1
|
Ventriglia S, Kalcheim C. From neural tube to spinal cord: The dynamic journey of the dorsal neuroepithelium. Dev Biol 2024; 511:26-38. [PMID: 38580174 DOI: 10.1016/j.ydbio.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/21/2024] [Accepted: 04/02/2024] [Indexed: 04/07/2024]
Abstract
In a developing embryo, formation of tissues and organs is remarkably precise in both time and space. Through cell-cell interactions, neighboring progenitors coordinate their activities, sequentially generating distinct types of cells. At present, we only have limited knowledge, rather than a systematic understanding, of the underlying logic and mechanisms responsible for cell fate transitions. The formation of the dorsal aspect of the spinal cord is an outstanding model to tackle these dynamics, as it first generates the peripheral nervous system and is later responsible for transmitting sensory information from the periphery to the brain and for coordinating local reflexes. This is reflected first by the ontogeny of neural crest cells, progenitors of the peripheral nervous system, followed by formation of the definitive roof plate of the central nervous system and specification of adjacent interneurons, then a transformation of roof plate into dorsal radial glia and ependyma lining the forming central canal. How do these peripheral and central neural branches segregate from common progenitors? How are dorsal radial glia established concomitant with transformation of the neural tube lumen into a central canal? How do the dorsal radial glia influence neighboring cells? This is only a partial list of questions whose clarification requires the implementation of experimental paradigms in which precise control of timing is crucial. Here, we outline some available answers and still open issues, while highlighting the contributions of avian models and their potential to address mechanisms of neural patterning and function.
Collapse
Affiliation(s)
- Susanna Ventriglia
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, 9112102, P.O.Box 12272, Israel.
| | - Chaya Kalcheim
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, 9112102, P.O.Box 12272, Israel.
| |
Collapse
|
2
|
Kahane N, Dahan-Barda Y, Kalcheim C. A Spatio-Temporal-Dependent Requirement of Sonic Hedgehog in the Early Development of Sclerotome-Derived Vertebrae and Ribs. Int J Mol Sci 2024; 25:5602. [PMID: 38891790 PMCID: PMC11171667 DOI: 10.3390/ijms25115602] [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: 04/15/2024] [Revised: 05/13/2024] [Accepted: 05/19/2024] [Indexed: 06/21/2024] Open
Abstract
Derived from axial structures, Sonic Hedgehog (Shh) is secreted into the paraxial mesoderm, where it plays crucial roles in sclerotome induction and myotome differentiation. Through conditional loss-of-function in quail embryos, we investigate the timing and impact of Shh activity during early formation of sclerotome-derived vertebrae and ribs, and of lateral mesoderm-derived sternum. To this end, Hedgehog interacting protein (Hhip) was electroporated at various times between days 2 and 5. While the vertebral body and rib primordium showed consistent size reduction, rib expansion into the somatopleura remained unaffected, and the sternal bud developed normally. Additionally, we compared these effects with those of locally inhibiting BMP activity. Transfection of Noggin in the lateral mesoderm hindered sternal bud formation. Unlike Hhip, BMP inhibition via Noggin or Smad6 induced myogenic differentiation of the lateral dermomyotome lip, while impeding the growth of the myotome/rib complex into the somatic mesoderm, thus affirming the role of the lateral dermomyotome epithelium in rib guidance. Overall, these findings underscore the continuous requirement for opposing gradients of Shh and BMP activity in the morphogenesis of proximal and distal flank skeletal structures, respectively. Future research should address the implications of these early interactions to the later morphogenesis and function of the musculo-skeletal system and of possible associated malformations.
Collapse
Affiliation(s)
| | | | - Chaya Kalcheim
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University of Jerusalem-Hadassah Medical School, P.O. Box 12272, Jerusalem 9112102, Israel; (N.K.); (Y.D.-B.)
| |
Collapse
|
3
|
Kotov A, Seal S, Alkobtawi M, Kappès V, Ruiz SM, Arbès H, Harland RM, Peshkin L, Monsoro-Burq AH. A time-resolved single-cell roadmap of the logic driving anterior neural crest diversification from neural border to migration stages. Proc Natl Acad Sci U S A 2024; 121:e2311685121. [PMID: 38683994 PMCID: PMC11087755 DOI: 10.1073/pnas.2311685121] [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: 07/20/2023] [Accepted: 03/12/2024] [Indexed: 05/02/2024] Open
Abstract
Neural crest cells exemplify cellular diversification from a multipotent progenitor population. However, the full sequence of early molecular choices orchestrating the emergence of neural crest heterogeneity from the embryonic ectoderm remains elusive. Gene-regulatory-networks (GRN) govern early development and cell specification toward definitive neural crest. Here, we combine ultradense single-cell transcriptomes with machine-learning and large-scale transcriptomic and epigenomic experimental validation of selected trajectories, to provide the general principles and highlight specific features of the GRN underlying neural crest fate diversification from induction to early migration stages using Xenopus frog embryos as a model. During gastrulation, a transient neural border zone state precedes the choice between neural crest and placodes which includes multiple converging gene programs. During neurulation, transcription factor connectome, and bifurcation analyses demonstrate the early emergence of neural crest fates at the neural plate stage, alongside an unbiased multipotent-like lineage persisting until epithelial-mesenchymal transition stage. We also decipher circuits driving cranial and vagal neural crest formation and provide a broadly applicable high-throughput validation strategy for investigating single-cell transcriptomes in vertebrate GRNs in development, evolution, and disease.
Collapse
Affiliation(s)
- Aleksandr Kotov
- Université Paris-Saclay, Département de Biologie, Faculté des Sciences d’Orsay, Signalisation Radiobiology and Cancer, CNRS UMR 3347, INSERM U1021, OrsayF-91405, France
- Institut Curie Research Division, Paris Science et Lettres Research University, OrsayF-91405, France
| | - Subham Seal
- Université Paris-Saclay, Département de Biologie, Faculté des Sciences d’Orsay, Signalisation Radiobiology and Cancer, CNRS UMR 3347, INSERM U1021, OrsayF-91405, France
- Institut Curie Research Division, Paris Science et Lettres Research University, OrsayF-91405, France
| | - Mansour Alkobtawi
- Université Paris-Saclay, Département de Biologie, Faculté des Sciences d’Orsay, Signalisation Radiobiology and Cancer, CNRS UMR 3347, INSERM U1021, OrsayF-91405, France
- Institut Curie Research Division, Paris Science et Lettres Research University, OrsayF-91405, France
| | - Vincent Kappès
- Université Paris-Saclay, Département de Biologie, Faculté des Sciences d’Orsay, Signalisation Radiobiology and Cancer, CNRS UMR 3347, INSERM U1021, OrsayF-91405, France
- Institut Curie Research Division, Paris Science et Lettres Research University, OrsayF-91405, France
| | - Sofia Medina Ruiz
- Molecular and Cell Biology Department, Genetics, Genomics and Development Division, University of California Berkeley, CA94720
| | - Hugo Arbès
- Université Paris-Saclay, Département de Biologie, Faculté des Sciences d’Orsay, Signalisation Radiobiology and Cancer, CNRS UMR 3347, INSERM U1021, OrsayF-91405, France
- Institut Curie Research Division, Paris Science et Lettres Research University, OrsayF-91405, France
| | - Richard M. Harland
- Molecular and Cell Biology Department, Genetics, Genomics and Development Division, University of California Berkeley, CA94720
| | - Leonid Peshkin
- Systems Biology Division, Harvard Medical School, Boston, MA02115
| | - Anne H. Monsoro-Burq
- Université Paris-Saclay, Département de Biologie, Faculté des Sciences d’Orsay, Signalisation Radiobiology and Cancer, CNRS UMR 3347, INSERM U1021, OrsayF-91405, France
- Institut Curie Research Division, Paris Science et Lettres Research University, OrsayF-91405, France
- Institut Universitaire de France, ParisF-75005, France
| |
Collapse
|
4
|
Kato Y, Yoshida S, Kato T. Missing pieces of the pituitary puzzle: participation of extra-adenohypophyseal placode-lineage cells in the adult pituitary gland. Cell Tissue Res 2023; 394:487-496. [PMID: 37650920 DOI: 10.1007/s00441-023-03829-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/14/2023] [Indexed: 09/01/2023]
Abstract
The pituitary gland is a major endocrine tissue composing of two distinct entities, the adenohypophysis (anterior pituitary, cranial placode origin) and the neurohypophysis (posterior pituitary, neural ectoderm origin), and plays important roles in maintaining vital homeostasis. This tissue is maintained by a slow, consistent cell-renewal system of adult stem/progenitor cells. Recent accumulating evidence shows that neural crest-, head mesenchyme-, and endoderm lineage cells invade during pituitary development and contribute to the maintenance of the adult pituitary gland. Based on these novel observations, this article discusses whether these lineage cells are involved in pituitary organogenesis, maintenance, regeneration, dysplasia, or tumors.
Collapse
Affiliation(s)
- Yukio Kato
- Institute for Endocrinology, Meiji University, 1-1-1 Higashi-Mita, Tama-Ku, Kawasaki, Kanagawa, 214-8571, Japan.
| | - Saishu Yoshida
- Department of Biochemistry, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Takako Kato
- Institute for Endocrinology, Meiji University, 1-1-1 Higashi-Mita, Tama-Ku, Kawasaki, Kanagawa, 214-8571, Japan
| |
Collapse
|
5
|
Candido-Ferreira IL, Lukoseviciute M, Sauka-Spengler T. Multi-layered transcriptional control of cranial neural crest development. Semin Cell Dev Biol 2022; 138:1-14. [PMID: 35941042 DOI: 10.1016/j.semcdb.2022.07.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 07/23/2022] [Accepted: 07/23/2022] [Indexed: 11/28/2022]
Abstract
The neural crest (NC) is an emblematic population of embryonic stem-like cells with remarkable migratory ability. These distinctive attributes have inspired the curiosity of developmental biologists for over 150 years, however only recently the regulatory mechanisms controlling the complex features of the NC have started to become elucidated at genomic scales. Regulatory control of NC development is achieved through combinatorial transcription factor binding and recruitment of associated transcriptional complexes to distal cis-regulatory elements. Together, they regulate when, where and to what extent transcriptional programmes are actively deployed, ultimately shaping ontogenetic processes. Here, we discuss how transcriptional networks control NC ontogeny, with a special emphasis on the molecular mechanisms underlying specification of the cephalic NC. We also cover emerging properties of transcriptional regulation revealed in diverse developmental systems, such as the role of three-dimensional conformation of chromatin, and how they are involved in the regulation of NC ontogeny. Finally, we highlight how advances in deciphering the NC transcriptional network have afforded new insights into the molecular basis of human diseases.
Collapse
Affiliation(s)
- Ivan L Candido-Ferreira
- University of Oxford, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford OX3 9DS, UK
| | - Martyna Lukoseviciute
- University of Oxford, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford OX3 9DS, UK
| | - Tatjana Sauka-Spengler
- University of Oxford, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford OX3 9DS, UK.
| |
Collapse
|
6
|
Rekler D, Kalcheim C. Completion of neural crest cell production and emigration is regulated by retinoic-acid-dependent inhibition of BMP signaling. eLife 2022; 11:72723. [PMID: 35394423 PMCID: PMC8993216 DOI: 10.7554/elife.72723] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 03/02/2022] [Indexed: 12/21/2022] Open
Abstract
Production and emigration of neural crest cells is a transient process followed by the emergence of the definitive roof plate. The mechanisms regulating the end of neural crest ontogeny are poorly understood. Whereas early crest development is stimulated by mesoderm-derived retinoic acid, we report that the end of the neural crest period is regulated by retinoic acid synthesized in the dorsal neural tube. Inhibition of retinoic acid signaling in the neural tube prevents the normal upregulation of BMP inhibitors in the nascent roof plate and prolongs the period of BMP responsiveness which otherwise ceases close to roof plate establishment. Consequently, neural crest production and emigration are extended well into the roof plate stage. In turn, extending the activity of neural crest-specific genes inhibits the onset of retinoic acid synthesis in roof plate suggesting a mutual repressive interaction between neural crest and roof plate traits. Although several roof plate-specific genes are normally expressed in the absence of retinoic acid signaling, roof plate and crest markers are co-expressed in single cells and this domain also contains dorsal interneurons. Hence, the cellular and molecular architecture of the roof plate is compromised. Collectively, our results demonstrate that neural tube-derived retinoic acid, via inhibition of BMP signaling, is an essential factor responsible for the end of neural crest generation and the proper segregation of dorsal neural lineages. The division between the central nervous system – formed by the brain and spinal cord – and the peripheral nervous system – which consists of the neurons that sense and relay information to and from the body – takes place early during embryonic development. Initially, the nervous system consists of a tube of cells called the neural tube. From the top region of this tube, some cells change their shape, exit the tube and migrate to different places in the developing body. These cells are called the ‘neural crest’, and they form many different structures, including the peripheral nervous system. Neural crest cells keep leaving the neural tube for a period of time, but after that, the neural tube stops producing them. At this point, the region of the neural tube that had been producing neural crest cells becomes the ‘roof plate’ of the central nervous system, a structure that is essential for the development of specific groups of neurons in the brain and spinal cord. In bird embryos, a protein called bone morphogenetic protein (BMP) is essential for neural crest production because it triggers the migration of these cells away from the neural tube. Before the roof plate is formed, the activity of BMP is blocked by proteins known as BMP inhibitors, which stop more cells from leaving the neural tube. Around the time when neural crest formation stops, another molecule called retinoic acid begins to be synthesized in the top region of the neural tube. Rekler and Kalcheim asked whether retinoic acid is involved in the transition from neural crest to roof plate. To test this hypothesis, Rekler and Kalcheim blocked the activity of retinoic acid in the neural tube of quail embryos at the time when they should stop producing neural crest cells. This resulted in embryos in which the neural tube keeps producing neural crest cells after the roof plate has formed. In these embryos, individual cells in the resulting ‘roof plate’ produced both proteins that are normally only found in neural crest cells, and proteins typically exclusive to the roof plate. This suggests that, in the absence of retinoic acid activity, the segregation of neural crest identity from roof plate identity is compromised. Rekler and Kalcheim also found that, in the embryos where retinoic acid activity had been blocked, the cells in the area where the roof plate should be produced virtually no BMP inhibitors, and exhibited extended BMP activity. This allowed neural crest cells to continue forming and migrating away from the neural tube well after the period when they would stop in a normal embryo. These results indicate that retinoic acid stops the production of neural crest cells by repressing BMP activity in the roof plate of the neural tube. Rekler and Kalcheim’s experiments shed light on the mechanisms that allow the central and peripheral nervous systems to become segregated. This could increase our understanding of the origin of several neurodevelopmental disorders, potentially providing insights into their treatment or prevention. Additionally, the process of neural crest production and exit from the neural tube is highly similar to the process of metastasis in many invasive cancers. Thus, by understanding how the production of neural crest cells is terminated, it may be possible to learn how to prevent malignant cancer cells from spreading through the body.
Collapse
Affiliation(s)
- Dina Rekler
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
| | - Chaya Kalcheim
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
7
|
Dawes JHP, Kelsh RN. Cell Fate Decisions in the Neural Crest, from Pigment Cell to Neural Development. Int J Mol Sci 2021; 22:13531. [PMID: 34948326 PMCID: PMC8706606 DOI: 10.3390/ijms222413531] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 11/17/2022] Open
Abstract
The neural crest shows an astonishing multipotency, generating multiple neural derivatives, but also pigment cells, skeletogenic and other cell types. The question of how this process is controlled has been the subject of an ongoing debate for more than 35 years. Based upon new observations of zebrafish pigment cell development, we have recently proposed a novel, dynamic model that we believe goes some way to resolving the controversy. Here, we will firstly summarize the traditional models and the conflicts between them, before outlining our novel model. We will also examine our recent dynamic modelling studies, looking at how these reveal behaviors compatible with the biology proposed. We will then outline some of the implications of our model, looking at how it might modify our views of the processes of fate specification, differentiation, and commitment.
Collapse
Affiliation(s)
- Jonathan H. P. Dawes
- Centre for Networks and Collective Behaviour, University of Bath, Bath BA2 7AY, UK;
- Department of Mathematical Sciences, University of Bath, Bath BA2 7AY, UK
| | - Robert N. Kelsh
- Centre for Mathematical Biology, University of Bath, Bath BA2 7AY, UK
- Department of Biology & Biochemistry, University of Bath, Bath BA2 7AY, UK
| |
Collapse
|
8
|
Colombo S, Petit V, Wagner RY, Champeval D, Yajima I, Gesbert F, Aktary Z, Davidson I, Delmas V, Larue L. Stabilization of β-catenin promotes melanocyte specification at the expense of the Schwann cell lineage. Development 2021; 149:274086. [PMID: 34878101 PMCID: PMC8917410 DOI: 10.1242/dev.194407] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 11/25/2021] [Indexed: 11/20/2022]
Abstract
The canonical Wnt/β-catenin pathway governs a multitude of developmental processes in various cell lineages, including the melanocyte lineage. Indeed, β-catenin regulates transcription of Mitf-M, the master regulator of this lineage. The first wave of melanocytes to colonize the skin is directly derived from neural crest cells, whereas the second wave of melanocytes is derived from Schwann cell precursors (SCPs). We investigated the influence of β-catenin in the development of melanocytes of the first and second waves by generating mice expressing a constitutively active form of β-catenin in cells expressing tyrosinase. Constitutive activation of β-catenin did not affect the development of truncal melanoblasts but led to marked hyperpigmentation of the paws. By activating β-catenin at various stages of development (E8.5-E11.5), we showed that the activation of β-catenin in bipotent SCPs favored melanoblast specification at the expense of Schwann cells in the limbs within a specific temporal window. Furthermore, in vitro hyperactivation of the Wnt/β-catenin pathway, which is required for melanocyte development, induces activation of Mitf-M, in turn repressing FoxD3 expression. In conclusion, β-catenin overexpression promotes SCP cell fate decisions towards the melanocyte lineage. Summary: Activation of β-catenin in bipotent Schwann cell precursors during a specific developmental window induces Mitf and represses FoxD3 to promote melanoblast cell fate at the expense of Schwann cells in limbs.
Collapse
Affiliation(s)
- Sophie Colombo
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, Orsay, France.,Univ Paris-Sud, Univ Paris-Saclay, CNRS UMR 3347, Orsay, France.,Equipes Labellisées Ligue Contre le Cancer, France
| | - Valérie Petit
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, Orsay, France.,Univ Paris-Sud, Univ Paris-Saclay, CNRS UMR 3347, Orsay, France.,Equipes Labellisées Ligue Contre le Cancer, France
| | - Roselyne Y Wagner
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, Orsay, France.,Univ Paris-Sud, Univ Paris-Saclay, CNRS UMR 3347, Orsay, France.,Equipes Labellisées Ligue Contre le Cancer, France
| | - Delphine Champeval
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, Orsay, France.,Univ Paris-Sud, Univ Paris-Saclay, CNRS UMR 3347, Orsay, France.,Equipes Labellisées Ligue Contre le Cancer, France
| | - Ichiro Yajima
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, Orsay, France.,Univ Paris-Sud, Univ Paris-Saclay, CNRS UMR 3347, Orsay, France.,Equipes Labellisées Ligue Contre le Cancer, France
| | - Franck Gesbert
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, Orsay, France.,Univ Paris-Sud, Univ Paris-Saclay, CNRS UMR 3347, Orsay, France.,Equipes Labellisées Ligue Contre le Cancer, France
| | - Zackie Aktary
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, Orsay, France.,Univ Paris-Sud, Univ Paris-Saclay, CNRS UMR 3347, Orsay, France.,Equipes Labellisées Ligue Contre le Cancer, France
| | - Irwin Davidson
- Equipes Labellisées Ligue Contre le Cancer, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UNISTRA, 1 Rue Laurent Fries, 67404 Illkirch Cedex. Department of Functional Genomics and Cancer, France
| | - Véronique Delmas
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, Orsay, France.,Univ Paris-Sud, Univ Paris-Saclay, CNRS UMR 3347, Orsay, France.,Equipes Labellisées Ligue Contre le Cancer, France
| | - Lionel Larue
- Institut Curie, PSL Research University, INSERM U1021, Normal and Pathological Development of Melanocytes, Orsay, France.,Univ Paris-Sud, Univ Paris-Saclay, CNRS UMR 3347, Orsay, France.,Equipes Labellisées Ligue Contre le Cancer, France
| |
Collapse
|
9
|
Sutton G, Kelsh RN, Scholpp S. Review: The Role of Wnt/β-Catenin Signalling in Neural Crest Development in Zebrafish. Front Cell Dev Biol 2021; 9:782445. [PMID: 34912811 PMCID: PMC8667473 DOI: 10.3389/fcell.2021.782445] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/16/2021] [Indexed: 12/20/2022] Open
Abstract
The neural crest (NC) is a multipotent cell population in vertebrate embryos with extraordinary migratory capacity. The NC is crucial for vertebrate development and forms a myriad of cell derivatives throughout the body, including pigment cells, neuronal cells of the peripheral nervous system, cardiomyocytes and skeletogenic cells in craniofacial tissue. NC induction occurs at the end of gastrulation when the multipotent population of NC progenitors emerges in the ectodermal germ layer in the neural plate border region. In the process of NC fate specification, fate-specific markers are expressed in multipotent progenitors, which subsequently adopt a specific fate. Thus, NC cells delaminate from the neural plate border and migrate extensively throughout the embryo until they differentiate into various cell derivatives. Multiple signalling pathways regulate the processes of NC induction and specification. This review explores the ongoing role of the Wnt/β-catenin signalling pathway during NC development, focusing on research undertaken in the Teleost model organism, zebrafish (Danio rerio). We discuss the function of the Wnt/β-catenin signalling pathway in inducing the NC within the neural plate border and the specification of melanocytes from the NC. The current understanding of NC development suggests a continual role of Wnt/β-catenin signalling in activating and maintaining the gene regulatory network during NC induction and pigment cell specification. We relate this to emerging models and hypotheses on NC fate restriction. Finally, we highlight the ongoing challenges facing NC research, current gaps in knowledge, and this field's potential future directions.
Collapse
Affiliation(s)
- Gemma Sutton
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Robert N. Kelsh
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Steffen Scholpp
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
10
|
Kelsh RN, Camargo Sosa K, Farjami S, Makeev V, Dawes JHP, Rocco A. Cyclical fate restriction: a new view of neural crest cell fate specification. Development 2021; 148:273451. [PMID: 35020872 DOI: 10.1242/dev.176057] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neural crest cells are crucial in development, not least because of their remarkable multipotency. Early findings stimulated two hypotheses for how fate specification and commitment from fully multipotent neural crest cells might occur, progressive fate restriction (PFR) and direct fate restriction, differing in whether partially restricted intermediates were involved. Initially hotly debated, they remain unreconciled, although PFR has become favoured. However, testing of a PFR hypothesis of zebrafish pigment cell development refutes this view. We propose a novel 'cyclical fate restriction' hypothesis, based upon a more dynamic view of transcriptional states, reconciling the experimental evidence underpinning the traditional hypotheses.
Collapse
Affiliation(s)
- Robert N Kelsh
- Department of Biology & Biochemistry, University of Bath, Bath, BA2 7AY, UK
| | - Karen Camargo Sosa
- Department of Biology & Biochemistry, University of Bath, Bath, BA2 7AY, UK
| | - Saeed Farjami
- Department of Microbial Sciences, FHMS, University of Surrey, Guildford, GU2 7XH, UK
| | - Vsevolod Makeev
- Department of Computational Systems Biology, Vavilov Institute of General Genetics, Russian Academy of Sciences, Ul. Gubkina 3, Moscow, 119991, Russian Federation.,Department of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701, Russian Federation
| | - Jonathan H P Dawes
- Department of Mathematical Sciences, University of Bath, Bath, BA2 7AY, UK
| | - Andrea Rocco
- Department of Microbial Sciences, FHMS, University of Surrey, Guildford, GU2 7XH, UK.,Department of Physics, FEPS, University of Surrey, Guildford, GU2 7XH, UK
| |
Collapse
|
11
|
Kato Y, Yoshida S, Kato T. New insights into the role and origin of pituitary S100β-positive cells. Cell Tissue Res 2021; 386:227-237. [PMID: 34550453 DOI: 10.1007/s00441-021-03523-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 09/07/2021] [Indexed: 01/16/2023]
Abstract
In the anterior pituitary, S100β protein (S100β) has been assumed to be a marker of folliculo-stellate cells, which are one of the non-hormone-producing cells existing in the parenchyma of the adult anterior lobe and are composed of subpopulations with various functions. However, recent accumulating studies on S100β-positive cells, including non-folliculo-stellate cells lining the marginal cell layer (MCL), have shown the novel aspect that most S100β-positive cells in the MCL and parenchyma of the adult anterior lobe are positive for sex determining region Y-box 2 (SOX2), a marker of pituitary stem/progenitor cells. From the viewpoint of SOX2-positive cells, the majority of these cells in the MCL and in the parenchyma are positive for S100β, suggesting that S100β plays a role in the large population of stem/progenitor cells in the anterior lobe of the adult pituitary. Reportedly, S100β/SOX2-double positive cells are able to differentiate into hormone-producing cells and various types of non-hormone-producing cells. Intriguingly, it has been demonstrated that extra-pituitary lineage cells invade the pituitary gland during prenatal pituitary organogenesis. Among them, two S100β-positive populations have been identified: one is SOX2-positive population which invades at the late embryonic period through the pituitary stalk and another is a SOX2-negative population that invades at the middle embryonic period through Atwell's recess. These two populations are likely the substantive origin of S100β-positive cells in the postnatal anterior pituitary, while S100β-positive cells emerging from oral ectoderm-derived cells remain unclear.
Collapse
Affiliation(s)
- Yukio Kato
- Institute for Endocrinology, Meiji University, 1-1-1 Higashi-mita, Tama-ku, Kawasaki, Kanagawa, 214-8571, Japan.
| | - Saishu Yoshida
- Department of Biochemistry, The Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Takako Kato
- Institute for Endocrinology, Meiji University, 1-1-1 Higashi-mita, Tama-ku, Kawasaki, Kanagawa, 214-8571, Japan
| |
Collapse
|
12
|
Wang YF, Liu C, Xu PF. Deciphering and reconstitution of positional information in the human brain development. ACTA ACUST UNITED AC 2021; 10:29. [PMID: 34467458 PMCID: PMC8408296 DOI: 10.1186/s13619-021-00091-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/02/2021] [Indexed: 12/29/2022]
Abstract
Organoid has become a novel in vitro model to research human development and relevant disorders in recent years. With many improvements on the culture protocols, current brain organoids could self-organize into a complicated three-dimensional organization that mimics most of the features of the real human brain at the molecular, cellular, and further physiological level. However, lacking positional information, an important characteristic conveyed by gradients of signaling molecules called morphogens, leads to the deficiency of spatiotemporally regulated cell arrangements and cell–cell interactions in the brain organoid development. In this review, we will overview the role of morphogen both in the vertebrate neural development in vivo as well as the brain organoid culture in vitro, the strategies to apply morphogen concentration gradients in the organoid system and future perspectives of the brain organoid technology.
Collapse
Affiliation(s)
- Yi-Fan Wang
- Women's Hospital, and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Institute of Zhejiang University and University of Edinburgh, Jiaxing, Zhejiang, China.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Dr, Singapore, 117599, Singapore
| | - Cong Liu
- Women's Hospital, and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Peng-Fei Xu
- Women's Hospital, and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
13
|
Ofek S, Wiszniak S, Kagan S, Tondl M, Schwarz Q, Kalcheim C. Notch signaling is a critical initiator of roof plate formation as revealed by the use of RNA profiling of the dorsal neural tube. BMC Biol 2021; 19:84. [PMID: 33892704 PMCID: PMC8063321 DOI: 10.1186/s12915-021-01014-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/25/2021] [Indexed: 12/31/2022] Open
Abstract
Background The dorsal domain of the neural tube is an excellent model to investigate the generation of complexity during embryonic development. It is a highly dynamic and multifaceted region being first transiently populated by prospective neural crest (NC) cells that sequentially emigrate to generate most of the peripheral nervous system. Subsequently, it becomes the definitive roof plate (RP) of the central nervous system. The RP, in turn, constitutes a patterning center for dorsal interneuron development. The factors underlying establishment of the definitive RP and its segregation from NC and dorsal interneurons are currently unknown. Results We performed a transcriptome analysis at trunk levels of quail embryos comparing the dorsal neural tube at premigratory NC and RP stages. This unraveled molecular heterogeneity between NC and RP stages, and within the RP itself. By implementing these genes, we asked whether Notch signaling is involved in RP development. First, we observed that Notch is active at the RP-interneuron interface. Furthermore, gain and loss of Notch function in quail and mouse embryos, respectively, revealed no effect on early NC behavior. Constitutive Notch activation caused a local downregulation of RP markers with a concomitant development of dI1 interneurons, as well as an ectopic upregulation of RP markers in the interneuron domain. Reciprocally, in mice lacking Notch activity, both the RP and dI1 interneurons failed to form and this was associated with expansion of the dI2 population. Conclusions Collectively, our results offer a new resource for defining specific cell types, and provide evidence that Notch is required to establish the definitive RP, and to determine the choice between RP and interneuron fates, but not the segregation of RP from NC.
Collapse
Affiliation(s)
- Shai Ofek
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University of Jerusalem-Hadassah Medical School, P.O.Box 12272, 9112102, Jerusalem, Israel
| | - Sophie Wiszniak
- Centre for Cancer Biology, University of South Australia and SA Pathology, North Terrace, Adelaide, SA, 5001, Australia
| | - Sarah Kagan
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University of Jerusalem-Hadassah Medical School, P.O.Box 12272, 9112102, Jerusalem, Israel
| | - Markus Tondl
- Centre for Cancer Biology, University of South Australia and SA Pathology, North Terrace, Adelaide, SA, 5001, Australia
| | - Quenten Schwarz
- Centre for Cancer Biology, University of South Australia and SA Pathology, North Terrace, Adelaide, SA, 5001, Australia.
| | - Chaya Kalcheim
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University of Jerusalem-Hadassah Medical School, P.O.Box 12272, 9112102, Jerusalem, Israel.
| |
Collapse
|
14
|
Rekler D, Kalcheim C. From Neural Crest to Definitive Roof Plate: The Dynamic Behavior of the Dorsal Neural Tube. Int J Mol Sci 2021; 22:3911. [PMID: 33920095 PMCID: PMC8070085 DOI: 10.3390/ijms22083911] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 01/11/2023] Open
Abstract
Research on the development of the dorsal neural tube is particularly challenging. In this highly dynamic domain, a temporal transition occurs between early neural crest progenitors that undergo an epithelial-to-mesenchymal transition and exit the neural primordium, and the subsequent roof plate, a resident epithelial group of cells that constitutes the dorsal midline of the central nervous system. Among other functions, the roof plate behaves as an organizing center for the generation of dorsal interneurons. Despite extensive knowledge of the formation, emigration and migration of neural crest progenitors, little is known about the mechanisms leading to the end of neural crest production and the transition into a roof plate stage. Are these two mutually dependent or autonomously regulated processes? Is the generation of roof plate and dorsal interneurons induced by neural tube-derived factors throughout both crest and roof plate stages, respectively, or are there differences in signaling properties and responsiveness as a function of time? In this review, we discuss distinctive characteristics of each population and possible mechanisms leading to the shift between the above cell types.
Collapse
Affiliation(s)
| | - Chaya Kalcheim
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University of Jerusalem-Hadassah Medical School, P.O.Box 12272, Jerusalem 9112102, Israel;
| |
Collapse
|
15
|
Yu L, Peng F, Dong X, Chen Y, Sun D, Jiang S, Deng C. Sex-Determining Region Y Chromosome-Related High-Mobility-Group Box 10 in Cancer: A Potential Therapeutic Target. Front Cell Dev Biol 2020; 8:564740. [PMID: 33344444 PMCID: PMC7744619 DOI: 10.3389/fcell.2020.564740] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 11/17/2020] [Indexed: 01/20/2023] Open
Abstract
Sex-determining region Y-related high mobility group-box 10 (SOX10), a member of the SOX family, has recently been highlighted as an essential transcriptional factor involved in developmental biology. Recently, the functionality of SOX 10 has been increasingly revealed by researchers worldwide. It has been reported that SOX10 significantly regulates the proliferation, migration, and apoptosis of tumors and is closely associated with the progression of cancer. In this review, we first introduce the basic background of the SOX family and SOX10 and then discuss the pathophysiological roles of SOX10 in cancer. Besides, we enumerate the application of SOX10 in the pathological diagnosis and therapeutic potential of cancer. Eventually, we summarize the potential directions and perspectives of SOX10 in neoplastic theranostics. The information compiled herein may assist in additional studies and increase the potential of SOX10 as a therapeutic target for cancer.
Collapse
Affiliation(s)
- Liming Yu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Fan Peng
- Department of Cardiology, Xijing Hopspital, The Airforce Military Medical University, Xi'an, China
| | - Xue Dong
- Outpatient Department of Liaoning Military Region, General Hospital of Northern Theater Command, Shenyang, China
| | - Ying Chen
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Dongdong Sun
- Department of Cardiology, Xijing Hopspital, The Airforce Military Medical University, Xi'an, China
| | - Shuai Jiang
- Department of Cardiology, Xijing Hopspital, The Airforce Military Medical University, Xi'an, China
| | - Chao Deng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
16
|
Abstract
Investigations of the cellular and molecular mechanisms that mediate the development of the autonomic nervous system have identified critical genes and signaling pathways that, when disrupted, cause disorders of the autonomic nervous system. This review summarizes our current understanding of how the autonomic nervous system emerges from the organized spatial and temporal patterning of precursor cell migration, proliferation, communication, and differentiation, and discusses potential clinical implications for developmental disorders of the autonomic nervous system, including familial dysautonomia, Hirschsprung disease, Rett syndrome, and congenital central hypoventilation syndrome.
Collapse
Affiliation(s)
- Frances Lefcort
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, Montana
| |
Collapse
|
17
|
Teixeira BL, Amarante-Silva D, Visoni SB, Garcez RC, Trentin AG. FGF2 Stimulates the Growth and Improves the Melanocytic Commitment of Trunk Neural Crest Cells. Cell Mol Neurobiol 2020; 40:383-393. [PMID: 31555941 PMCID: PMC11448768 DOI: 10.1007/s10571-019-00738-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 09/14/2019] [Indexed: 12/13/2022]
Abstract
Neural crest cells (NCCs) comprise a population of multipotent progenitors and stem cells at the origin of the peripheral nervous system (PNS) and melanocytes of skin, which are profoundly influenced by microenvironmental factors, among which is basic fibroblast growth factor 2 (FGF2). In this work, we further investigated the role of this growth factor in quail trunk NC morphogenesis and demonstrated its huge effect in NCC growth mainly by stimulating cell proliferation but also reducing cell death, despite that NCC migration from the neural tube explant was not affected. Moreover, following FGF2 treatment, reduced expression of the early NC markers Sox10 and FoxD3 and improved proliferation of HNK1-positive NCC were observed. Since these markers are involved in the regulation of glial and melanocytic fate of NC, the effect of FGF2 on NCC differentiation was investigated. Therefore, in the presence of FGF2, increased proportions of NCCs positives to the melanoblast marker Mitf as well as NCCs double stained to Mitf and BrdU were recorded. In addition, treatment with FGF2, followed by differentiation medium, resulted in increased expression of melanin and improved proportion of melanin-pigmented melanocytes without alteration in the glial marker Schwann myelin protein (SMP). Taken together, these data further reveal the important role of FGF2 in NCC proliferation, survival, and differentiation, particularly in melanocyte development. This is the first demonstration of FGF2 effects in melanocyte commitment of NC and in the proliferation of Mitf-positive melanoblasts. Elucidating the differentiation process of embryonic NCCs brings us a step closer to understanding the development of the PNS and then undertaking the search for advanced technologies to prevent, or treat, injuries caused by NC-related disorders, also known as neurocristopathies.
Collapse
Affiliation(s)
- Bianca Luise Teixeira
- Department of Cell Biology, Embryology and Genetics, Federal University of Santa Catarina, Florianopolis-SC, Campus Universitário,Trindade, Florianópolis, SC, 88040-900, Brazil
| | - Diego Amarante-Silva
- Department of Cell Biology, Embryology and Genetics, Federal University of Santa Catarina, Florianopolis-SC, Campus Universitário,Trindade, Florianópolis, SC, 88040-900, Brazil
| | - Silvia Beatriz Visoni
- Department of Cell Biology, Embryology and Genetics, Federal University of Santa Catarina, Florianopolis-SC, Campus Universitário,Trindade, Florianópolis, SC, 88040-900, Brazil
| | - Ricardo Castilho Garcez
- Department of Cell Biology, Embryology and Genetics, Federal University of Santa Catarina, Florianopolis-SC, Campus Universitário,Trindade, Florianópolis, SC, 88040-900, Brazil
| | - Andrea Gonçalves Trentin
- Department of Cell Biology, Embryology and Genetics, Federal University of Santa Catarina, Florianopolis-SC, Campus Universitário,Trindade, Florianópolis, SC, 88040-900, Brazil.
- National Institute of Science and Technology for Regenerative Medicine, Av. Carlos Chagas Filho, n°373, Rio De Janeiro, RJ, CEP: 21941902, Brazil.
| |
Collapse
|
18
|
Xi Y, Wang L, Liu H, Ma S, Li Y, Li L, Wang J, Chunchun H, Bai L, Mustafa A, He H. A 14-bp insertion in endothelin receptor B-like (EDNRB2) is associated with white plumage in Chinese geese. BMC Genomics 2020; 21:162. [PMID: 32066369 PMCID: PMC7027040 DOI: 10.1186/s12864-020-6562-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 02/07/2020] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Gang goose is a native species with gray plumage in Sichuan, China. As a result of overhunting, the number of gray Gang geese has decreased dramatically. To keep the species from extinction, conservation work for Gang geese was undertaken. In the process of pure breeding of gray Gang geese, approximately 2% of the offspring of each generation were white. This study aims to explain the genetic mechanism of this phenomenon and provide reliable molecular markers for goose-related plumage color breeding. RESULTS We used the method of pooled whole genome sequencing and Fst (fixation statistics) to identify the differentiation degree of alleles between gray Gang geese and white Gang geese from their offspring. In this way, EDNRB2, a key gene that affects the migration of melanoblasts, was identified. Then, the transcriptome was sequenced for the two geese plumage color populations, and the DEGs (differentially expressed genes) were analyzed. The results indicated that EDNRB2, as a possible candidate gene, had a significantly differential mRNA expression. In addition, a 14-bp insertion (NW_013185915.1: g. 750,748-750,735 insertion. CACAGGTGAGCTCT) in exon 3 of EDNRB2 was analyzed and found to have a significant association between gray geese and Chinese white breeds (P = 0.00), while this mutation was not found in European geese. Meanwhile, the insertion was homozygous in all the white geese we detected and heterozygous in gray geese, indicating that this mutation is recessive. Furthermore, this 14-bp insertion leads to a frameshift mutation in the EDNRB2 coding region and nonsense-mediated mRNA decay. CONCLUSION Our study strongly suggests that the 14-bp insertion in exon 3 of the EDNRB2 gene is associated with the white plumage phenotype in Chinese geese. This study is the first to investigate the relationship between EDNRB2 and white plumage in geese.
Collapse
Affiliation(s)
- Yang Xi
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, People’s Republic of China
| | - Lei Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, People’s Republic of China
| | - Hehe Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, People’s Republic of China
| | - Shengchao Ma
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, People’s Republic of China
| | - Yanying Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, People’s Republic of China
| | - Liang Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, People’s Republic of China
| | - Jiwen Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, People’s Republic of China
| | - Han Chunchun
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, People’s Republic of China
| | - Lili Bai
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, People’s Republic of China
| | - Ahsan Mustafa
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, People’s Republic of China
| | - Hua He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, People’s Republic of China
| |
Collapse
|
19
|
Mehrotra P, Tseropoulos G, Bronner ME, Andreadis ST. Adult tissue-derived neural crest-like stem cells: Sources, regulatory networks, and translational potential. Stem Cells Transl Med 2019; 9:328-341. [PMID: 31738018 PMCID: PMC7031649 DOI: 10.1002/sctm.19-0173] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 10/22/2019] [Accepted: 10/25/2019] [Indexed: 12/15/2022] Open
Abstract
Neural crest (NC) cells are a multipotent stem cell population that give rise to a diverse array of cell types in the body, including peripheral neurons, Schwann cells (SC), craniofacial cartilage and bone, smooth muscle cells, and melanocytes. NC formation and differentiation into specific lineages takes place in response to a set of highly regulated signaling and transcriptional events within the neural plate border. Premigratory NC cells initially are contained within the dorsal neural tube from which they subsequently emigrate, migrating to often distant sites in the periphery. Following their migration and differentiation, some NC‐like cells persist in adult tissues in a nascent multipotent state, making them potential candidates for autologous cell therapy. This review discusses the gene regulatory network responsible for NC development and maintenance of multipotency. We summarize the genes and signaling pathways that have been implicated in the differentiation of a postmigratory NC into mature myelinating SC. We elaborate on the signals and transcription factors involved in the acquisition of immature SC fate, axonal sorting of unmyelinated neuronal axons, and finally the path toward mature myelinating SC, which envelope axons within myelin sheaths, facilitating electrical signal propagation. The gene regulatory events guiding development of SC in vivo provides insights into means for differentiating NC‐like cells from adult human tissues into functional SC, which have the potential to provide autologous cell sources for the treatment of demyelinating and neurodegenerative disorders.
Collapse
Affiliation(s)
- Pihu Mehrotra
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, New York
| | - Georgios Tseropoulos
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, New York
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California
| | - Stelios T Andreadis
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, New York.,Center of Excellence in Bioinformatics and Life Sciences, Buffalo, New York.,Department of Biomedical Engineering, University at Buffalo, Buffalo, New York
| |
Collapse
|
20
|
Williams RM, Candido-Ferreira I, Repapi E, Gavriouchkina D, Senanayake U, Ling ITC, Telenius J, Taylor S, Hughes J, Sauka-Spengler T. Reconstruction of the Global Neural Crest Gene Regulatory Network In Vivo. Dev Cell 2019; 51:255-276.e7. [PMID: 31639368 PMCID: PMC6838682 DOI: 10.1016/j.devcel.2019.10.003] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 05/31/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023]
Abstract
Precise control of developmental processes is encoded in the genome in the form of gene regulatory networks (GRNs). Such multi-factorial systems are difficult to decode in vertebrates owing to their complex gene hierarchies and dynamic molecular interactions. Here we present a genome-wide in vivo reconstruction of the GRN underlying development of the multipotent neural crest (NC) embryonic cell population. By coupling NC-specific epigenomic and transcriptional profiling at population and single-cell levels with genome/epigenome engineering in vivo, we identify multiple regulatory layers governing NC ontogeny, including NC-specific enhancers and super-enhancers, novel trans-factors, and cis-signatures allowing reverse engineering of the NC-GRN at unprecedented resolution. Furthermore, identification and dissection of divergent upstream combinatorial regulatory codes has afforded new insights into opposing gene circuits that define canonical and neural NC fates early during NC ontogeny. Our integrated approach, allowing dissection of cell-type-specific regulatory circuits in vivo, has broad implications for GRN discovery and investigation.
Collapse
Affiliation(s)
- Ruth M Williams
- University of Oxford, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford OX3 9DS, UK
| | - Ivan Candido-Ferreira
- University of Oxford, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford OX3 9DS, UK
| | - Emmanouela Repapi
- University of Oxford, MRC Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, Oxford OX3 9DS, UK
| | - Daria Gavriouchkina
- University of Oxford, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford OX3 9DS, UK
| | - Upeka Senanayake
- University of Oxford, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford OX3 9DS, UK
| | - Irving T C Ling
- University of Oxford, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford OX3 9DS, UK; University of Oxford, Department of Paediatric Surgery, Children's Hospital Oxford, Oxford, UK
| | - Jelena Telenius
- University of Oxford, MRC Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, Oxford OX3 9DS, UK; University of Oxford, MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford OX3 9DS, UK
| | - Stephen Taylor
- University of Oxford, MRC Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, Oxford OX3 9DS, UK
| | - Jim Hughes
- University of Oxford, MRC Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, Oxford OX3 9DS, UK; University of Oxford, MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford OX3 9DS, UK
| | - Tatjana Sauka-Spengler
- University of Oxford, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford OX3 9DS, UK.
| |
Collapse
|
21
|
Wnt Signaling in Neural Crest Ontogenesis and Oncogenesis. Cells 2019; 8:cells8101173. [PMID: 31569501 PMCID: PMC6829301 DOI: 10.3390/cells8101173] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 02/06/2023] Open
Abstract
Neural crest (NC) cells are a temporary population of multipotent stem cells that generate a diverse array of cell types, including craniofacial bone and cartilage, smooth muscle cells, melanocytes, and peripheral neurons and glia during embryonic development. Defective neural crest development can cause severe and common structural birth defects, such as craniofacial anomalies and congenital heart disease. In the early vertebrate embryos, NC cells emerge from the dorsal edge of the neural tube during neurulation and then migrate extensively throughout the anterior-posterior body axis to generate numerous derivatives. Wnt signaling plays essential roles in embryonic development and cancer. This review summarizes current understanding of Wnt signaling in NC cell induction, delamination, migration, multipotency, and fate determination, as well as in NC-derived cancers.
Collapse
|
22
|
Kumar D, Nitzan E, Kalcheim C. YAP promotes neural crest emigration through interactions with BMP and Wnt activities. Cell Commun Signal 2019; 17:69. [PMID: 31228951 PMCID: PMC6589182 DOI: 10.1186/s12964-019-0383-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 06/04/2019] [Indexed: 12/12/2022] Open
Abstract
Background Premigratory neural crest progenitors undergo an epithelial-to-mesenchymal transition and leave the neural tube as motile cells. Previously, we showed that BMP generates trunk neural crest emigration through canonical Wnt signaling which in turn stimulates G1/S transition. The molecular network underlying this process is, however, not yet completely deciphered. Yes-associated-protein (YAP), an effector of the Hippo pathway, controls various aspects of development including cell proliferation, migration, survival and differentiation. In this study, we examined the possible involvement of YAP in neural crest emigration and its relationship with BMP and Wnt. Methods We implemented avian embryos in which levels of YAP gene activity were either reduced or upregulated by in ovo plasmid electroporation, and monitored effects on neural crest emigration, survival and proliferation. Neural crest-derived sensory neuron and melanocyte development were assessed upon gain of YAP function. Imunohistochemistry was used to assess YAP expression. In addition, the activity of specific signaling pathways including YAP, BMP and Wnt was monitored with specific reporters. Results We find that the Hippo pathway transcriptional co-activator YAP is expressed and is active in premigratory crest of avian embryos. Gain of YAP function stimulates neural crest emigration in vivo, and attenuating YAP inhibits cell exit. This is associated with an accumulation of FoxD3-expressing cells in the dorsal neural tube, with reduced proliferation, and enhanced apoptosis. Furthermore, gain of YAP function inhibits differentiation of Islet-1-positive sensory neurons and augments the number of EdnrB2-positive melanocytes. Using specific in vivo reporters, we show that loss of YAP function in the dorsal neural tube inhibits BMP and Wnt activities whereas gain of YAP function stimulates these pathways. Reciprocally, inhibition of BMP and Wnt signaling by noggin or Xdd1, respectively, downregulates YAP activity. In addition, YAP-dependent stimulation of neural crest emigration is compromised upon inhibition of either BMP or Wnt activities. Together, our results suggest a positive bidirectional cross talk between these pathways. Conclusions Our data show that YAP is necessary for emigration of neural crest progenitors. In addition, they incorporate YAP signaling into a BMP/Wnt-dependent molecular network responsible for emigration of trunk-level neural crest.
Collapse
Affiliation(s)
- Deepak Kumar
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, P.O. Box 12272, 91120, Jerusalem, Israel
| | - Erez Nitzan
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, P.O. Box 12272, 91120, Jerusalem, Israel
| | - Chaya Kalcheim
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, P.O. Box 12272, 91120, Jerusalem, Israel.
| |
Collapse
|
23
|
Lukoseviciute M, Gavriouchkina D, Williams RM, Hochgreb-Hagele T, Senanayake U, Chong-Morrison V, Thongjuea S, Repapi E, Mead A, Sauka-Spengler T. From Pioneer to Repressor: Bimodal foxd3 Activity Dynamically Remodels Neural Crest Regulatory Landscape In Vivo. Dev Cell 2019; 47:608-628.e6. [PMID: 30513303 PMCID: PMC6286384 DOI: 10.1016/j.devcel.2018.11.009] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 08/15/2018] [Accepted: 10/31/2018] [Indexed: 02/06/2023]
Abstract
The neural crest (NC) is a transient embryonic stem cell-like population characterized by its multipotency and broad developmental potential. Here, we perform NC-specific transcriptional and epigenomic profiling of foxd3-mutant cells in vivo to define the gene regulatory circuits controlling NC specification. Together with global binding analysis obtained by foxd3 biotin-ChIP and single cell profiles of foxd3-expressing premigratory NC, our analysis shows that, during early steps of NC formation, foxd3 acts globally as a pioneer factor to prime the onset of genes regulating NC specification and migration by re-arranging the chromatin landscape, opening cis-regulatory elements and reshuffling nucleosomes. Strikingly, foxd3 then gradually switches from an activator to its well-described role as a transcriptional repressor and potentially uses differential partners for each role. Taken together, these results demonstrate that foxd3 acts bimodally in the neural crest as a switch from “permissive” to “repressive” nucleosome and chromatin organization to maintain multipotency and define cell fates. FoxD3 primes neural crest specification by modulating distal enhancers FoxD3 represses a number of neural crest migration and differentiation genes In neural crest, FoxD3 acts to switch chromatin from “permissive” to “repressive” Distinctive gene regulatory mechanisms underlie the bimodal action of FoxD3
Collapse
Affiliation(s)
- Martyna Lukoseviciute
- Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Daria Gavriouchkina
- Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Ruth M Williams
- Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Tatiana Hochgreb-Hagele
- Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Upeka Senanayake
- Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Vanessa Chong-Morrison
- Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Supat Thongjuea
- Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Emmanouela Repapi
- MRC WIMM Centre for Computational Biology Research Group, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Adam Mead
- Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Tatjana Sauka-Spengler
- Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK.
| |
Collapse
|
24
|
Abstract
Neural crest cells are the embryonic precursors of most neurons and all glia of the peripheral nervous system, pigment cells, some endocrine components, and connective tissue of the head, face, neck, and heart. Following induction, crest cells undergo an epithelial to mesenchymal transition that enables them to migrate along specific pathways culminating in their phenotypic differentiation. Researching this unique embryonic population has revealed important understandings of basic biological and developmental principles. These principles are likely to assist in clarifying the etiology and help in finding strategies for the treatment of neural crest diseases, collectively termed neurocristopathies. The progress achieved in neural crest research is made feasible thanks to the continuous development of species-specific in vivo and in vitro paradigms and more recently the possibility to produce neural crest cells and specific derivatives from embryonic or induced pluripotent stem cells. All of the above assist us in elucidating mechanisms that regulate neural crest development using state-of-the art cellular, molecular, and imaging approaches.
Collapse
Affiliation(s)
- Chaya Kalcheim
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC), Hebrew University-Hadassah Medical School, Jerusalem, Israel.
- Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University-Hadassah Medical School, Jerusalem, Israel.
| |
Collapse
|
25
|
Dupin E, Calloni GW, Coelho-Aguiar JM, Le Douarin NM. The issue of the multipotency of the neural crest cells. Dev Biol 2018; 444 Suppl 1:S47-S59. [DOI: 10.1016/j.ydbio.2018.03.024] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 03/29/2018] [Accepted: 03/30/2018] [Indexed: 12/25/2022]
|
26
|
Davis EL, Davis AR, Gugala Z, Olmsted-Davis EA. Is heterotopic ossification getting nervous?: The role of the peripheral nervous system in heterotopic ossification. Bone 2018; 109:22-27. [PMID: 28716552 PMCID: PMC5768468 DOI: 10.1016/j.bone.2017.07.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 07/12/2017] [Accepted: 07/13/2017] [Indexed: 12/28/2022]
Abstract
Heterotopic ossification (HO), or de novo bone formation in soft tissue, is often observed following traumatic injury. Recent studies suggest that peripheral nerves may play a key functional role in this process. The results supporting a neurological basis for HO are examined in this article. Evidence supports the fact that BMPs released from bone matrix possess the capacity to induce HO. However, the process cannot be recapitulated using recombinant proteins without extremely high doses suggesting other components are required for this process. Study of injuries that increase risk for HO, i.e. amputation, hip replacement, elbow fracture, burn, and CNS injury suggests that a likely candidate is traumatic injury of adjacent peripheral nerves. Recent studies suggest neuroinflammation may play a key functional role, by its ability to open the blood-nerve barrier (BNB). Barrier opening is characterized by a change in permeability and is experimentally assessed by the ability of Evans blue dye to enter the endoneurium of peripheral nerves. A combination of BMP and barrier opening is required to activate bone progenitors in the endoneurial compartment. This process is referred to as "neurogenic HO".
Collapse
Affiliation(s)
- Eleanor L Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX 77030, United States
| | - Alan R Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX 77030, United States; Department of Pediatrics - Section Hematology/Oncology, Baylor College of Medicine, Houston, TX 77030, United States; Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX 77030, United States
| | - Zbigniew Gugala
- Department of Orthopedic Surgery and Rehabilitation, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Elizabeth A Olmsted-Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX 77030, United States; Department of Pediatrics - Section Hematology/Oncology, Baylor College of Medicine, Houston, TX 77030, United States; Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX 77030, United States.
| |
Collapse
|
27
|
Adachi E, Sakai K, Nishiuchi T, Imamura R, Sato H, Matsumoto K. Different growth and metastatic phenotypes associated with a cell-intrinsic change of Met in metastatic melanoma. Oncotarget 2018; 7:70779-70793. [PMID: 27683122 PMCID: PMC5342589 DOI: 10.18632/oncotarget.12221] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 09/13/2016] [Indexed: 12/19/2022] Open
Abstract
A dynamic phenotypic change contributes to the metastatic progression and drug resistance in malignant melanoma. Nevertheless, mechanisms for a phenotypic change have remained to be addressed. Here, we show that Met receptor expression changes in a cell-autonomous manner and can distinguish phenotypical differences in growth, as well as in metastatic and drug-resistant characteristics. In metastatic melanoma, the cells are composed of Met-low and Met-high populations. Met-low populations have stem-like gene expression profiles, are resistant to chemotherapeutic agents, and have shown abundant angiogenesis and rapid tumor growth in subcutaneous inoculation. Met-high populations have a differentiated phenotype, are relatively resistant to B-RAF inhibitor, and are highly metastatic to the lungs. Met plays a definitive role in lung metastasis because the lung metastasis of Met-high cells requires Met, and treatment of mice with the Met-containing exosomes from Met-high cells facilitates lung metastasis by Met-low cells. Clonal cell fate analysis showed the hierarchical phenotypical changes from Met-low to Met-high populations. Met-low cells either showed self-renewal or changed into Met-high cells, whereas Met-high cells remained Met-high. Clonal transition from Met-low to Met-high cells accompanied changes in the gene expression profile, in tumor growth, and in metastasis that were similar to those in Met-high cells. These findings indicate that malignant melanoma has the ability to undergo phenotypic change by a cell-intrinsic/autonomous mechanism that can be characterized by Met expression.
Collapse
Affiliation(s)
- Eri Adachi
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kakuma, Kanazawa 920-1192, Japan
| | - Katsuya Sakai
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kakuma, Kanazawa 920-1192, Japan
| | - Takumi Nishiuchi
- Division of Functional Genomics, Advanced Science Research Center, Kanazawa University, Kakuma, Kanazawa 920-0934, Japan
| | - Ryu Imamura
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kakuma, Kanazawa 920-1192, Japan
| | - Hiroki Sato
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kakuma, Kanazawa 920-1192, Japan
| | - Kunio Matsumoto
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kakuma, Kanazawa 920-1192, Japan
| |
Collapse
|
28
|
Kalcheim C. Neural crest emigration: From start to stop. Genesis 2018; 56:e23090. [DOI: 10.1002/dvg.23090] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 01/07/2018] [Accepted: 01/08/2018] [Indexed: 12/28/2022]
Affiliation(s)
- Chaya Kalcheim
- Department of Medical Neurobiology, IMRIC and ELSC; Hebrew University of Jerusalem-Hadassah Medical School; Jerusalem 9112102 Israel
| |
Collapse
|
29
|
Kastriti ME, Adameyko I. Specification, plasticity and evolutionary origin of peripheral glial cells. Curr Opin Neurobiol 2017; 47:196-202. [DOI: 10.1016/j.conb.2017.11.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 08/30/2017] [Accepted: 11/03/2017] [Indexed: 12/31/2022]
|
30
|
Ueharu H, Yoshida S, Kanno N, Horiguchi K, Nishimura N, Kato T, Kato Y. SOX10-positive cells emerge in the rat pituitary gland during late embryogenesis and start to express S100β. Cell Tissue Res 2017; 372:77-90. [PMID: 29130118 DOI: 10.1007/s00441-017-2724-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 10/26/2017] [Indexed: 12/19/2022]
Abstract
In the pituitary gland, S100β-positive cells localize in the neurohypophysis and adenohypophysis but the lineage of the two groups remains obscure. S100β is often observed in many neural crest-derived cell types. Therefore, in this study, we investigate the origin of pituitary S100β-positive cells by immunohistochemistry for SOX10, a potent neural crest cell marker, using S100β-green fluorescence protein-transgenic rats. On embryonic day 21.5, a SOX10-positive cell population, which was also positive for the stem/progenitor cell marker SOX2, emerged in the pituitary stalk and posterior lobe and subsequently expanded to create a rostral-caudal gradient on postnatal day 3 (P3). Thereafter, SOX10-positive cells appeared in the intermediate lobe by P15, localizing to the boundary facing the posterior lobe, the gap between the lobule structures and the marginal cell layer, a pituitary stem/progenitor cell niche. Subsequently, there was an increase in SOX10/S100β double-positive cells; some of these cells in the gap between the lobule structures showed extended cytoplasm containing F-actin, indicating a feature of migration activity. The proportion of SOX10-positive cells in the postnatal anterior lobe was lower than 0.025% but about half of them co-localized with the pituitary-specific progenitor cell marker PROP1. Collectively, the present study identified that one of the lineages of S100β-positive cells is a SOX10-positive one and that SOX10-positive cells express pituitary stem/progenitor cell marker genes.
Collapse
Affiliation(s)
- Hiroki Ueharu
- Division of Life Science, Graduate School of Agriculture, Meiji University, Kawasaki, Kanagawa, 214-8571, Japan
| | - Saishu Yoshida
- Institute of Reproduction and Endocrinology, Meiji University, Tokyo, Kanagawa, 214-8571, Japan
| | - Naoko Kanno
- Division of Life Science, Graduate School of Agriculture, Meiji University, Kawasaki, Kanagawa, 214-8571, Japan
| | - Kotaro Horiguchi
- Institute of Reproduction and Endocrinology, Meiji University, Tokyo, Kanagawa, 214-8571, Japan.,Laboratory of Anatomy and Cell Biology, Faculty of Health Sciences, Kyorin University, Mitaka, Tokyo, 181-8612, Japan
| | - Naoto Nishimura
- Division of Life Science, Graduate School of Agriculture, Meiji University, Kawasaki, Kanagawa, 214-8571, Japan
| | - Takako Kato
- Institute of Reproduction and Endocrinology, Meiji University, Tokyo, Kanagawa, 214-8571, Japan
| | - Yukio Kato
- Division of Life Science, Graduate School of Agriculture, Meiji University, Kawasaki, Kanagawa, 214-8571, Japan. .,Institute of Reproduction and Endocrinology, Meiji University, Tokyo, Kanagawa, 214-8571, Japan. .,Department of Life Science, School of Agriculture, Meiji University, 1-1-1 Higashi-Mita, Tama-Ku, Kawasaki, Kanagawa, 214-8571, Japan.
| |
Collapse
|
31
|
Delfino-Machín M, Madelaine R, Busolin G, Nikaido M, Colanesi S, Camargo-Sosa K, Law EWP, Toppo S, Blader P, Tiso N, Kelsh RN. Sox10 contributes to the balance of fate choice in dorsal root ganglion progenitors. PLoS One 2017; 12:e0172947. [PMID: 28253350 PMCID: PMC5333849 DOI: 10.1371/journal.pone.0172947] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 02/12/2017] [Indexed: 11/19/2022] Open
Abstract
The development of functional peripheral ganglia requires a balance of specification of both neuronal and glial components. In the developing dorsal root ganglia (DRGs), these components form from partially-restricted bipotent neuroglial precursors derived from the neural crest. Work in mouse and chick has identified several factors, including Delta/Notch signaling, required for specification of a balance of these components. We have previously shown in zebrafish that the Sry-related HMG domain transcription factor, Sox10, plays an unexpected, but crucial, role in sensory neuron fate specification in vivo. In the same study we described a novel Sox10 mutant allele, sox10baz1, in which sensory neuron numbers are elevated above those of wild-types. Here we investigate the origin of this neurogenic phenotype. We demonstrate that the supernumerary neurons are sensory neurons, and that enteric and sympathetic neurons are almost absent just as in classical sox10 null alleles; peripheral glial development is also severely abrogated in a manner similar to other sox10 mutant alleles. Examination of proliferation and apoptosis in the developing DRG reveals very low levels of both processes in wild-type and sox10baz1, excluding changes in the balance of these as an explanation for the overproduction of sensory neurons. Using chemical inhibition of Delta-Notch-Notch signaling we demonstrate that in embryonic zebrafish, as in mouse and chick, lateral inhibition during the phase of trunk DRG development is required to achieve a balance between glial and neuronal numbers. Importantly, however, we show that this mechanism is insufficient to explain quantitative aspects of the baz1 phenotype. The Sox10(baz1) protein shows a single amino acid substitution in the DNA binding HMG domain; structural analysis indicates that this change is likely to result in reduced flexibility in the HMG domain, consistent with sequence-specific modification of Sox10 binding to DNA. Unlike other Sox10 mutant proteins, Sox10(baz1) retains an ability to drive neurogenin1 transcription. We show that overexpression of neurogenin1 is sufficient to produce supernumerary DRG sensory neurons in a wild-type background, and can rescue the sensory neuron phenotype of sox10 morphants in a manner closely resembling the baz1 phenotype. We conclude that an imbalance of neuronal and glial fate specification results from the Sox10(baz1) protein's unique ability to drive sensory neuron specification whilst failing to drive glial development. The sox10baz1 phenotype reveals for the first time that a Notch-dependent lateral inhibition mechanism is not sufficient to fully explain the balance of neurons and glia in the developing DRGs, and that a second Sox10-dependent mechanism is necessary. Sox10 is thus a key transcription factor in achieving the balance of sensory neuronal and glial fates.
Collapse
Affiliation(s)
- Mariana Delfino-Machín
- Department of Biology and Biochemistry and Centre for Regenerative Medicine, University of Bath, Bath, United Kingdom
| | - Romain Madelaine
- Centre de Biologie du Développement (CBD, UMR5547), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | | | - Masataka Nikaido
- Department of Biology and Biochemistry and Centre for Regenerative Medicine, University of Bath, Bath, United Kingdom
| | - Sarah Colanesi
- Department of Biology and Biochemistry and Centre for Regenerative Medicine, University of Bath, Bath, United Kingdom
| | - Karen Camargo-Sosa
- Department of Biology and Biochemistry and Centre for Regenerative Medicine, University of Bath, Bath, United Kingdom
| | - Edward W. P. Law
- Department of Biology and Biochemistry and Centre for Regenerative Medicine, University of Bath, Bath, United Kingdom
| | - Stefano Toppo
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Patrick Blader
- Centre de Biologie du Développement (CBD, UMR5547), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Natascia Tiso
- Department of Biology, University of Padova, Padova, Italy
| | - Robert N. Kelsh
- Department of Biology and Biochemistry and Centre for Regenerative Medicine, University of Bath, Bath, United Kingdom
| |
Collapse
|
32
|
Eroh GD, Clayton FC, Florell SR, Cassidy PB, Chirife A, Marón CF, Valenzuela LO, Campbell MS, Seger J, Rowntree VJ, Leachman SA. Cellular and ultrastructural characterization of the grey-morph phenotype in southern right whales (Eubalaena australis). PLoS One 2017; 12:e0171449. [PMID: 28170433 PMCID: PMC5295704 DOI: 10.1371/journal.pone.0171449] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/20/2017] [Indexed: 11/18/2022] Open
Abstract
Southern right whales (SRWs, Eubalena australis) are polymorphic for an X-linked pigmentation pattern known as grey morphism. Most SRWs have completely black skin with white patches on their bellies and occasionally on their backs; these patches remain white as the whale ages. Grey morphs (previously referred to as partial albinos) appear mostly white at birth, with a splattering of rounded black marks; but as the whales age, the white skin gradually changes to a brownish grey color. The cellular and developmental bases of grey morphism are not understood. Here we describe cellular and ultrastructural features of grey-morph skin in relation to that of normal, wild-type skin. Melanocytes were identified histologically and counted, and melanosomes were measured using transmission electron microscopy. Grey-morph skin had fewer melanocytes when compared to wild-type skin, suggesting reduced melanocyte survival, migration, or proliferation in these whales. Grey-morph melanocytes had smaller melanosomes relative to wild-type skin, normal transport of melanosomes to surrounding keratinocytes, and normal localization of melanin granules above the keratinocyte nuclei. These findings indicate that SRW grey-morph pigmentation patterns are caused by reduced numbers of melanocytes in the skin, as well as by reduced amounts of melanin production and/or reduced sizes of mature melanosomes. Grey morphism is distinct from piebaldism and albinism found in other species, which are genetic pigmentation conditions resulting from the local absence of melanocytes, or the inability to synthesize melanin, respectively.
Collapse
Affiliation(s)
- Guy D. Eroh
- Huntsman Cancer Institute, Salt Lake City, Utah, United States of America
- University of Georgia, Athens, Georgia, United States of America
| | - Fred C. Clayton
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Scott R. Florell
- Department of Dermatology, University of Utah, Salt Lake City, Utah, United States of America
| | - Pamela B. Cassidy
- Huntsman Cancer Institute, Salt Lake City, Utah, United States of America
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Andrea Chirife
- Programa de Monitoreo Sanitario Ballena Franca Austral, Puerto Madryn, Chubut, Argentina
| | - Carina F. Marón
- Department of Biology, University of Utah, Salt Lake City, Utah, United States of America
- Instituto de Conservación de Ballenas, Buenos Aires, Argentina
| | - Luciano O. Valenzuela
- Department of Biology, University of Utah, Salt Lake City, Utah, United States of America
- Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Sociales, Universidad Nacional del Centro de la Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Michael S. Campbell
- Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States of America
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, United States of America
| | - Jon Seger
- Department of Biology, University of Utah, Salt Lake City, Utah, United States of America
| | - Victoria J. Rowntree
- Programa de Monitoreo Sanitario Ballena Franca Austral, Puerto Madryn, Chubut, Argentina
- Department of Biology, University of Utah, Salt Lake City, Utah, United States of America
- Instituto de Conservación de Ballenas, Buenos Aires, Argentina
- Ocean Alliance/Whale Conservation Institute, Gloucester, Massachusetts, United States of America
| | - Sancy A. Leachman
- Huntsman Cancer Institute, Salt Lake City, Utah, United States of America
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon, United States of America
- * E-mail:
| |
Collapse
|
33
|
Substrate-mediated reprogramming of human fibroblasts into neural crest stem-like cells and their applications in neural repair. Biomaterials 2016; 102:148-61. [DOI: 10.1016/j.biomaterials.2016.06.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 06/07/2016] [Indexed: 11/22/2022]
|
34
|
Kaucka M, Ivashkin E, Gyllborg D, Zikmund T, Tesarova M, Kaiser J, Xie M, Petersen J, Pachnis V, Nicolis SK, Yu T, Sharpe P, Arenas E, Brismar H, Blom H, Clevers H, Suter U, Chagin AS, Fried K, Hellander A, Adameyko I. Analysis of neural crest-derived clones reveals novel aspects of facial development. SCIENCE ADVANCES 2016; 2:e1600060. [PMID: 27493992 PMCID: PMC4972470 DOI: 10.1126/sciadv.1600060] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 06/29/2016] [Indexed: 05/05/2023]
Abstract
Cranial neural crest cells populate the future facial region and produce ectomesenchyme-derived tissues, such as cartilage, bone, dermis, smooth muscle, adipocytes, and many others. However, the contribution of individual neural crest cells to certain facial locations and the general spatial clonal organization of the ectomesenchyme have not been determined. We investigated how neural crest cells give rise to clonally organized ectomesenchyme and how this early ectomesenchyme behaves during the developmental processes that shape the face. Using a combination of mouse and zebrafish models, we analyzed individual migration, cell crowd movement, oriented cell division, clonal spatial overlapping, and multilineage differentiation. The early face appears to be built from multiple spatially defined overlapping ectomesenchymal clones. During early face development, these clones remain oligopotent and generate various tissues in a given location. By combining clonal analysis, computer simulations, mouse mutants, and live imaging, we show that facial shaping results from an array of local cellular activities in the ectomesenchyme. These activities mostly involve oriented divisions and crowd movements of cells during morphogenetic events. Cellular behavior that can be recognized as individual cell migration is very limited and short-ranged and likely results from cellular mixing due to the proliferation activity of the tissue. These cellular mechanisms resemble the strategy behind limb bud morphogenesis, suggesting the possibility of common principles and deep homology between facial and limb outgrowth.
Collapse
Affiliation(s)
- Marketa Kaucka
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Evgeny Ivashkin
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm SE-171 77, Sweden
- Research Center of Neurology, 125367 Moscow, Russia
| | - Daniel Gyllborg
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Tomas Zikmund
- Central European Institute of Technology, Brno University of Technology, 616 00 Brno, Czech Republic
| | - Marketa Tesarova
- Central European Institute of Technology, Brno University of Technology, 616 00 Brno, Czech Republic
| | - Jozef Kaiser
- Central European Institute of Technology, Brno University of Technology, 616 00 Brno, Czech Republic
| | - Meng Xie
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Julian Petersen
- Department of Molecular Neurosciences, Medical University of Vienna, Vienna 1190, Austria
| | - Vassilis Pachnis
- Division of Molecular Neurobiology, Medical Research Council National Institute for Medical Research, London NW7 1AA, UK
| | - Silvia K. Nicolis
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milano, Italy
| | - Tian Yu
- Department of Craniofacial Development and Stem Cell Biology, King’s College London Dental Institute, Guy’s Hospital, London SE1 9RT, UK
| | - Paul Sharpe
- Department of Craniofacial Development and Stem Cell Biology, King’s College London Dental Institute, Guy’s Hospital, London SE1 9RT, UK
| | - Ernest Arenas
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Hjalmar Brismar
- Science for Life Laboratory, Royal Institute of Technology, Solna 17121, Sweden
| | - Hans Blom
- Science for Life Laboratory, Royal Institute of Technology, Solna 17121, Sweden
| | - Hans Clevers
- Hubrecht Institute of the Royal Netherlands Academy of Arts and Sciences, Princess Maxima Centre and University Medical Centre Utrecht, 3584 Utrecht, Netherlands
| | - Ueli Suter
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich CH-8093, Switzerland
| | - Andrei S. Chagin
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Kaj Fried
- Department of Neuroscience, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Andreas Hellander
- Department of Information Technology, Uppsala University, Uppsala SE-751 05, Sweden
- Corresponding author. (I.A.); (A.H.)
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm SE-171 77, Sweden
- Department of Molecular Neurosciences, Medical University of Vienna, Vienna 1190, Austria
- Corresponding author. (I.A.); (A.H.)
| |
Collapse
|
35
|
Agarwalla PK, Koch MJ, Mordes DA, Codd PJ, Coumans JV. Pigmented Lesions of the Nervous System and the Neural Crest: Lessons From Embryology. Neurosurgery 2016; 78:142-55. [PMID: 26355366 DOI: 10.1227/neu.0000000000001010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Neurosurgeons encounter a number of pigmented tumors of the central nervous system in a variety of locations, including primary central nervous system melanoma, blue nevus of the spinal cord, and melanotic schwannoma. When examined through the lens of embryology, pigmented lesions share a unifying connection: They occur in structures that are neural crest cell derivatives. Here, we review the important progress made in the embryology of neural crest cells, present 3 cases of pigmented tumors of the nervous system, and discuss these clinical entities in the context of the development of melanoblasts. Pigmented lesions of the nervous system arise along neural crest cell migration routes and from neural crest-derived precursors. Awareness of the evolutionary clues of vertebrate pigmentation by the neurosurgical and neuro-oncological community at large is valuable for identifying pathogenic or therapeutic targets and for designing future research on nervous system pigmented lesions. When encountering such a lesion, clinicians should be aware of the embryological basis to direct additional evaluation, including genetic testing, and to work with the scientific community in better understanding these lesions and their relationship to neural crest developmental biology.
Collapse
Affiliation(s)
- Pankaj K Agarwalla
- Departments of *Neurosurgery and‡Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | | | | | |
Collapse
|
36
|
Nitzan E, Avraham O, Kahane N, Ofek S, Kumar D, Kalcheim C. Dynamics of BMP and Hes1/Hairy1 signaling in the dorsal neural tube underlies the transition from neural crest to definitive roof plate. BMC Biol 2016; 14:23. [PMID: 27012662 PMCID: PMC4806459 DOI: 10.1186/s12915-016-0245-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/10/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The dorsal midline region of the neural tube that results from closure of the neural folds is generally termed the roof plate (RP). However, this domain is highly dynamic and complex, and is first transiently inhabited by prospective neural crest (NC) cells that sequentially emigrate from the neuroepithelium. It only later becomes the definitive RP, the dorsal midline cells of the spinal cord. We previously showed that at the trunk level of the axis, prospective RP progenitors originate ventral to the premigratory NC and progressively reach the dorsal midline following NC emigration. However, the molecular mechanisms underlying the end of NC production and formation of the definitive RP remain virtually unknown. RESULTS Based on distinctive cellular and molecular traits, we have defined an initial NC and a subsequent RP stage, allowing us to investigate the mechanisms responsible for the transition between the two phases. We demonstrate that in spite of the constant production of BMP4 in the dorsal tube at both stages, RP progenitors only transiently respond to the ligand and lose competence shortly before they arrive at their final location. In addition, exposure of dorsal tube cells at the NC stage to high levels of BMP signaling induces premature RP traits, such as Hes1/Hairy1, while concomitantly inhibiting NC production. Reciprocally, early inhibition of BMP signaling prevents Hairy1 mRNA expression at the RP stage altogether, suggesting that BMP is both necessary and sufficient for the development of this RP-specific trait. Furthermore, when Hes1/Hairy1 is misexpressed at the NC stage, it inhibits BMP signaling and downregulates BMPR1A/Alk3 mRNA expression, transcription of BMP targets such as Foxd3, cell-cycle progression, and NC emigration. Reciprocally, Foxd3 inhibits Hairy1, suggesting that repressive cross-interactions at the level of, and downstream from, BMP ensure the temporal separation between both lineages. CONCLUSIONS Together, our data suggest that BMP signaling is important both for NC and RP formation. Given that these two structures develop sequentially, we speculate that the longer exposure of RP progenitors to BMP compared with that of premigratory NC cells may be translated into a higher signaling level in the former. This induces changes in responsiveness to BMP, most likely by downregulating the expression of Alk3 receptors and, consequently, of BMP-dependent downstream transcription factors, which exhibit spatial complementary expression patterns and mutually repress each other to generate alternative fates. This molecular dynamic is likely to account for the transition between the NC and definitive RP stages and thus be responsible for the segregation between central and peripheral lineages during neural development.
Collapse
Affiliation(s)
- Erez Nitzan
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, 9112102,, PO Box 12272,, Israel.,Present Address: Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Oshri Avraham
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, 9112102,, PO Box 12272,, Israel.,Present address: Department of Genetics, Washington University, St. Louis, MO, USA
| | - Nitza Kahane
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, 9112102,, PO Box 12272,, Israel
| | - Shai Ofek
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, 9112102,, PO Box 12272,, Israel
| | - Deepak Kumar
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, 9112102,, PO Box 12272,, Israel
| | - Chaya Kalcheim
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, 9112102,, PO Box 12272,, Israel.
| |
Collapse
|
37
|
Sanchez-Ferras O, Bernas G, Farnos O, Touré AM, Souchkova O, Pilon N. A direct role for murine Cdx proteins in the trunk neural crest gene regulatory network. Development 2016; 143:1363-74. [PMID: 26952979 DOI: 10.1242/dev.132159] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/18/2016] [Indexed: 12/28/2022]
Abstract
Numerous studies in chordates and arthropods currently indicate that Cdx proteins have a major ancestral role in the organization of post-head tissues. In urochordate embryos, Cdx loss-of-function has been shown to impair axial elongation, neural tube (NT) closure and pigment cell development. Intriguingly, in contrast to axial elongation and NT closure, a Cdx role in neural crest (NC)-derived melanocyte/pigment cell development has not been reported in any other chordate species. To address this, we generated a new conditional pan-Cdx functional knockdown mouse model that circumvents Cdx functional redundancy as well as the early embryonic lethality of Cdx mutants. Through directed inhibition in the neuroectoderm, we provide in vivo evidence that murine Cdx proteins impact melanocyte and enteric nervous system development by, at least in part, directly controlling the expression of the key early regulators of NC ontogenesis Pax3,Msx1 and Foxd3 Our work thus reveals a novel role for Cdx proteins at the top of the trunk NC gene regulatory network in the mouse, which appears to have been inherited from their ancestral ortholog.
Collapse
Affiliation(s)
- Oraly Sanchez-Ferras
- Molecular Genetics of Development Laboratory, Department of Biological Sciences and BioMed Research Center, University of Quebec at Montreal (UQAM), Montreal H2X 3Y7, Canada
| | - Guillaume Bernas
- Molecular Genetics of Development Laboratory, Department of Biological Sciences and BioMed Research Center, University of Quebec at Montreal (UQAM), Montreal H2X 3Y7, Canada
| | - Omar Farnos
- Molecular Genetics of Development Laboratory, Department of Biological Sciences and BioMed Research Center, University of Quebec at Montreal (UQAM), Montreal H2X 3Y7, Canada
| | - Aboubacrine M Touré
- Molecular Genetics of Development Laboratory, Department of Biological Sciences and BioMed Research Center, University of Quebec at Montreal (UQAM), Montreal H2X 3Y7, Canada
| | - Ouliana Souchkova
- Molecular Genetics of Development Laboratory, Department of Biological Sciences and BioMed Research Center, University of Quebec at Montreal (UQAM), Montreal H2X 3Y7, Canada
| | - Nicolas Pilon
- Molecular Genetics of Development Laboratory, Department of Biological Sciences and BioMed Research Center, University of Quebec at Montreal (UQAM), Montreal H2X 3Y7, Canada
| |
Collapse
|
38
|
Le Douarin NM, Dupin E. The Pluripotency of Neural Crest Cells and Their Role in Brain Development. Curr Top Dev Biol 2016; 116:659-78. [PMID: 26970647 DOI: 10.1016/bs.ctdb.2015.10.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The neural crest (NC) is, in the Chordate phylum, an innovation of vertebrates, which exhibits several original characteristics: its component cells are pluripotent and give rise to both ectodermal and mesodermal cell types. Moreover, during the early stages of neurogenesis, the NC cells exert a paracrine stimulating effect on the development of the preotic brain.
Collapse
Affiliation(s)
- Nicole M Le Douarin
- Collège de France, 3 rue d'Ulm, Paris, France; INSERM U968, Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR S 968, Institut de la Vision, Paris, France; CNRS, UMR 7210, Paris, France.
| | - Elisabeth Dupin
- INSERM U968, Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR S 968, Institut de la Vision, Paris, France; CNRS, UMR 7210, Paris, France
| |
Collapse
|
39
|
Reconciling diverse mammalian pigmentation patterns with a fundamental mathematical model. Nat Commun 2016; 7:10288. [PMID: 26732977 PMCID: PMC4729835 DOI: 10.1038/ncomms10288] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 11/26/2015] [Indexed: 12/18/2022] Open
Abstract
Bands of colour extending laterally from the dorsal to ventral trunk are a common feature of mouse chimeras. These stripes were originally taken as evidence of the directed dorsoventral migration of melanoblasts (the embryonic precursors of melanocytes) as they colonize the developing skin. Depigmented ‘belly spots' in mice with mutations in the receptor tyrosine kinase Kit are thought to represent a failure of this colonization, either due to impaired migration or proliferation. Tracing of single melanoblast clones, however, has revealed a diffuse distribution with high levels of axial mixing—hard to reconcile with directed migration. Here we construct an agent-based stochastic model calibrated by experimental measurements to investigate the formation of diffuse clones, chimeric stripes and belly spots. Our observations indicate that melanoblast colonization likely proceeds through a process of undirected migration, proliferation and tissue expansion, and that reduced proliferation is the cause of the belly spots in Kit mutants. How embryonic melanoblast behaviour influences adult pigmentation patterns and causes patterning defects is unclear. Here, Mort et al. construct a stochastic model parameterised experimentally to show that melanoblast migration is undirected and that reduced proliferation causes patterning defects.
Collapse
|
40
|
Kalcheim C. Epithelial-Mesenchymal Transitions during Neural Crest and Somite Development. J Clin Med 2015; 5:jcm5010001. [PMID: 26712793 PMCID: PMC4730126 DOI: 10.3390/jcm5010001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 12/09/2015] [Accepted: 12/14/2015] [Indexed: 01/14/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a central process during embryonic development that affects selected progenitor cells of all three germ layers. In addition to driving the onset of cellular migrations and subsequent tissue morphogenesis, the dynamic conversions of epithelium into mesenchyme and vice-versa are intimately associated with the segregation of homogeneous precursors into distinct fates. The neural crest and somites, progenitors of the peripheral nervous system and of skeletal tissues, respectively, beautifully illustrate the significance of EMT to the above processes. Ongoing studies progressively elucidate the gene networks underlying EMT in each system, highlighting the similarities and differences between them. Knowledge of the mechanistic logic of this normal ontogenetic process should provide important insights to the understanding of pathological conditions such as cancer metastasis, which shares some common molecular themes.
Collapse
Affiliation(s)
- Chaya Kalcheim
- Edmond and Lili Safra Center for Brain Sciences (ELSC), Department of Medical Neurobiology, Institute for Medical Research Israel-Canada (IMRIC), Hebrew University of Jerusalem-Hadassah Medical School, P.O. Box 12272, Jerusalem 9112102, Israel.
| |
Collapse
|
41
|
Understanding Melanocyte Stem Cells for Disease Modeling and Regenerative Medicine Applications. Int J Mol Sci 2015; 16:30458-69. [PMID: 26703580 PMCID: PMC4691150 DOI: 10.3390/ijms161226207] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 12/01/2015] [Accepted: 12/07/2015] [Indexed: 02/04/2023] Open
Abstract
Melanocytes in the skin play an indispensable role in the pigmentation of skin and its appendages. It is well known that the embryonic origin of melanocytes is neural crest cells. In adult skin, functional melanocytes are continuously repopulated by the differentiation of melanocyte stem cells (McSCs) residing in the epidermis of the skin. Many preceding studies have led to significant discoveries regarding the cellular and molecular characteristics of this unique stem cell population. The alteration of McSCs has been also implicated in several skin abnormalities and disease conditions. To date, our knowledge of McSCs largely comes from studying the stem cell niche of mouse hair follicles. Suggested by several anatomical differences between mouse and human skin, there could be distinct features associated with mouse and human McSCs as well as their niches in the skin. Recent advances in human pluripotent stem cell (hPSC) research have provided us with useful tools to potentially acquire a substantial amount of human McSCs and functional melanocytes for research and regenerative medicine applications. This review highlights recent studies and progress involved in understanding the development of cutaneous melanocytes and the regulation of McSCs.
Collapse
|
42
|
Kerosuo L, Nie S, Bajpai R, Bronner ME. Crestospheres: Long-Term Maintenance of Multipotent, Premigratory Neural Crest Stem Cells. Stem Cell Reports 2015; 5:499-507. [PMID: 26441305 PMCID: PMC4625028 DOI: 10.1016/j.stemcr.2015.08.017] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 08/27/2015] [Accepted: 08/28/2015] [Indexed: 01/12/2023] Open
Abstract
Premigratory neural crest cells comprise a transient, embryonic population that arises within the CNS, but subsequently migrates away and differentiates into many derivatives. Previously, premigratory neural crest could not be maintained in a multipotent, adhesive state without spontaneous differentiation. Here, we report conditions that enable maintenance of neuroepithelial “crestospheres” that self-renew and retain multipotency for weeks. Moreover, under differentiation conditions, these cells can form multiple derivatives in vitro and in vivo after transplantation into chick embryos. Similarly, human embryonic stem cells directed to a neural crest fate can be maintained as crestospheres and subsequently differentiated into several derivatives. By devising conditions that maintain the premigratory state in vitro, these results demonstrate that neuroepithelial neural crest precursors are capable of long-term self-renewal. This approach will help uncover mechanisms underlying their developmental potential, differentiation and, together with the induced pluripotent stem cell techniques, the pathology of human neurocristopathies. Long-term maintenance of premigratory chick neural crest cells as crestospheres A self-renewing population of multipotent neuroepithelial neural crest stem cells Crestospheres differentiate into neural crest derivatives in vitro and in vivo Long-term maintenance of human ESC-derived crestospheres for several weeks
Collapse
Affiliation(s)
- Laura Kerosuo
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Shuyi Nie
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Ruchi Bajpai
- Center for Craniofacial Molecular Biology and Department of Biochemistry, University of Southern California, Los Angeles, CA 90089, USA
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
43
|
Fortuna V, Pardanaud L, Brunet I, Ola R, Ristori E, Santoro MM, Nicoli S, Eichmann A. Vascular Mural Cells Promote Noradrenergic Differentiation of Embryonic Sympathetic Neurons. Cell Rep 2015; 11:1786-96. [PMID: 26074079 DOI: 10.1016/j.celrep.2015.05.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Revised: 04/14/2015] [Accepted: 05/13/2015] [Indexed: 11/25/2022] Open
Abstract
The sympathetic nervous system controls smooth muscle tone and heart rate in the cardiovascular system. Postganglionic sympathetic neurons (SNs) develop in close proximity to the dorsal aorta (DA) and innervate visceral smooth muscle targets. Here, we use the zebrafish embryo to ask whether the DA is required for SN development. We show that noradrenergic (NA) differentiation of SN precursors temporally coincides with vascular mural cell (VMC) recruitment to the DA and vascular maturation. Blocking vascular maturation inhibits VMC recruitment and blocks NA differentiation of SN precursors. Inhibition of platelet-derived growth factor receptor (PDGFR) signaling prevents VMC differentiation and also blocks NA differentiation of SN precursors. NA differentiation is normal in cloche mutants that are devoid of endothelial cells but have VMCs. Thus, PDGFR-mediated mural cell recruitment mediates neurovascular interactions between the aorta and sympathetic precursors and promotes their noradrenergic differentiation.
Collapse
Affiliation(s)
- Vitor Fortuna
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06510, USA; Health Science Institute, Federal University of Bahia, Salvador 40110-902, Brazil
| | - Luc Pardanaud
- CNRS UMR7241, INSERM U1050, Center for Interdisciplinary Research in Biology (CIRB), Collège de France, Paris 75005, France
| | - Isabelle Brunet
- CNRS UMR7241, INSERM U1050, Center for Interdisciplinary Research in Biology (CIRB), Collège de France, Paris 75005, France
| | - Roxana Ola
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Emma Ristori
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Massimo M Santoro
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Torino, Italy; VIB Vesalius Research Center, KU Leuven, 3000 Leuven, Belgium
| | - Stefania Nicoli
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06510, USA.
| | - Anne Eichmann
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06510, USA; CNRS UMR7241, INSERM U1050, Center for Interdisciplinary Research in Biology (CIRB), Collège de France, Paris 75005, France; Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
44
|
Li L, Li D, Liu L, Li S, Feng Y, Peng X, Gong Y. Endothelin Receptor B2 (EDNRB2) Gene Is Associated with Spot Plumage Pattern in Domestic Ducks (Anas platyrhynchos). PLoS One 2015; 10:e0125883. [PMID: 25955279 PMCID: PMC4425580 DOI: 10.1371/journal.pone.0125883] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 03/08/2015] [Indexed: 01/24/2023] Open
Abstract
Endothelin receptor B subtype 2 (EDNRB2) is a seven-transmembrane G-protein coupled receptor. In this study, we investigated EDNRB2 gene as a candidate gene for duck spot plumage pattern according to studies of chicken and Japanese quail. The entire coding region was cloned by the reverse transcription polymerase chain reaction (RT-PCR). Sequence analysis showed that duck EDNRB2 cDNA contained a 1311bp open reading frame and encoded a putative protein of 436 amino acids residues. The transcript shared 89%-90% identity with the counterparts in other avian species. A phylogenetic tree based on amino acid sequences showed that duck EDNRB2 was evolutionary conserved in avian clade. The entire coding region of EDNRB2 were sequenced in 20 spot and 20 non-spot ducks, and 13 SNPs were identified. Two of them (c.940G>A and c.995G>A) were non-synonymous substitutions, and were genotyped in 647 ducks representing non-spot and spot phenotypes. The c.995G>A mutation, which results in the amino acid substitution of Arg332His, was completely associated with the spot phenotype: all 152 spot ducks were carriers of the AA genotype and the other 495 individuals with non-spot phenotype were carriers of GA or GG genotype, respectively. Segregation in 17 GA×GG and 22 GA×GA testing combinations confirmed this association since the segregation ratios and genotypes of the offspring were in agreement with the hypothesis. In order to investigate the underlying mechanism of the spot phenotype, MITF gene was used as cell type marker of melanocyte progenitor cells while TYR and TYRP1 gene were used as cell type markers of mature melanocytes. Transcripts of MITF, TYR and TYRP1 gene with expected size were identified in all pigmented skin tissues while PCR products were not obtained from non-pigmented skin tissues. It was inferred that melanocytes are absent in non-pigmented skin tissues of spot ducks.
Collapse
Affiliation(s)
- Ling Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Dan Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Li Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Shijun Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yanping Feng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xiuli Peng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yanzhang Gong
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- * E-mail:
| |
Collapse
|
45
|
Pentimento: Neural Crest and the origin of mesectoderm. Dev Biol 2015; 401:37-61. [DOI: 10.1016/j.ydbio.2014.12.035] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 12/28/2014] [Accepted: 12/30/2014] [Indexed: 11/17/2022]
|
46
|
Abstract
Melanocyte development provides an excellent model for studying more complex developmental processes. Melanocytes have an apparently simple aetiology, differentiating from the neural crest and migrating through the developing embryo to specific locations within the skin and hair follicles, and to other sites in the body. The study of pigmentation mutations in the mouse provided the initial key to identifying the genes and proteins involved in melanocyte development. In addition, work on chicken has provided important embryological and molecular insights, whereas studies in zebrafish have allowed live imaging as well as genetic and transgenic approaches. This cross-species approach is powerful and, as we review here, has resulted in a detailed understanding of melanocyte development and differentiation, melanocyte stem cells and the role of the melanocyte lineage in diseases such as melanoma. Summary: This Review discusses melanocyte development and differentiation, melanocyte stem cells, and the role of the melanocyte lineage in diseases such as melanoma.
Collapse
Affiliation(s)
| | - Ian J Jackson
- MRC Human Genetics Unit and University of Edinburgh Cancer Research UK Cancer Centre, MRC Institute for Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - E Elizabeth Patton
- MRC Human Genetics Unit and Roslin Institute, University of Edinburgh, Edinburgh EH25 9RG, UK
| |
Collapse
|
47
|
Aoki H, Hara A, Kunisada T. White spotting phenotype induced by targeted REST disruption during neural crest specification to a melanocyte cell lineage. Genes Cells 2015; 20:439-49. [DOI: 10.1111/gtc.12235] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Accepted: 02/15/2015] [Indexed: 01/07/2023]
Affiliation(s)
- Hitomi Aoki
- Department of Tissue and Organ Development, Regeneration, and Advanced Medical Science; Gifu University Graduate School of Medicine; 1-1 Yanagido Gifu 501-1194 Japan
| | - Akira Hara
- Department of Tumor Pathology; Gifu University Graduate School of Medicine; 1-1 Yanagido Gifu 501-1194 Japan
| | - Takahiro Kunisada
- Department of Tissue and Organ Development, Regeneration, and Advanced Medical Science; Gifu University Graduate School of Medicine; 1-1 Yanagido Gifu 501-1194 Japan
| |
Collapse
|
48
|
Jacob C. Transcriptional control of neural crest specification into peripheral glia. Glia 2015; 63:1883-1896. [PMID: 25752517 DOI: 10.1002/glia.22816] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 01/29/2015] [Accepted: 02/20/2015] [Indexed: 12/20/2022]
Abstract
The neural crest is a transient migratory multipotent cell population that originates from the neural plate border and is formed at the end of gastrulation and during neurulation in vertebrate embryos. These cells give rise to many different cell types of the body such as chondrocytes, smooth muscle cells, endocrine cells, melanocytes, and cells of the peripheral nervous system including different subtypes of neurons and peripheral glia. Acquisition of lineage-specific markers occurs before or during migration and/or at final destination. What are the mechanisms that direct specification of neural crest cells into a specific lineage and how do neural crest cells decide on a specific migration route? Those are fascinating and complex questions that have existed for decades and are still in the research focus of developmental biologists. This review discusses transcriptional events and regulations occurring in neural crest cells and derived lineages, which control specification of peripheral glia, namely Schwann cell precursors that interact with peripheral axons and further differentiate into myelinating or nonmyelinating Schwann cells, satellite cells that remain tightly associated with neuronal cell bodies in sensory and autonomous ganglia, and olfactory ensheathing cells that wrap olfactory axons, both at the periphery in the olfactory mucosa and in the central nervous system in the olfactory bulb. Markers of the different peripheral glia lineages including intermediate multipotent cells such as boundary cap cells, as well as the functions of these specific markers, are also reviewed. Enteric ganglia, another type of peripheral glia, will not be discussed in this review. GLIA 2015;63:1883-1896.
Collapse
Affiliation(s)
- Claire Jacob
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
49
|
Premigratory and Migratory Neural Crest Cells Are Multipotent In Vivo. Cell Stem Cell 2015; 16:314-22. [DOI: 10.1016/j.stem.2015.02.017] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 12/13/2014] [Accepted: 02/19/2015] [Indexed: 01/09/2023]
|
50
|
Kam MKM, Lui VCH. Roles of Hoxb5 in the development of vagal and trunk neural crest cells. Dev Growth Differ 2015; 57:158-68. [PMID: 25703667 DOI: 10.1111/dgd.12199] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/05/2015] [Accepted: 01/09/2015] [Indexed: 12/22/2022]
Abstract
Neural crest cells (NC) are a group of multipotent stem cells uniquely present in vertebrates. They are destined to form various organs according to their anterior-posterior (A-P) levels of origin in the neural tube (NT). They develop into a wide spectrum of cell lineages under the influence of signaling cascades, neural plate border genes and NC specifier genes. Although this complex gene regulatory network (GRN) specifies the fate of NC and the combinatory action of Hox genes executed at the time of NC induction governs the patterning of NC for the formation of specific structures along the A-P axis, not much information on how GRN and Hox genes directly interact and orchestrate is available. This review summarizes recent findings on the multiple roles of Hoxb5 on the survival and cell lineage differentiation of vagal and trunk NC cells during early development, by direct transcriptional regulation of NC specifier genes (Sox9 and Foxd3) of the GRN. We will also review findings on the transcriptional regulation of Ret by Hoxb5 in the population of the vagal NC that are committed to the enteric neuron and glia lineages. Functional redundancy between Hox proteins (Hoxa5 and Hoxc5) from the same paralogue group as Hoxb5, and the cooperative effects of Hox cofactors, collaborators and transcription factors in the Hoxb5 transcriptional regulation of target genes will also be discussed.
Collapse
Affiliation(s)
- Mandy K M Kam
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | | |
Collapse
|