101
|
Falgairolle M, Puhl JG, Pujala A, Liu W, O'Donovan MJ. Motoneurons regulate the central pattern generator during drug-induced locomotor-like activity in the neonatal mouse. eLife 2017; 6. [PMID: 28671548 PMCID: PMC5550280 DOI: 10.7554/elife.26622] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 06/30/2017] [Indexed: 12/17/2022] Open
Abstract
Motoneurons are traditionally viewed as the output of the spinal cord that do not influence locomotor rhythmogenesis. We assessed the role of motoneuron firing during ongoing locomotor-like activity in neonatal mice expressing archaerhopsin-3 (Arch), halorhodopsin (eNpHR), or channelrhodopsin-2 (ChR2) in Choline acetyltransferase neurons (ChAT+) or Arch in LIM-homeodomain transcription factor Isl1+ neurons. Illumination of the lumbar cord in mice expressing eNpHR or Arch in ChAT+ or Isl1+ neurons, depressed motoneuron discharge, transiently decreased the frequency, and perturbed the phasing of the locomotor-like rhythm. When the light was turned off motoneuron firing and locomotor frequency both transiently increased. These effects were not due to cholinergic neurotransmission, persisted during partial blockade of gap junctions and were mediated, in part, by AMPAergic transmission. In spinal cords expressing ChR2, illumination increased motoneuron discharge and transiently accelerated the rhythm. We conclude that motoneurons provide feedback to the central pattern generator (CPG) during drug-induced locomotor-like activity. DOI:http://dx.doi.org/10.7554/eLife.26622.001
Collapse
Affiliation(s)
- Melanie Falgairolle
- Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, United States
| | - Joshua G Puhl
- Department of Entomology, University of Minnesota, Saint Paul, United States
| | - Avinash Pujala
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Wenfang Liu
- Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, United States
| | - Michael J O'Donovan
- Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, United States
| |
Collapse
|
102
|
Intron-specific patterns of divergence of lin-11 regulatory function in the C. elegans nervous system. Dev Biol 2017; 424:90-103. [DOI: 10.1016/j.ydbio.2017.02.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 02/08/2017] [Accepted: 02/12/2017] [Indexed: 12/19/2022]
|
103
|
ISLET1-Dependent β-Catenin/Hedgehog Signaling Is Required for Outgrowth of the Lower Jaw. Mol Cell Biol 2017; 37:MCB.00590-16. [PMID: 28069742 DOI: 10.1128/mcb.00590-16] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 01/04/2017] [Indexed: 12/30/2022] Open
Abstract
Mandibular patterning information initially resides in the epithelium during development. However, how transcriptional regulation of epithelium-derived signaling controls morphogenesis of the mandible remains elusive. Using ShhCre to target the mandibular epithelium, we ablated transcription factor Islet1, resulting in a distally truncated mandible via unbalanced cell apoptosis and decreased cell proliferation in the distal mesenchyme. Loss of Islet1 caused a lack of cartilage at the distal tip, leading the fusion of two growing mandibular elements surrounding the rostral process of Meckel's cartilage. Loss of Islet1 results in dysregulation of mesenchymal genes important for morphogenesis of the mandibular arch. We revealed that Islet1 is required for the activation of epithelial β-catenin signaling via repression of Wnt antagonists. Reactivation of β-catenin in the epithelium of the Islet1 mutant rescued mandibular morphogenesis through sonic hedgehog (SHH) signaling to the mesenchyme. Furthermore, overexpression of a transgenic hedgehog ligand in the epithelium also partially restored outgrowth of the mandible. These data reveal functional roles for an ISLET1-dependent network integrating β-catenin/SHH signals in mesenchymal cell survival and outgrowth of the mandible during development.
Collapse
|
104
|
The Isl1-Lhx3 Complex Promotes Motor Neuron Specification by Activating Transcriptional Pathways that Enhance Its Own Expression and Formation. eNeuro 2017; 4:eN-NWR-0349-16. [PMID: 28451636 PMCID: PMC5394944 DOI: 10.1523/eneuro.0349-16.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/27/2017] [Accepted: 03/11/2017] [Indexed: 01/27/2023] Open
Abstract
Motor neuron (MN) progenitor cells rapidly induce high expression of the transcription factors Islet-1 (Isl1), LIM-homeobox 3 (Lhx3), and the transcriptional regulator LMO4, as they differentiate. While these factors are critical for MN specification, the mechanisms regulating their precise temporal and spatial expression patterns are not well characterized. Isl1 and Lhx3 form the Isl1-Lhx3 complex, which induces the transcription of genes critical for MN specification and maturation. Here, we report that Isl1, Lhx3, and Lmo4 are direct target genes of the Isl1-Lhx3 complex. Our results show that specific genomic loci associated with these genes recruit the Isl1-Lhx3 complex to activate the transcription of Isl1, Lhx3, and Lmo4 in embryonic MNs of chick and mouse. These findings support a model in which the Isl1-Lhx3 complex amplifies its own expression through a potent autoregulatory feedback loop and simultaneously enhances the transcription of Lmo4. LMO4 blocks the formation of the V2 interneuron-specifying Lhx3 complex. In developing MNs, this action inhibits the expression of V2 interneuron genes and increases the pool of unbound Lhx3 available to incorporate into the Isl1-Lhx3 complex. Identifying the pathways that regulate the expression of these key factors provides important insights into the genetic strategies utilized to promote MN differentiation and maturation.
Collapse
|
105
|
Yousefi B, Sanooghi D, Faghihi F, Joghataei MT, Latifi N. Evaluation of motor neuron differentiation potential of human umbilical cord blood- derived mesenchymal stem cells, in vitro. J Chem Neuroanat 2017; 81:18-26. [PMID: 28153469 DOI: 10.1016/j.jchemneu.2017.01.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 12/25/2016] [Accepted: 01/22/2017] [Indexed: 10/20/2022]
Abstract
Many people suffer from spinal cord injuries annually. These deficits usually threaten the quality of life of patients. As a postpartum medically waste product, human Umbilical Cord Blood (UCB) is a rich source of stem cells with self- renewal properties and neural differentiation capacity which made it useful in regenerative medicine. Since there is no report on potential of human umbilical cord blood-derived mesenchymal stem cells into motor neurons, we set out to evaluate the differentiation properties of these cells into motor neuron-like cells through administration of Retinoic Acid(RA), Sonic Hedgehog(Shh) and BDNF using a three- step in vitro procedure. The results were evaluated using Real-time PCR, Flowcytometry and Immunocytochemistry for two weeks. Our data showed that the cells changed into bipolar morphology and could express markers related to motor neuron; including Hb-9, Pax-6, Islet-1, NF-H, ChAT at the level of mRNA and protein. We could also quantitatively evaluate the expression of Islet-1, ChAT and NF-H at 7 and 14days post- induction using flowcytometry. It is concluded that human UCB-MSCs is potent to express motor neuron- related markers in the presence of RA, Shh and BDNF through a three- step protocol; thus it could be a suitable cell candidate for regeneration of motor neurons in spinal cord injuries.
Collapse
Affiliation(s)
- Behnam Yousefi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Davood Sanooghi
- Department of Genetics, Faculty of Biological Sciences, Shahid Beheshti University, Tehran, Iran
| | - Faezeh Faghihi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Taghi Joghataei
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Nourahmad Latifi
- Department of Surgery, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
106
|
Allaway KC, Machold R. Developmental specification of forebrain cholinergic neurons. Dev Biol 2016; 421:1-7. [PMID: 27847324 DOI: 10.1016/j.ydbio.2016.11.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 11/10/2016] [Accepted: 11/11/2016] [Indexed: 01/17/2023]
Abstract
Striatal cholinergic interneurons and basal forebrain cholinergic projection neurons, which together comprise the forebrain cholinergic system, regulate attention, memory, reward pathways, and motor activity through the neuromodulation of multiple brain circuits. The importance of these neurons in the etiology of neurocognitive disorders has been well documented, but our understanding of their specification during embryogenesis is still incomplete. All forebrain cholinergic projection neurons and interneurons appear to share a common developmental origin in the embryonic ventral telencephalon, a region that also gives rise to GABAergic projection neurons and interneurons. Significant progress has been made in identifying the key intrinsic and extrinsic factors that promote a cholinergic fate in this precursor population. However, how cholinergic interneurons and projection neurons differentiate from one another during development, as well as how distinct developmental programs contribute to heterogeneity within those two classes, is not yet well understood. In this review we summarize the transcription factors and signaling molecules known to play a role in the specification and early development of striatal and basal forebrain cholinergic neurons. We also discuss the heterogeneity of these populations and its possible developmental origins.
Collapse
Affiliation(s)
- Kathryn C Allaway
- NYU Neuroscience Institute and the Department of Neuroscience and Physiology, Smilow Research Center, New York University School of Medicine, 522 First Avenue, New York, NY 10016, USA
| | - Robert Machold
- NYU Neuroscience Institute and the Department of Neuroscience and Physiology, Smilow Research Center, New York University School of Medicine, 522 First Avenue, New York, NY 10016, USA.
| |
Collapse
|
107
|
ISL1-based LIM complexes control Slit2 transcription in developing cranial motor neurons. Sci Rep 2016; 6:36491. [PMID: 27819291 PMCID: PMC5098159 DOI: 10.1038/srep36491] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 10/17/2016] [Indexed: 01/02/2023] Open
Abstract
LIM-homeodomain (HD) transcription factors form a multimeric complex and assign neuronal subtype identities, as demonstrated by the hexameric ISL1-LHX3 complex which gives rise to somatic motor (SM) neurons. However, the roles of combinatorial LIM code in motor neuron diversification and their subsequent differentiation is much less well understood. In the present study, we demonstrate that the ISL1 controls postmitotic cranial branchiomotor (BM) neurons including the positioning of the cell bodies and peripheral axon pathfinding. Unlike SM neurons, which transform into interneurons, BM neurons are normal in number and in marker expression in Isl1 mutant mice. Nevertheless, the movement of trigeminal and facial BM somata is stalled, and their peripheral axons are fewer or misrouted, with ectopic branches. Among genes whose expression level changes in previous ChIP-seq and microarray analyses in Isl1-deficient cell lines, we found that Slit2 transcript was almost absent from BM neurons of Isl1 mutants. Both ISL1-LHX3 and ISL1-LHX4 bound to the Slit2 enhancer and drove endogenous Slit2 expression in SM and BM neurons. Our findings suggest that combinations of ISL1 and LHX factors establish cell-type specificity and functional diversity in terms of motor neuron identities and/or axon development.
Collapse
|
108
|
Suzuki K, Matsumaru D, Matsushita S, Murashima A, Ludwig M, Reutter H, Yamada G. Epispadias and the associated embryopathies: genetic and developmental basis. Clin Genet 2016; 91:247-253. [PMID: 27649475 DOI: 10.1111/cge.12871] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/13/2016] [Accepted: 09/16/2016] [Indexed: 12/25/2022]
Abstract
The abnormalities in the urogenital organs are frequently observed as human developmental diseases. Among such diseases, the defects in the upper part of external genitalia are rather rare named epispadias. The cleft in the dorsal part of external genitalia often reaches to the urethra. In general, the urogenital abnormalities accompany defects in the adjacent tissues and organs. The ventral body wall and bladder can also be affected in the patients with dorsal defects of the external genitalia. Therefore, such multiple malformations are often classified as bladder exstrophy and epispadias complex (BEEC). Because of the lower frequency of such birth defects and their early embryonic development, animal models are required to analyze the pathogenic mechanisms and the functions of responsible genes. Mutant mouse analyses on various signal cascades for external genitalia and body wall development are increasingly performed. The genetic interactions between growth factors such as bone morphogenetic proteins (Bmp) and transcription factors such as Msx1/2 and Isl1 have been suggested to play roles for such organogenesis. The significance of epithelial-mesenchymal interaction (EMI) is suggested during development. In this review, we describe on such local interactions and developmental regulators. We also introduce some mutant mouse models displaying external genitalia-body wall abnormalities.
Collapse
Affiliation(s)
- K Suzuki
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University (WMU), Wakayama, Japan
| | - D Matsumaru
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University (WMU), Wakayama, Japan
| | - S Matsushita
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University (WMU), Wakayama, Japan
| | - A Murashima
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University (WMU), Wakayama, Japan.,Division of Human Embryology, Department of Anatomy, Iwate Medical University, Yahaba, Japan
| | - M Ludwig
- Department of Clinical Chemistry and Clinical Pharmacology, University Hospital of Bonn, Bonn, Germany
| | - H Reutter
- Institute of Human Genetics, University Hospital of Bonn, Bonn, Germany.,Department of Neonatology and Pediatric Intensive Care, University Hospital of Bonn, Bonn, Germany
| | - G Yamada
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University (WMU), Wakayama, Japan
| |
Collapse
|
109
|
Yang Z, Zhang Q, Lu Q, Jia Z, Chen P, Ma K, Wang W, Zhou C. ISL-1 promotes pancreatic islet cell proliferation by forming an ISL-1/Set7/9/PDX-1 complex. Cell Cycle 2016; 14:3820-9. [PMID: 26176407 PMCID: PMC4825718 DOI: 10.1080/15384101.2015.1069926] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Islet-1 (ISL-1), a LIM-homeodomain transcription factor, has been recently found to be essential for promoting postnatal pancreatic islet proliferation. However, the detailed mechanism has not yet been elucidated. In the present study, we investigated the mechanism by which ISL-1 promotes β-cell proliferation through regulation of CyclinD1 in HIT-T15 and NIT-1 cells, as well in rat islet mass. Our results provide the evidence that ISL-1 promotes adult pancreatic islet β-cell proliferation by activating CyclinD1 transcription through cooperation with Set7/9 and PDX-1 to form an ISL-1/Set7/9/PDX-1 complex. This complex functions in an ISL-1-dependent manner, with Set7/9 functioning not only as a histone methyltransferase, which increases the histone H3K4 tri-methylation of the CyclinD1 promoter region, but also an adaptor to bridge ISL-1 and PDX-1, while PDX-1 functions as a RNA pol II binding modulator. Furthermore, the formation of the ISL-1/Set7/9/PDX-1 complex is positively associated with insulin-like growth factor-1 treatment in NIT and HIT-T15 cells in vitro, while may be negatively correlated with age in vivo.
Collapse
Affiliation(s)
- Zhe Yang
- a Department of Biochemistry and Molecular Biology ; School of Basic Medical Sciences; Key Laboratory of Molecular Cardiovascular Sciences; Ministry of Education of China; Peking University ; Beijing , China
| | - Qiao Zhang
- a Department of Biochemistry and Molecular Biology ; School of Basic Medical Sciences; Key Laboratory of Molecular Cardiovascular Sciences; Ministry of Education of China; Peking University ; Beijing , China.,b Current address: Department of Biochemistry and Molecular Biology ; School of Basic Medical Sciences; Kunming Medical University ; Kunming , China
| | - Qin Lu
- a Department of Biochemistry and Molecular Biology ; School of Basic Medical Sciences; Key Laboratory of Molecular Cardiovascular Sciences; Ministry of Education of China; Peking University ; Beijing , China
| | - Zhuqing Jia
- a Department of Biochemistry and Molecular Biology ; School of Basic Medical Sciences; Key Laboratory of Molecular Cardiovascular Sciences; Ministry of Education of China; Peking University ; Beijing , China
| | - Ping Chen
- a Department of Biochemistry and Molecular Biology ; School of Basic Medical Sciences; Key Laboratory of Molecular Cardiovascular Sciences; Ministry of Education of China; Peking University ; Beijing , China
| | - Kangtao Ma
- a Department of Biochemistry and Molecular Biology ; School of Basic Medical Sciences; Key Laboratory of Molecular Cardiovascular Sciences; Ministry of Education of China; Peking University ; Beijing , China
| | - Weiping Wang
- a Department of Biochemistry and Molecular Biology ; School of Basic Medical Sciences; Key Laboratory of Molecular Cardiovascular Sciences; Ministry of Education of China; Peking University ; Beijing , China
| | - Chunyan Zhou
- a Department of Biochemistry and Molecular Biology ; School of Basic Medical Sciences; Key Laboratory of Molecular Cardiovascular Sciences; Ministry of Education of China; Peking University ; Beijing , China
| |
Collapse
|
110
|
Stallings CE, Kapali J, Ellsworth BS. Mouse Models of Gonadotrope Development. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 143:1-48. [PMID: 27697200 DOI: 10.1016/bs.pmbts.2016.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The pituitary gonadotrope is central to reproductive function. Gonadotropes develop in a systematic process dependent on signaling factors secreted from surrounding tissues and those produced within the pituitary gland itself. These signaling pathways are important for stimulating specific transcription factors that ultimately regulate the expression of genes and define gonadotrope identity. Proper gonadotrope development and ultimately gonadotrope function are essential for normal sexual maturation and fertility. Understanding the mechanisms governing differentiation programs of gonadotropes is important to improve treatment and molecular diagnoses for patients with gonadotrope abnormalities. Much of what is known about gonadotrope development has been elucidated from mouse models in which important factors contributing to gonadotrope development and function have been deleted, ectopically expressed, or modified. This chapter will focus on many of these mouse models and their contribution to our current understanding of gonadotrope development.
Collapse
Affiliation(s)
- C E Stallings
- Department of Physiology, School of Medicine, Southern Illinois University, Carbondale, IL, United States
| | - J Kapali
- Department of Physiology, School of Medicine, Southern Illinois University, Carbondale, IL, United States
| | - B S Ellsworth
- Department of Physiology, School of Medicine, Southern Illinois University, Carbondale, IL, United States.
| |
Collapse
|
111
|
Gergics P, Christian HC, Choo MS, Ajmal A, Camper SA. Gene Expression in Mouse Thyrotrope Adenoma: Transcription Elongation Factor Stimulates Proliferation. Endocrinology 2016; 157:3631-46. [PMID: 27580811 PMCID: PMC5007889 DOI: 10.1210/en.2016-1183] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Thyrotrope hyperplasia and hypertrophy are common responses to primary hypothyroidism. To understand the genetic regulation of these processes, we studied gene expression changes in the pituitaries of Cga(-/-) mice, which are deficient in the common α-subunit of TSH, LH, and FSH. These mice have thyrotrope hypertrophy and hyperplasia and develop thyrotrope adenoma. We report that cell proliferation is increased, but the expression of most stem cell markers is unchanged. The α-subunit is required for secretion of the glycoprotein hormone β-subunits, and mutants exhibit elevated expression of many genes involved in the unfolded protein response, consistent with dilation and stress of the endoplasmic reticulum. Mutants have elevated expression of transcription factors that are important in thyrotrope function, such as Gata2 and Islet 1, and those that stimulate proliferation, including Nupr1, E2f1, and Etv5. We characterized the expression and function of a novel, overexpressed gene, transcription elongation factor A (SII)-like 5 (Tceal5). Stable expression of Tceal5 in a pituitary progenitor cell line is sufficient to increase cell proliferation. Thus, Tceal5 may act as a proto-oncogene. This study provides a rich resource for comparing pituitary transcriptomes and an analysis of gene expression networks.
Collapse
Affiliation(s)
- Peter Gergics
- Department of Human Genetics (P.G., M.S.C., S.A.C.), University of Michigan, Ann Arbor, Michigan 48109; Department of Physiology, Anatomy and Genetics (H.C.C.), University of Oxford, Oxford OX3 0RZ, United Kingdom; and Department of Internal Medicine, Metabolism, Endocrinology and Diabetes (A.A.), University of Michigan, Ann Arbor, Michigan 48105
| | - Helen C Christian
- Department of Human Genetics (P.G., M.S.C., S.A.C.), University of Michigan, Ann Arbor, Michigan 48109; Department of Physiology, Anatomy and Genetics (H.C.C.), University of Oxford, Oxford OX3 0RZ, United Kingdom; and Department of Internal Medicine, Metabolism, Endocrinology and Diabetes (A.A.), University of Michigan, Ann Arbor, Michigan 48105
| | - Monica S Choo
- Department of Human Genetics (P.G., M.S.C., S.A.C.), University of Michigan, Ann Arbor, Michigan 48109; Department of Physiology, Anatomy and Genetics (H.C.C.), University of Oxford, Oxford OX3 0RZ, United Kingdom; and Department of Internal Medicine, Metabolism, Endocrinology and Diabetes (A.A.), University of Michigan, Ann Arbor, Michigan 48105
| | - Adnan Ajmal
- Department of Human Genetics (P.G., M.S.C., S.A.C.), University of Michigan, Ann Arbor, Michigan 48109; Department of Physiology, Anatomy and Genetics (H.C.C.), University of Oxford, Oxford OX3 0RZ, United Kingdom; and Department of Internal Medicine, Metabolism, Endocrinology and Diabetes (A.A.), University of Michigan, Ann Arbor, Michigan 48105
| | - Sally A Camper
- Department of Human Genetics (P.G., M.S.C., S.A.C.), University of Michigan, Ann Arbor, Michigan 48109; Department of Physiology, Anatomy and Genetics (H.C.C.), University of Oxford, Oxford OX3 0RZ, United Kingdom; and Department of Internal Medicine, Metabolism, Endocrinology and Diabetes (A.A.), University of Michigan, Ann Arbor, Michigan 48105
| |
Collapse
|
112
|
Anderson C, Khan MAF, Wong F, Solovieva T, Oliveira NMM, Baldock RA, Tickle C, Burt DW, Stern CD. A strategy to discover new organizers identifies a putative heart organizer. Nat Commun 2016; 7:12656. [PMID: 27557800 DOI: 10.1038/ncomms12656] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/19/2016] [Indexed: 11/09/2022] Open
Abstract
Organizers are regions of the embryo that can both induce new fates and impart pattern on other regions. So far, surprisingly few organizers have been discovered, considering the number of patterned tissue types generated during development. This may be because their discovery has relied on transplantation and ablation experiments. Here we describe a new approach, using chick embryos, to discover organizers based on a common gene expression signature, and use it to uncover the anterior intestinal portal (AIP) endoderm as a putative heart organizer. We show that the AIP can induce cardiac identity from non-cardiac mesoderm and that it can pattern this by specifying ventricular and suppressing atrial regional identity. We also uncover some of the signals responsible. The method holds promise as a tool to discover other novel organizers acting during development.
Collapse
Affiliation(s)
- Claire Anderson
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Mohsin A F Khan
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Frances Wong
- Department of Genomics and Genetics, The Roslin Institute, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG Scotland, UK
| | - Tatiana Solovieva
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Nidia M M Oliveira
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Richard A Baldock
- Biomedical Systems Analysis Section, MRC Human Genetics Unit, IGMM, University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Cheryll Tickle
- Department of Biology &Biochemistry, University of Bath, Bath BA2 7AY, UK
| | - Dave W Burt
- Department of Genomics and Genetics, The Roslin Institute, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG Scotland, UK
| | - Claudio D Stern
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
113
|
Wu Y, Tao J, Wang YL, Hu WJ, Han X, Cheng WW. A genotype-phenotype correlation study reveals that a non-coding RNA might be associated with cardiovascular anomalies in fetuses with WHS. Prenat Diagn 2016; 36:979-981. [PMID: 27501084 DOI: 10.1002/pd.4884] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 07/18/2016] [Accepted: 07/30/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Yi Wu
- Prenatal Diagnosis Center, International Peace Maternal & Child Health Hospital, Medical College, Shanghai JiaoTong University, Shanghai, China
| | - Jiong Tao
- Department of Reproductive Genetics, International Peace Maternal & Child Health Hospital, Medical College, Shanghai JiaoTong University, Shanghai, China
| | - Yan-Lin Wang
- Prenatal Diagnosis Center, International Peace Maternal & Child Health Hospital, Medical College, Shanghai JiaoTong University, Shanghai, China
| | - Wen-Jing Hu
- Department of Reproductive Genetics, International Peace Maternal & Child Health Hospital, Medical College, Shanghai JiaoTong University, Shanghai, China
| | - Xu Han
- Department of Reproductive Genetics, International Peace Maternal & Child Health Hospital, Medical College, Shanghai JiaoTong University, Shanghai, China
| | - Wei-Wei Cheng
- Prenatal Diagnosis Center, International Peace Maternal & Child Health Hospital, Medical College, Shanghai JiaoTong University, Shanghai, China.
| |
Collapse
|
114
|
Clovis YM, Seo SY, Kwon JS, Rhee JC, Yeo S, Lee JW, Lee S, Lee SK. Chx10 Consolidates V2a Interneuron Identity through Two Distinct Gene Repression Modes. Cell Rep 2016; 16:1642-1652. [PMID: 27477290 DOI: 10.1016/j.celrep.2016.06.100] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 04/29/2016] [Accepted: 06/29/2016] [Indexed: 12/22/2022] Open
Abstract
During development, two cell types born from closely related progenitor pools often express identical transcriptional regulators despite their completely distinct characteristics. This phenomenon implies the need for a mechanism that operates to segregate the identities of the two cell types throughout differentiation after initial fate commitment. To understand this mechanism, we investigated the fate specification of spinal V2a interneurons, which share important developmental genes with motor neurons (MNs). We demonstrate that the paired homeodomain factor Chx10 functions as a critical determinant for V2a fate and is required to consolidate V2a identity in postmitotic neurons. Chx10 actively promotes V2a fate, downstream of the LIM-homeodomain factor Lhx3, while concomitantly suppressing the MN developmental program by preventing the MN-specific transcription complex from binding and activating MN genes. This dual activity enables Chx10 to effectively separate the V2a and MN pathways. Our study uncovers a widely applicable gene regulatory principle for segregating related cell fates.
Collapse
Affiliation(s)
- Yoanne M Clovis
- Pediatric Neuroscience Research Program, Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| | - So Yeon Seo
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Korea
| | - Ji-Sun Kwon
- Pediatric Neuroscience Research Program, Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Jennifer C Rhee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Korea
| | - Sujeong Yeo
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Korea
| | - Jae W Lee
- Pediatric Neuroscience Research Program, Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Seunghee Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Korea.
| | - Soo-Kyung Lee
- Pediatric Neuroscience Research Program, Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA; Vollum Institute, Oregon Health and Science University, Portland, OR 97239, USA.
| |
Collapse
|
115
|
Luo H, Cui S, Chen D, Liu J, Liu Z. Immunohistochemical Detection of Islet-1 and Neuronal Nitric Oxide Synthase in the Dorsal Root Ganglia (DRG) of Sheep Fetuses During Gestation. J Histochem Cytochem 2016; 52:797-803. [PMID: 15150288 DOI: 10.1369/jhc.4a6273.2004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This study first investigated the ontogeny of Islet-1 and neuronal nitric oxide synthase (nNOS) expression and their co-localization in the DRG of sheep fetuses during gestation by immunohistochemistry (IHC). The results showed that Islet-1 and nNOS were located in the nuclei and cytoplasm of DRG neurons, respectively. The relative percentages of Islet-1-immunopositive (Islet-1+) neurons accounting for the total DRG neurons were 90%, 79%, 66%, and 53% at days 60, 90, and 120 of gestation and postnatally, respectively. The percentage of nNOS-immunopositive (nNOS+) neurons was 94% at day 60 and declined to ∼30% at day 90, with no obvious further change until the postnatal period. Dual IHC showed that ∼69% Islet-1+ neurons express nNOS at day 60 of gestation. This proportion declined to ∼24% at day 90, after which there was no significant change until birth. We also observed that most Islet-1+ and nNOS+ neurons belonged to small and medium-sized DRG neurons from day 90 of gestation to the postnatal period. These results suggest that both Islet-1 and nNOS are important for the differentiation and maintenance of some specific phenotypes of DRG neurons during late gestation of sheep fetuses, although the related mechanisms need to be further elucidated. (J Histochem Cytochem 52:797–803, 2004)
Collapse
Affiliation(s)
- Haoshu Luo
- Department of Animal Physiology, College of Biological Sciences and Faculty of Veterinary Medicine, China Agricultural University, Beijing, People's Republic of China
| | | | | | | | | |
Collapse
|
116
|
Lien HW, Yuan RY, Chou CM, Chen YC, Hung CC, Hu CH, Hwang SPL, Hwang PP, Shen CN, Chen CL, Cheng CH, Huang CJ. Zebrafish cyclin Dx is required for development of motor neuron progenitors, and its expression is regulated by hypoxia-inducible factor 2α. Sci Rep 2016; 6:28297. [PMID: 27323909 PMCID: PMC4915019 DOI: 10.1038/srep28297] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 06/01/2016] [Indexed: 01/31/2023] Open
Abstract
Cyclins play a central role in cell-cycle regulation; in mammals, the D family of cyclins consists of cyclin D1, D2, and D3. In Xenopus, only homologs of cyclins D1 and D2 have been reported, while a novel cyclin, cyclin Dx (ccndx), was found to be required for the maintenance of motor neuron progenitors during embryogenesis. It remains unknown whether zebrafish possess cyclin D3 or cyclin Dx. In this study, we identified a zebrafish ccndx gene encoding a protein which can form a complex with Cdk4. Through whole-mount in situ hybridization, we observed that zccndx mRNA is expressed in the motor neurons of hindbrain and spinal cord during development. Analysis of a 4-kb promoter sequence of the zccndx gene revealed the presence of HRE sites, which can be regulated by HIF2α. Morpholino knockdown of zebrafish Hif2α and cyclin Dx resulted in the abolishment of isl1 and oligo2 expression in the precursors of motor neurons, and also disrupted axon growth. Overexpression of cyclin Dx mRNA in Hif2α morphants partially rescued zccndx expression. Taken together, our data indicate that zebrafish cyclin Dx plays a role in maintaining the precursors of motor neurons.
Collapse
Affiliation(s)
- Huang-Wei Lien
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 115, Taiwan
| | - Rey-Yue Yuan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 115, Taiwan
| | - Chih-Ming Chou
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 115, Taiwan
| | - Yi-Chung Chen
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 115, Taiwan
| | - Chin-Chun Hung
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Chin-Hwa Hu
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung 20224, Taiwan
| | - Sheng-Ping L Hwang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 115, Taiwan
| | - Pung-Pung Hwang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 115, Taiwan
| | - Chia-Ning Shen
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Chih-Lung Chen
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Chia-Hsiung Cheng
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 115, Taiwan
| | - Chang-Jen Huang
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
117
|
Galloway DA, Moore CS. miRNAs As Emerging Regulators of Oligodendrocyte Development and Differentiation. Front Cell Dev Biol 2016; 4:59. [PMID: 27379236 PMCID: PMC4911355 DOI: 10.3389/fcell.2016.00059] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 06/03/2016] [Indexed: 12/21/2022] Open
Abstract
Chronic demyelination is a hallmark of neurological disorders such as multiple sclerosis (MS) and several leukodystrophies. In the central nervous system (CNS), remyelination is a regenerative process that is often inadequate during these pathological states. In the MS context, in situ evidence suggests that remyelination is mediated by populations of oligodendrocyte progenitor cells (OPCs) that proliferate, migrate, and differentiate into mature, myelin-producing oligodendrocytes at sites of demyelinated lesions. The molecular programming of OPCs into mature oligodendrocytes is governed by a myriad of complex intracellular signaling pathways that modulate this process. Recent research has demonstrated the importance of specific and short non-coding RNAs, known as microRNAs (miRNAs), in regulating OPC differentiation and remyelination. Fortunately, it may be possible to take advantage of numerous developmental studies (both human and rodent) that have previously characterized miRNA expression profiles from the early neural progenitor cell to the late myelin-producing oligodendrocyte. Here we review much of the work to date and discuss the impact of miRNAs on OPC and oligodendrocyte biology. Additionally, we consider the potential for miRNA-mediated therapy in the context of remyelination and brain repair.
Collapse
Affiliation(s)
- Dylan A Galloway
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland St. John's, NL, Canada
| | - Craig S Moore
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland St. John's, NL, Canada
| |
Collapse
|
118
|
The LIM-homeodomain transcription factor Islet2a promotes angioblast migration. Dev Biol 2016; 414:181-92. [PMID: 27126199 DOI: 10.1016/j.ydbio.2016.04.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 03/25/2016] [Accepted: 04/24/2016] [Indexed: 02/04/2023]
Abstract
Angioblasts of the developing vascular system require many signaling inputs to initiate their migration, proliferation and differentiation into endothelial cells. What is less studied is which intrinsic cell factors interpret these extrinsic signals. Here, we show the Lim homeodomain transcription factor islet2a (isl2a) is expressed in the lateral posterior mesoderm prior to angioblast migration. isl2a deficient angioblasts show disorganized migration to the midline to form axial vessels and fail to spread around the tailbud of the embryo. Isl2a morphants have fewer vein cells and decreased vein marker expression. We demonstrate that isl2a is required cell autonomously in angioblasts to promote their incorporation into the vein, and is permissive for vein identity. Knockout of isl2a results in decreased migration and proliferation of angioblasts during intersegmental artery growth. Since Notch signaling controls both artery-vein identity and tip-stalk cell formation, we explored the interaction of isl2a and Notch. We find that isl2a expression is negatively regulated by Notch activity, and that isl2a positively regulates flt4, a VEGF-C receptor repressed by Notch during angiogenesis. Thus Isl2a may act as an intermediate between Notch signaling and genetic programs controlling angioblast number and migration, placing it as a novel transcriptional regulator of early angiogenesis.
Collapse
|
119
|
Bohuslavova R, Dodd N, Macova I, Chumak T, Horak M, Syka J, Fritzsch B, Pavlinkova G. Pax2-Islet1 Transgenic Mice Are Hyperactive and Have Altered Cerebellar Foliation. Mol Neurobiol 2016; 54:1352-1368. [PMID: 26843111 PMCID: PMC5310572 DOI: 10.1007/s12035-016-9716-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 01/12/2016] [Indexed: 12/18/2022]
Abstract
The programming of cell fate by transcription factors requires precise regulation of their time and level of expression. The LIM-homeodomain transcription factor Islet1 (Isl1) is involved in cell-fate specification of motor neurons, and it may play a similar role in the inner ear. In order to study its role in the regulation of vestibulo-motor development, we investigated a transgenic mouse expressing Isl1 under the Pax2 promoter control (Tg+/−). The transgenic mice show altered level, time, and place of expression of Isl1 but are viable. However, Tg+/− mice exhibit hyperactivity, including circling behavior, and progressive age-related decline in hearing, which has been reported previously. Here, we describe the molecular and morphological changes in the cerebellum and vestibular system that may cause the hyperactivity of Tg+/− mice. The transgene altered the formation of folia in the cerebellum, the distribution of calretinin labeled unipolar brush cells, and reduced the size of the cerebellum, inferior colliculus, and saccule. Age-related progressive reduction of calbindin expression was detected in Purkinje cells in the transgenic cerebella. The hyperactivity of Tg+/− mice is reduced upon the administration of picrotoxin, a non-competitive channel blocker for the γ-aminobutyric acid (GABA) receptor chloride channels. This suggests that the overexpression of Isl1 significantly affects the functions of GABAergic neurons. We demonstrate that the overexpression of Isl1 affects the development and function of the cerebello-vestibular system, resulting in hyperactivity.
Collapse
Affiliation(s)
- Romana Bohuslavova
- Institute of Biotechnology CAS, Prumyslova 595, Vestec, Prague-West District, 25242, Czech Republic
| | - Nicole Dodd
- Institute of Biotechnology CAS, Prumyslova 595, Vestec, Prague-West District, 25242, Czech Republic
| | - Iva Macova
- Institute of Biotechnology CAS, Prumyslova 595, Vestec, Prague-West District, 25242, Czech Republic
| | - Tetyana Chumak
- Institute of Experimental Medicine CAS, Prague, Czech Republic
| | - Martin Horak
- Institute of Physiology CAS, Prague, Czech Republic
| | - Josef Syka
- Institute of Experimental Medicine CAS, Prague, Czech Republic
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, IA, USA
| | - Gabriela Pavlinkova
- Institute of Biotechnology CAS, Prumyslova 595, Vestec, Prague-West District, 25242, Czech Republic.
| |
Collapse
|
120
|
Dorn T, Goedel A, Lam JT, Haas J, Tian Q, Herrmann F, Bundschu K, Dobreva G, Schiemann M, Dirschinger R, Guo Y, Kühl SJ, Sinnecker D, Lipp P, Laugwitz KL, Kühl M, Moretti A. Direct nkx2-5 transcriptional repression of isl1 controls cardiomyocyte subtype identity. Stem Cells 2016; 33:1113-29. [PMID: 25524439 PMCID: PMC6750130 DOI: 10.1002/stem.1923] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 10/29/2014] [Accepted: 11/08/2014] [Indexed: 12/31/2022]
Abstract
During cardiogenesis, most myocytes arise from cardiac progenitors expressing the transcription factors Isl1 and Nkx2-5. Here, we show that a direct repression of Isl1 by Nkx2-5 is necessary for proper development of the ventricular myocardial lineage. Overexpression of Nkx2-5 in mouse embryonic stem cells (ESCs) delayed specification of cardiac progenitors and inhibited expression of Isl1 and its downstream targets in Isl1(+) precursors. Embryos deficient for Nkx2-5 in the Isl1(+) lineage failed to downregulate Isl1 protein in cardiomyocytes of the heart tube. We demonstrated that Nkx2-5 directly binds to an Isl1 enhancer and represses Isl1 transcriptional activity. Furthermore, we showed that overexpression of Isl1 does not prevent cardiac differentiation of ESCs and in Xenopus laevis embryos. Instead, it leads to enhanced specification of cardiac progenitors, earlier cardiac differentiation, and increased cardiomyocyte number. Functional and molecular characterization of Isl1-overexpressing cardiomyocytes revealed higher beating frequencies in both ESC-derived contracting areas and Xenopus Isl1-gain-of-function hearts, which associated with upregulation of nodal-specific genes and downregulation of transcripts of working myocardium. Immunocytochemistry of cardiomyocyte lineage-specific markers demonstrated a reduction of ventricular cells and an increase of cells expressing the pacemaker channel Hcn4. Finally, optical action potential imaging of single cardiomyocytes combined with pharmacological approaches proved that Isl1 overexpression in ESCs resulted in normally electrophysiologically functional cells, highly enriched in the nodal subtype at the expense of the ventricular lineage. Our findings provide an Isl1/Nkx2-5-mediated mechanism that coordinately regulates the specification of cardiac progenitors toward the different myocardial lineages and ensures proper acquisition of myocyte subtype identity.
Collapse
Affiliation(s)
- Tatjana Dorn
- I. Medizinische Klinik und Poliklinik, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
121
|
Nam H, Lee S. Identification of STAM1 as a novel effector of ventral projection of spinal motor neurons. Development 2016; 143:2334-43. [DOI: 10.1242/dev.135848] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 05/04/2016] [Indexed: 12/27/2022]
Abstract
During spinal cord development, motor neuron (MN) axons exit the spinal cord ventrally, although the molecular basis for this process remains poorly understood. STAM1 and Hrs form a complex involved with endosomal targeting of cargo proteins, including the chemokine receptor CXCR4. Interestingly, the absence of CXCR4 signaling in spinal MNs is known to enforce improper extension of the axons into the dorsal side of the spinal cord. Here we report that the MN-specific Isl1-Lhx3 complex directly transactivates the Stam1 gene and STAM1 functions in determining the ventral spinal MN axonal projections. STAM1 is co-expressed with Hrs in embryonic spinal MNs, and knock-down of STAM1 in the developing chick spinal cord results in down-regulation of the expression of CXCR4, accompanied by dorsally projecting motor axons. Interestingly, overexpression of STAM1 or CXCR4 also results in dorsal projection of motor axons, suggesting that proper CXCR4 protein level is critical for the ventral motor axon trajectory. Our results reveal a critical regulatory axis for the ventral axonal trajectory of developing spinal MNs, consisting of the Isl1-Lhx3 complex, STAM1 and CXCR4.
Collapse
Affiliation(s)
- Heejin Nam
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Korea
| | - Seunghee Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Korea
| |
Collapse
|
122
|
Generating Diverse Spinal Motor Neuron Subtypes from Human Pluripotent Stem Cells. Stem Cells Int 2015; 2016:1036974. [PMID: 26823667 PMCID: PMC4707335 DOI: 10.1155/2016/1036974] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 09/14/2015] [Indexed: 12/18/2022] Open
Abstract
Resolving the mechanisms underlying human neuronal diversification remains a major challenge in developmental and applied neurobiology. Motor neurons (MNs) represent a diverse pool of neuronal subtypes exhibiting differential vulnerability in different human neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA). The ability to predictably manipulate MN subtype lineage restriction from human pluripotent stem cells (PSCs) will form the essential basis to establishing accurate, clinically relevant in vitro disease models. I first overview motor neuron developmental biology to provide some context for reviewing recent studies interrogating pathways that influence the generation of MN diversity. I conclude that motor neurogenesis from PSCs provides a powerful reductionist model system to gain insight into the developmental logic of MN subtype diversification and serves more broadly as a leading exemplar of potential strategies to resolve the molecular basis of neuronal subclass differentiation within the nervous system. These studies will in turn permit greater mechanistic understanding of differential MN subtype vulnerability using in vitro human disease models.
Collapse
|
123
|
Harada H, Omi M, Sato T, Nakamura H. Pea3 determines the isthmus region at the downstream of Fgf8-Ras-ERK signaling pathway. Dev Growth Differ 2015; 57:657-66. [DOI: 10.1111/dgd.12254] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/25/2015] [Accepted: 10/25/2015] [Indexed: 01/25/2023]
Affiliation(s)
- Hidekiyo Harada
- Department of Molecular Neurobiology; Institute of Development, Aging & Cancer and Graduate School of Life Sciences; Tohoku University; Sendai 980-8575 Miyagi Japan
| | - Minoru Omi
- Department of Molecular Neurobiology; Institute of Development, Aging & Cancer and Graduate School of Life Sciences; Tohoku University; Sendai 980-8575 Miyagi Japan
| | - Tatsuya Sato
- Department of Molecular Neurobiology; Institute of Development, Aging & Cancer and Graduate School of Life Sciences; Tohoku University; Sendai 980-8575 Miyagi Japan
| | - Harukazu Nakamura
- Department of Molecular Neurobiology; Institute of Development, Aging & Cancer and Graduate School of Life Sciences; Tohoku University; Sendai 980-8575 Miyagi Japan
| |
Collapse
|
124
|
Zhu H. Forkhead box transcription factors in embryonic heart development and congenital heart disease. Life Sci 2015; 144:194-201. [PMID: 26656470 DOI: 10.1016/j.lfs.2015.12.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 11/24/2015] [Accepted: 12/01/2015] [Indexed: 12/31/2022]
Abstract
Embryonic heart development is a very complicated process regulated precisely by a network composed of many genes and signaling pathways in time and space. Forkhead box (Fox, FOX) proteins are a family of transcription factors characterized by the presence of an evolutionary conserved "forkhead"or "winged-helix" DNA-binding domain and able to organize temporal and spatial gene expression during development. They are involved in a wide variety of cellular processes, such as cell cycle progression, proliferation, differentiation, migration, metabolism and DNA damage response. An abundance of studies in model organisms and systems has established that Foxa2, Foxc1/c2, Foxh1 and Foxm1, Foxos and Foxps are important components of the signaling pathways that instruct cardiogenesis and embryonic heart development, playing paramount roles in heart development. The previous studies also have demonstrated that mutations in some of the forkhead box genes and the aberrant expression of forkhead box gene are heavily implicated in the congenital heart disease (CHD) of humans. This review primarily focuses on the current understanding of heart development regulated by forkhead box transcription factors and molecular genetic mechanisms by which forkhead box factors modulate heart development during embryogenesis and organogenesis. This review also summarizes human CHD related mutations in forkhead box genes as well as the abnormal expression of forkhead box gene, and discusses additional possible regulatory mechanisms of the forkhead box genes during embryonic heart development that warrant further investigation.
Collapse
Affiliation(s)
- Hong Zhu
- Department of Biomedical Engineering, College of Biology, Hunan University, 1 Denggao Road, Yuelu District, Changsha, Hunan 410082, PR China.
| |
Collapse
|
125
|
Yang SL, Yang M, Herrlinger S, Liang C, Lai F, Chen JF. MiR-302/367 regulate neural progenitor proliferation, differentiation timing, and survival in neurulation. Dev Biol 2015; 408:140-50. [PMID: 26441343 DOI: 10.1016/j.ydbio.2015.09.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 09/26/2015] [Accepted: 09/26/2015] [Indexed: 11/16/2022]
Abstract
How neural progenitor cell (NPC) behaviors are temporally controlled in early developing embryos remains undefined. The in vivo functions of microRNAs (miRNAs) in early mammalian development remain largely unknown. Mir-302/367 is a miRNA cluster that encodes miR-367 and four miR-302 members (miR302a-d). We show that miR-302b is highly expressed in early neuroepithelium and its expression decline as development progresses. We generated a mir-302/367 knockout mouse model and found that deletion of mir-302/367 results in an early embryonic lethality and open neural tube defect (NTD). NPCs exhibit enhanced proliferation, precocious differentiation, and decreased cell survival in mutant embryos. Furthermore, we identified Fgf15, Cyclin D1, and D2 as direct targets of miR-302 in NPCs in vivo, and their expression is enhanced in mutant NPCs. Ectopic expression of Cyclin D1 and D2 increases NPC proliferation, while FGF19 (human ortholog of Fgf15) overexpression leads to an increase of NPC differentiation. Thus, these findings reveal essential roles of miR-302/367 in orchestrating gene expression and NPC behaviors in neurulation; they also point to miRNAs as critical genetic components associated with neural tube formation.
Collapse
Affiliation(s)
- Si-Lu Yang
- Department of Genetics, Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Mei Yang
- 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
| | - Chen Liang
- Department of Genetics, Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Fan Lai
- Biochemistry & Molecular Biology, University of Miami, Miami, FL 33136, USA
| | - Jian-Fu Chen
- Department of Genetics, Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
126
|
Kim N, Park C, Jeong Y, Song MR. Functional Diversification of Motor Neuron-specific Isl1 Enhancers during Evolution. PLoS Genet 2015; 11:e1005560. [PMID: 26447474 PMCID: PMC4598079 DOI: 10.1371/journal.pgen.1005560] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 09/09/2015] [Indexed: 11/19/2022] Open
Abstract
Functional diversification of motor neurons has occurred in order to selectively control the movements of different body parts including head, trunk and limbs. Here we report that transcription of Isl1, a major gene necessary for motor neuron identity, is controlled by two enhancers, CREST1 (E1) and CREST2 (E2) that allow selective gene expression of Isl1 in motor neurons. Introduction of GFP reporters into the chick neural tube revealed that E1 is active in hindbrain motor neurons and spinal cord motor neurons, whereas E2 is active in the lateral motor column (LMC) of the spinal cord, which controls the limb muscles. Genome-wide ChIP-Seq analysis combined with reporter assays showed that Phox2 and the Isl1-Lhx3 complex bind to E1 and drive hindbrain and spinal cord-specific expression of Isl1, respectively. Interestingly, Lhx3 alone was sufficient to activate E1, and this may contribute to the initiation of Isl1 expression when progenitors have just developed into motor neurons. E2 was induced by onecut 1 (OC-1) factor that permits Isl1 expression in LMCm neurons. Interestingly, the core region of E1 has been conserved in evolution, even in the lamprey, a jawless vertebrate with primitive motor neurons. All E1 sequences from lamprey to mouse responded equally well to Phox2a and the Isl1-Lhx3 complex. Conversely, E2, the enhancer for limb-innervating motor neurons, was only found in tetrapod animals. This suggests that evolutionarily-conserved enhancers permit the diversification of motor neurons. During evolution, motor neurons became specialized to control movements of different body parts including head, trunk and limbs. Here we report that two enhancers of Isl1, E1 and E2, are active together with transcription factors in motor neurons. Surprisingly, E1 and its response to transcription factors has been conserved in evolution from the lamprey to man, whereas E2 is only found in animals with limbs. Our study provides an evolutionary example of how functional diversification of motor neurons is achieved by a dynamic interplay between enhancers and transcription factors.
Collapse
Affiliation(s)
- Namhee Kim
- School of Life Sciences, Cell Dynamics Research Center, Gwangju Institute of Science and Technology, Oryong-dong, Buk-gu, Gwangju, Republic of Korea
| | - Chungoo Park
- School of Biological Sciences and Technology, Chonnam National University, Yongbong-ro, Buk-gu, Gwangju, Republic of Korea
| | - Yongsu Jeong
- Department of Genetic Engineering, College of Life Sciences and Graduate School of Biotechnology, Kyung Hee University, Yongin-si, Republic of Korea
| | - Mi-Ryoung Song
- School of Life Sciences, Cell Dynamics Research Center, Gwangju Institute of Science and Technology, Oryong-dong, Buk-gu, Gwangju, Republic of Korea
- * E-mail:
| |
Collapse
|
127
|
Bui TV, Stifani N, Panek I, Farah C. Genetically identified spinal interneurons integrating tactile afferents for motor control. J Neurophysiol 2015; 114:3050-63. [PMID: 26445867 DOI: 10.1152/jn.00522.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 09/28/2015] [Indexed: 11/22/2022] Open
Abstract
Our movements are shaped by our perception of the world as communicated by our senses. Perception of sensory information has been largely attributed to cortical activity. However, a prior level of sensory processing occurs in the spinal cord. Indeed, sensory inputs directly project to many spinal circuits, some of which communicate with motor circuits within the spinal cord. Therefore, the processing of sensory information for the purpose of ensuring proper movements is distributed between spinal and supraspinal circuits. The mechanisms underlying the integration of sensory information for motor control at the level of the spinal cord have yet to be fully described. Recent research has led to the characterization of spinal neuron populations that share common molecular identities. Identification of molecular markers that define specific populations of spinal neurons is a prerequisite to the application of genetic techniques devised to both delineate the function of these spinal neurons and their connectivity. This strategy has been used in the study of spinal neurons that receive tactile inputs from sensory neurons innervating the skin. As a result, the circuits that include these spinal neurons have been revealed to play important roles in specific aspects of motor function. We describe these genetically identified spinal neurons that integrate tactile information and the contribution of these studies to our understanding of how tactile information shapes motor output. Furthermore, we describe future opportunities that these circuits present for shedding light on the neural mechanisms of tactile processing.
Collapse
Affiliation(s)
- Tuan V Bui
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada; Center for Neural Dynamics, University of Ottawa, Ottawa, Ontario, Canada; and
| | - Nicolas Stifani
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Izabela Panek
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Carl Farah
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
128
|
Modeling amyotrophic lateral sclerosis in pure human iPSc-derived motor neurons isolated by a novel FACS double selection technique. Neurobiol Dis 2015; 82:269-280. [DOI: 10.1016/j.nbd.2015.06.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 05/30/2015] [Accepted: 06/17/2015] [Indexed: 01/01/2023] Open
|
129
|
Zhou C, Yang G, Chen M, He L, Xiang L, Ricupero C, Mao JJ, Ling J. Lhx6 and Lhx8: cell fate regulators and beyond. FASEB J 2015; 29:4083-91. [PMID: 26148970 PMCID: PMC4566936 DOI: 10.1096/fj.14-267500] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 06/22/2015] [Indexed: 12/11/2022]
Abstract
As transcription factors of the lines (LIN)-11/Islet (Isl)-1/mitosis entry checkpoint (MEC)-3 (LIM)-homeobox subfamily, LIM homeobox (Lhx)6 and -8 are remarkably conserved and involved in the morphogenesis of multiple organ systems. Lhx6 and -8 play overlapping and distinctive roles, but in general act as cell fate mediators and in turn are regulated by several transcriptional factors, such as sonic hedgehog, fibroblast growth factors, and wingless-int (Wnt)/β-catenin. In this review, we first summarize Lhx6 and -8 distributions in development and then explore how Lhx6 and -8 act as transcription factors and coregulators of cell lineage specification. Known Lhx6 and -8 functions and targets are outlined in neurogenesis, craniofacial development, and germ cell differentiation. The underlying mechanisms of Lhx6 and -8 in regulating cell fate remain elusive. Whether Lhx6 and -8 affect functions in tissues and organs other than neural, craniofacial, oocytes, and germ cells is largely unexplored. Taken together, Lhx6 and -8 are important regulators of cell lineage specification and may act as one of the pivotal mediators of stem cell fate. Undoubtedly, future investigations of Lhx6 and -8 biology will continue to yield fascinating insights into tissue development and homeostasis, in addition to their putative roles in tissue regeneration and ageing.
Collapse
Affiliation(s)
- Chen Zhou
- *Center for Craniofacial Regeneration, Columbia University Medical Center, New York, New York, USA; Guanghua School of Stomatology, Hospital of Stomatology, and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Guodong Yang
- *Center for Craniofacial Regeneration, Columbia University Medical Center, New York, New York, USA; Guanghua School of Stomatology, Hospital of Stomatology, and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Mo Chen
- *Center for Craniofacial Regeneration, Columbia University Medical Center, New York, New York, USA; Guanghua School of Stomatology, Hospital of Stomatology, and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Ling He
- *Center for Craniofacial Regeneration, Columbia University Medical Center, New York, New York, USA; Guanghua School of Stomatology, Hospital of Stomatology, and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Lusai Xiang
- *Center for Craniofacial Regeneration, Columbia University Medical Center, New York, New York, USA; Guanghua School of Stomatology, Hospital of Stomatology, and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Christopher Ricupero
- *Center for Craniofacial Regeneration, Columbia University Medical Center, New York, New York, USA; Guanghua School of Stomatology, Hospital of Stomatology, and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Jeremy J Mao
- *Center for Craniofacial Regeneration, Columbia University Medical Center, New York, New York, USA; Guanghua School of Stomatology, Hospital of Stomatology, and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Junqi Ling
- *Center for Craniofacial Regeneration, Columbia University Medical Center, New York, New York, USA; Guanghua School of Stomatology, Hospital of Stomatology, and Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
130
|
Sajgo S, Ali S, Popescu O, Badea TC. Dynamic expression of transcription factor Brn3b during mouse cranial nerve development. J Comp Neurol 2015; 524:1033-61. [PMID: 26356988 DOI: 10.1002/cne.23890] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 08/18/2015] [Accepted: 08/31/2015] [Indexed: 01/23/2023]
Abstract
During development, transcription factor combinatorial codes define a large variety of morphologically and physiologically distinct neurons. Such a combinatorial code has been proposed for the differentiation of projection neurons of the somatic and visceral components of cranial nerves. It is possible that individual neuronal cell types are not specified by unique transcription factors but rather emerge through the intersection of their expression domains. Brn3a, Brn3b, and Brn3c, in combination with each other and/or transcription factors of other families, can define subgroups of retinal ganglion cells (RGC), spiral and vestibular ganglia, inner ear and vestibular hair cell neurons in the vestibuloacoustic system, and groups of somatosensory neurons in the dorsal root ganglia. The present study investigates the expression and potential role of the Brn3b transcription factor in cranial nerves and associated nuclei of the brainstem. We report the dynamic expression of Brn3b in the somatosensory component of cranial nerves II, V, VII, and VIII and visceromotor nuclei of nerves VII, IX, and X as well as other brainstem nuclei during different stages of development into adult stage. We find that genetically identified Brn3b(KO) RGC axons show correct but delayed pathfinding during the early stages of embryonic development. However, loss of Brn3b does not affect the anatomy of the other cranial nerves normally expressing this transcription factor.
Collapse
Affiliation(s)
- Szilard Sajgo
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, 20892.,Molecular Biology Center, Interdisciplinary Research Institute on Bio-Nano-Science, Babes-Bolyai University, Cluj-Napoca, Cluj, 400084, Romania
| | - Seid Ali
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, 20892
| | - Octavian Popescu
- Molecular Biology Center, Interdisciplinary Research Institute on Bio-Nano-Science, Babes-Bolyai University, Cluj-Napoca, Cluj, 400084, Romania.,Institute of Biology, Romanian Academy, Bucharest, 060031, Romania
| | | |
Collapse
|
131
|
Expression and function of the LIM-homeodomain transcription factor Islet-1 in the developing and mature vertebrate retina. Exp Eye Res 2015; 138:22-31. [DOI: 10.1016/j.exer.2015.06.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 06/24/2015] [Accepted: 06/25/2015] [Indexed: 11/19/2022]
|
132
|
Castinetti F, Brinkmeier ML, Mortensen AH, Vella KR, Gergics P, Brue T, Hollenberg AN, Gan L, Camper SA. ISL1 Is Necessary for Maximal Thyrotrope Response to Hypothyroidism. Mol Endocrinol 2015; 29:1510-21. [PMID: 26296153 DOI: 10.1210/me.2015-1192] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
ISLET1 is a homeodomain transcription factor necessary for development of the pituitary, retina, motor neurons, heart, and pancreas. Isl1-deficient mice (Isl1(-/-)) die early during embryogenesis at embryonic day 10.5 due to heart defects, and at that time, they have an undersized pituitary primordium. ISL1 is expressed in differentiating pituitary cells in early embryogenesis. Here, we report the cell-specific expression of ISL1 and assessment of its role in gonadotropes and thyrotropes. Isl1 expression is elevated in pituitaries of Cga(-/-) mice, a model of hypothyroidism with thyrotrope hypertrophy and hyperplasia. Thyrotrope-specific disruption of Isl1 with Tshb-cre is permissive for normal serum TSH, but T4 levels are decreased, suggesting decreased thyrotrope function. Inducing hypothyroidism in normal mice causes a reduction in T4 levels and dramatically elevated TSH response, but mice with thyrotrope-specific disruption of Isl1 have a blunted TSH response. In contrast, deletion of Isl1 in gonadotropes with an Lhb-cre transgene has no obvious effect on gonadotrope function or fertility. These results show that ISL1 is necessary for maximal thyrotrope response to hypothyroidism, in addition to its role in development of Rathke's pouch.
Collapse
Affiliation(s)
- F Castinetti
- Human Genetics, University of Michigan (F.C., M.L.B., A.H.M., P.G., S.A.C.), Ann Arbor, Michigan 48109; Beth Israel Deaconess Medical Center (K.R.V., A.N.H.), Harvard University, Boston, Massachusetts 02215; Aix-Marseille University (F.C., T.B.), Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, Centre National de la Recherche Scientifique, Faculté de Médecine de Marseille, and Assistance Publique-Hôpitaux de Marseille, Department of Endocrinology, Hôpital de la Timone, Marseille, France 13000; and University of Rochester School of Medicine and Dentistry (L.G.), Rochester, New York 14642
| | - M L Brinkmeier
- Human Genetics, University of Michigan (F.C., M.L.B., A.H.M., P.G., S.A.C.), Ann Arbor, Michigan 48109; Beth Israel Deaconess Medical Center (K.R.V., A.N.H.), Harvard University, Boston, Massachusetts 02215; Aix-Marseille University (F.C., T.B.), Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, Centre National de la Recherche Scientifique, Faculté de Médecine de Marseille, and Assistance Publique-Hôpitaux de Marseille, Department of Endocrinology, Hôpital de la Timone, Marseille, France 13000; and University of Rochester School of Medicine and Dentistry (L.G.), Rochester, New York 14642
| | - A H Mortensen
- Human Genetics, University of Michigan (F.C., M.L.B., A.H.M., P.G., S.A.C.), Ann Arbor, Michigan 48109; Beth Israel Deaconess Medical Center (K.R.V., A.N.H.), Harvard University, Boston, Massachusetts 02215; Aix-Marseille University (F.C., T.B.), Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, Centre National de la Recherche Scientifique, Faculté de Médecine de Marseille, and Assistance Publique-Hôpitaux de Marseille, Department of Endocrinology, Hôpital de la Timone, Marseille, France 13000; and University of Rochester School of Medicine and Dentistry (L.G.), Rochester, New York 14642
| | - K R Vella
- Human Genetics, University of Michigan (F.C., M.L.B., A.H.M., P.G., S.A.C.), Ann Arbor, Michigan 48109; Beth Israel Deaconess Medical Center (K.R.V., A.N.H.), Harvard University, Boston, Massachusetts 02215; Aix-Marseille University (F.C., T.B.), Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, Centre National de la Recherche Scientifique, Faculté de Médecine de Marseille, and Assistance Publique-Hôpitaux de Marseille, Department of Endocrinology, Hôpital de la Timone, Marseille, France 13000; and University of Rochester School of Medicine and Dentistry (L.G.), Rochester, New York 14642
| | - P Gergics
- Human Genetics, University of Michigan (F.C., M.L.B., A.H.M., P.G., S.A.C.), Ann Arbor, Michigan 48109; Beth Israel Deaconess Medical Center (K.R.V., A.N.H.), Harvard University, Boston, Massachusetts 02215; Aix-Marseille University (F.C., T.B.), Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, Centre National de la Recherche Scientifique, Faculté de Médecine de Marseille, and Assistance Publique-Hôpitaux de Marseille, Department of Endocrinology, Hôpital de la Timone, Marseille, France 13000; and University of Rochester School of Medicine and Dentistry (L.G.), Rochester, New York 14642
| | - T Brue
- Human Genetics, University of Michigan (F.C., M.L.B., A.H.M., P.G., S.A.C.), Ann Arbor, Michigan 48109; Beth Israel Deaconess Medical Center (K.R.V., A.N.H.), Harvard University, Boston, Massachusetts 02215; Aix-Marseille University (F.C., T.B.), Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, Centre National de la Recherche Scientifique, Faculté de Médecine de Marseille, and Assistance Publique-Hôpitaux de Marseille, Department of Endocrinology, Hôpital de la Timone, Marseille, France 13000; and University of Rochester School of Medicine and Dentistry (L.G.), Rochester, New York 14642
| | - A N Hollenberg
- Human Genetics, University of Michigan (F.C., M.L.B., A.H.M., P.G., S.A.C.), Ann Arbor, Michigan 48109; Beth Israel Deaconess Medical Center (K.R.V., A.N.H.), Harvard University, Boston, Massachusetts 02215; Aix-Marseille University (F.C., T.B.), Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, Centre National de la Recherche Scientifique, Faculté de Médecine de Marseille, and Assistance Publique-Hôpitaux de Marseille, Department of Endocrinology, Hôpital de la Timone, Marseille, France 13000; and University of Rochester School of Medicine and Dentistry (L.G.), Rochester, New York 14642
| | - L Gan
- Human Genetics, University of Michigan (F.C., M.L.B., A.H.M., P.G., S.A.C.), Ann Arbor, Michigan 48109; Beth Israel Deaconess Medical Center (K.R.V., A.N.H.), Harvard University, Boston, Massachusetts 02215; Aix-Marseille University (F.C., T.B.), Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, Centre National de la Recherche Scientifique, Faculté de Médecine de Marseille, and Assistance Publique-Hôpitaux de Marseille, Department of Endocrinology, Hôpital de la Timone, Marseille, France 13000; and University of Rochester School of Medicine and Dentistry (L.G.), Rochester, New York 14642
| | - S A Camper
- Human Genetics, University of Michigan (F.C., M.L.B., A.H.M., P.G., S.A.C.), Ann Arbor, Michigan 48109; Beth Israel Deaconess Medical Center (K.R.V., A.N.H.), Harvard University, Boston, Massachusetts 02215; Aix-Marseille University (F.C., T.B.), Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille, Centre National de la Recherche Scientifique, Faculté de Médecine de Marseille, and Assistance Publique-Hôpitaux de Marseille, Department of Endocrinology, Hôpital de la Timone, Marseille, France 13000; and University of Rochester School of Medicine and Dentistry (L.G.), Rochester, New York 14642
| |
Collapse
|
133
|
Critical Roles of the LIM Domains of Lhx3 in Recruiting Coactivators to the Motor Neuron-Specifying Isl1-Lhx3 Complex. Mol Cell Biol 2015; 35:3579-89. [PMID: 26260513 DOI: 10.1128/mcb.00335-15] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 08/03/2015] [Indexed: 11/20/2022] Open
Abstract
During spinal cord development, the LIM domains of the LIM homeodomain factor Lhx3 bind to either the LIM cofactor nuclear LIM interactor (NLI) or another LIM homeodomain factor, Isl1, assembling the tetrameric V2 interneuron-specifying Lhx3 complex (2NLI:2Lhx3) or the hexameric motor neuron-specifying Isl1-Lhx3 complex (2NLI:2Isl1:2Lhx3). However, the detailed molecular basis by which the Lhx3-LIM domains contribute to motor neuron specification still remains poorly understood. Here, we show that the Lhx3-LIM domains are essential for recruiting transcriptional coactivators to the Isl1-Lhx3 complex. Using a yeast genetic screening system, we identify Lhx3 point mutants that bind to NLI but not Isl1. Accordingly, these mutants fail to assemble the Isl1-Lhx3 complex. However, their interaction with coactivators is relatively intact, and they are fully functional in the Lhx3 complex and V2 interneuron specification. Interestingly, when these Lhx3 mutants are directly fused to Isl1, their transcriptional activity in the Isl1-Lhx3 complex is restored. We further show that this restoration reflects an unexpected role of the Lhx3-LIM domains, likely together with Isl1, to form an interaction interface for coactivators. Our results suggest that the Lhx3-LIM domains play critical roles in transactivation of the Isl1-Lhx3 complex by not only directing the assembly of the Isl1-Lhx3 complex but also recruiting coactivators to the complex.
Collapse
|
134
|
Abstract
PURPOSE OF REVIEW This review will discuss recent advances in understanding mouse and human pancreatic islet cell development, novel concepts related to β cell dysfunction and improved approaches for replenishing β cells to treat diabetes. RECENT FINDINGS Considerable knowledge about pancreatic islet development and function has been gained using model systems with subsequent validation in human tissues. Recently, several rodent studies have revealed that differentiated adult islet cells retain remarkable plasticity and can be converted to other islet cell types by perturbing their transcription factor profiles. Furthermore, significant advances have been made in the generation of β-like cells from stem cell populations. Therefore, the generation of functionally mature β cells by the in-situ conversion of non-β cell populations or by the directed differentiation of human pluripotent stem cells could represent novel mechanisms for replenishing β cells in diabetic patients. SUMMARY The overall conservation between mouse and human pancreatic development, islet physiology and etiology of diabetes encourages the translation of novel β cell replacement therapies to humans. Further deciphering the molecular mechanisms that direct islet cell regeneration, plasticity and function could improve and expand the β cell replacement strategies for treating diabetes.
Collapse
Affiliation(s)
- Anthony I Romer
- Department of Genetics and Development, Columbia University, New York, New York, USA
| | | |
Collapse
|
135
|
Huettl RE, Eckstein S, Stahl T, Petricca S, Ninkovic J, Götz M, Huber AB. Functional dissection of the Pax6 paired domain: Roles in neural tube patterning and peripheral nervous system development. Dev Biol 2015; 413:86-103. [PMID: 26187199 DOI: 10.1016/j.ydbio.2015.07.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 06/21/2015] [Accepted: 07/11/2015] [Indexed: 10/23/2022]
Abstract
During development of the CNS, stem and progenitor cell proliferation, cell fate designation, and patterning decisions are tightly regulated by interdependent networks of key transcriptional regulators. In a genetic approach we analyzed divergent functionality of the PAI and RED sub-domains of the Pax6 Paired domain (PD) during progenitor zone formation, motor and interneuron development, and peripheral connectivity at distinct levels within the neural tube: within the hindbrain, mutation of the PAI sub-domain severely affected patterning of the p3 and pMN domains and establishment of the corresponding motor neurons. Exit point designation of hypoglossal axons was disturbed in embryos harboring either mutations in the PD sub-domains or containing a functional Pax6 Null allele. At brachial spinal levels, we propose a selective involvement of the PAI sub-domain during patterning of ventral p2 and pMN domains, critically disturbing generation of specific motor neuron subtypes and increasing V2 interneuron numbers. Our findings present a novel aspect of how Pax6 not only utilizes its modular structure to perform distinct functions via its paired and homeodomain. Individual sub-domains can exert distinct functions, generating a new level of complexity for transcriptional regulation by one single transcription factor not only in dorso-ventral, but also rostro-caudal neural tube patterning.
Collapse
Affiliation(s)
- Rosa-Eva Huettl
- Institute of Developmental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Simone Eckstein
- Institute of Developmental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Tessa Stahl
- Institute of Stem Cell Research, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Stefania Petricca
- Institute of Stem Cell Research, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Jovica Ninkovic
- Institute of Stem Cell Research, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Magdalena Götz
- Institute of Stem Cell Research, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Andrea B Huber
- Institute of Developmental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany.
| |
Collapse
|
136
|
Russ JB, Kaltschmidt JA. From induction to conduction: how intrinsic transcriptional priming of extrinsic neuronal connectivity shapes neuronal identity. Open Biol 2015; 4:rsob.140144. [PMID: 25297387 PMCID: PMC4221895 DOI: 10.1098/rsob.140144] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Every behaviour of an organism relies on an intricate and vastly diverse network of neurons whose identity and connectivity must be specified with extreme precision during development. Intrinsically, specification of neuronal identity depends heavily on the expression of powerful transcription factors that direct numerous features of neuronal identity, including especially properties of neuronal connectivity, such as dendritic morphology, axonal targeting or synaptic specificity, ultimately priming the neuron for incorporation into emerging circuitry. As the neuron's early connectivity is established, extrinsic signals from its pre- and postsynaptic partners feedback on the neuron to further refine its unique characteristics. As a result, disruption of one component of the circuitry during development can have vital consequences for the proper identity specification of its synaptic partners. Recent studies have begun to harness the power of various transcription factors that control neuronal cell fate, including those that specify a neuron's subtype-specific identity, seeking insight for future therapeutic strategies that aim to reconstitute damaged circuitry through neuronal reprogramming.
Collapse
Affiliation(s)
- Jeffrey B Russ
- Weill Cornell/Rockefeller University/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10065, USA Neuroscience Program, Weill Cornell Medical College, New York, NY 10065, USA Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Julia A Kaltschmidt
- Neuroscience Program, Weill Cornell Medical College, New York, NY 10065, USA Cell and Developmental Biology Program, Weill Cornell Medical College, New York, NY 10065, USA Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| |
Collapse
|
137
|
Rosin JM, Kurrasch DM, Cobb J. Shox2 is required for the proper development of the facial motor nucleus and the establishment of the facial nerves. BMC Neurosci 2015; 16:39. [PMID: 26156498 PMCID: PMC4495855 DOI: 10.1186/s12868-015-0176-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/12/2015] [Indexed: 11/10/2022] Open
Abstract
Background Axons from the visceral motor neurons (vMNs) project from nuclei in the hindbrain to innervate autonomic ganglia and branchial arch-derived muscles. Although much is known about the events that govern specification of somatic motor neurons, the genetic pathways responsible for the development of vMNs are less well characterized. We know that vMNs, like all motor neurons, depend on sonic hedgehog signaling for their generation. Similarly, the paired-like homeobox 2b (Phox2b) gene, which is expressed in both proliferating progenitors and post-mitotic motor neurons, is essential for the development of vMNs. Given that our previous study identified a novel role for the short stature homeobox 2 (Shox2) gene in the hindbrain, and since SHOX2 has been shown to regulate transcription of islet 1 (Isl1), an important regulator of vMN development, we sought to determine whether Shox2 is required for the proper development of the facial motor nucleus. Results Using a Nestin-Cre driver, we show that elimination of Shox2 throughout the brain results in elevated cell death in the facial motor nucleus at embryonic day 12.5 (E12.5) and E14.5, which correlates with impaired axonal projection properties of vMNs. We also observed changes in the spatial expression of the vMN cell fate factors Isl1 and Phox2b, and concomitant defects in Shh and Ptch1 expression in Shox2 mutants. Furthermore, we demonstrate that elimination of Shox2 results in the loss of dorsomedial and ventromedial subnuclei by postnatal day 0 (P0), which may explain the changes in physical activity and impaired feeding/nursing behavior in Shox2 mutants. Conclusions Combined, our data show that Shox2 is required for development of the facial motor nucleus and its associated facial (VII) nerves, and serves as a new molecular tool to probe the genetic programs of this understudied hindbrain region. Electronic supplementary material The online version of this article (doi:10.1186/s12868-015-0176-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jessica M Rosin
- Department of Biological Sciences, University of Calgary, 2500 University Drive N.W., BI286D, Calgary, AB, T2N 1N4, Canada.
| | - Deborah M Kurrasch
- Department of Medical Genetics, Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive N.W., Room HS2275, Calgary, AB, T2N 4N1, Canada.
| | - John Cobb
- Department of Biological Sciences, University of Calgary, 2500 University Drive N.W., BI286D, Calgary, AB, T2N 1N4, Canada.
| |
Collapse
|
138
|
Pancreatic regeneration: basic research and gene regulation. Surg Today 2015; 46:633-40. [PMID: 26148809 DOI: 10.1007/s00595-015-1215-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 05/19/2015] [Indexed: 12/28/2022]
|
139
|
Lu DC, Niu T, Alaynick WA. Molecular and cellular development of spinal cord locomotor circuitry. Front Mol Neurosci 2015; 8:25. [PMID: 26136656 PMCID: PMC4468382 DOI: 10.3389/fnmol.2015.00025] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/30/2015] [Indexed: 01/20/2023] Open
Abstract
The spinal cord of vertebrate animals is comprised of intrinsic circuits that are capable of sensing the environment and generating complex motor behaviors. There are two major perspectives for understanding the biology of this complicated structure. The first approaches the spinal cord from the point of view of function and is based on classic and ongoing research in electrophysiology, adult behavior, and spinal cord injury. The second view considers the spinal cord from a developmental perspective and is founded mostly on gene expression and gain-of-function and loss-of-function genetic experiments. Together these studies have uncovered functional classes of neurons and their lineage relationships. In this review, we summarize our knowledge of developmental classes, with an eye toward understanding the functional roles of each group.
Collapse
Affiliation(s)
- Daniel C Lu
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - Tianyi Niu
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - William A Alaynick
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| |
Collapse
|
140
|
Chumak T, Bohuslavova R, Macova I, Dodd N, Buckiova D, Fritzsch B, Syka J, Pavlinkova G. Deterioration of the Medial Olivocochlear Efferent System Accelerates Age-Related Hearing Loss in Pax2-Isl1 Transgenic Mice. Mol Neurobiol 2015; 53:2368-83. [PMID: 25990412 DOI: 10.1007/s12035-015-9215-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 05/07/2015] [Indexed: 11/28/2022]
Abstract
The development, maturation, and maintenance of the inner ear are governed by temporal and spatial expression cascades of transcription factors that form a gene regulatory network. ISLET1 (ISL1) may be one of the major players in this cascade, and in order to study its role in the regulation of inner ear development, we produced a transgenic mouse overexpressing Isl1 under the Pax2 promoter. Pax2-regulated ISL1 overexpression increases the embryonic ISL1(+) domain and induces accelerated nerve fiber extension and branching in E12.5 embryos. Despite these gains in early development, the overexpression of ISL1 impairs the maintenance and function of hair cells of the organ of Corti. Mutant mice exhibit hyperactivity, circling behavior, and progressive age-related decline in hearing functions, which is reflected in reduced otoacoustic emissions (DPOAEs) followed by elevated hearing thresholds. The reduction of the amplitude of DPOAEs in transgenic mice was first detected at 1 month of age. By 6-9 months of age, DPOAEs completely disappeared, suggesting a functional inefficiency of outer hair cells (OHCs). The timing of DPOAE reduction coincides with the onset of the deterioration of cochlear efferent terminals. In contrast to these effects on efferents, we only found a moderate loss of OHCs and spiral ganglion neurons. For the first time, our results show that the genetic alteration of the medial olivocochlear (MOC) efferent system induces an early onset of age-related hearing loss. Thus, the neurodegeneration of the MOC system could be a contributing factor to the pathology of age-related hearing loss.
Collapse
Affiliation(s)
- Tetyana Chumak
- Institute of Experimental Medicine, CAS, Prague, Czechia
| | - Romana Bohuslavova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology, CAS, Prague 4, CZ-142 20, Prague, Czechia
| | - Iva Macova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology, CAS, Prague 4, CZ-142 20, Prague, Czechia
| | - Nicole Dodd
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology, CAS, Prague 4, CZ-142 20, Prague, Czechia
| | | | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, IA, 52242, USA
| | - Josef Syka
- Institute of Experimental Medicine, CAS, Prague, Czechia
| | - Gabriela Pavlinkova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology, CAS, Prague 4, CZ-142 20, Prague, Czechia.
| |
Collapse
|
141
|
Jarrar W, Dias JM, Ericson J, Arnold HH, Holz A. Nkx2.2 and Nkx2.9 are the key regulators to determine cell fate of branchial and visceral motor neurons in caudal hindbrain. PLoS One 2015; 10:e0124408. [PMID: 25919494 PMCID: PMC4412715 DOI: 10.1371/journal.pone.0124408] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 03/05/2015] [Indexed: 12/12/2022] Open
Abstract
Cranial motor nerves in vertebrates are comprised of the three principal subtypes of branchial, visceral, and somatic motor neurons, which develop in typical patterns along the anteroposterior and dorsoventral axes of hindbrain. Here we demonstrate that the formation of branchial and visceral motor neurons critically depends on the transcription factors Nkx2.2 and Nkx2.9, which together determine the cell fate of neuronal progenitor cells. Disruption of both genes in mouse embryos results in complete loss of the vagal and spinal accessory motor nerves, and partial loss of the facial and glossopharyngeal motor nerves, while the purely somatic hypoglossal and abducens motor nerves are not diminished. Cell lineage analysis in a genetically marked mouse line reveals that alterations of cranial nerves in Nkx2.2; Nkx2.9 double-deficient mouse embryos result from changes of cell fate in neuronal progenitor cells. As a consequence progenitors of branchiovisceral motor neurons in the ventral p3 domain of hindbrain are transformed to somatic motor neurons, which use ventral exit points to send axon trajectories to their targets. Cell fate transformation is limited to the caudal hindbrain, as the trigeminal nerve is not affected in double-mutant embryos suggesting that Nkx2.2 and Nkx2.9 proteins play no role in the development of branchiovisceral motor neurons in hindbrain rostral to rhombomere 4.
Collapse
Affiliation(s)
- Wassan Jarrar
- Cell and Molecular Biology, Zoological Institute, University of Braunschweig, Braunschweig, Germany
| | - Jose M. Dias
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Johan Ericson
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Hans-Henning Arnold
- Cell and Molecular Biology, Zoological Institute, University of Braunschweig, Braunschweig, Germany
- * E-mail: (AH); (HHA)
| | - Andreas Holz
- Cell and Molecular Biology, Zoological Institute, University of Braunschweig, Braunschweig, Germany
- * E-mail: (AH); (HHA)
| |
Collapse
|
142
|
Pathania R, Ramachandran S, Elangovan S, Padia R, Yang P, Cinghu S, Veeranan-Karmegam R, Arjunan P, Gnana-Prakasam JP, Sadanand F, Pei L, Chang CS, Choi JH, Shi H, Manicassamy S, Prasad PD, Sharma S, Ganapathy V, Jothi R, Thangaraju M. DNMT1 is essential for mammary and cancer stem cell maintenance and tumorigenesis. Nat Commun 2015; 6:6910. [PMID: 25908435 PMCID: PMC4410389 DOI: 10.1038/ncomms7910] [Citation(s) in RCA: 182] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/09/2015] [Indexed: 02/07/2023] Open
Abstract
Mammary stem/progenitor cells (MaSCs) maintain self-renewal of the mammary epithelium during puberty and pregnancy. DNA methylation provides a potential epigenetic mechanism for maintaining cellular memory during self-renewal. Although DNA methyltransferases (DNMTs) are dispensable for embryonic stem cell maintenance, their role in maintaining MaSCs and cancer stem cells (CSCs) in constantly replenishing mammary epithelium is unclear. Here we show that DNMT1 is indispensable for MaSC maintenance. Furthermore, we find that DNMT1 expression is elevated in mammary tumors, and mammary gland-specific DNMT1 deletion protects mice from mammary tumorigenesis by limiting the CSC pool. Through genome-scale methylation studies, we identify ISL1 as a direct DNMT1 target, hypermethylated and downregulated in mammary tumors and CSCs. DNMT inhibition or ISL1 expression in breast cancer cells limits CSC population. Altogether, our studies uncover an essential role for DNMT1 in MaSC and CSC maintenance and identify DNMT1-ISL1 axis as a potential therapeutic target for breast cancer treatment.
Collapse
Affiliation(s)
- Rajneesh Pathania
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Sabarish Ramachandran
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Selvakumar Elangovan
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Ravi Padia
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Pengyi Yang
- System Biology Section, Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | - Senthilkumar Cinghu
- System Biology Section, Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | - Rajalakshmi Veeranan-Karmegam
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Pachiappan Arjunan
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Jaya P Gnana-Prakasam
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Fulzele Sadanand
- Department of Orthopedic Surgery, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Lirong Pei
- Department of Pathology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Chang-Sheng Chang
- Department of Pathology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Jeong-Hyeon Choi
- Department of Biostatistics, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA.,Cancer Research Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Huidong Shi
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA.,Cancer Research Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Santhakumar Manicassamy
- Cancer Research Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Puttur D Prasad
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA.,Cancer Research Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Suash Sharma
- Department of Pathology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA.,Cancer Research Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Vadivel Ganapathy
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA.,Cancer Research Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Raja Jothi
- System Biology Section, Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | - Muthusamy Thangaraju
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA.,Cancer Research Center, Medical College of Georgia, Georgia Regents University, Augusta, Georgia 30912, USA
| |
Collapse
|
143
|
Allan DW, Thor S. Transcriptional selectors, masters, and combinatorial codes: regulatory principles of neural subtype specification. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2015; 4:505-28. [PMID: 25855098 PMCID: PMC4672696 DOI: 10.1002/wdev.191] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 03/04/2015] [Accepted: 03/04/2015] [Indexed: 01/08/2023]
Abstract
The broad range of tissue and cellular diversity of animals is generated to a large extent by the hierarchical deployment of sequence-specific transcription factors and co-factors (collectively referred to as TF's herein) during development. Our understanding of these developmental processes has been facilitated by the recognition that the activities of many TF's can be meaningfully described by a few functional categories that usefully convey a sense for how the TF's function, and also provides a sense for the regulatory organization of the developmental processes in which they participate. Here, we draw on examples from studies in Caenorhabditis elegans, Drosophila melanogaster, and vertebrates to discuss how the terms spatial selector, temporal selector, tissue/cell type selector, terminal selector and combinatorial code may be usefully applied to categorize the activities of TF's at critical steps of nervous system construction. While we believe that these functional categories are useful for understanding the organizational principles by which TF's direct nervous system construction, we however caution against the assumption that a TF's function can be solely or fully defined by any single functional category. Indeed, most TF's play diverse roles within different functional categories, and their roles can blur the lines we draw between these categories. Regardless, it is our belief that the concepts discussed here are helpful in clarifying the regulatory complexities of nervous system development, and hope they prove useful when interpreting mutant phenotypes, designing future experiments, and programming specific neuronal cell types for use in therapies. WIREs Dev Biol 2015, 4:505–528. doi: 10.1002/wdev.191 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Douglas W Allan
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Stefan Thor
- Department of Clinical and Experimental Medicine, Linkoping University, Linkoping, Sweden
| |
Collapse
|
144
|
Lee H, Kim M, Kim N, Macfarlan T, Pfaff SL, Mastick GS, Song MR. Slit and Semaphorin signaling governed by Islet transcription factors positions motor neuron somata within the neural tube. Exp Neurol 2015; 269:17-27. [PMID: 25843547 DOI: 10.1016/j.expneurol.2015.03.024] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 03/25/2015] [Accepted: 03/27/2015] [Indexed: 01/15/2023]
Abstract
Motor neurons send out axons to peripheral muscles while their cell bodies remain in the ventral spinal cord. The unique configuration of motor neurons spanning the border between the CNS and PNS has been explained by structural barriers such as boundary cap (BC) cells, basal lamina and radial glia. However, mechanisms in motor neurons that retain their position have not been addressed yet. Here we demonstrate that the Islet1 (Isl1) and Islet2 (Isl2) transcription factors, which are essential for acquisition of motor neuron identity, also contribute to restrict motor neurons within the neural tube. In mice that lack both Isl1 and Isl2, large numbers of motor neurons exited the neural tube, even prior to the appearance of BC cells at the ventral exit points. Transcriptional profiling of motor neurons derived from Isl1 null embryonic stem cells revealed that transcripts of major genes involved in repulsive mechanisms were misregulated. Particularly, expression of Neuropilin1 (Npr1) and Slit2 mRNA was diminished in Islet mutant mice, and these could be target genes of the Islet proteins. Consistent with this mechanism, Robo and Slit mutations in mice and knockdown of Npr1 and Slit2 in chick embryos caused motor neurons to migrate to the periphery. Together, our study suggests that Islet genes engage Robo-Slit and Neuropilin-Semaphorin signaling in motor neurons to retain motor somata within the CNS.
Collapse
Affiliation(s)
- Hojae Lee
- School of Life Sciences, Cell Dynamics Research Center, Gwangju Institute of Science and Technology, Oryong-dong, Buk-gu, Gwangju 500-712, Republic of Korea
| | - Minkyung Kim
- Department of Biology, University of Nevada, Reno, NV 89557, USA
| | - Namhee Kim
- School of Life Sciences, Cell Dynamics Research Center, Gwangju Institute of Science and Technology, Oryong-dong, Buk-gu, Gwangju 500-712, Republic of Korea
| | - Todd Macfarlan
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Samuel L Pfaff
- Gene Expression Laboratory and the Howard Hughes Medical Institute, The Salk Institute for Biological Studies, 10010 North Torrey Pines, La Jolla, CA 92037, USA
| | - Grant S Mastick
- Department of Biology, University of Nevada, Reno, NV 89557, USA
| | - Mi-Ryoung Song
- School of Life Sciences, Cell Dynamics Research Center, Gwangju Institute of Science and Technology, Oryong-dong, Buk-gu, Gwangju 500-712, Republic of Korea.
| |
Collapse
|
145
|
Gergics P, Brinkmeier ML, Camper SA. Lhx4 deficiency: increased cyclin-dependent kinase inhibitor expression and pituitary hypoplasia. Mol Endocrinol 2015; 29:597-612. [PMID: 25668206 PMCID: PMC4399274 DOI: 10.1210/me.2014-1380] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 02/06/2015] [Indexed: 12/30/2022] Open
Abstract
Defects in the Lhx4, Lhx3, and Pitx2 genes can cause combined pituitary hormone deficiency and pituitary hypoplasia in both humans and mice. Not much is known about the mechanism underlying hypoplasia in these mutants beyond generally increased cell death and poorly maintained proliferation. We identified both common and unique abnormalities in developmental regulation of key cell cycle regulator gene expression in each of these three mutants. All three mutants exhibit reduced expression of the proliferative marker Ki67 and the transitional marker p57. We discovered that expression of the cyclin-dependent kinase inhibitor 1a (Cdkn1a or p21) is expanded dorsally in the pituitary primordium of both Lhx3 and Lhx4 mutants. Uniquely, Lhx4 mutants exhibit reduced cyclin D1 expression and have auxiliary pouch-like structures. We show evidence for indirect and direct effects of LHX4 on p21 expression in αT3-1 pituitary cells. In summary, Lhx4 is necessary for efficient pituitary progenitor cell proliferation and restriction of p21 expression.
Collapse
Affiliation(s)
- Peter Gergics
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan 48109
| | | | | |
Collapse
|
146
|
Islet 1 specifies the identity of hypothalamic melanocortin neurons and is critical for normal food intake and adiposity in adulthood. Proc Natl Acad Sci U S A 2015; 112:E1861-70. [PMID: 25825735 DOI: 10.1073/pnas.1500672112] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Food intake and body weight regulation depend on proper expression of the proopiomelanocortin gene (Pomc) in a group of neurons located in the mediobasal hypothalamus of all vertebrates. These neurons release POMC-encoded melanocortins, which are potent anorexigenic neuropeptides, and their absence from mice or humans leads to hyperphagia and severe obesity. Although the pathophysiology of hypothalamic POMC neurons is well understood, the genetic program that establishes the neuronal melanocortinergic phenotype and maintains a fully functional neuronal POMC phenotype throughout adulthood remains unknown. Here, we report that the early expression of the LIM-homeodomain transcription factor Islet 1 (ISL1) in the developing hypothalamus promotes the terminal differentiation of melanocortinergic neurons and is essential for hypothalamic Pomc expression since its initial onset and throughout the entire lifetime. We detected ISL1 in the prospective hypothalamus just before the onset of Pomc expression and, from then on, Pomc and Isl1 coexpress. ISL1 binds in vitro and in vivo to critical homeodomain binding DNA motifs present in the neuronal Pomc enhancers nPE1 and nPE2, and mutations of these sites completely disrupt the ability of these enhancers to drive reporter gene expression to hypothalamic POMC neurons in transgenic mice and zebrafish. ISL1 is necessary for hypothalamic Pomc expression during mouse and zebrafish embryogenesis. Furthermore, conditional Isl1 inactivation from POMC neurons impairs Pomc expression, leading to hyperphagia and obesity. Our results demonstrate that ISL1 specifies the identity of hypothalamic melanocortin neurons and is required for melanocortin-induced satiety and normal adiposity throughout the entire lifespan.
Collapse
|
147
|
Giszter SF. Spinal primitives and intra-spinal micro-stimulation (ISMS) based prostheses: a neurobiological perspective on the "known unknowns" in ISMS and future prospects. Front Neurosci 2015; 9:72. [PMID: 25852454 PMCID: PMC4367173 DOI: 10.3389/fnins.2015.00072] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 02/18/2014] [Indexed: 11/13/2022] Open
Abstract
The current literature on Intra-Spinal Micro-Stimulation (ISMS) for motor prostheses is reviewed in light of neurobiological data on spinal organization, and a neurobiological perspective on output motor modularity, ISMS maps, stimulation combination effects, and stability. By comparing published data in these areas, the review identifies several gaps in current knowledge that are crucial to the development of effective intraspinal neuroprostheses. Gaps can be categorized into a lack of systematic and reproducible details of: (a) Topography and threshold for ISMS across the segmental motor system, the topography of autonomic recruitment by ISMS, and the coupling relations between these two types of outputs in practice. (b) Compositional rules for ISMS motor responses tested across the full range of the target spinal topographies. (c) Rules for ISMS effects' dependence on spinal cord state and neural dynamics during naturally elicited or ISMS triggered behaviors. (d) Plasticity of the compositional rules for ISMS motor responses, and understanding plasticity of ISMS topography in different spinal cord lesion states, disease states, and following rehabilitation. All these knowledge gaps to a greater or lesser extent require novel electrode technology in order to allow high density chronic recording and stimulation. The current lack of this technology may explain why these prominent gaps in the ISMS literature currently exist. It is also argued that given the "known unknowns" in the current ISMS literature, it may be prudent to adopt and develop control schemes that can manage the current results with simple superposition and winner-take-all interactions, but can also incorporate the possible plastic and stochastic dynamic interactions that may emerge in fuller analyses over longer terms, and which have already been noted in some simpler model systems.
Collapse
Affiliation(s)
- Simon F Giszter
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Drexel University Philadelphia, PA, USA ; School of Biomedical Engineering and Health Systems, Drexel University Philadelphia, PA, USA
| |
Collapse
|
148
|
Draaken M, Knapp M, Pennimpede T, Schmidt JM, Ebert AK, Rösch W, Stein R, Utsch B, Hirsch K, Boemers TM, Mangold E, Heilmann S, Ludwig KU, Jenetzky E, Zwink N, Moebus S, Herrmann BG, Mattheisen M, Nöthen MM, Ludwig M, Reutter H. Genome-wide association study and meta-analysis identify ISL1 as genome-wide significant susceptibility gene for bladder exstrophy. PLoS Genet 2015; 11:e1005024. [PMID: 25763902 PMCID: PMC4357422 DOI: 10.1371/journal.pgen.1005024] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 01/26/2015] [Indexed: 11/18/2022] Open
Abstract
The bladder exstrophy-epispadias complex (BEEC) represents the severe end of the uro-rectal malformation spectrum, and is thought to result from aberrant embryonic morphogenesis of the cloacal membrane and the urorectal septum. The most common form of BEEC is isolated classic bladder exstrophy (CBE). To identify susceptibility loci for CBE, we performed a genome-wide association study (GWAS) of 110 CBE patients and 1,177 controls of European origin. Here, an association was found with a region of approximately 220kb on chromosome 5q11.1. This region harbors the ISL1 (ISL LIM homeobox 1) gene. Multiple markers in this region showed evidence for association with CBE, including 84 markers with genome-wide significance. We then performed a meta-analysis using data from a previous GWAS by our group of 98 CBE patients and 526 controls of European origin. This meta-analysis also implicated the 5q11.1 locus in CBE risk. A total of 138 markers at this locus reached genome-wide significance in the meta-analysis, and the most significant marker (rs9291768) achieved a P value of 2.13 × 10-12. No other locus in the meta-analysis achieved genome-wide significance. We then performed murine expression analyses to follow up this finding. Here, Isl1 expression was detected in the genital region within the critical time frame for human CBE development. Genital regions with Isl1 expression included the peri-cloacal mesenchyme and the urorectal septum. The present study identified the first genome-wide significant locus for CBE at chromosomal region 5q11.1, and provides strong evidence for the hypothesis that ISL1 is the responsible candidate gene in this region.
Collapse
Affiliation(s)
- Markus Draaken
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
- * E-mail:
| | - Michael Knapp
- Institute of Medical Biometry, Informatics, and Epidemiology, University of Bonn, Bonn, Germany
- * E-mail:
| | - Tracie Pennimpede
- Department of Developmental Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
- * E-mail:
| | | | - Anne-Karolin Ebert
- Department of Urology and Pediatric Urology, University Hospital of Ulm, Germany
| | - Wolfgang Rösch
- Department of Pediatric Urology, St. Hedwig Hospital Barmherzige Brüder, Regensburg, Germany
| | - Raimund Stein
- Department of Urology, Division of Pediatric Urology, University of Mainz, Mainz, Germany
| | - Boris Utsch
- Department of General Pediatrics and Neonatology, Justus Liebig University, Giessen, Germany
| | - Karin Hirsch
- Department of Urology, Division of Paediatric Urology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas M. Boemers
- Department of Pediatric Surgery and Pediatric Urology, Children’s Hospital of Cologne, Cologne, Germany
| | | | - Stefanie Heilmann
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Kerstin U. Ludwig
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Ekkehart Jenetzky
- Department of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany
- Department of Child and Adolescent Psychiatry and Psychotherapy, Johannes-Gutenberg University, Mainz, Germany
| | - Nadine Zwink
- Department of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany
| | - Susanne Moebus
- Institute of Medical Informatics, Biometry, and Epidemiology, University Hospital of Essen, University Duisburg-Essen, Essen, Germany
| | - Bernhard G. Herrmann
- Department of Developmental Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Manuel Mattheisen
- Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts, United States of America
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Genomic Mathematics, University of Bonn, Bonn, Germany
| | - Markus M. Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Michael Ludwig
- Department of Clinical Chemistry and Clinical Pharmacology, University of Bonn, Bonn, Germany
| | - Heiko Reutter
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Neonatology, Children's Hospital, University of Bonn, Bonn, Germany
- * E-mail:
| |
Collapse
|
149
|
Engrailed 1 mediates correct formation of limb innervation through two distinct mechanisms. PLoS One 2015; 10:e0118505. [PMID: 25710467 PMCID: PMC4340014 DOI: 10.1371/journal.pone.0118505] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 01/19/2015] [Indexed: 12/24/2022] Open
Abstract
Engrailed-1 (En1) is expressed in the ventral ectoderm of the developing limb where it plays an instructive role in the dorsal-ventral patterning of the forelimb. Besides its well-described role as a transcription factor in regulating gene expression through its DNA-binding domain, En1 may also be secreted to form an extracellular gradient, and directly impact on the formation of the retinotectal map. We show here that absence of En1 causes mispatterning of the forelimb and thus defects in the dorsal-ventral pathfinding choice of motor axons in vivo. In addition, En1 but not En2 also has a direct and specific repulsive effect on motor axons of the lateral aspect of the lateral motor column (LMC) but not on medial LMC projections. Moreover, an ectopic dorsal source of En1 pushes lateral LMC axons to the ventral limb in vivo. Thus, En1 controls the establishment of limb innervation through two distinct molecular mechanisms.
Collapse
|
150
|
Butler SJ, Bronner ME. From classical to current: analyzing peripheral nervous system and spinal cord lineage and fate. Dev Biol 2015; 398:135-46. [PMID: 25446276 PMCID: PMC4845735 DOI: 10.1016/j.ydbio.2014.09.033] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 09/22/2014] [Accepted: 09/25/2014] [Indexed: 01/13/2023]
Abstract
During vertebrate development, the central (CNS) and peripheral nervous systems (PNS) arise from the neural plate. Cells at the margin of the neural plate give rise to neural crest cells, which migrate extensively throughout the embryo, contributing to the majority of neurons and all of the glia of the PNS. The rest of the neural plate invaginates to form the neural tube, which expands to form the brain and spinal cord. The emergence of molecular cloning techniques and identification of fluorophores like Green Fluorescent Protein (GFP), together with transgenic and electroporation technologies, have made it possible to easily visualize the cellular and molecular events in play during nervous system formation. These lineage-tracing techniques have precisely demonstrated the migratory pathways followed by neural crest cells and increased knowledge about their differentiation into PNS derivatives. Similarly, in the spinal cord, lineage-tracing techniques have led to a greater understanding of the regional organization of multiple classes of neural progenitor and post-mitotic neurons along the different axes of the spinal cord and how these distinct classes of neurons assemble into the specific neural circuits required to realize their various functions. Here, we review how both classical and modern lineage and marker analyses have expanded our knowledge of early peripheral nervous system and spinal cord development.
Collapse
Affiliation(s)
- Samantha J Butler
- Department of Neurobiology, TLSB 3129, 610 Charles E Young Drive East, University of California, Los Angeles, Los Angeles, CA 90095-7239, USA; Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| | - Marianne E Bronner
- Department of Neurobiology, TLSB 3129, 610 Charles E Young Drive East, University of California, Los Angeles, Los Angeles, CA 90095-7239, USA; Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|