1
|
Shan W, Li J, Philpot Z, Zuo Z. Carboxyl Terminal Modulator Protein Induces Cell Senescence and Is Upregulated With Aging by Zic2 in Rats. J Cell Physiol 2025; 240:e70007. [PMID: 39888066 PMCID: PMC11780686 DOI: 10.1002/jcp.70007] [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: 04/09/2024] [Revised: 12/21/2024] [Accepted: 01/17/2025] [Indexed: 02/01/2025]
Abstract
Carboxyl terminal modulator protein (CTMP) may be involved in various physiological and pathological processes, such as inflammation, tumor growth, and cardiac hypertrophy. Our recent study has shown that CTMP is increased with aging and plays a role in determining brain ischemic tolerance. However, it is not known how CTMP expression with aging is regulated and whether the changed CTMP expression has an effect on cell senescence. Here, cells that stably overexpressed CTMP were generated and cell senescence biomarkers were determined. The brains of Fischer 344 male rats were harvested for Western blot analysis and immunostaining to detect CTMP and the Zinc finger protein Zic2. The regulations of CTMP expression by Zic2 were examined by promoter activity assays. Increasing CTMP enhanced cells expressing senescence-associated β-galactosidase staining but without expression of Ki67, decreased cell proliferation and colony formation, and increased cells with condensed DNA of more than one pair of homologous chromosomes caused by senescence. Zic2 was decreased with aging in rats. Zic2 and CTMP were mainly expressed in the neurons in rats. Similarly, CTMP protein was expressed in the neurons of human brain. An anti-Zic2 antibody immunoprecipitated DNA fragments of ctmp gene. Zic2 inhibited the activity of presumptive ctmp promoter. Overexpressing Zic2 decreased CTMP in cells. These results suggest that CTMP induces cell senescence and that Zic2 is a suppressor of CTMP expression. The decrease of Zic2 contributes to CTMP increase with aging.
Collapse
Affiliation(s)
- Weiran Shan
- Department of AnesthesiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Jun Li
- Department of AnesthesiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Zachary Philpot
- Department of AnesthesiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
- Idaho College of Osteopathic MedicineMeridianIdahoUSA
| | - Zhiyi Zuo
- Department of AnesthesiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| |
Collapse
|
2
|
Huber PB, Rao A, LaBonne C. BET activity plays an essential role in control of stem cell attributes in Xenopus. Development 2024; 151:dev202990. [PMID: 38884356 PMCID: PMC11266789 DOI: 10.1242/dev.202990] [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: 04/26/2024] [Accepted: 05/31/2024] [Indexed: 06/18/2024]
Abstract
Neural crest cells are a stem cell population unique to vertebrate embryos that retains broad multi-germ layer developmental potential through neurulation. Much remains to be learned about the genetic and epigenetic mechanisms that control the potency of neural crest cells. Here, we examine the role that epigenetic readers of the BET (bromodomain and extra terminal) family play in controlling the potential of pluripotent blastula and neural crest cells. We find that inhibiting BET activity leads to loss of pluripotency at blastula stages and a loss of neural crest at neurula stages. We compare the effects of HDAC (an eraser of acetylation marks) and BET (a reader of acetylation) inhibition and find that they lead to similar cellular outcomes through distinct effects on the transcriptome. Interestingly, loss of BET activity in cells undergoing lineage restriction is coupled to increased expression of genes linked to pluripotency and prolongs the competence of initially pluripotent cells to transit to a neural progenitor state. Together these findings advance our understanding of the epigenetic control of pluripotency and the formation of the vertebrate neural crest.
Collapse
Affiliation(s)
- Paul B. Huber
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
- National Institute for Theory and Mathematics in Biology, Northwestern University, Evanston, IL 60208, USA
| | - Anjali Rao
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Carole LaBonne
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
- National Institute for Theory and Mathematics in Biology, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
3
|
Martinez JL, Piciw JG, Crockett M, Sorci IA, Makwana N, Sirois CL, Giffin-Rao Y, Bhattacharyya A. Transcriptional consequences of trisomy 21 on neural induction. Front Cell Neurosci 2024; 18:1341141. [PMID: 38357436 PMCID: PMC10865501 DOI: 10.3389/fncel.2024.1341141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024] Open
Abstract
Introduction Down syndrome, caused by trisomy 21, is a complex developmental disorder associated with intellectual disability and reduced growth of multiple organs. Structural pathologies are present at birth, reflecting embryonic origins. A fundamental unanswered question is how an extra copy of human chromosome 21 contributes to organ-specific pathologies that characterize individuals with Down syndrome, and, relevant to the hallmark intellectual disability in Down syndrome, how trisomy 21 affects neural development. We tested the hypothesis that trisomy 21 exerts effects on human neural development as early as neural induction. Methods Bulk RNA sequencing was performed on isogenic trisomy 21 and euploid human induced pluripotent stem cells (iPSCs) at successive stages of neural induction: embryoid bodies at Day 6, early neuroectoderm at Day 10, and differentiated neuroectoderm at Day 17. Results Gene expression analysis revealed over 1,300 differentially expressed genes in trisomy 21 cells along the differentiation pathway compared to euploid controls. Less than 5% of the gene expression changes included upregulated chromosome 21 encoded genes at every timepoint. Genes involved in specific growth factor signaling pathways (WNT and Notch), metabolism (including oxidative stress), and extracellular matrix were altered in trisomy 21 cells. Further analysis uncovered heterochronic expression of genes. Conclusion Trisomy 21 impacts discrete developmental pathways at the earliest stages of neural development. The results suggest that metabolic dysfunction arises early in embryogenesis in trisomy 21 and may affect development and function more broadly.
Collapse
Affiliation(s)
- José L. Martinez
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Jennifer G. Piciw
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, United States
- Medical Scientist Training Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Madeline Crockett
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Isabella A. Sorci
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Nikunj Makwana
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Carissa L. Sirois
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
| | | | - Anita Bhattacharyya
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
4
|
Thiery AP, Buzzi AL, Hamrud E, Cheshire C, Luscombe NM, Briscoe J, Streit A. scRNA-sequencing in chick suggests a probabilistic model for cell fate allocation at the neural plate border. eLife 2023; 12:e82717. [PMID: 37530410 PMCID: PMC10425176 DOI: 10.7554/elife.82717] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 08/01/2023] [Indexed: 08/03/2023] Open
Abstract
The vertebrate 'neural plate border' is a transient territory located at the edge of the neural plate containing precursors for all ectodermal derivatives: the neural plate, neural crest, placodes and epidermis. Elegant functional experiments in a range of vertebrate models have provided an in-depth understanding of gene regulatory interactions within the ectoderm. However, these experiments conducted at tissue level raise seemingly contradictory models for fate allocation of individual cells. Here, we carry out single cell RNA sequencing of chick ectoderm from primitive streak to neurulation stage, to explore cell state diversity and heterogeneity. We characterise the dynamics of gene modules, allowing us to model the order of molecular events which take place as ectodermal fates segregate. Furthermore, we find that genes previously classified as neural plate border 'specifiers' typically exhibit dynamic expression patterns and are enriched in either neural, neural crest or placodal fates, revealing that the neural plate border should be seen as a heterogeneous ectodermal territory and not a discrete transitional transcriptional state. Analysis of neural, neural crest and placodal markers reveals that individual NPB cells co-express competing transcriptional programmes suggesting that their ultimate identify is not yet fixed. This population of 'border located undecided progenitors' (BLUPs) gradually diminishes as cell fate decisions take place. Considering our findings, we propose a probabilistic model for cell fate choice at the neural plate border. Our data suggest that the probability of a progenitor's daughters to contribute to a given ectodermal derivative is related to the balance of competing transcriptional programmes, which in turn are regulated by the spatiotemporal position of a progenitor.
Collapse
Affiliation(s)
- Alexandre P Thiery
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College LondonLondonUnited Kingdom
| | - Ailin Leticia Buzzi
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College LondonLondonUnited Kingdom
| | - Eva Hamrud
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College LondonLondonUnited Kingdom
| | - Chris Cheshire
- Bioinformatics and Computational Biology Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Nicholas M Luscombe
- Bioinformatics and Computational Biology Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - James Briscoe
- Bioinformatics and Computational Biology Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Andrea Streit
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College LondonLondonUnited Kingdom
| |
Collapse
|
5
|
Ahmad MH, Ghosh B, Rizvi MA, Ali M, Kaur L, Mondal AC. Neural crest cells development and neuroblastoma progression: Role of Wnt signaling. J Cell Physiol 2023; 238:306-328. [PMID: 36502519 DOI: 10.1002/jcp.30931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 09/19/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022]
Abstract
Neuroblastoma (NB) is one of the most common heterogeneous extracranial cancers in infancy that arises from neural crest (NC) cells of the sympathetic nervous system. The Wnt signaling pathway, both canonical and noncanonical pathway, is a highly conserved signaling pathway that regulates the development and differentiation of the NC cells during embryogenesis. Reports suggest that aberrant activation of Wnt ligands/receptors in Wnt signaling pathways promote progression and relapse of NB. Wnt signaling pathways regulate NC induction and migration in a similar manner; it regulates proliferation and metastasis of NB. Inhibiting the Wnt signaling pathway or its ligands/receptors induces apoptosis and abrogates proliferation and tumorigenicity in all major types of NB cells. Here, we comprehensively discuss the Wnt signaling pathway and its mechanisms in regulating the development of NC and NB pathogenesis. This review highlights the implications of aberrant Wnt signaling in the context of etiology, progression, and relapse of NB. We have also described emerging strategies for Wnt-based therapies against the progression of NB that will provide new insights into the development of Wnt-based therapeutic strategies for NB.
Collapse
Affiliation(s)
- Mir Hilal Ahmad
- School of Life Sciences, Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India.,Genome Biology Lab, Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Balaram Ghosh
- Department of Clinical Pharmacology, Midnapore Medical College & Hospital, West Bengal, Medinipur, India
| | - Moshahid Alam Rizvi
- Genome Biology Lab, Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Mansoor Ali
- School of Life Sciences, Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Loveleena Kaur
- Division of Cancer Pharmacology, Indian Institute of Integrative Medicine (IIIM), Srinagar, India
| | - Amal Chandra Mondal
- School of Life Sciences, Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
6
|
Affiliation(s)
- Yeri Alice Rim
- CiSTEM Laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ji Hyeon Ju
- CiSTEM Laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea. .,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea.
| |
Collapse
|
7
|
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
|
8
|
Prasad MS, Charney RM, García-Castro MI. Specification and formation of the neural crest: Perspectives on lineage segregation. Genesis 2019; 57:e23276. [PMID: 30576078 PMCID: PMC6570420 DOI: 10.1002/dvg.23276] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 12/17/2018] [Accepted: 12/18/2018] [Indexed: 12/21/2022]
Abstract
The neural crest is a fascinating embryonic population unique to vertebrates that is endowed with remarkable differentiation capacity. Thought to originate from ectodermal tissue, neural crest cells generate neurons and glia of the peripheral nervous system, and melanocytes throughout the body. However, the neural crest also generates many ectomesenchymal derivatives in the cranial region, including cell types considered to be of mesodermal origin such as cartilage, bone, and adipose tissue. These ectomesenchymal derivatives play a critical role in the formation of the vertebrate head, and are thought to be a key attribute at the center of vertebrate evolution and diversity. Further, aberrant neural crest cell development and differentiation is the root cause of many human pathologies, including cancers, rare syndromes, and birth malformations. In this review, we discuss the current findings of neural crest cell ontogeny, and consider tissue, cell, and molecular contributions toward neural crest formation. We further provide current perspectives into the molecular network involved during the segregation of the neural crest lineage.
Collapse
Affiliation(s)
- Maneeshi S Prasad
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California
| | - Rebekah M Charney
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California
| | - Martín I García-Castro
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California
| |
Collapse
|
9
|
Nadadhur AG, Leferink PS, Holmes D, Hinz L, Cornelissen-Steijger P, Gasparotto L, Heine VM. Patterning factors during neural progenitor induction determine regional identity and differentiation potential in vitro. Stem Cell Res 2018; 32:25-34. [PMID: 30172094 DOI: 10.1016/j.scr.2018.08.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 08/13/2018] [Accepted: 08/22/2018] [Indexed: 12/20/2022] Open
Abstract
The neural tube consists of neural progenitors (NPs) that acquire different characteristics during gestation due to patterning factors. However, the influence of such patterning factors on human pluripotent stem cells (hPSCs) during in vitro neural differentiation is often unclear. This study compared neural induction protocols involving in vitro patterning with single SMAD inhibition (SSI), retinoic acid (RA) administration and dual SMAD inhibition (DSI). While the derived NP cells expressed known NP markers, they differed in their NP expression profile and differentiation potential. Cortical neuronal cells generated from 1) SSI NPs exhibited less mature neuronal phenotypes, 2) RA NPs exhibited an increased GABAergic phenotype, and 3) DSI NPs exhibited greater expression of glutamatergic lineage markers. Further, although all NPs generated astrocytes, astrocytes derived from the RA-induced NPs had the highest GFAP expression. Differences between NP populations included differential expression of regional identity markers HOXB4, LBX1, OTX1 and GSX2, which persisted into mature neural cell stages. This study suggests that patterning factors regulate how potential NPs may differentiate into specific neuronal and glial cell types in vitro. This challenges the utility of generic neural induction procedures, while highlighting the importance of carefully selecting specific NP protocols.
Collapse
Affiliation(s)
- Aishwarya G Nadadhur
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands
| | - Prisca S Leferink
- Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands
| | - Dwayne Holmes
- Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands
| | - Lisa Hinz
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands
| | - Paulien Cornelissen-Steijger
- Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands
| | - Lisa Gasparotto
- Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands
| | - Vivi M Heine
- Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands; Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands.
| |
Collapse
|
10
|
Wnt3a Ectopic Expression Interferes Axonal Projection and Motor Neuron Positioning During the Chicken Spinal Cord Development. J Mol Neurosci 2018; 64:619-630. [PMID: 29574664 DOI: 10.1007/s12031-018-1060-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 03/13/2018] [Indexed: 12/29/2022]
Abstract
The formation of dorsal-ventral axis of the spinal cord is controlled largely by dorsal signals such as Wnts (which are members of the wingless + MMTV integrants, Int family), besides ventral signals such as sonic hedgehog (Shh). Wnt3a, one of the Wnt family members, is involved in multiple cellular functions, including self-renewal, proliferation, differentiation, and motility. Here, we aim to study the mechanism of the regulation of chicken spinal cord patterning by Wnt3a. In this study, Wnt3a was ectopically expressed in the spinal cord of developing chicken embryos by in ovo electroporation. The results of immunofluorescent staining revealed that Wnt3a ectopic expression caused the abnormality of commissural axonal projection and the formation of nerve fibers was interrupted. It is worth noting that neurons in the ventricular zone, especially motor neurons, could not migrate laterally after the Wnt3a overexpression, which led to the malformation of motor column. In addition, we found that neurons could not protrude axons outwardly after overexpression of Wnt3a in the spinal cord. It was also found that Wnt3a overexpression inhibited the outgrowth of processes in culturing SH-SY5Y cells. In conclusion, we proposed that Wnt3a regulates neuronal morphology, which subsequently disrupts axonal projection and motor neuron positioning during spinal cord development.
Collapse
|
11
|
Brafman D, Willert K. Wnt/β-catenin signaling during early vertebrate neural development. Dev Neurobiol 2017; 77:1239-1259. [PMID: 28799266 DOI: 10.1002/dneu.22517] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/24/2017] [Accepted: 08/09/2017] [Indexed: 12/29/2022]
Abstract
The vertebrate central nervous system (CNS) is comprised of vast number of distinct cell types arranged in a highly organized manner. This high degree of complexity is achieved by cellular communication, including direct cell-cell contact, cell-matrix interactions, and cell-growth factor signaling. Among the several developmental signals controlling the development of the CNS, Wnt proteins have emerged as particularly critical and, hence, have captivated the attention of many researchers. With Wnts' evolutionarily conserved function as primordial symmetry breaking signals, these proteins and their downstream effects are responsible for simultaneously establishing cellular diversity and tissue organization. With their expansive repertoire of secreted agonists and antagonists, cell surface receptors, signaling cascades and downstream biological effects, Wnts are ideally suited to control the complex processes underlying vertebrate neural development. In this review, we will describe the mechanisms by which Wnts exert their potent effects on cells and tissues and highlight the many roles of Wnt signaling during neural development, starting from the initial induction of the neural plate, the subsequent patterning along the embryonic axes, to the intricately organized structure of the CNS. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 1239-1259, 2017.
Collapse
Affiliation(s)
- David Brafman
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona, 85287
| | - Karl Willert
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA, 92093-0695
| |
Collapse
|
12
|
Time-Course Gene Expression Profiling Reveals a Novel Role of Non-Canonical WNT Signaling During Neural Induction. Sci Rep 2016; 6:32600. [PMID: 27600186 PMCID: PMC5013468 DOI: 10.1038/srep32600] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 08/10/2016] [Indexed: 12/22/2022] Open
Abstract
The process of neuroepithelial differentiation from human pluripotent stem cells (PSCs) resembles in vivo neuroectoderm induction in the temporal course, morphogenesis, and biochemical changes. This in vitro model is therefore well-suited to reveal previously unknown molecular mechanisms underlying neural induction in humans. By transcriptome analysis of cells along PSC differentiation to early neuroepithelia at day 6 and definitive neuroepithelia at day 10, we found downregulation of genes that are associated with TGF-β and canonical WNT/β-CATENIN signaling, confirming the roles of classical signaling in human neural induction. Interestingly, WNT/Ca2+ signaling was upregulated. Pharmacological inhibition of the downstream effector of WNT/Ca2+ pathway, Ca2+/calmodulin-dependent protein kinase II (CaMKII), led to an inhibition of the neural marker PAX6 and upregulation of epidermal marker K18, suggesting that Ca2+/CaMKII signaling promotes neural induction by preventing the alternative epidermal fate. In addition, our analyses revealed known and novel expression patterns of genes that are involved in DNA methylation, histone modification, as well as epithelial-mesenchymal transition, highlighting potential roles of those genes and signaling pathways during neural differentiation.
Collapse
|
13
|
Mašek J, Machoň O, Kořínek V, Taketo MM, Kozmik Z. Tcf7l1 protects the anterior neural fold from adopting the neural crest fate. Development 2016; 143:2206-16. [DOI: 10.1242/dev.132357] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 04/21/2016] [Indexed: 12/11/2022]
Abstract
The neural crest (NC) is crucial for the evolutionary diversification of vertebrates. NC cells are induced at the neural plate border by the coordinated action of several signaling pathways, including Wnt/β-catenin. NC cells are normally generated in the posterior neural plate border, whereas the anterior neural fold is devoid of NC cells. Using the mouse model, we show here that active repression of Wnt/β-catenin signaling is required for maintenance of neuroepithelial identity in the anterior neural fold and for inhibition of NC induction. Conditional inactivation of Tcf7l1, a transcriptional repressor of Wnt target genes, leads to aberrant activation of Wnt/β-catenin signaling in the anterior neuroectoderm and its conversion into NC. This reduces the developing prosencephalon without affecting the anterior-posterior neural character. Thus, Tcf7l1 defines the border between the NC and the prospective forebrain via restriction of the Wnt/β-catenin signaling gradient.
Collapse
Affiliation(s)
- Jan Mašek
- Institute of Molecular Genetics, Academy of Science of the Czech Republic, Prague 142 20, Czech Republic
| | - Ondřej Machoň
- Institute of Molecular Genetics, Academy of Science of the Czech Republic, Prague 142 20, Czech Republic
| | - Vladimír Kořínek
- Institute of Molecular Genetics, Academy of Science of the Czech Republic, Prague 142 20, Czech Republic
| | - M. Mark Taketo
- Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Zbyněk Kozmik
- Institute of Molecular Genetics, Academy of Science of the Czech Republic, Prague 142 20, Czech Republic
| |
Collapse
|
14
|
Insulin-like factor regulates neural induction through an IGF1 receptor-independent mechanism. Sci Rep 2015; 5:11603. [PMID: 26112133 PMCID: PMC4481404 DOI: 10.1038/srep11603] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 05/29/2015] [Indexed: 01/15/2023] Open
Abstract
Insulin receptor (IR) and insulin-like growth factor-1 receptor (IGF1R) signalling is required for normal embryonic growth and development. Previous reports indicated that the IGF/IGF1R/MAPK pathway contributes to neural induction and the IGF/IGF1R/PI3K/Akt pathway to eye development. Here, we report the isolation of insulin3 encoding a novel insulin-like ligand involved in neural induction. Insulin3 has a similar structure to pro-insulin and mature IGF ligands, but cannot activate the IGF1 receptor. However, similar to IGFs, Insulin3 induced the gene expression of an anterior neural marker, otx2, and enlarged anterior head structures by inhibiting Wnt signalling. Insulin3 are predominantly localised to the endoplasmic reticulum when otx2 is induced by insulin3. Insulin3 reduced extracellular Wnts and cell surface localised Lrp6. These results suggest that Insulin3 is a novel cell-autonomous inhibitor of Wnt signalling. This study provides the first evidence that an insulin-like factor regulates neural induction through an IGF1R-independent mechanism.
Collapse
|
15
|
Zhang X, Cheong SM, Amado NG, Reis AH, MacDonald BT, Zebisch M, Jones EY, Abreu JG, He X. Notum is required for neural and head induction via Wnt deacylation, oxidation, and inactivation. Dev Cell 2015; 32:719-30. [PMID: 25771893 PMCID: PMC4375027 DOI: 10.1016/j.devcel.2015.02.014] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 02/17/2015] [Accepted: 02/17/2015] [Indexed: 11/21/2022]
Abstract
Secreted Wnt morphogens are essential for embryogenesis and homeostasis and require a lipid/palmitoleoylate modification for receptor binding and activity. Notum is a secreted Wnt antagonist that belongs to the α/β hydrolase superfamily, but its mechanism of action and roles in vertebrate embryogenesis are not fully understood. Here, we report that Notum hydrolyzes the Wnt palmitoleoylate adduct extracellularly, resulting in inactivated Wnt proteins that form oxidized oligomers incapable of receptor binding. Thus, Notum is a Wnt deacylase, and palmitoleoylation is obligatory for the Wnt structure that maintains its active monomeric conformation. Notum is expressed in naive ectoderm and neural plate in Xenopus and is required for neural and head induction. These findings suggest that Notum is a prerequisite for the "default" neural fate and that distinct mechanisms of Wnt inactivation by the Tiki protease in the Organizer and the Notum deacylase in presumptive neuroectoderm orchestrate vertebrate brain development.
Collapse
Affiliation(s)
- Xinjun Zhang
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Seong-Moon Cheong
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Nathalia G Amado
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Alice H Reis
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Bryan T MacDonald
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Matthias Zebisch
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - E Yvonne Jones
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Jose Garcia Abreu
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Xi He
- Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
16
|
Pera EM, Acosta H, Gouignard N, Climent M, Arregi I. Active signals, gradient formation and regional specificity in neural induction. Exp Cell Res 2013; 321:25-31. [PMID: 24315941 DOI: 10.1016/j.yexcr.2013.11.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 11/21/2013] [Accepted: 11/23/2013] [Indexed: 01/01/2023]
Abstract
The question of how the vertebrate embryo gives rise to a nervous system is of paramount interest in developmental biology. Neural induction constitutes the earliest step in this process and is tightly connected with development of the embryonic body axes. In the Xenopus embryo, perpendicular gradients of BMP and Wnt signals pattern the dorsoventral and anteroposterior body axes. Both pathways need to be inhibited to allow anterior neural induction to occur. FGF8 and IGF are active neural inducers that together with BMP and Wnt signals are integrated at the level of Smad 1/5/8 phosphorylation. Hedgehog (Hh) also contributes to anterior neural induction. Suppressor-of-fused plays an important role in intertwining the Hh and Wnt pathways. Distinct mechanisms are discussed that establish morphogen gradients and integrate retinoic acid and FGF signals during posterior development. These findings not only improve our understanding of regional specification in neural induction, but have profound implications for mammalian stem cell research and regenerative medicine.
Collapse
Affiliation(s)
- Edgar M Pera
- Lund Stem Cell Center, BMC, B12, Klinikgatan 26, Lund University, S-221 84 Lund, Sweden.
| | - Helena Acosta
- Lund Stem Cell Center, BMC, B12, Klinikgatan 26, Lund University, S-221 84 Lund, Sweden
| | - Nadège Gouignard
- Lund Stem Cell Center, BMC, B12, Klinikgatan 26, Lund University, S-221 84 Lund, Sweden
| | - Maria Climent
- Lund Stem Cell Center, BMC, B12, Klinikgatan 26, Lund University, S-221 84 Lund, Sweden
| | - Igor Arregi
- Lund Stem Cell Center, BMC, B12, Klinikgatan 26, Lund University, S-221 84 Lund, Sweden
| |
Collapse
|
17
|
Signaling pathways regulating ectodermal cell fate choices. Exp Cell Res 2013; 321:11-6. [PMID: 23939346 DOI: 10.1016/j.yexcr.2013.08.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 08/01/2013] [Indexed: 01/23/2023]
Abstract
Although embryonic patterning and early development of the nervous system have been studied for decades, our understanding of how signals instruct ectodermal derivatives to acquire specific identities has only recently started to form a coherent picture. In this mini-review, we summarize recent findings and models of how a handful of well-known secreted signals influence progenitor cells in successive binary decisions to adopt various cell type specific differentiation programs.
Collapse
|
18
|
The Xenopus Tgfbi is required for embryogenesis through regulation of canonical Wnt signalling. Dev Biol 2013; 379:16-27. [DOI: 10.1016/j.ydbio.2012.11.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 09/18/2012] [Accepted: 11/12/2012] [Indexed: 10/27/2022]
|
19
|
Yardley N, García-Castro MI. FGF signaling transforms non-neural ectoderm into neural crest. Dev Biol 2012; 372:166-77. [PMID: 23000357 PMCID: PMC3541687 DOI: 10.1016/j.ydbio.2012.09.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Revised: 07/29/2012] [Accepted: 09/04/2012] [Indexed: 10/27/2022]
Abstract
The neural crest arises at the border between the neural plate and the adjacent non-neural ectoderm. It has been suggested that both neural and non-neural ectoderm can contribute to the neural crest. Several studies have examined the molecular mechanisms that regulate neural crest induction in neuralized tissues or the neural plate border. Here, using the chick as a model system, we address the molecular mechanisms by which non-neural ectoderm generates neural crest. We report that in response to FGF the non-neural ectoderm can ectopically express several early neural crest markers (Pax7, Msx1, Dlx5, Sox9, FoxD3, Snail2, and Sox10). Importantly this response to FGF signaling can occur without inducing ectopic mesodermal tissues. Furthermore, the non-neural ectoderm responds to FGF by expressing the prospective neural marker Sox3, but it does not express definitive markers of neural or anterior neural (Sox2 and Otx2) tissues. These results suggest that the non-neural ectoderm can launch the neural crest program in the absence of mesoderm, without acquiring definitive neural character. Finally, we report that prior to the upregulation of these neural crest markers, the non-neural ectoderm upregulates both BMP and Wnt molecules in response to FGF. Our results provide the first effort to understand the molecular events leading to neural crest development via the non-neural ectoderm in amniotes and present a distinct response to FGF signaling.
Collapse
Affiliation(s)
- Nathan Yardley
- KBT 1100, Department of Molecular, Cellular, and Developmental Biology, Yale University, PO Box 208103, New Haven, Connecticut 06520-8103, USA
| | - Martín I. García-Castro
- KBT 1100, Department of Molecular, Cellular, and Developmental Biology, Yale University, PO Box 208103, New Haven, Connecticut 06520-8103, USA
| |
Collapse
|
20
|
Beccari L, Marco-Ferreres R, Bovolenta P. The logic of gene regulatory networks in early vertebrate forebrain patterning. Mech Dev 2012; 130:95-111. [PMID: 23111324 DOI: 10.1016/j.mod.2012.10.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 10/09/2012] [Indexed: 01/19/2023]
Abstract
The vertebrate forebrain or prosencephalon is patterned at the beginning of neurulation into four major domains: the telencephalic, hypothalamic, retinal and diencephalic anlagen. These domains will then give rise to the majority of the brain structures involved in sensory integration and the control of higher intellectual and homeostatic functions. Understanding how forebrain pattering arises has thus attracted the interest of developmental neurobiologists for decades. As a result, most of its regulators have been identified and their hierarchical relationship is now the object of active investigation. Here, we summarize the main morphogenetic pathways and transcription factors involved in forebrain specification and propose the backbone of a possible gene regulatory network (GRN) governing its specification, taking advantage of the GRN principles elaborated by pioneer studies in simpler organisms. We will also discuss this GRN and its operational logic in the context of the remarkable morphological and functional diversification that the forebrain has undergone during evolution.
Collapse
Affiliation(s)
- Leonardo Beccari
- Centro de Biología Molecular "Severo Ochoa", CSIC-UAM, c/Nicolas Cabrera, 1, Madrid 28049, Spain
| | | | | |
Collapse
|
21
|
Dual functions of DP1 promote biphasic Wnt-on and Wnt-off states during anteroposterior neural patterning. EMBO J 2012; 31:3384-97. [PMID: 22773187 DOI: 10.1038/emboj.2012.181] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2012] [Accepted: 06/15/2012] [Indexed: 01/14/2023] Open
Abstract
DP1, a dimerization partner protein of the transcription factor E2F, is known to inhibit Wnt/β-catenin signalling along with E2F, although the function of DP1 itself was not well characterized. Here, we present a novel dual regulatory mechanism of Wnt/β-catenin signalling by DP1 independent from E2F. DP1 negatively regulates Wnt/β-catenin signalling by inhibiting Dvl-Axin interaction and by enhancing poly-ubiquitination of β-catenin. In contrast, DP1 positively modulates the signalling upon Wnt stimulation, via increasing cytosolic β-catenin and antagonizing the kinase activity of NLK. In Xenopus embryos, DP1 exerts both positive and negative roles in Wnt/β-catenin signalling during anteroposterior neural patterning. From subcellular localization analyses, we suggest that the dual roles of DP1 in Wnt/β-catenin signalling are endowed by differential nucleocytoplasmic localizations. We propose that these dual functions of DP1 can promote and stabilize biphasic Wnt-on and Wnt-off states in response to a gradual gradient of Wnt/β-catenin signalling to determine differential cell fates.
Collapse
|
22
|
Dang LTH, Wong L, Tropepe V. Zfhx1b induces a definitive neural stem cell fate in mouse embryonic stem cells. Stem Cells Dev 2012; 21:2838-51. [PMID: 22594450 DOI: 10.1089/scd.2011.0593] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Inducing a stable and predictable program of neural cell fate in pluripotent cells in vitro is an important goal for utilizing these cells for modeling human disease mechanisms. However, the extent to which in vitro neural specification recapitulates in vivo neural specification remains to be fully established. We previously demonstrated that in the mouse embryo, activation of fibroblast growth factor (FGF) signalling promotes definitive neural stem cell (NSC) development through the upregulation of the transcription factor Zfhx1b. Here, we asked whether Zfhx1b is similarly required during neural lineage development of embryonic stem (ES) cells. Zfhx1b gene expression is rapidly upregulated in mouse ES cells cultured in a permissive neural-inducing environment, compared to ES cells in a standard pluripotency maintenance environment, and is potentiated by FGF signalling. However, overexpression of Zfhx1b in ES cells in maintenance conditions, containing serum and leukemia inhibitory factor (LIF), is sufficient to induce Sox1 expression, a marker found in neural precursors and to promote definitive NSC colony formation. Knockdown of Zfhx1b in ES cells using siRNA did not affect the initial transition of ES cells to a neural cell fate, but did diminish the ability of these neural cells to develop further into definitive NSCs. Thus, our findings using ES cells are congruent with evidence from mouse embryos and support a model, whereby intercellular FGF signaling induces Zfhx1b, which promotes the development of definitive NSCs subsequent to an initial neural specification event that is independent of this pathway.
Collapse
Affiliation(s)
- Lan T H Dang
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | | | | |
Collapse
|
23
|
Steventon B, Mayor R. Early neural crest induction requires an initial inhibition of Wnt signals. Dev Biol 2012; 365:196-207. [PMID: 22394485 PMCID: PMC3657187 DOI: 10.1016/j.ydbio.2012.02.029] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Revised: 01/31/2012] [Accepted: 02/20/2012] [Indexed: 01/28/2023]
Abstract
Neural crest (NC) induction is a long process that continues through gastrula and neurula stages. In order to reveal additional stages of NC induction we performed a series of explants where different known inducing tissues were taken along with the prospective NC. Interestingly the dorso-lateral marginal zone (DLMZ) is only able to promote the expression of a subset of neural plate border (NPB) makers without the presence of specific NC markers. We then analysed the temporal requirement for BMP and Wnt signals for the NPB genes Hairy2a and Dlx5, compared to the expression of neural plate (NP) and NC genes. Although the NP is sensitive to BMP levels at early gastrula stages, Hairy2a/Dlx5 expression is unaffected. Later, the NP becomes insensitive to BMP levels at late gastrulation when NC markers require an inhibition. The NP requires an inhibition of Wnt signals prior to gastrulation, but becomes insensitive during early gastrula stages when Hairy2a/Dlx5 requires an inhibition of Wnt signalling. An increase in Wnt signalling is then important for the switch from NPB to NC at late gastrula stages. In addition to revealing an additional distinct signalling event in NC induction, this work emphasizes the importance of integrating both timing and levels of signalling activity during the patterning of complex tissues such as the vertebrate ectoderm.
Collapse
Affiliation(s)
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
24
|
Stuhlmiller TJ, García-Castro MI. Current perspectives of the signaling pathways directing neural crest induction. Cell Mol Life Sci 2012; 69:3715-37. [PMID: 22547091 PMCID: PMC3478512 DOI: 10.1007/s00018-012-0991-8] [Citation(s) in RCA: 167] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Revised: 03/12/2012] [Accepted: 04/02/2012] [Indexed: 01/05/2023]
Abstract
The neural crest is a migratory population of embryonic cells with a tremendous potential to differentiate and contribute to nearly every organ system in the adult body. Over the past two decades, an incredible amount of research has given us a reasonable understanding of how these cells are generated. Neural crest induction involves the combinatorial input of multiple signaling pathways and transcription factors, and is thought to occur in two phases from gastrulation to neurulation. In the first phase, FGF and Wnt signaling induce NC progenitors at the border of the neural plate, activating the expression of members of the Msx, Pax, and Zic families, among others. In the second phase, BMP, Wnt, and Notch signaling maintain these progenitors and bring about the expression of definitive NC markers including Snail2, FoxD3, and Sox9/10. In recent years, additional signaling molecules and modulators of these pathways have been uncovered, creating an increasingly complex regulatory network. In this work, we provide a comprehensive review of the major signaling pathways that participate in neural crest induction, with a focus on recent developments and current perspectives. We provide a simplified model of early neural crest development and stress similarities and differences between four major model organisms: Xenopus, chick, zebrafish, and mouse.
Collapse
Affiliation(s)
- Timothy J Stuhlmiller
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06520-8103, USA
| | | |
Collapse
|
25
|
Maéno M, Komiyama K, Matsuzaki Y, Hosoya J, Kurihara S, Sakata H, Izutsu Y. Distinct mechanisms control the timing of differentiation of two myeloid populations in Xenopus ventral blood islands. Dev Growth Differ 2012; 54:187-201. [PMID: 22470938 DOI: 10.1111/j.1440-169x.2011.01321.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Previous study has suggested that distinct populations of myeloid cells exist in the anterior ventral blood islands (aVBI) and posterior ventral blood islands (pVBI) in Xenopus neurula embryo. However, details for differentiation programs of these two populations have not been elucidated. In the present study, we examined the role of Wnt, vascular endothelial growth factor (VEGF) and fibroblast growth factor signals in the regulation of myeloid cell differentiation in the dorsal marginal zone and ventral marginal zone explants that are the sources of myeloid cells in the aVBI and pVBI. We found that regulation of Wnt activity is essential for the differentiation of myeloid cells in the aVBI but is not required for the differentiation of myeloid cells in the pVBI. Endogenous activity of the VEGF signal is necessary for differentiation of myeloid cells in the pVBI but is not involved in the differentiation of myeloid cells in the aVBI. Overall results reveal that distinct mechanisms are involved in the myeloid, erythroid and endothelial cell differentiation in the aVBI and pVBI.
Collapse
Affiliation(s)
- Mitsugu Maéno
- Department of Biology, Faculty of Science, Niigata University, 8050 Ikarashi-2, Nishi-ku, Niigata 950-2181, Japan.
| | | | | | | | | | | | | |
Collapse
|
26
|
Indian hedgehog signaling is required for proper formation, maintenance and migration of Xenopus neural crest. Dev Biol 2012; 364:99-113. [PMID: 22309705 DOI: 10.1016/j.ydbio.2012.01.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2011] [Revised: 12/30/2011] [Accepted: 01/23/2012] [Indexed: 11/23/2022]
Abstract
Neural crest induction is the result of the combined action at the neural plate border of FGF, BMP, and Wnt signals from the neural plate, mesoderm and nonneural ectoderm. In this work we show that the expression of Indian hedgehog (Ihh, formerly named Banded hedgehog) and members of the Hedgehog pathway occurs at the prospective neural fold, in the premigratory and migratory neural crest. We performed a functional analysis that revealed the requirement of Ihh signaling in neural crest development. During the early steps of neural crest induction loss of function experiments with antisense morpholino or locally grafted cyclopamine-loaded beads suppressed the expression of early neural crest markers concomitant with the increase in neural and epidermal markers. We showed that changes in Ihh activity produced no alterations in either cell proliferation or apoptosis, suggesting that this signal involves cell fate decisions. A temporal analysis showed that Hedgehog is continuously required not only in the early and late specification but also during the migration of the neural crest. We also established that the mesodermal source of Ihh is important to maintain specification and also to support the migratory process. By a combination of embryological and molecular approaches our results demonstrated that Ihh signaling drives in the migration of neural crest cells by autocrine or paracrine mechanisms. Finally, the abrogation of Ihh signaling strongly affected only the formation of cartilages derived from the neural crest, while no effects were observed on melanocytes. Taken together, our results provide insights into the role of the Ihh cell signaling pathway during the early steps of neural crest development.
Collapse
|
27
|
Pourebrahim R, Houtmeyers R, Ghogomu S, Janssens S, Thelie A, Tran HT, Langenberg T, Vleminckx K, Bellefroid E, Cassiman JJ, Tejpar S. Transcription factor Zic2 inhibits Wnt/β-catenin protein signaling. J Biol Chem 2011; 286:37732-40. [PMID: 21908606 DOI: 10.1074/jbc.m111.242826] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Zic transcription factors play critical roles during embryonic development. Mutations in the ZIC2 gene are associated with human holoprosencephaly, but the etiology is still unclear. Here, we report a novel function for ZIC2 as a regulator of β-catenin·TCF4-mediated transcription. We show that ZIC2 can bind directly to the DNA-binding high mobility group box of TCF4 via its zinc finger domain and inhibit the transcriptional activity of the β-catenin·TCF4 complex. However, the binding of TCF4 to DNA was not affected by ZIC2. Zic2 RNA injection completely inhibited β-catenin-induced axis duplication in Xenopus embryos and strongly blocked the ability of β-catenin to induce expression of known Wnt targets in animal caps. Moreover, Zic2 knockdown in transgenic Xenopus Wnt reporter embryos led to ectopic Wnt signaling activity mainly at the midbrain-hindbrain boundary. Together, our results demonstrate a previously unknown role for ZIC2 as a transcriptional regulator of the β-catenin·TCF4 complex.
Collapse
Affiliation(s)
- Rasoul Pourebrahim
- Department of Human Genetics, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Wnt signaling pathways control lineage specification in vertebrate embryos and regulate pluripotency in embryonic stem (ES) cells, but how the balance between progenitor self-renewal and differentiation is achieved during axis specification and tissue patterning remains highly controversial. The context- and stage-specific effects of the different Wnt pathways produce complex and sometimes opposite outcomes that help to generate embryonic cell diversity. Although the results of recent studies of the Wnt/β-catenin pathway in ES cells appear to be surprising and controversial, they converge on the same conserved mechanism that leads to the inactivation of TCF3-mediated repression.
Collapse
Affiliation(s)
- Sergei Y Sokol
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029, USA.
| |
Collapse
|
29
|
Min TH, Kriebel M, Hou S, Pera EM. The dual regulator Sufu integrates Hedgehog and Wnt signals in the early Xenopus embryo. Dev Biol 2011; 358:262-76. [PMID: 21839734 DOI: 10.1016/j.ydbio.2011.07.035] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Revised: 07/01/2011] [Accepted: 07/28/2011] [Indexed: 12/14/2022]
Abstract
Hedgehog (Hh) and Wnt proteins are important signals implicated in several aspects of embryonic development, including the early development of the central nervous system. We found that Xenopus Suppressor-of-fused (XSufu) affects neural induction and patterning by regulating the Hh/Gli and Wnt/β-catenin pathways. Microinjection of XSufu mRNA induced expansion of the epidermis at the expense of neural plate tissue and caused enlargement of the eyes. An antisense morpholino oligonucleotide against XSufu had the opposite effect. Interestingly, both gain- and loss-of-function experiments resulted in a posterior shift of brain markers, suggesting a biphasic effect of XSufu on anteroposterior patterning. XSufu blocked early Wnt/β-catenin signaling, as indicated by the suppression of XWnt8-induced secondary axis formation in mRNA-injected embryos, and activation of Wnt target genes in XSufu-MO-injected ectodermal explants. We show that XSufu binds to XGli1 and Xβ-catenin. In Xenopus embryos and mouse embryonic fibroblasts, Gli1 inhibits Wnt signaling under overexpression of β-catenin, whereas β-catenin stimulates Hh signaling under overexpression of Gli1. Notably, endogenous Sufu is critically involved in this crosstalk. The results suggest that XSufu may act as a common regulator of Hh and Wnt signaling and contribute to intertwining the two pathways.
Collapse
Affiliation(s)
- Tan H Min
- Stem Cell Center, Lund University, 221 84 Lund, Sweden
| | | | | | | |
Collapse
|
30
|
Denis JA, Rochon-Beaucourt C, Champon B, Pietu G. Global Transcriptional Profiling of Neural and Mesenchymal Progenitors Derived from Human Embryonic Stem Cells Reveals Alternative Developmental Signaling Pathways. Stem Cells Dev 2011; 20:1395-409. [DOI: 10.1089/scd.2010.0331] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Jérôme Alexandre Denis
- INSERM/UEVE U-861, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, Evry Cedex, France
| | - Christelle Rochon-Beaucourt
- INSERM/UEVE U-861, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, Evry Cedex, France
| | - Benoite Champon
- CECS/AFM, I-STEM, Centre d'Etude des Cellules Souches, Evry Cedex, France
| | - Geneviève Pietu
- INSERM/UEVE U-861, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, Evry Cedex, France
| |
Collapse
|
31
|
Yan B, Neilson KM, Moody SA. Microarray identification of novel downstream targets of FoxD4L1/D5, a critical component of the neural ectodermal transcriptional network. Dev Dyn 2011; 239:3467-80. [PMID: 21069826 DOI: 10.1002/dvdy.22485] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
FoxD4L1/D5 is a forkhead transcription factor that functions as both a transcriptional activator and repressor. FoxD4L1/D5 acts upstream of several other neural transcription factors to maintain neural fate, regulate neural plate patterning, and delay the expression of neural differentiation factors. To identify a more complete list of downstream genes that participate in these earliest steps of neural ectodermal development, we carried out a microarray analysis comparing gene expression in control animal cap ectodermal explants (ACs), which will form epidermis, to that in FoxD4L1/D5-expressing ACs. Forty-four genes were tested for validation by RT-PCR of ACs and/or in situ hybridization assays in embryos; 86% of those genes up-regulated and 100% of those genes down-regulated in the microarray were altered accordingly in one of these independent assays. Eleven of these 44 genes are of unknown function, and we provide herein their developmental expression patterns to begin to reveal their roles in ectodermal development.
Collapse
Affiliation(s)
- Bo Yan
- Department of Anatomy and Regenerative Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | | | | |
Collapse
|
32
|
Engberg N, Kahn M, Petersen DR, Hansson M, Serup P. Retinoic acid synthesis promotes development of neural progenitors from mouse embryonic stem cells by suppressing endogenous, Wnt-dependent nodal signaling. Stem Cells 2010; 28:1498-509. [PMID: 20665854 DOI: 10.1002/stem.479] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Embryonic stem (ES) cells differentiate spontaneously toward a neuroectodermal fate in serum-free, adherent monocultures. Here, we show that this spontaneous neural fate requires retinoic acid (RA) synthesis. We monitor ES cells containing reporter genes for markers of the early neural plate as well as the primitive streak and its progeny to determine the cell fates induced when RA signaling is perturbed. We demonstrate that the spontaneous neural commitment of mouse ES cells requires endogenous RA production from vitamin A (vitA) in the medium. Formation of neural progenitors is inhibited by removing vitA from the medium, by inhibiting the enzymes that catalyze the synthesis of RA, or by inhibiting RA receptors. We show that subnanomolar concentrations of RA restore neuroectodermal differentiation when RA synthesis is blocked. We demonstrate that a neural to mesodermal fate change occurring when RA signaling is inhibited is dependent on Nodal-, Wnt-, and fibroblast growth factor-signaling. We show that Nodal suppresses neural development in a Wnt-dependent manner and that Wnt-mediated inhibition of neural development is reversed by inhibition of Nodal signaling. Together, our results show that neural induction in ES cells requires RA at subnanomolar levels to suppress Nodal signaling and suggest that the mechanism by which Wnt signaling suppresses neural development is through facilitation of Nodal signaling.
Collapse
Affiliation(s)
- Nina Engberg
- Department of Stem Cell Biology, Hagedorn Research Institute, Gentofte, Denmark
| | | | | | | | | |
Collapse
|
33
|
Lin HH, Bell E, Uwanogho D, Perfect LW, Noristani H, Bates TJD, Snetkov V, Price J, Sun YM. Neuronatin promotes neural lineage in ESCs via Ca(2+) signaling. Stem Cells 2010; 28:1950-60. [PMID: 20872847 PMCID: PMC3003906 DOI: 10.1002/stem.530] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Accepted: 09/04/2010] [Indexed: 12/19/2022]
Abstract
Neural induction is the first step in the formation of the vertebrate central nervous system. The emerging consensus of the mechanisms underlying neural induction is the combined influences from inhibiting bone morphogenetic protein (BMP) signaling and activating fibroblast growth factor (FGF)/Erk signaling, which act extrinsically via either autocrine or paracrine fashions. However, do intrinsic forces (cues) exist and do they play decisive roles in neural induction? These questions remain to be answered. Here, we have identified a novel neural initiator, neuronatin (Nnat), which acts as an intrinsic factor to promote neural fate in mammals and Xenopus. ESCs lacking this intrinsic factor fail to undergo neural induction despite the inhibition of the BMP pathway. We show that Nnat initiates neural induction in ESCs through increasing intracellular Ca(2+) ([Ca(2+) ](i)) by antagonizing Ca(2+) -ATPase isoform 2 (sarco/endoplasmic reticulum Ca(2+) -ATPase isoform 2) in the endoplasmic reticulum, which in turn increases the phosphorylation of Erk1/2 and inhibits the BMP4 pathway and leads to neural induction in conjunction with FGF/Erk pathway.
Collapse
Affiliation(s)
- Hsuan-Hwai Lin
- Institute of Psychiatry, King's College London, Centre for the Cellular Basis of Behaviour LondonUnited Kingdom
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical CenterTaipei, Taiwan, Republic of China
| | - Esther Bell
- MRC Centre for Developmental Neurobiology, Kings College London, Guy's CampusLondon, United Kingdom
| | - Dafe Uwanogho
- Institute of Psychiatry, King's College London, Centre for the Cellular Basis of Behaviour LondonUnited Kingdom
| | - Leo W Perfect
- Institute of Psychiatry, King's College London, Centre for the Cellular Basis of Behaviour LondonUnited Kingdom
| | - Harun Noristani
- Institute of Psychiatry, King's College London, Centre for the Cellular Basis of Behaviour LondonUnited Kingdom
| | - Thomas J D Bates
- MRC Centre for Developmental Neurobiology, Kings College London, Guy's CampusLondon, United Kingdom
| | - Vladimir Snetkov
- Department of Asthma, Allergy and Respiratory Science, Franklin-Wilkins Building, King's College LondonLondon, United Kingdom
| | - Jack Price
- Institute of Psychiatry, King's College London, Centre for the Cellular Basis of Behaviour LondonUnited Kingdom
| | - Yuh-Man Sun
- Institute of Psychiatry, King's College London, Centre for the Cellular Basis of Behaviour LondonUnited Kingdom
| |
Collapse
|
34
|
Klymkowsky MW, Rossi CC, Artinger KB. Mechanisms driving neural crest induction and migration in the zebrafish and Xenopus laevis. Cell Adh Migr 2010; 4:595-608. [PMID: 20962584 PMCID: PMC3011258 DOI: 10.4161/cam.4.4.12962] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Accepted: 07/09/2010] [Indexed: 01/09/2023] Open
Abstract
The neural crest is an evolutionary adaptation, with roots in the formation of mesoderm. Modification of neural crest behavior has been is critical for the evolutionary diversification of the vertebrates and defects in neural crest underlie a range of human birth defects. There has been a tremendous increase in our knowledge of the molecular, cellular, and inductive interactions that converge on defining the neural crest and determining its behavior. While there is a temptation to look for simple models to explain neural crest behavior, the reality is that the system is complex in its circuitry. In this review, our goal is to identify the broad features of neural crest origins (developmentally) and migration (cellularly) using data from the zebrafish (teleost) and Xenopus laevis (tetrapod amphibian) in order to illuminate where general mechanisms appear to be in play, and equally importantly, where disparities in experimental results suggest areas of profitable study.
Collapse
Affiliation(s)
- Michael W Klymkowsky
- Department of Molecular, Cellular and Developmental Biology; University of Colorado Boulder; Boulder, CO USA
| | - Christy Cortez Rossi
- Department of Craniofacial Biology; University of Colorado Denver; School of Dental Medicine; Aurora, CO USA
| | - Kristin Bruk Artinger
- Department of Craniofacial Biology; University of Colorado Denver; School of Dental Medicine; Aurora, CO USA
| |
Collapse
|
35
|
Paraxial T-box genes, Tbx6 and Tbx1, are required for cranial chondrogenesis and myogenesis. Dev Biol 2010; 346:170-80. [DOI: 10.1016/j.ydbio.2010.07.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 06/12/2010] [Accepted: 07/06/2010] [Indexed: 12/18/2022]
|
36
|
Wang Y, Fu Y, Gao L, Zhu G, Liang J, Gao C, Huang B, Fenger U, Niehrs C, Chen YG, Wu W. Xenopus skip modulates Wnt/beta-catenin signaling and functions in neural crest induction. J Biol Chem 2010; 285:10890-901. [PMID: 20103590 PMCID: PMC2856295 DOI: 10.1074/jbc.m109.058347] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Revised: 12/30/2009] [Indexed: 11/06/2022] Open
Abstract
The beta-catenin-lymphoid enhancer factor (LEF) protein complex is the key mediator of canonical Wnt signaling and initiates target gene transcription upon ligand stimulation. In addition to beta-catenin and LEF themselves, many other proteins have been identified as necessary cofactors. Here we report that the evolutionally conserved splicing factor and transcriptional co-regulator, SKIP/SNW/NcoA62, forms a ternary complex with LEF1 and HDAC1 and mediates the repression of target genes. Loss-of-function studies showed that SKIP is obligatory for Wnt signaling-induced target gene transactivation, suggesting an important role of SKIP in the canonical Wnt signaling. Consistent with its involvement in beta-catenin signaling, the C-terminally truncated forms of SKIP are able to stabilize beta-catenin and enhance Wnt signaling. In Xenopus embryos, both overexpression and knockdown of Skip lead to reduced neural crest induction, consistent with down-regulated Wnt signaling in both cases. Our results indicate that SKIP is a novel component of the beta-catenin transcriptional complex.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/antagonists & inhibitors
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Blotting, Western
- Chromatin Immunoprecipitation
- Embryo, Nonmammalian/cytology
- Embryo, Nonmammalian/metabolism
- Gene Expression Regulation, Developmental
- Gene Library
- HeLa Cells
- Humans
- Immunoenzyme Techniques
- Luciferases/metabolism
- Mice
- Neural Crest/cytology
- Neural Crest/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/pharmacology
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Wnt1 Protein/genetics
- Wnt1 Protein/metabolism
- Xenopus laevis
- beta Catenin/genetics
- beta Catenin/metabolism
Collapse
Affiliation(s)
- Ying Wang
- From the School of Life Sciences
- Protein Science Laboratory of the Ministry of Education, and
| | - Yu Fu
- From the School of Life Sciences
- Protein Science Laboratory of the Ministry of Education, and
| | - Lei Gao
- From the School of Life Sciences
- Protein Science Laboratory of the Ministry of Education, and
| | - Guixin Zhu
- From the School of Life Sciences
- Protein Science Laboratory of the Ministry of Education, and
| | - Juan Liang
- From the School of Life Sciences
- Protein Science Laboratory of the Ministry of Education, and
| | - Chan Gao
- From the School of Life Sciences
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Tsinghua University, Beijing 100084, China and
| | - Binlu Huang
- From the School of Life Sciences
- Protein Science Laboratory of the Ministry of Education, and
| | - Ursula Fenger
- the Division of Molecular Embryology, German Cancer Research Center, Im Neuenheimer Feld 581, D-69120 Heidelberg, Germany
| | - Christof Niehrs
- the Division of Molecular Embryology, German Cancer Research Center, Im Neuenheimer Feld 581, D-69120 Heidelberg, Germany
| | - Ye-Guang Chen
- From the School of Life Sciences
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Tsinghua University, Beijing 100084, China and
| | - Wei Wu
- From the School of Life Sciences
- Protein Science Laboratory of the Ministry of Education, and
| |
Collapse
|
37
|
Koenig SF, Brentle S, Hamdi K, Fichtner D, Wedlich D, Gradl D. En2, Pax2/5 and Tcf-4 transcription factors cooperate in patterning the Xenopus brain. Dev Biol 2010; 340:318-28. [DOI: 10.1016/j.ydbio.2010.02.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2009] [Revised: 02/01/2010] [Accepted: 02/10/2010] [Indexed: 11/25/2022]
|
38
|
Schlosser G. Making senses development of vertebrate cranial placodes. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2010; 283:129-234. [PMID: 20801420 DOI: 10.1016/s1937-6448(10)83004-7] [Citation(s) in RCA: 142] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Cranial placodes (which include the adenohypophyseal, olfactory, lens, otic, lateral line, profundal/trigeminal, and epibranchial placodes) give rise to many sense organs and ganglia of the vertebrate head. Recent evidence suggests that all cranial placodes may be developmentally related structures, which originate from a common panplacodal primordium at neural plate stages and use similar regulatory mechanisms to control developmental processes shared between different placodes such as neurogenesis and morphogenetic movements. After providing a brief overview of placodal diversity, the present review summarizes current evidence for the existence of a panplacodal primordium and discusses the central role of transcription factors Six1 and Eya1 in the regulation of processes shared between different placodes. Upstream signaling events and transcription factors involved in early embryonic induction and specification of the panplacodal primordium are discussed next. I then review how individual placodes arise from the panplacodal primordium and present a model of multistep placode induction. Finally, I briefly summarize recent advances concerning how placodal neurons and sensory cells are specified, and how morphogenesis of placodes (including delamination and migration of placode-derived cells and invagination) is controlled.
Collapse
Affiliation(s)
- Gerhard Schlosser
- Zoology, School of Natural Sciences & Martin Ryan Institute, National University of Ireland, Galway, Ireland
| |
Collapse
|
39
|
Zhang Q, Zheng K, Ma S, Tong Y, Luo C. Goldfish β-catenin cell-autonomously inhibits the expression of early neural development regulating gene vsx1. CHINESE SCIENCE BULLETIN-CHINESE 2010. [DOI: 10.1007/s11434-009-0714-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
40
|
BMP inhibition initiates neural induction via FGF signaling and Zic genes. Proc Natl Acad Sci U S A 2009; 106:17437-42. [PMID: 19805078 DOI: 10.1073/pnas.0906352106] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Neural induction is the process that initiates nervous system development in vertebrates. Two distinct models have been put forward to describe this phenomenon in molecular terms. The default model states that ectoderm cells are fated to become neural in absence of instruction, and do so when bone morphogenetic protein (BMP) signals are abolished. A more recent view implicates a conserved role for FGF signaling that collaborates with BMP inhibition to allow neural fate specification. Using the Xenopus embryo, we obtained evidence that may unite the 2 views. We show that a dominant-negative R-Smad, Smad5-somitabun-unlike the other BMP inhibitors used previously-can trigger conversion of Xenopus epidermis into neural tissue in vivo. However, it does so only if FGF activity is uncompromised. We report that this activity may be encoded by FGF4, as its expression is activated upon BMP inhibition, and its knockdown suppresses endogenous, as well as ectopic, neural induction by Smad5-somitabun. Supporting the importance of FGF instructive activity, we report the isolation of 2 immediate early neural targets, zic3 and foxD5a. Conversely, we found that zic1 can be activated by BMP inhibition in the absence of translation. Finally, Zic1 and Zic3 are required together for definitive neural fate acquisition, both in ectopic and endogenous situations. We propose to merge the previous models into a unique one whereby neural induction is controlled by BMP inhibition, which activates directly, and, via FGF instructive activity, early neural regulators such as Zic genes.
Collapse
|
41
|
Rogers C, Moody SA, Casey E. Neural induction and factors that stabilize a neural fate. BIRTH DEFECTS RESEARCH. PART C, EMBRYO TODAY : REVIEWS 2009; 87:249-62. [PMID: 19750523 PMCID: PMC2756055 DOI: 10.1002/bdrc.20157] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The neural ectoderm of vertebrates forms when the bone morphogenetic protein (BMP) signaling pathway is suppressed. Herein, we review the molecules that directly antagonize extracellular BMP and the signaling pathways that further contribute to reduce BMP activity in the neural ectoderm. Downstream of neural induction, a large number of "neural fate stabilizing" (NFS) transcription factors are expressed in the presumptive neural ectoderm, developing neural tube and ultimately in neural stem cells. Herein, we review what is known about their activities during normal development to maintain a neural fate and regulate neural differentiation. Further elucidation of how the NFS genes interact to regulate neural specification and differentiation should ultimately prove useful for regulating the expansion and differentiation of neural stem and progenitor cells.
Collapse
Affiliation(s)
| | - Sally A. Moody
- Department of Anatomy and Regenerative Biology, The George Washington University
| | - Elena Casey
- Department of Biology, Georgetown University
| |
Collapse
|
42
|
Wang X, Lee JE, Dorsky RI. Identification of Wnt-responsive cells in the zebrafish hypothalamus. Zebrafish 2009; 6:49-58. [PMID: 19374548 DOI: 10.1089/zeb.2008.0570] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
In all vertebrate brains, there is a period of widespread embryonic neurogenesis followed by specific regional neurogenesis that continues into adult stages. The Wnt signaling pathway, which is essential for numerous developmental processes, has also been suggested to be involved in neurogenesis. To help investigate the exact roles of canonical Wnt signaling in neurogenesis, here we examine the identity of Wnt-responsive cells in the zebrafish hypothalamus. This tissue is a useful diencephalic neurogenesis model containing evolutionarily conserved populations of neurons. We first performed in situ hybridization to show the expression patterns of Tcf family members and a canonical Wnt signaling reporter in the 50 hpf embryonic hypothalamus and larval/adult hypothalamus. We then used immunohistochemistry to identify the cell types of Wnt-responsive and Lef1-positive cells in both 50 hpf embryonic and adult hypothalamus. Our results indicate that Wnt-responsive cells in the hypothalamus are likely to be both mitotic progenitors and postmitotic precursors at embryonic stages, but only precursors at the adult stage. These data suggest that canonical Wnt signaling may be functionally required for maintenance of neural progenitor and precursor pools in the embryo, and for ongoing neurogenesis in the adult zebrafish.
Collapse
Affiliation(s)
- Xu Wang
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah 84132, USA
| | | | | |
Collapse
|
43
|
Zoltewicz JS, Ashique AM, Choe Y, Lee G, Taylor S, Phamluong K, Solloway M, Peterson AS. Wnt signaling is regulated by endoplasmic reticulum retention. PLoS One 2009; 4:e6191. [PMID: 19593386 PMCID: PMC2703784 DOI: 10.1371/journal.pone.0006191] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2009] [Accepted: 06/10/2009] [Indexed: 12/11/2022] Open
Abstract
Precise regulation of Wnt signaling is important in many contexts, as in development of the vertebrate forebrain, where excessive or ectopic Wnt signaling leads to severe brain defects. Mutation of the widely expressed oto gene causes loss of the anterior forebrain during mouse embryogenesis. Here we report that oto is the mouse ortholog of the gpi deacylase gene pgap1, and that the endoplasmic reticulum (ER)-resident Oto protein has a novel and deacylase-independent function during Wnt maturation. Oto increases the hydrophobicities of Wnt3a and Wnt1 by promoting the addition of glycophosphatidylinositol (gpi)-like anchors to these Wnts, which results in their retention in the ER. We also report that oto-deficient embryos exhibit prematurely robust Wnt activity in the Wnt1 domain of the early neural plate. We examine the effect of low oto expression on Wnt1 in vitro by knocking down endogenous oto expression in 293 and M14 melanoma cells using shRNA. Knockdown of oto results in increased Wnt1 secretion which is correlated with greatly enhanced canonical Wnt activity. These data indicate that oto deficiency increases Wnt signaling in vivo and in vitro. Finally, we address the mechanism of Oto-mediated Wnt retention under oto-abundant conditions, by cotransfecting Wnt1 with gpi-specific phospholipase D (GPI-PLD). The presence of GPI-PLD in the secretory pathway results in increased secretion of soluble Wnt1, suggesting that the gpi-like anchor lipids on Wnt1 mediate its retention in the ER. These data now provide a mechanistic framework for understanding the forebrain defects in oto mice, and support a role for Oto-mediated Wnt regulation during early brain development. Our work highlights a critical role for ER retention in regulating Wnt signaling in the mouse embryo, and gives insight into the notoriously inefficient secretion of Wnts.
Collapse
Affiliation(s)
- J. Susie Zoltewicz
- Ernest Gallo Clinic & Research Center, Emeryville, California, United States of America
- * E-mail: (JSZ); (ASP)
| | - Amir M. Ashique
- Ernest Gallo Clinic & Research Center, Emeryville, California, United States of America
| | - Youngshik Choe
- Ernest Gallo Clinic & Research Center, Emeryville, California, United States of America
| | - Gena Lee
- Ernest Gallo Clinic & Research Center, Emeryville, California, United States of America
| | - Stacy Taylor
- Ernest Gallo Clinic & Research Center, Emeryville, California, United States of America
| | - Khanhky Phamluong
- Ernest Gallo Clinic & Research Center, Emeryville, California, United States of America
| | - Mark Solloway
- Ernest Gallo Clinic & Research Center, Emeryville, California, United States of America
| | - Andrew S. Peterson
- Ernest Gallo Clinic & Research Center, Emeryville, California, United States of America
- * E-mail: (JSZ); (ASP)
| |
Collapse
|
44
|
Kim H, Cheong SM, Ryu J, Jung HJ, Jho EH, Han JK. Xenopus Wntless and the retromer complex cooperate to regulate XWnt4 secretion. Mol Cell Biol 2009; 29:2118-28. [PMID: 19223472 PMCID: PMC2663306 DOI: 10.1128/mcb.01503-08] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2008] [Revised: 10/27/2008] [Accepted: 02/02/2009] [Indexed: 11/20/2022] Open
Abstract
Wnt signaling is implicated in a variety of developmental and pathological processes. The molecular mechanisms governing the secretion of Wnt ligands remain to be elucidated. Wntless, an evolutionarily conserved multipass transmembrane protein, is a dedicated secretion factor of Wnt proteins that participates in Drosophila melanogaster embryogenesis. In this study, we show that Xenopus laevis Wntless (XWntless) regulates the secretion of a specific Wnt ligand, XWnt4, and that this regulation is specifically required for eye development in Xenopus. Moreover, the Retromer complex is required for XWntless recycling to regulate the XWnt4-mediated eye development. Inhibition of Retromer function by Vps35 morpholino (MO) resulted in various Wnt deficiency phenotypes, affecting mesoderm induction, gastrulation cell movements, neural induction, neural tube closure, and eye development. Overexpression of XWntless led to the rescue of Vps35 MO-mediated eye defects but not other deficiencies. These results collectively suggest that XWntless and the Retromer complex are required for the efficient secretion of XWnt4, facilitating its role in Xenopus eye development.
Collapse
Affiliation(s)
- Hyunjoon Kim
- Division of Molecular and Life Sciences, Pohang University of Science and Technology, Pohang, Kyungbuk 790-784, Republic of Korea
| | | | | | | | | | | |
Collapse
|
45
|
Steventon B, Araya C, Linker C, Kuriyama S, Mayor R. Differential requirements of BMP and Wnt signalling during gastrulation and neurulation define two steps in neural crest induction. Development 2009; 136:771-9. [PMID: 19176585 DOI: 10.1242/dev.029017] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The neural crest is induced by a combination of secreted signals. Although previous models of neural crest induction have proposed a step-wise activation of these signals, the actual spatial and temporal requirement has not been analysed. Through analysing the role of the mesoderm we show for the first time that specification of neural crest requires two temporally and chemically different steps: first, an induction at the gastrula stage dependent on signals arising from the dorsolateral mesoderm; and second, a maintenance step at the neurula stage dependent on signals from tissues adjacent to the neural crest. By performing tissue recombination experiments and using specific inhibitors of different inductive signals, we show that the first inductive step requires Wnt activation and BMP inhibition, whereas the later maintenance step requires activation of both pathways. This change in BMP necessity from BMP inhibition at gastrula to BMP activation at neurula stages is further supported by the dynamic expression of BMP4 and its antagonists, and is confirmed by direct measurements of BMP activity in the neural crest cells. The differential requirements of BMP activity allow us to propose an explanation for apparently discrepant results between chick and frog experiments. The demonstration that Wnt signals are required for neural crest induction by mesoderm solves an additional long-standing controversy. Finally, our results emphasise the importance of considering the order of exposure to signals during an inductive event.
Collapse
Affiliation(s)
- Ben Steventon
- Department of Cell and Developmental Biology, University College London, London, UK
| | | | | | | | | |
Collapse
|
46
|
Gaulden J, Reiter JF. Neur-ons and neur-offs: regulators of neural induction in vertebrate embryos and embryonic stem cells. Hum Mol Genet 2008; 17:R60-6. [PMID: 18632699 DOI: 10.1093/hmg/ddn119] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Although the spatial and temporal orchestration of early vertebrate embryogenesis is missing from cell culture systems, recent work suggests that many of the same signals affecting neural induction in vertebrate embryos also regulate embryonic stem (ES) cell neurogenesis. One key regulatory mechanism involved in both in vivo and in vitro neural induction is the inhibition of bone morphogenetic protein (BMP) signals. Wnts and Fibroblast Growth Factors represent additional regulatory influences, which may affect the adoption of neural fates through both BMP-dependent and BMP-independent mechanisms. Insights into neural induction in vivo help to guide paradigms for promoting neural differentiation by ES cells. Conversely, insights into the mechanisms by which ES cells adopt neural fates may provide an improved understanding of neural induction in the early embryo.
Collapse
Affiliation(s)
- Julie Gaulden
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, CA 94158-2324, USA
| | | |
Collapse
|
47
|
van Venrooy S, Fichtner D, Kunz M, Wedlich D, Gradl D. Cold-inducible RNA binding protein (CIRP), a novel XTcf-3 specific target gene regulates neural development in Xenopus. BMC DEVELOPMENTAL BIOLOGY 2008; 8:77. [PMID: 18687117 PMCID: PMC2527318 DOI: 10.1186/1471-213x-8-77] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2008] [Accepted: 08/07/2008] [Indexed: 12/11/2022]
Abstract
BACKGROUND As nuclear mediators of wnt/beta-catenin signaling, Lef/Tcf transcription factors play important roles in development and disease. Although it is well established, that the four vertebrate Lef/Tcfs have unique functional properties, most studies unite Lef-1, Tcf-1, Tcf-3 and Tcf-4 and reduce their function to uniformly transduce wnt/beta-catenin signaling for activating wnt target genes. In order to discriminate target genes regulated by XTcf-3 from those regulated by XTcf-4 or Lef/Tcfs in general, we performed a subtractive screen, using neuralized Xenopus animal cap explants. RESULTS We identified cold-inducible RNA binding protein (CIRP) as novel XTcf-3 specific target gene. Furthermore, we show that knockdown of XTcf-3 by injection of an antisense morpholino oligonucleotide results in a general broadening of the anterior neural tissue. Depletion of XCIRP by antisense morpholino oligonucleotide injection leads to a reduced stability of mRNA and an enlargement of the anterior neural plate similar to the depletion of XTcf-3. CONCLUSION Distinct steps in neural development are differentially regulated by individual Lef/Tcfs. For proper development of the anterior brain XTcf-3 and the Tcf-subtype specific target XCIRP appear indispensable. Thus, regulation of anterior neural development, at least in part, depends on mRNA stabilization by the novel XTcf-3 target gene XCIRP.
Collapse
|
48
|
Sox3 regulates both neural fate and differentiation in the zebrafish ectoderm. Dev Biol 2008; 320:289-301. [DOI: 10.1016/j.ydbio.2008.05.542] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2007] [Revised: 05/14/2008] [Accepted: 05/15/2008] [Indexed: 01/21/2023]
|
49
|
Chang C, Harland RM. Neural induction requires continued suppression of both Smad1 and Smad2 signals during gastrulation. Development 2008; 134:3861-72. [PMID: 17933792 DOI: 10.1242/dev.007179] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Vertebrate neural induction requires inhibition of bone morphogenetic protein (BMP) signaling in the ectoderm. However, whether inhibition of BMP signaling is sufficient to induce neural tissues in vivo remains controversial. Here we have addressed why inhibition of BMP/Smad1 signaling does not induce neural markers efficiently in Xenopus ventral ectoderm, and show that suppression of both Smad1 and Smad2 signals is sufficient to induce neural markers. Manipulations that inhibit both Smad1 and Smad2 pathways, including a truncated type IIB activin receptor, Smad7 and Ski, induce early neural markers and inhibit epidermal genes in ventral ectoderm; and co-expression of BMP inhibitors with a truncated activin/nodal-specific type IB activin receptor leads to efficient neural induction. Conversely, stimulation of Smad2 signaling in the neural plate at gastrula stages results in inhibition of neural markers, disruption of the neural tube and reduction of head structures, with conversion of neural to neural crest and mesodermal fates. The ability of activated Smad2 to block neural induction declines by the end of gastrulation. Our results indicate that prospective neural cells are poised to respond to Smad2 and Smad1 signals to adopt mesodermal and non-neural ectodermal fates even at gastrula stages, after the conventionally assigned end of mesodermal competence, so that continued suppression of both mesoderm- and epidermis-inducing Smad signals leads to efficient neural induction.
Collapse
Affiliation(s)
- Chenbei Chang
- Department of Cell Biology, MCLM 360, University of Alabama at Birmingham, Birmingham, AL 35294-0005, USA.
| | | |
Collapse
|
50
|
Rogers CD, Archer TC, Cunningham DD, Grammer TC, Casey EMS. Sox3 expression is maintained by FGF signaling and restricted to the neural plate by Vent proteins in the Xenopus embryo. Dev Biol 2007; 313:307-19. [PMID: 18031719 DOI: 10.1016/j.ydbio.2007.10.023] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2007] [Revised: 10/03/2007] [Accepted: 10/18/2007] [Indexed: 11/30/2022]
Abstract
The formation of the nervous system is initiated when ectodermal cells adopt the neural fate. Studies in Xenopus demonstrate that inhibition of BMP results in the formation of neural tissue. However, the molecular mechanism driving the expression of early neural genes in response to this inhibition is unknown. Moreover, controversy remains regarding the sufficiency of BMP inhibition for neural induction. To address these questions, we performed a detailed analysis of the regulation of the soxB1 gene, sox3, one of the earliest genes expressed in the neuroectoderm. Using ectodermal explant assays, we analyzed the role of BMP, Wnt and FGF signaling in the regulation of sox3 and the closely related soxB1 gene, sox2. Our results demonstrate that both sox3 and sox2 are induced in response to BMP antagonism, but by distinct mechanisms and that the activation of both genes is independent of FGF signaling. However, both require FGF for the maintenance of their expression. Finally, sox3 genomic elements were identified and characterized and an element required for BMP-mediated repression via Vent proteins was identified through the use of transgenesis and computational analysis. Interestingly, none of the elements required for sox3 expression were identified in the sox2 locus. Together our data indicate that two closely related genes have unique mechanisms of gene regulation at the onset of neural development.
Collapse
Affiliation(s)
- Crystal D Rogers
- Department of Biology, Georgetown University, Washington, DC 20057, USA
| | | | | | | | | |
Collapse
|