201
|
Heavner WE, Andoniadou CL, Pevny LH. Establishment of the neurogenic boundary of the mouse retina requires cooperation of SOX2 and WNT signaling. Neural Dev 2014; 9:27. [PMID: 25488119 PMCID: PMC4295269 DOI: 10.1186/1749-8104-9-27] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 11/14/2014] [Indexed: 12/03/2022] Open
Abstract
Background Eye development in vertebrates relies on the critical regulation of SOX2 expression. Humans with mutations in SOX2 often suffer from eye defects including anophthalmia (no eye) and microphthalmia (small eye). In mice, deletion of Sox2 in optic cup progenitor cells results in loss of neural competence and cell fate conversion of the neural retina to a non-neurogenic fate, specifically the acquisition of fate associated with progenitors of the ciliary epithelium. This fate is also promoted with constitutive expression of stabilized β-Catenin in the optic cup, where the WNT pathway is up-regulated. We addressed whether SOX2 co-ordinates the neurogenic boundary of the retina through modulating the WNT/β-Catenin pathway by using a genetic approach in the mouse. Results Upon deletion of Sox2 in the optic cup, response to WNT signaling was expanded, correlating with loss of neural competence, cell fate conversion of the neural retina to ciliary epithelium primordium and, in addition, increased cell cycle time of optic cup progenitors. Removal of Ctnnb1 rescued the cell fate conversion; however, the loss of neural competence and the proliferation defect resulting from lack of SOX2 were not overcome. Lastly, central Sox2-deficient optic cup progenitor cells exhibited WNT-independent up-regulation of D-type Cyclins. Conclusion We propose two distinct roles for SOX2 in the developing retina. Our findings suggest that SOX2 antagonizes the WNT pathway to maintain a neurogenic fate and, in contrast, regulates cycling of optic cup progenitors in a WNT-independent manner. Given that WNT signaling acting upstream of SOX2 has been implicated in the tumorigenicity of embryonic stem cell-derived retinal progenitor cells, our results distinguish the endogenous role of WNT signaling in early optic cup patterning and support a WNT-independent role for SOX2 in maintaining retinal progenitor cell proliferation. Electronic supplementary material The online version of this article (doi:10.1186/1749-8104-9-27) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Whitney E Heavner
- UNC Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA.
| | | | | |
Collapse
|
202
|
Kicheva A, Bollenbach T, Ribeiro A, Valle HP, Lovell-Badge R, Episkopou V, Briscoe J. Coordination of progenitor specification and growth in mouse and chick spinal cord. Science 2014; 345:1254927. [PMID: 25258086 PMCID: PMC4228193 DOI: 10.1126/science.1254927] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Development requires tissue growth as well as cell diversification. To address how these processes are coordinated, we analyzed the development of molecularly distinct domains of neural progenitors in the mouse and chick neural tube. We show that during development, these domains undergo changes in size that do not scale with changes in overall tissue size. Our data show that domain proportions are first established by opposing morphogen gradients and subsequently controlled by domain-specific regulation of differentiation rate but not differences in proliferation rate. Regulation of differentiation rate is key to maintaining domain proportions while accommodating both intra- and interspecies variations in size. Thus, the sequential control of progenitor specification and differentiation elaborates pattern without requiring that signaling gradients grow as tissues expand.
Collapse
Affiliation(s)
- Anna Kicheva
- Medical Research Council (MRC), National Institute for Medical Research, The Ridgeway, Mill Hill, London, NW71AA, UK
| | - Tobias Bollenbach
- Institute of Science and Technology (IST) Austria, Am Campus 1, A - 3400 Klosterneuburg, Austria
| | - Ana Ribeiro
- Medical Research Council (MRC), National Institute for Medical Research, The Ridgeway, Mill Hill, London, NW71AA, UK
| | - Helena Pérez Valle
- Medical Research Council (MRC), National Institute for Medical Research, The Ridgeway, Mill Hill, London, NW71AA, UK. Imperial College London, UK
| | - Robin Lovell-Badge
- Medical Research Council (MRC), National Institute for Medical Research, The Ridgeway, Mill Hill, London, NW71AA, UK. Department of Biochemistry, The University of Hong Kong, 3/F Laboratory Block, Faculty of Medicine Building, 21 Sassoon Road, Hong Kong. Division of Biosciences, Faculty of Life Sciences, University College London, UK
| | - Vasso Episkopou
- Division of Brain Sciences, Faculty of Medicine, Imperial College London, UK
| | - James Briscoe
- Medical Research Council (MRC), National Institute for Medical Research, The Ridgeway, Mill Hill, London, NW71AA, UK.
| |
Collapse
|
203
|
Houlihan SL, Feng Y. The scaffold protein Nde1 safeguards the brain genome during S phase of early neural progenitor differentiation. eLife 2014; 3:e03297. [PMID: 25245017 PMCID: PMC4170211 DOI: 10.7554/elife.03297] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 09/01/2014] [Indexed: 12/14/2022] Open
Abstract
Successfully completing the S phase of each cell cycle ensures genome integrity. Impediment of DNA replication can lead to DNA damage and genomic disorders. In this study, we show a novel function for NDE1, whose mutations cause brain developmental disorders, in safeguarding the genome through S phase during early steps of neural progenitor fate restrictive differentiation. Nde1 mutant neural progenitors showed catastrophic DNA double strand breaks concurrent with the DNA replication. This evoked DNA damage responses, led to the activation of p53-dependent apoptosis, and resulted in the reduction of neurons in cortical layer II/III. We discovered a nuclear pool of Nde1, identified the interaction of Nde1 with cohesin and its associated chromatin remodeler, and showed that stalled DNA replication in Nde1 mutants specifically occurred in mid-late S phase at heterochromatin domains. These findings suggest that NDE1-mediated heterochromatin replication is indispensible for neuronal differentiation, and that the loss of NDE1 function may lead to genomic neurological disorders.
Collapse
Affiliation(s)
- Shauna L Houlihan
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, United States
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, United States
- Driskill Graduate Program, Northwestern University Feinberg School of Medicine, Chicago, United States
| | - Yuanyi Feng
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, United States
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, United States
| |
Collapse
|
204
|
Gonsalvez DG, Li-Yuen-Fong M, Cane KN, Stamp LA, Young HM, Anderson CR. Different neural crest populations exhibit diverse proliferative behaviors. Dev Neurobiol 2014; 75:287-301. [DOI: 10.1002/dneu.22229] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 08/16/2014] [Accepted: 09/02/2014] [Indexed: 01/02/2023]
Affiliation(s)
- David G. Gonsalvez
- Department of Anatomy and Neuroscience; University of Melbourne; Victoria 3010 Australia
| | - Mathew Li-Yuen-Fong
- Department of Anatomy and Neuroscience; University of Melbourne; Victoria 3010 Australia
| | - Kylie N. Cane
- Department of Anatomy and Neuroscience; University of Melbourne; Victoria 3010 Australia
| | - Lincon A. Stamp
- Department of Anatomy and Neuroscience; University of Melbourne; Victoria 3010 Australia
| | - Heather M. Young
- Department of Anatomy and Neuroscience; University of Melbourne; Victoria 3010 Australia
| | - Colin R. Anderson
- Department of Anatomy and Neuroscience; University of Melbourne; Victoria 3010 Australia
| |
Collapse
|
205
|
Orosco LA, Ross AP, Cates SL, Scott SE, Wu D, Sohn J, Pleasure D, Pleasure SJ, Adamopoulos IE, Zarbalis KS. Loss of Wdfy3 in mice alters cerebral cortical neurogenesis reflecting aspects of the autism pathology. Nat Commun 2014; 5:4692. [PMID: 25198012 PMCID: PMC4159772 DOI: 10.1038/ncomms5692] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 07/15/2014] [Indexed: 01/07/2023] Open
Abstract
Autism spectrum disorders (ASDs) are complex and heterogeneous developmental disabilities affecting an ever-increasing number of children worldwide. The diverse manifestations and complex, largely genetic aetiology of ASDs pose a major challenge to the identification of unifying neuropathological features. Here we describe the neurodevelopmental defects in mice that carry deleterious alleles of the Wdfy3 gene, recently recognized as causative in ASDs. Loss of Wdfy3 leads to a regionally enlarged cerebral cortex resembling early brain overgrowth described in many children on the autism spectrum. In addition, affected mouse mutants display migration defects of cortical projection neurons, a recognized cause of epilepsy, which is significantly comorbid with autism. Our analysis of affected mouse mutants defines an important role for Wdfy3 in regulating neural progenitor divisions and neural migration in the developing brain. Furthermore, Wdfy3 is essential for cerebral expansion and functional organization while its loss-of-function results in pathological changes characteristic of ASDs.
Collapse
Affiliation(s)
- Lori A Orosco
- 1] Department of Pathology and Laboratory Medicine, University of California at Davis, Sacramento, California 95817, USA [2] Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Northern California, 2425 Stockton Boulevard, Sacramento, California 95817, USA
| | - Adam P Ross
- 1] Department of Pathology and Laboratory Medicine, University of California at Davis, Sacramento, California 95817, USA [2] Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Northern California, 2425 Stockton Boulevard, Sacramento, California 95817, USA
| | - Staci L Cates
- 1] Department of Pathology and Laboratory Medicine, University of California at Davis, Sacramento, California 95817, USA [2] Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Northern California, 2425 Stockton Boulevard, Sacramento, California 95817, USA
| | - Sean E Scott
- 1] Department of Pathology and Laboratory Medicine, University of California at Davis, Sacramento, California 95817, USA [2] Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Northern California, 2425 Stockton Boulevard, Sacramento, California 95817, USA
| | - Dennis Wu
- 1] Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Northern California, 2425 Stockton Boulevard, Sacramento, California 95817, USA [2] Department of Internal Medicine, Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, California 95616, USA
| | - Jiho Sohn
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Northern California, 2425 Stockton Boulevard, Sacramento, California 95817, USA
| | - David Pleasure
- 1] Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Northern California, 2425 Stockton Boulevard, Sacramento, California 95817, USA [2] Departments of Neurology and Pediatrics, University of California at Davis, Sacramento, California 95817, USA
| | - Samuel J Pleasure
- Department of Neurology, Programs in Neuroscience, Developmental and Stem Cell Biology, UCSF Institute for Regeneration Medicine, University of California at San Francisco, Sandler Neurosciences Center, Box 3206, 675 Nelson Rising Lane, Room 214, San Francisco, California 94158, USA
| | - Iannis E Adamopoulos
- 1] Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Northern California, 2425 Stockton Boulevard, Sacramento, California 95817, USA [2] Department of Internal Medicine, Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, California 95616, USA
| | - Konstantinos S Zarbalis
- 1] Department of Pathology and Laboratory Medicine, University of California at Davis, Sacramento, California 95817, USA [2] Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Northern California, 2425 Stockton Boulevard, Sacramento, California 95817, USA
| |
Collapse
|
206
|
Aguiar DP, Sghari S, Creuzet S. The facial neural crest controls fore- and midbrain patterning by regulating Foxg1 expression through Smad1 activity. Development 2014; 141:2494-505. [PMID: 24917504 DOI: 10.1242/dev.101790] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The facial neural crest (FNC), a pluripotent embryonic structure forming craniofacial structures, controls the activity of brain organisers and stimulates cerebrum growth. To understand how the FNC conveys its trophic effect, we have studied the role of Smad1, which encodes an intracellular transducer, to which multiple signalling pathways converge, in the regulation of Foxg1. Foxg1 is a transcription factor essential for telencephalic specification, the mutation of which leads to microcephaly and mental retardation. Smad1 silencing, based on RNA interference (RNAi), was performed in pre-migratory FNC cells. Soon after electroporation of RNAi molecules, Smad1 inactivation abolished the expression of Foxg1 in the chick telencephalon, resulting in dramatic microcephaly and partial holoprosencephaly. In addition, the depletion of Foxg1 activity altered the expression Otx2 and Foxa2 in di/mesencephalic neuroepithelium. However, when mutated forms of Smad1 mediating Fgf and Wnt signalling were transfected into FNC cells, these defects were overcome. We also show that, downstream of Smad1 activity, Dkk1, a Wnt antagonist produced by the FNC, initiated the specification of the telencephalon by regulating Foxg1 activity. Additionally, the activity of Cerberus in FNC-derived mesenchyme synergised with Dkk1 to control Foxg1 expression and maintain the balance between Otx2 and Foxa2.
Collapse
Affiliation(s)
- Diego P Aguiar
- Institut de Neurobiologie, Laboratoire de Neurobiologie et Développement, Avenue de la Terrasse, Gif-sur-Yvette 91198, France
| | - Soufien Sghari
- Institut de Neurobiologie, Laboratoire de Neurobiologie et Développement, Avenue de la Terrasse, Gif-sur-Yvette 91198, France
| | - Sophie Creuzet
- Institut de Neurobiologie, Laboratoire de Neurobiologie et Développement, Avenue de la Terrasse, Gif-sur-Yvette 91198, France
| |
Collapse
|
207
|
Kumamoto T, Hanashima C. Neuronal subtype specification in establishing mammalian neocortical circuits. Neurosci Res 2014; 86:37-49. [PMID: 25019611 DOI: 10.1016/j.neures.2014.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 06/21/2014] [Accepted: 06/23/2014] [Indexed: 11/28/2022]
Abstract
The functional integrity of the neocortical circuit relies on the precise production of diverse neuron populations and their assembly during development. In recent years, extensive progress has been made in the understanding of the mechanisms that control differentiation of each neuronal type within the neocortex. In this review, we address how the elaborate neocortical cytoarchitecture is established from a simple neuroepithelium based on recent studies examining the spatiotemporal mechanisms of neuronal subtype specification. We further discuss the critical events that underlie the conversion of the stem amniotes cerebrum to a mammalian-type neocortex, and extend these key findings in the light of mammalian evolution to understand how the neocortex in humans evolved from common ancestral mammals.
Collapse
Affiliation(s)
- Takuma Kumamoto
- Laboratory for Neocortical Development, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Carina Hanashima
- Laboratory for Neocortical Development, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan; Department of Biology, Graduate School of Science, Kobe University, Kobe 657-8501, Japan.
| |
Collapse
|
208
|
Hellbach N, Weise SC, Vezzali R, Wahane SD, Heidrich S, Roidl D, Pruszak J, Esser JS, Vogel T. Neural deletion of Tgfbr2 impairs angiogenesis through an altered secretome. Hum Mol Genet 2014; 23:6177-90. [PMID: 24990151 PMCID: PMC4222361 DOI: 10.1093/hmg/ddu338] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Simultaneous generation of neural cells and that of the nutrient-supplying vasculature during brain development is called neurovascular coupling. We report on a transgenic mouse with impaired transforming growth factor β (TGFβ)-signalling in forebrain-derived neural cells using a Foxg1-cre knock-in to drive the conditional knock-out of the Tgfbr2. Although the expression of FOXG1 is assigned to neural progenitors and neurons of the telencephalon, Foxg1cre/+;Tgfbr2flox/flox (Tgfbr2-cKO) mutants displayed intracerebral haemorrhage. Blood vessels exhibited an atypical, clustered appearance were less in number and displayed reduced branching. Vascular endothelial growth factor (VEGF) A, insulin-like growth factor (IGF) 1, IGF2, TGFβ, inhibitor of DNA binding (ID) 1, thrombospondin (THBS) 2, and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) 1 were altered in either expression levels or tissue distribution. Accordingly, human umbilical vein endothelial cells (HUVEC) displayed branching defects after stimulation with conditioned medium (CM) that was derived from primary neural cultures of the ventral and dorsal telencephalon of Tgfbr2-cKO. Supplementing CM of Tgfbr2-cKO with VEGFA rescued these defects, but application of TGFβ aggravated them. HUVEC showed reduced migration towards CM of mutants compared with controls. Supplementing the CM with growth factors VEGFA, fibroblast growth factor (FGF) 2 and IGF1 partially restored HUVEC migration. In contrast, TGFβ supplementation further impaired migration of HUVEC. We observed differences along the dorso-ventral axis of the telencephalon with regard to the impact of these factors on the phenotype. Together these data establish a TGFBR2-dependent molecular crosstalk between neural and endothelial cells during brain vessel development. These findings will be useful to further elucidate neurovascular interaction in general and to understand pathologies of the blood vessel system such as intracerebral haemorrhages, hereditary haemorrhagic telangiectasia, Alzheimeŕs disease, cerebral amyloid angiopathy or tumour biology.
Collapse
Affiliation(s)
- Nicole Hellbach
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, University of Freiburg, 79104 Freiburg, Germany, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Stefan C Weise
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, University of Freiburg, 79104 Freiburg, Germany, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Riccardo Vezzali
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, University of Freiburg, 79104 Freiburg, Germany, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Shalaka D Wahane
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, University of Freiburg, 79104 Freiburg, Germany, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Stefanie Heidrich
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Deborah Roidl
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Jan Pruszak
- Emmy Noether-Group for Stem Cell Biology, Department of Molecular Embryology, Institute of Anatomy and Cell Biology, University of Freiburg, 79104 Freiburg, Germany, Center for Biological Signaling Studies (BIOSS), University of Freiburg, 79104 Freiburg, Germany and
| | - Jennifer S Esser
- Department of Cardiology and Angiology I, University Heart Center Freiburg, 79106 Freiburg, Germany
| | - Tanja Vogel
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, University of Freiburg, 79104 Freiburg, Germany,
| |
Collapse
|
209
|
Woldehawariat G, Martinez PE, Hauser P, Hoover DM, Drevets WWC, McMahon FJ. Corpus callosum size is highly heritable in humans, and may reflect distinct genetic influences on ventral and rostral regions. PLoS One 2014; 9:e99980. [PMID: 24968245 PMCID: PMC4072678 DOI: 10.1371/journal.pone.0099980] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 05/21/2014] [Indexed: 01/17/2023] Open
Abstract
Anatomical differences in the corpus callosum have been found in various psychiatric disorders, but data on the genetic contributions to these differences have been limited. The current study used morphometric MRI data to assess the heritability of corpus callosum size and the genetic correlations among anatomical sub-regions of the corpus callosum among individuals with and without mood disorders. The corpus callosum (CC) was manually segmented at the mid-sagittal plane in 42 women (healthy, n = 14; major depressive disorder, n = 15; bipolar disorder, n = 13) and their 86 child or adolescent offspring. Four anatomical sub-regions (CC-genu, CC2, CC3 and CC-splenium) and total CC were measured and analyzed. Heritability and genetic correlations were estimated using a variance components method, with adjustment for age, sex, diagnosis, and diagnosis x age, where appropriate. Significant heritability was found for several CC sub-regions (P<0.01), with estimated values ranging from 48% (splenium) to 67% (total CC). There were strong and significant genetic correlations among most sub regions. Correlations between the genu and mid-body, between the genu and total corpus callosum, and between anterior and mid body were all >90%, but no significant genetic correlations were detected between ventral and rostral regions in this sample. Genetic factors play an important role in corpus callosum size among individuals. Distinct genetic factors seem to be involved in caudal and rostral regions, consistent with the divergent functional specialization of these brain areas.
Collapse
Affiliation(s)
- Girma Woldehawariat
- Genetic Basis of Mood & Anxiety Disorders Section, Human Genetics Branch, National Institute of Mental Health, NIH, DHHS, Bethesda, Maryland, United States of America
| | - Pedro E. Martinez
- Section on Behavioral Endocrinology, National Institute of Mental Health, NIH, DHHS, Bethesda, Maryland, Unites States of America
| | - Peter Hauser
- VISN 22 Network Office, Long Beach, California, United States of America
| | - David M. Hoover
- Center for Information Technology, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Wayne W. C. Drevets
- Laureate Institute for Brain Research and the University of Oklahoma College of Medicine, Tulsa, Oklahoma, United States of America
| | - Francis J. McMahon
- Genetic Basis of Mood & Anxiety Disorders Section, Human Genetics Branch, National Institute of Mental Health, NIH, DHHS, Bethesda, Maryland, United States of America
| |
Collapse
|
210
|
Mutations in Eml1 lead to ectopic progenitors and neuronal heterotopia in mouse and human. Nat Neurosci 2014; 17:923-33. [DOI: 10.1038/nn.3729] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 04/29/2014] [Indexed: 12/17/2022]
|
211
|
Fang P, Zeng P, Wang Z, Liu M, Xu W, Dai J, Zhao X, Zhang D, Liang D, Chen X, Shi S, Zhang M, Wang L, Qiao Z, Shi H. Estimated Diversity of Messenger RNAs in Each Murine Spermatozoa and Their Potential Function During Early Zygotic Development1. Biol Reprod 2014; 90:94. [DOI: 10.1095/biolreprod.114.117788] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
212
|
Fish JL, Sklar RS, Woronowicz KC, Schneider RA. Multiple developmental mechanisms regulate species-specific jaw size. Development 2014; 141:674-84. [PMID: 24449843 DOI: 10.1242/dev.100107] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Variation in jaw size during evolution has been crucial for the adaptive radiation of vertebrates, yet variation in jaw size during development is often associated with disease. To test the hypothesis that early developmental events regulating neural crest (NC) progenitors contribute to species-specific differences in size, we investigated mechanisms through which two avian species, duck and quail, achieve their remarkably different jaw size. At early stages, duck exhibit an anterior shift in brain regionalization yielding a shorter, broader, midbrain. We find no significant difference in the total number of pre-migratory NC; however, duck concentrate their pre-migratory NC in the midbrain, which contributes to an increase in size of the post-migratory NC population allocated to the mandibular arch. Subsequent differences in proliferation lead to a progressive increase in size of the duck mandibular arch relative to that of quail. To test the role of pre-migratory NC progenitor number in regulating jaw size, we reduced and augmented NC progenitors. In contrast to previous reports of regeneration by NC precursors, we find that neural fold extirpation results in a loss of NC precursors. Despite this reduction in their numbers, post-migratory NC progenitors compensate, producing a symmetric and normal-sized jaw. Our results suggest that evolutionary modification of multiple aspects of NC cell biology, including NC allocation within the jaw primordia and NC-mediated proliferation, have been important to the evolution of jaw size. Furthermore, our finding of NC post-migratory compensatory mechanisms potentially extends the developmental time frame for treatments of disease or injury associated with NC progenitor loss.
Collapse
Affiliation(s)
- Jennifer L Fish
- University of California, 513 Parnassus Ave, S-1159 San Francisco, CA 94143, USA
| | | | | | | |
Collapse
|
213
|
A novel function for Foxm1 in interkinetic nuclear migration in the developing telencephalon and anxiety-related behavior. J Neurosci 2014; 34:1510-22. [PMID: 24453338 DOI: 10.1523/jneurosci.2549-13.2014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Interkinetic nuclear migration (INM) is a key feature of cortical neurogenesis. INM functions to maximize the output of the neuroepithelium, and more importantly, balance the self-renewal and differentiation of the progenitors. Although INM has been reported to be highly correlated with the cell cycle, little is known about the effects of cell cycle regulators on INM. In this study, by crossing Foxm1(fl/fl) mice with Emx1-Cre line, we report that a conditional disruption of forkhead transcription factor M1 (Foxm1) in dorsal telencephalon results in abnormal cell cycle progression, leading to impaired INM through the downregulation of Cyclin b1 and Cdc25b. The impairment of INM disturbs the synchronization of apical progenitors (APs) and promotes the transition from APs to basal progenitors (BPs) in a cell-autonomous fashion. Moreover, ablation of Foxm1 causes anxiety-related behaviors in adulthood. Thus, this study provides evidence of linkages among the cell cycle regulator Foxm1, INM, and adult behavior.
Collapse
|
214
|
Range R. Specification and positioning of the anterior neuroectoderm in deuterostome embryos. Genesis 2014; 52:222-34. [PMID: 24549984 DOI: 10.1002/dvg.22759] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 02/10/2014] [Accepted: 02/14/2014] [Indexed: 02/01/2023]
Abstract
The molecular mechanisms used by deuterostome embryos (vertebrates, urochordates, cephalochordates, hemichordates, and echinoderms) to specify and then position the anterior neuroectoderm (ANE) along the anterior-posterior axis are incompletely understood. Studies in several deuterostome embryos suggest that the ANE is initially specified by an early, broad regulatory state. Then, a posterior-to-anterior wave of respecification restricts this broad ANE potential to the anterior pole. In vertebrates, sea urchins and hemichordates a posterior-anterior gradient of Wnt/β-catenin signaling plays an essential and conserved role in this process. Recent data collected from the basal deuterostome sea urchin embryo suggests that positioning the ANE to the anterior pole involves more than the Wnt/β-catenin pathway, instead relying on the integration of information from the Wnt/β-catenin, Wnt/JNK, and Wnt/PKC pathways. Moreover, comparison of functional and expression data from the ambulacrarians, invertebrate chordates, and vertebrates strongly suggests that this Wnt network might be an ANE positioning mechanism shared by all deuterostomes.
Collapse
Affiliation(s)
- Ryan Range
- Department of Biological Sciences, Mississippi State University, Mississippi State, Mississippi
| |
Collapse
|
215
|
Hippenmeyer S. Molecular pathways controlling the sequential steps of cortical projection neuron migration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 800:1-24. [PMID: 24243097 DOI: 10.1007/978-94-007-7687-6_1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Coordinated migration of newly-born neurons to their target territories is essential for correct neuronal circuit assembly in the developing brain. Although a cohort of signaling pathways has been implicated in the regulation of cortical projection neuron migration, the precise molecular mechanisms and how a balanced interplay of cell-autonomous and non-autonomous functions of candidate signaling molecules controls the discrete steps in the migration process, are just being revealed. In this chapter, I will focally review recent advances that improved our understanding of the cell-autonomous and possible cell-nonautonomous functions of the evolutionarily conserved LIS1/NDEL1-complex in regulating the sequential steps of cortical projection neuron migration. I will then elaborate on the emerging concept that the Reelin signaling pathway, acts exactly at precise stages in the course of cortical projection neuron migration. Lastly, I will discuss how finely tuned transcriptional programs and downstream effectors govern particular aspects in driving radial migration at discrete stages and how they regulate the precise positioning of cortical projection neurons in the developing cerebral cortex.
Collapse
Affiliation(s)
- Simon Hippenmeyer
- Developmental Neurobiology, IST Austria (Institute of Science and Technology Austria), Am Campus 1, A-3400, Klosterneuburg, Austria,
| |
Collapse
|
216
|
Mort RL, Ford MJ, Sakaue-Sawano A, Lindstrom NO, Casadio A, Douglas AT, Keighren MA, Hohenstein P, Miyawaki A, Jackson IJ. Fucci2a: a bicistronic cell cycle reporter that allows Cre mediated tissue specific expression in mice. Cell Cycle 2014; 13:2681-96. [PMID: 25486356 PMCID: PMC4613862 DOI: 10.4161/15384101.2015.945381] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 06/04/2014] [Indexed: 01/01/2023] Open
Abstract
Markers of cell cycle stage allow estimation of cell cycle dynamics in cell culture and during embryonic development. The Fucci system incorporates genetically encoded probes that highlight G1 and S/G2/M phases of the cell cycle allowing live imaging. However the available mouse models that incorporate Fucci are beset by problems with transgene inactivation, varying expression level, lack of conditional potential and/or the need to maintain separate transgenes-there is no transgenic mouse model that solves all these problems. To address these shortfalls we re-engineered the Fucci system to create 2 bicistronic Fucci variants incorporating both probes fused using the Thosea asigna virus 2A (T2A) self cleaving peptide. We characterize these variants in stable 3T3 cell lines. One of the variants (termed Fucci2a) faithfully recapitulated the nuclear localization and cell cycle stage specific florescence of the original Fucci system. We go on to develop a conditional mouse allele (R26Fucci2aR) carefully designed for high, inducible, ubiquitous expression allowing investigation of cell cycle status in single cell lineages within the developing embryo. We demonstrate the utility of R26Fucci2aR for live imaging by using high resolution confocal microscopy of ex vivo lung, kidney and neural crest development. Using our 3T3 system we describe and validate a method to estimate cell cycle times from relatively short time-lapse sequences that we then apply to our neural crest data. The Fucci2a system and the R26Fucci2aR mouse model are compelling new tools for the investigation of cell cycle dynamics in cell culture and during mouse embryonic development.
Collapse
Key Words
- BrdU, 5-bromo-2′-deoxyuridine
- DAPI, 4′, 6-diamidino-2-phenylindole
- DMEM, Dulbeccos modified eagle medium
- ECACC, European Collection of Cell Cultures
- EMMA, European Mouse Mutant Archive
- FACS, Fluorescence-activated cell sorting
- Fucci
- Fucci, Fluorescent Ubiquitination-based Cell Cycle Indicator
- Fucci2
- Fucci2a
- GMEM, Glasgow minimum essential medium
- IRES, Internal ribosomal entry site
- LIF, leukemia inhibitory factor
- RBDB, Riken Bioresource Center DNA Bank
- T2A, Thosea asigna virus 2A peptide
- cell cycle
- hESC, Human embryonic stem cell
- kidney
- lung
- mAG, Monomeric Azami Green
- mESC, Mouse embryonic stem cell
- mKO2, Monomeric Kusabira Orange
- melanoblast
Collapse
Affiliation(s)
- Richard Lester Mort
- MRC Human Genetics Unit; MRC IGMM; University of Edinburgh; Western General Hospital Edinburgh; Scotland, UK
| | - Matthew Jonathan Ford
- MRC Human Genetics Unit; MRC IGMM; University of Edinburgh; Western General Hospital Edinburgh; Scotland, UK
| | - Asako Sakaue-Sawano
- Laboratory for Cell Function and Dynamics; Advanced Technology Development Group; Brain Science Institute; RIKEN; Wako-city, Saitama, Japan
| | - Nils Olof Lindstrom
- The Roslin Institute; The University of Edinburgh; Easter Bush, Midlothian; Scotland, UK
| | - Angela Casadio
- MRC Human Genetics Unit; MRC IGMM; University of Edinburgh; Western General Hospital Edinburgh; Scotland, UK
| | - Adam Thomas Douglas
- MRC Human Genetics Unit; MRC IGMM; University of Edinburgh; Western General Hospital Edinburgh; Scotland, UK
| | - Margaret Anne Keighren
- MRC Human Genetics Unit; MRC IGMM; University of Edinburgh; Western General Hospital Edinburgh; Scotland, UK
| | - Peter Hohenstein
- MRC Human Genetics Unit; MRC IGMM; University of Edinburgh; Western General Hospital Edinburgh; Scotland, UK
- The Roslin Institute; The University of Edinburgh; Easter Bush, Midlothian; Scotland, UK
| | - Atsushi Miyawaki
- Laboratory for Cell Function and Dynamics; Advanced Technology Development Group; Brain Science Institute; RIKEN; Wako-city, Saitama, Japan
| | - Ian James Jackson
- MRC Human Genetics Unit; MRC IGMM; University of Edinburgh; Western General Hospital Edinburgh; Scotland, UK
- The Roslin Institute; The University of Edinburgh; Easter Bush, Midlothian; Scotland, UK
| |
Collapse
|
217
|
Nonomura K, Yamaguchi Y, Hamachi M, Koike M, Uchiyama Y, Nakazato K, Mochizuki A, Sakaue-Sawano A, Miyawaki A, Yoshida H, Kuida K, Miura M. Local Apoptosis Modulates Early Mammalian Brain Development through the Elimination of Morphogen-Producing Cells. Dev Cell 2013; 27:621-34. [DOI: 10.1016/j.devcel.2013.11.015] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 11/05/2013] [Accepted: 11/20/2013] [Indexed: 02/07/2023]
|
218
|
Mascelli S, Barla A, Raso A, Mosci S, Nozza P, Biassoni R, Morana G, Huber M, Mircean C, Fasulo D, Noy K, Wittemberg G, Pignatelli S, Piatelli G, Cama A, Garré ML, Capra V, Verri A. Molecular fingerprinting reflects different histotypes and brain region in low grade gliomas. BMC Cancer 2013; 13:387. [PMID: 23947815 PMCID: PMC3765921 DOI: 10.1186/1471-2407-13-387] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 08/06/2013] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Paediatric low-grade gliomas (LGGs) encompass a heterogeneous set of tumours of different histologies, site of lesion, age and gender distribution, growth potential, morphological features, tendency to progression and clinical course. Among LGGs, Pilocytic astrocytomas (PAs) are the most common central nervous system (CNS) tumours in children. They are typically well-circumscribed, classified as grade I by the World Health Organization (WHO), but recurrence or progressive disease occurs in about 10-20% of cases. Despite radiological and neuropathological features deemed as classic are acknowledged, PA may present a bewildering variety of microscopic features. Indeed, tumours containing both neoplastic ganglion and astrocytic cells occur at a lower frequency. METHODS Gene expression profiling on 40 primary LGGs including PAs and mixed glial-neuronal tumours comprising gangliogliomas (GG) and desmoplastic infantile gangliogliomas (DIG) using Affymetrix array platform was performed. A biologically validated machine learning workflow for the identification of microarray-based gene signatures was devised. The method is based on a sparsity inducing regularization algorithm l₁l₂ that selects relevant variables and takes into account their correlation. The most significant genetic signatures emerging from gene-chip analysis were confirmed and validated by qPCR. RESULTS We identified an expression signature composed by a biologically validated list of 15 genes, able to distinguish infratentorial from supratentorial LGGs. In addition, a specific molecular fingerprinting distinguishes the supratentorial PAs from those originating in the posterior fossa. Lastly, within supratentorial tumours, we also identified a gene expression pattern composed by neurogenesis, cell motility and cell growth genes which dichotomize mixed glial-neuronal tumours versus PAs. Our results reinforce previous observations about aberrant activation of the mitogen-activated protein kinase (MAPK) pathway in LGGs, but still point to an active involvement of TGF-beta signaling pathway in the PA development and pick out some hitherto unreported genes worthy of further investigation for the mixed glial-neuronal tumours. CONCLUSIONS The identification of a brain region-specific gene signature suggests that LGGs, with similar pathological features but located at different sites, may be distinguishable on the basis of cancer genetics. Molecular fingerprinting seems to be able to better sub-classify such morphologically heterogeneous tumours and it is remarkable that mixed glial-neuronal tumours are strikingly separated from PAs.
Collapse
Affiliation(s)
- Samantha Mascelli
- Neurosurgery Unit, Istituto Giannina Gaslini, via G. Gaslini 5, 16147, Genoa, Italy
| | - Annalisa Barla
- DISI - Department of Computer Science, Università degli Studi di Genova, Via Dodecaneso, 35-16146, Genoa, Italy
| | - Alessandro Raso
- Neurosurgery Unit, Istituto Giannina Gaslini, via G. Gaslini 5, 16147, Genoa, Italy
| | - Sofia Mosci
- DISI - Department of Computer Science, Università degli Studi di Genova, Via Dodecaneso, 35-16146, Genoa, Italy
| | - Paolo Nozza
- Pathology Unit, Istituto Giannina Gaslini, via G. Gaslini 5, 16147, Genoa, Italy
| | - Roberto Biassoni
- Molecular Medicine Unit, Istituto Giannina Gaslini, via G. Gaslini 5, 16147, Genoa, Italy
| | - Giovanni Morana
- Neuroradiology Unit, Istituto Giannina Gaslini, via G. Gaslini 5, 16147, Genoa, Italy
| | - Martin Huber
- Siemens AG, Corporate Technology, Freyeslebenstr. 1, 91058, Erlangen, Germany
| | - Cristian Mircean
- Siemens AG, Corporate Technology, Freyeslebenstr. 1, 91058, Erlangen, Germany
| | - Daniel Fasulo
- SCR - Siemens Corporate Research, Princeton, NJ, USA
| | - Karin Noy
- SCR - Siemens Corporate Research, Princeton, NJ, USA
| | | | - Sara Pignatelli
- Neuro-oncology Unit, Istituto Giannina Gaslini, via G. Gaslini 5, 16147, Genoa, Italy
| | - Gianluca Piatelli
- Neurosurgery Unit, Istituto Giannina Gaslini, via G. Gaslini 5, 16147, Genoa, Italy
| | - Armando Cama
- Neurosurgery Unit, Istituto Giannina Gaslini, via G. Gaslini 5, 16147, Genoa, Italy
| | - Maria Luisa Garré
- Neuro-oncology Unit, Istituto Giannina Gaslini, via G. Gaslini 5, 16147, Genoa, Italy
| | - Valeria Capra
- Neurosurgery Unit, Istituto Giannina Gaslini, via G. Gaslini 5, 16147, Genoa, Italy
| | - Alessandro Verri
- DISI - Department of Computer Science, Università degli Studi di Genova, Via Dodecaneso, 35-16146, Genoa, Italy
| |
Collapse
|
219
|
Nomura T, Gotoh H, Ono K. Changes in the regulation of cortical neurogenesis contribute to encephalization during amniote brain evolution. Nat Commun 2013; 4:2206. [DOI: 10.1038/ncomms3206] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 06/27/2013] [Indexed: 12/30/2022] Open
|
220
|
Inactivation of mTORC1 in the developing brain causes microcephaly and affects gliogenesis. J Neurosci 2013; 33:7799-810. [PMID: 23637172 DOI: 10.1523/jneurosci.3294-12.2013] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) regulates cell growth in response to various intracellular and extracellular signals. It assembles into two multiprotein complexes: the rapamycin-sensitive mTOR complex 1 (mTORC1) and the rapamycin-insensitive mTORC2. In this study, we inactivated mTORC1 in mice by deleting the gene encoding raptor in the progenitors of the developing CNS. Mice are born but never feed and die within a few hours. The brains deficient for raptor show a microcephaly starting at E17.5 that is the consequence of a reduced cell number and cell size. Changes in cell cycle length during late cortical development and increased cell death both contribute to the reduction in cell number. Neurospheres derived from raptor-deficient brains are smaller, and differentiation of neural progenitors into glia but not into neurons is inhibited. The differentiation defect is paralleled by decreased Stat3 signaling, which is a target of mTORC1 and has been implicated in gliogenesis. Together, our results show that postnatal survival, overall brain growth, and specific aspects of brain development critically depend on mTORC1 function.
Collapse
|
221
|
Pax6 exerts regional control of cortical progenitor proliferation via direct repression of Cdk6 and hypophosphorylation of pRb. Neuron 2013; 78:269-84. [PMID: 23622063 PMCID: PMC3898967 DOI: 10.1016/j.neuron.2013.02.012] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2013] [Indexed: 11/22/2022]
Abstract
The mechanisms by which early spatiotemporal expression patterns of transcription factors such as Pax6 regulate cortical progenitors in a region-specific manner are poorly understood. Pax6 is expressed in a gradient across the developing cortex and is essential for normal corticogenesis. We found that constitutive or conditional loss of Pax6 increases cortical progenitor proliferation by amounts that vary regionally with normal Pax6 levels. We compared the gene expression profiles of equivalent Pax6-expressing progenitors isolated from Pax6+/+ and Pax6−/− cortices and identified many negatively regulated cell-cycle genes, including Cyclins and Cdks. Biochemical assays indicated that Pax6 directly represses Cdk6 expression. Cyclin/Cdk repression inhibits retinoblastoma protein (pRb) phosphorylation, thereby limiting the transcription of genes that directly promote the mechanics of the cell cycle, and we found that Pax6 inhibits pRb phosphorylation and represses genes involved in DNA replication. Our results indicate that Pax6’s modulation of cortical progenitor cell cycles is regional and direct. Pax6 loss increases cortical progenitor proliferation by region-specific amounts The size of this effect correlates directly with normal Pax6 expression levels Expression of many key cell-cycle regulators is increased in the absence of Pax6 Pax6 directly represses Cdk6 expression and controls pRb phosphorylation
Collapse
|
222
|
Fotaki V, Smith R, Pratt T, Price DJ. Foxg1 is required to limit the formation of ciliary margin tissue and Wnt/β-catenin signalling in the developing nasal retina of the mouse. Dev Biol 2013; 380:299-313. [PMID: 23624311 PMCID: PMC3722486 DOI: 10.1016/j.ydbio.2013.04.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 04/15/2013] [Accepted: 04/16/2013] [Indexed: 12/23/2022]
Abstract
The ciliary margin (CM) develops in the peripheral retina and gives rise to the iris and the ciliary body. The Wnt/β-catenin signalling pathway has been implicated in ciliary margin development. Here, we tested the hypothesis that in the developing mouse retina Foxg1 is responsible for suppressing the Wnt/β-catenin pathway and restricting CM development. We showed that there is excess CM tissue in Foxg1−/− null embryos and this expansion is more pronounced in the nasal retina where Foxg1 normally shows its highest expression levels. Results on expression of a reporter allele for Wnt/β-catenin signalling and of Lef1, a target of Wnt/β-catenin signalling, displayed significant upregulation of this pathway in Foxg1−/− nulls at embryonic days 12.5 and 14.5. Interestingly, this upregulation was observed specifically in the nasal retina, where normally very few Wnt-responsive cells are observed. These results indicate a suppressive role of Foxg1 on this signalling pathway. Our results reveal a new role of Foxg1 in limiting CM development in the nasal peripheral retina and add a new molecular player in the developmental network involved in CM specification. Foxg1 is expressed in a nasal-high to temporal-low gradient in developing retina. Ciliary margin expansion is observed nasally in the Foxg1−/− mutant retina. Wnt/β-catenin signalling is upregulated in the Foxg1−/− peripheral retina nasally. A new role of Foxg1 in controlling ciliary margin development is proposed.
Collapse
Affiliation(s)
- Vassiliki Fotaki
- University of Edinburgh, Centre for Integrative Physiology, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK.
| | | | | | | |
Collapse
|
223
|
Functional implications of hippocampal adult neurogenesis in intellectual disabilities. Amino Acids 2013; 45:113-31. [DOI: 10.1007/s00726-013-1489-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 03/15/2013] [Indexed: 12/19/2022]
|
224
|
Liang H, Xiao G, Yin H, Hippenmeyer S, Horowitz JM, Ghashghaei HT. Neural development is dependent on the function of specificity protein 2 in cell cycle progression. Development 2013; 140:552-61. [PMID: 23293287 DOI: 10.1242/dev.085621] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Faithful progression through the cell cycle is crucial to the maintenance and developmental potential of stem cells. Here, we demonstrate that neural stem cells (NSCs) and intermediate neural progenitor cells (NPCs) employ a zinc-finger transcription factor specificity protein 2 (Sp2) as a cell cycle regulator in two temporally and spatially distinct progenitor domains. Differential conditional deletion of Sp2 in early embryonic cerebral cortical progenitors, and perinatal olfactory bulb progenitors disrupted transitions through G1, G2 and M phases, whereas DNA synthesis appeared intact. Cell-autonomous function of Sp2 was identified by deletion of Sp2 using mosaic analysis with double markers, which clearly established that conditional Sp2-null NSCs and NPCs are M phase arrested in vivo. Importantly, conditional deletion of Sp2 led to a decline in the generation of NPCs and neurons in the developing and postnatal brains. Our findings implicate Sp2-dependent mechanisms as novel regulators of cell cycle progression, the absence of which disrupts neurogenesis in the embryonic and postnatal brain.
Collapse
Affiliation(s)
- Huixuan Liang
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
| | | | | | | | | | | |
Collapse
|
225
|
Larue L, de Vuyst F, Delmas V. Modeling melanoblast development. Cell Mol Life Sci 2013; 70:1067-79. [PMID: 22915137 PMCID: PMC11113344 DOI: 10.1007/s00018-012-1112-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 07/17/2012] [Accepted: 07/30/2012] [Indexed: 12/14/2022]
Abstract
Melanoblasts are a particular type of cell that displays extensive cellular proliferation during development to contribute to the skin. There are only a few melanoblast founders, initially located just dorsal to the neural tube, and they sequentially colonize the dermis, epidermis, and hair follicles. In each compartment, melanoblasts are exposed to a wide variety of developmental cues that regulate their expansion. The colonization of the dermis and epidermis by melanoblasts involves substantial proliferation to generate thousands of cells or more from a few founders within a week of development. This review addresses the cellular and molecular events occurring during melanoblast development. We focus on intrinsic and extrinsic factors that control melanoblast proliferation. We also present a robust mathematical model for estimating the doubling-time of dermal and epidermal melanoblasts for all coat color phenotypes from black to white.
Collapse
Affiliation(s)
- Lionel Larue
- Institut Curie, Centre de Recherche, Developmental Genetics of Melanocytes, 91405, Orsay, France.
| | | | | |
Collapse
|
226
|
Song M, Mohamad O, Chen D, Yu SP. Coordinated development of voltage-gated Na+ and K+ currents regulates functional maturation of forebrain neurons derived from human induced pluripotent stem cells. Stem Cells Dev 2013; 22:1551-63. [PMID: 23259973 DOI: 10.1089/scd.2012.0556] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Like embryonic stem (ES) cells, human induced pluripotent stem (hiPS) cells can differentiate into neuronal cells. However, it is unclear how their exquisite neuronal function is electrophysiologically coordinated during differentiation and whether they are functionally identical to human ES cell-derived neurons. In this study, we differentiated hiPS and ES cells into pyramidal-like neurons and conducted electrophysiological characterization over the 4-week terminal differentiation period. The human neuron-like cells express forebrain pyramidal cell markers NeuN, neurofilament, the microtubule-associated protein 2 (MAP2), the paired box protein Pax-6 (PAX6), Tuj1, and the forkhead box protein G1 (FoxG1). The size of developing neurons increased continuously during the 4-week culture, and cell-resting membrane potentials (RMPs) underwent a negative shift from -40 to -70 mV. Expression of the muscarinic receptor-modulated K(+) currents (IM) participated in the development of cell RMPs and controlled excitability. Immature neurons at week 1 could only fire abortive action potentials (APs) and the frequency of AP firing progressively increased with neuronal maturation. Interestingly, the developmental change of voltage-gated Na(+) current (INa) did not correlate with the change in the AP firing frequency. On the other hand, the transient outward K(+) current (IA), but not the delayed rectifier current (IK) contributed to the high frequency firing of APs. Synaptic activities were observed throughout the 4-week development. These morphological and electrophysiological features were almost identical between iPS and ES cell-derived neurons. This is the first systematic investigation showing functional evidence that hiPS cell-derived neurons possess similar neuronal activities as ES cell-derived neurons. These data support that iPS cell-derived neural progenitor cells have the potential for replacing lost neurons in cell-based therapy.
Collapse
Affiliation(s)
- Mingke Song
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | |
Collapse
|
227
|
Beccari L, Marco-Ferreres R, Bovolenta P. The logic of gene regulatory networks in early vertebrate forebrain patterning. Mech Dev 2012; 130:95-111. [PMID: 23111324 DOI: 10.1016/j.mod.2012.10.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 10/09/2012] [Indexed: 01/19/2023]
Abstract
The vertebrate forebrain or prosencephalon is patterned at the beginning of neurulation into four major domains: the telencephalic, hypothalamic, retinal and diencephalic anlagen. These domains will then give rise to the majority of the brain structures involved in sensory integration and the control of higher intellectual and homeostatic functions. Understanding how forebrain pattering arises has thus attracted the interest of developmental neurobiologists for decades. As a result, most of its regulators have been identified and their hierarchical relationship is now the object of active investigation. Here, we summarize the main morphogenetic pathways and transcription factors involved in forebrain specification and propose the backbone of a possible gene regulatory network (GRN) governing its specification, taking advantage of the GRN principles elaborated by pioneer studies in simpler organisms. We will also discuss this GRN and its operational logic in the context of the remarkable morphological and functional diversification that the forebrain has undergone during evolution.
Collapse
Affiliation(s)
- Leonardo Beccari
- Centro de Biología Molecular "Severo Ochoa", CSIC-UAM, c/Nicolas Cabrera, 1, Madrid 28049, Spain
| | | | | |
Collapse
|
228
|
Measurement and 3D-visualization of cell-cycle length using double labelling with two thymidine analogues applied in early heart development. PLoS One 2012; 7:e47719. [PMID: 23091641 PMCID: PMC3473012 DOI: 10.1371/journal.pone.0047719] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 09/14/2012] [Indexed: 11/19/2022] Open
Abstract
Organ development is a complex spatial process in which local differences in cell proliferation rate play a key role. Understanding this role requires the measurement of the length of the cell cycle at every position of the three-dimensional (3D) structure. This measurement can be accomplished by exposing the developing embryo to two different thymidine analogues for two different durations immediately followed by tissue fixation. This paper presents a method and a dedicated computer program to measure the resulting labelling indices and subsequently calculate and visualize local cell cycle lengths within the 3D morphological context of a developing organ. By applying this method to the developing heart, we show a large difference in cell cycle lengths between the early heart tube and the adjacent mesenchyme of the pericardial wall. Later in development, a local increase in cell size was found to be associated with a decrease in cell cycle length in the region where the chamber myocardium starts to develop. The combined application of halogenated-thymidine double exposure and image processing enables the automated study of local cell cycle parameters in single specimens in a full 3D context. It can be applied in a wide range of research fields ranging from embryonic development to tissue regeneration and cancer research.
Collapse
|
229
|
Miyoshi G, Fishell G. Dynamic FoxG1 expression coordinates the integration of multipolar pyramidal neuron precursors into the cortical plate. Neuron 2012; 74:1045-58. [PMID: 22726835 DOI: 10.1016/j.neuron.2012.04.025] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2012] [Indexed: 01/20/2023]
Abstract
Pyramidal cells of the cerebral cortex are born in the ventricular zone and migrate through the intermediate zone to enter into the cortical plate. In the intermediate zone, these migrating precursors move tangentially and initiate the extension of their axons by transiently adopting a characteristic multipolar morphology. We observe that expression of the forkhead transcription factor FoxG1 is dynamically regulated during this transitional period. By utilizing conditional genetic strategies, we show that the downregulation of FoxG1 at the beginning of the multipolar cell phase induces Unc5D expression, the timing of which ultimately determines the laminar identity of pyramidal neurons. In addition, we demonstrate that the re-expression of FoxG1 is required for cells to transit out of the multipolar cell phase and to enter into the cortical plate. Thus, the dynamic expression of FoxG1 during migration within the intermediate zone is essential for the proper assembly of the cerebral cortex.
Collapse
Affiliation(s)
- Goichi Miyoshi
- NYU Neuroscience Institute, Department of Physiology and Neuroscience, Smilow Research Center, New York University School of Medicine, 522 First Avenue, New York, NY 10016, USA
| | | |
Collapse
|
230
|
Ellaway CJ, Ho G, Bettella E, Knapman A, Collins F, Hackett A, McKenzie F, Darmanian A, Peters GB, Fagan K, Christodoulou J. 14q12 microdeletions excluding FOXG1 give rise to a congenital variant Rett syndrome-like phenotype. Eur J Hum Genet 2012; 21:522-7. [PMID: 22968132 DOI: 10.1038/ejhg.2012.208] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Rett syndrome is a clinically defined neurodevelopmental disorder almost exclusively affecting females. Usually sporadic, Rett syndrome is caused by mutations in the X-linked MECP2 gene in ∼90-95% of classic cases and 40-60% of individuals with atypical Rett syndrome. Mutations in the CDKL5 gene have been associated with the early-onset seizure variant of Rett syndrome and mutations in FOXG1 have been associated with the congenital Rett syndrome variant. We report the clinical features and array CGH findings of three atypical Rett syndrome patients who had severe intellectual impairment, early-onset developmental delay, postnatal microcephaly and hypotonia. In addition, the females had a seizure disorder, agenesis of the corpus callosum and subtle dysmorphism. All three were found to have an interstitial deletion of 14q12. The deleted region in common included the PRKD1 gene but not the FOXG1 gene. Gene expression analysis suggested a decrease in FOXG1 levels in two of the patients. Screening of 32 atypical Rett syndrome patients did not identify any pathogenic mutations in the PRKD1 gene, although a previously reported frameshift mutation affecting FOXG1 (c.256dupC, p.Gln86ProfsX35) was identified in a patient with the congenital Rett syndrome variant. There is phenotypic overlap between congenital Rett syndrome variants with FOXG1 mutations and the clinical presentation of our three patients with this 14q12 microdeletion, not encompassing the FOXG1 gene. We propose that the primary defect in these patients is misregulation of the FOXG1 gene rather than a primary abnormality of PRKD1.
Collapse
Affiliation(s)
- Carolyn J Ellaway
- Western Sydney Genetics Program, Children's Hospital at Westmead, Sydney, New South Wales, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
231
|
Disrupted ERK signaling during cortical development leads to abnormal progenitor proliferation, neuronal and network excitability and behavior, modeling human neuro-cardio-facial-cutaneous and related syndromes. J Neurosci 2012; 32:8663-77. [PMID: 22723706 DOI: 10.1523/jneurosci.1107-12.2012] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Genetic disorders arising from copy number variations in the ERK (extracellular signal-regulated kinase) MAP (mitogen-activated protein) kinases or mutations in their upstream regulators that result in neuro-cardio-facial-cutaneous syndromes are associated with developmental abnormalities, cognitive deficits, and autism. We developed murine models of these disorders by deleting the ERKs at the beginning of neurogenesis and report disrupted cortical progenitor generation and proliferation, which leads to altered cytoarchitecture of the postnatal brain in a gene-dose-dependent manner. We show that these changes are due to ERK-dependent dysregulation of cyclin D1 and p27(Kip1), resulting in cell cycle elongation, favoring neurogenic over self-renewing divisions. The precocious neurogenesis causes premature progenitor pool depletion, altering the number and distribution of pyramidal neurons. Importantly, loss of ERK2 alters the intrinsic excitability of cortical neurons and contributes to perturbations in global network activity. These changes are associated with elevated anxiety and impaired working and hippocampal-dependent memory in these mice. This study provides a novel mechanistic insight into the basis of cortical malformation which may provide a potential link to cognitive deficits in individuals with altered ERK activity.
Collapse
|
232
|
Danesin C, Houart C. A Fox stops the Wnt: implications for forebrain development and diseases. Curr Opin Genet Dev 2012; 22:323-30. [DOI: 10.1016/j.gde.2012.05.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 04/17/2012] [Accepted: 05/15/2012] [Indexed: 10/28/2022]
|
233
|
Abstract
During embryonic development, cells must divide to produce appropriate numbers, but later must exit the cell cycle to allow differentiation. How these processes of proliferation and differentiation are co-ordinated during embryonic development has been poorly understood until recently. However, a number of studies have now given an insight into how the cell cycle machinery, including cyclins, CDKs (cyclin-dependent kinases), CDK inhibitors and other cell cycle regulators directly influence mechanisms that control cell fate and differentiation. Conversely, examples are emerging of transcriptional regulators that are better known for their role in driving the differentiated phenotype, which also play complementary roles in controlling cell cycle progression. The present review will summarise our current understanding of the mechanisms co-ordinating the cell cycle and differentiation in the developing nervous system, where these links have been, perhaps, most extensively studied.
Collapse
|
234
|
Wei B, Huang Z, He S, Sun C, You Y, Liu F, Yang Z. The onion skin-like organization of the septum arises from multiple embryonic origins to form multiple adult neuronal fates. Neuroscience 2012; 222:110-23. [PMID: 22820053 DOI: 10.1016/j.neuroscience.2012.07.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 07/09/2012] [Accepted: 07/10/2012] [Indexed: 01/28/2023]
Abstract
In the past several decades, tremendous progress has been achieved through developmental studies of the central nervous system structures such as the cerebral cortex. The septum, which receives reciprocal connections from a variety of brain structures, contains diverse projection neurons but few interneurons. However, the mechanisms underlying its development remain poorly understood. Here we show that the septum is organized into an onion skin-like structure composed of five groups of neurons. These neurons are parvalbumin, choline acetyltransferase, neuronal nitric oxide synthase, calretinin and calbindin immunoreactive. Using the BrdU birth-dating method, we found that these five groups of neurons in the septum are grossly generated following an outside-in pattern. Interestingly, the distinct molecular identities of these neuronal subtypes correspond to their heterogeneous subpallial origins. Using three specific transgenic mouse lines and focal in utero electroporation of Cre-reporter plasmid, we showed that septal neurons originate from not only local progenitor regions but also neighboring progenitor regions including the medial ganglionic eminence and preoptic area. Thus, the neuronal diversity of the septum is achieved through both temporal and spatial control. Our results also suggest that multiple neuronal subtypes arrive to the septum through both radial and tangential migration. Based on these findings, we proposed a novel developmental model involving multiple spatial-temporal origins of septal neurons. This study presents new perspectives for comprehensively exploring septal functions in brain circuits.
Collapse
Affiliation(s)
- B Wei
- Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, People's Republic of China.
| | | | | | | | | | | | | |
Collapse
|
235
|
Carlin D, Sepich D, Grover VK, Cooper MK, Solnica-Krezel L, Inbal A. Six3 cooperates with Hedgehog signaling to specify ventral telencephalon by promoting early expression of Foxg1a and repressing Wnt signaling. Development 2012; 139:2614-24. [PMID: 22736245 PMCID: PMC3383232 DOI: 10.1242/dev.076018] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2012] [Indexed: 01/18/2023]
Abstract
Six3 exerts multiple functions in the development of anterior neural tissue of vertebrate embryos. Whereas complete loss of Six3 function in the mouse results in failure of forebrain formation, its hypomorphic mutations in human and mouse can promote holoprosencephaly (HPE), a forebrain malformation that results, at least in part, from abnormal telencephalon development. However, the roles of Six3 in telencephalon patterning and differentiation are not well understood. To address the role of Six3 in telencephalon development, we analyzed zebrafish embryos deficient in two out of three Six3-related genes, six3b and six7, representing a partial loss of Six3 function. We found that telencephalon forms in six3b;six7-deficient embryos; however, ventral telencephalic domains are smaller and dorsal domains are larger. Decreased cell proliferation or excess apoptosis cannot account for the ventral deficiency. Instead, six3b and six7 are required during early segmentation for specification of ventral progenitors, similar to the role of Hedgehog (Hh) signaling in telencephalon development. Unlike in mice, we observe that Hh signaling is not disrupted in embryos with reduced Six3 function. Furthermore, six3b overexpression is sufficient to compensate for loss of Hh signaling in isl1- but not nkx2.1b-positive cells, suggesting a novel Hh-independent role for Six3 in telencephalon patterning. We further find that Six3 promotes ventral telencephalic fates through transient regulation of foxg1a expression and repression of the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Dan Carlin
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Diane Sepich
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Vandana K. Grover
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Michael K. Cooper
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lilianna Solnica-Krezel
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Adi Inbal
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA
- Department of Medical Neurobiology, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| |
Collapse
|
236
|
Yip DJ, Corcoran CP, Alvarez-Saavedra M, DeMaria A, Rennick S, Mears AJ, Rudnicki MA, Messier C, Picketts DJ. Snf2l regulates Foxg1-dependent progenitor cell expansion in the developing brain. Dev Cell 2012; 22:871-8. [PMID: 22516202 DOI: 10.1016/j.devcel.2012.01.020] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 01/11/2012] [Accepted: 01/26/2012] [Indexed: 01/21/2023]
Abstract
Balancing progenitor cell self-renewal and differentiation is essential for brain development and is regulated by the activity of chromatin remodeling complexes. Nevertheless, linking chromatin changes to specific pathways that control cortical histogenesis remains a challenge. Here we identify a genetic interaction between the chromatin remodeler Snf2l and Foxg1, a key regulator of neurogenesis. Snf2l mutant mice exhibit forebrain hypercellularity arising from increased Foxg1 expression, increased progenitor cell expansion, and delayed differentiation. We demonstrate that Snf2l binds to the Foxg1 locus at midneurogenesis and that the phenotype is rescued by reducing Foxg1 dosage, thus revealing that Snf2l and Foxg1 function antagonistically to regulate brain size.
Collapse
Affiliation(s)
- Darren J Yip
- Regenerative Medicine Program, Ottawa Hospital Research Institute, and Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
237
|
Miranda RC. MicroRNAs and Fetal Brain Development: Implications for Ethanol Teratology during the Second Trimester Period of Neurogenesis. Front Genet 2012; 3:77. [PMID: 22623924 PMCID: PMC3353139 DOI: 10.3389/fgene.2012.00077] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 04/23/2012] [Indexed: 11/29/2022] Open
Abstract
Maternal ethanol consumption during pregnancy can lead to a stereotypic cluster of fetal craniofacial, cardiovascular, skeletal, and neurological deficits that are collectively termed the fetal alcohol spectrum disorder (FASD). Fetal ethanol exposure is a leading non-genetic cause of mental retardation. Mechanisms underlying the etiology of ethanol teratology are varied and complex. This review will focus on the developing brain as an important and vulnerable ethanol target. Near the end of the first trimester, and during the second trimester, fetal neural stem cells (NSCs) produce most of the neurons of the adult brain, and ethanol has been shown to influence NSC renewal and maturation. We will discuss the neural developmental and teratological implications of the biogenesis and function of microRNAs (miRNAs), a class of small non-protein-coding RNAs that control the expression of gene networks by translation repression. A small but growing body of research has identified ethanol-sensitive miRNAs at different stages of NSC and brain maturation. While many miRNAs appear to be vulnerable to ethanol at specific developmental stages, a few, like the miR-9 family, appear to exhibit broad vulnerability to ethanol across multiple stages of NSC differentiation. An assessment of the regulation and function of these miRNAs provides important clues about the mechanisms that underlie fetal vulnerability to alterations in the maternal-fetal environment and yields insights into the genesis of FASD.
Collapse
|
238
|
Pauly MC, Piroth T, Döbrössy M, Nikkhah G. Restoration of the striatal circuitry: from developmental aspects toward clinical applications. Front Cell Neurosci 2012; 6:16. [PMID: 22529778 PMCID: PMC3329876 DOI: 10.3389/fncel.2012.00016] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 03/23/2012] [Indexed: 12/20/2022] Open
Abstract
In the basal ganglia circuitry, the striatum is a highly complex structure coordinating motor and cognitive functions and it is severely affected in Huntington's disease (HD) patients. Transplantation of fetal ganglionic eminence (GE) derived precursor cells aims to restore neural circuitry in the degenerated striatum of HD patients. Pre-clinical transplantation in genetic and lesion HD animal models has increased our knowledge of graft vs. host interactions, and clinical studies have been shown to successfully reduce motor and cognitive effects caused by the disease. Investigating the molecular mechanisms of striatal neurogenesis is a key research target, since novel strategies aim on generating striatal neurons by differentiating embryonic stem cells or by reprogramming somatic cells as alternative cell source for neural transplantation.
Collapse
Affiliation(s)
- Marie-Christin Pauly
- Division of Stereotactic Neurosurgery, Department of General Neurosurgery, University Freiburg - Medical Center Freiburg im Breisgau, Germany
| | | | | | | |
Collapse
|
239
|
Mairet-Coello G, Tury A, Van Buskirk E, Robinson K, Genestine M, DiCicco-Bloom E. p57(KIP2) regulates radial glia and intermediate precursor cell cycle dynamics and lower layer neurogenesis in developing cerebral cortex. Development 2012; 139:475-87. [PMID: 22223678 DOI: 10.1242/dev.067314] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
During cerebral cortex development, precise control of precursor cell cycle length and cell cycle exit is required for balanced precursor pool expansion and layer-specific neurogenesis. Here, we defined the roles of cyclin-dependent kinase inhibitor (CKI) p57(KIP2), an important regulator of G1 phase, using deletion mutant mice. Mutant mice displayed macroencephaly associated with cortical hyperplasia during late embryogenesis and postnatal development. Embryonically, proliferation of radial glial cells (RGC) and intermediate precursors (IPC) was increased, expanding both populations, with greater effect on IPCs. Furthermore, cell cycle re-entry was increased during early corticogenesis, whereas cell cycle exit was augmented at middle stage. Consequently, neurogenesis was reduced early, whereas it was enhanced during later development. In agreement, the timetable of early neurogenesis, indicated by birthdating analysis, was delayed. Cell cycle dynamics analyses in mutants indicated that p57(KIP2) regulates cell cycle length in both RGCs and IPCs. By contrast, related CKI p27(KIP1) controlled IPC proliferation exclusively. Furthermore, p57(KIP2) deficiency markedly increased RGC and IPC divisions at E14.5, whereas p27(KIP1) increased IPC proliferation at E16.5. Consequently, loss of p57(KIP2) increased primarily layer 5-6 neuron production, whereas loss of p27(KIP1) increased neurons specifically in layers 2-5. In conclusion, our observations suggest that p57(KIP2) and p27(KIP1) control neuronal output for distinct cortical layers by regulating different stages of precursor proliferation, and support a model in which IPCs contribute to both lower and upper layer neuron generation.
Collapse
Affiliation(s)
- Georges Mairet-Coello
- Department of Neuroscience and Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, NJ 08554, USA
| | | | | | | | | | | |
Collapse
|
240
|
Taniguchi K, Anderson AE, Sutherland AE, Wotton D. Loss of Tgif function causes holoprosencephaly by disrupting the SHH signaling pathway. PLoS Genet 2012; 8:e1002524. [PMID: 22383895 PMCID: PMC3285584 DOI: 10.1371/journal.pgen.1002524] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 12/21/2011] [Indexed: 01/23/2023] Open
Abstract
Holoprosencephaly (HPE) is a severe human genetic disease affecting craniofacial development, with an incidence of up to 1/250 human conceptions and 1.3 per 10,000 live births. Mutations in the Sonic Hedgehog (SHH) gene result in HPE in humans and mice, and the Shh pathway is targeted by other mutations that cause HPE. However, at least 12 loci are associated with HPE in humans, suggesting that defects in other pathways contribute to this disease. Although the TGIF1 (TG-interacting factor) gene maps to the HPE4 locus, and heterozygous loss of function TGIF1 mutations are associated with HPE, mouse models have not yet explained how loss of Tgif1 causes HPE. Using a conditional Tgif1 allele, we show that mouse embryos lacking both Tgif1 and the related Tgif2 have HPE-like phenotypes reminiscent of Shh null embryos. Eye and nasal field separation is defective, and forebrain patterning is disrupted in embryos lacking both Tgifs. Early anterior patterning is relatively normal, but expression of Shh is reduced in the forebrain, and Gli3 expression is up-regulated throughout the neural tube. Gli3 acts primarily as an antagonist of Shh function, and the introduction of a heterozygous Gli3 mutation into embryos lacking both Tgif genes partially rescues Shh signaling, nasal field separation, and HPE. Tgif1 and Tgif2 are transcriptional repressors that limit Transforming Growth Factor β/Nodal signaling, and we show that reducing Nodal signaling in embryos lacking both Tgifs reduces the severity of HPE and partially restores the output of Shh signaling. Together, these results support a model in which Tgif function limits Nodal signaling to maintain the appropriate output of the Shh pathway in the forebrain. These data show for the first time that Tgif1 mutation in mouse contributes to HPE pathogenesis and provide evidence that this is due to disruption of the Shh pathway. Holoprosencephaly (HPE) is a devastating genetic disease affecting human brain development. HPE affects more than 1/8,000 live births and up to 1/250 conceptions. Several genetic loci are associated with HPE, and the mutated genes have been identified at some. We have analyzed the role of the TGIF1 gene, which is present at one of these loci (the HPE4 locus) and is mutated in a subset of human HPE patients. We show that Tgif1 mutations in mice cause HPE when combined with a mutation in the closely related Tgif2 gene. This provides the first evidence from model organisms that TGIF1 is in fact the gene at the HPE4 locus that causes HPE when mutated. The Sonic Hedgehog signaling pathway is the best understood pathway in the pathogenesis of HPE, and mutation of the Sonic Hedgehog gene in both humans and mice causes HPE. We show that mutations in Tgif1 and Tgif2 in mice cause HPE by disrupting the Sonic Hedgehog signaling pathway, further emphasizing the importance of this pathway for normal brain development. Thus we confirm TGIF1 as an HPE gene and provide genetic evidence that Tgif1 mutations cause HPE by disrupting the interplay of the Nodal and Sonic Hedgehog pathways.
Collapse
Affiliation(s)
- Kenichiro Taniguchi
- Department of Biochemistry and Molecular Genetics and Center for Cell Signaling, University of Virginia, Charlottesville, Virginia, United States of America
| | - Anoush E. Anderson
- Department of Biochemistry and Molecular Genetics and Center for Cell Signaling, University of Virginia, Charlottesville, Virginia, United States of America
| | - Ann E. Sutherland
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, United States of America
| | - David Wotton
- Department of Biochemistry and Molecular Genetics and Center for Cell Signaling, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
241
|
Peco E, Escude T, Agius E, Sabado V, Medevielle F, Ducommun B, Pituello F. The CDC25B phosphatase shortens the G2 phase of neural progenitors and promotes efficient neuron production. Development 2012; 139:1095-104. [PMID: 22318230 DOI: 10.1242/dev.068569] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
During embryonic development, changes in cell cycle kinetics have been associated with neurogenesis. This observation suggests that specific cell cycle regulators may be recruited to modify cell cycle dynamics and influence the decision between proliferation and differentiation. In the present study, we investigate the role of core positive cell cycle regulators, the CDC25 phosphatases, in this process. We report that, in the developing chicken spinal cord, only CDC25A is expressed in domains where neural progenitors undergo proliferative self-renewing divisions, whereas the combinatorial expression of CDC25A and CDC25B correlates remarkably well with areas where neurogenesis occurs. We also establish that neural progenitors expressing both CDC25A and CDC25B have a shorter G2 phase than those expressing CDC25A alone. We examine the functional relevance of these correlations using an RNAi-based method that allows us to knock down CDC25B efficiently and specifically. Reducing CDC25B expression results in a specific lengthening of the G2 phase, whereas the S-phase length and the total cell cycle time are not significantly modified. This modification of cell cycle kinetics is associated with a reduction in neuron production that is due to the altered conversion of proliferating neural progenitor cells to post-mitotic neurons. Thus, expression of CDC25B in neural progenitors has two functions: to change cell cycle kinetics and in particular G2-phase length and also to promote neuron production, identifying new roles for this phosphatase during neurogenesis.
Collapse
Affiliation(s)
- Emilie Peco
- Université de Toulouse, CBD, 118 route de Narbonne, F-31062 Toulouse, France
| | | | | | | | | | | | | |
Collapse
|
242
|
Molecular regulation of striatal development: a review. ANATOMY RESEARCH INTERNATIONAL 2012; 2012:106529. [PMID: 22567304 PMCID: PMC3335634 DOI: 10.1155/2012/106529] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 10/07/2011] [Indexed: 01/26/2023]
Abstract
The central nervous system is composed of the brain and the spinal cord. The brain is a complex organ that processes and coordinates activities of the body in bilaterian, higher-order animals. The development of the brain mirrors its complex function as it requires intricate genetic signalling at specific times, and deviations from this can lead to brain malformations such as anencephaly. Research into how the CNS is specified and patterned has been studied extensively in chick, fish, frog, and mice, but findings from the latter will be emphasised here as higher-order mammals show most similarity to the human brain. Specifically, we will focus on the embryonic development of an important forebrain structure, the striatum (also known as the dorsal striatum or neostriatum). Over the past decade, research on striatal development in mice has led to an influx of new information about the genes involved, but the precise orchestration between the genes, signalling molecules, and transcription factors remains unanswered. We aim to summarise what is known to date about the tightly controlled network of interacting genes that control striatal development. This paper will discuss early telencephalon patterning and dorsal ventral patterning with specific reference to the genes involved in striatal development.
Collapse
|
243
|
Alfonso J, Le Magueresse C, Zuccotti A, Khodosevich K, Monyer H. Diazepam Binding Inhibitor Promotes Progenitor Proliferation in the Postnatal SVZ by Reducing GABA Signaling. Cell Stem Cell 2012; 10:76-87. [DOI: 10.1016/j.stem.2011.11.011] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Revised: 08/31/2011] [Accepted: 11/10/2011] [Indexed: 02/01/2023]
|
244
|
Runx1 promotes neuronal differentiation in dorsal root ganglion. Mol Cell Neurosci 2012; 49:23-31. [PMID: 21906677 DOI: 10.1016/j.mcn.2011.08.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 08/02/2011] [Accepted: 08/19/2011] [Indexed: 11/21/2022] Open
Abstract
Transcription factor Runx1 controls the cell type specification of peptidergic and nonpeptidergic nociceptive dorsal root ganglion (DRG) neurons by repressing TrkA and calcitonin gene-related peptide (CGRP) expression and activating Ret expression during late embryonic and early postnatal periods (Chen et al., 2006b; Kramer et al., 2006; Yoshikawa et al., 2007). Because Runx1 is expressed in DRG from early developmental stages, we examined the roles of Runx1 in the proliferation and the neuronal differentiation of DRG cells. We used transgenic Runx1-deficient (Runx1(-/-)::Tg) mice which are rescued from early embryonic lethality by selective expression of Runx1 in hematopoietic cells under the control of GATA-1 promoter. We found that TrkA-expressing (TrkA(+)) DRG neurons were decreased at embryonic day (E) 12.5 in contrast to the previous study showing that TrkA(+) DRG neurons were increased at E17.5 in Runx1(-/-)::Tg mice (Yoshikawa et al., 2007). The number of DRG neurons which express neuronal markers Hu, NeuN and Islet1 was also reduced in Runx1(-/-)::Tg mice at E12.5, suggesting that the neuronal differentiation was suppressed in these mice. The cell cycle analysis using BrdU/IDU revealed that the number of DRG cells in S-phase and G2/M-phase was increased in Runx1(-/-)::Tg mice at E12.5, while the length of S-phase was not changed between Runx1(+/+)::Tg and Runx1(-/-)::Tg mice, suggesting that Runx1 negatively controls the proliferation of DRG progenitor cell subpopulation in early embryonic period. Hes1 is a negative regulator of neuronal differentiation (Ishibashi et al., 1995; Tomita et al., 1996), and we found that the number of Hes1(+) DRG cells was increased in Runx1(-/-)::Tg mice at E12.5. In summary, the present study suggests a novel function that Runx1 activates the neuronal differentiation of DRG cell subpopulation through the repression of Hes1 expression in early embryonic period.
Collapse
|
245
|
Viegas P, Nicoleau C, Perrier AL. Derivation of striatal neurons from human stem cells. PROGRESS IN BRAIN RESEARCH 2012. [DOI: 10.1016/b978-0-444-59575-1.00017-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
246
|
Auger H, Thuret R, El Yakoubi W, Papalopulu N. A bromodeoxyuridine (BrdU) based protocol for characterizing proliferating progenitors in Xenopus embryos. Methods Mol Biol 2012; 917:461-75. [PMID: 22956104 DOI: 10.1007/978-1-61779-992-1_26] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BrdU is a thymidine analog that is incorporated into DNA during the S-phase of the cell cycle. BrdU incorporation can be used to quantify the number of cells that are in S-phase in the time period that BrdU is available. Thus, BrdU incorporation is an essential method in the quantitative analysis of cell proliferation, during normal embryonic development or after experimental manipulation. It is a reliable and versatile method that can be easily combined with immunohistochemistry and in situ hybridization to relate cell proliferation with gene expression. BrdU incorporation has been used in all model organisms; here, we describe a protocol adapted for use in Xenopus embryos.
Collapse
Affiliation(s)
- Hélène Auger
- Faculty of Life Sciences, University of Manchester, Manchester, England, UK
| | | | | | | |
Collapse
|
247
|
De Filippis R, Pancrazi L, Bjørgo K, Rosseto A, Kleefstra T, Grillo E, Panighini A, Cardarelli F, Meloni I, Ariani F, Mencarelli MA, Hayek J, Renieri A, Costa M, Mari F. Expanding the phenotype associated with FOXG1 mutations and in vivo FoxG1 chromatin-binding dynamics. Clin Genet 2011; 82:395-403. [PMID: 22091895 DOI: 10.1111/j.1399-0004.2011.01810.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mutations in the Forkhead box G1 (FOXG1) gene, a brain specific transcriptional factor, are responsible for the congenital variant of Rett syndrome. Until now FOXG1 point mutations have been reported in 12 Rett patients. Recently seven additional patients have been reported with a quite homogeneous severe phenotype designated as the FOXG1 syndrome. Here we describe two unrelated patients with a de novo FOXG1 point mutation, p.Gln46X and p.Tyr400X, respectively, having a milder phenotype and sharing a distinctive facial appearance. Although FoxG1 action depends critically on its binding to chromatin, very little is known about the dynamics of this process. Using fluorescence recovery after photobleaching, we showed that most of the GFP-FoxG1 fusion protein associates reversibly to chromatin whereas the remaining fraction is bound irreversibly. Furthermore, we showed that the two pathologic derivatives of FoxG1 described in this paper present a dramatic alteration in chromatin affinity and irreversibly bound fraction in comparison with Ser323fsX325 mutant (associated with a severe phenotype) and wild type Foxg1 protein. Our observations suggest that alterations in the kinetics of FoxG1 binding to chromatin might contribute to the pathological effects of FOXG1 mutations.
Collapse
Affiliation(s)
- R De Filippis
- Medical Genetics, Department of Biotechnology, University of Siena, Siena, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
248
|
Passemard S, El Ghouzzi V, Nasser H, Verney C, Vodjdani G, Lacaud A, Lebon S, Laburthe M, Robberecht P, Nardelli J, Mani S, Verloes A, Gressens P, Lelièvre V. VIP blockade leads to microcephaly in mice via disruption of Mcph1-Chk1 signaling. J Clin Invest 2011; 121:3071-87. [PMID: 21737879 DOI: 10.1172/jci43824] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Accepted: 05/11/2011] [Indexed: 01/14/2023] Open
Abstract
Autosomal recessive primary microcephaly (MCPH) is a genetic disorder that causes a reduction of cortical outgrowth without severe interference with cortical patterning. It is associated with mutations in a number of genes encoding protein involved in mitotic spindle formation and centrosomal activities or cell cycle control. We have shown previously that blocking vasoactive intestinal peptide (VIP) during gestation in mice by using a VIP antagonist (VA) results in microcephaly. Here, we have shown that the cortical abnormalities caused by prenatal VA administration mimic the phenotype described in MCPH patients and that VIP blockade during neurogenesis specifically disrupts Mcph1 signaling. VA administration reduced neuroepithelial progenitor proliferation by increasing cell cycle length and promoting cell cycle exit and premature neuronal differentiation. Quantitative RT-PCR and Western blot showed that VA downregulated Mcph1. Inhibition of Mcph1 expression led to downregulation of Chk1 and reduction of Chk1 kinase activity. The inhibition of Mcph1 and Chk1 affected the expression of a specific subset of cell cycle–controlling genes and turned off neural stem cell proliferation in neurospheres. Furthermore, in vitro silencing of either Mcph1 or Chk1 in neurospheres mimicked VA-induced inhibition of cell proliferation. These results demonstrate that VIP blockade induces microcephaly through Mcph1 signaling and suggest that VIP/Mcph1/Chk1 signaling is key for normal cortical development.
Collapse
|
249
|
Young A, Machacek DW, Dhara SK, MacLeish PR, Benveniste M, Dodla MC, Sturkie CD, Stice SL. Ion channels and ionotropic receptors in human embryonic stem cell derived neural progenitors. Neuroscience 2011; 192:793-805. [PMID: 21672611 PMCID: PMC3166353 DOI: 10.1016/j.neuroscience.2011.04.039] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Revised: 04/14/2011] [Accepted: 04/15/2011] [Indexed: 11/24/2022]
Abstract
Human neural progenitor cells differentiated from human embryonic stem cells offer a potential cell source for studying neurodegenerative diseases and for drug screening assays. Previously, we demonstrated that human neural progenitors could be maintained in a proliferative state with the addition of leukemia inhibitory factor and basic fibroblast growth factor. Here we demonstrate that 96 h after removal of basic fibroblast growth factor the neural progenitor cell culture was significantly altered and cell replication halted. Fourteen days after the removal of basic fibroblast growth factor, most cells expressed microtubule-associated protein 2 and TUJ1, markers characterizing a post-mitotic neuronal phenotype as well as neural developmental markers Cdh2 and Gbx2. Real-time PCR was performed to determine the ionotropic receptor subunit expression profile. Differentiated neural progenitors express subunits of glutamatergic, GABAergic, nicotinic, purinergic and transient receptor potential receptors. In addition, sodium and calcium channel subunits were also expressed. Functionally, virtually all the hNP cells tested under whole-cell voltage clamp exhibited delayed rectifier potassium channel currents and some differentiated cells exhibited tetrodotoxin-sensitive, voltage-dependent sodium channel current. Action potentials could also be elicited by currents injection under whole-cell current clamp in a minority of cells. These results indicate that removing basic fibroblast growth factor from the neural progenitor cell cultures leads to a post-mitotic state, and has the capability to produce excitable cells that can generate action potentials, a landmark characteristic of a neuronal phenotype. This is the first report of an efficient and simple means of generating human neuronal cells for ionotropic receptor assays and ultimately for electrically active human neural cell assays for drug discovery.
Collapse
Affiliation(s)
- Amber Young
- Regenerative Bioscience Center, 425 River Rd Room 450, Athens, GA 30602 USA
| | - Dave W. Machacek
- Aruna Biomedical, Athens, GA 30602, 425 River Rd Room 430, Athens, GA 30602 USA
| | - Sujoy K. Dhara
- Regenerative Bioscience Center, 425 River Rd Room 450, Athens, GA 30602 USA
| | - Peter R. MacLeish
- Neuroscience Institute, Morehouse School of Medicine, 720 Westview Drive, SW, Atlanta, GA 30310
| | - Morris Benveniste
- Neuroscience Institute, Morehouse School of Medicine, 720 Westview Drive, SW, Atlanta, GA 30310
| | - Mahesh C. Dodla
- Regenerative Bioscience Center, 425 River Rd Room 450, Athens, GA 30602 USA
| | - Carla D. Sturkie
- Regenerative Bioscience Center, 425 River Rd Room 450, Athens, GA 30602 USA
| | - Steven L. Stice
- Regenerative Bioscience Center, 425 River Rd Room 450, Athens, GA 30602 USA
- Aruna Biomedical, Athens, GA 30602, 425 River Rd Room 430, Athens, GA 30602 USA
| |
Collapse
|
250
|
Yu T, Yaguchi Y, Echevarria D, Martinez S, Basson MA. Sprouty genes prevent excessive FGF signalling in multiple cell types throughout development of the cerebellum. Development 2011; 138:2957-68. [PMID: 21693512 PMCID: PMC3119305 DOI: 10.1242/dev.063784] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2011] [Indexed: 01/06/2023]
Abstract
Fibroblast growth factors (FGFs) and regulators of the FGF signalling pathway are expressed in several cell types within the cerebellum throughout its development. Although much is known about the function of this pathway during the establishment of the cerebellar territory during early embryogenesis, the role of this pathway during later developmental stages is still poorly understood. Here, we investigated the function of sprouty genes (Spry1, Spry2 and Spry4), which encode feedback antagonists of FGF signalling, during cerebellar development in the mouse. Simultaneous deletion of more than one of these genes resulted in a number of defects, including mediolateral expansion of the cerebellar vermis, reduced thickness of the granule cell layer and abnormal foliation. Analysis of cerebellar development revealed that the anterior cerebellar neuroepithelium in the early embryonic cerebellum was expanded and that granule cell proliferation during late embryogenesis and early postnatal development was reduced. We show that the granule cell proliferation deficit correlated with reduced sonic hedgehog (SHH) expression and signalling. A reduction in Fgfr1 dosage during development rescued these defects, confirming that the abnormalities are due to excess FGF signalling. Our data indicate that sprouty acts both cell autonomously in granule cell precursors and non-cell autonomously to regulate granule cell number. Taken together, our data demonstrate that FGF signalling levels have to be tightly controlled throughout cerebellar development in order to maintain the normal development of multiple cell types.
Collapse
Affiliation(s)
- Tian Yu
- Department of Craniofacial Development, King's College London, London SE1 9RT, UK
| | - Yuichiro Yaguchi
- Department of Craniofacial Development, King's College London, London SE1 9RT, UK
- Department of Otorhinolaryngology, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Diego Echevarria
- Instituto de Neurociencias de Alicante, UMH-CSIC, 03550-San Juan de Alicante, Spain
| | - Salvador Martinez
- Instituto de Neurociencias de Alicante, UMH-CSIC, 03550-San Juan de Alicante, Spain
| | - M. Albert Basson
- Department of Craniofacial Development, King's College London, London SE1 9RT, UK
- MRC Centre for Developmental Neurobiology, King's College London, London SE1 1UL, UK
| |
Collapse
|