151
|
Kumamoto T, Hanashima C. Evolutionary conservation and conversion of Foxg1 function in brain development. Dev Growth Differ 2017; 59:258-269. [PMID: 28581027 DOI: 10.1111/dgd.12367] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 05/01/2017] [Accepted: 05/01/2017] [Indexed: 12/31/2022]
Abstract
Among the forkhead box protein family, Foxg1 is a unique transcription factor that plays pleiotropic and non-redundant roles in vertebrate brain development. The emergence of the telencephalon at the rostral end of the neural tube and its subsequent expansion that is mediated by Foxg1 was a key reason for the vertebrate brain to acquire higher order information processing, where Foxg1 is repetitively used in the sequential events of telencephalic development to control multi-steps of brain circuit formation ranging from cell cycle control to neuronal differentiation in a clade- and species-specific manner. The objective of this review is to discuss how the evolutionary changes in cis- and trans-regulatory network that is mediated by a single transcription factor has contributed to determining the fundamental vertebrate brain structure and its divergent roles in instructing species-specific neuronal circuitry and functional specialization.
Collapse
Affiliation(s)
- Takuma Kumamoto
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U968, CNRS UMR 7210, Institut de la Vision, 75012, Paris, France
| | - Carina Hanashima
- Faculty of Education and Integrated Arts and Sciences, Waseda University, Shinjuku-ku, Tokyo, 162-8480, Japan.,Graduate School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, 162-8480, Japan.,Laboratory for Neocortical Development, RIKEN Center for Developmental Biology, Kobe, 650-0047, Japan
| |
Collapse
|
152
|
Suzuki T, Morishita Y. A quantitative approach to understanding vertebrate limb morphogenesis at the macroscopic tissue level. Curr Opin Genet Dev 2017; 45:108-114. [PMID: 28502890 DOI: 10.1016/j.gde.2017.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/13/2017] [Accepted: 04/13/2017] [Indexed: 02/01/2023]
Abstract
To understand organ morphogenetic mechanisms, it is essential to clarify how spatiotemporally-regulated molecular/cellular dynamics causes physical tissue deformation. In the case of vertebrate limb development, while some of the genes and oriented cell behaviors underlying morphogenesis have been revealed, tissue deformation dynamics remains incompletely understood. We here introduce our recent work on the reconstruction of tissue deformation dynamics in chick limb development from cell lineage tracing data. This analysis has revealed globally-aligned anisotropic tissue deformation along the proximo-distal axis not only in the distal region but also in the whole limb bud. This result points to a need, as a future challenge, to find oriented molecular/cellular behaviors for realizing the observed anisotropic tissue deformation in both proximal and distal regions, which will lead to systems understanding of limb morphogenesis.
Collapse
Affiliation(s)
- Takayuki Suzuki
- Division of Biological Science, Graduate School of Science, Nagoya University Furo-cho, Chikusa-ku, Nagoya 464-8602, Japan.
| | - Yoshihiro Morishita
- Laboratory for Developmental Morphogeometry, RIKEN Quantitative Biology Center, Kobe 650-0047, Japan.
| |
Collapse
|
153
|
Fused cerebral organoids model interactions between brain regions. Nat Methods 2017; 14:743-751. [PMID: 28504681 PMCID: PMC5540177 DOI: 10.1038/nmeth.4304] [Citation(s) in RCA: 493] [Impact Index Per Article: 70.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 05/04/2017] [Indexed: 02/08/2023]
Abstract
Human brain development involves complex interactions between different areas, including long distance neuronal migration or formation of major axonal tracts. 3D cerebral organoids allow the growth of diverse brain regions in vitro, but the random arrangement of regional identities limits the reliable analysis of complex phenotypes. Here, we describe a co-culture method combining various brain regions of choice within one organoid tissue. By fusing organoids specified toward dorsal and ventral forebrain, we generate a dorsal-ventral axis. Using fluorescent reporters, we demonstrate robust directional GABAergic interneuron migration from ventral into dorsal forebrain. We describe methodology for time-lapse imaging of human interneuron migration that is inhibited by the CXCR4 antagonist AMD3100. Our results demonstrate that cerebral organoid fusion cultures can model complex interactions between different brain regions. Combined with reprogramming technology, fusions offer the possibility to analyze complex neurodevelopmental defects using cells from neurological disease patients, and to test potential therapeutic compounds.
Collapse
|
154
|
Hwang Y, Futran M, Hidalgo D, Pop R, Iyer DR, Scully R, Rhind N, Socolovsky M. Global increase in replication fork speed during a p57 KIP2-regulated erythroid cell fate switch. SCIENCE ADVANCES 2017; 3:e1700298. [PMID: 28560351 PMCID: PMC5446218 DOI: 10.1126/sciadv.1700298] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 03/28/2017] [Indexed: 06/07/2023]
Abstract
Cell cycle regulators are increasingly implicated in cell fate decisions, such as the acquisition or loss of pluripotency and self-renewal potential. The cell cycle mechanisms that regulate these cell fate decisions are largely unknown. We studied an S phase-dependent cell fate switch, in which murine early erythroid progenitors transition in vivo from a self-renewal state into a phase of active erythroid gene transcription and concurrent maturational cell divisions. We found that progenitors are dependent on p57KIP2-mediated slowing of replication forks for self-renewal, a novel function for cyclin-dependent kinase inhibitors. The switch to differentiation entails rapid down-regulation of p57KIP2 with a consequent global increase in replication fork speed and an abruptly shorter S phase. Our work suggests that cell cycles with specialized global DNA replication dynamics are integral to the maintenance of specific cell states and to cell fate decisions.
Collapse
Affiliation(s)
- Yung Hwang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Melinda Futran
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Daniel Hidalgo
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Ramona Pop
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Divya Ramalingam Iyer
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Ralph Scully
- Division of Hematology-Oncology, Department of Medicine, and Cancer Research Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| | - Nicholas Rhind
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Merav Socolovsky
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|
155
|
Bulstrode H, Johnstone E, Marques-Torrejon MA, Ferguson KM, Bressan RB, Blin C, Grant V, Gogolok S, Gangoso E, Gagrica S, Ender C, Fotaki V, Sproul D, Bertone P, Pollard SM. Elevated FOXG1 and SOX2 in glioblastoma enforces neural stem cell identity through transcriptional control of cell cycle and epigenetic regulators. Genes Dev 2017; 31:757-773. [PMID: 28465359 PMCID: PMC5435889 DOI: 10.1101/gad.293027.116] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 03/30/2017] [Indexed: 12/11/2022]
Abstract
Glioblastoma multiforme (GBM) is an aggressive brain tumor driven by cells with hallmarks of neural stem (NS) cells. GBM stem cells frequently express high levels of the transcription factors FOXG1 and SOX2. Here we show that increased expression of these factors restricts astrocyte differentiation and can trigger dedifferentiation to a proliferative NS cell state. Transcriptional targets include cell cycle and epigenetic regulators (e.g., Foxo3, Plk1, Mycn, Dnmt1, Dnmt3b, and Tet3). Foxo3 is a critical repressed downstream effector that is controlled via a conserved FOXG1/SOX2-bound cis-regulatory element. Foxo3 loss, combined with exposure to the DNA methylation inhibitor 5-azacytidine, enforces astrocyte dedifferentiation. DNA methylation profiling in differentiating astrocytes identifies changes at multiple polycomb targets, including the promoter of Foxo3 In patient-derived GBM stem cells, CRISPR/Cas9 deletion of FOXG1 does not impact proliferation in vitro; however, upon transplantation in vivo, FOXG1-null cells display increased astrocyte differentiation and up-regulate FOXO3. In contrast, SOX2 ablation attenuates proliferation, and mutant cells cannot be expanded in vitro. Thus, FOXG1 and SOX2 operate in complementary but distinct roles to fuel unconstrained self-renewal in GBM stem cells via transcriptional control of core cell cycle and epigenetic regulators.
Collapse
Affiliation(s)
- Harry Bulstrode
- Medical Research Council (MRC) Centre for Regenerative Medicine
- Edinburgh Cancer Research UK Cancer Centre, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
| | - Ewan Johnstone
- Wellcome Trust-MRC Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, United Kingdom
| | - Maria Angeles Marques-Torrejon
- Medical Research Council (MRC) Centre for Regenerative Medicine
- Edinburgh Cancer Research UK Cancer Centre, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
| | - Kirsty M Ferguson
- Medical Research Council (MRC) Centre for Regenerative Medicine
- Edinburgh Cancer Research UK Cancer Centre, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
| | - Raul Bardini Bressan
- Medical Research Council (MRC) Centre for Regenerative Medicine
- Edinburgh Cancer Research UK Cancer Centre, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
| | - Carla Blin
- Medical Research Council (MRC) Centre for Regenerative Medicine
- Edinburgh Cancer Research UK Cancer Centre, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
| | - Vivien Grant
- Medical Research Council (MRC) Centre for Regenerative Medicine
- Edinburgh Cancer Research UK Cancer Centre, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
| | - Sabine Gogolok
- Medical Research Council (MRC) Centre for Regenerative Medicine
- Edinburgh Cancer Research UK Cancer Centre, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
| | - Ester Gangoso
- Medical Research Council (MRC) Centre for Regenerative Medicine
- Edinburgh Cancer Research UK Cancer Centre, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
| | - Sladjana Gagrica
- Department of Cancer Biology, UCL Cancer Institute, University College London, London WC1E 6BT, United Kingdom
| | - Christine Ender
- Department of Cancer Biology, UCL Cancer Institute, University College London, London WC1E 6BT, United Kingdom
| | - Vassiliki Fotaki
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom
| | - Duncan Sproul
- MRC Human Genetics Unit
- Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Paul Bertone
- Wellcome Trust-MRC Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, United Kingdom
| | - Steven M Pollard
- Medical Research Council (MRC) Centre for Regenerative Medicine
- Edinburgh Cancer Research UK Cancer Centre, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
| |
Collapse
|
156
|
Differentiation of human telencephalic progenitor cells into MSNs by inducible expression of Gsx2 and Ebf1. Proc Natl Acad Sci U S A 2017; 114:E1234-E1242. [PMID: 28137879 DOI: 10.1073/pnas.1611473114] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Medium spiny neurons (MSNs) are a key population in the basal ganglia network, and their degeneration causes a severe neurodegenerative disorder, Huntington's disease. Understanding how ventral neuroepithelial progenitors differentiate into MSNs is critical for regenerative medicine to develop specific differentiation protocols using human pluripotent stem cells. Studies performed in murine models have identified some transcriptional determinants, including GS Homeobox 2 (Gsx2) and Early B-cell factor 1 (Ebf1). Here, we have generated human embryonic stem (hES) cell lines inducible for these transcription factors, with the aims of (i) studying their biological role in human neural progenitors and (ii) incorporating TF conditional expression in a developmental-based protocol for generating MSNs from hES cells. Using this approach, we found that Gsx2 delays cell-cycle exit and reduces Pax6 expression, whereas Ebf1 promotes neuronal differentiation. Moreover, we found that Gsx2 and Ebf1 combined overexpression in hES cells achieves high yields of MSNs, expressing Darpp32 and Ctip2, in vitro as well in vivo after transplantation. We show that hES-derived striatal progenitors can be transplanted in animal models and can differentiate and integrate into the host, extending fibers over a long distance.
Collapse
|
157
|
Joven A, Wang H, Pinheiro T, Hameed LS, Belnoue L, Simon A. Cellular basis of brain maturation and acquisition of complex behaviors in salamanders. Development 2017; 145:dev.160051. [DOI: 10.1242/dev.160051] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 11/20/2017] [Indexed: 12/22/2022]
Abstract
The overall bauplan of the tetrapod brain is highly conserved, yet significant variations exist among species in terms of brain size, structural composition and cellular diversity. Understanding processes underlying neural and behavioral development in a wide range of species is important both from an evolutionary developmental perspective as well as for the identification of cell sources with post-developmental neurogenic potential. Here we characterize germinal processes in the brain of Notophthalmus viridescens and Pleurodeles waltl during both development and adulthood. Using a combination of cell tracking tools, including clonal analyses in new transgenic salamander lines we examine the origin of neural stem and progenitor cells found in the adult brain, determine regional variability in cell cycle length of progenitor cells, and show spatio-temporally orchestrated neurogenesis. We analyze how maturation of different brain regions and neuronal subpopulations are linked to the acquisition of complex behaviors, and how these behaviors are altered upon chemical ablation of dopamine neurons. Our data analyzed from an evolutionary perspective reveal both common and species-specific processes in tetrapod brain formation and function.
Collapse
Affiliation(s)
- Alberto Joven
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Heng Wang
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Tiago Pinheiro
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - L. Shahul Hameed
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Laure Belnoue
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - András Simon
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
158
|
RNA activation of haploinsufficient Foxg1 gene in murine neocortex. Sci Rep 2016; 6:39311. [PMID: 27995975 PMCID: PMC5172352 DOI: 10.1038/srep39311] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 11/22/2016] [Indexed: 11/18/2022] Open
Abstract
More than one hundred distinct gene hemizygosities are specifically linked to epilepsy, mental retardation, autism, schizophrenia and neuro-degeneration. Radical repair of these gene deficits via genome engineering is hardly feasible. The same applies to therapeutic stimulation of the spared allele by artificial transactivators. Small activating RNAs (saRNAs) offer an alternative, appealing approach. As a proof-of-principle, here we tested this approach on the Rett syndrome-linked, haploinsufficient, Foxg1 brain patterning gene. We selected a set of artificial small activating RNAs (saRNAs) upregulating it in neocortical precursors and their derivatives. Expression of these effectors achieved a robust biological outcome. saRNA-driven activation (RNAa) was limited to neural cells which normally express Foxg1 and did not hide endogenous gene tuning. saRNAs recognized target chromatin through a ncRNA stemming from it. Gene upregulation required Ago1 and was associated to RNApolII enrichment throughout the Foxg1 locus. Finally, saRNA delivery to murine neonatal brain replicated Foxg1-RNAa in vivo.
Collapse
|
159
|
Geng X, Acosta S, Lagutin O, Gil HJ, Oliver G. Six3 dosage mediates the pathogenesis of holoprosencephaly. Development 2016; 143:4462-4473. [PMID: 27770010 PMCID: PMC5201039 DOI: 10.1242/dev.132142] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 10/12/2016] [Indexed: 01/06/2023]
Abstract
Holoprosencephaly (HPE) is defined as the incomplete separation of the two cerebral hemispheres. The pathology of HPE is variable and, based on the severity of the defect, HPE is divided into alobar, semilobar, and lobar. Using a novel hypomorphic Six3 allele, we demonstrate in mice that variability in Six3 dosage results in different HPE phenotypes. Furthermore, we show that whereas the semilobar phenotype results from severe downregulation of Shh expression in the rostral diencephalon ventral midline, the alobar phenotype is caused by downregulation of Foxg1 expression in the anterior neural ectoderm. Consistent with these results, in vivo activation of the Shh signaling pathway rescued the semilobar phenotype but not the alobar phenotype. Our findings show that variations in Six3 dosage result in different forms of HPE.
Collapse
Affiliation(s)
- Xin Geng
- Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Sandra Acosta
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL 60611, USA
| | - Oleg Lagutin
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Hyea Jin Gil
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL 60611, USA
| | - Guillermo Oliver
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
160
|
Santos-Durán GN, Ferreiro-Galve S, Menuet A, Quintana-Urzainqui I, Mazan S, Rodríguez-Moldes I, Candal E. The Shark Alar Hypothalamus: Molecular Characterization of Prosomeric Subdivisions and Evolutionary Trends. Front Neuroanat 2016; 10:113. [PMID: 27932958 PMCID: PMC5121248 DOI: 10.3389/fnana.2016.00113] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 11/08/2016] [Indexed: 12/31/2022] Open
Abstract
The hypothalamus is an important physiologic center of the vertebrate brain involved in the elaboration of individual and species survival responses. To better understand the ancestral organization of the alar hypothalamus we revisit previous data on ScOtp, ScDlx2/5, ScTbr1, ScNkx2.1 expression and Pax6 immunoreactivity jointly with new data on ScNeurog2, ScLhx9, ScLhx5, and ScNkx2.8 expression, in addition to immunoreactivity to serotonin (5-HT) and doublecortin (DCX) in the catshark Scyliorhinus canicula, a key species for this purpose since cartilaginous fishes are basal representatives of gnathostomes (jawed vertebrates). Our study revealed a complex genoarchitecture for the chondrichthyan alar hypothalamus. We identified terminal (rostral) and peduncular (caudal) subdivisions in the prosomeric paraventricular and subparaventricular areas (TPa/PPa and TSPa/PSPa, respectively) evidenced by the expression pattern of developmental genes like ScLhx5 (TPa) and immunoreactivity against Pax6 (PSPa) and 5-HT (PPa and PSPa). Dorso-ventral subdivisions were only evidenced in the SPa (SPaD, SPaV; respectively) by means of Pax6 and ScNkx2.8 (respectively). Interestingly, ScNkx2.8 expression overlaps over the alar-basal boundary, as Nkx2.2 does in other vertebrates. Our results reveal evidences for the existence of different groups of tangentially migrated cells expressing ScOtp, Pax6, and ScDlx2. The genoarchitectonic comparative analysis suggests alternative interpretations of the rostral-most alar plate in prosomeric terms and reveals a conserved molecular background for the vertebrate alar hypothalamus likely acquired before/during the agnathan-gnathostome transition, on which Otp, Pax6, Lhx5, and Neurog2 are expressed in the Pa while Dlx and Nkx2.2/Nkx2.8 are expressed in the SPa.
Collapse
Affiliation(s)
- Gabriel N Santos-Durán
- Grupo BRAINSHARK, Departamento de Biología Funcional, Universidade de Santiago de Compostela Santiago de Compostela, Spain
| | - Susana Ferreiro-Galve
- Grupo BRAINSHARK, Departamento de Biología Funcional, Universidade de Santiago de Compostela Santiago de Compostela, Spain
| | - Arnaud Menuet
- CNRS, UMR 7355, University of Orleans Orleans, France
| | - Idoia Quintana-Urzainqui
- Grupo BRAINSHARK, Departamento de Biología Funcional, Universidade de Santiago de CompostelaSantiago de Compostela, Spain; Centre for Integrative Physiology, University of EdinburghEdinburgh, UK
| | - Sylvie Mazan
- Sorbonne Universités, UPMC, CNRS UMR7232 Biologie Intégrative des Organismes Marins, Observatoire Océanologique Banyuls sur Mer, France
| | - Isabel Rodríguez-Moldes
- Grupo BRAINSHARK, Departamento de Biología Funcional, Universidade de Santiago de Compostela Santiago de Compostela, Spain
| | - Eva Candal
- Grupo BRAINSHARK, Departamento de Biología Funcional, Universidade de Santiago de Compostela Santiago de Compostela, Spain
| |
Collapse
|
161
|
Choi H, Song J, Park G, Kim J. Modeling of Autism Using Organoid Technology. Mol Neurobiol 2016; 54:7789-7795. [DOI: 10.1007/s12035-016-0274-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 10/30/2016] [Indexed: 01/01/2023]
|
162
|
Loughran AJ, Tuomanen EI. Blood borne: bacterial components in mother's blood influence fetal development. INFLAMMATION AND CELL SIGNALING 2016; 3. [PMID: 28280750 PMCID: PMC5340199 DOI: 10.14800/ics.1421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Bacterial or viral infection of the mother during the course of pregnancy can cross the placenta and actively infect the fetus. However, especially for bacteria, it is more common for mothers to experience an infection that can be treated without overt fetal infection. In this setting, it is less well understood what the risk to fetal development is, particularly in terms of neurological development. This research highlight reviews recent findings indicating that bacterial components generated during infection of the mother can cross the placenta and activate the fetal innate immune system resulting in changes in the course of brain development and subsequent progression to postnatal cognitive disorders. Bacterial cell wall is a ubiquitous bacterial PAMP (pathogen-associated molecular pattern) known to activate inflammation through the stimulation of TLR2. Cell wall is released from bacteria during antibiotic treatment and new work shows that embryos exposed to cell wall from the mother demonstrate anomalous proliferation of neuronal precursor cells in a TLR2 dependent manner. Such proliferation increases the neuronal density of the cortical plate and alters brain architecture. Although there is no fetal death, subsequent cognitive development is significantly impaired. This model system suggests that bacterial infection of the mother and its treatment can impact fetal brain development and requires greater understanding to potentially eliminate a risk factor for cognitive disorders such as autism.
Collapse
Affiliation(s)
- Allister J Loughran
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Elaine I Tuomanen
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| |
Collapse
|
163
|
Shao Q, Herrlinger S, Yang SL, Lai F, Moore JM, Brindley MA, Chen JF. Zika virus infection disrupts neurovascular development and results in postnatal microcephaly with brain damage. Development 2016; 143:4127-4136. [PMID: 27729407 DOI: 10.1242/dev.143768] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 10/04/2016] [Indexed: 12/16/2022]
Abstract
Zika virus (ZIKV) infection of pregnant women can result in fetal brain abnormalities. It has been established that ZIKV disrupts neural progenitor cells (NPCs) and leads to embryonic microcephaly. However, the fate of other cell types in the developing brain and their contributions to ZIKV-associated brain abnormalities remain largely unknown. Using intracerebral inoculation of embryonic mouse brains, we found that ZIKV infection leads to postnatal growth restriction including microcephaly. In addition to cell cycle arrest and apoptosis of NPCs, ZIKV infection causes massive neuronal death and axonal rarefaction, which phenocopy fetal brain abnormalities in humans. Importantly, ZIKV infection leads to abnormal vascular density and diameter in the developing brain, resulting in a leaky blood-brain barrier (BBB). Massive neuronal death and BBB leakage indicate brain damage, which is further supported by extensive microglial activation and astrogliosis in virally infected brains. Global gene analyses reveal dysregulation of genes associated with immune responses in virus-infected brains. Thus, our data suggest that ZIKV triggers a strong immune response and disrupts neurovascular development, resulting in postnatal microcephaly with extensive brain damage.
Collapse
Affiliation(s)
- Qiang Shao
- Department of Genetics, Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Stephanie Herrlinger
- Department of Genetics, Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Si-Lu Yang
- Department of Genetics, Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Fan Lai
- University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Department of Human Genetics, Miami, FL 33136, USA
| | - Julie M Moore
- Department of Infectious Diseases and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA
| | - Melinda A Brindley
- Department of Infectious Diseases, Department of Population Health and Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA
| | - Jian-Fu Chen
- Department of Genetics, Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
164
|
Fededa JP, Esk C, Mierzwa B, Stanyte R, Yuan S, Zheng H, Ebnet K, Yan W, Knoblich JA, Gerlich DW. MicroRNA-34/449 controls mitotic spindle orientation during mammalian cortex development. EMBO J 2016; 35:2386-2398. [PMID: 27707753 PMCID: PMC5109238 DOI: 10.15252/embj.201694056] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 08/18/2016] [Accepted: 09/06/2016] [Indexed: 01/07/2023] Open
Abstract
Correct orientation of the mitotic spindle determines the plane of cellular cleavage and is crucial for organ development. In the developing cerebral cortex, spindle orientation defects result in severe neurodevelopmental disorders, but the precise mechanisms that control this important event are not fully understood. Here, we use a combination of high-content screening and mouse genetics to identify the miR-34/449 family as key regulators of mitotic spindle orientation in the developing cerebral cortex. By screening through all cortically expressed miRNAs in HeLa cells, we show that several members of the miR-34/449 family control mitotic duration and spindle rotation. Analysis of miR-34/449 knockout (KO) mouse embryos demonstrates significant spindle misorientation phenotypes in cortical progenitors, resulting in an excess of radial glia cells at the expense of intermediate progenitors and a significant delay in neurogenesis. We identify the junction adhesion molecule-A (JAM-A) as a key target for miR-34/449 in the developing cortex that might be responsible for those defects. Our data indicate that miRNA-dependent regulation of mitotic spindle orientation is crucial for cell fate specification during mammalian neurogenesis.
Collapse
Affiliation(s)
- Juan Pablo Fededa
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA) Vienna Biocenter (VBC), Vienna, Austria
| | - Christopher Esk
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA) Vienna Biocenter (VBC), Vienna, Austria
| | - Beata Mierzwa
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA) Vienna Biocenter (VBC), Vienna, Austria
| | - Rugile Stanyte
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA) Vienna Biocenter (VBC), Vienna, Austria
| | - Shuiqiao Yuan
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| | - Huili Zheng
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| | - Klaus Ebnet
- Institute-associated Research Group "Cell Adhesion and Cell Polarity", Institute of Medical Biochemistry, ZMBE, Münster, Germany
| | - Wei Yan
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| | - Juergen A Knoblich
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA) Vienna Biocenter (VBC), Vienna, Austria
| | - Daniel W Gerlich
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA) Vienna Biocenter (VBC), Vienna, Austria
| |
Collapse
|
165
|
Radhakrishnan B, Alwin Prem Anand A. Role of miRNA-9 in Brain Development. J Exp Neurosci 2016; 10:101-120. [PMID: 27721656 PMCID: PMC5053108 DOI: 10.4137/jen.s32843] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 09/01/2016] [Accepted: 09/07/2016] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) are a class of small regulatory RNAs involved in gene regulation. The regulation is effected by either translational inhibition or transcriptional silencing. In vertebrates, the importance of miRNA in development was discovered from mice and zebrafish dicer knockouts. The miRNA-9 (miR-9) is one of the most highly expressed miRNAs in the early and adult vertebrate brain. It has diverse functions within the developing vertebrate brain. In this article, the role of miR-9 in the developing forebrain (telencephalon and diencephalon), midbrain, hindbrain, and spinal cord of vertebrate species is highlighted. In the forebrain, miR-9 is necessary for the proper development of dorsoventral telencephalon by targeting marker genes expressed in the telencephalon. It regulates proliferation in telencephalon by regulating Foxg1, Pax6, Gsh2, and Meis2 genes. The feedback loop regulation between miR-9 and Nr2e1/Tlx helps in neuronal migration and differentiation. Targeting Foxp1 and Foxp2, and Map1b by miR-9 regulates the radial migration of neurons and axonal development. In the organizers, miR-9 is inversely regulated by hairy1 and Fgf8 to maintain zona limitans interthalamica and midbrain–hindbrain boundary (MHB). It maintains the MHB by inhibiting Fgf signaling genes and is involved in the neurogenesis of the midbrain–hindbrain by regulating Her genes. In the hindbrain, miR-9 modulates progenitor proliferation and differentiation by regulating Her genes and Elav3. In the spinal cord, miR-9 modulates the regulation of Foxp1 and Onecut1 for motor neuron development. In the forebrain, midbrain, and hindbrain, miR-9 is necessary for proper neuronal progenitor maintenance, neurogenesis, and differentiation. In vertebrate brain development, miR-9 is involved in regulating several region-specific genes in a spatiotemporal pattern.
Collapse
Affiliation(s)
| | - A Alwin Prem Anand
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Tübingen, Germany
| |
Collapse
|
166
|
Time-Course Gene Expression Profiling Reveals a Novel Role of Non-Canonical WNT Signaling During Neural Induction. Sci Rep 2016; 6:32600. [PMID: 27600186 PMCID: PMC5013468 DOI: 10.1038/srep32600] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 08/10/2016] [Indexed: 12/22/2022] Open
Abstract
The process of neuroepithelial differentiation from human pluripotent stem cells (PSCs) resembles in vivo neuroectoderm induction in the temporal course, morphogenesis, and biochemical changes. This in vitro model is therefore well-suited to reveal previously unknown molecular mechanisms underlying neural induction in humans. By transcriptome analysis of cells along PSC differentiation to early neuroepithelia at day 6 and definitive neuroepithelia at day 10, we found downregulation of genes that are associated with TGF-β and canonical WNT/β-CATENIN signaling, confirming the roles of classical signaling in human neural induction. Interestingly, WNT/Ca2+ signaling was upregulated. Pharmacological inhibition of the downstream effector of WNT/Ca2+ pathway, Ca2+/calmodulin-dependent protein kinase II (CaMKII), led to an inhibition of the neural marker PAX6 and upregulation of epidermal marker K18, suggesting that Ca2+/CaMKII signaling promotes neural induction by preventing the alternative epidermal fate. In addition, our analyses revealed known and novel expression patterns of genes that are involved in DNA methylation, histone modification, as well as epithelial-mesenchymal transition, highlighting potential roles of those genes and signaling pathways during neural differentiation.
Collapse
|
167
|
Dee A, Li K, Heng X, Guo Q, Li JYH. Regulation of self-renewing neural progenitors by FGF/ERK signaling controls formation of the inferior colliculus. Development 2016; 143:3661-3673. [PMID: 27578777 DOI: 10.1242/dev.138537] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 08/22/2016] [Indexed: 11/20/2022]
Abstract
The embryonic tectum displays an anteroposterior gradient in development and produces the superior colliculus and inferior colliculus. Studies suggest that partition of the tectum is controlled by different strengths and durations of FGF signals originated from the so-called isthmic organizer at the mid/hindbrain junction; however, the underlying mechanism is unclear. We show that deleting Ptpn11, which links FGF with the ERK pathway, prevents inferior colliculus formation by depleting a previously uncharacterized stem cell zone. The stem-zone loss is attributed to shortening of S phase and acceleration of cell cycle exit and neurogenesis. Expression of a constitutively active Mek1 (Mek1DD), the known ERK activator, restores the tectal stem zone and the inferior colliculus without Ptpn11. By contrast, Mek1DD expression fails to rescue the tectal stem zone and the inferior colliculus in the absence of Fgf8 and the isthmic organizer, indicating that FGF and Mek1DD initiate qualitatively and/or quantitatively distinctive signaling. Together, our data show that the formation of the inferior colliculus relies on the provision of new cells from the tectal stem zone. Furthermore, distinctive ERK signaling mediates Fgf8 in the control of cell survival, tissue polarity and cytogenetic gradient during the development of the tectum.
Collapse
Affiliation(s)
- Alexander Dee
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06030-6403, USA
| | - Kairong Li
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06030-6403, USA
| | - Xin Heng
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06030-6403, USA
| | - Qiuxia Guo
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06030-6403, USA
| | - James Y H Li
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06030-6403, USA .,Institute for Systems Genomics, School of Medicine, University of Connecticut Health, Farmington, CT 06030, USA
| |
Collapse
|
168
|
Ernst C. Proliferation and Differentiation Deficits are a Major Convergence Point for Neurodevelopmental Disorders. Trends Neurosci 2016; 39:290-299. [DOI: 10.1016/j.tins.2016.03.001] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 03/01/2016] [Accepted: 03/02/2016] [Indexed: 12/13/2022]
|
169
|
Denham M, Hasegawa K, Menheniott T, Rollo B, Zhang D, Hough S, Alshawaf A, Febbraro F, Ighaniyan S, Leung J, Elliott DA, Newgreen DF, Pera MF, Dottori M. Multipotent caudal neural progenitors derived from human pluripotent stem cells that give rise to lineages of the central and peripheral nervous system. Stem Cells 2016; 33:1759-70. [PMID: 25753817 PMCID: PMC5347855 DOI: 10.1002/stem.1991] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Revised: 12/25/2014] [Accepted: 01/17/2015] [Indexed: 01/22/2023]
Abstract
The caudal neural plate is a distinct region of the embryo that gives rise to major progenitor lineages of the developing central and peripheral nervous system, including neural crest and floor plate cells. We show that dual inhibition of the glycogen synthase kinase 3β and activin/nodal pathways by small molecules differentiate human pluripotent stem cells (hPSCs) directly into a preneuroepithelial progenitor population we named “caudal neural progenitors” (CNPs). CNPs coexpress caudal neural plate and mesoderm markers, and, share high similarities to embryonic caudal neural plate cells in their lineage differentiation potential. Exposure of CNPs to BMP2/4, sonic hedgehog, or FGF2 signaling efficiently directs their fate to neural crest/roof plate cells, floor plate cells, and caudally specified neuroepithelial cells, respectively. Neural crest derived from CNPs differentiated to neural crest derivatives and demonstrated extensive migratory properties in vivo. Importantly, we also determined the key extrinsic factors specifying CNPs from human embryonic stem cell include FGF8, canonical WNT, and IGF1. Our studies are the first to identify a multipotent neural progenitor derived from hPSCs, that is the precursor for major neural lineages of the embryonic caudal neural tube. Stem Cells2015;33:1759–1770
Collapse
Affiliation(s)
- Mark Denham
- Department of Anatomy and Neurosciences, University of Melbourne, Melbourne, Australia.,Danish Research Institute of Translational Neuroscience, Aarhus University, Aarhus, Denmark
| | - Kouichi Hasegawa
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan.,InStem, NCBS, Bangalore, Karnataka, India
| | | | - Ben Rollo
- Murdoch Children's Research Institute, Melbourne, Australia
| | | | - Shelley Hough
- Department of Anatomy and Neurosciences, University of Melbourne, Melbourne, Australia
| | - Abdullah Alshawaf
- Centre for Neural Engineering, Department of Electrical and Electronic Engineering, University of Melbourne, Melbourne, Australia
| | - Fabia Febbraro
- Danish Research Institute of Translational Neuroscience, Aarhus University, Aarhus, Denmark
| | | | - Jessie Leung
- Department of Anatomy and Neurosciences, University of Melbourne, Melbourne, Australia
| | | | | | - Martin F Pera
- Department of Anatomy and Neurosciences, University of Melbourne, Melbourne, Australia.,Walter and Eliza Hall Institute, Melbourne, Australia.,Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
| | - Mirella Dottori
- Department of Anatomy and Neurosciences, University of Melbourne, Melbourne, Australia.,Centre for Neural Engineering, Department of Electrical and Electronic Engineering, University of Melbourne, Melbourne, Australia
| |
Collapse
|
170
|
Humann J, Mann B, Gao G, Moresco P, Ramahi J, Loh LN, Farr A, Hu Y, Durick-Eder K, Fillon SA, Smeyne RJ, Tuomanen EI. Bacterial Peptidoglycan Traverses the Placenta to Induce Fetal Neuroproliferation and Aberrant Postnatal Behavior. Cell Host Microbe 2016; 19:388-99. [PMID: 26962947 PMCID: PMC4787272 DOI: 10.1016/j.chom.2016.02.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 02/04/2016] [Accepted: 02/21/2016] [Indexed: 11/26/2022]
Abstract
Maternal infection during pregnancy is associated with adverse outcomes for the fetus, including postnatal cognitive disorders. However, the underlying mechanisms are obscure. We find that bacterial cell wall peptidoglycan (CW), a universal PAMP for TLR2, traverses the murine placenta into the developing fetal brain. In contrast to adults, CW-exposed fetal brains did not show any signs of inflammation or neuronal death. Instead, the neuronal transcription factor FoxG1 was induced, and neuroproliferation leading to a 50% greater density of neurons in the cortical plate was observed. Bacterial infection of pregnant dams, followed by antibiotic treatment, which releases CW, yielded the same result. Neuroproliferation required TLR2 and was recapitulated in vitro with fetal neuronal precursor cells and TLR2/6, but not TLR2/1, ligands. The fetal neuroproliferative response correlated with abnormal cognitive behavior in CW-exposed pups following birth. Thus, the bacterial CW-TLR2 signaling axis affects fetal neurodevelopment and may underlie postnatal cognitive disorders.
Collapse
Affiliation(s)
- Jessica Humann
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Beth Mann
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Geli Gao
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Philip Moresco
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Joseph Ramahi
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Lip Nam Loh
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Arden Farr
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yunming Hu
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kelly Durick-Eder
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Sophie A Fillon
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Richard J Smeyne
- Department of Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Elaine I Tuomanen
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
171
|
Wang W, Jossin Y, Chai G, Lien WH, Tissir F, Goffinet AM. Feedback regulation of apical progenitor fate by immature neurons through Wnt7-Celsr3-Fzd3 signalling. Nat Commun 2016; 7:10936. [PMID: 26939553 PMCID: PMC4786577 DOI: 10.1038/ncomms10936] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 02/03/2016] [Indexed: 11/09/2022] Open
Abstract
Sequential generation of neurons and glial cells during development is critical for the wiring and function of the cerebral cortex. This process requires accurate coordination of neural progenitor cell (NPC) fate decisions, by NPC-autonomous mechanisms as well as by negative feedback from neurons. Here, we show that neurogenesis is protracted and gliogenesis decreased in mice with mutations of genes Celsr3 and Fzd3. This phenotype is not due to gene inactivation in progenitors, but rather in immature cortical neurons. Mutant neurons are unable to upregulate expression of Jag1 in response to cortical Wnt7, resulting in blunted activation of Notch signalling in NPC. Thus, Celsr3 and Fzd3 enable immature neurons to respond to Wnt7, upregulate Jag1 and thereby facilitate feedback signals that tune the timing of NPC fate decisions via Notch activation.
Collapse
Affiliation(s)
- Wei Wang
- Université catholique de Louvain, Institute of Neuroscience, 73 Avenue Mounier, Box B1.7316, 1200 Brussels, Belgium
| | - Yves Jossin
- Université catholique de Louvain, Institute of Neuroscience, 73 Avenue Mounier, Box B1.7316, 1200 Brussels, Belgium
| | - Guoliang Chai
- Université catholique de Louvain, Institute of Neuroscience, 73 Avenue Mounier, Box B1.7316, 1200 Brussels, Belgium
| | - Wen-Hui Lien
- Université catholique de Louvain, de Duve Institute, Avenue Hippocrate 74, Box B1.74.09, 1200 Brussels, Belgium
| | - Fadel Tissir
- Université catholique de Louvain, Institute of Neuroscience, 73 Avenue Mounier, Box B1.7316, 1200 Brussels, Belgium
| | - Andre M Goffinet
- Université catholique de Louvain, Institute of Neuroscience, 73 Avenue Mounier, Box B1.7316, 1200 Brussels, Belgium.,WELBIO, 6 Avenue Pasteur, 1300 Wavre, Belgium
| |
Collapse
|
172
|
Retinoic Acid Receptor β Controls Development of Striatonigral Projection Neurons through FGF-Dependent and Meis1-Dependent Mechanisms. J Neurosci 2016; 35:14467-75. [PMID: 26511239 DOI: 10.1523/jneurosci.1278-15.2015] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The mammalian striatum controls sensorimotor and psychoaffective functions through coordinated activities of its two striatonigral and striatopallidal output pathways. Here we show that retinoic acid receptor β (RARβ) controls development of a subpopulation of GABAergic, Gad65-positive striatonigral projection neurons. In Rarb(-/-) knock-out mice, concomitant reduction of Gad65, dopamine receptor D1 (Drd1), and substance P expression at different phases of prenatal development was associated with reduced number of Drd1-positive cells at birth, in contrast to normal numbers of striatopallidal projection neurons expressing dopamine receptor D2. Fate mapping using BrdU pulse-chase experiments revealed that such deficits may originate from compromised proliferation of late-born striosomal neurons and lead to decreased number of Drd1-positive cells retaining BrdU in postnatal day (P) 0 Rarb(-/-) striatum. Reduced expression of Fgf3 in the subventricular zone of the lateral ganglionic eminence (LGE) at embryonic day 13.5 may underlie such deficits by inducing premature differentiation of neuronal progenitors, as illustrated by reduced expression of the proneural gene Ascl1 (Mash1) and increased expression of Meis1, a marker of postmitotic LGE neurons. In agreement with a critical role of FGF3 in this control, reduced number of Ascl1-expressing neural progenitors, and a concomitant increase of Meis1-expressing cells, were observed in primary cell cultures of Rarb(-/-) LGE. This defect was normalized by addition of fibroblast growth factor (FGF). Such data point to role of Meis1 in striatal development, also supported by reduced neuronal differentiation in the LGE of Meis1(-/-) embryos. Our data unveil a novel mechanism of development of striatonigral projection neurons involving retinoic acid and FGF, two signals required for positioning the boundaries of Meis1-expressing cells.
Collapse
|
173
|
Frullanti E, Amabile S, Lolli MG, Bartolini A, Livide G, Landucci E, Mari F, Vaccarino FM, Ariani F, Massimino L, Renieri A, Meloni I. Altered expression of neuropeptides in FoxG1-null heterozygous mutant mice. Eur J Hum Genet 2016; 24:252-7. [PMID: 25966633 PMCID: PMC4717204 DOI: 10.1038/ejhg.2015.79] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 03/13/2015] [Accepted: 03/19/2015] [Indexed: 12/17/2022] Open
Abstract
Foxg1 gene encodes for a transcription factor essential for telencephalon development in the embryonic mammalian forebrain. Its complete absence is embryonic lethal while Foxg1 heterozygous mice are viable but display microcephaly, altered hippocampal neurogenesis and behavioral and cognitive deficiencies. In order to evaluate the effects of Foxg1 alteration in adult brain, we performed expression profiling in total brains from Foxg1+/- heterozygous mutants and wild-type littermates. We identified statistically significant differences in expression levels for 466 transcripts (P<0.001), 29 of which showed a fold change ≥ 1.5. Among the differentially expressed genes was found a group of genes expressed in the basal ganglia and involved in the control of movements. A relevant (three to sevenfold changes) and statistically significant increase of expression, confirmed by qRT-PCR, was found in two highly correlated genes with expression restricted to the hypothalamus: Oxytocin (Oxt) and Arginine vasopressin (Avp). These neuropeptides have an important role in maternal and social behavior, and their alteration is associated with impaired social interaction and autistic behavior. In addition, Neuronatin (Nnat) levels appear significantly higher both in Foxg1+/- whole brain and in hippocampal neurons after silencing Foxg1, strongly suggesting that it is directly or indirectly repressed by Foxg1. During fetal and neonatal brain development, Nnat may regulate neuronal excitability, receptor trafficking and calcium-dependent signaling and, in the adult brain, it is predominantly expressed in parvalbumin-positive GABAergic interneurons. Overall, these results implicate the overexpression of a group of neuropeptides in the basal ganglia, hypothalamus, cortex and hippocampus in the pathogenesis FOXG1 behavioral impairments.
Collapse
Affiliation(s)
- Elisa Frullanti
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | | | | | - Anna Bartolini
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Gabriella Livide
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | | | - Francesca Mari
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
- Medical Genetics, University of Siena, Siena, Italy
| | - Flora M Vaccarino
- Child Study Center and Department of Neurobiology, Yale University, New Haven, CT, USA
| | - Francesca Ariani
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
- Medical Genetics, University of Siena, Siena, Italy
| | | | - Alessandra Renieri
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
- Medical Genetics, University of Siena, Siena, Italy
| | | |
Collapse
|
174
|
Precious SV, Kelly CM, Allen ND, Rosser AE. Can manipulation of differentiation conditions eliminate proliferative cells from a population of ES cell-derived forebrain cells? NEUROGENESIS 2016; 3:e1127311. [PMID: 27606335 PMCID: PMC4973593 DOI: 10.1080/23262133.2015.1127311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 11/24/2015] [Accepted: 11/30/2015] [Indexed: 11/12/2022]
Abstract
There is preliminary evidence that implantation of primary fetal striatal cells provides functional benefit in patients with Huntington's disease, a neurodegenerative condition resulting in loss of medium-sized spiny neurons (MSN) of the striatum. Scarcity of primary fetal tissue means it is important to identify a renewable source of cells from which to derive donor MSNs. Embryonic stem (ES) cells, which predominantly default to telencephalic-like precursors in chemically defined medium (CDM), offer a potentially inexhaustible supply of cells capable of generating the desired neurons. Using an ES cell line, with the forebrain marker FoxG1 tagged to the LacZ reporter, we assessed effects of known developmental factors on the yield of forebrain-like precursor cells in CDM suspension culture. Addition of FGF2, but not DKK1, increased the proportion of FoxG1-expressing cells at day 8 of neural induction. Oct4 was expressed at day 8, but was undetectable by day 16. Differentiation of day 16 precursors generated GABA-expressing neurons, with few DARPP32 positive MSNs. Transplantation of day 8 precursor cells into quinolinic acid-lesioned striata resulted in generation of teratomas. However, transplantation of day 16 precursors yielded grafts expressing neuronal markers including NeuN, calbindin and parvalbumin, but no DARPP32 6 weeks post-transplantation. Manipulation of fate of ES cells requires optimization of both concentration and timing of addition of factors to culture systems to generate the desired phenotypes. Furthermore, we highlight the value of increasing the precursor phase of ES cell suspension culture when directing differentiation toward forebrain fate, so as to dramatically reduce the risk of teratoma formation.
Collapse
Affiliation(s)
- Sophie V Precious
- Brain Repair Group, School of Biosciences, Cardiff University , Cardiff, UK
| | - Claire M Kelly
- Brain Repair Group, School of Biosciences, Cardiff University , Cardiff, UK
| | - Nicholas D Allen
- Genetics Group, School of Biosciences, Cardiff University , Cardiff, UK
| | - Anne E Rosser
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, UK; Departments of Neurology and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, UK
| |
Collapse
|
175
|
Reconciling diverse mammalian pigmentation patterns with a fundamental mathematical model. Nat Commun 2016; 7:10288. [PMID: 26732977 PMCID: PMC4729835 DOI: 10.1038/ncomms10288] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 11/26/2015] [Indexed: 12/18/2022] Open
Abstract
Bands of colour extending laterally from the dorsal to ventral trunk are a common feature of mouse chimeras. These stripes were originally taken as evidence of the directed dorsoventral migration of melanoblasts (the embryonic precursors of melanocytes) as they colonize the developing skin. Depigmented ‘belly spots' in mice with mutations in the receptor tyrosine kinase Kit are thought to represent a failure of this colonization, either due to impaired migration or proliferation. Tracing of single melanoblast clones, however, has revealed a diffuse distribution with high levels of axial mixing—hard to reconcile with directed migration. Here we construct an agent-based stochastic model calibrated by experimental measurements to investigate the formation of diffuse clones, chimeric stripes and belly spots. Our observations indicate that melanoblast colonization likely proceeds through a process of undirected migration, proliferation and tissue expansion, and that reduced proliferation is the cause of the belly spots in Kit mutants. How embryonic melanoblast behaviour influences adult pigmentation patterns and causes patterning defects is unclear. Here, Mort et al. construct a stochastic model parameterised experimentally to show that melanoblast migration is undirected and that reduced proliferation causes patterning defects.
Collapse
|
176
|
Thuret R, Auger H, Papalopulu N. Analysis of neural progenitors from embryogenesis to juvenile adult in Xenopus laevis reveals biphasic neurogenesis and continuous lengthening of the cell cycle. Biol Open 2015; 4:1772-81. [PMID: 26621828 PMCID: PMC4736028 DOI: 10.1242/bio.013391] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Xenopus laevis is a prominent model system for studying neural development, but our understanding of the long-term temporal dynamics of neurogenesis remains incomplete. Here, we present the first continuous description of neurogenesis in X. laevis, covering the entire period of development from the specification of neural ectoderm during gastrulation to juvenile frog. We have used molecular markers to identify progenitors and neurons, short-term bromodeoxyuridine (BrdU) incorporation to map the generation of newborn neurons and dual pulse S-phase labelling to characterise changes in their cell cycle length. Our study revealed the persistence of Sox3-positive progenitor cells from the earliest stages of neural development through to the juvenile adult. Two periods of intense neuronal generation were observed, confirming the existence of primary and secondary waves of neurogenesis, punctuated by a period of quiescence before metamorphosis and culminating in another period of quiescence in the young adult. Analysis of multiple parameters indicates that neural progenitors alternate between global phases of differentiation and amplification and that, regardless of their behaviour, their cell cycle lengthens monotonically during development, at least at the population level.
Collapse
Affiliation(s)
- Raphaël Thuret
- Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Hélène Auger
- Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Nancy Papalopulu
- Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
177
|
FOXG1-Dependent Dysregulation of GABA/Glutamate Neuron Differentiation in Autism Spectrum Disorders. Cell 2015; 162:375-390. [PMID: 26186191 DOI: 10.1016/j.cell.2015.06.034] [Citation(s) in RCA: 761] [Impact Index Per Article: 84.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 03/27/2015] [Accepted: 05/29/2015] [Indexed: 12/19/2022]
Abstract
Autism spectrum disorder (ASD) is a disorder of brain development. Most cases lack a clear etiology or genetic basis, and the difficulty of re-enacting human brain development has precluded understanding of ASD pathophysiology. Here we use three-dimensional neural cultures (organoids) derived from induced pluripotent stem cells (iPSCs) to investigate neurodevelopmental alterations in individuals with severe idiopathic ASD. While no known underlying genomic mutation could be identified, transcriptome and gene network analyses revealed upregulation of genes involved in cell proliferation, neuronal differentiation, and synaptic assembly. ASD-derived organoids exhibit an accelerated cell cycle and overproduction of GABAergic inhibitory neurons. Using RNA interference, we show that overexpression of the transcription factor FOXG1 is responsible for the overproduction of GABAergic neurons. Altered expression of gene network modules and FOXG1 are positively correlated with symptom severity. Our data suggest that a shift toward GABAergic neuron fate caused by FOXG1 is a developmental precursor of ASD.
Collapse
|
178
|
Patriarchi T, Amabile S, Frullanti E, Landucci E, Lo Rizzo C, Ariani F, Costa M, Olimpico F, W Hell J, M Vaccarino F, Renieri A, Meloni I. Imbalance of excitatory/inhibitory synaptic protein expression in iPSC-derived neurons from FOXG1(+/-) patients and in foxg1(+/-) mice. Eur J Hum Genet 2015; 24:871-80. [PMID: 26443267 DOI: 10.1038/ejhg.2015.216] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Revised: 08/24/2015] [Accepted: 09/01/2015] [Indexed: 01/12/2023] Open
Abstract
Rett syndrome (RTT) is a severe neurodevelopmental disorder associated with mutations in either MECP2, CDKL5 or FOXG1. The precise molecular mechanisms that lead to the pathogenesis of RTT have yet to be elucidated. We recently reported that expression of GluD1 (orphan glutamate receptor δ-1 subunit) is increased in iPSC-derived neurons obtained from patients with mutations in either MECP2 or CDKL5. GluD1 controls synaptic differentiation and shifts the balance between excitatory and inhibitory synapses toward the latter. Thus, an increase in GluD1 might be a critical factor in the etiology of RTT by affecting the excitatory/inhibitory balance in the developing brain. To test this hypothesis, we generated iPSC-derived neurons from FOXG1(+/-) patients. We analyzed mRNA and protein levels of GluD1 together with key markers of excitatory and inhibitory synapses in these iPSC-derived neurons and in Foxg1(+/-) mouse fetal (E11.5) and adult (P70) brains. We found strong correlation between iPSC-derived neurons and fetal mouse brains, where GluD1 and inhibitory synaptic markers (GAD67 and GABA AR-α1) were increased, whereas the levels of a number of excitatory synaptic markers (VGLUT1, GluA1, GluN1 and PSD-95) were decreased. In adult mice, GluD1 was decreased along with all GABAergic and glutamatergic markers. Our findings further the understanding of the etiology of RTT by introducing a new pathological event occurring in the brain of FOXG1(+/-) patients during embryonic development and its time-dependent shift toward a general decrease in brain synapses.
Collapse
Affiliation(s)
- Tommaso Patriarchi
- Medical Genetics, University of Siena, Siena, Italy.,Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | | | - Elisa Frullanti
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | | | | | - Francesca Ariani
- Medical Genetics, University of Siena, Siena, Italy.,Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Mario Costa
- Institute of Neuroscience, Italian National Research Council, Pisa, Italy.,Scuola Normale Superiore, Pisa, Italy
| | - Francesco Olimpico
- Institute of Neuroscience, Italian National Research Council, Pisa, Italy
| | - Johannes W Hell
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | - Flora M Vaccarino
- Child Study Center, Yale University School of Medicine, New Haven, CT, USA.,Department of Neurobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Alessandra Renieri
- Medical Genetics, University of Siena, Siena, Italy.,Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | | |
Collapse
|
179
|
Hernández-Bejarano M, Gestri G, Spawls L, Nieto-López F, Picker A, Tada M, Brand M, Bovolenta P, Wilson SW, Cavodeassi F. Opposing Shh and Fgf signals initiate nasotemporal patterning of the zebrafish retina. Development 2015; 142:3933-42. [PMID: 26428010 PMCID: PMC4712879 DOI: 10.1242/dev.125120] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 09/21/2015] [Indexed: 01/12/2023]
Abstract
The earliest known determinants of retinal nasotemporal identity are the transcriptional regulators Foxg1, which is expressed in the prospective nasal optic vesicle, and Foxd1, which is expressed in the prospective temporal optic vesicle. Previous work has shown that, in zebrafish, Fgf signals from the dorsal forebrain and olfactory primordia are required to specify nasal identity in the dorsal, prospective nasal, optic vesicle. Here, we show that Hh signalling from the ventral forebrain is required for specification of temporal identity in the ventral optic vesicle and is sufficient to induce temporal character when activated in the prospective nasal retina. Consequently, the evaginating optic vesicles become partitioned into prospective nasal and temporal domains by the opposing actions of Fgfs and Shh emanating from dorsal and ventral domains of the forebrain primordium. In absence of Fgf activity, foxd1 expression is established irrespective of levels of Hh signalling, indicating that the role of Shh in promoting foxd1 expression is only required in the presence of Fgf activity. Once the spatially complementary expression of foxd1 and foxg1 is established, the boundary between expression domains is maintained by mutual repression between Foxd1 and Foxg1. Summary: In the fish eye, Hh signalling from the ventral forebrain regulates spatial identity in the retina by promoting foxd1 expression. This role is required only in the presence of Fgf activity.
Collapse
Affiliation(s)
| | - Gaia Gestri
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1 6BT, UK
| | - Lana Spawls
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1 6BT, UK
| | - Francisco Nieto-López
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), C/Nicolás Cabrera 1, 28049, Madrid, Spain
| | - Alexander Picker
- Center of Regenerative Therapies Dresden (CRTD), Biotechnology Center, Dresden University of Technology, 01062 Dresden, Germany
| | - Masazumi Tada
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1 6BT, UK
| | - Michael Brand
- Center of Regenerative Therapies Dresden (CRTD), Biotechnology Center, Dresden University of Technology, 01062 Dresden, Germany
| | - Paola Bovolenta
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), C/Nicolás Cabrera 1, 28049, Madrid, Spain CIBER de Enfermedades Raras (CIBERER), C/Nicolás Cabrera 1, 28049, Madrid, Spain
| | - Stephen W Wilson
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1 6BT, UK
| | - Florencia Cavodeassi
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), C/Nicolás Cabrera 1, 28049, Madrid, Spain CIBER de Enfermedades Raras (CIBERER), C/Nicolás Cabrera 1, 28049, Madrid, Spain
| |
Collapse
|
180
|
McMahon KQ, Papandreou A, Ma M, Barry BJ, Mirzaa GM, Dobyns WB, Scott RH, Trump N, Kurian MA, Paciorkowski AR. Familial recurrences of FOXG1-related disorder: Evidence for mosaicism. Am J Med Genet A 2015; 167A:3096-102. [PMID: 26364767 DOI: 10.1002/ajmg.a.37353] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 08/13/2015] [Indexed: 12/18/2022]
Abstract
FOXG1-related disorders are caused by heterozygous mutations in FOXG1 and result in a spectrum of neurodevelopmental phenotypes including postnatal microcephaly, intellectual disability with absent speech, epilepsy, chorea, and corpus callosum abnormalities. The recurrence risk for de novo mutations in FOXG1-related disorders is assumed to be low. Here, we describe three unrelated sets of full siblings with mutations in FOXG1 (c.515_577del63, c.460dupG, and c.572T > G), representing familial recurrence of the disorder. In one family, we have documented maternal somatic mosaicism for the FOXG1 mutation, and all of the families presumably represent parental gonadal (or germline) mosaicism. To our knowledge, mosaicism has not been previously reported in FOXG1-related disorders. Therefore, this report provides evidence that germline mosaicism for FOXG1 mutations is a likely explanation for familial recurrence and should be considered during recurrence risk counseling for families of children with FOXG1-related disorders.
Collapse
Affiliation(s)
- Kelly Q McMahon
- Department of Neurology, University of Rochester Medical Center, Rochester, New York
| | - Apostolos Papandreou
- Developmental Neurosciences, UCL-Institute of Child Health, London, United Kingdom.,Department of Neurology, Great Ormond Street Hospital, London, United Kingdom.,Genetics and Genomics Medicine, UCL-Institute of Child Health, London, United Kingdom
| | - Mandy Ma
- University of Buffalo School of Medicine, Buffalo, New York
| | | | - Ghayda M Mirzaa
- Department of Pediatrics, Division of Genetic Medicine, University of Washington, Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington
| | - William B Dobyns
- Department of Pediatrics, Division of Genetic Medicine, University of Washington, Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington
| | - Richard H Scott
- Genetics and Genomics Medicine, UCL-Institute of Child Health, London, United Kingdom.,North East Thames Regional Genetics Service, Great Ormond Street Hospital, London, United Kingdom
| | - Natalie Trump
- North East Thames Regional Genetics Service, Great Ormond Street Hospital, London, United Kingdom
| | - Manju A Kurian
- Developmental Neurosciences, UCL-Institute of Child Health, London, United Kingdom.,Department of Neurology, Great Ormond Street Hospital, London, United Kingdom
| | - Alex R Paciorkowski
- Department of Neurology, University of Rochester Medical Center, Rochester, New York.,Departments of Pediatrics and Biomedical Genetics, Center for Neural Development and Disease, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
181
|
Genomic DISC1 Disruption in hiPSCs Alters Wnt Signaling and Neural Cell Fate. Cell Rep 2015; 12:1414-29. [PMID: 26299970 DOI: 10.1016/j.celrep.2015.07.061] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 07/08/2015] [Accepted: 07/29/2015] [Indexed: 02/08/2023] Open
Abstract
Genetic and clinical association studies have identified disrupted in schizophrenia 1 (DISC1) as a candidate risk gene for major mental illness. DISC1 is interrupted by a balanced chr(1;11) translocation in a Scottish family in which the translocation predisposes to psychiatric disorders. We investigate the consequences of DISC1 interruption in human neural cells using TALENs or CRISPR-Cas9 to target the DISC1 locus. We show that disruption of DISC1 near the site of the translocation results in decreased DISC1 protein levels because of nonsense-mediated decay of long splice variants. This results in an increased level of canonical Wnt signaling in neural progenitor cells and altered expression of fate markers such as Foxg1 and Tbr2. These gene expression changes are rescued by antagonizing Wnt signaling in a critical developmental window, supporting the hypothesis that DISC1-dependent suppression of basal Wnt signaling influences the distribution of cell types generated during cortical development.
Collapse
|
182
|
Pandit T, Jidigam VK, Patthey C, Gunhaga L. Neural retina identity is specified by lens-derived BMP signals. Development 2015; 142:1850-9. [PMID: 25968316 PMCID: PMC4440930 DOI: 10.1242/dev.123653] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The eye has served as a classical model to study cell specification and tissue induction for over a century. Nevertheless, the molecular mechanisms that regulate the induction and maintenance of eye-field cells, and the specification of neural retina cells are poorly understood. Moreover, within the developing anterior forebrain, how prospective eye and telencephalic cells are differentially specified is not well defined. In the present study, we have analyzed these issues by manipulating signaling pathways in intact chick embryo and explant assays. Our results provide evidence that at blastula stages, BMP signals inhibit the acquisition of eye-field character, but from neural tube/optic vesicle stages, BMP signals from the lens are crucial for the maintenance of eye-field character, inhibition of dorsal telencephalic cell identity and specification of neural retina cells. Subsequently, our results provide evidence that a Rax2-positive eye-field state is not sufficient for the progress to a neural retina identity, but requires BMP signals. In addition, our results argue against any essential role of Wnt or FGF signals during the specification of neural retina cells, but provide evidence that Wnt signals together with BMP activity are sufficient to induce cells of retinal pigment epithelial character. We conclude that BMP activity emanating from the lens ectoderm maintains eye-field identity, inhibits telencephalic character and induces neural retina cells. Our findings link the requirement of the lens ectoderm for neural retina specification with the molecular mechanism by which cells in the forebrain become specified as neural retina by BMP activity. SUMMARY: BMP signals from the lens are crucial to maintain eye-field character, inhibit dorsal telencephalic cell identity, and specificy neural retina cells in chick embryos.
Collapse
Affiliation(s)
- Tanushree Pandit
- Umeå Centre for Molecular Medicine, Umeå University, Umeå 901 87, Sweden
| | - Vijay K Jidigam
- Umeå Centre for Molecular Medicine, Umeå University, Umeå 901 87, Sweden
| | - Cedric Patthey
- Umeå Centre for Molecular Medicine, Umeå University, Umeå 901 87, Sweden
| | - Lena Gunhaga
- Umeå Centre for Molecular Medicine, Umeå University, Umeå 901 87, Sweden
| |
Collapse
|
183
|
Heide M, Zhang Y, Zhou X, Zhao T, Miquelajáuregui A, Varela-Echavarría A, Alvarez-Bolado G. Lhx5 controls mamillary differentiation in the developing hypothalamus of the mouse. Front Neuroanat 2015; 9:113. [PMID: 26321924 PMCID: PMC4536661 DOI: 10.3389/fnana.2015.00113] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 07/30/2015] [Indexed: 12/30/2022] Open
Abstract
Acquisition of specific neuronal identity by individual brain nuclei is a key step in brain development. However, how the mechanisms that confer neuronal identity are integrated with upstream regional specification networks is still mysterious. Expression of Sonic hedgehog (Shh), is required for hypothalamic specification and is later downregulated by Tbx3 to allow for the differentiation of the tubero-mamillary region. In this region, the mamillary body (MBO), is a large neuronal aggregate essential for memory formation. To clarify how MBO identity is acquired after regional specification, we investigated Lhx5, a transcription factor with restricted MBO expression. We first generated a hypomorph allele of Lhx5—in homozygotes, the MBO disappears after initial specification. Intriguingly, in these mutants, Tbx3 was downregulated and the Shh expression domain abnormally extended. Microarray analysis and chromatin immunoprecipitation indicated that Lhx5 appears to be involved in Shh downregulation through Tbx3 and activates several MBO-specific regulator and effector genes. Finally, by tracing the caudal hypothalamic cell lineage we show that, in the Lhx5 mutant, at least some MBO cells are present but lack characteristic marker expression. Our work shows how the Lhx5 locus contributes to integrate regional specification pathways with downstream acquisition of neuronal identity in the MBO.
Collapse
Affiliation(s)
- Michael Heide
- Institute of Anatomy and Cell Biology, University of Heidelberg Heidelberg, Germany
| | - Yuanfeng Zhang
- Institute of Anatomy and Cell Biology, University of Heidelberg Heidelberg, Germany
| | - Xunlei Zhou
- Institute of Anatomy and Cell Biology, University of Heidelberg Heidelberg, Germany
| | - Tianyu Zhao
- Key Laboratory of Oral Disease and Biomedical Sciences, Stomatological Hospital, Chongqing Medical University Chongqing, China
| | - Amaya Miquelajáuregui
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México Querétaro, Mexico
| | - Alfredo Varela-Echavarría
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México Querétaro, Mexico
| | | |
Collapse
|
184
|
Wong FK, Fei JF, Mora-Bermúdez F, Taverna E, Haffner C, Fu J, Anastassiadis K, Stewart AF, Huttner WB. Sustained Pax6 Expression Generates Primate-like Basal Radial Glia in Developing Mouse Neocortex. PLoS Biol 2015; 13:e1002217. [PMID: 26252244 PMCID: PMC4529158 DOI: 10.1371/journal.pbio.1002217] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 06/30/2015] [Indexed: 11/21/2022] Open
Abstract
The evolutionary expansion of the neocortex in mammals has been linked to enlargement of the subventricular zone (SVZ) and increased proliferative capacity of basal progenitors (BPs), notably basal radial glia (bRG). The transcription factor Pax6 is known to be highly expressed in primate, but not mouse, BPs. Here, we demonstrate that sustaining Pax6 expression selectively in BP-genic apical radial glia (aRG) and their BP progeny of embryonic mouse neocortex suffices to induce primate-like progenitor behaviour. Specifically, we conditionally expressed Pax6 by in utero electroporation using a novel, Tis21–CreERT2 mouse line. This expression altered aRG cleavage plane orientation to promote bRG generation, increased cell-cycle re-entry of BPs, and ultimately increased upper-layer neuron production. Upper-layer neuron production was also increased in double-transgenic mouse embryos with sustained Pax6 expression in the neurogenic lineage. Strikingly, increased BPs existed not only in the SVZ but also in the intermediate zone of the neocortex of these double-transgenic mouse embryos. In mutant mouse embryos lacking functional Pax6, the proportion of bRG among BPs was reduced. Our data identify specific Pax6 effects in BPs and imply that sustaining this Pax6 function in BPs could be a key aspect of SVZ enlargement and, consequently, the evolutionary expansion of the neocortex. "Humanizing" the expression of the transcription factor Pax6 in cortical progenitors in the developing mouse brain is sufficient to endow these progenitors with a primate-like proliferative capacity. During development, neural progenitors generate all cells that make up the mammalian brain. Differences in brain size among the various mammalian species are attributed to differences in the abundance and proliferative capacity of a specific class of neural progenitors called basal progenitors. Among these, a specific progenitor type called basal radial glia is thought to have played an important role during evolution in the expansion of the neocortex, the part of the brain associated with higher cognitive functions like conscious thought and language. In the neocortex, the expression of the transcription factor Pax6 in basal progenitors is low in rodents, but high in primates, including humans. In this study, we aimed to mimic the elevated expression pattern of Pax6 seen in humans in basal progenitors of the embryonic mouse neocortex. To this end, we generated a novel, transgenic mouse line that allows sustained expression of the Pax6 gene in basal progenitors. This elevated expression resulted in an increase in the generation of basal radial glia, in the proliferative capacity of basal progenitors, and, ultimately, in the number of neurons produced. Our findings demonstrate that altering the expression of a single transcription factor from a mouse to a human-like pattern suffices to induce a primate-like proliferative behaviour in neural progenitors, which is thought to underlie the evolutionary expansion of the neocortex.
Collapse
Affiliation(s)
- Fong Kuan Wong
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Ji-Feng Fei
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | - Elena Taverna
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Christiane Haffner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Jun Fu
- Biotechnology Center of the Technische Universität Dresden, Dresden, Germany
| | | | - A. Francis Stewart
- Biotechnology Center of the Technische Universität Dresden, Dresden, Germany
| | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- * E-mail:
| |
Collapse
|
185
|
Abstract
High-content imaging is a powerful tool for determining cell phenotypes at the single cell level. Characterising the effect of small molecules on cell cycle distribution is important for understanding their mechanism of action especially in oncology drug discovery but also for understanding potential toxicology liabilities. Here, a high-throughput phenotypic assay utilising the PerkinElmer Operetta high-content imager and Harmony software to determine cell cycle distribution is described. PhenoLOGIC, a machine learning algorithm within Harmony software was employed to robustly separate single cells from cell clumps. DNA content, EdU incorporation and pHH3 (S10) expression levels were subsequently utilised to separate cells into the various phases of the cell cycle. The assay is amenable to multiplexing with an additional pharmacodynamic marker to assess cell cycle changes within a specific cellular sub-population. Using this approach, the cell cycle distribution of γH2AX positive nuclei was determined following treatment with DNA damaging agents. Likewise, the assay can be multiplexed with Ki67 to determine the fraction of quiescent cells and with BrdU dual labelling to determine S-phase duration. This methodology therefore provides a relatively cheap, quick and high-throughput phenotypic method for determining accurate cell cycle distribution for small molecule mechanism of action and drug toxicity studies.
Collapse
|
186
|
Fimiani C, Goina E, Mallamaci A. Upregulating endogenous genes by an RNA-programmable artificial transactivator. Nucleic Acids Res 2015; 43:7850-64. [PMID: 26152305 PMCID: PMC4652751 DOI: 10.1093/nar/gkv682] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 06/22/2015] [Indexed: 11/12/2022] Open
Abstract
To promote expression of endogenous genes ad libitum, we developed a novel, programmable transcription factor prototype. Kept together via an MS2 coat protein/RNA interface, it includes a fixed, polypeptidic transactivating domain and a variable RNA domain that recognizes the desired gene. Thanks to this device, we specifically upregulated five genes, in cell lines and primary cultures of murine pallial precursors. Gene upregulation was small, however sufficient to robustly inhibit neuronal differentiation. The transactivator interacted with target gene chromatin via its RNA cofactor. Its activity was restricted to cells in which the target gene is normally transcribed. Our device might be useful for specific applications. However for this purpose, it will require an improvement of its transactivation power as well as a better characterization of its target specificity and mechanism of action.
Collapse
Affiliation(s)
- Cristina Fimiani
- Laboratory of Cerebral Cortex Development, SISSA, Trieste, 34136, Italy
| | - Elisa Goina
- Laboratory of Cerebral Cortex Development, SISSA, Trieste, 34136, Italy
| | | |
Collapse
|
187
|
Green Synthesis of Silver and Titanium Dioxide Nanoparticles Using Euphorbia prostrata Extract Shows Shift from Apoptosis to G0/G1 Arrest followed by Necrotic Cell Death in Leishmania donovani. Antimicrob Agents Chemother 2015; 59:4782-99. [PMID: 26033724 DOI: 10.1128/aac.00098-15] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 05/18/2015] [Indexed: 11/20/2022] Open
Abstract
The aim of the present study was to synthesize silver (Ag) and titanium dioxide (TiO2) nanoparticles (NPs) using green synthesis from aqueous leaf extract of Euphorbia prostrata as antileishmanial agents and to explore the underlying molecular mechanism of induced cell death. In vitro antileishmanial activity of synthesized NPs was tested against promastigotes of Leishmania donovani by alamarBlue and propidium iodide uptake assays. Antileishmanial activity of synthesized NPs on intracellular amastigotes was assessed by Giemsa staining. The leishmanicidal effect of synthesized Ag NPs was further confirmed by DNA fragmentation assay and by cell cycle progression and transmission electron microscopy (TEM) of the treated parasites. TEM analysis of the synthesized Ag NPs showed a spherical shape with an average size of 12.82 ± 2.50 nm, and in comparison to synthesized TiO2 NPs, synthesized Ag NPs were found to be most active against Leishmania parasites after 24 h exposure, with 50% inhibitory concentrations (IC50) of 14.94 μg/ml and 3.89 μg/ml in promastigotes and intracellular amastigotes, respectively. A significant increase in G0/G1 phase of the cell cycle with a subsequent decrease in S (synthesis) and G2/M phases compared to controls was observed. The growth-inhibitory effect of synthesized Ag NPs was attributed to increased length of S phase. A decreased reactive oxygen species level was also observed, which could be responsible for the caspase-independent shift from apoptosis (G0/G1 arrest) to massive necrosis. High-molecular-weight DNA fragmentation as a positive consequence of necrotic cell death was also visualized. We also report that the unique trypanothione/trypanothione reductase (TR) system of Leishmania cells was significantly inhibited by synthesized Ag NPs. The green-synthesized Ag NPs may provide promising leads for the development of cost-effective and safer alternative treatment against visceral leishmaniasis.
Collapse
|
188
|
Morishita Y, Kuroiwa A, Suzuki T. Quantitative analysis of tissue deformation dynamics reveals three characteristic growth modes and globally aligned anisotropic tissue deformation during chick limb development. Development 2015; 142:1672-83. [PMID: 25858459 PMCID: PMC4419272 DOI: 10.1242/dev.109728] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Accepted: 03/02/2015] [Indexed: 01/30/2023]
Abstract
Tissue-level characterization of deformation dynamics is crucial for understanding organ morphogenetic mechanisms, especially the interhierarchical links among molecular activities, cellular behaviors and tissue/organ morphogenetic processes. Limb development is a well-studied topic in vertebrate organogenesis. Nevertheless, there is still little understanding of tissue-level deformation relative to molecular and cellular dynamics. This is mainly because live recording of detailed cell behaviors in whole tissues is technically difficult. To overcome this limitation, by applying a recently developed Bayesian approach, we here constructed tissue deformation maps for chick limb development with high precision, based on snapshot lineage tracing using dye injection. The precision of the constructed maps was validated with a clear statistical criterion. From the geometrical analysis of the map, we identified three characteristic tissue growth modes in the limb and showed that they are consistent with local growth factor activity and cell cycle length. In particular, we report that SHH signaling activity changes dynamically with developmental stage and strongly correlates with the dynamic shift in the tissue growth mode. We also found anisotropic tissue deformation along the proximal-distal axis. Morphogenetic simulation and experimental studies suggested that this directional tissue elongation, and not local growth, has the greatest impact on limb shaping. This result was supported by the novel finding that anisotropic tissue elongation along the proximal-distal axis occurs independently of cell proliferation. Our study marks a pivotal point for multi-scale system understanding in vertebrate development. Summary: Chick limb morphogenesis occurs through issue elongation independently of cell proliferation, with three tissue growth modes correlated with SHH expression.
Collapse
Affiliation(s)
- Yoshihiro Morishita
- Laboratory for Developmental Morphogeometry, RIKEN Quantitative Biology Center, Kobe 650-0047, Japan RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Atsushi Kuroiwa
- Division of Biological Science, Graduate School of Science, Nagoya University Furo-cho, Chikusa-ku, Nagoya 464-8602, Japan
| | - Takayuki Suzuki
- Division of Biological Science, Graduate School of Science, Nagoya University Furo-cho, Chikusa-ku, Nagoya 464-8602, Japan
| |
Collapse
|
189
|
Peyre E, Silva CG, Nguyen L. Crosstalk between intracellular and extracellular signals regulating interneuron production, migration and integration into the cortex. Front Cell Neurosci 2015; 9:129. [PMID: 25926769 PMCID: PMC4396449 DOI: 10.3389/fncel.2015.00129] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/19/2015] [Indexed: 11/29/2022] Open
Abstract
During embryogenesis, cortical interneurons are generated by ventral progenitors located in the ganglionic eminences of the telencephalon. They travel along multiple tangential paths to populate the cortical wall. As they reach this structure they undergo intracortical dispersion to settle in their final destination. At the cellular level, migrating interneurons are highly polarized cells that extend and retract processes using dynamic remodeling of microtubule and actin cytoskeleton. Different levels of molecular regulation contribute to interneuron migration. These include: (1) Extrinsic guidance cues distributed along migratory streams that are sensed and integrated by migrating interneurons; (2) Intrinsic genetic programs driven by specific transcription factors that grant specification and set the timing of migration for different subtypes of interneurons; (3) Adhesion molecules and cytoskeletal elements/regulators that transduce molecular signalings into coherent movement. These levels of molecular regulation must be properly integrated by interneurons to allow their migration in the cortex. The aim of this review is to summarize our current knowledge of the interplay between microenvironmental signals and cell autonomous programs that drive cortical interneuron porduction, tangential migration, and intergration in the developing cerebral cortex.
Collapse
Affiliation(s)
- Elise Peyre
- GIGA-Neurosciences, University of Liège Liège, Belgium ; Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège Liège, Belgium
| | - Carla G Silva
- GIGA-Neurosciences, University of Liège Liège, Belgium ; Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège Liège, Belgium
| | - Laurent Nguyen
- GIGA-Neurosciences, University of Liège Liège, Belgium ; Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège Liège, Belgium ; Wallon Excellence in Lifesciences and Biotechnology, University of Liège Liège, Belgium
| |
Collapse
|
190
|
Sabherwal N, Thuret R, Lea R, Stanley P, Papalopulu N. aPKC phosphorylates p27Xic1, providing a mechanistic link between apicobasal polarity and cell-cycle control. Dev Cell 2015; 31:559-71. [PMID: 25490266 PMCID: PMC4262734 DOI: 10.1016/j.devcel.2014.10.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 09/05/2014] [Accepted: 10/29/2014] [Indexed: 11/30/2022]
Abstract
During the development of the nervous system, apicobasally polarized stem cells are characterized by a shorter cell cycle than nonpolar progenitors, leading to a lower differentiation potential of these cells. However, how polarization might be directly linked to the kinetics of the cell cycle is not understood. Here, we report that apicobasally polarized neuroepithelial cells in Xenopus laevis have a shorter cell cycle than nonpolar progenitors, consistent with mammalian systems. We show that the apically localized serine/threonine kinase aPKC directly phosphorylates an N-terminal site of the cell-cycle inhibitor p27Xic1 and reduces its ability to inhibit the cyclin-dependent kinase 2 (Cdk2), leading to shortening of G1 and S phases. Overexpression of activated aPKC blocks the neuronal differentiation-promoting activity of p27Xic1. These findings provide a direct mechanistic link between apicobasal polarity and the cell cycle, which may explain how proliferation is favored over differentiation in polarized neural stem cells. aPKC shortens G1 and S phases of cell cycle by phosphorylating p27Xic1 Phosphorylated p27Xic1 exhibits weaker binding to and inhibition of Cdk2 p27Xic1 promotes neuronal differentiation and elongates cell cycle via G1 phase Effects of p27Xic1 on neuronal differentiation are rescued by activated aPKC
Collapse
Affiliation(s)
- Nitin Sabherwal
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK.
| | - Raphael Thuret
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Robert Lea
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Peter Stanley
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Nancy Papalopulu
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK.
| |
Collapse
|
191
|
Boyd JL, Skove SL, Rouanet JP, Pilaz LJ, Bepler T, Gordân R, Wray GA, Silver DL. Human-chimpanzee differences in a FZD8 enhancer alter cell-cycle dynamics in the developing neocortex. Curr Biol 2015; 25:772-779. [PMID: 25702574 DOI: 10.1016/j.cub.2015.01.041] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 08/28/2014] [Accepted: 01/16/2015] [Indexed: 12/14/2022]
Abstract
The human neocortex differs from that of other great apes in several notable regards, including altered cell cycle, prolonged corticogenesis, and increased size [1-5]. Although these evolutionary changes most likely contributed to the origin of distinctively human cognitive faculties, their genetic basis remains almost entirely unknown. Highly conserved non-coding regions showing rapid sequence changes along the human lineage are candidate loci for the development and evolution of uniquely human traits. Several studies have identified human-accelerated enhancers [6-14], but none have linked an expression difference to a specific organismal trait. Here we report the discovery of a human-accelerated regulatory enhancer (HARE5) of FZD8, a receptor of the Wnt pathway implicated in brain development and size [15, 16]. Using transgenic mice, we demonstrate dramatic differences in human and chimpanzee HARE5 activity, with human HARE5 driving early and robust expression at the onset of corticogenesis. Similar to HARE5 activity, FZD8 is expressed in neural progenitors of the developing neocortex [17-19]. Chromosome conformation capture assays reveal that HARE5 physically and specifically contacts the core Fzd8 promoter in the mouse embryonic neocortex. To assess the phenotypic consequences of HARE5 activity, we generated transgenic mice in which Fzd8 expression is under control of orthologous enhancers (Pt-HARE5::Fzd8 and Hs-HARE5::Fzd8). In comparison to Pt-HARE5::Fzd8, Hs-HARE5::Fzd8 mice showed marked acceleration of neural progenitor cell cycle and increased brain size. Changes in HARE5 function unique to humans thus alter the cell-cycle dynamics of a critical population of stem cells during corticogenesis and may underlie some distinctive anatomical features of the human brain.
Collapse
Affiliation(s)
- J Lomax Boyd
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Stephanie L Skove
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jeremy P Rouanet
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Louis-Jan Pilaz
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Tristan Bepler
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA
| | - Raluca Gordân
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA; Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC 27710, USA
| | - Gregory A Wray
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708, USA; Department of Biology, Duke University, Durham, NC 27708, USA; Department of Evolutionary Anthropology, Duke University, Durham, NC 27708, USA
| | - Debra L Silver
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Duke Institute for Brain Sciences, Durham, NC 27710 USA.
| |
Collapse
|
192
|
Jin J, Peng Q, Hou Z, Jiang M, Wang X, Langseth AJ, Tao M, Barker PB, Mori S, Bergles DE, Ross CA, Detloff PJ, Zhang J, Duan W. Early white matter abnormalities, progressive brain pathology and motor deficits in a novel knock-in mouse model of Huntington's disease. Hum Mol Genet 2015; 24:2508-27. [PMID: 25609071 DOI: 10.1093/hmg/ddv016] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 01/19/2015] [Indexed: 12/21/2022] Open
Abstract
White matter abnormalities have been reported in premanifest Huntington's disease (HD) subjects before overt striatal neuronal loss, but whether the white matter changes represent a necessary step towards further pathology and the underlying mechanism of these changes remains unknown. Here, we characterized a novel knock-in mouse model that expresses mouse HD gene homolog (Hdh) with extended CAG repeat- HdhQ250, which was derived from the selective breeding of HdhQ150 mice. HdhQ250 mice manifest an accelerated and robust phenotype compared with its parent line. HdhQ250 mice exhibit progressive motor deficits, reduction in striatal and cortical volume, accumulation of mutant huntingtin aggregation, decreased levels of DARPP32 and BDNF and altered striatal metabolites. The abnormalities detected in this mouse model are reminiscent of several aspects of human HD. In addition, disturbed myelination was evident in postnatal Day 14 HdhQ250 mouse brain, including reduced levels of myelin regulatory factor and myelin basic protein, and decreased numbers of myelinated axons in the corpus callosum. Thinner myelin sheaths, indicated by increased G-ratio of myelin, were also detected in the corpus callosum of adult HdhQ250 mice. Moreover, proliferation of oligodendrocyte precursor cells is altered by mutant huntingtin both in vitro and in vivo. Our data indicate that this model is suitable for understanding comprehensive pathogenesis of HD in white matter and gray matter as well as developing therapeutics for HD.
Collapse
Affiliation(s)
- Jing Jin
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences
| | - Qi Peng
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences
| | | | - Mali Jiang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences
| | - Xin Wang
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY 10065, USA
| | | | - Michael Tao
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences
| | | | | | | | - Christopher A Ross
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA, Department of Neuroscience, Department of Neurology and Pharmacology and Molecular Sciences and
| | - Peter J Detloff
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35242, USA
| | | | - Wenzhen Duan
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA, Department of Neuroscience,
| |
Collapse
|
193
|
Najas S, Arranz J, Lochhead PA, Ashford AL, Oxley D, Delabar JM, Cook SJ, Barallobre MJ, Arbonés ML. DYRK1A-mediated Cyclin D1 Degradation in Neural Stem Cells Contributes to the Neurogenic Cortical Defects in Down Syndrome. EBioMedicine 2015; 2:120-34. [PMID: 26137553 PMCID: PMC4484814 DOI: 10.1016/j.ebiom.2015.01.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 01/16/2015] [Accepted: 01/16/2015] [Indexed: 01/02/2023] Open
Abstract
Alterations in cerebral cortex connectivity lead to intellectual disability and in Down syndrome, this is associated with a deficit in cortical neurons that arises during prenatal development. However, the pathogenic mechanisms that cause this deficit have not yet been defined. Here we show that the human DYRK1A kinase on chromosome 21 tightly regulates the nuclear levels of Cyclin D1 in embryonic cortical stem (radial glia) cells, and that a modest increase in DYRK1A protein in transgenic embryos lengthens the G1 phase in these progenitors. These alterations promote asymmetric proliferative divisions at the expense of neurogenic divisions, producing a deficit in cortical projection neurons that persists in postnatal stages. Moreover, radial glial progenitors in the Ts65Dn mouse model of Down syndrome have less Cyclin D1, and Dyrk1a is the triplicated gene that causes both early cortical neurogenic defects and decreased nuclear Cyclin D1 levels in this model. These data provide insights into the mechanisms that couple cell cycle regulation and neuron production in cortical neural stem cells, emphasizing that the deleterious effect of DYRK1A triplication in the formation of the cerebral cortex begins at the onset of neurogenesis, which is relevant to the search for early therapeutic interventions in Down syndrome.
Collapse
Affiliation(s)
- Sònia Najas
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 08028 Barcelona, Spain
| | - Juan Arranz
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 08028 Barcelona, Spain
| | - Pamela A. Lochhead
- Signalling Programme, The Babraham Institute, Babraham Research Campus, CB22 3AT Cambridge, UK
| | - Anne L. Ashford
- Signalling Programme, The Babraham Institute, Babraham Research Campus, CB22 3AT Cambridge, UK
| | - David Oxley
- Proteomics Group, The Babraham Institute, Babraham Research Campus, CB22 3AT Cambridge, UK
| | - Jean M. Delabar
- Sorbonne Universités, UPMC Univ Paris 06, Inserm, CNRS, UM 75, U 1127, UMR 7225, ICM, 75013 Paris, France
| | - Simon J. Cook
- Signalling Programme, The Babraham Institute, Babraham Research Campus, CB22 3AT Cambridge, UK
| | - María José Barallobre
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 08028 Barcelona, Spain
| | - Maria L. Arbonés
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 08028 Barcelona, Spain
| |
Collapse
|
194
|
Watanabe N, Kageyama R, Ohtsuka T. Hbp1 regulates the timing of neuronal differentiation during cortical development by controlling cell cycle progression. Development 2015; 142:2278-90. [DOI: 10.1242/dev.120477] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 05/26/2015] [Indexed: 11/20/2022]
Abstract
In the developing mammalian brain, neural stem cells (NSCs) initially expand the progenitor pool by symmetric divisions. NSCs then shift from symmetric to asymmetric division and commence neurogenesis. Although the precise mechanisms regulating the developmental timing of this transition have not been fully elucidated, gradual elongation in the length of the cell cycle and coinciding accumulation of determinants that promote neuronal differentiation may function as a biological clock that regulates the onset of asymmetric division and neurogenesis. We conducted gene expression profiling of embryonic NSCs in the cortical regions and found that expression of high mobility group box transcription factor 1 (Hbp1) was upregulated during neurogenic stages. Induced conditional knockout mice of Hbp1 generated by crossing with Nestin-CreERT2 mice exhibited a remarkable dilatation of the telencephalic vesicles with a tangentially expanded ventricular zone and a thinner cortical plate containing reduced numbers of neurons. In these Hbp1-deficient mouse embryos, neural stem/progenitor cells continued to divide with a shorter cell cycle length. And downstream target genes of the Wnt signaling, such as cyclin D1 and c-jun, were upregulated in the germinal zone of the cortical regions. These results indicate that Hbp1 plays a critical role in regulating the timing of cortical neurogenesis by elongating the cell cycle and is essential for normal cortical development.
Collapse
Affiliation(s)
- Naoki Watanabe
- Institute for Virus Research, Kyoto University, Shogoin-Kawahara, Sakyo-ku, Kyoto 606-8507, Japan
- Japan Science and Technology Agency, Core Research for Evolutional Science and Technology (CREST), 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Ryoichiro Kageyama
- Institute for Virus Research, Kyoto University, Shogoin-Kawahara, Sakyo-ku, Kyoto 606-8507, Japan
- Japan Science and Technology Agency, Core Research for Evolutional Science and Technology (CREST), 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
- World Premier International Research Initiative-Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida, Sakyo-ku, Kyoto 606-8501, Japan
| | - Toshiyuki Ohtsuka
- Institute for Virus Research, Kyoto University, Shogoin-Kawahara, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
195
|
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: 24] [Impact Index Per Article: 2.4] [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
|
196
|
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: 104] [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
|
197
|
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: 3.7] [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
|
198
|
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: 1.3] [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
|
199
|
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: 6.4] [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
|
200
|
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: 3.0] [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
|