneous and carry a small population of progenitor cells that can produce various subtypes of breast cancer. SATB2 (special AT-rich binding protein-2) is a newly identified transcription factor and epigenetic regulator. It is highly expressed in embryonic stem cells, but not in adult tissues, and regulates pluripotency-maintaining factors. However, the molecular mechanisms by which SATB2 induces transformation of human mammary epithelial cells (HMECs) leading to malignant phenotype are unknown. The main goal of this paper is to examine the molecular mechanisms by which SATB2 induces cellular transformation of HMECs into cells that are capable of self-renewal. SATB2-transformed HMECs gain the phenotype of breast progenitor cells by expressing markers of stem cells, pluripotency-maintaining factor, and epithelial to mesenchymal transition. SATB2 is highly expressed in human breast cancer cell lines, primary mammary tissues and cancer stem cells (CSCs), but not in HMECs and normal breast tissues. Chromatin Immunoprecipitation assays demonstrate that SATB2 can directly bind to promoters of Bcl-2, c-Myc, Nanog, Klf4, and XIAP, suggesting a role of SATB2 in regulation of pluripotency, cell survival and proliferation. Furthermore, inhibition of SATB2 by shRNA in breast cancer cell lines and CSCs attenuates cell proliferation and EMT phenotype. Our results suggest that SATB2 induces dedifferentiation/transformation of mature HMECs into progenitor-like cells.
Collapse
|
258
|
Cipriani S, Journiac N, Nardelli J, Verney C, Delezoide AL, Guimiot F, Gressens P, Adle-Biassette H. Dynamic Expression Patterns of Progenitor and Neuron Layer Markers in the Developing Human Dentate Gyrus and Fimbria. Cereb Cortex 2017; 27:358-372. [PMID: 26443441 PMCID: PMC5894254 DOI: 10.1093/cercor/bhv223] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The molecular mechanisms that orchestrate the development of the human dentate gyrus are not known. In this study, we characterized the formation of human dentate and fimbrial progenitors and postmitotic neurons from 9 gestational weeks (GW9) to GW25. PAX6+ progenitor cells remained proliferative until GW16 in the dentate ventricular zone. By GW11, the secondary dentate matrix had developed in the intermediate zone, surrounding the dentate anlage and streaming toward the subpial layer. This secondary matrix contained proliferating PAX6+ and/or TBR2+ progenitors. In parallel, SOX2+ and PAX6+ fimbrial cells were detected approaching the dentate anlage, representing a possible source of extra-dentate progenitors. By GW16, when the granule cell layer could be delineated, a hilar matrix containing PAX6+ and some TBR2+ progenitors had become identifiable. By GW25, when the 2 limbs of the granule cell layer had formed, the secondary dentate matrix was reduced to a pool of progenitors at the fimbrio-dentate junction. Although human dentate development recapitulates key steps previously described in rodents, differences seemed to emerge in neuron layer markers expression. Further studies are necessary to better elucidate their role in dentate formation and connectivity.
Collapse
Affiliation(s)
- Sara Cipriani
- INSERM UMR 1141, Hôpital Robert-Debré, Paris, France
- Faculté de Médecine Denis Diderot, Université Paris 7, Paris, France
| | - Nathalie Journiac
- INSERM UMR 1141, Hôpital Robert-Debré, Paris, France
- Faculté de Médecine Denis Diderot, Université Paris 7, Paris, France
| | - Jeannette Nardelli
- INSERM UMR 1141, Hôpital Robert-Debré, Paris, France
- Faculté de Médecine Denis Diderot, Université Paris 7, Paris, France
| | - Catherine Verney
- INSERM UMR 1141, Hôpital Robert-Debré, Paris, France
- Faculté de Médecine Denis Diderot, Université Paris 7, Paris, France
| | - Anne-Lise Delezoide
- INSERM UMR 1141, Hôpital Robert-Debré, Paris, France
- Faculté de Médecine Denis Diderot, Université Paris 7, Paris, France
- Service de Biologie du Développement, Hôpital Robert-Debré, APHP, Paris, France
| | - Fabien Guimiot
- INSERM UMR 1141, Hôpital Robert-Debré, Paris, France
- Faculté de Médecine Denis Diderot, Université Paris 7, Paris, France
- Service de Biologie du Développement, Hôpital Robert-Debré, APHP, Paris, France
| | - Pierre Gressens
- INSERM UMR 1141, Hôpital Robert-Debré, Paris, France
- Faculté de Médecine Denis Diderot, Université Paris 7, Paris, France
| | - Homa Adle-Biassette
- INSERM UMR 1141, Hôpital Robert-Debré, Paris, France
- Faculté de Médecine Denis Diderot, Université Paris 7, Paris, France
- Service d'Anatomie et de Cytologie Pathologiques, Hôpital Lariboisère, APHP, Paris, France
| |
Collapse
|
259
|
Jaitner C, Reddy C, Abentung A, Whittle N, Rieder D, Delekate A, Korte M, Jain G, Fischer A, Sananbenesi F, Cera I, Singewald N, Dechant G, Apostolova G. Satb2 determines miRNA expression and long-term memory in the adult central nervous system. eLife 2016; 5. [PMID: 27897969 PMCID: PMC5207769 DOI: 10.7554/elife.17361] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 11/28/2016] [Indexed: 01/09/2023] Open
Abstract
SATB2 is a risk locus for schizophrenia and encodes a DNA-binding protein that regulates higher-order chromatin configuration. In the adult brain Satb2 is almost exclusively expressed in pyramidal neurons of two brain regions important for memory formation, the cerebral cortex and the CA1-hippocampal field. Here we show that Satb2 is required for key hippocampal functions since deletion of Satb2 from the adult mouse forebrain prevents the stabilization of synaptic long-term potentiation and markedly impairs long-term fear and object discrimination memory. At the molecular level, we find that synaptic activity and BDNF up-regulate Satb2, which itself binds to the promoters of coding and non-coding genes. Satb2 controls the hippocampal levels of a large cohort of miRNAs, many of which are implicated in synaptic plasticity and memory formation. Together, our findings demonstrate that Satb2 is critically involved in long-term plasticity processes in the adult forebrain that underlie the consolidation and stabilization of context-linked memory. DOI:http://dx.doi.org/10.7554/eLife.17361.001
Collapse
Affiliation(s)
- Clemens Jaitner
- Institute for Neuroscience, Medical University of Innsbruck, Innsbruck, Austria
| | - Chethan Reddy
- Institute for Neuroscience, Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas Abentung
- Institute for Neuroscience, Medical University of Innsbruck, Innsbruck, Austria
| | - Nigel Whittle
- Department of Pharmacology and Toxicology, University of Innsbruck, Innsbruck, Austria
| | - Dietmar Rieder
- Division of Bioinformatics, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Andrea Delekate
- Zoological Institute, Technical University Braunschweig, Braunschweig, Germany
| | - Martin Korte
- Zoological Institute, Technical University Braunschweig, Braunschweig, Germany.,AG Neuroinflammation and Neurodegeneration (NIND), Braunschweig, Germany
| | - Gaurav Jain
- Research Group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Göttingen, Germany.,Research Group for Complex Neurodegenerative Disorders, German Center for Neurodegenerative Diseases, Göttingen, Germany
| | - Andre Fischer
- Research Group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Göttingen, Germany.,Department of Psychiatry and Psychotherapy, University Medical Center, German Center for Neurodegenerative Diseases, Göttingen, Germany
| | - Farahnaz Sananbenesi
- Research Group for Complex Neurodegenerative Disorders, German Center for Neurodegenerative Diseases, Göttingen, Germany
| | - Isabella Cera
- Institute for Neuroscience, Medical University of Innsbruck, Innsbruck, Austria
| | - Nicolas Singewald
- Department of Pharmacology and Toxicology, University of Innsbruck, Innsbruck, Austria
| | - Georg Dechant
- Institute for Neuroscience, Medical University of Innsbruck, Innsbruck, Austria
| | - Galina Apostolova
- Institute for Neuroscience, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
260
|
Caubit X, Gubellini P, Andrieux J, Roubertoux PL, Metwaly M, Jacq B, Fatmi A, Had-Aissouni L, Kwan KY, Salin P, Carlier M, Liedén A, Rudd E, Shinawi M, Vincent-Delorme C, Cuisset JM, Lemaitre MP, Abderrehamane F, Duban B, Lemaitre JF, Woolf AS, Bockenhauer D, Severac D, Dubois E, Zhu Y, Sestan N, Garratt AN, Lydia Kerkerian-Le G, Fasano L. TSHZ3 deletion causes an autism syndrome and defects in cortical projection neurons. Nat Genet 2016; 48:1359-1369. [PMID: 27668656 PMCID: PMC5083212 DOI: 10.1038/ng.3681] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/29/2016] [Indexed: 12/12/2022]
Abstract
TSHZ3, which encodes a zinc-finger transcription factor, was recently positioned as a hub gene in a module of the genes with the highest expression in the developing human neocortex, but its functions remained unknown. Here we identify TSHZ3 as the critical region for a syndrome associated with heterozygous deletions at 19q12-q13.11, which includes autism spectrum disorder (ASD). In Tshz3-null mice, differentially expressed genes include layer-specific markers of cerebral cortical projection neurons (CPNs), and the human orthologs of these genes are strongly associated with ASD. Furthermore, mice heterozygous for Tshz3 show functional changes at synapses established by CPNs and exhibit core ASD-like behavioral abnormalities. These findings highlight essential roles for Tshz3 in CPN development and function, whose alterations can account for ASD in the newly defined TSHZ3 deletion syndrome.
Collapse
Affiliation(s)
| | | | - Joris Andrieux
- Institut de génétique médicale, Hôpital Jeanne de Flandre, CHRU Lille, France
| | | | | | - Bernard Jacq
- Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Ahmed Fatmi
- Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | | | - Kenneth Y Kwan
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Molecular & Behavioral Neuroscience Institute (MBNI), Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Pascal Salin
- Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | | | - Agne Liedén
- Karolinska University Hospital Solna, Clinical Genetics Unit, Stockholm, Sweden
| | - Eva Rudd
- Karolinska University Hospital Solna, Clinical Genetics Unit, Stockholm, Sweden
| | - Marwan Shinawi
- Department of Pediatrics, Division of Genetics and Genomic Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | | | | | - Bénédicte Duban
- Centre de cytogénétique, Hôpital Saint Vincent de Paul, GHICL, UCL, Lille, France
| | | | - Adrian S Woolf
- Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Manchester Academic Health Science Centre and the Royal Manchester Children's and St Mary's Hospitals, Manchester, UK
| | | | - Dany Severac
- MGX-Montpellier GenomiX, c/o Institut de Génomique Fonctionnelle, Montpellier, France
| | - Emeric Dubois
- MGX-Montpellier GenomiX, c/o Institut de Génomique Fonctionnelle, Montpellier, France
| | - Ying Zhu
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Nenad Sestan
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Alistair N Garratt
- Institute of Cell Biology and Neurobiology, Center for Anatomy, Charité University Hospital Berlin, Berlin, Germany
| | | | | |
Collapse
|
261
|
Zarate YA, Fish JL. SATB2-associated syndrome: Mechanisms, phenotype, and practical recommendations. Am J Med Genet A 2016; 173:327-337. [PMID: 27774744 PMCID: PMC5297989 DOI: 10.1002/ajmg.a.38022] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 09/29/2016] [Indexed: 12/11/2022]
Abstract
The SATB2‐associated syndrome is a recently described syndrome characterized by developmental delay/intellectual disability with absent or limited speech development, craniofacial abnormalities, behavioral problems, dysmorphic features, and palatal and dental abnormalities. Alterations of the SATB2 gene can result from a variety of different mechanisms that include contiguous deletions, intragenic deletions and duplications, translocations with secondary gene disruption, and point mutations. The multisystemic nature of this syndrome demands a multisystemic approach and we propose evaluation and management guidelines. The SATB2‐associated syndrome registry has now been started and that will allow gathering further clinical information and refining the provided surveillance recommendations. © 2016 The Authors. American Journal of Medical Genetics Part A Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yuri A Zarate
- Section of Genetics and Metabolism, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Jennifer L Fish
- Department of Biological Sciences, University of Massachusetts Lowell, Lowell, Massachusetts
| |
Collapse
|
262
|
Welniarz Q, Dusart I, Roze E. The corticospinal tract: Evolution, development, and human disorders. Dev Neurobiol 2016; 77:810-829. [PMID: 27706924 DOI: 10.1002/dneu.22455] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 09/18/2016] [Accepted: 09/19/2016] [Indexed: 01/22/2023]
Abstract
The corticospinal tract (CST) plays a major role in cortical control of spinal cord activity. In particular, it is the principal motor pathway for voluntary movements. Here, we discuss: (i) the anatomic evolution and development of the CST across mammalian species, focusing on its role in motor functions; (ii) the molecular mechanisms regulating corticospinal tract formation and guidance during mouse development; and (iii) human disorders associated with abnormal CST development. A comparison of CST anatomy and development across mammalian species first highlights important similarities. In particular, most CST axons cross the anatomical midline at the junction between the brainstem and spinal cord, forming the pyramidal decussation. Reorganization of the pattern of CST projections to the spinal cord during evolution led to improved motor skills. Studies of the molecular mechanisms involved in CST formation and guidance in mice have identified several factors that act synergistically to ensure proper formation of the CST at each step of development. Human CST developmental disorders can result in a reduction of the CST, or in guidance defects associated with abnormal CST anatomy. These latter disorders result in altered midline crossing at the pyramidal decussation or in the spinal cord, but spare the rest of the CST. Careful appraisal of clinical manifestations associated with CST malformations highlights the critical role of the CST in the lateralization of motor control. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 810-829, 2017.
Collapse
Affiliation(s)
- Quentin Welniarz
- Institut du Cerveau et de la Moelle épinière, Sorbonne Universités, UPMC Univ Paris 06, INSERM U 1127, CNRS UMR 7225, F-75013, Paris, France.,Institut de Biologie Paris Seine, Neuroscience Paris Seine, Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, F-75005, Paris, France
| | - Isabelle Dusart
- Institut de Biologie Paris Seine, Neuroscience Paris Seine, Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, F-75005, Paris, France
| | - Emmanuel Roze
- Institut du Cerveau et de la Moelle épinière, Sorbonne Universités, UPMC Univ Paris 06, INSERM U 1127, CNRS UMR 7225, F-75013, Paris, France.,Département des Maladies du Système Nerveux, AP-HP, Hôpital de la Salpêtrière, Paris, France
| |
Collapse
|
263
|
Li Y, Liu YH, Hu YY, Chen L, Li JM. Special AT-rich sequence-binding protein 2 acts as a negative regulator of stemness in colorectal cancer cells. World J Gastroenterol 2016; 22:8528-8539. [PMID: 27784965 PMCID: PMC5064034 DOI: 10.3748/wjg.v22.i38.8528] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 07/29/2016] [Accepted: 08/19/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To find the mechanisms by which special AT-rich sequence-binding protein 2 (SATB2) influences colorectal cancer (CRC) metastasis.
METHODS Cell growth assay, colony-forming assay, cell adhesion assay and cell migration assay were used to evaluate the biological characteristics of CRC cells with gain or loss of SATB2. Sphere formation assay was used to detect the self-renewal ability of CRC cells. The mRNA expression of stem cell markers in CRC cells with upregulated or downregulated SATB2 expression was detected by quantitative real-time polymerase chain reaction. Chromatin immunoprecipitation (ChIP) was used to verify the binding loci of SATB2 on genomic sequences of stem cell markers. The Cancer Genome Atlas (TCGA) database and our clinical samples were analyzed to find the correlation between SATB2 and some key stem cell markers.
RESULTS Downregulation of SATB2 led to an aggressive phenotype in SW480 and DLD-1 cells, which was characterized by increased migration and invasion abilities. Overexpression of SATB2 suppressed the migration and invasion abilities in SW480 and SW620 cells. Using sequential sphere formation assay to detect the self-renewal abilities of CRC cells, we found more secondary sphere formation but not primary sphere formation in SW480 and DLD-1 cells after SATB2 expression was knocked down. Moreover, most markers for stem cells such as CD133, CD44, AXIN2, MEIS2 and NANOG were increased in cells with SATB2 knockdown and decreased in cells with SATB2 overexpression. ChIP assay showed that SATB2 bound to regulatory elements of CD133, CD44, MEIS2 and AXIN2 genes. Using TCGA database and our clinical samples, we found that SATB2 was correlated with some key stem cell markers including CD44 and CD24 in clinical tissues of CRC patients.
CONCLUSION SATB2 can directly bind to the regulatory elements in the genetic loci of several stem cell markers and consequently inhibit the progression of CRC by negatively regulating stemness of CRC cells.
Collapse
|
264
|
Fededa JP, Esk C, Mierzwa B, Stanyte R, Yuan S, Zheng H, Ebnet K, Yan W, Knoblich JA, Gerlich DW. MicroRNA-34/449 controls mitotic spindle orientation during mammalian cortex development. EMBO J 2016; 35:2386-2398. [PMID: 27707753 PMCID: PMC5109238 DOI: 10.15252/embj.201694056] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 08/18/2016] [Accepted: 09/06/2016] [Indexed: 01/07/2023] Open
Abstract
Correct orientation of the mitotic spindle determines the plane of cellular cleavage and is crucial for organ development. In the developing cerebral cortex, spindle orientation defects result in severe neurodevelopmental disorders, but the precise mechanisms that control this important event are not fully understood. Here, we use a combination of high-content screening and mouse genetics to identify the miR-34/449 family as key regulators of mitotic spindle orientation in the developing cerebral cortex. By screening through all cortically expressed miRNAs in HeLa cells, we show that several members of the miR-34/449 family control mitotic duration and spindle rotation. Analysis of miR-34/449 knockout (KO) mouse embryos demonstrates significant spindle misorientation phenotypes in cortical progenitors, resulting in an excess of radial glia cells at the expense of intermediate progenitors and a significant delay in neurogenesis. We identify the junction adhesion molecule-A (JAM-A) as a key target for miR-34/449 in the developing cortex that might be responsible for those defects. Our data indicate that miRNA-dependent regulation of mitotic spindle orientation is crucial for cell fate specification during mammalian neurogenesis.
Collapse
Affiliation(s)
- Juan Pablo Fededa
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA) Vienna Biocenter (VBC), Vienna, Austria
| | - Christopher Esk
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA) Vienna Biocenter (VBC), Vienna, Austria
| | - Beata Mierzwa
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA) Vienna Biocenter (VBC), Vienna, Austria
| | - Rugile Stanyte
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA) Vienna Biocenter (VBC), Vienna, Austria
| | - Shuiqiao Yuan
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| | - Huili Zheng
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| | - Klaus Ebnet
- Institute-associated Research Group "Cell Adhesion and Cell Polarity", Institute of Medical Biochemistry, ZMBE, Münster, Germany
| | - Wei Yan
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| | - Juergen A Knoblich
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA) Vienna Biocenter (VBC), Vienna, Austria
| | - Daniel W Gerlich
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA) Vienna Biocenter (VBC), Vienna, Austria
| |
Collapse
|
265
|
Arlotta P, Hobert O. Homeotic Transformations of Neuronal Cell Identities. Trends Neurosci 2016; 38:751-762. [PMID: 26596501 DOI: 10.1016/j.tins.2015.10.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 10/13/2015] [Accepted: 10/18/2015] [Indexed: 11/20/2022]
Abstract
Homeosis is classically defined as the transformation of one body part into something that resembles another body part. We propose here to broaden the concept of homeosis to the many neuronal cell identity transformations that have been uncovered over the past few years upon removal of specific regulatory factors in organisms from Caenorhabditis elegans to Drosophila, zebrafish, and mice. The concept of homeosis provides a framework for the evolution of cell type diversity in the brain.
Collapse
Affiliation(s)
- Paola Arlotta
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA.
| | - Oliver Hobert
- Department of Biology and Department of Biochemistry and Molecular Biophysics, Howard Hughes Medical Institute, Columbia University, New York, NY, USA.
| |
Collapse
|
266
|
Chen X, Petit EI, Dobrenis K, Sze JY. Spatiotemporal SERT expression in cortical map development. Neurochem Int 2016; 98:129-37. [PMID: 27282696 PMCID: PMC4969137 DOI: 10.1016/j.neuint.2016.05.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 05/03/2016] [Accepted: 05/17/2016] [Indexed: 11/21/2022]
Abstract
The cerebral cortex is organized into morphologically distinct areas that provide biological frameworks underlying perception, cognition, and behavior. Profiling mouse and human cortical transcriptomes have revealed temporal-specific differential gene expression modules in distinct neocortical areas during cortical map establishment. However, the biological roles of spatiotemporal gene expression in cortical patterning and how cortical topographic gene expression is regulated are largely unknown. Here, we characterize temporal- and spatial-defined expression of serotonin (5-HT) transporter (SERT) in glutamatergic neurons during sensory map development in mice. SERT is transiently expressed in glutamatergic thalamic neurons projecting to sensory cortices and in pyramidal neurons in the prefrontal cortex (PFC) and hippocampus (HPC) during the period that lays down the basic functional neural circuits. We previously identified that knockout of SERT in the thalamic neurons blocks 5-HT uptake by their thalamocortical axons, resulting in excessive 5-HT signaling that impairs sensory map architecture. In contrast, here we show that selective SERT knockout in the PFC and HPC neurons does not perturb sensory map patterning. These data suggest that transient SERT expression in specific glutamatergic neurons provides area-specific instructions for cortical map patterning. Hence, genetic and pharmacological manipulations of this SERT function could illuminate the fundamental genetic programming of cortex-specific maps and biological roles of temporal-specific cortical topographic gene expression in normal development and mental disorders.
Collapse
Affiliation(s)
- Xiaoning Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Emilie I Petit
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kostantin Dobrenis
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA; Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ji Ying Sze
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
267
|
Chao MW, Chen CP, Yang YH, Chuang YC, Chu TY, Tseng CY. N-acetylcysteine attenuates lipopolysaccharide-induced impairment in lamination of Ctip2-and Tbr1- expressing cortical neurons in the developing rat fetal brain. Sci Rep 2016; 6:32373. [PMID: 27577752 PMCID: PMC5006028 DOI: 10.1038/srep32373] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 08/03/2016] [Indexed: 02/02/2023] Open
Abstract
Oxidative stress and inflammatory insults are the major instigating events of bacterial intrauterine infection that lead to fetal brain injury. The purpose of this study is to investigate the remedial effects of N-acetyl-cysteine (NAC) for inflammation-caused deficits in brain development. We found that lipopolysaccharide (LPS) induced reactive oxygen species (ROS) production by RAW264.7 cells. Macrophage-conditioned medium caused noticeable cortical cell damage, specifically in cortical neurons. LPS at 25 μg/kg caused more than 75% fetal loss in rats. An increase in fetal cortical thickness was noted in the LPS-treated group. In the enlarged fetal cortex, laminar positioning of the early born cortical cells expressing Tbr1 and Ctip2 was disrupted, with a scattered distribution. The effect was similar, but minor, in later born Satb2-expressing cortical cells. NAC protected against LPS-induced neuron toxicity in vitro and counteracted pregnancy loss and alterations in thickness and lamination of the neocortex in vivo. Fetal loss and abnormal fetal brain development were due to LPS-induced ROS production. NAC is an effective protective agent against LPS-induced damage. This finding highlights the key therapeutic impact of NAC in LPS-caused abnormal neuronal laminar distribution during brain development.
Collapse
Affiliation(s)
- Ming-Wei Chao
- Department of Bioscience Technology, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
| | - Chie-Pein Chen
- Division of High Risk Pregnancy, Mackay Memorial Hospital, Taipei, Taiwan
| | - Yu-Hsiu Yang
- Department of Biomedical Engineering, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
| | - Yu-Chen Chuang
- Department of Biomedical Engineering, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
| | - Tzu-Yun Chu
- Division of High Risk Pregnancy, Mackay Memorial Hospital, Taipei, Taiwan
| | - Chia-Yi Tseng
- Department of Biomedical Engineering, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
- International Master Program of Biomedical Material and Technology, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
- Center for Nano-Technology, Chung Yuan Christian University, Zhongli district, Taoyuan City, Taiwan
| |
Collapse
|
268
|
Hilde KL, Levine AJ, Hinckley CA, Hayashi M, Montgomery JM, Gullo M, Driscoll SP, Grosschedl R, Kohwi Y, Kohwi-Shigematsu T, Pfaff SL. Satb2 Is Required for the Development of a Spinal Exteroceptive Microcircuit that Modulates Limb Position. Neuron 2016; 91:763-776. [PMID: 27478017 DOI: 10.1016/j.neuron.2016.07.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 05/11/2016] [Accepted: 06/07/2016] [Indexed: 12/12/2022]
Abstract
Motor behaviors such as walking or withdrawing the limb from a painful stimulus rely upon integrative multimodal sensory circuitry to generate appropriate muscle activation patterns. Both the cellular components and the molecular mechanisms that instruct the assembly of the spinal sensorimotor system are poorly understood. Here we characterize the connectivity pattern of a sub-population of lamina V inhibitory sensory relay neurons marked during development by the nuclear matrix and DNA binding factor Satb2 (ISR(Satb2)). ISR(Satb2) neurons receive inputs from multiple streams of sensory information and relay their outputs to motor command layers of the spinal cord. Deletion of the Satb2 transcription factor from ISR(Satb2) neurons perturbs their cellular position, molecular profile, and pre- and post-synaptic connectivity. These alterations are accompanied by abnormal limb hyperflexion responses to mechanical and thermal stimuli and during walking. Thus, Satb2 is a genetic determinant that mediates proper circuit development in a core sensory-to-motor spinal network.
Collapse
Affiliation(s)
- Kathryn L Hilde
- Gene Expression Laboratory and the Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines, La Jolla, CA, 92037, USA
| | - Ariel J Levine
- Gene Expression Laboratory and the Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines, La Jolla, CA, 92037, USA
| | - Christopher A Hinckley
- Gene Expression Laboratory and the Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines, La Jolla, CA, 92037, USA
| | - Marito Hayashi
- Gene Expression Laboratory and the Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines, La Jolla, CA, 92037, USA
| | - Jessica M Montgomery
- Gene Expression Laboratory and the Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines, La Jolla, CA, 92037, USA
| | - Miriam Gullo
- Gene Expression Laboratory and the Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines, La Jolla, CA, 92037, USA
| | - Shawn P Driscoll
- Gene Expression Laboratory and the Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines, La Jolla, CA, 92037, USA
| | - Rudolf Grosschedl
- Max Planck Institute of Immunobiology and Epigenetics, Department of Cellular and Molecular Immunology, 79108 Freiburg, Germany
| | - Yoshinori Kohwi
- Department of Orofacial Sciences, University of California, San Francisco, CA 94143, USA
| | | | - Samuel L Pfaff
- Gene Expression Laboratory and the Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines, La Jolla, CA, 92037, USA.
| |
Collapse
|
269
|
Diodato A, Ruinart de Brimont M, Yim YS, Derian N, Perrin S, Pouch J, Klatzmann D, Garel S, Choi GB, Fleischmann A. Molecular signatures of neural connectivity in the olfactory cortex. Nat Commun 2016; 7:12238. [PMID: 27426965 PMCID: PMC4960301 DOI: 10.1038/ncomms12238] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 06/14/2016] [Indexed: 01/01/2023] Open
Abstract
The ability to target subclasses of neurons with defined connectivity is crucial for uncovering neural circuit functions. The olfactory (piriform) cortex is thought to generate odour percepts and memories, and odour information encoded in piriform is routed to target brain areas involved in multimodal sensory integration, cognition and motor control. However, it remains unknown if piriform outputs are spatially organized, and if distinct output channels are delineated by different gene expression patterns. Here we identify genes selectively expressed in different layers of the piriform cortex. Neural tracing experiments reveal that these layer-specific piriform genes mark different subclasses of neurons, which project to distinct target areas. Interestingly, these molecular signatures of connectivity are maintained in reeler mutant mice, in which neural positioning is scrambled. These results reveal that a predictive link between a neuron's molecular identity and connectivity in this cortical circuit is determined independent of its spatial position. The piriform cortex projects to multiple brain regions involved in diverse aspects of olfactory behavior but information about the organization of these outputs is lacking. Here the authors show that piriform neurons exhibit layer specific gene expression patterns that also define distinct projection targets.
Collapse
Affiliation(s)
- Assunta Diodato
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, and CNRS, UMR 7241 and INSERM U1050, F-75005 Paris, France
| | - Marion Ruinart de Brimont
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, and CNRS, UMR 7241 and INSERM U1050, F-75005 Paris, France
| | - Yeong Shin Yim
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Nicolas Derian
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U959, Immunology-Immunopathology-Immunotherapy (I3), and AP-HP, Clinical Investigation Center in Biotherapy, Hôpital Pitié-Salpêtrière, F-75013 Paris, France
| | - Sandrine Perrin
- École Normale Supérieure, Institut de Biologie de l'ENS, Plateforme Génomique, and INSERM U1024, CNRS UMR 8197, F-75005 Paris, France
| | - Juliette Pouch
- École Normale Supérieure, Institut de Biologie de l'ENS, Plateforme Génomique, and INSERM U1024, CNRS UMR 8197, F-75005 Paris, France
| | - David Klatzmann
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U959, Immunology-Immunopathology-Immunotherapy (I3), and AP-HP, Clinical Investigation Center in Biotherapy, Hôpital Pitié-Salpêtrière, F-75013 Paris, France
| | - Sonia Garel
- École Normale Supérieure, Institut de Biologie de l'ENS, and INSERM U1024, CNRS UMR 8197, F-75005 Paris, France
| | - Gloria B Choi
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Alexander Fleischmann
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, and CNRS, UMR 7241 and INSERM U1050, F-75005 Paris, France
| |
Collapse
|
270
|
Larimer P, Spatazza J, Espinosa JS, Tang Y, Kaneko M, Hasenstaub AR, Stryker MP, Alvarez-Buylla A. Caudal Ganglionic Eminence Precursor Transplants Disperse and Integrate as Lineage-Specific Interneurons but Do Not Induce Cortical Plasticity. Cell Rep 2016; 16:1391-1404. [PMID: 27425623 DOI: 10.1016/j.celrep.2016.06.071] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 05/04/2016] [Accepted: 06/16/2016] [Indexed: 10/21/2022] Open
Abstract
The maturation of inhibitory GABAergic cortical circuits regulates experience-dependent plasticity. We recently showed that the heterochronic transplantation of parvalbumin (PV) or somatostatin (SST) interneurons from the medial ganglionic eminence (MGE) reactivates ocular dominance plasticity (ODP) in the postnatal mouse visual cortex. Might other types of interneurons similarly induce cortical plasticity? Here, we establish that caudal ganglionic eminence (CGE)-derived interneurons, when transplanted into the visual cortex of neonatal mice, migrate extensively in the host brain and acquire laminar distribution, marker expression, electrophysiological properties, and visual response properties like those of host CGE interneurons. Although transplants from the anatomical CGE do induce ODP, we found that this plasticity reactivation is mediated by a small fraction of MGE-derived cells contained in the transplant. These findings demonstrate that transplanted CGE cells can successfully engraft into the postnatal mouse brain and confirm the unique role of MGE lineage neurons in the induction of ODP.
Collapse
Affiliation(s)
- Phillip Larimer
- Center for Integrative Neuroscience, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA 94158, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA; Coleman Memorial Laboratory, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Julien Spatazza
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Juan Sebastian Espinosa
- Center for Integrative Neuroscience, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA 94158, USA; Department of Physiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yunshuo Tang
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Megumi Kaneko
- Center for Integrative Neuroscience, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA 94158, USA; Department of Physiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Andrea R Hasenstaub
- Center for Integrative Neuroscience, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA 94158, USA; Coleman Memorial Laboratory, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael P Stryker
- Center for Integrative Neuroscience, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA 94158, USA; Department of Physiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Arturo Alvarez-Buylla
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
271
|
Sahu SK, Fritz A, Tiwari N, Kovacs Z, Pouya A, Wüllner V, Bora P, Schacht T, Baumgart J, Peron S, Berninger B, Tiwari VK, Methner A. TOX3 regulates neural progenitor identity. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:833-40. [DOI: 10.1016/j.bbagrm.2016.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 03/11/2016] [Accepted: 04/07/2016] [Indexed: 01/19/2023]
|
272
|
Ji L, Kim NH, Huh SO, Rhee HJ. Depletion of Inositol Polyphosphate 4-Phosphatase II Suppresses Callosal Axon Formation in the Developing Mice. Mol Cells 2016; 39:501-7. [PMID: 27109423 PMCID: PMC4916402 DOI: 10.14348/molcells.2016.0058] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 03/24/2016] [Accepted: 03/25/2016] [Indexed: 02/02/2023] Open
Abstract
The corpus callosum is a bundle of nerve fibers that connects the two cerebral hemispheres and is essential for coordinated transmission of information between them. Disruption of early stages of callosal development can cause agenesis of the corpus callosum (AgCC), including both complete and partial callosal absence, causing mild to severe cognitive impairment. Despite extensive studies, the etiology of AgCC remains to be clarified due to the complicated mechanism involved in generating AgCC. The biological function of PI3K signaling including phosphatidylinositol-3,4,5-trisphosphate is well established in diverse biochemical processes including axon and dendrite morphogenesis, but the function of the closely related phosphatidylinositol-3,4,-bisphosphate (PI(3,4)P2) signaling, particularly in the nervous system, is largely unknown. Here, we provide the first report on the role of inositol polyphosphate 4-phosphatase II (INPP4B), a PI(3,4)P2 metabolizing 4-phosphatase in the regulation of callosal axon formation. Depleting INPP4B by in utero electroporation suppressed medially directed callosal axon formation. Moreover, depletion of INPP4B significantly attenuated formation of Satb2-positive pyramidal neurons and axon polarization in cortical neurons during cortical development. Taken together, these data suggest that INPP4B plays a role in the regulating callosal axon formation by controlling axon polarization and the Satb2-positive pyramidal neuron population. Dysregulation of INPP4B during cortical development may be implicated in the generation of partial AgCC.
Collapse
Affiliation(s)
- Liting Ji
- Department of Pharmacology, College of Medicine, Hallym University, Chunchon 200-702,
Korea
| | - Nam-Ho Kim
- Department of Pharmacology, College of Medicine, Hallym University, Chunchon 200-702,
Korea
| | - Sung-Oh Huh
- Department of Pharmacology, College of Medicine, Hallym University, Chunchon 200-702,
Korea
- Institute of Natural Medicine, Hallym University, Chunchon 200-702,
Korea
| | - Hae Jin Rhee
- Institute of Natural Medicine, Hallym University, Chunchon 200-702,
Korea
| |
Collapse
|
273
|
Tonchev AB, Tuoc TC, Rosenthal EH, Studer M, Stoykova A. Zbtb20 modulates the sequential generation of neuronal layers in developing cortex. Mol Brain 2016; 9:65. [PMID: 27282384 PMCID: PMC4901408 DOI: 10.1186/s13041-016-0242-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 05/21/2016] [Indexed: 11/25/2022] Open
Abstract
Background During corticogenesis, genetic programs encoded in progenitor cells at different developmental stages and inherited in postmitotic neurons specify distinct layer and area identities. Transcription factor Zbtb20 has been shown to play a role for hippocampal development but whether it is implicated in mammalian neocortical morphogenesis remains unknown. Results Here, we report that during embyogenesis transcription factor Zbtb20 has a dynamic spatio-temporal expression pattern in mitotic cortical progenitors through which it modulates the sequential generation of cortical neuronal layer identities. Zbtb20 knock out mice exhibited enhanced populations of early born L6-L4 neuronal subtypes and a dramatic reduction of the late born L3/L2 neurons. This defect was due to a temporal misbalance in the production of earlier versus later born neurons, leading to a progressive diminishing of the progenitor pool for the generation of L3-L2 neurons. Zbtb20 implements these temporal effects in part by binding to promoter of the orphan nuclear receptor CoupTF1/Nr2f1. In addition to its effects exerted in cortical progenitors, the postmitotic expression of Zbtb20 in L3/L2 neurons starting at birth may contribute to their proper differentiation and migration. Conclusions Our findings reveal Zbtb20 as a novel temporal regulator for the generation of layer-specific neuronal identities. Electronic supplementary material The online version of this article (doi:10.1186/s13041-016-0242-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anton B Tonchev
- Molecular Developmental Neurobiology Laboratory, Max Planck Institute of Biophysical Chemistry, Am Fassberg, 37077, Gottingen, Germany. .,Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), 37075, Göttingen, Germany. .,Department of Anatomy, Histology and Embryology, Medical University-Varna, Varna, Bulgaria.
| | - Tran Cong Tuoc
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), 37075, Göttingen, Germany.,Molecular Neurobiology Group, Institute of Neuroanatomy, University of Goettingen Medical Center, Goettingen, Germany
| | - Eva H Rosenthal
- Molecular Developmental Neurobiology Laboratory, Max Planck Institute of Biophysical Chemistry, Am Fassberg, 37077, Gottingen, Germany
| | - Michèle Studer
- University Nice Sophia Antipolis, iBV, UMR 7277, F-06108, Nice, France.,Inserm, iBV, U1091, F-06108, Nice, France
| | - Anastassia Stoykova
- Molecular Developmental Neurobiology Laboratory, Max Planck Institute of Biophysical Chemistry, Am Fassberg, 37077, Gottingen, Germany. .,Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), 37075, Göttingen, Germany. .,Department of Anatomy, Histology and Embryology, Medical University-Varna, Varna, Bulgaria.
| |
Collapse
|
274
|
Liu J, Wu X, Zhang H, Qiu R, Yoshikawa K, Lu Q. Prospective separation and transcriptome analyses of cortical projection neurons and interneurons based on lineage tracing by Tbr2 (Eomes)-GFP/Dcx-mRFP reporters. Dev Neurobiol 2016; 76:587-99. [PMID: 26248544 PMCID: PMC4744584 DOI: 10.1002/dneu.22332] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 07/29/2015] [Accepted: 07/31/2015] [Indexed: 12/23/2022]
Abstract
In the cerebral cortex, projection neurons and interneurons work coordinately to establish neural networks for normal cortical functions. While the specific mechanisms that control productions of projection neurons and interneurons are beginning to be revealed, a global characterization of the molecular differences between these two neuron types is crucial for a more comprehensive understanding of their developmental specifications and functions. In this study, using lineage tracing power of combining Tbr2(Eomes)-GFP and Dcx-mRFP reporter mice, we prospectively separated intermediate progenitor cell (IPC)-derived neurons (IPNs) from non-IPC-derived neurons (non-IPNs) of the embryonic cerebral cortex. Molecular characterizations revealed that IPNs and non-IPNs were enriched with projection neurons and interneurons, respectively. Expression profiling documented cell-specific genes including differentially expressed transcriptional regulators that might be involved in cellular specifications, for instance, our data found that SOX1 and SOX2, which were known for important functions in neural stem/progenitor cells, continued to be expressed by interneurons but not by projection neurons. Transcriptome analyses of cortical neurons isolated at different stages of neurogenesis revealed distinct temporal patterns of expression of genes involved in early-born or late-born neuron specification. These data present a resource useful for further investigation of the molecular regulations and functions of projection neurons and interneurons.
Collapse
Affiliation(s)
- Jiancheng Liu
- Department of Neurosciences, Beckman Research Institute of City of
Hope, Duarte, CA 91010, USA
| | - Xiwei Wu
- Department of Molecular and Cellular Biology, Beckman Research
Institute of City of Hope, Duarte, CA 91010, USA
| | - Heying Zhang
- Department of Neurosciences, Beckman Research Institute of City of
Hope, Duarte, CA 91010, USA
| | - Runxiang Qiu
- Department of Neurosciences, Beckman Research Institute of City of
Hope, Duarte, CA 91010, USA
| | - Kazuaki Yoshikawa
- Laboratory of Regulation of Neuronal Development, Institute for
Protein Research, Osaka University, Suita, Osaka, Japan
| | - Qiang Lu
- Department of Neurosciences, Beckman Research Institute of City of
Hope, Duarte, CA 91010, USA
| |
Collapse
|
275
|
Amamoto R, Huerta VGL, Takahashi E, Dai G, Grant AK, Fu Z, Arlotta P. Adult axolotls can regenerate original neuronal diversity in response to brain injury. eLife 2016; 5. [PMID: 27156560 PMCID: PMC4861602 DOI: 10.7554/elife.13998] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 04/07/2016] [Indexed: 12/11/2022] Open
Abstract
The axolotl can regenerate multiple organs, including the brain. It remains, however, unclear whether neuronal diversity, intricate tissue architecture, and axonal connectivity can be regenerated; yet, this is critical for recovery of function and a central aim of cell replacement strategies in the mammalian central nervous system. Here, we demonstrate that, upon mechanical injury to the adult pallium, axolotls can regenerate several of the populations of neurons present before injury. Notably, regenerated neurons acquire functional electrophysiological traits and respond appropriately to afferent inputs. Despite the ability to regenerate specific, molecularly-defined neuronal subtypes, we also uncovered previously unappreciated limitations by showing that newborn neurons organize within altered tissue architecture and fail to re-establish the long-distance axonal tracts and circuit physiology present before injury. The data provide a direct demonstration that diverse, electrophysiologically functional neurons can be regenerated in axolotls, but challenge prior assumptions of functional brain repair in regenerative species. DOI:http://dx.doi.org/10.7554/eLife.13998.001 Humans and other mammals have a very limited ability to regenerate new neurons in the brain to replace those that have been injured or damaged. In striking contrast, some animals like fish and salamanders are capable of filling in injured brain regions with new neurons. This is a complex task, as the brain is composed of many different types of neurons that are connected to each other in a highly organized manner across both short and long distances. The extent to which even the most regenerative species can build new brain regions was not known. Understanding any limitations will help to set realistic expectations for the success of potential treatments that aim to replace neurons in mammals. Amamoto et al. found that the brain of the axolotl, a species of salamander, could selectively regenerate the specific types of neurons that were damaged. This finding suggests that the brain is able to somehow sense which types of neurons are injured. The new neurons were able to mature into functional neurons, but they were limited in their ability to reconnect to their original, distant target neurons. More research is now needed to investigate how the axolotl brain recognizes which types of neurons have been damaged. It will also be important to understand which cells respond to the injury to give rise to the new neurons that fill the injury site, and to uncover the molecules that are important for governing this regenerative process. DOI:http://dx.doi.org/10.7554/eLife.13998.002
Collapse
Affiliation(s)
- Ryoji Amamoto
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, United States
| | | | - Emi Takahashi
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, United States
| | - Guangping Dai
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States
| | - Aaron K Grant
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, United States
| | - Zhanyan Fu
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, United States
| | - Paola Arlotta
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, United States.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, United States
| |
Collapse
|
276
|
Ctip1 Regulates the Balance between Specification of Distinct Projection Neuron Subtypes in Deep Cortical Layers. Cell Rep 2016; 15:999-1012. [PMID: 27117402 DOI: 10.1016/j.celrep.2016.03.064] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 02/24/2016] [Accepted: 03/16/2016] [Indexed: 01/11/2023] Open
Abstract
The molecular linkage between neocortical projection neuron subtype and area development, which enables the establishment of functional areas by projection neuron populations appropriate for specific sensory and motor functions, is poorly understood. Here, we report that Ctip1 controls precision of neocortical development by regulating subtype identity in deep-layer projection neurons. Ctip1 is expressed by postmitotic callosal and corticothalamic projection neurons but is excluded over embryonic development from corticospinal motor neurons, which instead express its close relative, Ctip2. Loss of Ctip1 function results in a striking bias in favor of subcerebral projection neuron development in sensory cortex at the expense of corticothalamic and deep-layer callosal development, while misexpression of Ctip1 in vivo represses subcerebral gene expression and projections. As we report in a paired paper, Ctip1 also controls acquisition of sensory area identity. Therefore, Ctip1 couples subtype and area specification, enabling specific functional areas to organize precise ratios of appropriate output projections.
Collapse
|
277
|
Mühlebner A, Iyer AM, van Scheppingen J, Anink JJ, Jansen FE, Veersema TJ, Braun KP, Spliet WGM, van Hecke W, Söylemezoğlu F, Feucht M, Krsek P, Zamecnik J, Bien CG, Polster T, Coras R, Blümcke I, Aronica E. Specific pattern of maturation and differentiation in the formation of cortical tubers in tuberous sclerosis omplex (TSC): evidence from layer-specific marker expression. J Neurodev Disord 2016; 8:9. [PMID: 27042238 PMCID: PMC4818922 DOI: 10.1186/s11689-016-9142-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 03/08/2016] [Indexed: 03/12/2023] Open
Abstract
BACKGROUND Tuberous sclerosis complex (TSC) is a multisystem disorder that results from mutations in the TSC1 or TSC2 genes, leading to constitutive activation of the mammalian target of rapamycin (mTOR) signaling pathway. Cortical tubers represent typical lesions of the central nervous system (CNS) in TSC. The pattern of cortical layering disruption observed in brain tissue of TSC patients is not yet fully understood, and little is known about the origin and phenotype of individual abnormal cell types recognized in tubers. METHODS In the present study, we aimed to characterize dysmorphic neurons (DNs) and giant cells (GCs) of cortical tubers using neocortical layer-specific markers (NeuN, SMI32, Tbr1, Satb2, Cux2, ER81, and RORβ) and to compare the features with the histo-morphologically similar focal cortical dysplasia (FCD) type IIb. We studied a cohort of nine surgically resected cortical tubers, five FCD type IIb, and four control samples using immunohistochemistry and in situ hybridization. RESULTS Cortical tuber displayed a prominent cell loss in all cortical layers. Moreover, we observed altered proportions of layer-specific markers within the dysplastic region. DNs, in both tubers and FCD type IIb, were found positive for different cortical layer markers, regardless of their laminar location, and their immunophenotype resembles that of cortical projection neurons. CONCLUSIONS These findings demonstrate that, similar to FCD type IIb, cortical layering is markedly disturbed in cortical tubers of TSC patients. Distribution of these disturbances is comparable in all tubers and suggests a dysmaturation affecting early and late migratory patterns, with a more severe impairment of the late stage of maturation.
Collapse
Affiliation(s)
- Angelika Mühlebner
- Department of (Neuro) Pathology, Academic Medical Center, Amsterdam, The Netherlands ; Department of Pediatrics, Medical University Vienna, Vienna, Austria
| | - Anand M Iyer
- Department of (Neuro) Pathology, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Jasper J Anink
- Department of (Neuro) Pathology, Academic Medical Center, Amsterdam, The Netherlands
| | - Floor E Jansen
- Department of Pediatric Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tim J Veersema
- Department of Pediatric Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Kees P Braun
- Department of Pediatric Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wim G M Spliet
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wim van Hecke
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Figen Söylemezoğlu
- Department of Pathology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Martha Feucht
- Department of Pediatrics, Medical University Vienna, Vienna, Austria
| | - Pavel Krsek
- Department of Neurology, Charles University, 2nd Faculty of Medicine, Motol University Hospital, Prague, Czech Republic
| | - Josef Zamecnik
- Department of Pathology and Molecular Medicine, Charles University, 2nd Faculty of Medicine, Motol University Hospital, Prague, Czech Republic
| | | | - Tilman Polster
- Epilepsy Centre Bethel, Krankenhaus Mara, Bielefeld, Germany
| | - Roland Coras
- Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | - Ingmar Blümcke
- Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | - Eleonora Aronica
- Department of (Neuro) Pathology, Academic Medical Center, Amsterdam, The Netherlands ; Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands ; Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands
| |
Collapse
|
278
|
Massart R, Mignon V, Stanic J, Munoz-Tello P, Becker JAJ, Kieffer BL, Darmon M, Sokoloff P, Diaz J. Developmental and adult expression patterns of the G-protein-coupled receptor GPR88 in the rat: Establishment of a dual nuclear-cytoplasmic localization. J Comp Neurol 2016; 524:2776-802. [PMID: 26918661 DOI: 10.1002/cne.23991] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 02/22/2016] [Accepted: 02/23/2016] [Indexed: 01/31/2023]
Abstract
GPR88 is a neuronal cerebral orphan G-protein-coupled receptor (GPCR) that has been linked to various psychiatric disorders. However, no extensive description of its localization has been provided so far. Here, we investigate the spatiotemporal expression of the GPR88 in prenatal and postnatal rat tissues by using in situ hybridization and immunohistochemistry. GPR88 protein was initially detected at embryonic day 16 (E16) in the striatal primordium. From E16-E20 to adulthood, the highest expression levels of both protein and mRNA were observed in striatum, olfactory tubercle, nucleus accumbens, amygdala, and neocortex, whereas in spinal cord, pons, and medulla GPR88 expression remains discrete. We observed an intracellular redistribution of GPR88 during cortical lamination. In the cortical plate of the developing cortex, GPR88 presents a classical GPCR plasma membrane/cytoplasmic localization that shifts, on the day of birth, to nuclei of neurons progressively settling in layers V to II. This intranuclear localization remains throughout adulthood and was also detected in monkey and human cortex as well as in the amygdala and hypothalamus of rats. Apart from the central nervous system, GPR88 was transiently expressed at high levels in peripheral tissues, including adrenal cortex (E16-E21) and cochlear ganglia (E19-P3), and also at moderate levels in retina (E18-E19) and spleen (E21-P7). The description of the GPR88 anatomical expression pattern may provide precious functional insights into this novel receptor. Furthermore, the GRP88 nuclear localization suggests nonclassical GPCR modes of action of the protein that could be relevant for cortical development and psychiatric disorders. J. Comp. Neurol. 524:2776-2802, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Renaud Massart
- INSERM UMR894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014, Paris, France.,Neurology-Psychiatry Department, Pierre Fabre Research Institute, 81100, Castres, France
| | - Virginie Mignon
- INSERM UMR894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006, Paris, France
| | - Jennifer Stanic
- INSERM UMR894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014, Paris, France
| | - Paola Munoz-Tello
- INSERM UMR894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014, Paris, France
| | - Jerôme A J Becker
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, CNRS, INSERM, 67400, Illkirch-Graffenstaden, France
| | - Brigitte L Kieffer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, CNRS, INSERM, 67400, Illkirch-Graffenstaden, France
| | - Michèle Darmon
- INSERM UMR894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014, Paris, France
| | - Pierre Sokoloff
- Neurology-Psychiatry Department, Pierre Fabre Research Institute, 81100, Castres, France
| | - Jorge Diaz
- INSERM UMR894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006, Paris, France
| |
Collapse
|
279
|
Mutually repressive interaction between Brn1/2 and Rorb contributes to the establishment of neocortical layer 2/3 and layer 4. Proc Natl Acad Sci U S A 2016; 113:3371-6. [PMID: 26951672 DOI: 10.1073/pnas.1515949113] [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] [Indexed: 12/20/2022] Open
Abstract
Although several molecules have been shown to play important roles in subtype specification of neocortical neurons, the entire mechanism involved in the specification, in particular, of upper cortical plate (UCP) neurons still remains unclear. The UCP, which is responsible for intracortical connections in the neocortex, comprises histologically, functionally, and molecularly different layer 2/3 (L2/3) and L4. Here, we report the essential interactions between two types of transcription factors, Rorb (RAR-related orphan receptor beta) and Brn1/2 (Brain-1/Brain-2), for UCP specification. We found that Brn2 expression was detected in all upper layers in the immature UCP, but was subsequently restricted to L2/3, accompanied by up-regulation of Rorb in L4, suggesting demarcation of L2/3 and L4 during cortical maturation. Rorb indeed inhibited Brn2 expression and the expression of other L2/3 characteristics, revealed by ectopic expression and knockdown studies. Moreover, this inhibition occurred through direct binding of Rorb to the Brn2 locus. Conversely, Brn1/2 also inhibited Rorb expression and the expression of several L4 characteristics. Together, these results suggest that a mutually repressive mechanism exists between Brn1/2 and Rorb expression and that the established expression of Brn1/2 and Rorb further specifies those neurons into L2/3 and L4, respectively, during UCP maturation.
Collapse
|
280
|
SATB2 Expression Distinguishes Ovarian Metastases of Colorectal and Appendiceal Origin From Primary Ovarian Tumors of Mucinous or Endometrioid Type. Am J Surg Pathol 2016; 40:419-32. [DOI: 10.1097/pas.0000000000000553] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
281
|
Oishi K, Nakagawa N, Tachikawa K, Sasaki S, Aramaki M, Hirano S, Yamamoto N, Yoshimura Y, Nakajima K. Identity of neocortical layer 4 neurons is specified through correct positioning into the cortex. eLife 2016; 5. [PMID: 26880563 PMCID: PMC4764574 DOI: 10.7554/elife.10907] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 01/28/2016] [Indexed: 11/13/2022] Open
Abstract
Many cell-intrinsic mechanisms have been shown to regulate neuronal subtype specification in the mammalian neocortex. However, how much cell environment is crucial for subtype determination still remained unclear. Here, we show that knockdown of Protocadherin20 (Pcdh20), which is expressed in post-migratory neurons of layer 4 (L4) lineage, caused the cells to localize in L2/3. The ectopically positioned "future L4 neurons" lost their L4 characteristics but acquired L2/3 characteristics. Knockdown of a cytoskeletal protein in the future L4 neurons, which caused random disruption of positioning, also showed that those accidentally located in L4 acquired the L4 characteristics. Moreover, restoration of positioning of the Pcdh20-knockdown neurons into L4 rescued the specification failure. We further suggest that the thalamocortical axons provide a positional cue to specify L4 identity. These results suggest that the L4 identity is not completely determined at the time of birth but ensured by the surrounding environment after appropriate positioning.
Collapse
Affiliation(s)
- Koji Oishi
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Nao Nakagawa
- Division of Visual Information Processing, National Institute for Physiological Sciences, National Institutes for Natural Sciences, Okazaki, Japan.,Department of Physiological Sciences, Graduate University for Advanced Studies, Okazaki, Japan
| | - Kashiko Tachikawa
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Shinji Sasaki
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Michihiko Aramaki
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Shinji Hirano
- Department of Cell Biology, Kansai Medical University, Osaka, Japan
| | - Nobuhiko Yamamoto
- Laboratory of Cellular and Molecular Neurobiology, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Yumiko Yoshimura
- Division of Visual Information Processing, National Institute for Physiological Sciences, National Institutes for Natural Sciences, Okazaki, Japan.,Department of Physiological Sciences, Graduate University for Advanced Studies, Okazaki, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
282
|
CRISPR/Cas9-mediated gene knockout in the mouse brain using in utero electroporation. Sci Rep 2016; 6:20611. [PMID: 26857612 PMCID: PMC4746659 DOI: 10.1038/srep20611] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 01/08/2016] [Indexed: 01/23/2023] Open
Abstract
The CRISPR/Cas9 system has recently been adapted for generating knockout mice to investigate physiological functions and pathological mechanisms. Here, we report a highly efficient procedure for brain-specific disruption of genes of interest in vivo. We constructed pX330 plasmids expressing humanized Cas9 and single-guide RNAs (sgRNAs) against the Satb2 gene, which encodes an AT-rich DNA-binding transcription factor and is responsible for callosal axon projections in the developing mouse brain. We first confirmed that these constructs efficiently induced double-strand breaks (DSBs) in target sites of exogenous plasmids both in vitro and in vivo. We then found that the introduction of pX330-Satb2 into the developing mouse brain using in utero electroporation led to a dramatic reduction of Satb2 expression in the transfected cerebral cortex, suggesting DSBs had occurred in the Satb2 gene with high efficiency. Furthermore, we found that Cas9-mediated targeting of the Satb2 gene induced abnormalities in axonal projection patterns, which is consistent with the phenotypes previously observed in Satb2 mutant mice. Introduction of pX330-NeuN using our procedure also resulted in the efficient disruption of the NeuN gene. Thus, our procedure combining the CRISPR/Cas9 system and in utero electroporation is an effective and rapid approach to achieve brain-specific gene knockout in vivo.
Collapse
|
283
|
Xing L, Larsen RS, Bjorklund GR, Li X, Wu Y, Philpot BD, Snider WD, Newbern JM. Layer specific and general requirements for ERK/MAPK signaling in the developing neocortex. eLife 2016; 5. [PMID: 26848828 PMCID: PMC4758957 DOI: 10.7554/elife.11123] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 01/04/2016] [Indexed: 12/11/2022] Open
Abstract
Aberrant signaling through the Raf/MEK/ERK (ERK/MAPK) pathway causes pathology in a family of neurodevelopmental disorders known as 'RASopathies' and is implicated in autism pathogenesis. Here, we have determined the functions of ERK/MAPK signaling in developing neocortical excitatory neurons. Our data reveal a critical requirement for ERK/MAPK signaling in the morphological development and survival of large Ctip2+ neurons in layer 5. Loss of Map2k1/2 (Mek1/2) led to deficits in corticospinal tract formation and subsequent corticospinal neuron apoptosis. ERK/MAPK hyperactivation also led to reduced corticospinal axon elongation, but was associated with enhanced arborization. ERK/MAPK signaling was dispensable for axonal outgrowth of layer 2/3 callosal neurons. However, Map2k1/2 deletion led to reduced expression of Arc and enhanced intrinsic excitability in both layers 2/3 and 5, in addition to imbalanced synaptic excitation and inhibition. These data demonstrate selective requirements for ERK/MAPK signaling in layer 5 circuit development and general effects on cortical pyramidal neuron excitability. DOI:http://dx.doi.org/10.7554/eLife.11123.001 In the nervous system, cells called neurons form networks that relay information in the form of electrical signals around the brain and the rest of the body. Typically, an electrical signal travels from branch-like structures at one end of the cell, through the cell body and then along a long fiber called an axon to reach junctions with another neurons. The connections between neurons start to form as the nervous system develops in the embryo, and any errors or delays in this process can cause severe neurological disorders and intellectual disabilities. For example, genetic mutations affecting a communication system within cells known as the ERK/MAPK pathway can lead to a family of syndromes called the “RASopathies”. Abnormalities in this pathway may also contribute to certain types of autism. However, it is not clear how alterations to the ERK/MAPK pathway cause these conditions. Xing et al. investigated whether ERK/MAPK signaling regulates the formation of connections between neurons and the activity of neurons in mouse brains. The experiments showed that the growth of axons that extend from an area of the brain called the cerebral cortex towards the spinal cord are particularly sensitive to changes in the level of signaling through the ERK/MAPK pathway. On the other hand, inhibiting the pathway has relatively little effect on the growth of axons within the cerebral cortex. Further experiments showed that many neurons in the cerebral cortex require the ERK/MAPK pathway to activate genes that alter neuronal activity and the strength of the connections between neurons. Xing et al.’s findings suggest that defects in the connections between the cerebral cortex and different regions of the nervous system may contribute to the symptoms observed in patients with conditions linked to alterations in ERK/MAPK activity. Future studies will focus on understanding the molecular mechanisms by which ERK/MAPK pathway influences the organization and activity of neuron circuits during the development of the nervous system. DOI:http://dx.doi.org/10.7554/eLife.11123.002
Collapse
Affiliation(s)
- Lei Xing
- University of North Carolina Neuroscience Center, The University of North Carolina School of Medicine, Chapel Hill, United States
| | - Rylan S Larsen
- Allen Institute for Brain Science, Seattle, United States
| | | | - Xiaoyan Li
- University of North Carolina Neuroscience Center, The University of North Carolina School of Medicine, Chapel Hill, United States
| | - Yaohong Wu
- University of North Carolina Neuroscience Center, The University of North Carolina School of Medicine, Chapel Hill, United States
| | - Benjamin D Philpot
- University of North Carolina Neuroscience Center, The University of North Carolina School of Medicine, Chapel Hill, United States.,Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States.,Carolina Institute for Developmental Disabilities, The University of North Carolina School of Medicine, Chapel Hill, United States
| | - William D Snider
- University of North Carolina Neuroscience Center, The University of North Carolina School of Medicine, Chapel Hill, United States.,Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States.,Carolina Institute for Developmental Disabilities, The University of North Carolina School of Medicine, Chapel Hill, United States
| | - Jason M Newbern
- School of Life Sciences, Arizona State University, Tempe, United States
| |
Collapse
|
284
|
Harb K, Magrinelli E, Nicolas CS, Lukianets N, Frangeul L, Pietri M, Sun T, Sandoz G, Grammont F, Jabaudon D, Studer M, Alfano C. Area-specific development of distinct projection neuron subclasses is regulated by postnatal epigenetic modifications. eLife 2016; 5:e09531. [PMID: 26814051 PMCID: PMC4744182 DOI: 10.7554/elife.09531] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 12/13/2015] [Indexed: 12/25/2022] Open
Abstract
During cortical development, the identity of major classes of long-distance projection neurons is established by the expression of molecular determinants, which become gradually restricted and mutually exclusive. However, the mechanisms by which projection neurons acquire their final properties during postnatal stages are still poorly understood. In this study, we show that the number of neurons co-expressing Ctip2 and Satb2, respectively involved in the early specification of subcerebral and callosal projection neurons, progressively increases after birth in the somatosensory cortex. Ctip2/Satb2 postnatal co-localization defines two distinct neuronal subclasses projecting either to the contralateral cortex or to the brainstem suggesting that Ctip2/Satb2 co-expression may refine their properties rather than determine their identity. Gain- and loss-of-function approaches reveal that the transcriptional adaptor Lmo4 drives this maturation program through modulation of epigenetic mechanisms in a time- and area-specific manner, thereby indicating that a previously unknown genetic program postnatally promotes the acquisition of final subtype-specific features.
Collapse
Affiliation(s)
- Kawssar Harb
- Institut de Biologie Valrose, University of Nice Sophia Antipolis, Nice, France.,Institut de Biologie Valrose, Institut national de la santé et de la recherche médicale, Nice, France.,Centre national de la recherche scientifique, Institut de Biologie Valrose, Nice, France
| | - Elia Magrinelli
- Institut de Biologie Valrose, University of Nice Sophia Antipolis, Nice, France.,Institut de Biologie Valrose, Institut national de la santé et de la recherche médicale, Nice, France.,Centre national de la recherche scientifique, Institut de Biologie Valrose, Nice, France
| | - Céline S Nicolas
- Institut de Biologie Valrose, University of Nice Sophia Antipolis, Nice, France.,Institut de Biologie Valrose, Institut national de la santé et de la recherche médicale, Nice, France.,Centre national de la recherche scientifique, Institut de Biologie Valrose, Nice, France
| | - Nikita Lukianets
- Institut de Biologie Valrose, University of Nice Sophia Antipolis, Nice, France.,Institut de Biologie Valrose, Institut national de la santé et de la recherche médicale, Nice, France.,Centre national de la recherche scientifique, Institut de Biologie Valrose, Nice, France
| | - Laura Frangeul
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Mariel Pietri
- Institut de Biologie Valrose, University of Nice Sophia Antipolis, Nice, France.,Institut de Biologie Valrose, Institut national de la santé et de la recherche médicale, Nice, France.,Centre national de la recherche scientifique, Institut de Biologie Valrose, Nice, France
| | - Tao Sun
- Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, United States
| | - Guillaume Sandoz
- Institut de Biologie Valrose, University of Nice Sophia Antipolis, Nice, France.,Institut de Biologie Valrose, Institut national de la santé et de la recherche médicale, Nice, France.,Centre national de la recherche scientifique, Institut de Biologie Valrose, Nice, France
| | - Franck Grammont
- Institut de Biologie Valrose, University of Nice Sophia Antipolis, Nice, France.,Laboratoire J.A. Dieudonné, Nice, France
| | - Denis Jabaudon
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Michele Studer
- Institut de Biologie Valrose, University of Nice Sophia Antipolis, Nice, France.,Institut de Biologie Valrose, Institut national de la santé et de la recherche médicale, Nice, France.,Centre national de la recherche scientifique, Institut de Biologie Valrose, Nice, France
| | - Christian Alfano
- Institut de Biologie Valrose, University of Nice Sophia Antipolis, Nice, France.,Institut de Biologie Valrose, Institut national de la santé et de la recherche médicale, Nice, France.,Centre national de la recherche scientifique, Institut de Biologie Valrose, Nice, France
| |
Collapse
|
285
|
Rodríguez-Tornos FM, Briz CG, Weiss LA, Sebastián-Serrano A, Ares S, Navarrete M, Frangeul L, Galazo M, Jabaudon D, Esteban JA, Nieto M. Cux1 Enables Interhemispheric Connections of Layer II/III Neurons by Regulating Kv1-Dependent Firing. Neuron 2016; 89:494-506. [PMID: 26804994 DOI: 10.1016/j.neuron.2015.12.020] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 04/23/2015] [Accepted: 12/01/2015] [Indexed: 01/19/2023]
Abstract
Neuronal subtype-specific transcription factors (TFs) instruct key features of neuronal function and connectivity. Activity-dependent mechanisms also contribute to wiring and circuit assembly, but whether and how they relate to TF-directed neuronal differentiation is poorly investigated. Here we demonstrate that the TF Cux1 controls the formation of the layer II/III corpus callosum (CC) projections through the developmental transcriptional regulation of Kv1 voltage-dependent potassium channels and the resulting postnatal switch to a Kv1-dependent firing mode. Loss of Cux1 function led to a decrease in the expression of Kv1 transcripts, aberrant firing responses, and selective loss of CC contralateral innervation. Firing and innervation were rescued by re-expression of Kv1 or postnatal reactivation of Cux1. Knocking down Kv1 mimicked Cux1-mediated CC axonal loss. These findings reveal that activity-dependent processes are central bona fide components of neuronal TF-differentiation programs and establish the importance of intrinsic firing modes in circuit assembly within the neocortex.
Collapse
Affiliation(s)
| | - Carlos G Briz
- Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain
| | - Linnea A Weiss
- Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain
| | - Alvaro Sebastián-Serrano
- Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain
| | - Saúl Ares
- Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain; Departamento de Matemáticas Universidad Carlos III de Madrid, Grupo Interdisciplinar de Sistemas Complejos (GISC), 28911 Leganés, Madrid, Spain
| | - Marta Navarrete
- Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Científicas (CSIC-UAM), 28049 Madrid, Spain
| | - Laura Frangeul
- Department of Basic Neurosciences, University of Geneva, 1211 Geneva 4, Switzerland
| | - Maria Galazo
- HSCRB Harvard University, Cambridge, MA 02138, USA
| | - Denis Jabaudon
- Department of Basic Neurosciences, University of Geneva, 1211 Geneva 4, Switzerland
| | - José A Esteban
- Centro de Biología Molecular "Severo Ochoa," Consejo Superior de Investigaciones Científicas (CSIC-UAM), 28049 Madrid, Spain
| | - Marta Nieto
- Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain.
| |
Collapse
|
286
|
Lee JS, Yoo Y, Lim BC, Kim KJ, Choi M, Chae JH. SATB2-associated syndrome presenting with Rett-like phenotypes. Clin Genet 2016; 89:728-32. [PMID: 26596517 DOI: 10.1111/cge.12698] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 11/18/2015] [Accepted: 11/19/2015] [Indexed: 11/30/2022]
Abstract
The SATB2-associated syndrome (SAS) was proposed recently, after the SATB2 gene was initially discovered to be associated with isolated cleft palate. This syndrome is characterized by intellectual disability with delayed speech development, facial dysmorphism, cleft or high-arched palate, and dentition problems. Here, we describe two novel SATB2 sequence variants in two unrelated patients presenting with Rett-like phenotypes. We performed trio-based whole-exome sequencing in a 17-month-old girl presenting with severe retardation and Rett-like phenotypes, which revealed a de novo missense variant in SATB2 (p.Glu396Gln). Moreover, targeted sequencing of the SATB2 gene was performed in a 2-year-old girl with severe psychomotor retardation, facial hypotonia, and cleft palate who also exhibited some features of Rett syndrome. A nonsense variant in SATB2 was identified in this patient (p.Arg459*). This study expanded the clinical and genetic spectrum of SAS. SATB2 variants should be considered in cases with psychomotor retardation alone or in any cases with Rett-like phenotypes, regardless of the typical features of SAS such as cleft palate.
Collapse
Affiliation(s)
- J S Lee
- Department of Pediatrics, Gachon University Gil Medical Center, Incheon, South Korea
| | - Y Yoo
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - B C Lim
- Department of Pediatrics, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - K J Kim
- Department of Pediatrics, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - M Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - J-H Chae
- Department of Pediatrics, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
287
|
Nechiporuk T, McGann J, Mullendorff K, Hsieh J, Wurst W, Floss T, Mandel G. The REST remodeling complex protects genomic integrity during embryonic neurogenesis. eLife 2016; 5:e09584. [PMID: 26745185 PMCID: PMC4728133 DOI: 10.7554/elife.09584] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 10/20/2015] [Indexed: 01/01/2023] Open
Abstract
The timely transition from neural progenitor to post-mitotic neuron requires down-regulation and loss of the neuronal transcriptional repressor, REST. Here, we have used mice containing a gene trap in the Rest gene, eliminating transcription from all coding exons, to remove REST prematurely from neural progenitors. We find that catastrophic DNA damage occurs during S-phase of the cell cycle, with long-term consequences including abnormal chromosome separation, apoptosis, and smaller brains. Persistent effects are evident by latent appearance of proneural glioblastoma in adult mice deleted additionally for the tumor suppressor p53 protein (p53). A previous line of mice deleted for REST in progenitors by conventional gene targeting does not exhibit these phenotypes, likely due to a remaining C-terminal peptide that still binds chromatin and recruits co-repressors. Our results suggest that REST-mediated chromatin remodeling is required in neural progenitors for proper S-phase dynamics, as part of its well-established role in repressing neuronal genes until terminal differentiation. DOI:http://dx.doi.org/10.7554/eLife.09584.001 In the brain, cells called neurons connect to each other to form complex networks through which information is rapidly processed. These cells start to form in the developing brains of animal embryos when “neural” stem cells divide in a process called neurogenesis. For this process to proceed normally, particular genes in the stem cells have to be switched on or off at different times. This ensures that the protein products of the genes are only made when they are needed. Proteins called transcription factors can bind to DNA to activate or inactivate particular genes; for example, a transcription factor called REST inactivates thousands of genes that are needed by neurons. During neurogenesis, the production of REST normally declines, and some studies have shown that if the production of this protein is artificially increased, the formation of neurons is delayed. However, other studies suggest that REST may not play a major role in neurogenesis. Here, Nechiporuk et al. re-examine the role of REST in mice. The experiments used genetically modified mice in which the gene that encodes REST was prematurely switched off in neural stem cells. Compared with normal mice, these mutant mice had much smaller brains that contained fewer neurons because the stem cells stopped dividing earlier than normal. Unexpectedly, many genes that are normally switched off by REST, were not significantly changed, while genes that are not normally regulated by REST – such as the gene that encodes a protein called p53 – were active. It is known from previous work that p53 is expressed when cells are exposed to harmful conditions that can damage DNA. This helps to prevent cells from becoming cancerous. Nechiporuk et al. found that cells that lacked REST had higher levels of DNA damage than normal cells due to errors during the process of copying DNA before a cell divides. Furthermore, when both REST and p53 were absent, the neural stem cells became cancerous and formed tumors in the mice. Nechiporuk et al.’s findings suggest that REST protects the DNA of genes that are needed for neurons to form and work properly. The new challenge is to understand where in the genome the damage is occurring. DOI:http://dx.doi.org/10.7554/eLife.09584.002
Collapse
Affiliation(s)
- Tamilla Nechiporuk
- Vollum Institute, Howard Hughes Medical Institute, Oregon Health and Science University, Portland, United States
| | - James McGann
- Vollum Institute, Howard Hughes Medical Institute, Oregon Health and Science University, Portland, United States
| | - Karin Mullendorff
- Vollum Institute, Howard Hughes Medical Institute, Oregon Health and Science University, Portland, United States
| | - Jenny Hsieh
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, United States.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany.,Technische Universität München, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Ludwig-Maximilians-Universität, Munich, Germany
| | - Thomas Floss
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Gail Mandel
- Vollum Institute, Howard Hughes Medical Institute, Oregon Health and Science University, Portland, United States
| |
Collapse
|
288
|
Nikouei K, Muñoz-Manchado AB, Hjerling-Leffler J. BCL11B/CTIP2 is highly expressed in GABAergic interneurons of the mouse somatosensory cortex. J Chem Neuroanat 2016; 71:1-5. [DOI: 10.1016/j.jchemneu.2015.12.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 11/22/2015] [Accepted: 12/11/2015] [Indexed: 11/25/2022]
|
289
|
Gascón S, Murenu E, Masserdotti G, Ortega F, Russo GL, Petrik D, Deshpande A, Heinrich C, Karow M, Robertson SP, Schroeder T, Beckers J, Irmler M, Berndt C, Angeli JPF, Conrad M, Berninger B, Götz M. Identification and Successful Negotiation of a Metabolic Checkpoint in Direct Neuronal Reprogramming. Cell Stem Cell 2015; 18:396-409. [PMID: 26748418 DOI: 10.1016/j.stem.2015.12.003] [Citation(s) in RCA: 276] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 11/07/2015] [Accepted: 12/10/2015] [Indexed: 11/16/2022]
Abstract
Despite the widespread interest in direct neuronal reprogramming, the mechanisms underpinning fate conversion remain largely unknown. Our study revealed a critical time point after which cells either successfully convert into neurons or succumb to cell death. Co-transduction with Bcl-2 greatly improved negotiation of this critical point by faster neuronal differentiation. Surprisingly, mutants with reduced or no affinity for Bax demonstrated that Bcl-2 exerts this effect by an apoptosis-independent mechanism. Consistent with a caspase-independent role, ferroptosis inhibitors potently increased neuronal reprogramming by inhibiting lipid peroxidation occurring during fate conversion. Genome-wide expression analysis confirmed that treatments promoting neuronal reprogramming elicit an anti-oxidative stress response. Importantly, co-expression of Bcl-2 and anti-oxidative treatments leads to an unprecedented improvement in glial-to-neuron conversion after traumatic brain injury in vivo, underscoring the relevance of these pathways in cellular reprograming irrespective of cell type in vitro and in vivo.
Collapse
Affiliation(s)
- Sergio Gascón
- Physiological Genomics, Biomedical Center Ludwig-Maximilians-University Munich, 80336 Munich, Germany; Institute for Stem Cell Research, Helmholtz Center Munich, 85764 Neuherberg, Germany.
| | - Elisa Murenu
- Physiological Genomics, Biomedical Center Ludwig-Maximilians-University Munich, 80336 Munich, Germany; Institute for Stem Cell Research, Helmholtz Center Munich, 85764 Neuherberg, Germany
| | - Giacomo Masserdotti
- Physiological Genomics, Biomedical Center Ludwig-Maximilians-University Munich, 80336 Munich, Germany; Institute for Stem Cell Research, Helmholtz Center Munich, 85764 Neuherberg, Germany
| | - Felipe Ortega
- Physiological Genomics, Biomedical Center Ludwig-Maximilians-University Munich, 80336 Munich, Germany; Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, 55128 Mainz, Germany; Biochemistry and Molecular Biology Department, Faculty of Veterinary Medicine, Complutense University, Avenue Puerta de Hierro, 28040 Madrid, Spain
| | - Gianluca L Russo
- Physiological Genomics, Biomedical Center Ludwig-Maximilians-University Munich, 80336 Munich, Germany; Institute for Stem Cell Research, Helmholtz Center Munich, 85764 Neuherberg, Germany
| | - David Petrik
- Physiological Genomics, Biomedical Center Ludwig-Maximilians-University Munich, 80336 Munich, Germany; Institute for Stem Cell Research, Helmholtz Center Munich, 85764 Neuherberg, Germany
| | - Aditi Deshpande
- Physiological Genomics, Biomedical Center Ludwig-Maximilians-University Munich, 80336 Munich, Germany
| | - Christophe Heinrich
- Physiological Genomics, Biomedical Center Ludwig-Maximilians-University Munich, 80336 Munich, Germany
| | - Marisa Karow
- Physiological Genomics, Biomedical Center Ludwig-Maximilians-University Munich, 80336 Munich, Germany
| | - Stephen P Robertson
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, 9016 Dunedin, New Zealand
| | - Timm Schroeder
- Research Unit Stem Cell Dynamics, Helmholtz Center Munich, Neuherberg, 85764 Neuherberg, Germany
| | - Johannes Beckers
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Institute of Experimental Genetics, Helmholtz Center Munich GmbH, 85764 Neuherberg, Germany; Center of Life and Food Sciences Weihenstephan, Technical University Munich, 85354 Freising, Germany
| | - Martin Irmler
- Institute of Experimental Genetics, Helmholtz Center Munich GmbH, 85764 Neuherberg, Germany
| | - Carsten Berndt
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Merowingerplatz 1a, 40225 Düsseldorf, Germany
| | | | - Marcus Conrad
- Institute of Developmental Genetics, Helmholtz Center Munich, 85764 Neuherberg, Germany
| | - Benedikt Berninger
- Physiological Genomics, Biomedical Center Ludwig-Maximilians-University Munich, 80336 Munich, Germany; Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, 55128 Mainz, Germany; Focus Program Translational Neuroscience, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| | - Magdalena Götz
- Physiological Genomics, Biomedical Center Ludwig-Maximilians-University Munich, 80336 Munich, Germany; Institute for Stem Cell Research, Helmholtz Center Munich, 85764 Neuherberg, Germany; Excellence Cluster of Systems Neurology (SYNERGY), 80336 Munich, Germany.
| |
Collapse
|
290
|
Daviaud N, Chen K, Huang Y, Friedel RH, Zou H. Impaired cortical neurogenesis in plexin-B1 and -B2 double deletion mutant. Dev Neurobiol 2015; 76:882-99. [PMID: 26579598 DOI: 10.1002/dneu.22364] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 10/12/2015] [Accepted: 11/16/2015] [Indexed: 11/08/2022]
Abstract
Mammalian cortical expansion is tightly controlled by fine-tuning of proliferation and differentiation of neural progenitors in a region-specific manner. How extrinsic cues interface with cell-intrinsic programs to balance proliferative versus neurogenic decisions remains an unsolved question. We examined the function of Semaphorin receptors Plexin-B1 and -B2 in corticogenesis by generating double mutants, whereby Plexin-B2 was conditionally ablated in the developing brain in a Plexin-B1 null mutant background. Absence of both Plexin-Bs resulted in cortical thinning, particularly in the caudomedial cortex. Plexin-B1/B2 double, but not single, mutants exhibited a reduced neural progenitor pool, attributable to decreased proliferation and an altered division mode favoring cell cycle exit. This resulted in deficient production of neurons throughout the neurogenic period, proportionally affecting all cortical laminae. Consistent with the in vivo data, cultured neural progenitors lacking both Plexin-B1 and -B2 displayed decreased proliferative capacity and increased spontaneous differentiation. Our study therefore defines a novel function of Plexin-B1 and -B2 in transmitting extrinsic signals to maintain proliferative and undifferentiated states of neural progenitors. As single mutants displayed no apparent cortical defects, we conclude that Plexin-B1 and -B2 play redundant or compensatory roles during forebrain development to ensure proper neuronal production and neocortical expansion. © 2015 Wiley Periodicals, Inc. Develop Neurobiol 76: 882-899, 2016.
Collapse
Affiliation(s)
- Nicolas Daviaud
- Fishberg Department of Neuroscience and Friedman Brain Institute, New York, New York 10029
| | - Karen Chen
- Fishberg Department of Neuroscience and Friedman Brain Institute, New York, New York 10029
| | - Yong Huang
- Fishberg Department of Neuroscience and Friedman Brain Institute, New York, New York 10029
| | - Roland H Friedel
- Fishberg Department of Neuroscience and Friedman Brain Institute, New York, New York 10029.,Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Hongyan Zou
- Fishberg Department of Neuroscience and Friedman Brain Institute, New York, New York 10029.,Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| |
Collapse
|
291
|
Lim CS, Yang JE, Lee YK, Lee K, Lee JA, Kaang BK. Understanding the molecular basis of autism in a dish using hiPSCs-derived neurons from ASD patients. Mol Brain 2015; 8:57. [PMID: 26419846 PMCID: PMC4589208 DOI: 10.1186/s13041-015-0146-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 09/11/2015] [Indexed: 12/20/2022] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder characterized by deficits in social cognition, language development, and repetitive/restricted behaviors. Due to the complexity and heterogeneity of ASD and lack of a proper human cellular model system, the pathophysiological mechanism of ASD during the developmental process is largely unknown. However, recent progress in induced pluripotent stem cell (iPSC) technology as well as in vitro neural differentiation techniques have allowed us to functionally characterize neurons and analyze cortical development during neural differentiation. These technical advances will increase our understanding of the pathogenic mechanisms of heterogeneous ASD and help identify molecular biomarkers for patient stratification as well as personalized medicine. In this review, we summarize our current knowledge of iPSC generation, differentiation of specific neuronal subtypes from iPSCs, and phenotypic characterizations of human ASD patient-derived iPSC models. Finally, we discuss the current limitations of iPSC technology and future directions of ASD pathophysiology studies using iPSCs.
Collapse
Affiliation(s)
- Chae-Seok Lim
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Gwanangno 599, Seoul, Gwanak-gu, 151-747, Korea
| | - Jung-Eun Yang
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Gwanangno 599, Seoul, Gwanak-gu, 151-747, Korea
| | - You-Kyung Lee
- Department of Biological Sciences and Biotechnology, College of Life Science and NanoTechnology, Hannam University, Jeonmin-dong 461-6, Daejeon, Yuseong-gu, 305-811, Korea
| | - Kyungmin Lee
- Department of Anatomy, Kyungpook National University Graduate School of Medicine, Dongin-dong 2-101, Daegu, Jung-gu, 700-422, Korea
| | - Jin-A Lee
- Department of Biological Sciences and Biotechnology, College of Life Science and NanoTechnology, Hannam University, Jeonmin-dong 461-6, Daejeon, Yuseong-gu, 305-811, Korea.
| | - Bong-Kiun Kaang
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Gwanangno 599, Seoul, Gwanak-gu, 151-747, Korea.
| |
Collapse
|
292
|
Bcl11a (Ctip1) Controls Migration of Cortical Projection Neurons through Regulation of Sema3c. Neuron 2015; 87:311-25. [PMID: 26182416 DOI: 10.1016/j.neuron.2015.06.023] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 05/04/2015] [Accepted: 06/16/2015] [Indexed: 01/12/2023]
Abstract
During neocortical development, neurons undergo polarization, oriented migration, and layer-type-specific differentiation. The transcriptional programs underlying these processes are not completely understood. Here, we show that the transcription factor Bcl11a regulates polarity and migration of upper layer neurons. Bcl11a-deficient late-born neurons fail to correctly switch from multipolar to bipolar morphology, resulting in impaired radial migration. We show that the expression of Sema3c is increased in migrating Bcl11a-deficient neurons and that Bcl11a is a direct negative regulator of Sema3c transcription. In vivo gain-of-function and rescue experiments demonstrate that Sema3c is a major downstream effector of Bcl11a required for the cell polarity switch and for the migration of upper layer neurons. Our data uncover a novel Bcl11a/Sema3c-dependent regulatory pathway used by migrating cortical neurons.
Collapse
|
293
|
Abstract
The neocortex is the part of the brain responsible for execution of higher-order brain functions, including cognition, sensory perception, and sophisticated motor control. During evolution, the neocortex has developed an unparalleled neuronal diversity, which still remains partly unclassified and unmapped at the functional level. Here, we broadly review the structural blueprint of the neocortex and discuss the current classification of its neuronal diversity. We then cover the principles and mechanisms that build neuronal diversity during cortical development and consider the impact of neuronal class-specific identity in shaping cortical connectivity and function.
Collapse
Affiliation(s)
- Simona Lodato
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138; ,
| | - Paola Arlotta
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138; ,
| |
Collapse
|
294
|
Abstract
The neocortex is the part of the brain responsible for execution of higher-order brain functions, including cognition, sensory perception, and sophisticated motor control. During evolution, the neocortex has developed an unparalleled neuronal diversity, which still remains partly unclassified and unmapped at the functional level. Here, we broadly review the structural blueprint of the neocortex and discuss the current classification of its neuronal diversity. We then cover the principles and mechanisms that build neuronal diversity during cortical development and consider the impact of neuronal class-specific identity in shaping cortical connectivity and function.
Collapse
Affiliation(s)
- Simona Lodato
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138; ,
| | - Paola Arlotta
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138; ,
| |
Collapse
|
295
|
Bayam E, Sahin GS, Guzelsoy G, Guner G, Kabakcioglu A, Ince-Dunn G. Genome-wide target analysis of NEUROD2 provides new insights into regulation of cortical projection neuron migration and differentiation. BMC Genomics 2015; 16:681. [PMID: 26341353 PMCID: PMC4560887 DOI: 10.1186/s12864-015-1882-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 08/26/2015] [Indexed: 12/03/2022] Open
Abstract
Background Cellular differentiation programs are controlled, to a large extent, by the combinatorial functioning of specific transcription factors. Cortical projection neurons constitute the major excitatory neuron population within the cortex and mediate long distance communication between the cortex and other brain regions. Our understanding of effector transcription factors and their downstream transcriptional programs that direct the differentiation process of cortical projection neurons is far from complete. Results In this study, we carried out a ChIP-Seq (chromatin-immunoprecipitation and sequencing) analysis of NEUROD2, an effector transcription factor expressed in lineages of cortical projection neurons during the peak of cortical excitatory neurogenesis. Our results suggest that during cortical development NEUROD2 targets key genes that are required for Reelin signaling, a major pathway that regulates the migration of neurons from germinal zones to their final layers of residence within the cortex. We also find that NEUROD2 binds to a large set of genes with functions in layer-specific differentiation and in axonal pathfinding of cortical projection neurons. Conclusions Our analysis of in vivo NEUROD2 target genes offers mechanistic insight into signaling pathways that regulate neuronal migration and axon guidance and identifies genes that are likely to be required for proper cortical development. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1882-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Efil Bayam
- Molecular Biology and Genetics Department, Koç University, Rumeli Feneri Yolu, Istanbul, 34450, Turkey.
| | - Gulcan Semra Sahin
- Molecular Biology and Genetics Department, Koç University, Rumeli Feneri Yolu, Istanbul, 34450, Turkey. .,Present Address: Neuroscience Department, Washington State University, Pullman, WA, 99164, USA.
| | - Gizem Guzelsoy
- Molecular Biology and Genetics Department, Koç University, Rumeli Feneri Yolu, Istanbul, 34450, Turkey.
| | - Gokhan Guner
- Molecular Biology and Genetics Department, Koç University, Rumeli Feneri Yolu, Istanbul, 34450, Turkey.
| | - Alkan Kabakcioglu
- Physics Department, Koç University, Rumeli Feneri Yolu, Istanbul, 34450, Turkey.
| | - Gulayse Ince-Dunn
- Molecular Biology and Genetics Department, Koç University, Rumeli Feneri Yolu, Istanbul, 34450, Turkey.
| |
Collapse
|
296
|
Molecular Biomarkers for Embryonic and Adult Neural Stem Cell and Neurogenesis. BIOMED RESEARCH INTERNATIONAL 2015; 2015:727542. [PMID: 26421301 PMCID: PMC4569757 DOI: 10.1155/2015/727542] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 11/19/2014] [Indexed: 02/07/2023]
Abstract
The procedure of neurogenesis has made numerous achievements in the past decades, during which various molecular biomarkers have been emerging and have been broadly utilized for the investigation of embryonic and adult neural stem cell (NSC). Nevertheless, there is not a consistent and systematic illustration to depict the functional characteristics of the specific markers expressed in distinct cell types during the different stages of neurogenesis. Here we gathered and generalized a series of NSC biomarkers emerging during the procedures of embryonic and adult neural stem cell, which may be used to identify the subpopulation cells with distinguishing characters in different timeframes of neurogenesis. The identifications of cell patterns will provide applications to the detailed investigations of diverse developmental cell stages and the extents of cell differentiation, which will facilitate the tracing of cell time-course and fate determination of specific cell types and promote the further and literal discoveries of embryonic and adult neurogenesis. Meanwhile, via the utilization of comprehensive applications under the aiding of the systematic knowledge framework, researchers may broaden their insights into the derivation and establishment of novel technologies to analyze the more detailed process of embryogenesis and adult neurogenesis.
Collapse
|
297
|
Mutual regulation between Satb2 and Fezf2 promotes subcerebral projection neuron identity in the developing cerebral cortex. Proc Natl Acad Sci U S A 2015; 112:11702-7. [PMID: 26324926 DOI: 10.1073/pnas.1504144112] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Generation of distinct cortical projection neuron subtypes during development relies in part on repression of alternative neuron identities. It was reported that the special AT-rich sequence-binding protein 2 (Satb2) is required for proper development of callosal neuron identity and represses expression of genes that are essential for subcerebral axon development. Surprisingly, Satb2 has recently been shown to be necessary for subcerebral axon development. Here, we unravel a previously unidentified mechanism underlying this paradox. We show that SATB2 directly activates transcription of forebrain embryonic zinc finger 2 (Fezf2) and SRY-box 5 (Sox5), genes essential for subcerebral neuron development. We find that the mutual regulation between Satb2 and Fezf2 enables Satb2 to promote subcerebral neuron identity in layer 5 neurons, and to repress subcerebral characters in callosal neurons. Thus, Satb2 promotes the development of callosal and subcerebral neurons in a cell context-dependent manner.
Collapse
|
298
|
Centrosomin represses dendrite branching by orienting microtubule nucleation. Nat Neurosci 2015; 18:1437-45. [PMID: 26322925 DOI: 10.1038/nn.4099] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 08/04/2015] [Indexed: 02/07/2023]
Abstract
Neuronal dendrite branching is fundamental for building nervous systems. Branch formation is genetically encoded by transcriptional programs to create dendrite arbor morphological diversity for complex neuronal functions. In Drosophila sensory neurons, the transcription factor Abrupt represses branching via an unknown effector pathway. Targeted screening for branching-control effectors identified Centrosomin, the primary centrosome-associated protein for mitotic spindle maturation. Centrosomin repressed dendrite branch formation and was used by Abrupt to simplify arbor branching. Live imaging revealed that Centrosomin localized to the Golgi cis face and that it recruited microtubule nucleation to Golgi outposts for net retrograde microtubule polymerization away from nascent dendrite branches. Removal of Centrosomin enabled the engagement of wee Augmin activity to promote anterograde microtubule growth into the nascent branches, leading to increased branching. The findings reveal that polarized targeting of Centrosomin to Golgi outposts during elaboration of the dendrite arbor creates a local system for guiding microtubule polymerization.
Collapse
|
299
|
Tantirigama MLS, Oswald MJ, Clare AJ, Wicky HE, Day RC, Hughes SM, Empson RM. Fezf2 expression in layer 5 projection neurons of mature mouse motor cortex. J Comp Neurol 2015; 524:829-45. [PMID: 26234885 DOI: 10.1002/cne.23875] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 07/29/2015] [Accepted: 07/30/2015] [Indexed: 12/24/2022]
Abstract
The mature cerebral cortex contains a wide diversity of neuron phenotypes. This diversity is specified during development by neuron-specific expression of key transcription factors, some of which are retained for the life of the animal. One of these key developmental transcription factors that is also retained in the adult is Fezf2, but the neuron types expressing it in the mature cortex are unknown. With a validated Fezf2-Gfp reporter mouse, whole-cell electrophysiology with morphology reconstruction, cluster analysis, in vivo retrograde labeling, and immunohistochemistry, we identify a heterogeneous population of Fezf2(+) neurons in both layer 5A and layer 5B of the mature motor cortex. Functional electrophysiology identified two distinct subtypes of Fezf2(+) neurons that resembled pyramidal tract projection neurons (PT-PNs) and intratelencephalic projection neurons (IT-PNs). Retrograde labeling confirmed the former type to include corticospinal projection neurons (CSpPNs) and corticothalamic projection neurons (CThPNs), whereas the latter type included crossed corticostriatal projection neurons (cCStrPNs) and crossed-corticocortical projection neurons (cCCPNs). The two Fezf2(+) subtypes expressed either CTIP2 or SATB2 to distinguish their physiological identity and confirmed that specific expression combinations of key transcription factors persist in the mature motor cortex. Our findings indicate a wider role for Fezf2 within gene expression networks that underpin the diversity of layer 5 cortical projection neurons.
Collapse
Affiliation(s)
- Malinda L S Tantirigama
- Department of Physiology, Brain Health Research Centre, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand, 9054
| | - Manfred J Oswald
- Department of Physiology, Brain Health Research Centre, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand, 9054
| | - Alison J Clare
- Department of Biochemistry, Brain Health Research Centre, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand, 9054
| | - Hollie E Wicky
- Department of Biochemistry, Brain Health Research Centre, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand, 9054
| | - Robert C Day
- Department of Biochemistry, Brain Health Research Centre, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand, 9054
| | - Stephanie M Hughes
- Department of Biochemistry, Brain Health Research Centre, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand, 9054
| | - Ruth M Empson
- Department of Physiology, Brain Health Research Centre, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand, 9054
| |
Collapse
|
300
|
Inoue M, Iwai R, Yamanishi E, Yamagata K, Komabayashi-Suzuki M, Honda A, Komai T, Miyachi H, Kitano S, Watanabe C, Teshima W, Mizutani KI. Deletion of Prdm8 impairs development of upper-layer neocortical neurons. Genes Cells 2015; 20:758-70. [PMID: 26283595 DOI: 10.1111/gtc.12274] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 06/15/2015] [Indexed: 01/17/2023]
Abstract
Upper-layer (UL) neocortical neurons are the most prominent distinguishing features of the mammalian neocortex compared with those of the avian dorsal cortex and are vastly expanded in primates. However, little is known about the identities of the genes that control the specification of UL neurons. Here, we found that Prdm8, a member of the PR (PRDI-BF1 and RIZ homology) domain protein family, was specifically expressed in the postnatal UL neocortex, particular those in late-born RORß-positive layer IV neurons. We generated homozygous Prdm8 knockout (Prdm8 KO) mice and found that the deletion of Prdm8 causes growth retardation and a reduced brain weight, although the brain weight-to-body weight ratio is unchanged at postnatal day 8 (P8). Immunohistochemistry showed that the relative UL thickness, but not the thickness of the deep layer (DL), was significantly reduced in Prdm8 KO mice compared with wild-type (WT) mice. In addition, we found that a number of late-born Brn2-positive UL neurons were significantly decreased in Prdm8 KO mice. To identify genes regulated by Prdm8 during neocortical development, we compared expression profiling analysis in Prdm8 KO and WT mice, and identified some candidate genes. These results suggest that the proper expression of Prdm8 is required for the normal development and construction of UL neurons in the mammalian neocortex.
Collapse
Affiliation(s)
- Mayuko Inoue
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto, 619-0225, Japan.,Institute for Virus Research, Kyoto University, Kyoto, 606-8507, Japan
| | - Ryota Iwai
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto, 619-0225, Japan
| | - Emiko Yamanishi
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto, 619-0225, Japan
| | - Kazuyuki Yamagata
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto, 619-0225, Japan
| | - Mariko Komabayashi-Suzuki
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto, 619-0225, Japan
| | - Aya Honda
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto, 619-0225, Japan
| | - Tae Komai
- Institute for Virus Research, Kyoto University, Kyoto, 606-8507, Japan
| | - Hitoshi Miyachi
- Institute for Virus Research, Kyoto University, Kyoto, 606-8507, Japan
| | - Satsuki Kitano
- Institute for Virus Research, Kyoto University, Kyoto, 606-8507, Japan
| | - Chisato Watanabe
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto, 619-0225, Japan
| | - Waka Teshima
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto, 619-0225, Japan
| | - Ken-ichi Mizutani
- Laboratory of Neural Differentiation, Graduate School of Brain Science, Doshisha University, Kyoto, 619-0225, Japan.,Japan Science and Technology Agency, PRESTO, Saitama, 332-0012, Japan
| |
Collapse
|