1
|
Fritzsch B, Glover JC. Gene networks and the evolution of olfactory organs, eyes, hair cells and motoneurons: a view encompassing lancelets, tunicates and vertebrates. Front Cell Dev Biol 2024; 12:1340157. [PMID: 38533086 PMCID: PMC10963430 DOI: 10.3389/fcell.2024.1340157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/26/2024] [Indexed: 03/28/2024] Open
Abstract
Key developmental pathways and gene networks underlie the formation of sensory cell types and structures involved in chemosensation, vision and mechanosensation, and of the efferents these sensory inputs can activate. We describe similarities and differences in these pathways and gene networks in selected species of the three main chordate groups, lancelets, tunicates, and vertebrates, leading to divergent development of olfactory receptors, eyes, hair cells and motoneurons. The lack of appropriately posited expression of certain transcription factors in lancelets and tunicates prevents them from developing vertebrate-like olfactory receptors and eyes, although they generate alternative structures for chemosensation and vision. Lancelets and tunicates lack mechanosensory cells associated with the sensation of acoustic stimuli, but have gravisensitive organs and ciliated epidermal sensory cells that may (and in some cases clearly do) provide mechanosensation and thus the capacity to respond to movement relative to surrounding water. Although functionally analogous to the vertebrate vestibular apparatus and lateral line, homology is questionable due to differences in the expression of the key transcription factors Neurog and Atoh1/7, on which development of vertebrate hair cells depends. The vertebrate hair cell-bearing inner ear and lateral line thus likely represent major evolutionary advances specific to vertebrates. Motoneurons develop in vertebrates under the control of the ventral signaling molecule hedgehog/sonic hedgehog (Hh,Shh), against an opposing inhibitory effect mediated by dorsal signaling molecules. Many elements of Shh-signaling and downstream genes involved in specifying and differentiating motoneurons are also exhibited by lancelets and tunicates, but the repertoire of MNs in vertebrates is broader, indicating greater diversity in motoneuron differentiation programs.
Collapse
Affiliation(s)
- Bernd Fritzsch
- Department of Biological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Joel C. Glover
- Sars International Centre for Marine Molecular Biology, University of Bergen, Bergen, Norway
- Laboratory of Neural Development and Optical Recording (NDEVOR), Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
2
|
Kokkorakis N, Douka K, Nalmpanti A, Politis PK, Zagoraiou L, Matsas R, Gaitanou M. Mirk/Dyrk1B controls ventral spinal cord development via Shh pathway. Cell Mol Life Sci 2024; 81:70. [PMID: 38294527 PMCID: PMC10830675 DOI: 10.1007/s00018-023-05097-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 12/14/2023] [Accepted: 12/17/2023] [Indexed: 02/01/2024]
Abstract
Cross-talk between Mirk/Dyrk1B kinase and Sonic hedgehog (Shh)/Gli pathway affects physiology and pathology. Here, we reveal a novel role for Dyrk1B in regulating ventral progenitor and neuron subtypes in the embryonic chick spinal cord (SC) via the Shh pathway. Using in ovo gain-and-loss-of-function approaches at E2, we report that Dyrk1B affects the proliferation and differentiation of neuronal progenitors at E4 and impacts on apoptosis specifically in the motor neuron (MN) domain. Especially, Dyrk1B overexpression decreases the numbers of ventral progenitors, MNs, and V2a interneurons, while the pharmacological inhibition of endogenous Dyrk1B kinase activity by AZ191 administration increases the numbers of ventral progenitors and MNs. Mechanistically, Dyrk1B overexpression suppresses Shh, Gli2 and Gli3 mRNA levels, while conversely, Shh, Gli2 and Gli3 transcription is increased in the presence of Dyrk1B inhibitor AZ191 or Smoothened agonist SAG. Most importantly, in phenotype rescue experiments, SAG restores the Dyrk1B-mediated dysregulation of ventral progenitors. Further at E6, Dyrk1B affects selectively the medial lateral motor neuron column (LMCm), consistent with the expression of Shh in this region. Collectively, these observations reveal a novel regulatory function of Dyrk1B kinase in suppressing the Shh/Gli pathway and thus affecting ventral subtypes in the developing spinal cord. These data render Dyrk1B a possible therapeutic target for motor neuron diseases.
Collapse
Affiliation(s)
- N Kokkorakis
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Hellenic Pasteur Institute, Athens, Greece
- Division of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - K Douka
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Hellenic Pasteur Institute, Athens, Greece
| | - A Nalmpanti
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Hellenic Pasteur Institute, Athens, Greece
- Athens International Master's Programme in Neurosciences, Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - P K Politis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- School of Medicine, European University Cyprus, Nicosia, Cyprus
| | - L Zagoraiou
- School of Medicine, European University Cyprus, Nicosia, Cyprus
| | - R Matsas
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Hellenic Pasteur Institute, Athens, Greece
| | - M Gaitanou
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Hellenic Pasteur Institute, Athens, Greece.
| |
Collapse
|
3
|
Xie Y, Harwell CC, Garcia ADR. Astrocyte Development in the Rodent. ADVANCES IN NEUROBIOLOGY 2024; 39:51-67. [PMID: 39190071 DOI: 10.1007/978-3-031-64839-7_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Astrocytes have gained increasing recognition as key elements of a broad array of nervous system functions. These include essential roles in synapse formation and elimination, synaptic modulation, maintenance of the blood-brain barrier, energetic support, and neural repair after injury or disease of the nervous system. Nevertheless, our understanding of mechanisms underlying astrocyte development and maturation remains far behind that of neurons and oligodendrocytes. Early efforts to understand astrocyte development focused primarily on their specification from embryonic progenitors and the molecular mechanisms driving the switch from neuron to glial production. Considerably, less is known about postnatal stages of astrocyte development, the period during which they are predominantly generated and mature. Notably, this period is coincident with synapse formation and the emergence of nascent neural circuits. Thus, a greater understanding of astrocyte development is likely to shed new light on the formation and maturation of synapses and circuits. Here, we highlight key foundational principles of embryonic and postnatal astrocyte development, focusing largely on what is known from rodent studies.
Collapse
Affiliation(s)
- Yajun Xie
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, USA
| | - Corey C Harwell
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA, USA
| | - A Denise R Garcia
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
4
|
Kim YI, O'Rourke R, Sagerström CG. scMultiome analysis identifies embryonic hindbrain progenitors with mixed rhombomere identities. eLife 2023; 12:e87772. [PMID: 37947350 PMCID: PMC10662952 DOI: 10.7554/elife.87772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 11/09/2023] [Indexed: 11/12/2023] Open
Abstract
Rhombomeres serve to position neural progenitors in the embryonic hindbrain, thereby ensuring appropriate neural circuit formation, but the molecular identities of individual rhombomeres and the mechanism whereby they form has not been fully established. Here, we apply scMultiome analysis in zebrafish to molecularly resolve all rhombomeres for the first time. We find that rhombomeres become molecularly distinct between 10hpf (end of gastrulation) and 13hpf (early segmentation). While the embryonic hindbrain transiently contains alternating odd- versus even-type rhombomeres, our scMultiome analyses do not detect extensive odd versus even molecular characteristics in the early hindbrain. Instead, we find that each rhombomere displays a unique gene expression and chromatin profile. Prior to the appearance of distinct rhombomeres, we detect three hindbrain progenitor clusters (PHPDs) that correlate with the earliest visually observed segments in the hindbrain primordium that represent prospective rhombomere r2/r3 (possibly including r1), r4, and r5/r6, respectively. We further find that the PHPDs form in response to Fgf and RA morphogens and that individual PHPD cells co-express markers of multiple mature rhombomeres. We propose that the PHPDs contain mixed-identity progenitors and that their subdivision into individual rhombomeres requires the resolution of mixed transcription and chromatin states.
Collapse
Affiliation(s)
- Yong-Il Kim
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Medical SchoolAuroraUnited States
| | - Rebecca O'Rourke
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Medical SchoolAuroraUnited States
| | - Charles G Sagerström
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Medical SchoolAuroraUnited States
| |
Collapse
|
5
|
Wang W, Wang T, Gao Y, Liang G, Pu Y, Zhang J. Model of neural development by differentiating human induced pluripotent stem cells into neural progenitor cells to study the neurodevelopmental toxicity of lead. Food Chem Toxicol 2023; 179:113947. [PMID: 37467947 DOI: 10.1016/j.fct.2023.113947] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/04/2023] [Accepted: 07/12/2023] [Indexed: 07/21/2023]
Abstract
Lead (Pb) exposure causes immeasurable damage to multiple human systems, particularly the central nervous system (CNS). In this study, human induced pluripotent stem cells (hiPSCs) were differentiated into neural progenitor cells (NPCs) to investigate the neurotoxic effects of Pb. The hiPSCs were treated with 0, 0.5, 1.0, 2.5, 5.0 and 10.0 μmol/L Pb for 7 days, whereas embryoid bodies (EBs) and NPCs were treated with 0, 0.1, 0.5, and 1.0 μmol/L Pb for 7 days. Pb exposure disrupted the cell cycle and caused apoptosis in hiPSCs, EBs, and NPCs. Besides, Pb inhibited the differentiation of NPCs and EBs. Whole exome sequencing revealed 2509, 2413, and 1984 single nucleotide variants (SNVs) caused by Pb in hiPSCs, EBs, and NPCs, respectively. The common mutation sites in the exon region were mostly nonsynonymous mutations. We identified 18, 19, and 18 common deleterious mutations in hiPSCs, EBs, and NPCs, respectively. Additionally, Online Mendelian Inheritance in Man database analysis revealed 30, 20, and 13 genes related to CNS disorders in hiPSCs, EBs, and NPCs, respectively. Our findings suggest that this in vitro model may supplement animal models and be applied to the study of neurodevelopmental toxicity in the future.
Collapse
Affiliation(s)
- Wei Wang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Tong Wang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Yu Gao
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Geyu Liang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Juan Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China, School of Public Health, Southeast University, Nanjing, 210009, China; Jiangsu Institute for Sports and Health (JISH), Nanjing, 211100, China.
| |
Collapse
|
6
|
Perrault EN, Shireman JM, Ali ES, Lin P, Preddy I, Park C, Budhiraja S, Baisiwala S, Dixit K, James CD, Heiland DH, Ben-Sahra I, Pott S, Basu A, Miska J, Ahmed AU. Ribonucleotide reductase regulatory subunit M2 drives glioblastoma TMZ resistance through modulation of dNTP production. SCIENCE ADVANCES 2023; 9:eade7236. [PMID: 37196077 PMCID: PMC10191446 DOI: 10.1126/sciadv.ade7236] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 04/13/2023] [Indexed: 05/19/2023]
Abstract
During therapy, adaptations driven by cellular plasticity are partly responsible for driving the inevitable recurrence of glioblastoma (GBM). To investigate plasticity-induced adaptation during standard-of-care chemotherapy temozolomide (TMZ), we performed in vivo single-cell RNA sequencing in patient-derived xenograft (PDX) tumors of GBM before, during, and after therapy. Comparing single-cell transcriptomic patterns identified distinct cellular populations present during TMZ therapy. Of interest was the increased expression of ribonucleotide reductase regulatory subunit M2 (RRM2), which we found to regulate dGTP and dCTP production vital for DNA damage response during TMZ therapy. Furthermore, multidimensional modeling of spatially resolved transcriptomic and metabolomic analysis in patients' tissues revealed strong correlations between RRM2 and dGTP. This supports our data that RRM2 regulates the demand for specific dNTPs during therapy. In addition, treatment with the RRM2 inhibitor 3-AP (Triapine) enhances the efficacy of TMZ therapy in PDX models. We present a previously unidentified understanding of chemoresistance through critical RRM2-mediated nucleotide production.
Collapse
Affiliation(s)
- Ella N. Perrault
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jack M. Shireman
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Eunus S. Ali
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Peiyu Lin
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Isabelle Preddy
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Cheol Park
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Shreya Budhiraja
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Shivani Baisiwala
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Karan Dixit
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - C. David James
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Dieter H Heiland
- Microenvironment and Immunology Research Laboratory, Medical-Center, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Medical-Center, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), partner site Freiburg, Freiburg, Germany
| | - Issam Ben-Sahra
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Sebastian Pott
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Anindita Basu
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Jason Miska
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Atique U. Ahmed
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
7
|
Marques GS, Teles-Reis J, Konstantinides N, Brito PH, Homem CCF. Asynchronous transcription and translation of neurotransmitter-related genes characterize the initial stages of neuronal maturation in Drosophila. PLoS Biol 2023; 21:e3002115. [PMID: 37205703 PMCID: PMC10234549 DOI: 10.1371/journal.pbio.3002115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 06/01/2023] [Accepted: 04/06/2023] [Indexed: 05/21/2023] Open
Abstract
Neuron specification and maturation are essential for proper central nervous system development. However, the precise mechanisms that govern neuronal maturation, essential to shape and maintain neuronal circuitry, remain poorly understood. Here, we analyse early-born secondary neurons in the Drosophila larval brain, revealing that the early maturation of secondary neurons goes through 3 consecutive phases: (1) Immediately after birth, neurons express pan-neuronal markers but do not transcribe terminal differentiation genes; (2) Transcription of terminal differentiation genes, such as neurotransmitter-related genes VGlut, ChAT, or Gad1, starts shortly after neuron birth, but these transcripts are, however, not translated; (3) Translation of neurotransmitter-related genes only begins several hours later in mid-pupa stages in a coordinated manner with animal developmental stage, albeit in an ecdysone-independent manner. These results support a model where temporal regulation of transcription and translation of neurotransmitter-related genes is an important mechanism to coordinate neuron maturation with brain development.
Collapse
Affiliation(s)
- Graça S. Marques
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa; Lisboa, Portugal
| | - José Teles-Reis
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa; Lisboa, Portugal
| | | | - Patrícia H. Brito
- Applied Molecular Biosciences Unit-UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal
| | - Catarina C. F. Homem
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa; Lisboa, Portugal
| |
Collapse
|
8
|
Kim YI, O’Rourke R, Sagerström CG. scMultiome analysis identifies embryonic hindbrain progenitors with mixed rhombomere identities. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.27.525932. [PMID: 36747868 PMCID: PMC9900950 DOI: 10.1101/2023.01.27.525932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Rhombomeres serve to position neural progenitors in the embryonic hindbrain, thereby ensuring appropriate neural circuit formation, but the molecular identities of individual rhombomeres and the mechanism whereby they form have not been fully established. Here we apply scMultiome analysis in zebrafish to molecularly resolve all rhombomeres for the first time. We find that rhombomeres become molecularly distinct between 10hpf (end of gastrulation) and 13hpf (early segmentation). While the mature hindbrain consists of alternating odd- versus even-type rhombomeres, our scMultiome analyses do not detect extensive odd versus even characteristics in the early hindbrain. Instead, we find that each rhombomere displays a unique gene expression and chromatin profile. Prior to the appearance of distinct rhombomeres, we detect three hindbrain progenitor clusters (PHPDs) that correlate with the earliest visually observed segments in the hindbrain primordium and that represent prospective rhombomere r2/r3 (possibly including r1), r4 and r5/r6, respectively. We further find that the PHPDs form in response to Fgf and RA morphogens and that individual PHPD cells co-express markers of multiple mature rhombomeres. We propose that the PHPDs contain mixed-identity progenitors and that their subdivision into individual mature rhombomeres requires resolution of mixed transcription and chromatin states.
Collapse
Affiliation(s)
| | | | - Charles G. Sagerström
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Medical School, 12801 E. 17th Avenue, Aurora, CO 80045
| |
Collapse
|
9
|
Abu-El-Rub E, Khasawneh RR, Almahasneh FA, Aloud BM, Zegallai HM. The Molecular and Functional Changes of Neural Stem Cells in Alzheimer's Disease: Can They be Reinvigorated to Conduct Neurogenesis. Curr Stem Cell Res Ther 2023; 18:580-594. [PMID: 36045542 DOI: 10.2174/1574888x17666220831105257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/04/2022] [Accepted: 07/19/2022] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is considered one of the most complicated neurodegenerative disorders, and it is associated with progressive memory loss and remarkable neurocognitive dysfunction that negatively impacts the ability to perform daily living activities. AD accounts for an estimated 60-80% of dementia cases. AD's previously known pathological basis is the deposition of amyloid β (Aβ) aggregates and the formation of neurofibrillary tangles by tau hyperphosphorylation in the cell bodies of neurons that are located in the hippocampus, neocortex, and certain other regions of the cerebral hemispheres and limbic system. The lack of neurotransmitter acetylcholine and the activation of oxidative stress cascade may also contribute to the pathogenesis of AD. These pathological events can lead to irreversible loss of neuronal networks and the emergence of memory impairment and cognitive dysfunction that can engender an abnormal change in the personality. AD cannot be cured, and to some extent, the prescribed medications can only manage the symptoms associated with this disease. Several studies have reported that the regenerative abilities of neural stem/progenitor cells (NSCs) remarkably decline in AD, which disturbs the balancing power to control its progression. Exogenous infusion or endogenous activation of NSCs may be the ultimate solution to restore the neuronal networks in the brain of AD patients and regenerate the damaged areas responsible for memory and cognition. In this mini-review, we will touch upon the fate of NSCs in AD and the utilization of neurogenesis using modified NSCs to restore cognitive functions in AD.
Collapse
Affiliation(s)
- Ejlal Abu-El-Rub
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| | - Ramada R Khasawneh
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| | - Fatimah A Almahasneh
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| | - Basma Milad Aloud
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
| | - Hana M Zegallai
- Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, Canada
- DREAM, Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
10
|
Barak M, Fedorova V, Pospisilova V, Raska J, Vochyanova S, Sedmik J, Hribkova H, Klimova H, Vanova T, Bohaciakova D. Human iPSC-Derived Neural Models for Studying Alzheimer's Disease: from Neural Stem Cells to Cerebral Organoids. Stem Cell Rev Rep 2022; 18:792-820. [PMID: 35107767 PMCID: PMC8930932 DOI: 10.1007/s12015-021-10254-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2021] [Indexed: 12/05/2022]
Abstract
During the past two decades, induced pluripotent stem cells (iPSCs) have been widely used to study mechanisms of human neural development, disease modeling, and drug discovery in vitro. Especially in the field of Alzheimer’s disease (AD), where this treatment is lacking, tremendous effort has been put into the investigation of molecular mechanisms behind this disease using induced pluripotent stem cell-based models. Numerous of these studies have found either novel regulatory mechanisms that could be exploited to develop relevant drugs for AD treatment or have already tested small molecules on in vitro cultures, directly demonstrating their effect on amelioration of AD-associated pathology. This review thus summarizes currently used differentiation strategies of induced pluripotent stem cells towards neuronal and glial cell types and cerebral organoids and their utilization in modeling AD and potential drug discovery.
Collapse
Affiliation(s)
- Martin Barak
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Veronika Fedorova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Veronika Pospisilova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Jan Raska
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Simona Vochyanova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Jiri Sedmik
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
- International Clinical Research Center, St. Anne's Faculty Hospital Brno, Brno, Czech Republic
| | - Hana Hribkova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Hana Klimova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Tereza Vanova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
- International Clinical Research Center, St. Anne's Faculty Hospital Brno, Brno, Czech Republic
| | - Dasa Bohaciakova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic.
- International Clinical Research Center, St. Anne's Faculty Hospital Brno, Brno, Czech Republic.
| |
Collapse
|
11
|
Gupta S, Butler SJ. Getting in touch with your senses: Mechanisms specifying sensory interneurons in the dorsal spinal cord. WIREs Mech Dis 2021; 13:e1520. [PMID: 34730293 PMCID: PMC8459260 DOI: 10.1002/wsbm.1520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/10/2021] [Accepted: 01/11/2021] [Indexed: 11/18/2022]
Abstract
The spinal cord is functionally and anatomically divided into ventrally derived motor circuits and dorsally derived somatosensory circuits. Sensory stimuli originating either at the periphery of the body, or internally, are relayed to the dorsal spinal cord where they are processed by distinct classes of sensory dorsal interneurons (dIs). dIs convey sensory information, such as pain, heat or itch, either to the brain, and/or to the motor circuits to initiate the appropriate response. They also regulate the intensity of sensory information and are the major target for the opioid analgesics. While the developmental mechanisms directing ventral and dorsal cell fates have been hypothesized to be similar, more recent research has suggested that dI fates are specified by novel mechanisms. In this review, we will discuss the molecular events that specify dorsal neuronal patterning in the spinal cord, thereby generating diverse dI identities. We will then discuss how this molecular understanding has led to the development of robust stem cell methods to derive multiple spinal cell types, including the dIs, and the implication of these studies for treating spinal cord injuries and neurodegenerative diseases. This article is categorized under: Neurological Diseases > Stem Cells and Development.
Collapse
Affiliation(s)
- Sandeep Gupta
- Department of NeurobiologyUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Samantha J. Butler
- Department of NeurobiologyUniversity of California, Los AngelesLos AngelesCaliforniaUSA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell ResearchUniversity of California, Los AngelesLos AngelesCaliforniaUSA
- Intellectual and Developmental Disabilities Research CenterUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| |
Collapse
|
12
|
From Bipotent Neuromesodermal Progenitors to Neural-Mesodermal Interactions during Embryonic Development. Int J Mol Sci 2021; 22:ijms22179141. [PMID: 34502050 PMCID: PMC8431582 DOI: 10.3390/ijms22179141] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 11/17/2022] Open
Abstract
To ensure the formation of a properly patterned embryo, multiple processes must operate harmoniously at sequential phases of development. This is implemented by mutual interactions between cells and tissues that together regulate the segregation and specification of cells, their growth and morphogenesis. The formation of the spinal cord and paraxial mesoderm derivatives exquisitely illustrate these processes. Following early gastrulation, while the vertebrate body elongates, a population of bipotent neuromesodermal progenitors resident in the posterior region of the embryo generate both neural and mesodermal lineages. At later stages, the somitic mesoderm regulates aspects of neural patterning and differentiation of both central and peripheral neural progenitors. Reciprocally, neural precursors influence the paraxial mesoderm to regulate somite-derived myogenesis and additional processes by distinct mechanisms. Central to this crosstalk is the activity of the axial notochord, which, via sonic hedgehog signaling, plays pivotal roles in neural, skeletal muscle and cartilage ontogeny. Here, we discuss the cellular and molecular basis underlying this complex developmental plan, with a focus on the logic of sonic hedgehog activities in the coordination of the neural-mesodermal axis.
Collapse
|
13
|
Pardieck J, Harb M, Sakiyama-Elbert S. Induction of Ventral Spinal V0 Interneurons from Mouse Embryonic Stem Cells. Stem Cells Dev 2021; 30:816-829. [PMID: 34139881 DOI: 10.1089/scd.2021.0003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The ventral spinal population of V0 interneurons (INs) contributes to the coordinated movements directed by spinal central pattern generators (CPGs), including respiratory circuits and left-right alternation in locomotion. One challenge in studying V0 INs has been the limited number of cells that can be isolated from primary sources for basic research or therapeutic use. However, derivation from a pluripotent source, such as has been done recently for other IN populations, could resolve this issue. However, there is currently no protocol to specifically derive V0 interneurons from pluripotent cell types. To generate an induction protocol, mouse embryonic stem cells (mESCs) were grown in suspension culture and then exposed to retinoic acid (RA) and collected at different time points to measure mRNA expression of the V0 progenitor transcription factor marker, Dbx1, and postmitotic transcription factor marker, Evx1. The cultures were also exposed to the sonic hedgehog signaling pathway agonist purmorphamine (purm) and the Notch signaling pathway inhibitor N-{N-(3,5-difluorophenacetyl-L-alanyl)}-(S)-phenylglycine-t-butyl-ester (DAPT) to determine if either of these pathways contribute to V0 IN induction, specifically the ventral (V0V) subpopulation. From the various parameters tested, the final protocol that generated the greatest percentage of cells expressing V0V IN markers was an 8-day protocol using 4 days of suspension culture to form embryoid bodies followed by addition of 1 μM RA from days 4 to 8, 100 nM purm from days 4 to 6, and 5 μM DAPT from days 6 to 8. This protocol will allow investigators to obtain V0 IN cultures for use in in vitro studies, such as those examining CPG microcircuits, electrophysiological characterization, or even for transplantation studies in injury or disease models.
Collapse
Affiliation(s)
- Jennifer Pardieck
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Manwal Harb
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA
| | | |
Collapse
|
14
|
Katsuyama T, Kadoya M, Shirai M, Sasai N. Sox14 is essential for initiation of neuronal differentiation in the chick spinal cord. Dev Dyn 2021; 251:350-361. [PMID: 34181293 DOI: 10.1002/dvdy.392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The neural tube comprises several different types of progenitors and postmitotic neurons that co-ordinately act with each other to play integrated functions. Its development consists of two phases: proliferation of progenitor cells and differentiation into postmitotic neurons. How progenitor cells differentiate into each corresponding neuron is an important question for understanding the mechanisms of neuronal development. RESULTS Here we introduce one of the Sox transcription factors, Sox14, which plays an essential role in the promotion of neuronal differentiation. Sox14 belongs to the SoxB2 subclass and its expression starts in the progenitor regions before neuronal differentiation is initiated at the trunk level of the neural tube. After neuronal differentiation is initiated, Sox14 expression gradually becomes confined to the V2a region of the neural tube, where Chx10 is co-expressed. Overexpression of Sox14 restricts progenitor cell proliferation. Conversely, the blockade of Sox14 expression by the RNAi strategy inhibits V2a neuron differentiation and causes expansion of the progenitor domain. We further found that Sox14 acted as a transcriptional activator. CONCLUSIONS Sox14 acts as a modulator of cell proliferation and is essential for initiation of neuronal differentiation in the chick neural tube.
Collapse
Affiliation(s)
- Taiki Katsuyama
- Developmental Biomedical Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Minori Kadoya
- Developmental Biomedical Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Manabu Shirai
- Omics Research Center (ORC), National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Noriaki Sasai
- Developmental Biomedical Science, Nara Institute of Science and Technology, Ikoma, Japan
| |
Collapse
|
15
|
Ho EK, Stearns T. Hedgehog signaling and the primary cilium: implications for spatial and temporal constraints on signaling. Development 2021; 148:dev195552. [PMID: 33914866 PMCID: PMC8126410 DOI: 10.1242/dev.195552] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The mechanisms of vertebrate Hedgehog signaling are linked to the biology of the primary cilium, an antenna-like organelle that projects from the surface of most vertebrate cell types. Although the advantages of restricting signal transduction to cilia are often noted, the constraints imposed are less frequently considered, and yet they are central to how Hedgehog signaling operates in developing tissues. In this Review, we synthesize current understanding of Hedgehog signal transduction, ligand secretion and transport, and cilia dynamics to explore the temporal and spatial constraints imposed by the primary cilium on Hedgehog signaling in vivo.
Collapse
Affiliation(s)
- Emily K. Ho
- Department of Developmental Biology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Tim Stearns
- Department of Biology, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
16
|
Curry RN, Glasgow SM. The Role of Neurodevelopmental Pathways in Brain Tumors. Front Cell Dev Biol 2021; 9:659055. [PMID: 34012965 PMCID: PMC8127784 DOI: 10.3389/fcell.2021.659055] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
Disruptions to developmental cell signaling pathways and transcriptional cascades have been implicated in tumor initiation, maintenance and progression. Resurgence of aberrant neurodevelopmental programs in the context of brain tumors highlights the numerous parallels that exist between developmental and oncologic mechanisms. A deeper understanding of how dysregulated developmental factors contribute to brain tumor oncogenesis and disease progression will help to identify potential therapeutic targets for these malignancies. In this review, we summarize the current literature concerning developmental signaling cascades and neurodevelopmentally-regulated transcriptional programs. We also examine their respective contributions towards tumor initiation, maintenance, and progression in both pediatric and adult brain tumors and highlight relevant differentiation therapies and putative candidates for prospective treatments.
Collapse
Affiliation(s)
- Rachel N. Curry
- Department of Neuroscience, Baylor College of Medicine, Center for Cell and Gene Therapy, Houston, TX, United States
- Integrative Molecular and Biomedical Sciences, Graduate School of Biomedical Sciences, Baylor College of Medicine, Houston, TX, United States
| | - Stacey M. Glasgow
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, San Diego, CA, United States
- Neurosciences Graduate Program, University of California, San Diego, San Diego, CA, United States
- Biomedical Sciences Graduate Program, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
17
|
May EA, Sroka TJ, Mick DU. Phosphorylation and Ubiquitylation Regulate Protein Trafficking, Signaling, and the Biogenesis of Primary Cilia. Front Cell Dev Biol 2021; 9:664279. [PMID: 33912570 PMCID: PMC8075051 DOI: 10.3389/fcell.2021.664279] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/09/2021] [Indexed: 12/30/2022] Open
Abstract
The primary cilium is a solitary, microtubule-based membrane protrusion extending from the surface of quiescent cells that senses the cellular environment and triggers specific cellular responses. The functions of primary cilia require not only numerous different components but also their regulated interplay. The cilium performs highly dynamic processes, such as cell cycle-dependent assembly and disassembly as well as delivery, modification, and removal of signaling components to perceive and process external signals. On a molecular level, these processes often rely on a stringent control of key modulatory proteins, of which the activity, localization, and stability are regulated by post-translational modifications (PTMs). While an increasing number of PTMs on ciliary components are being revealed, our knowledge on the identity of the modifying enzymes and their modulation is still limited. Here, we highlight recent findings on cilia-specific phosphorylation and ubiquitylation events. Shedding new light onto the molecular mechanisms that regulate the sensitive equilibrium required to maintain and remodel primary cilia functions, we discuss their implications for cilia biogenesis, protein trafficking, and cilia signaling processes.
Collapse
Affiliation(s)
- Elena A May
- Center of Human and Molecular Biology (ZHMB), Saarland University School of Medicine, Homburg, Germany.,Center for Molecular Signaling (PZMS), Department of Medical Biochemistry and Molecular Biology, Saarland University School of Medicine, Homburg, Germany
| | - Tommy J Sroka
- Center of Human and Molecular Biology (ZHMB), Saarland University School of Medicine, Homburg, Germany.,Center for Molecular Signaling (PZMS), Department of Medical Biochemistry and Molecular Biology, Saarland University School of Medicine, Homburg, Germany
| | - David U Mick
- Center of Human and Molecular Biology (ZHMB), Saarland University School of Medicine, Homburg, Germany.,Center for Molecular Signaling (PZMS), Department of Medical Biochemistry and Molecular Biology, Saarland University School of Medicine, Homburg, Germany
| |
Collapse
|
18
|
Mouilleau V, Vaslin C, Robert R, Gribaudo S, Nicolas N, Jarrige M, Terray A, Lesueur L, Mathis MW, Croft G, Daynac M, Rouiller-Fabre V, Wichterle H, Ribes V, Martinat C, Nedelec S. Dynamic extrinsic pacing of the HOX clock in human axial progenitors controls motor neuron subtype specification. Development 2021; 148:148/6/dev194514. [PMID: 33782043 DOI: 10.1242/dev.194514] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 02/16/2021] [Indexed: 12/17/2022]
Abstract
Rostro-caudal patterning of vertebrates depends on the temporally progressive activation of HOX genes within axial stem cells that fuel axial embryo elongation. Whether the pace of sequential activation of HOX genes, the 'HOX clock', is controlled by intrinsic chromatin-based timing mechanisms or by temporal changes in extrinsic cues remains unclear. Here, we studied HOX clock pacing in human pluripotent stem cell-derived axial progenitors differentiating into diverse spinal cord motor neuron subtypes. We show that the progressive activation of caudal HOX genes is controlled by a dynamic increase in FGF signaling. Blocking the FGF pathway stalled induction of HOX genes, while a precocious increase of FGF, alone or with GDF11 ligand, accelerated the HOX clock. Cells differentiated under accelerated HOX induction generated appropriate posterior motor neuron subtypes found along the human embryonic spinal cord. The pacing of the HOX clock is thus dynamically regulated by exposure to secreted cues. Its manipulation by extrinsic factors provides synchronized access to multiple human neuronal subtypes of distinct rostro-caudal identities for basic and translational applications.This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Vincent Mouilleau
- Institut du Fer à Moulin, 75005 Paris, France.,Inserm, UMR-S 1270, 75005 Paris, France.,Sorbonne Université, Science and Engineering Faculty, 75005 Paris, France.,I-STEM, UMR 861, Inserm, UEPS, 91100 Corbeil-Essonnes, France
| | - Célia Vaslin
- Institut du Fer à Moulin, 75005 Paris, France.,Inserm, UMR-S 1270, 75005 Paris, France.,Sorbonne Université, Science and Engineering Faculty, 75005 Paris, France
| | - Rémi Robert
- Institut du Fer à Moulin, 75005 Paris, France.,Inserm, UMR-S 1270, 75005 Paris, France.,Sorbonne Université, Science and Engineering Faculty, 75005 Paris, France
| | - Simona Gribaudo
- Institut du Fer à Moulin, 75005 Paris, France.,Inserm, UMR-S 1270, 75005 Paris, France.,Sorbonne Université, Science and Engineering Faculty, 75005 Paris, France
| | - Nour Nicolas
- Laboratory of Development of the Gonads, Unit of Genetic Stability, Stem Cells and Radiation, UMR 967, INSERM, CEA/DSV/iRCM/SCSR, Université Paris Diderot, Sorbonne Paris Cité, Université Paris-Sud, Université Paris-Saclay, Fontenay aux Roses F-92265, France
| | - Margot Jarrige
- I-STEM, UMR 861, Inserm, UEPS, 91100 Corbeil-Essonnes, France
| | - Angélique Terray
- Institut du Fer à Moulin, 75005 Paris, France.,Inserm, UMR-S 1270, 75005 Paris, France.,Sorbonne Université, Science and Engineering Faculty, 75005 Paris, France
| | - Léa Lesueur
- I-STEM, UMR 861, Inserm, UEPS, 91100 Corbeil-Essonnes, France
| | - Mackenzie W Mathis
- Departments of Pathology and Cell Biology, Neuroscience, and Neurology, Center for Motor Neuron Biology and Disease, Columbia Stem Cell Initiative, Columbia University Medical Center, New York, NY 10032, USA
| | - Gist Croft
- Departments of Pathology and Cell Biology, Neuroscience, and Neurology, Center for Motor Neuron Biology and Disease, Columbia Stem Cell Initiative, Columbia University Medical Center, New York, NY 10032, USA
| | - Mathieu Daynac
- Institut du Fer à Moulin, 75005 Paris, France.,Inserm, UMR-S 1270, 75005 Paris, France.,Sorbonne Université, Science and Engineering Faculty, 75005 Paris, France
| | - Virginie Rouiller-Fabre
- Laboratory of Development of the Gonads, Unit of Genetic Stability, Stem Cells and Radiation, UMR 967, INSERM, CEA/DSV/iRCM/SCSR, Université Paris Diderot, Sorbonne Paris Cité, Université Paris-Sud, Université Paris-Saclay, Fontenay aux Roses F-92265, France
| | - Hynek Wichterle
- Departments of Pathology and Cell Biology, Neuroscience, and Neurology, Center for Motor Neuron Biology and Disease, Columbia Stem Cell Initiative, Columbia University Medical Center, New York, NY 10032, USA
| | - Vanessa Ribes
- Université de Paris, CNRS, Institut Jacques Monod, 15 rue Hélène Brion, 75013 Paris, France
| | - Cécile Martinat
- I-STEM, UMR 861, Inserm, UEPS, 91100 Corbeil-Essonnes, France
| | - Stéphane Nedelec
- Institut du Fer à Moulin, 75005 Paris, France .,Inserm, UMR-S 1270, 75005 Paris, France.,Sorbonne Université, Science and Engineering Faculty, 75005 Paris, France
| |
Collapse
|
19
|
Jacobs CT, Huang P. Complex crosstalk of Notch and Hedgehog signalling during the development of the central nervous system. Cell Mol Life Sci 2021; 78:635-644. [PMID: 32880661 PMCID: PMC11072263 DOI: 10.1007/s00018-020-03627-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/23/2020] [Accepted: 08/20/2020] [Indexed: 01/20/2023]
Abstract
The development of the vertebrate central nervous system (CNS) is tightly regulated by many highly conserved cell signalling pathways. These pathways ensure that differentiation and migration events occur in a specific and spatiotemporally restricted manner. Two of these pathways, Notch and Hedgehog (Hh) signalling, have been shown to form a complex web of interaction throughout different stages of CNS development. Strikingly, some processes employ Notch signalling to regulate Hh response, while others utilise Hh signalling to modulate Notch response. Notch signalling functions upstream of Hh response through controlling the trafficking of integral pathway components as well as through modulating protein levels and transcription of downstream transcriptional factors. In contrast, Hh signalling regulates Notch response by either indirectly controlling expression of key Notch ligands and regulatory proteins or directly through transcriptional control of canonical Notch target genes. Here, we review these interactions and demonstrate the level of interconnectivity between the pathways, highlighting context-dependent modes of crosstalk. Since many other developmental signalling pathways are active in these tissues, it is likely that the interplay between Notch and Hh signalling is not only an example of signalling crosstalk but also functions as a component of a wider, multi-pathway signalling network.
Collapse
Affiliation(s)
- Craig T Jacobs
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada
| | - Peng Huang
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
20
|
Li X, Li Y, Li S, Li H, Yang C, Lin J. The role of Shh signalling pathway in central nervous system development and related diseases. Cell Biochem Funct 2020; 39:180-189. [PMID: 32840890 DOI: 10.1002/cbf.3582] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/07/2020] [Accepted: 08/01/2020] [Indexed: 12/15/2022]
Abstract
Sonic hedgehog (Shh) plays important roles in developmental of vertebrate animal central nervous system (CNS), and Gli is its downstream signal molecule. Shh signalling is essential for pattern formation, cell-fate specification, axon guidance, proliferation, survival and differentiation of neurons in CNS development. The abnormal signalling pathway of Shh leads to the occurrence of many nervous system diseases. The mechanism of Shh signalling is complex and remains incompletely understood. Nevertheless, studies have revealed that Shh signalling pathway is classified into canonical and non-canonical pathways. Here we review the role of the Shh signalling pathway and its impact in CNS development and related diseases. Specifically, we discuss the role of Shh in the spinal cord and brain development, cell differentiation and proliferation in CNS and related diseases such as brain tumour, Parkinson's diseases, epilepsy, autism, depression and traumatic brain injury. We also highlight future directions of research that could help to clarify the mechanisms and consequences of Shh signalling in the process of CNS development and related diseases. SIGNIFICANCE OF THE STUDY: This review summarized the role of Shh signalling pathway in CNS development and related diseases such as brain tumour, Parkinson's diseases, epilepsy, autism, depression and traumatic brain injury. It also presented the author's opinions on the future research direction of Shh signalling pathway.
Collapse
Affiliation(s)
- Xiaoying Li
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Yunxiao Li
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Shuanqing Li
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Han Li
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Ciqing Yang
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Juntang Lin
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
21
|
Wullimann MF, Umeasalugo KE. Sonic hedgehog expression in zebrafish forebrain identifies the teleostean pallidal signaling center and shows preglomerular complex and posterior tubercular dopamine cells to arise from shh cells. J Comp Neurol 2019; 528:1321-1348. [PMID: 31760659 DOI: 10.1002/cne.24825] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/15/2019] [Accepted: 11/16/2019] [Indexed: 12/16/2022]
Abstract
Ventralization, a major patterning process in the developing vertebrate neural tube (central nervous system, CNS), depends on Sonic hedgehog (SHH) as a main signaling morphogen. We studied the CNS of late larval and young adult zebrafish in a transgenic shh-GFP line revealing increased neuroanatomical detail due to the progressed differentiation state compared to earlier stages. Some major findings emerge from the present study. (a) shh -GFP is still expressed along the adult zebrafish CNS neuraxis in most locations seen in larvae. (b) We newly identify a ventroposterior shh pallidal domain representing the basal telencephalic signaling center important for basal ganglia development known in other vertebrates (i.e., the anterior entopeduncular area-basal medial ganglionic eminence of mammals). (c) We further show late-emerging shh-GFP positive radial glia cells in the medial zone of the dorsal telencephalon (i.e., the teleostan pallial amygdala). (d) Immunostains for tyrosine hydroxylase demonstrate that there is selective colocalization in adult dopamine cells with shh-GFP in the posterior tuberculum, including in projection cells to striatum, which represents a striking parallel to amniote mesodiencephalic dopamine cell origin from shh expressing floor plate cells. (e) There is no colocalization of shh and islet1 as shown by respective shh-GFP and islet1-GFP lines. (f) The only radially far migrated shh-GFP cells are located in the preglomerular area. (g) There are no adult cerebellar and tectal shh-GFP cells confirming their exclusive role during early development as previously reported by our laboratory.
Collapse
Affiliation(s)
- Mario F Wullimann
- Department Biology II, Division of Neurobiology, Ludwig-Maximilians-Universität München (LMU Munich), Munich, Germany
| | - Kosisochukwu E Umeasalugo
- Department Biology II, Division of Neurobiology, Ludwig-Maximilians-Universität München (LMU Munich), Munich, Germany
| |
Collapse
|
22
|
George S, Hamblin MR, Abrahamse H. Differentiation of Mesenchymal Stem Cells to Neuroglia: in the Context of Cell Signalling. Stem Cell Rev Rep 2019; 15:814-826. [PMID: 31515658 PMCID: PMC6925073 DOI: 10.1007/s12015-019-09917-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The promise of engineering specific cell types from stem cells and rebuilding damaged or diseased tissues has fascinated stem cell researchers and clinicians over last few decades. Mesenchymal Stem Cells (MSCs) have the potential to differentiate into non-mesodermal cells, particularly neural-lineage, consisting of neurons and glia. These multipotent adult stem cells can be used for implementing clinical trials in neural repair. Ongoing research identifies several molecular mechanisms involved in the speciation of neuroglia, which are tightly regulated and interconnected by various components of cell signalling machinery. Growing MSCs with multiple inducers in culture media will initiate changes on intricately interlinked cell signalling pathways and processes. Net result of these signal flow on cellular architecture is also dependent on the type of ligands and stem cells investigated in vitro. However, our understanding about this dynamic signalling machinery is limited and confounding, especially with spheroid structures, neurospheres and organoids. Therefore, the results for differentiating neurons and glia in vitro have been inconclusive, so far. Added to this complication, we have no convincing evidence about the electrical conductivity and functionality status generated in differentiating neurons and glia. This review has taken a step forward to tailor the information on differentiating neuroglia with the common methodologies, in practice.
Collapse
Affiliation(s)
- Sajan George
- Laser Research Centre, University of Johannesburg, P.O. Box 17011, Doornfontein, 2028, South Africa
| | - Michael R Hamblin
- Laser Research Centre, University of Johannesburg, P.O. Box 17011, Doornfontein, 2028, South Africa
- Wellman Centre for Photomedicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, 02115, USA
| | - Heidi Abrahamse
- Laser Research Centre, University of Johannesburg, P.O. Box 17011, Doornfontein, 2028, South Africa.
| |
Collapse
|
23
|
Nagarajan B, Harder A, Japp A, Häberlein F, Mingardo E, Kleinert H, Yilmaz Ö, Zoons A, Rau B, Christ A, Kubitscheck U, Eiberger B, Sandhoff R, Eckhardt M, Hartmann D, Odermatt B. CNS myelin protein 36K regulates oligodendrocyte differentiation through Notch. Glia 2019; 68:509-527. [PMID: 31702067 DOI: 10.1002/glia.23732] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 12/12/2022]
Abstract
In contrast to humans and other mammals, zebrafish can successfully regenerate and remyelinate central nervous system (CNS) axons following injury. In addition to common myelin proteins found in mammalian myelin, 36K protein is a major component of teleost fish CNS myelin. Although 36K is one of the most abundant proteins in zebrafish brain, its function remains unknown. Here we investigate the function of 36K using translation-blocking Morpholinos. Morphant larvae showed fewer dorsally migrated oligodendrocyte precursor cells as well as upregulation of Notch ligand. A gamma secretase inhibitor, which prevents activation of Notch, could rescue oligodendrocyte precursor cell numbers in 36K morphants, suggesting that 36K regulates initial myelination through inhibition of Notch signaling. Since 36K like other short chain dehydrogenases might act on lipids, we performed thin layer chromatography and mass spectrometry of lipids and found changes in lipid composition in 36K morphant larvae. Altogether, we suggest that during early development 36K regulates membrane lipid composition, thereby altering the amount of transmembrane Notch ligands and the efficiency of intramembrane gamma secretase processing of Notch and thereby influencing oligodendrocyte precursor cell differentiation and further myelination. Further studies on the role of 36K short chain dehydrogenase in oligodendrocyte precursor cell differentiation during remyelination might open up new strategies for remyelination therapies in human patients.
Collapse
Affiliation(s)
- Bhuvaneswari Nagarajan
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany
| | - Alexander Harder
- Institute of Physical and Theoretical Chemistry, University of Bonn, Bonn, Germany
| | - Anna Japp
- Institute of Neuropathology, University Clinics, University of Bonn, Bonn, Germany
| | - Felix Häberlein
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany.,Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Enrico Mingardo
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany
| | - Henning Kleinert
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany
| | - Öznur Yilmaz
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany
| | - Angelika Zoons
- Institute of Anatomy, Division of Neuroanatomy, University Clinics, University of Bonn, Bonn, Germany
| | - Birgit Rau
- Institute of Anatomy, Division of Neuroanatomy, University Clinics, University of Bonn, Bonn, Germany
| | - Andrea Christ
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany
| | - Ulrich Kubitscheck
- Institute of Physical and Theoretical Chemistry, University of Bonn, Bonn, Germany
| | - Britta Eiberger
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany
| | - Roger Sandhoff
- Lipid Pathobiochemistry Group, German Cancer Research Centre, Heidelberg, Germany
| | - Matthias Eckhardt
- Institute of Biochemistry and Molecular Biology, University Clinics, University of Bonn, Bonn, Germany
| | - Dieter Hartmann
- Institute of Anatomy, Division of Neuroanatomy, University Clinics, University of Bonn, Bonn, Germany
| | - Benjamin Odermatt
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany.,Institute of Anatomy, Division of Neuroanatomy, University Clinics, University of Bonn, Bonn, Germany
| |
Collapse
|
24
|
Arai Y, Cwetsch AW, Coppola E, Cipriani S, Nishihara H, Kanki H, Saillour Y, Freret-Hodara B, Dutriaux A, Okada N, Okano H, Dehay C, Nardelli J, Gressens P, Shimogori T, D’Onofrio G, Pierani A. Evolutionary Gain of Dbx1 Expression Drives Subplate Identity in the Cerebral Cortex. Cell Rep 2019; 29:645-658.e5. [DOI: 10.1016/j.celrep.2019.09.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/12/2019] [Accepted: 09/04/2019] [Indexed: 10/25/2022] Open
|
25
|
Jacobs CT, Huang P. Notch signalling maintains Hedgehog responsiveness via a Gli-dependent mechanism during spinal cord patterning in zebrafish. eLife 2019; 8:49252. [PMID: 31453809 PMCID: PMC6733594 DOI: 10.7554/elife.49252] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 08/19/2019] [Indexed: 01/11/2023] Open
Abstract
Spinal cord patterning is orchestrated by multiple cell signalling pathways. Neural progenitors are maintained by Notch signalling, whereas ventral neural fates are specified by Hedgehog (Hh) signalling. However, how dynamic interactions between Notch and Hh signalling drive the precise pattern formation is still unknown. We applied the PHRESH (PHotoconvertible REporter of Signalling History) technique to analyse cell signalling dynamics in vivo during zebrafish spinal cord development. This approach reveals that Notch and Hh signalling display similar spatiotemporal kinetics throughout spinal cord patterning. Notch signalling functions upstream to control Hh response of neural progenitor cells. Using gain- and loss-of-function tools, we demonstrate that this regulation occurs not at the level of upstream regulators or primary cilia, but rather at the level of Gli transcription factors. Our results indicate that Notch signalling maintains Hh responsiveness of neural progenitors via a Gli-dependent mechanism in the spinal cord.
Collapse
Affiliation(s)
- Craig T Jacobs
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Peng Huang
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
| |
Collapse
|
26
|
Di Bella DJ, Carcagno AL, Bartolomeu ML, Pardi MB, Löhr H, Siegel N, Hammerschmidt M, Marín-Burgin A, Lanuza GM. Ascl1 Balances Neuronal versus Ependymal Fate in the Spinal Cord Central Canal. Cell Rep 2019; 28:2264-2274.e3. [DOI: 10.1016/j.celrep.2019.07.087] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/07/2019] [Accepted: 07/23/2019] [Indexed: 01/04/2023] Open
|
27
|
Zimmermann JA, Schaffer DV. Engineering biomaterials to control the neural differentiation of stem cells. Brain Res Bull 2019; 150:50-60. [DOI: 10.1016/j.brainresbull.2019.05.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/09/2019] [Accepted: 05/09/2019] [Indexed: 12/13/2022]
|
28
|
Yang C, Li S, Li X, Li H, Li Y, Zhang C, Lin J. Effect of sonic hedgehog on motor neuron positioning in the spinal cord during chicken embryonic development. J Cell Mol Med 2019; 23:3549-3562. [PMID: 30834718 PMCID: PMC6484327 DOI: 10.1111/jcmm.14254] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 02/04/2019] [Accepted: 02/06/2019] [Indexed: 12/02/2022] Open
Abstract
Sonic hedgehog (SHH) is a vertebrate homologue of the secreted Drosophila protein hedgehog and is expressed by the notochord and floor plate in the developing spinal cord. Sonic hedgehog provides signals relevant for positional information, cell proliferation and possibly cell survival, depending on the time and location of expression. Although the role of SHH in providing positional information in the neural tube has been experimentally proven, the underlying mechanism remains unclear. In this study, in ovo electroporation was employed in the chicken spinal cord during chicken embryo development. Electroporation was conducted at stage 17 (E2.5), after electroporation the embryos were continued incubating to stage 28 (E6) for sampling, tissue fixation with 4% paraformaldehyde and frozen sectioning. Sonic hedgehog and related protein expressions were detected by in situ hybridization and fluorescence immunohistochemistry and the results were analysed after microphotography. Our results indicate that the ectopic expression of SHH leads to ventralization in the spinal cord during chicken embryonic development by inducing abnormalities in the structure of the motor column and motor neuron integration. In addition, ectopic SHH expression inhibits the expression of dorsal transcription factors and commissural axon projections. The correct location of SHH expression is vital to the formation of the motor column. Ectopic expression of SHH in the spinal cord not only affects the positioning of motor neurons, but also induces abnormalities in the structure of the motor column. It leads to ventralization in the spinal cord, resulting in the formation of more ventral neurons forming during neuronal formation.
Collapse
Affiliation(s)
- Ciqing Yang
- Xinxiang Key Laboratory of Neural Development, Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang, China
| | - Shuanqing Li
- Xinxiang Key Laboratory of Neural Development, Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang, China
| | - Xiaoying Li
- Xinxiang Key Laboratory of Neural Development, Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Han Li
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang, China.,Advanced Medical and Dental Institute, University Sains Malaysia, Bertam, Penang, Malaysia
| | - Yunxiao Li
- Xinxiang Key Laboratory of Neural Development, Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang, China
| | - Chen Zhang
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Juntang Lin
- Xinxiang Key Laboratory of Neural Development, Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang, China.,College of Biomedical Engineering, Xinxiang Medical University, Xinxiang, China.,Institute of Anatomy I, Jena University Hospital, Jena, Germany
| |
Collapse
|
29
|
Baeuml SW, Biechl D, Wullimann MF. Adult islet1 Expression Outlines Ventralized Derivatives Along Zebrafish Neuraxis. Front Neuroanat 2019; 13:19. [PMID: 30863287 PMCID: PMC6399416 DOI: 10.3389/fnana.2019.00019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 02/01/2019] [Indexed: 01/16/2023] Open
Abstract
Signals issued by dorsal roof and ventral floor plates, respectively, underlie the major patterning process of dorsalization and ventralization during vertebrate neural tube development. The ventrally produced morphogen Sonic hedgehog (SHH) is crucial for vertebrate hindbrain and spinal motor neuron development. One diagnostic gene for motor neurons is the LIM/homeodomain gene islet1, which has additional ventral expression domains extending into mid- and forebrain. In order to corroborate motor neuron development and, in particular, to improve on the identification of poorly documented zebrafish forebrain islet1 populations, we studied adult brains of transgenic islet1-GFP zebrafish (3 and 6 months). This molecular neuroanatomical analysis was supported by immunostaining these brains for tyrosine hydroxylase (TH) or choline acetyltransferase (ChAT), respectively, revealing zebrafish catecholaminergic and cholinergic neurons. The present analysis of ChAT and islet1-GFP label confirms ongoing adult expression of islet1 in zebrafish (basal plate) midbrain, hindbrain, and spinal motor neurons. In contrast, non-motor cholinergic systems lack islet1 expression. Additional presumed basal plate islet1 positive systems are described in detail, aided by TH staining which is particularly informative in the diencephalon. Finally, alar plate zebrafish forebrain systems with islet1 expression are described (i.e., thalamus, preoptic region, and subpallium). We conclude that adult zebrafish continue to express islet1 in the same brain systems as in the larva. Further, pending functional confirmation we hypothesize that the larval expression of sonic hedgehog (shh) might causally underlie much of adult islet1 expression because it explains findings beyond ventrally located systems, for example regarding shh expression in the zona limitans intrathalamica and correlated islet1-GFP expression in the thalamus.
Collapse
Affiliation(s)
- Stephan W Baeuml
- Division of Neurobiology, Department Biology II, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Daniela Biechl
- Division of Neurobiology, Department Biology II, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Mario F Wullimann
- Division of Neurobiology, Department Biology II, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
30
|
Lara-Ramirez R, Pérez-González C, Anselmi C, Patthey C, Shimeld SM. A Notch-regulated proliferative stem cell zone in the developing spinal cord is an ancestral vertebrate trait. Development 2019; 146:dev.166595. [PMID: 30552127 DOI: 10.1242/dev.166595] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 11/26/2018] [Indexed: 01/01/2023]
Abstract
Vertebrates have evolved the most sophisticated nervous systems we know. These differ from the nervous systems of invertebrates in several ways, including the evolution of new cell types, and the emergence and elaboration of patterning mechanisms to organise cells in time and space. Vertebrates also generally have many more cells in their central nervous systems than invertebrates, and an increase in neural cell number may have contributed to the sophisticated anatomy of the brain and spinal cord. Here, we study how increased cell number evolved in the vertebrate central nervous system, investigating the regulation of cell proliferation in the lamprey spinal cord. Markers of proliferation show that a ventricular progenitor zone is found throughout the lamprey spinal cord. We show that inhibition of Notch signalling disrupts the maintenance of this zone. When Notch is blocked, progenitor cells differentiate precociously, the proliferative ventricular zone is lost and differentiation markers become expressed throughout the spinal cord. Comparison with other chordates suggests that the emergence of a persistent Notch-regulated proliferative progenitor zone was a crucial step for the evolution of vertebrate spinal cord complexity.
Collapse
Affiliation(s)
- Ricardo Lara-Ramirez
- Department of Zoology, University of Oxford, 11a Mansfield Road, Oxford OX1 3SZ, UK
| | | | - Chiara Anselmi
- Department of Zoology, University of Oxford, 11a Mansfield Road, Oxford OX1 3SZ, UK
| | - Cedric Patthey
- Department of Zoology, University of Oxford, 11a Mansfield Road, Oxford OX1 3SZ, UK
| | - Sebastian M Shimeld
- Department of Zoology, University of Oxford, 11a Mansfield Road, Oxford OX1 3SZ, UK
| |
Collapse
|
31
|
Andrews MG, Kong J, Novitch BG, Butler SJ. New perspectives on the mechanisms establishing the dorsal-ventral axis of the spinal cord. Curr Top Dev Biol 2018; 132:417-450. [PMID: 30797516 DOI: 10.1016/bs.ctdb.2018.12.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Distinct classes of neurons arise at different positions along the dorsal-ventral axis of the spinal cord leading to spinal neurons being segregated along this axis according to their physiological properties and functions. Thus, the neurons associated with motor control are generally located in, or adjacent to, the ventral horn whereas the interneurons (INs) that mediate sensory activities are present within the dorsal horn. Here, we review classic and recent studies examining the developmental mechanisms that establish the dorsal-ventral axis in the embryonic spinal cord. Intriguingly, while the cellular organization of the dorsal and ventral halves of the spinal cord looks superficially similar during early development, the underlying molecular mechanisms that establish dorsal vs ventral patterning are markedly distinct. For example, the ventral spinal cord is patterned by the actions of a single growth factor, sonic hedgehog (Shh) acting as a morphogen, i.e., concentration-dependent signal. Recent studies have shed light on the mechanisms by which the spatial and temporal gradient of Shh is transduced by cells to elicit the generation of different classes of ventral INs, and motor neurons (MNs). In contrast, the dorsal spinal cord is patterned by the action of multiple factors, most notably by members of the bone morphogenetic protein (BMP) and Wnt families. While less is known about dorsal patterning, recent studies have suggested that the BMPs do not act as morphogens to specify dorsal IN identities as previously proposed, rather each BMP has signal-specific activities. Finally, we consider the promise that elucidation of these mechanisms holds for neural repair.
Collapse
Affiliation(s)
- Madeline G Andrews
- Department of Neurobiology, University of California, Los Angeles, CA, United States; Neuroscience Graduate Program, University of California, Los Angeles, CA, United States
| | - Jennifer Kong
- Department of Neurobiology, University of California, Los Angeles, CA, United States; Neuroscience Graduate Program, University of California, Los Angeles, CA, United States
| | - Bennett G Novitch
- Department of Neurobiology, University of California, Los Angeles, CA, United States; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, United States
| | - Samantha J Butler
- Department of Neurobiology, University of California, Los Angeles, CA, United States; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, United States.
| |
Collapse
|
32
|
Nadadhur AG, Leferink PS, Holmes D, Hinz L, Cornelissen-Steijger P, Gasparotto L, Heine VM. Patterning factors during neural progenitor induction determine regional identity and differentiation potential in vitro. Stem Cell Res 2018; 32:25-34. [PMID: 30172094 DOI: 10.1016/j.scr.2018.08.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 08/13/2018] [Accepted: 08/22/2018] [Indexed: 12/20/2022] Open
Abstract
The neural tube consists of neural progenitors (NPs) that acquire different characteristics during gestation due to patterning factors. However, the influence of such patterning factors on human pluripotent stem cells (hPSCs) during in vitro neural differentiation is often unclear. This study compared neural induction protocols involving in vitro patterning with single SMAD inhibition (SSI), retinoic acid (RA) administration and dual SMAD inhibition (DSI). While the derived NP cells expressed known NP markers, they differed in their NP expression profile and differentiation potential. Cortical neuronal cells generated from 1) SSI NPs exhibited less mature neuronal phenotypes, 2) RA NPs exhibited an increased GABAergic phenotype, and 3) DSI NPs exhibited greater expression of glutamatergic lineage markers. Further, although all NPs generated astrocytes, astrocytes derived from the RA-induced NPs had the highest GFAP expression. Differences between NP populations included differential expression of regional identity markers HOXB4, LBX1, OTX1 and GSX2, which persisted into mature neural cell stages. This study suggests that patterning factors regulate how potential NPs may differentiate into specific neuronal and glial cell types in vitro. This challenges the utility of generic neural induction procedures, while highlighting the importance of carefully selecting specific NP protocols.
Collapse
Affiliation(s)
- Aishwarya G Nadadhur
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands
| | - Prisca S Leferink
- Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands
| | - Dwayne Holmes
- Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands
| | - Lisa Hinz
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands
| | - Paulien Cornelissen-Steijger
- Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands
| | - Lisa Gasparotto
- Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands
| | - Vivi M Heine
- Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands; Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands.
| |
Collapse
|
33
|
Kawano R, Ohta K, Lupo G. Cadherin-7 enhances Sonic Hedgehog signalling by preventing Gli3 repressor formation during neural tube patterning. Open Biol 2018; 7:rsob.170225. [PMID: 29263249 PMCID: PMC5746549 DOI: 10.1098/rsob.170225] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 11/09/2017] [Indexed: 01/17/2023] Open
Abstract
Sonic Hedgehog (Shh) is a ventrally enriched morphogen controlling dorsoventral patterning of the neural tube. In the dorsal spinal cord, Gli3 protein bound to suppressor-of-fused (Sufu) is converted into Gli3 repressor (Gli3R), which inhibits Shh-target genes. Activation of Shh signalling prevents Gli3R formation, promoting neural tube ventralization. We show that cadherin-7 (Cdh7) expression in the intermediate spinal cord region is required to delimit the boundary between the ventral and the dorsal spinal cord. We demonstrate that Cdh7 functions as a receptor for Shh and enhances Shh signalling. Binding of Shh to Cdh7 promotes its aggregation on the cell membrane and association of Cdh7 with Gli3 and Sufu. These interactions prevent Gli3R formation and cause Gli3 protein degradation. We propose that Shh can act through Cdh7 to limit intracellular movement of Gli3 protein and production of Gli3R, thus eliciting more efficient activation of Gli-dependent signalling.
Collapse
Affiliation(s)
- Rie Kawano
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, Oita, Japan .,Global COE 'Cell Fate Regulation Research and Education Unit', Kumamoto University, Kumamoto, Japan.,Division of Developmental Neurobiology, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kunimasa Ohta
- Division of Developmental Neurobiology, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan.,International Research Core for Stem Cell-based Developmental Medicine, Kumamoto University, Kumamoto, Japan.,Japan Agency for Medical Research and Development (AMED), Tokyo, Japan
| | - Giuseppe Lupo
- Department of Chemistry, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
34
|
Cardozo MJ, Mysiak KS, Becker T, Becker CG. Reduce, reuse, recycle – Developmental signals in spinal cord regeneration. Dev Biol 2017; 432:53-62. [DOI: 10.1016/j.ydbio.2017.05.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 02/03/2017] [Accepted: 05/11/2017] [Indexed: 02/06/2023]
|
35
|
Hashimoto H, Jiang W, Yoshimura T, Moon KH, Bok J, Ikenaka K. Strong sonic hedgehog signaling in the mouse ventral spinal cord is not required for oligodendrocyte precursor cell (OPC) generation but is necessary for correct timing of its generation. Neurochem Int 2017; 119:178-183. [PMID: 29122585 DOI: 10.1016/j.neuint.2017.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/31/2017] [Accepted: 11/01/2017] [Indexed: 12/15/2022]
Abstract
In the mouse neural tube, sonic hedgehog (Shh) secreted from the floor plate (FP) and the notochord (NC) regulates ventral patterning of the neural tube, and later is essential for the generation of oligodendrocyte precursor cells (OPCs). During early development, the NC is adjacent to the neural tube and induces ventral domains in it, including the FP. In the later stage of development, during gliogenesis in the spinal cord, the pMN domain receives strong Shh signaling input. While this is considered to be essential for the generation of OPCs, the actual role of this strong input in OPC generation remains unclear. Here we studied OPC generation in bromi mutant mice which show abnormal ciliary structure. Shh signaling occurs within cilia and has been reported to be weak in bromi mutants. At E11.5, accumulation of Patched1 mRNA, a Shh signaling reporter, is observed in the pMN domain of wild type but not bromi mutants, whereas expression of Gli1 mRNA, another Shh reporter, disappeared. Thus, Shh signaling input to the pMN domain at E12.5 was reduced in bromi mutant mice. In these mutants, induction of the FP structure was delayed and its size was reduced compared to wild type mice. Furthermore, while the p3 and pMN domains were induced, the length of the Nkx2.2-positive region and the number of Olig2-positive cells decreased. The number of OPCs was also significantly decreased in the E12.5 and E14.5 bromi mutant spinal cord. In contrast, motor neuron (MN) production, detected by HB9 expression, significantly increased. It is likely that the transition from MN production to OPC generation in the pMN domain is impaired in bromi mutant mice. These results suggest that strong Shh input to the pMN domain is not required for OPC generation but is essential for producing a sufficient number of OPCs.
Collapse
Affiliation(s)
- Hirokazu Hashimoto
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Aichi 444-8787, Japan; Department of Physiological Sciences, School of Life Sciences, SOKENDAI (The Graduate University for Advanced Studies), Hayama, Kanagawa 240-0193, Japan
| | - Wen Jiang
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Aichi 444-8787, Japan; Department of Physiological Sciences, School of Life Sciences, SOKENDAI (The Graduate University for Advanced Studies), Hayama, Kanagawa 240-0193, Japan
| | - Takeshi Yoshimura
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Aichi 444-8787, Japan; Department of Physiological Sciences, School of Life Sciences, SOKENDAI (The Graduate University for Advanced Studies), Hayama, Kanagawa 240-0193, Japan
| | - Kyeong-Hye Moon
- Department of Anatomy, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea; BK21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| | - Jinwoong Bok
- Department of Anatomy, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea; Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea; BK21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| | - Kazuhiro Ikenaka
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Aichi 444-8787, Japan; Department of Physiological Sciences, School of Life Sciences, SOKENDAI (The Graduate University for Advanced Studies), Hayama, Kanagawa 240-0193, Japan.
| |
Collapse
|
36
|
Andrews MG, Del Castillo LM, Ochoa-Bolton E, Yamauchi K, Smogorzewski J, Butler SJ. BMPs direct sensory interneuron identity in the developing spinal cord using signal-specific not morphogenic activities. eLife 2017; 6. [PMID: 28925352 PMCID: PMC5605194 DOI: 10.7554/elife.30647] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 08/24/2017] [Indexed: 02/06/2023] Open
Abstract
The Bone Morphogenetic Protein (BMP) family reiteratively signals to direct disparate cellular fates throughout embryogenesis. In the developing dorsal spinal cord, multiple BMPs are required to specify sensory interneurons (INs). Previous studies suggested that the BMPs act as concentration-dependent morphogens to direct IN identity, analogous to the manner in which sonic hedgehog patterns the ventral spinal cord. However, it remains unresolved how multiple BMPs would cooperate to establish a unified morphogen gradient. Our studies support an alternative model: BMPs have signal-specific activities directing particular IN fates. Using chicken and mouse models, we show that the identity, not concentration, of the BMP ligand directs distinct dorsal identities. Individual BMPs promote progenitor patterning or neuronal differentiation by their activation of different type I BMP receptors and distinct modulations of the cell cycle. Together, this study shows that a 'mix and match' code of BMP signaling results in distinct classes of sensory INs.
Collapse
Affiliation(s)
- Madeline G Andrews
- Department of Neurobiology, University of California, Los Angeles, United States.,Neuroscience Graduate Program, University of California, Los Angeles, United States.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, United States
| | - Lorenzo M Del Castillo
- Department of Neurobiology, University of California, Los Angeles, United States.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, United States.,CIRM Bridges to Research Program, California State University, Northridge, United States
| | - Eliana Ochoa-Bolton
- Department of Neurobiology, University of California, Los Angeles, United States.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, United States.,CIRM Bridges to Research Program, California State University, Northridge, United States
| | - Ken Yamauchi
- Department of Neurobiology, University of California, Los Angeles, United States.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, United States
| | - Jan Smogorzewski
- Department of Dermatology, University of Southern California, California, United States
| | - Samantha J Butler
- Department of Neurobiology, University of California, Los Angeles, United States.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, United States
| |
Collapse
|
37
|
Oliveira MAP, Balling R, Smidt MP, Fleming RMT. Embryonic development of selectively vulnerable neurons in Parkinson's disease. NPJ Parkinsons Dis 2017; 3:21. [PMID: 28685157 PMCID: PMC5484687 DOI: 10.1038/s41531-017-0022-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 05/24/2017] [Accepted: 06/01/2017] [Indexed: 02/07/2023] Open
Abstract
A specific set of brainstem nuclei are susceptible to degeneration in Parkinson's disease. We hypothesise that neuronal vulnerability reflects shared phenotypic characteristics that confer selective vulnerability to degeneration. Neuronal phenotypic specification is mainly the cumulative result of a transcriptional regulatory program that is active during the development. By manual curation of the developmental biology literature, we comprehensively reconstructed an anatomically resolved cellular developmental lineage for the adult neurons in five brainstem regions that are selectively vulnerable to degeneration in prodromal or early Parkinson's disease. We synthesised the literature on transcription factors that are required to be active, or required to be inactive, in the development of each of these five brainstem regions, and at least two differentially vulnerable nuclei within each region. Certain transcription factors, e.g., Ascl1 and Lmx1b, seem to be required for specification of many brainstem regions that are susceptible to degeneration in early Parkinson's disease. Some transcription factors can even distinguish between differentially vulnerable nuclei within the same brain region, e.g., Pitx3 is required for specification of the substantia nigra pars compacta, but not the ventral tegmental area. We do not suggest that Parkinson's disease is a developmental disorder. In contrast, we consider identification of shared developmental trajectories as part of a broader effort to identify the molecular mechanisms that underlie the phenotypic features that are shared by selectively vulnerable neurons. Systematic in vivo assessment of fate determining transcription factors should be completed for all neuronal populations vulnerable to degeneration in early Parkinson's disease.
Collapse
Affiliation(s)
- Miguel A. P. Oliveira
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6 Avenue du Swing, Belvaux, L-4362 Luxembourg
| | - Rudi Balling
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6 Avenue du Swing, Belvaux, L-4362 Luxembourg
| | - Marten P. Smidt
- Department of Molecular Neuroscience, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Sciencepark 904, 1098 XH Amsterdam, The Netherlands
| | - Ronan M. T. Fleming
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6 Avenue du Swing, Belvaux, L-4362 Luxembourg
| |
Collapse
|
38
|
Laussu J, Audouard C, Kischel A, Assis-Nascimento P, Escalas N, Liebl DJ, Soula C, Davy A. Eph/Ephrin Signaling Controls Progenitor Identities In The Ventral Spinal Cord. Neural Dev 2017; 12:10. [PMID: 28595615 PMCID: PMC5463316 DOI: 10.1186/s13064-017-0087-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 05/31/2017] [Indexed: 02/01/2023] Open
Abstract
Background In the vertebrate spinal cord, motor neurons (MN) are generated in stereotypical numbers from a pool of dedicated progenitors (pMN) whose number depends on signals that control their specification but also their proliferation and differentiation rates. Although the initial steps of pMN specification have been extensively studied, how pMN numbers are regulated over time is less well characterized. Results Here, we show that ephrinB2 and ephrinB3 are differentially expressed in progenitor domains in the ventral spinal cord with several Eph receptors more broadly expressed. Genetic loss-of-function analyses show that ephrinB2 and ephrinB3 inversely control pMN numbers and that these changes in progenitor numbers correlate with changes in motor neuron numbers. Detailed phenotypic analyses by immunostaining and genetic interaction studies between ephrinB2 and Shh indicate that changes in pMN numbers in ephrin mutants are due to alteration in progenitor identity at late stages of development. Conclusions Altogether our data reveal that Eph:ephrin signaling is required to control progenitor identities in the ventral spinal cord. Electronic supplementary material The online version of this article (doi:10.1186/s13064-017-0087-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Julien Laussu
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 118 Route de Narbonne, 31062, Toulouse, France.,Present address: CRBM, 1919 route de Mende, 34293, Montpellier, France
| | - Christophe Audouard
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 118 Route de Narbonne, 31062, Toulouse, France
| | - Anthony Kischel
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 118 Route de Narbonne, 31062, Toulouse, France
| | - Poincyane Assis-Nascimento
- University of Miami Miller School of Medicine, The Miami Project to Cure Paralysis, 1095 NW 14th Terrace, Miami, FL, R-48, USA
| | - Nathalie Escalas
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 118 Route de Narbonne, 31062, Toulouse, France
| | - Daniel J Liebl
- University of Miami Miller School of Medicine, The Miami Project to Cure Paralysis, 1095 NW 14th Terrace, Miami, FL, R-48, USA
| | - Cathy Soula
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 118 Route de Narbonne, 31062, Toulouse, France
| | - Alice Davy
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 118 Route de Narbonne, 31062, Toulouse, France.
| |
Collapse
|
39
|
Zhang Y, Alvarez-Bolado G. Differential developmental strategies by Sonic hedgehog in thalamus and hypothalamus. J Chem Neuroanat 2016; 75:20-7. [DOI: 10.1016/j.jchemneu.2015.11.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 11/25/2015] [Indexed: 12/11/2022]
|
40
|
Ohta S, Wang B, Mansour SL, Schoenwolf GC. BMP regulates regional gene expression in the dorsal otocyst through canonical and non-canonical intracellular pathways. Development 2016; 143:2228-37. [PMID: 27151948 DOI: 10.1242/dev.137133] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 04/27/2016] [Indexed: 12/13/2022]
Abstract
The inner ear consists of two otocyst-derived, structurally and functionally distinct components: the dorsal vestibular and ventral auditory compartments. BMP signaling is required to form the vestibular compartment, but how it complements other required signaling molecules and acts intracellularly is unknown. Using spatially and temporally controlled delivery of signaling pathway regulators to developing chick otocysts, we show that BMP signaling regulates the expression of Dlx5 and Hmx3, both of which encode transcription factors essential for vestibular formation. However, although BMP regulates Dlx5 through the canonical SMAD pathway, surprisingly, it regulates Hmx3 through a non-canonical pathway involving both an increase in cAMP-dependent protein kinase A activity and the GLI3R to GLI3A ratio. Thus, both canonical and non-canonical BMP signaling establish the precise spatiotemporal expression of Dlx5 and Hmx3 during dorsal vestibular development. The identification of the non-canonical pathway suggests an intersection point between BMP and SHH signaling, which is required for ventral auditory development.
Collapse
Affiliation(s)
- Sho Ohta
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84132-3401, USA
| | - Baolin Wang
- Department of Cell and Developmental Biology and Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Suzanne L Mansour
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84132-3401, USA Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112-5330, USA
| | - Gary C Schoenwolf
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84132-3401, USA
| |
Collapse
|
41
|
Keratan Sulfate Regulates the Switch from Motor Neuron to Oligodendrocyte Generation During Development of the Mouse Spinal Cord. Neurochem Res 2016; 41:450-62. [DOI: 10.1007/s11064-016-1861-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 02/02/2016] [Accepted: 02/03/2016] [Indexed: 10/22/2022]
|
42
|
Petracca YL, Sartoretti MM, Di Bella DJ, Marin-Burgin A, Carcagno AL, Schinder AF, Lanuza GM. The late and dual origin of cerebrospinal fluid-contacting neurons in the mouse spinal cord. Development 2016; 143:880-91. [PMID: 26839365 DOI: 10.1242/dev.129254] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 01/25/2016] [Indexed: 12/16/2022]
Abstract
Considerable progress has been made in understanding the mechanisms that control the production of specialized neuronal types. However, how the timing of differentiation contributes to neuronal diversity in the developing spinal cord is still a pending question. In this study, we show that cerebrospinal fluid-contacting neurons (CSF-cNs), an anatomically discrete cell type of the ependymal area, originate from surprisingly late neurogenic events in the ventral spinal cord. CSF-cNs are identified by the expression of the transcription factors Gata2 and Gata3, and the ionic channels Pkd2l1 and Pkd1l2. Contrasting with Gata2/3(+) V2b interneurons, differentiation of CSF-cNs is independent of Foxn4 and takes place during advanced developmental stages previously assumed to be exclusively gliogenic. CSF-cNs are produced from two distinct dorsoventral regions of the mouse spinal cord. Most CSF-cNs derive from progenitors circumscribed to the late-p2 and the oligodendrogenic (pOL) domains, whereas a second subset of CSF-cNs arises from cells bordering the floor plate. The development of these two subgroups of CSF-cNs is differentially controlled by Pax6, they adopt separate locations around the postnatal central canal and they display electrophysiological differences. Our results highlight that spatiotemporal mechanisms are instrumental in creating neural cell diversity in the ventral spinal cord to produce distinct classes of interneurons, motoneurons, CSF-cNs, glial cells and ependymal cells.
Collapse
Affiliation(s)
- Yanina L Petracca
- Developmental Neurobiology Lab, Instituto Leloir and Consejo Nacional de Investigaciones Científicas y Técnicas (IIBBA-CONICET), Avenida Patricias Argentinas 435, Buenos Aires 1405, Argentina
| | - Maria Micaela Sartoretti
- Developmental Neurobiology Lab, Instituto Leloir and Consejo Nacional de Investigaciones Científicas y Técnicas (IIBBA-CONICET), Avenida Patricias Argentinas 435, Buenos Aires 1405, Argentina
| | - Daniela J Di Bella
- Developmental Neurobiology Lab, Instituto Leloir and Consejo Nacional de Investigaciones Científicas y Técnicas (IIBBA-CONICET), Avenida Patricias Argentinas 435, Buenos Aires 1405, Argentina
| | - Antonia Marin-Burgin
- Neuronal Plasticity Lab, Instituto Leloir and Consejo Nacional de Investigaciones Científicas y Técnicas (IIBBA-CONICET), Avenida Patricias Argentinas 435, Buenos Aires 1405, Argentina
| | - Abel L Carcagno
- Developmental Neurobiology Lab, Instituto Leloir and Consejo Nacional de Investigaciones Científicas y Técnicas (IIBBA-CONICET), Avenida Patricias Argentinas 435, Buenos Aires 1405, Argentina
| | - Alejandro F Schinder
- Neuronal Plasticity Lab, Instituto Leloir and Consejo Nacional de Investigaciones Científicas y Técnicas (IIBBA-CONICET), Avenida Patricias Argentinas 435, Buenos Aires 1405, Argentina
| | - Guillermo M Lanuza
- Developmental Neurobiology Lab, Instituto Leloir and Consejo Nacional de Investigaciones Científicas y Técnicas (IIBBA-CONICET), Avenida Patricias Argentinas 435, Buenos Aires 1405, Argentina
| |
Collapse
|
43
|
Thélie A, Desiderio S, Hanotel J, Quigley I, Van Driessche B, Rodari A, Borromeo MD, Kricha S, Lahaye F, Croce J, Cerda-Moya G, Ordoño Fernandez J, Bolle B, Lewis KE, Sander M, Pierani A, Schubert M, Johnson JE, Kintner CR, Pieler T, Van Lint C, Henningfeld KA, Bellefroid EJ, Van Campenhout C. Prdm12 specifies V1 interneurons through cross-repressive interactions with Dbx1 and Nkx6 genes in Xenopus. Development 2016; 142:3416-28. [PMID: 26443638 DOI: 10.1242/dev.121871] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
V1 interneurons are inhibitory neurons that play an essential role in vertebrate locomotion. The molecular mechanisms underlying their genesis remain, however, largely undefined. Here, we show that the transcription factor Prdm12 is selectively expressed in p1 progenitors of the hindbrain and spinal cord in the frog embryo, and that a similar restricted expression profile is observed in the nerve cord of other vertebrates as well as of the cephalochordate amphioxus. Using frog, chick and mice, we analyzed the regulation of Prdm12 and found that its expression in the caudal neural tube is dependent on retinoic acid and Pax6, and that it is restricted to p1 progenitors, due to the repressive action of Dbx1 and Nkx6-1/2 expressed in the adjacent p0 and p2 domains. Functional studies in the frog, including genome-wide identification of its targets by RNA-seq and ChIP-Seq, reveal that vertebrate Prdm12 proteins act as a general determinant of V1 cell fate, at least in part, by directly repressing Dbx1 and Nkx6 genes. This probably occurs by recruiting the methyltransferase G9a, an activity that is not displayed by the amphioxus Prdm12 protein. Together, these findings indicate that Prdm12 promotes V1 interneurons through cross-repressive interactions with Dbx1 and Nkx6 genes, and suggest that this function might have only been acquired after the split of the vertebrate and cephalochordate lineages.
Collapse
Affiliation(s)
- Aurore Thélie
- Laboratory of Developmental Genetics, Université Libre de Bruxelles (ULB), Institute of Molecular Biology and Medecine (IBMM) and ULB Neuroscience Institute, Gosselies B-6041, Belgium
| | - Simon Desiderio
- Laboratory of Developmental Genetics, Université Libre de Bruxelles (ULB), Institute of Molecular Biology and Medecine (IBMM) and ULB Neuroscience Institute, Gosselies B-6041, Belgium
| | - Julie Hanotel
- Laboratory of Developmental Genetics, Université Libre de Bruxelles (ULB), Institute of Molecular Biology and Medecine (IBMM) and ULB Neuroscience Institute, Gosselies B-6041, Belgium
| | - Ian Quigley
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | | | - Anthony Rodari
- Laboratory of Molecular Virology, ULB, IBMM, Gosselies B-6041, Belgium
| | - Mark D Borromeo
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sadia Kricha
- Laboratory of Developmental Genetics, Université Libre de Bruxelles (ULB), Institute of Molecular Biology and Medecine (IBMM) and ULB Neuroscience Institute, Gosselies B-6041, Belgium
| | - François Lahaye
- Sorbonne Universités, UPMC Université Paris 06, CNRS UMR 7009, Laboratoire de Biologie du Développement de Villefranche-sur-Mer (UMR 7009), Observatoire Océanologique de Villefranche-sur-Mer, Villefranche-sur-Mer 06230, France
| | - Jenifer Croce
- Sorbonne Universités, UPMC Université Paris 06, CNRS UMR 7009, Laboratoire de Biologie du Développement de Villefranche-sur-Mer (UMR 7009), Observatoire Océanologique de Villefranche-sur-Mer, Villefranche-sur-Mer 06230, France
| | - Gustavo Cerda-Moya
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Jesús Ordoño Fernandez
- Laboratory of Developmental Genetics, Université Libre de Bruxelles (ULB), Institute of Molecular Biology and Medecine (IBMM) and ULB Neuroscience Institute, Gosselies B-6041, Belgium
| | - Barbara Bolle
- Laboratory of Developmental Genetics, Université Libre de Bruxelles (ULB), Institute of Molecular Biology and Medecine (IBMM) and ULB Neuroscience Institute, Gosselies B-6041, Belgium
| | - Katharine E Lewis
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY 13244, USA
| | - Maike Sander
- Departments of Pediatrics and Cellular and Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093-0695, USA
| | - Alessandra Pierani
- Génétique et développement du cortex cerebral, Institut Jacques Monod, CNRS UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, Paris Cedex 13 75205, France
| | - Michael Schubert
- Sorbonne Universités, UPMC Université Paris 06, CNRS UMR 7009, Laboratoire de Biologie du Développement de Villefranche-sur-Mer (UMR 7009), Observatoire Océanologique de Villefranche-sur-Mer, Villefranche-sur-Mer 06230, France
| | - Jane E Johnson
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Christopher R Kintner
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Tomas Pieler
- Department of Developmental Biochemistry, Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University of Göttingen, 37077 Göttingen, Germany
| | - Carine Van Lint
- Laboratory of Molecular Virology, ULB, IBMM, Gosselies B-6041, Belgium
| | - Kristine A Henningfeld
- Department of Developmental Biochemistry, Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University of Göttingen, 37077 Göttingen, Germany
| | - Eric J Bellefroid
- Laboratory of Developmental Genetics, Université Libre de Bruxelles (ULB), Institute of Molecular Biology and Medecine (IBMM) and ULB Neuroscience Institute, Gosselies B-6041, Belgium
| | - Claude Van Campenhout
- Laboratory of Developmental Genetics, Université Libre de Bruxelles (ULB), Institute of Molecular Biology and Medecine (IBMM) and ULB Neuroscience Institute, Gosselies B-6041, Belgium
| |
Collapse
|
44
|
Huang S, O'Donovan KJ, Turner EE, Zhong J, Ginty DD. Extrinsic and intrinsic signals converge on the Runx1/CBFβ transcription factor for nonpeptidergic nociceptor maturation. eLife 2015; 4:e10874. [PMID: 26418744 PMCID: PMC4657622 DOI: 10.7554/elife.10874] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 09/28/2015] [Indexed: 01/16/2023] Open
Abstract
The generation of diverse neuronal subtypes involves specification of neural progenitors and, subsequently, postmitotic neuronal differentiation, a relatively poorly understood process. Here, we describe a mechanism whereby the neurotrophic factor NGF and the transcription factor Runx1 coordinate postmitotic differentiation of nonpeptidergic nociceptors, a major nociceptor subtype. We show that the integrity of a Runx1/CBFβ holocomplex is crucial for NGF-dependent nonpeptidergic nociceptor maturation. NGF signals through the ERK/MAPK pathway to promote expression of Cbfb but not Runx1 prior to maturation of nonpeptidergic nociceptors. In contrast, transcriptional initiation of Runx1 in nonpeptidergic nociceptor precursors is dependent on the homeodomain transcription factor Islet1, which is largely dispensable for Cbfb expression. Thus, an NGF/TrkA-MAPK-CBFβ pathway converges with Islet1-Runx1 signaling to promote Runx1/CBFβ holocomplex formation and nonpeptidergic nociceptor maturation. Convergence of extrinsic and intrinsic signals to control heterodimeric transcription factor complex formation provides a robust mechanism for postmitotic neuronal subtype specification. DOI:http://dx.doi.org/10.7554/eLife.10874.001 Animals detect and respond to their environment using their sensory nervous system, which forms through a complex, multi-step process. A precursor nerve cell’s fate is set early in its development, and determines the different nerve types it can become. As development progresses, sensory nerve cells develop further into distinct subtypes that perform particular tasks, such as responding to touch or pain. Nociceptors are the specialised sensory nerves that respond to potentially harmful stimuli. They form two distinct subtypes: peptidergic nerves detect potentially dangerous temperatures, whereas non-peptidergic nerves detect potentially dangerous mechanical sensations. Both subtypes originate from the same precursor nerve cell and both initially depend on an external molecule called NGF for their development and survival. During their development, non-peptidergic neurons stop responding to NGF and start producing a protein called Runx1, considered to be the ‘master regulator’ of non-peptidergic nerve cell development. Runx1 works by forming a complex with another protein called CBFbβ, and this complex activates a program of gene expression that is specific to non-peptidergic nerves. However it was unclear how an external signal, like NGF, can coordinate with or influence a nerve cell’s internal genetic program during the nerve’s development. It was also not known whether NGF and Runx1 interact with each other. By studying non-peptidergic nerve cell development in mice that lack NGF, Runx1 and other associated proteins, Huang et al. have now established the sequence of events that regulate the development of this nerve cell subtype. Two signalling pathways converge to switch on non-peptidergic nerve cell development. An NGF-driven signalling pathway activates the production of CBFβ, while another protein binds to the Runx1 gene to switch it on. This leads to the production of the Runx1 and CBFβ proteins that complex together to activate the non-peptidergic neuronal genetic program. These findings demonstrate how two different mechanisms converge to produce the component parts of a complex, which then activates a genetic program that drives the development of a particular neuronal subtype. Whether this mechanism is involved in determining the fate of other cell types remains a question for future work. DOI:http://dx.doi.org/10.7554/eLife.10874.002
Collapse
Affiliation(s)
- Siyi Huang
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, United States.,Department of Neuroscience, Howard Hughes Medical Institute, Johns Hopkins School of Medicine, Baltimore, United States
| | - Kevin J O'Donovan
- Burke Medical Research Institute, Weill Medical College of Cornell University, White Plains, United States
| | - Eric E Turner
- Seattle Children's Hospital, Seattle Children's Research Institute, Seattle, United States
| | - Jian Zhong
- Burke Medical Research Institute, Weill Medical College of Cornell University, White Plains, United States
| | - David D Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, United States.,Department of Neuroscience, Howard Hughes Medical Institute, Johns Hopkins School of Medicine, Baltimore, United States
| |
Collapse
|
45
|
Stasiulewicz M, Gray SD, Mastromina I, Silva JC, Björklund M, Seymour PA, Booth D, Thompson C, Green RJ, Hall EA, Serup P, Dale JK. A conserved role for Notch signaling in priming the cellular response to Shh through ciliary localisation of the key Shh transducer Smo. Development 2015; 142:2291-303. [PMID: 25995356 PMCID: PMC4510595 DOI: 10.1242/dev.125237] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 05/11/2015] [Indexed: 01/13/2023]
Abstract
Notochord-derived Sonic Hedgehog (Shh) is essential for dorsoventral patterning of the overlying neural tube. Increasing concentration and duration of Shh signal induces progenitors to acquire progressively more ventral fates. We show that Notch signalling augments the response of neuroepithelial cells to Shh, leading to the induction of higher expression levels of the Shh target gene Ptch1 and subsequently induction of more ventral cell fates. Furthermore, we demonstrate that activated Notch1 leads to pronounced accumulation of Smoothened (Smo) within primary cilia and elevated levels of full-length Gli3. Finally, we show that Notch activity promotes longer primary cilia both in vitro and in vivo. Strikingly, these Notch-regulated effects are Shh independent. These data identify Notch signalling as a novel modulator of Shh signalling that acts mechanistically via regulation of ciliary localisation of key components of its transduction machinery. Highlighted article: Shh signalling controls dorso-ventral cell fate in the neural tube. Notch regulates ciliary architecture and localisation of key Shh pathway components, thus sensitising cells to Shh.
Collapse
Affiliation(s)
- Magdalena Stasiulewicz
- Division of Cell and Developmental Biology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, Scotland, UK
| | - Shona D Gray
- Division of Cell and Developmental Biology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, Scotland, UK
| | - Ioanna Mastromina
- Division of Cell and Developmental Biology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, Scotland, UK
| | - Joana C Silva
- Division of Cell and Developmental Biology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, Scotland, UK
| | - Mia Björklund
- Division of Cell and Developmental Biology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, Scotland, UK
| | - Philip A Seymour
- Division of Cell and Developmental Biology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, Scotland, UK The Danish Stem Cell Center, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen DK-2200, Denmark
| | - David Booth
- Division of Cell and Developmental Biology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, Scotland, UK
| | - Calum Thompson
- Division of Cell and Developmental Biology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, Scotland, UK
| | - Richard J Green
- Division of Cell and Developmental Biology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, Scotland, UK
| | - Emma A Hall
- Division of Cell and Developmental Biology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, Scotland, UK MRC Human Genetics, Institute for Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Palle Serup
- Division of Cell and Developmental Biology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, Scotland, UK The Danish Stem Cell Center, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen DK-2200, Denmark
| | - J Kim Dale
- Division of Cell and Developmental Biology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, Scotland, UK
| |
Collapse
|
46
|
Hudson C, Sirour C, Yasuo H. Snail mediates medial-lateral patterning of the ascidian neural plate. Dev Biol 2015; 403:172-9. [PMID: 25962578 DOI: 10.1016/j.ydbio.2015.04.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 04/20/2015] [Accepted: 04/24/2015] [Indexed: 11/30/2022]
Abstract
The ascidian neural plate exhibits a regular, grid-like arrangement of cells. Patterning of the neural plate across the medial-lateral axis is initiated by bilateral sources of Nodal signalling, such that Nodal signalling induces expression of lateral neural plate genes and represses expression of medial neural plate genes. One of the earliest lateral neural plate genes induced by Nodal signals encodes the transcription factor Snail. Here, we show that Snail is a critical downstream factor mediating this Nodal-dependent patterning. Using gain and loss of function approaches, we show that Snail is required to repress medial neural plate gene expression at neural plate stages and to maintain the lateral neural tube genetic programme at later stages. A comparison of these results to those obtained following Nodal gain and loss of function indicates that Snail mediates a subset of Nodal functions. Consistently, overexpression of Snail can partially rescue a Nodal inhibition phenotype. We conclude that Snail is an early component of the gene regulatory network, initiated by Nodal signals, that patterns the ascidian neural plate.
Collapse
Affiliation(s)
- Clare Hudson
- CNRS, Laboratoire de Biologie du Développement de Villefranche-sur-mer, Observatoire Océanologique, 06230 Villefranche-sur-mer, France; Sorbonne Universités, UPMC Univ Paris 06, Laboratoire de Biologie du Développement de Villefranche-sur-mer, Observatoire Océanologique, 06230 Villefranche-sur-mer, France.
| | - Cathy Sirour
- CNRS, Laboratoire de Biologie du Développement de Villefranche-sur-mer, Observatoire Océanologique, 06230 Villefranche-sur-mer, France; Sorbonne Universités, UPMC Univ Paris 06, Laboratoire de Biologie du Développement de Villefranche-sur-mer, Observatoire Océanologique, 06230 Villefranche-sur-mer, France
| | - Hitoyoshi Yasuo
- CNRS, Laboratoire de Biologie du Développement de Villefranche-sur-mer, Observatoire Océanologique, 06230 Villefranche-sur-mer, France; Sorbonne Universités, UPMC Univ Paris 06, Laboratoire de Biologie du Développement de Villefranche-sur-mer, Observatoire Océanologique, 06230 Villefranche-sur-mer, France
| |
Collapse
|
47
|
Enriquez J, Venkatasubramanian L, Baek M, Peterson M, Aghayeva U, Mann RS. Specification of individual adult motor neuron morphologies by combinatorial transcription factor codes. Neuron 2015; 86:955-970. [PMID: 25959734 DOI: 10.1016/j.neuron.2015.04.011] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Revised: 03/12/2015] [Accepted: 04/04/2015] [Indexed: 11/27/2022]
Abstract
How the highly stereotyped morphologies of individual neurons are genetically specified is not well understood. We identify six transcription factors (TFs) expressed in a combinatorial manner in seven post-mitotic adult leg motor neurons (MNs) that are derived from a single neuroblast in Drosophila. Unlike TFs expressed in mitotically active neuroblasts, these TFs do not regulate each other's expression. Removing the activity of a single TF resulted in specific morphological defects, including muscle targeting and dendritic arborization, and in a highly specific walking defect in adult flies. In contrast, when the expression of multiple TFs was modified, nearly complete transformations in MN morphologies were generated. These results show that the morphological characteristics of a single neuron are dictated by a combinatorial code of morphology TFs (mTFs). mTFs function at a previously unidentified regulatory tier downstream of factors acting in the NB but independently of factors that act in terminally differentiated neurons.
Collapse
Affiliation(s)
- Jonathan Enriquez
- Department of Biochemistry and Molecular Biophysics, Columbia University, HHSC 1108, 701 W. 168(th) Street, New York, NY 10032, USA.
| | - Lalanti Venkatasubramanian
- Department of Biochemistry and Molecular Biophysics, Columbia University, HHSC 1108, 701 W. 168(th) Street, New York, NY 10032, USA
| | - Myungin Baek
- Department of Biochemistry and Molecular Biophysics, Columbia University, HHSC 1108, 701 W. 168(th) Street, New York, NY 10032, USA
| | - Meredith Peterson
- Department of Biochemistry and Molecular Biophysics, Columbia University, HHSC 1108, 701 W. 168(th) Street, New York, NY 10032, USA
| | - Ulkar Aghayeva
- Department of Biochemistry and Molecular Biophysics, Columbia University, HHSC 1108, 701 W. 168(th) Street, New York, NY 10032, USA
| | - Richard S Mann
- Department of Biochemistry and Molecular Biophysics, Columbia University, HHSC 1108, 701 W. 168(th) Street, New York, NY 10032, USA.
| |
Collapse
|
48
|
Kong JH, Yang L, Dessaud E, Chuang K, Moore DM, Rohatgi R, Briscoe J, Novitch BG. Notch activity modulates the responsiveness of neural progenitors to sonic hedgehog signaling. Dev Cell 2015; 33:373-87. [PMID: 25936505 PMCID: PMC4449290 DOI: 10.1016/j.devcel.2015.03.005] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 12/29/2014] [Accepted: 03/03/2015] [Indexed: 01/20/2023]
Abstract
Throughout the developing nervous system, neural stem and progenitor cells give rise to diverse classes of neurons and glia in a spatially and temporally coordinated manner. In the ventral spinal cord, much of this diversity emerges through the morphogen actions of Sonic hedgehog (Shh). Interpretation of the Shh gradient depends on both the amount of ligand and duration of exposure, but the mechanisms permitting prolonged responses to Shh are not well understood. We demonstrate that Notch signaling plays an essential role in this process, enabling neural progenitors to attain sufficiently high levels of Shh pathway activity needed to direct the ventral-most cell fates. Notch activity regulates subcellular localization of the Shh receptor Patched1, gating the translocation of the key effector Smoothened to primary cilia and its downstream signaling activities. These data reveal an unexpected role for Notch shaping the interpretation of the Shh morphogen gradient and influencing cell fate determination. Changes in Notch signaling alter the dorsoventral identity of neural progenitors Activation and inactivation of Notch signaling alter cellular responses to Shh Notch activity is required for efficient trafficking of Smo to primary cilia Notch activity regulates the subcellular distribution of the Shh receptor Ptch1
Collapse
Affiliation(s)
- Jennifer H Kong
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Linlin Yang
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Eric Dessaud
- The Francis Crick Institute, Mill Hill Laboratory, London NW7 1AA, UK
| | - Katherine Chuang
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Destaye M Moore
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Rajat Rohatgi
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - James Briscoe
- The Francis Crick Institute, Mill Hill Laboratory, London NW7 1AA, UK
| | - Bennett G Novitch
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
49
|
Singh S, Solecki DJ. Polarity transitions during neurogenesis and germinal zone exit in the developing central nervous system. Front Cell Neurosci 2015; 9:62. [PMID: 25852469 PMCID: PMC4349153 DOI: 10.3389/fncel.2015.00062] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 02/10/2015] [Indexed: 11/14/2022] Open
Abstract
During neural development, billions of neurons differentiate, polarize, migrate and form synapses in a precisely choreographed sequence. These precise developmental events are accompanied by discreet transitions in cellular polarity. While radial glial neural stem cells are highly polarized, transiently amplifying neural progenitors are less polarized after delaminating from their parental stem cell. Moreover, preceding their radial migration to a final laminar position neural progenitors re-adopt a polarized morphology before they embarking on their journey along a glial guide to the destination where they will fully mature. In this review, we will compare and contrast the key polarity transitions of cells derived from a neuroepithelium to the well-characterized polarity transitions that occur in true epithelia. We will highlight recent advances in the field that shows that neuronal progenitor delamination from germinal zone (GZ) niche shares similarities to an epithelial-mesenchymal transition. Moreover, studies in the cerebellum suggest the acquisition of radial migration and polarity in transiently amplifying neural progenitors share similarities to mesenchymal-epithelial transitions. Where applicable, we will compare and contrast the precise molecular mechanisms used by epithelial cells and neuronal progenitors to control plasticity in cell polarity during their distinct developmental programs.
Collapse
Affiliation(s)
- Shalini Singh
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital Memphis, TN, USA
| | - David J Solecki
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital Memphis, TN, USA
| |
Collapse
|
50
|
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
|