1
|
Kukreja K, Jia BZ, McGeary SE, Patel N, Megason SG, Klein AM. Cell state transitions are decoupled from cell division during early embryo development. Nat Cell Biol 2024; 26:2035-2045. [PMID: 39516639 DOI: 10.1038/s41556-024-01546-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 09/23/2024] [Indexed: 11/16/2024]
Abstract
As tissues develop, cells divide and differentiate concurrently. Conflicting evidence shows that cell division is either dispensable or required for formation of cell types. Here, to determine the role of cell division in differentiation, we arrested the cell cycle in zebrafish embryos using two independent approaches and profiled them at single-cell resolution. We show that cell division is dispensable for differentiation of all embryonic tissues from early gastrulation to the end of segmentation. However, arresting cell division does slow down differentiation in some cell types, and it induces global stress responses. While differentiation is robust to blocking cell division, the proportions of cells across cell states are not, but show evidence of partial compensation. This work clarifies our understanding of the role of cell division in development and showcases the utility of combining embryo-wide perturbations with single-cell RNA sequencing to uncover the role of common biological processes across multiple tissues.
Collapse
Affiliation(s)
- Kalki Kukreja
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | - Bill Z Jia
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Sean E McGeary
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Nikit Patel
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sean G Megason
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Allon M Klein
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Alaiz Noya M, Berti F, Dietrich S. Comprehensive expression analysis for the core cell cycle regulators in the chicken embryo reveals novel tissue-specific synexpression groups and similarities and differences with expression in mouse, frog and zebrafish. J Anat 2022; 241:42-66. [PMID: 35146756 PMCID: PMC9178385 DOI: 10.1111/joa.13629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 12/07/2021] [Accepted: 01/05/2022] [Indexed: 11/29/2022] Open
Abstract
The core cell cycle machinery is conserved from yeast to humans, and hence it is assumed that all vertebrates share the same set of players. Yet during vertebrate evolution, the genome was duplicated twice, followed by a further genome duplication in teleost fish. Thereafter, distinct genes were retained in different vertebrate lineages; some individual gene duplications also occurred. To which extent these diversifying tendencies were compensated by retaining the same expression patterns across homologous genes is not known. This study for the first time undertook a comprehensive expression analysis for the core cell cycle regulators in the chicken, focusing in on early neurula and pharyngula stages of development, with the latter representing the vertebrate phylotypic stage. We also compared our data with published data for the mouse, Xenopus and zebrafish, the other established vertebrate models. Our work shows that, while many genes are expressed widely, some are upregulated or specifically expressed in defined tissues of the chicken embryo, forming novel synexpression groups with markers for distinct developmental pathways. Moreover, we found that in the neural tube and in the somite, mRNAs of some of the genes investigated accumulate in a specific subcellular localisation, pointing at a novel link between the site of mRNA translation, cell cycle control and interkinetic nuclear movements. Finally, we show that expression patterns of orthologous genes may differ in the four vertebrate models. Thus, for any study investigating cell proliferation, cell differentiation, tissue regeneration, stem cell behaviour and cancer/cancer therapy, it has to be carefully examined which of the observed effects are due to the specific model organism used, and which can be generalised.
Collapse
Affiliation(s)
- Marta Alaiz Noya
- Institute for Biomedical and Biomolecular Science (IBBS), School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK.,Instituto de Neurociencias de Alicante, Universidad Miguel Hernández - Consejo Superior de Investigaciones Científicas, Alicante, Spain
| | - Federica Berti
- Institute for Biomedical and Biomolecular Science (IBBS), School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK.,Life Sciences Solutions, Thermo Fisher Scientific, Monza, Italy
| | - Susanne Dietrich
- Institute for Biomedical and Biomolecular Science (IBBS), School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| |
Collapse
|
3
|
Ta AC, Huang LC, McKeown CR, Bestman JE, Van Keuren-Jensen K, Cline HT. Temporal and spatial transcriptomic dynamics across brain development in Xenopus laevis tadpoles. G3 (BETHESDA, MD.) 2022; 12:jkab387. [PMID: 34751375 PMCID: PMC8728038 DOI: 10.1093/g3journal/jkab387] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/26/2021] [Indexed: 11/13/2022]
Abstract
Amphibian metamorphosis is a transitional period that involves significant changes in the cell-type populations and biological processes occurring in the brain. Analysis of gene expression dynamics during this process may provide insight into the molecular events underlying these changes. We conducted differential gene expression analyses of the developing Xenopus laevis tadpole brain during this period in two ways: first, over stages of the development in the midbrain and, second, across regions of the brain at a single developmental stage. We found that genes pertaining to positive regulation of neural progenitor cell proliferation as well as known progenitor cell markers were upregulated in the midbrain prior to metamorphic climax; concurrently, expression of cell cycle timing regulators decreased across this period, supporting the notion that cell cycle lengthening contributes to a decrease in proliferation by the end of metamorphosis. We also found that at the start of metamorphosis, neural progenitor populations appeared to be similar across the fore-, mid-, and hindbrain regions. Genes pertaining to negative regulation of differentiation were upregulated in the spinal cord compared to the rest of the brain, however, suggesting that different programs may regulate neurogenesis there. Finally, we found that regulation of biological processes like cell fate commitment and synaptic signaling follow similar trajectories in the brain across early tadpole metamorphosis and mid- to late-embryonic mouse development. By comparing expression across both temporal and spatial conditions, we have been able to illuminate cell-type and biological pathway dynamics in the brain during metamorphosis.
Collapse
Affiliation(s)
- Aaron C Ta
- Neuroscience Department and The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Department of Neuroscience, University of California, San Diego, La Jolla, CA 92037, USA
| | - Lin-Chien Huang
- Neuroscience Department and The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Caroline R McKeown
- Neuroscience Department and The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jennifer E Bestman
- Neuroscience Department and The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | - Hollis T Cline
- Neuroscience Department and The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
4
|
Chen L, Zhang M, Fang L, Yang X, Cao N, Xu L, Shi L, Cao Y. Coordinated regulation of the ribosome and proteasome by PRMT1 in the maintenance of neural stemness in cancer cells and neural stem cells. J Biol Chem 2021; 297:101275. [PMID: 34619150 PMCID: PMC8546425 DOI: 10.1016/j.jbc.2021.101275] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/19/2021] [Accepted: 09/30/2021] [Indexed: 12/17/2022] Open
Abstract
Previous studies suggested that cancer cells resemble neural stem/progenitor cells in regulatory network, tumorigenicity, and differentiation potential, and that neural stemness might represent the ground or basal state of differentiation and tumorigenicity. The neural ground state is reflected in the upregulation and enrichment of basic cell machineries and developmental programs, such as cell cycle, ribosomes, proteasomes, and epigenetic factors, in cancers and in embryonic neural or neural stem cells. However, how these machineries are concertedly regulated is unclear. Here, we show that loss of neural stemness in cancer or neural stem cells via muscle-like differentiation or neuronal differentiation, respectively, caused downregulation of ribosome and proteasome components and major epigenetic factors, including PRMT1, EZH2, and LSD1. Furthermore, inhibition of PRMT1, an oncoprotein that is enriched in neural cells during embryogenesis, caused neuronal-like differentiation, downregulation of a similar set of proteins downregulated by differentiation, and alteration of subcellular distribution of ribosome and proteasome components. By contrast, PRMT1 overexpression led to an upregulation of these proteins. PRMT1 interacted with these components and protected them from degradation via recruitment of the deubiquitinase USP7, also known to promote cancer and enriched in embryonic neural cells, thereby maintaining a high level of epigenetic factors that maintain neural stemness, such as EZH2 and LSD1. Taken together, our data indicate that PRMT1 inhibition resulted in repression of cell tumorigenicity. We conclude that PRMT1 coordinates ribosome and proteasome activity to match the needs for high production and homeostasis of proteins that maintain stemness in cancer and neural stem cells.
Collapse
Affiliation(s)
- Lu Chen
- Research Institute of Nanjing University in Shenzhen, Shenzhen, China; MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China; Jiangsu Key Laboratory of Molecular Medicine of the Medical School, Nanjing University, Nanjing, China
| | - Min Zhang
- Research Institute of Nanjing University in Shenzhen, Shenzhen, China; MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China; Jiangsu Key Laboratory of Molecular Medicine of the Medical School, Nanjing University, Nanjing, China
| | - Lei Fang
- Jiangsu Key Laboratory of Molecular Medicine of the Medical School, Nanjing University, Nanjing, China
| | - Xiaoli Yang
- Research Institute of Nanjing University in Shenzhen, Shenzhen, China; MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China; Jiangsu Key Laboratory of Molecular Medicine of the Medical School, Nanjing University, Nanjing, China
| | - Ning Cao
- Research Institute of Nanjing University in Shenzhen, Shenzhen, China; MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China; Jiangsu Key Laboratory of Molecular Medicine of the Medical School, Nanjing University, Nanjing, China
| | - Liyang Xu
- MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China; Jiangsu Key Laboratory of Molecular Medicine of the Medical School, Nanjing University, Nanjing, China
| | - Lihua Shi
- MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China; Jiangsu Key Laboratory of Molecular Medicine of the Medical School, Nanjing University, Nanjing, China
| | - Ying Cao
- Research Institute of Nanjing University in Shenzhen, Shenzhen, China; MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, China; Jiangsu Key Laboratory of Molecular Medicine of the Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
5
|
Neural is Fundamental: Neural Stemness as the Ground State of Cell Tumorigenicity and Differentiation Potential. Stem Cell Rev Rep 2021; 18:37-55. [PMID: 34714532 DOI: 10.1007/s12015-021-10275-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2021] [Indexed: 01/07/2023]
Abstract
Tumorigenic cells are similar to neural stem cells or embryonic neural cells in regulatory networks, tumorigenicity and pluripotent differentiation potential. By integrating the evidence from developmental biology, tumor biology and evolution, I will make a detailed discussion on the observations and propose that neural stemness underlies two coupled cell properties, tumorigenicity and pluripotent differentiation potential. Neural stemness property of tumorigenic cells can hopefully integrate different observations/concepts underlying tumorigenesis. Neural stem cells and tumorigenic cells share regulatory networks; both exhibit neural stemness, tumorigenicity and pluripotent differentiation potential; both depend on expression or activation of ancestral genes; both rely primarily on aerobic glycolytic metabolism; both can differentiate into various cells/tissues that are derived from three germ layers, leading to tumor formation resembling severely disorganized or more degenerated process of embryonic tissue differentiation; both are enriched in long genes with more splice variants that provide more plastic scaffolds for cell differentiation, etc. Neural regulatory networks, which include higher levels of basic machineries of cell physiological functions and developmental programs, work concertedly to define a basic state with fast cell cycle and proliferation. This is predestined by the evolutionary advantage of neural state, the ground or initial state for multicellularity with adaptation to an ancient environment. Tumorigenesis might represent a process of restoration of neural ground state, thereby restoring a state with fast proliferation and pluripotent differentiation potential in somatic cells. Tumorigenesis and pluripotent differentiation potential might be better understood from understanding neural stemness, and cancer therapy should benefit more from targeting neural stemness.
Collapse
|
6
|
Philpott A. The Use of Xenopus for Cell Biology Applications. Cold Spring Harb Protoc 2021; 2021:pdb.top105528. [PMID: 33782096 DOI: 10.1101/pdb.top105528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Problems of cell biology and the molecular controls underpinning them have been studied in the remarkably versatile Xenopus systems for many years. This versatility is showcased in several accompanying protocols, which are introduced here. One protocol demonstrates how the Xenopus embryonic ectoderm can be used to study the effects of mechanical cell deformation; another illustrates how the developing eye can be used as a platform for determining cell-cycle length. Two protocols show how extracts from Xenopus embryos can be exploited to characterize the behavior of specific intracellular proteins-specifically, to determine protein phosphorylation status and the ability to bind to chromatin. Finally, because specific antibodies to Xenopus proteins are pivotal reagents for cell biology and biochemistry applications, four protocols describing how to generate, purify, and assay the specificity of antibodies raised against Xenopus proteins are included in hopes of stimulating the expansion of these critical resources across the Xenopus community.
Collapse
Affiliation(s)
- Anna Philpott
- Department of Oncology, University of Cambridge, Cambridge, CB2 0XZ, United Kingdom; .,Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, CB2 0AW, United Kingdom
| |
Collapse
|
7
|
Lien HW, Yuan RY, Chou CM, Chen YC, Hung CC, Hu CH, Hwang SPL, Hwang PP, Shen CN, Chen CL, Cheng CH, Huang CJ. Zebrafish cyclin Dx is required for development of motor neuron progenitors, and its expression is regulated by hypoxia-inducible factor 2α. Sci Rep 2016; 6:28297. [PMID: 27323909 PMCID: PMC4915019 DOI: 10.1038/srep28297] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 06/01/2016] [Indexed: 01/31/2023] Open
Abstract
Cyclins play a central role in cell-cycle regulation; in mammals, the D family of cyclins consists of cyclin D1, D2, and D3. In Xenopus, only homologs of cyclins D1 and D2 have been reported, while a novel cyclin, cyclin Dx (ccndx), was found to be required for the maintenance of motor neuron progenitors during embryogenesis. It remains unknown whether zebrafish possess cyclin D3 or cyclin Dx. In this study, we identified a zebrafish ccndx gene encoding a protein which can form a complex with Cdk4. Through whole-mount in situ hybridization, we observed that zccndx mRNA is expressed in the motor neurons of hindbrain and spinal cord during development. Analysis of a 4-kb promoter sequence of the zccndx gene revealed the presence of HRE sites, which can be regulated by HIF2α. Morpholino knockdown of zebrafish Hif2α and cyclin Dx resulted in the abolishment of isl1 and oligo2 expression in the precursors of motor neurons, and also disrupted axon growth. Overexpression of cyclin Dx mRNA in Hif2α morphants partially rescued zccndx expression. Taken together, our data indicate that zebrafish cyclin Dx plays a role in maintaining the precursors of motor neurons.
Collapse
Affiliation(s)
- Huang-Wei Lien
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 115, Taiwan
| | - Rey-Yue Yuan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 115, Taiwan
| | - Chih-Ming Chou
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 115, Taiwan
| | - Yi-Chung Chen
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 115, Taiwan
| | - Chin-Chun Hung
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Chin-Hwa Hu
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung 20224, Taiwan
| | - Sheng-Ping L Hwang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 115, Taiwan
| | - Pung-Pung Hwang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 115, Taiwan
| | - Chia-Ning Shen
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Chih-Lung Chen
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Chia-Hsiung Cheng
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 115, Taiwan
| | - Chang-Jen Huang
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
8
|
Sun T, Li W, Ling S. miR-30c and semaphorin 3A determine adult neurogenesis by regulating proliferation and differentiation of stem cells in the subventricular zones of mouse. Cell Prolif 2016; 49:270-80. [PMID: 27198082 DOI: 10.1111/cpr.12261] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 03/31/2016] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES Mechanisms that regulate proliferation of adult neural stem cells are largely unknown. Here, we have investigated the role of microR-30c (miR-30c) and its target, semaphoring 3A (sema3A), in regulating adult neurogenesis and mechanisms underlying this process. MATERIALS AND METHODS In situ hybridization, immunofluorescence and quantitative real-time PCR were used to assess complementary expression patterns of miR-30c and sema3A in mice. Effects of miR-30c in the subventricular zone (SVZ) were examined by stereotaxic injection of up- and down-regulating lentiviruses. 5'-bromo-2'-deoxyuridine labelling was performed to investigate effects of miR-30c and sema3A on adult neurogenesis. Real-time cell assays, morphological analysis and cell cycle measurements were used to reveal the mechanisms by which miR-30c and sema3A regulate adult neurogenesis. RESULTS Expression of miR-30c negatively correlated with that of sema3A in neurons, and levels of miR-30c and sema3A correlated positively with numbers of newborn cells in the SVZ and rostral migration stream. miR-30c and sema3A affected adult neurogenesis by regulating proliferation and differentiation, as well as cycles of stem cells in the SVZ. CONCLUSIONS miR-30c and sema3A regulate adult neurogenesis by controlling proliferation and differentiation of stem cells in the SVZ. This finding reveals a novel regulatory mechanism of adult neurogenesis.
Collapse
Affiliation(s)
- Tingting Sun
- Institute of Neuroscience and Anatomy, Zhejiang University, School of Medicine, Hangzhou, 310058, China
| | - Weiyun Li
- Institute of Neuroscience and Anatomy, Zhejiang University, School of Medicine, Hangzhou, 310058, China
| | - Shucai Ling
- Institute of Neuroscience and Anatomy, Zhejiang University, School of Medicine, Hangzhou, 310058, China
| |
Collapse
|
9
|
The Inner Nuclear Membrane Protein Nemp1 Is a New Type of RanGTP-Binding Protein in Eukaryotes. PLoS One 2015; 10:e0127271. [PMID: 25946333 PMCID: PMC4422613 DOI: 10.1371/journal.pone.0127271] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 04/13/2015] [Indexed: 12/04/2022] Open
Abstract
The inner nuclear membrane (INM) protein Nemp1/TMEM194A has previously been suggested to be involved in eye development in Xenopus, and contains two evolutionarily conserved sequences in the transmembrane domains (TMs) and the C-terminal region, named region A and region B, respectively. To elucidate the molecular nature of Nemp1, we analyzed its interacting proteins through those conserved regions. First, we found that Nemp1 interacts with itself and lamin through the TMs and region A, respectively. Colocalization of Nemp1 and lamin at the INM suggests that the interaction with lamin participates in the INM localization of Nemp1. Secondly, through yeast two-hybrid screening using region B as bait, we identified the small GTPase Ran as a probable Nemp1-binding partner. GST pulldown and co-immunoprecipitation assays using region B and Ran mutants revealed that region B binds directly to the GTP-bound Ran through its effector domain. Immunostaining experiments using transfected COS-7 cells revealed that full-length Nemp1 recruits Ran near the nuclear envelope, suggesting a role for Nemp1 in the accumulation of RanGTP at the nuclear periphery. At the neurula-to-tailbud stages of Xenopus embryos, nemp1 expression overlapped with ran in several regions including the eye vesicles. Co-knockdown using antisense morpholino oligos for nemp1 and ran caused reduction of cell densities and severe eye defects more strongly than either single knockdown alone, suggesting their functional interaction. Finally we show that Arabidopsis thaliana Nemp1-orthologous proteins interact with A. thaliana Ran, suggesting their evolutionally conserved physical and functional interactions possibly in basic cellular functions including nuclear transportation. Taken together, we conclude that Nemp1 represents a new type of RanGTP-binding protein.
Collapse
|
10
|
Hardwick LJA, Ali FR, Azzarelli R, Philpott A. Cell cycle regulation of proliferation versus differentiation in the central nervous system. Cell Tissue Res 2014; 359:187-200. [PMID: 24859217 PMCID: PMC4284380 DOI: 10.1007/s00441-014-1895-8] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 04/10/2014] [Indexed: 01/07/2023]
Abstract
Formation of the central nervous system requires a period of extensive progenitor cell proliferation, accompanied or closely followed by differentiation; the balance between these two processes in various regions of the central nervous system gives rise to differential growth and cellular diversity. The correlation between cell cycle lengthening and differentiation has been reported across several types of cell lineage and from diverse model organisms, both in vivo and in vitro. Furthermore, different cell fates might be determined during different phases of the preceding cell cycle, indicating direct cell cycle influences on both early lineage commitment and terminal cell fate decisions. Significant advances have been made in the last decade and have revealed multi-directional interactions between the molecular machinery regulating the processes of cell proliferation and neuronal differentiation. Here, we first introduce the modes of proliferation in neural progenitor cells and summarise evidence linking cell cycle length and neuronal differentiation. Second, we describe the manner in which components of the cell cycle machinery can have additional and, sometimes, cell-cycle-independent roles in directly regulating neurogenesis. Finally, we discuss the way that differentiation factors, such as proneural bHLH proteins, can promote either progenitor maintenance or differentiation according to the cellular environment. These intricate connections contribute to precise coordination and the ultimate division versus differentiation decision.
Collapse
Affiliation(s)
- Laura J A Hardwick
- Department of Oncology, Hutchison/MRC Research Centre, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
| | | | | | | |
Collapse
|
11
|
Abstract
The G1 cyclins play a pivotal role in regulation of cell differentiation and proliferation. The mechanisms underlying their cell-specific roles are incompletely understood. Here, we show that a G1 cyclin, cyclin D2 (CycD2), enhances the activity of transcription factor GATA4, a key regulator of cardiomyocyte growth and differentiation. GATA4 recruits CycD2 to its target promoters, and their interaction results in synergistic activation of GATA-dependent transcription. This effect is specific to CycD2 because CycD1 is unable to potentiate activity of GATA4 and is CDK-independent. GATA4 physically interacts with CycD2 through a discreet N-terminal activation domain that is essential for the cardiogenic activity of GATA4. Human mutations in this domain that are linked to congenital heart disease interfere with CycD2-GATA4 synergy. Cardiogenesis assays in Xenopus embryos indicate that CycD2 enhances the cardiogenic function of GATA4. Together, our data uncover a role for CycD2 as a cardiogenic coactivator of GATA4 and suggest a paradigm for cell-specific effects of cyclin Ds.
Collapse
|
12
|
Loss of Xenopus cadherin-11 leads to increased Wnt/β-catenin signaling and up-regulation of target genes c-myc and cyclin D1 in neural crest. Dev Biol 2013; 383:132-45. [PMID: 23958437 DOI: 10.1016/j.ydbio.2013.08.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 07/18/2013] [Accepted: 08/09/2013] [Indexed: 01/15/2023]
Abstract
Xenopus cadherin-11 (Xcadherin-11) is an exceptional cadherin family member, which is predominantly expressed in cranial neural crest cells (NCCs). Apart from mediating cell-cell adhesion it promotes cranial NCC migration by initiating filopodia and lamellipodia formation. Here, we demonstrate an unexpected function of Xcadherin-11 in NCC specification by interfering with canonical Wnt/β-catenin signaling. Loss-of-function experiments, using a specific antisense morpholino oligonucleotide against Xcadherin-11, display a nuclear β-catenin localization in cranial NCCs and a broader expression domain of the proto-oncogene cyclin D1 which proceeds c-myc up-regulation. Additionally, we observe an enhanced NCC proliferation and an expansion of specific NCC genes like AP2 and Sox10. Thereby, we could allocate NCC proliferation and specification to different gene functions. To clarify which domain in Xcadherin-11 is required for early NCC development we tested different deletion mutants for their rescue ability in Xcadherin-11 morphants. We identified the cytoplasmic tail, specifically the β-catenin binding domain, to be necessary for proper NCC development. We propose that Xcadherin-11 is necessary for controlled NCC proliferation and early NCC specification in tuning the expression of the canonical Wnt/β-catenin target genes cyclin D1 and c-myc by regulating the concentration of the nuclear pool of β-catenin.
Collapse
|
13
|
Aldiri I, Moore KB, Hutcheson DA, Zhang J, Vetter ML. Polycomb repressive complex PRC2 regulates Xenopus retina development downstream of Wnt/β-catenin signaling. Development 2013; 140:2867-78. [PMID: 23739135 DOI: 10.1242/dev.088096] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The histone methyltransferase complex PRC2 controls key steps in developmental transitions and cell fate choices; however, its roles in vertebrate eye development remain unknown. Here, we report that in Xenopus, PRC2 regulates the progression of retinal progenitors from proliferation to differentiation. We show that the PRC2 core components are enriched in retinal progenitors and downregulated in differentiated cells. Knockdown of the PRC2 core component Ezh2 leads to reduced retinal progenitor proliferation, in part due to upregulation of the Cdk inhibitor p15(Ink4b). In addition, although PRC2 knockdown does not alter eye patterning, retinal progenitor gene expression or expression of the neural competence factor Sox2, it does cause suppression of proneural bHLH gene expression, indicating that PRC2 is crucial for the initiation of neural differentiation in the retina. Consistent with this, knocking down or blocking PRC2 function constrains the generation of most retinal neural cell types and promotes a Müller glial cell fate decision. We also show that Wnt/β-catenin signaling acting through the receptor Frizzled 5, but independent of Sox2, regulates expression of key PRC2 subunits in the developing retina. This is consistent with a role for this pathway in coordinating proliferation and the transition to neurogenesis in the Xenopus retina. Our data establish PRC2 as a regulator of proliferation and differentiation during eye development.
Collapse
Affiliation(s)
- Issam Aldiri
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | | | | | | | | |
Collapse
|
14
|
Nath K, Fisher C, Elinson RP. Expression of cyclin D1, cyclin D2, and N-myc in embryos of the direct developing frog Eleutherodactylus coqui, with a focus on limbs. Gene Expr Patterns 2013; 13:142-9. [PMID: 23473789 PMCID: PMC3657300 DOI: 10.1016/j.gep.2013.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 02/18/2013] [Accepted: 02/23/2013] [Indexed: 11/16/2022]
Abstract
Species of frogs that develop directly have removed the tadpole from their ontogeny and form adult structures precociously. To see whether cell cycle regulators could be involved in this altered embryogenesis, we examined the expression of ccnd1, ccnd2, and mycn in embryos of the direct developing frog, Eleutherodactylus coqui. Notable differences compared to embryos of Xenopus laevis, a species with a tadpole, included prominent expression of ccnd2 in the midbrain and ccnd1 in the mandibular neural crest. The former may contribute to the precocious appearance of the adult-type visual system and the latter to the adult-type jaw. Large domains of ccnd2 and mycn presage the early appearance of limb buds, and ccnd1 and mycn are implicated in digit development.
Collapse
Affiliation(s)
- Kimberly Nath
- Department of Biological Sciences, Duquesne University, 600 Forbes
Avenue, Pittsburgh, PA 15282, U.S.A
| | - Cara Fisher
- Department of Biological Sciences, Duquesne University, 600 Forbes
Avenue, Pittsburgh, PA 15282, U.S.A
| | - Richard P. Elinson
- Department of Biological Sciences, Duquesne University, 600 Forbes
Avenue, Pittsburgh, PA 15282, U.S.A
| |
Collapse
|
15
|
Xue XY, Harris WA. Using myc genes to search for stem cells in the ciliary margin of the Xenopus retina. Dev Neurobiol 2012; 72:475-90. [PMID: 21465669 DOI: 10.1002/dneu.20887] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The ciliary marginal zone (CMZ) of fish and frog retinas contains cells that proliferate throughout postembryonic development as the retina grows with increasing body size, indicating the presence of stem cells in this region. However, neither the location nor the molecular identity of retinal stem cells has been identified. Here, we show in Xenopus that c-myc and n-myc are sequentially expressed both during development and in the post-embryonic retina. The c-myc+/n-myc- cells near the extreme periphery of the CMZ cycle more slowly and preferentially retain DNA label compared to their more central cmyc+/n-myc+ neighbors which cycle rapidly and preferentially dilute DNA label. During retinal development c-myc is functionally required earlier than n-myc, and n-myc expression depends on earlier c-myc expression. The expression of c-myc but not n-myc in the CMZ depends on growth factor signaling. Our results suggest that c-myc+/n-myc- cells in the far peripheral CMZ are candidates for a niche-dependent population of retinal stem cells that give rise to more centrally located and rapidly dividing n-myc+ progenitors of more limited proliferative potential. Analysis of homologues of these genes in the zebrafish CMZ suggests that the transition from c-myc to n-myc expression might be conserved in other lower vertebrates whose retinas growth throughout life.
Collapse
Affiliation(s)
- Xiao Yan Xue
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | | |
Collapse
|
16
|
Luo J, Uribe RA, Hayton S, Calinescu AA, Gross JM, Hitchcock PF. Midkine-A functions upstream of Id2a to regulate cell cycle kinetics in the developing vertebrate retina. Neural Dev 2012; 7:33. [PMID: 23111152 PMCID: PMC3531272 DOI: 10.1186/1749-8104-7-33] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 08/31/2012] [Indexed: 02/04/2023] Open
Abstract
Background Midkine is a small heparin binding growth factor expressed in numerous tissues during development. The unique midkine gene in mammals has two paralogs in zebrafish: midkine-a (mdka) and midkine-b (mdkb). In the zebrafish retina, during both larval development and adult photoreceptor regeneration, mdka is expressed in retinal stem and progenitor cells and functions as a molecular component of the retina’s stem cell niche. In this study, loss-of-function and conditional overexpression were used to investigate the function of Mdka in the retina of the embryonic zebrafish. Results The results show that during early retinal development Mdka functions to regulate cell cycle kinetics. Following targeted knockdown of Mdka synthesis, retinal progenitors cycle more slowly, and this results in microphthalmia, a diminished rate of cell cycle exit and a temporal delay of cell cycle exit and neuronal differentiation. In contrast, Mdka overexpression results in acceleration of the cell cycle and retinal overgrowth. Mdka gain-of-function, however, does not temporally advance cell cycle exit. Experiments to identify a potential Mdka signaling pathway show that Mdka functions upstream of the HLH regulatory protein, Id2a. Gene expression analysis shows Mdka regulates id2a expression, and co-injection of Mdka morpholinos and id2a mRNA rescues the Mdka loss-of-function phenotype. Conclusions These data show that in zebrafish, Mdka resides in a shared Id2a pathway to regulate cell cycle kinetics in retinal progenitors. This is the first study to demonstrate the function of Midkine during retinal development and adds Midkine to the list of growth factors that transcriptionally regulate Id proteins.
Collapse
Affiliation(s)
- Jing Luo
- Department of Ophthalmology and Visual Sciences, University of Michigan, W, K, Kellogg Eye Center, 1000 Wall Street, Ann Arbor, MI 48105-0714, USA
| | | | | | | | | | | |
Collapse
|
17
|
Abstract
During embryonic development, cells must divide to produce appropriate numbers, but later must exit the cell cycle to allow differentiation. How these processes of proliferation and differentiation are co-ordinated during embryonic development has been poorly understood until recently. However, a number of studies have now given an insight into how the cell cycle machinery, including cyclins, CDKs (cyclin-dependent kinases), CDK inhibitors and other cell cycle regulators directly influence mechanisms that control cell fate and differentiation. Conversely, examples are emerging of transcriptional regulators that are better known for their role in driving the differentiated phenotype, which also play complementary roles in controlling cell cycle progression. The present review will summarise our current understanding of the mechanisms co-ordinating the cell cycle and differentiation in the developing nervous system, where these links have been, perhaps, most extensively studied.
Collapse
|
18
|
Elkouby YM, Polevoy H, Gutkovich YE, Michaelov A, Frank D. A hindbrain-repressive Wnt3a/Meis3/Tsh1 circuit promotes neuronal differentiation and coordinates tissue maturation. Development 2012; 139:1487-97. [PMID: 22399680 DOI: 10.1242/dev.072934] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
During development, early inducing programs must later be counterbalanced for coordinated tissue maturation. In Xenopus laevis embryos, activation of the Meis3 transcription factor by a mesodermal Wnt3a signal lies at the core of the hindbrain developmental program. We now identify a hindbrain restricting circuit, surprisingly comprising the hindbrain inducers Wnt3a and Meis3, and Tsh1 protein. Functional and biochemical analyses show that upon Tsh1 induction by strong Wnt3a/Meis3 feedback loop activity, the Meis3-Tsh1 transcription complex represses the Meis3 promoter, allowing cell cycle exit and neuron differentiation. Meis3 protein exhibits a conserved dual-role in hindbrain development, both inducing neural progenitors and maintaining their proliferative state. In this regulatory circuit, the Tsh1 co-repressor controls transcription factor gene expression that modulates cell cycle exit, morphogenesis and differentiation, thus coordinating neural tissue maturation. This newly identified Wnt/Meis/Tsh circuit could play an important role in diverse developmental and disease processes.
Collapse
Affiliation(s)
- Yaniv M Elkouby
- Department of Biochemistry, The Rappaport Family Institute for Research in the Medical Sciences, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | | | | | | | | |
Collapse
|
19
|
Brandt Y, Mitchell T, Wu Y, Hartley RS. Developmental downregulation of Xenopus cyclin E is phosphorylation and nuclear import dependent and is mediated by ubiquitination. Dev Biol 2011; 355:65-76. [PMID: 21539834 DOI: 10.1016/j.ydbio.2011.04.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2010] [Revised: 03/16/2011] [Accepted: 04/14/2011] [Indexed: 11/29/2022]
Abstract
Cyclins are regulatory subunits that bind to and activate catalytic Cdks. Cyclin E associates with Cdk2 to mediate the G1/S transition of the cell cycle. Cyclin E is overexpressed in breast, lung, skin, gastrointestinal, cervical, and ovarian cancers. Its overexpression correlates with poor patient prognosis and is involved in the etiology of breast cancer. We have been studying how cyclin E is normally downregulated during development in order to determine if disruption of similar mechanisms could either contribute to its overexpression in cancer, or be exploited to decrease its expression. In Xenopus laevis embryos, cyclin E protein level is high and constant until its abrupt destabilization by an undefined mechanism after the 12th cell cycle, which corresponds to the midblastula transition (MBT) and remodeling of the embryonic to the adult cell cycle. Since degradation of mammalian cyclin E is regulated by the ubiquitin proteasome system and is phosphorylation dependent, we examined the role of phosphorylation in Xenopus cyclin E turnover. We show that similarly to human cyclin E, phosphorylation of serine 398 and threonine 394 plays a role in cyclin E turnover at the MBT. Immunofluorescence analysis shows that cyclin E relocalizes from the cytoplasm to the nucleus preceding its degradation. When nuclear import is inhibited, cyclin E stability is markedly increased after the MBT. To investigate whether degradation of Xenopus cyclin E is mediated by the proteasomal pathway, we used proteasome inhibitors and observed a progressive accumulation of cyclin E in the cytoplasm after the MBT. Ubiquitination of cyclin E precedes its proteasomal degradation at the MBT. These results show that cyclin E destruction at the MBT requires both phosphorylation and nuclear import, as well as proteasomal activity.
Collapse
|
20
|
Horb LD, Horb ME. BrunoL1 regulates endoderm proliferation through translational enhancement of cyclin A2 mRNA. Dev Biol 2010; 345:156-69. [PMID: 20633547 DOI: 10.1016/j.ydbio.2010.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Revised: 06/12/2010] [Accepted: 07/02/2010] [Indexed: 10/19/2022]
Abstract
Developmental control of proliferation relies on tight regulation of protein expression. Although this has been well studied in early embryogenesis, how the cell cycle is regulated during organogenesis is not well understood. Bruno-Like RNA binding proteins bind to consensus sequences in the 3'UTR of specific mRNAs and repress protein translation, but much of this functional information is derived from studies on mainly two members, Drosophila Bruno and vertebrate BrunoL2 (CUGBP1). There are however, six vertebrate and three Drosophila Bruno family members, but less is known about these other family members, and none have been shown to function in the endoderm. We recently identified BrunoL1 as a dorsal pancreas enriched gene, and in this paper we define BrunoL1 function in Xenopus endoderm development. We find that, in contrast to other Bruno-Like proteins, BrunoL1 acts to enhance rather than repress translation. We demonstrate that BrunoL1 regulates proliferation of endoderm cells through translational control of cyclin A2 mRNA. Specifically BrunoL1 enhanced translation of cyclin A2 through binding consensus Bruno Response Elements (BREs) in its 3'UTR. We compared the ability of other Bruno-Like proteins, both vertebrate and invertebrate, to stimulate translation via the cyclin A2 3'UTR and found that only Drosophila Bru-3 had similar activity. In addition, we also found that both BrunoL1 and Bru-3 enhanced translation of mRNAs containing the 3'UTRs of Drosophila oskar or cyclin A, which have been well characterized to mediate repression. Lastly, we show that it is the Linker region of BrunoL1 that is both necessary and sufficient for this activity. These results are the first example of BRE-dependent translational enhancement and are the first demonstration in vertebrates of Bruno-Like proteins regulating translation through BREs.
Collapse
Affiliation(s)
- Lori Dawn Horb
- Laboratory of Molecular Organogenesis, Institut de Recherches Cliniques de Montréal, Montreal, QC, Canada
| | | |
Collapse
|
21
|
Thomson AM, Gillespie PJ, Blow JJ. Replication factory activation can be decoupled from the replication timing program by modulating Cdk levels. ACTA ACUST UNITED AC 2010; 188:209-21. [PMID: 20083602 PMCID: PMC2812520 DOI: 10.1083/jcb.200911037] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Cdk activity can differentially regulate the number of active replication factories, replication rates, and the rate of progression through the timing program during S phase. In the metazoan replication timing program, clusters of replication origins located in different subchromosomal domains fire at different times during S phase. We have used Xenopus laevis egg extracts to drive an accelerated replication timing program in mammalian nuclei. Although replicative stress caused checkpoint-induced slowing of the timing program, inhibition of checkpoint kinases in an unperturbed S phase did not accelerate it. Lowering cyclin-dependent kinase (Cdk) activity slowed both replication rate and progression through the timing program, whereas raising Cdk activity increased them. Surprisingly, modest alteration of Cdk activity changed the amount of DNA synthesized during different stages of the timing program. This was associated with a change in the number of active replication factories, whereas the distribution of origins within active factories remained relatively normal. The ability of Cdks to differentially effect replication initiation, factory activation, and progression through the timing program provides new insights into the way that chromosomal DNA replication is organized during S phase.
Collapse
Affiliation(s)
- Alexander M Thomson
- Wellcome Trust Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, UK
| | | | | |
Collapse
|
22
|
Almeida AD, Wise HM, Hindley CJ, Slevin MK, Hartley RS, Philpott A. The F-box protein Cdc4/Fbxw7 is a novel regulator of neural crest development in Xenopus laevis. Neural Dev 2010; 5:1. [PMID: 20047651 PMCID: PMC2819241 DOI: 10.1186/1749-8104-5-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2009] [Accepted: 01/04/2010] [Indexed: 11/13/2022] Open
Abstract
Background The neural crest is a unique population of cells that arise in the vertebrate ectoderm at the neural plate border after which they migrate extensively throughout the embryo, giving rise to a wide range of derivatives. A number of proteins involved in neural crest development have dynamic expression patterns, and it is becoming clear that ubiquitin-mediated protein degradation is partly responsible for this. Results Here we demonstrate a novel role for the F-box protein Cdc4/Fbxw7 in neural crest development. Two isoforms of Xenopus laevis Cdc4 were identified, and designated xCdc4α and xCdc4β. These are highly conserved with vertebrate Cdc4 orthologs, and the Xenopus proteins are functionally equivalent in terms of their ability to degrade Cyclin E, an established vertebrate Cdc4 target. Blocking xCdc4 function specifically inhibited neural crest development at an early stage, prior to expression of c-Myc, Snail2 and Snail. Conclusions We demonstrate that Cdc4, an ubiquitin E3 ligase subunit previously identified as targeting primarily cell cycle regulators for proteolysis, has additional roles in control of formation of the neural crest. Hence, we identify Cdc4 as a protein with separable but complementary functions in control of cell proliferation and differentiation.
Collapse
Affiliation(s)
- Alexandra D Almeida
- Department of Oncology, University of Cambridge, Hutchison-MRC Research Centre, Addenbrookes Hospital, Hills Road, Cambridge, CB2 0XZ, UK
| | | | | | | | | | | |
Collapse
|
23
|
Self-regulation of Stat3 activity coordinates cell-cycle progression and neural crest specification. EMBO J 2009; 29:55-67. [PMID: 19851287 DOI: 10.1038/emboj.2009.313] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Accepted: 09/25/2009] [Indexed: 01/13/2023] Open
Abstract
A complex set of extracellular signals is required for neural crest (NC) specification. However, how these signals function to coordinate cell-cycle progression and differentiation remains poorly understood. Here, we report in Xenopus a role for the transcription factor signal transducers and activators of transcription-3 (Stat3) in this process downstream of FGF signalling. Depletion of Stat3 inhibits NC gene expression and cell proliferation, whereas overexpression expands the NC domain and promotes cell proliferation. Stat3 is phosphorylated and activated in ectodermal cells by FGFs through binding with FGFR4. Stat3 activation is also modulated by Hairy2 and Id3 proteins that, respectively, facilitate and disrupt Stat3-FGFR4 complex formation. Furthermore, distinct levels of Stat3 activity control Hairy2 and Id3 transcription, leading to Stat3 self-regulation. Finally, high Stat3 activity maintains cells in an undifferentiated state, whereas low activity promotes cell proliferation and NC differentiation. Together, our data suggest that Stat3, downstream of FGFs and under the positive and negative feedback regulation of Hairy2 and Id3, plays an essential role in the coordination of cell-cycle progression and differentiation during NC specification.
Collapse
|
24
|
Agathocleous M, Iordanova I, Willardsen MI, Xue XY, Vetter ML, Harris WA, Moore KB. A directional Wnt/beta-catenin-Sox2-proneural pathway regulates the transition from proliferation to differentiation in the Xenopus retina. Development 2009; 136:3289-99. [PMID: 19736324 DOI: 10.1242/dev.040451] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Progenitor cells in the central nervous system must leave the cell cycle to become neurons and glia, but the signals that coordinate this transition remain largely unknown. We previously found that Wnt signaling, acting through Sox2, promotes neural competence in the Xenopus retina by activating proneural gene expression. We now report that Wnt and Sox2 inhibit neural differentiation through Notch activation. Independently of Sox2, Wnt stimulates retinal progenitor proliferation and this, when combined with the block on differentiation, maintains retinal progenitor fates. Feedback inhibition by Sox2 on Wnt signaling and by the proneural transcription factors on Sox2 mean that each element of the core pathway activates the next element and inhibits the previous one, providing a directional network that ensures retinal cells make the transition from progenitors to neurons and glia.
Collapse
Affiliation(s)
- Michalis Agathocleous
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | | | | | | | | | | | | |
Collapse
|
25
|
Movassagh M, Philpott A. Cardiac differentiation in Xenopus requires the cyclin-dependent kinase inhibitor, p27Xic1. Cardiovasc Res 2008; 79:436-47. [PMID: 18442987 PMCID: PMC2492727 DOI: 10.1093/cvr/cvn105] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Aims Cyclin-dependent kinase inhibitors (CDKIs) play a critical role in negatively regulating the proliferation of cardiomyocytes, although their role in cardiac differentiation remains largely undetermined. We have shown that the most prominent CDKI in Xenopus, p27Xic1(Xic1), plays a role in neuronal and myotome differentiation beyond its ability to arrest the cell cycle. Thus, we investigated whether it plays a similar role in cardiomyocyte differentiation. Methods and results Xenopus laevis embryos were sectioned, and whole-mount antibody staining and immunofluorescence studies were carried out to determine the total number and percentage of differentiated cardiomyocytes in mitosis. Capped RNA and/or translation-blocking Xic1 morpholino antisense oligonucleotides (Xic1Mo) were microinjected into embryos, and their role on cardiac differentiation was assessed by in situ hybridization and/or PCR. We show that cell-cycling post-gastrulation is not essential for cardiac differentiation in Xenopus embryos, and conversely that some cells can express markers of cardiac differentiation even when still in cycle. A targeted knock-down of Xic1 protein by Xic1Mo microinjection decreases the expression of markers of cardiac differentiation, which can be partially rescued by co-injection of full-length Xic1 RNA, demonstrating that Xic1 is essential for heart formation. Furthermore, using deleted and mutant forms of Xic1, we show that neither its abilities to inhibit the cell cycle nor the great majority of CDK kinase activity are essential for Xic1’s function in cardiomyocyte differentiation, an activity that resides in the N-terminus of the molecule. Conclusion Altogether, our results demonstrate that the CDKI Xic1 is required in Xenopus cardiac differentiation, and that this function is localized at its N-terminus, but it is distinct from its ability to arrest the cell cycle and inhibit overall CDK kinase activity. Hence, these results suggest that CDKIs play an important direct role in driving cardiomyocyte differentiation in addition to cell-cycle regulation.
Collapse
Affiliation(s)
- Mehregan Movassagh
- Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Addenbrooke's Hospital, Hills Road, Cambridge CB2 2XZ, UK
| | | |
Collapse
|
26
|
Philpott A, Yew PR. The Xenopus cell cycle: an overview. Mol Biotechnol 2008; 39:9-19. [PMID: 18266114 DOI: 10.1007/s12033-008-9033-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Accepted: 12/28/2007] [Indexed: 01/03/2023]
Abstract
Oocytes, eggs and embryos from the frog Xenopus laevis have been an important model system for studying cell-cycle regulation for several decades. First, progression through meiosis in the oocyte has been extensively investigated. Oocyte maturation has been shown to involve complex networks of signal transduction pathways, culminating in the cyclic activation and inactivation of Maturation Promoting Factor (MPF), composed of cyclin B and cdc2. After fertilisation, the early embryo undergoes rapid simplified cell cycles which have been recapitulated in cell-free extracts of Xenopus eggs. Experimental manipulation of these extracts has given a wealth of biochemical information about the cell cycle, particularly concerning DNA replication and mitosis. Finally, cells of older embryos adopt a more somatic-type cell cycle and have been used to study the balance between cell cycle and differentiation during development.
Collapse
Affiliation(s)
- Anna Philpott
- Department of Oncology, Hutchison/MRC Research Centre, Addenbrooke's Hospital, University of Cambridge, Hills Road, Cambridge, CB2 0XZ, England.
| | | |
Collapse
|
27
|
Hashiguchi A, Hitachi K, Inui M, Okabayashi K, Asashima M. TSC-box is essential for the nuclear localization and antiproliferative effect of XTSC-22. Dev Growth Differ 2007; 49:197-204. [PMID: 17394598 DOI: 10.1111/j.1440-169x.2007.00908.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Transforming growth factor-beta1-stimulated clone 22 (TSC-22) encodes a leucine zipper-containing protein that is highly conserved among various species. Mammalian TSC-22 is a potential tumor suppressor gene. It translocates into nuclei and suppresses cell division upon antiproliferative stimuli. In human colon carcinoma cells, TSC-22 inhibits cell growth by upregulating expression of the p21 gene, a cyclin-dependent kinase (Cdk) inhibitor. We previously showed that the Xenopus laevis homologue of the TSC-22 gene (XTSC-22) is required for cell movement during gastrulation through cell cycle regulation. In this report, we investigated the molecular mechanism of the antiproliferative effect of XTSC-22. Reverse transcriptase-polymerase chain reaction (RT-PCR) analysis suggested that XTSC-22 did not affect the expression levels of the p21 family of Cdk inhibitors or other cell cycle regulators. Analysis of deletion mutants of XTSC-22 revealed that nuclear localization of the N-terminal TSC-box is necessary for cell cycle inhibition by XTSC-22. Further experiments suggested that p27Xic1, a key Cdk inhibitor in Xenopus, interacts with XTSC-22. Because p27Xic1 is a cell cycle inhibitor with a nuclear localization signal, it is possible that XTSC-22 suppresses cell division by translocating into the nucleus with p27Xic1, where it may potentiate the intranuclear action of p27Xic1.
Collapse
Affiliation(s)
- Akiko Hashiguchi
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | | | | | | | | |
Collapse
|
28
|
Cosgrove RA, Philpott A. Cell cycling and differentiation do not require the retinoblastoma protein during early Xenopus development. Dev Biol 2007; 303:311-24. [PMID: 17188261 DOI: 10.1016/j.ydbio.2006.11.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2006] [Revised: 11/03/2006] [Accepted: 11/08/2006] [Indexed: 11/23/2022]
Abstract
The retinoblastoma protein (pRb) is a central regulator of the cell cycle, controlling passage through G1 phase. Moreover, pRb has also been shown to play a direct role in the differentiation of multiple tissues, including nerve and muscle. Rb null mice display embryonic lethality, although recent data have indicated that at least some of these defects are due to placental insufficiency. To investigate this further, we have examined the role of pRb in early development of the frog Xenopus laevis, which develops without the need for a placenta. Surprisingly, we see that loss of pXRb has no effect on either cell cycling or differentiation of neural or muscle tissue, while overexpression of pXRb similarly has no effects. We demonstrate that, in fact, pXRb is maintained in a hyperphosphorylated and therefore inactive state early in development. Therefore, Rb protein is not required for cell cycle control or differentiation in early embryos, indicating unusual control of these G1/G0 events at this developmental stage.
Collapse
Affiliation(s)
- Ruth A Cosgrove
- Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Addenbrookes Hospital, Hills Road, Cambridge CB2 0XZ, UK
| | | |
Collapse
|
29
|
Chen JA, Chu ST, Amaya E. Maintenance of motor neuron progenitors in Xenopus requires a novel localized cyclin. EMBO Rep 2007; 8:287-92. [PMID: 17304238 PMCID: PMC1808035 DOI: 10.1038/sj.embor.7400903] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2006] [Revised: 11/27/2006] [Accepted: 12/19/2006] [Indexed: 12/22/2022] Open
Abstract
The ventral spinal cord contains a pool of motor neuron progenitors (pMNs), which sequentially generate motor neurons and oligodendrocytes in the embryo. The mechanisms responsible for the maintenance of pMNs are not clearly understood. We have identified a novel cyclin, cyclin Dx (ccndx), which is specifically expressed in pMNs in Xenopus. Here, we show that inhibition of ccndx causes paralysis in embryos. Furthermore, we show that maintenance of pMNs requires ccndx function. In addition, inhibition of ccndx results in the specific loss of differentiated motor neurons. However, the expression of interneuron or sensory neuron markers is unaffected in these embryos, suggesting that the role of ccndx is specifically to maintain pMNs. Thus, we have identified, for the first time, a tissue-specific cell-cycle regulator that is essential for the maintenance of a pool of neural progenitors in the vertebrate spinal cord.
Collapse
Affiliation(s)
- Jun-An Chen
- The Wellcome Trust/Cancer Research UK Gurdon Institute and Department of Zoology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Sin-Tak Chu
- Institute of Biological Chemistry, Academia Sinica, Post Box 23-106, Taipei, Taiwan
| | - Enrique Amaya
- The Wellcome Trust/Cancer Research UK Gurdon Institute and Department of Zoology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
- The Healing Foundation Centre, Michael Smith Building, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
- Tel: 44 161 275 1716; Fax: 44 161 275 1505; E-mail:
| |
Collapse
|
30
|
Liu QY, Wu ZL, Lv WJ, Yan YC, Li YP. Developmental expression of Cyclin H and Cdk7 in zebrafish: the essential role of Cyclin H during early embryo development. Cell Res 2007; 17:163-73. [PMID: 17287831 DOI: 10.1038/sj.cr.7310144] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Cyclin-dependent kinase 7 (Cdk7) is the catalytic subunit of the metazoan Cdk-activating kinase (CAK). Activation of Cdk7 requires its association with a regulatory subunit, Cyclin H. Although the Cdk7/Cyclin H complex has been implicated in the regulation of RNA polymerase in several species, the precise function of their orthologs in zebrafish has not been fully elucidated. In this study, we isolated from zebrafish blastula embryos two cDNAs encoding the orthologs of human Cyclin H and Cdk7, and examined the role of Cdk7/Cyclin H in zebrafish embryogenesis. Sequence analysis showed that the zebrafish Cyclin H and Cdk7 cDNAs encode proteins with 65% and 86% identity to the respective human orthologs. RT-PCR and whole-mount in situ hybridization analyses of their expression in unfertilized eggs, embryos and organs of adult fish suggested that Cyclin H and Cdk7 messages are maternally loaded. Our data also showed that their transcripts were detected throughout development. Distribution of Cyclin H transcripts was found to be ubiquitous during early stages of development and become restricted to the anterior neural tube, brain, eyes, procreate tissues, liver and heart by 5 days post-fertilization. Expression of a dominant-negative form of Cyclin H delayed the onset of zygotic transcription in the early embryo, resulting in apoptosis at 5 hours post-fertilization and leading to sever defects in tissues normally exhibiting high levels of Cyclin H expression. These results implicate Cyclin H in the regulation of the transcriptional machinery during midblastula transition and suggest that it is an essential gene in early zebrafish larval development.
Collapse
Affiliation(s)
- Qing Yun Liu
- Lab of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Science, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | | | | | | | | |
Collapse
|
31
|
Locker M, Agathocleous M, Amato MA, Parain K, Harris WA, Perron M. Hedgehog signaling and the retina: insights into the mechanisms controlling the proliferative properties of neural precursors. Genes Dev 2006; 20:3036-48. [PMID: 17079690 PMCID: PMC1620016 DOI: 10.1101/gad.391106] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Hedgehog signaling has been linked to cell proliferation in a variety of systems; however, its effects on the cell cycle have not been closely studied. In the vertebrate retina, Hedgehog's effects are controversial, with some reports emphasizing increased proliferation and others pointing to a role in cell cycle exit. Here we demonstrate a novel role for Hedgehog signaling in speeding up the cell cycle in the developing retina by reducing the length of G1 and G2 phases. These fast cycling cells tend to exit the cell cycle early. Conversely, retinal progenitors with blocked Hedgehog signaling cycle more slowly, with longer G1 and G2 phases, and remain in the cell cycle longer. Hedgehog may modulate cell cycle kinetics through activation of the key cell cycle activators cyclin D1, cyclin A2, cyclin B1, and cdc25C. These findings support a role for Hedgehog in regulating the conversion from slow cycling stem cells to fast cycling transient amplifying progenitors that are closer to cell cycle exit.
Collapse
Affiliation(s)
- Morgane Locker
- Laboratoire Gènes, Développement et Neurogenèse, Université Paris XI, UMR Centre national de la Recherche Scientifique (CNRS) 8080, Orsay, France
| | | | | | | | | | | |
Collapse
|
32
|
Ueno S, Kono R, Iwao Y. PTEN is required for the normal progression of gastrulation by repressing cell proliferation after MBT in Xenopus embryos. Dev Biol 2006; 297:274-83. [PMID: 16919259 DOI: 10.1016/j.ydbio.2006.06.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2006] [Revised: 05/30/2006] [Accepted: 06/01/2006] [Indexed: 11/16/2022]
Abstract
PTEN phosphatase mediates several developmental cues involving cell proliferation, growth, death, and migration. We investigated the function of the PTEN gene at the transition from the cell proliferation state to morphogenesis around the midblastula transition (MBT) and gastrulation in Xenopus embryos. An immunoblotting analysis indicated that PTEN expresses constantly through embryogenesis. By up- or down-regulating PTEN activity using overexpression of the active form or C terminus of PTEN before MBT, we induced elongation of the cell cycle time just before MBT or maintained its speed even after MBT, respectively. The disruption of the cell cycle time by changing the activity of PTEN delayed gastrulation after MBT. In addition, PTEN began to localize to the plasma membranes and nuclei at MBT. Overexpression of a membrane-localizing mutant of PTEN caused dephosphorylation of Akt, whereas overexpression of the C terminus of PTEN caused phosphorylation of Akt and inhibited the localization of EGFP-PTEN to the plasma membranes and nuclei. These results indicate that an appropriate PTEN activity, probably regulated by its differential localization, is necessary for coordinating cell proliferation and early morphogenesis.
Collapse
Affiliation(s)
- Shuichi Ueno
- Department of Biological Science, Faculty of Science, Yamaguchi University, 753-8512 Yamaguchi, Japan.
| | | | | |
Collapse
|
33
|
Abstract
Despite the critical importance of TBX5 in normal development and disease, relatively little is known about the mechanisms by which TBX5 functions in the embryonic heart. Our present studies demonstrate that TBX5 is necessary to control the length of the embryonic cardiac cell cycle, with depletion of TBX5 leading to cardiac cell cycle arrest in late G(1)- or early S-phase. Blocking cell cycle progression by TBX5 depletion leads to a decrease in cardiac cell number, an alteration in the timing of the cardiac differentiation program, defects in cardiac sarcomere formation, and ultimately, to cardiac programmed cell death. In these studies we have also established that terminally differentiated cardiomyocytes retain the capacity to undergo cell division. We further show that TBX5 is sufficient to determine the length of the embryonic cardiac cell cycle and the timing of the cardiac differentiation program. Thus, these studies establish a role for TBX5 in regulating the progression of the cardiac cell cycle.
Collapse
Affiliation(s)
- Sarah C. Goetz
- Carolina Cardiovascular Biology Center, 5109 Neuroscience Research Building, Chapel Hill, NC 27599-7126, USA
- Department of Biology, Fordham Hall, UNC-Chapel Hill, Chapel Hill, NC 27599-3280, USA
| | - Daniel D. Brown
- Carolina Cardiovascular Biology Center, 5109 Neuroscience Research Building, Chapel Hill, NC 27599-7126, USA
- Department of Biology, Fordham Hall, UNC-Chapel Hill, Chapel Hill, NC 27599-3280, USA
| | - Frank L. Conlon
- Carolina Cardiovascular Biology Center, 5109 Neuroscience Research Building, Chapel Hill, NC 27599-7126, USA
- Department of Biology, Fordham Hall, UNC-Chapel Hill, Chapel Hill, NC 27599-3280, USA
- Department of Genetics, Fordham Hall, UNC-Chapel Hill, Chapel Hill, NC 27599-3280, USA
- *Author for correspondence (e-mail: )
| |
Collapse
|
34
|
Hashiguchi A, Okabayashi K, Asashima M. Role of TSC-22 during early embryogenesis in Xenopus laevis. Dev Growth Differ 2005; 46:535-44. [PMID: 15610143 DOI: 10.1111/j.1440-169x.2004.00770.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Transforming growth factor-beta1-stimulated clone 22 (TSC-22) encodes a leucine zipper-containing protein that is highly conserved. During mouse embryogenesis, TSC-22 is expressed at the site of epithelial-mesenchymal interaction. Here, we isolated Xenopus laevis TSC-22 (XTSC-22) and analyzed its function in early development. XTSC-22 mRNA was first detected in the ectoderm of late blastulae. Translational knockdown using XTSC-22 antisense morpholino oligonucleotides (XTSC-22-MO) caused a severe delay in blastopore closure in gastrulating embryos. This was not due to mesoderm induction or convergent-extension, as confirmed by whole-mount in situ hybridization and animal cap assay. Cell lineage tracing revealed that migration of ectoderm cells toward blastopore was disrupted in XTSC-22-depleted embryos, and these embryos had a marked increase in the number of dividing cells. In contrast, cell division was suppressed in XTSC-22 mRNA-injected embryos. Co-injection of XTSC-22-MO and mRNA encoding p27Xic1, which inhibits cell cycle promotion by binding cyclin/Cdk complexes, reversed aberrant cell division. This was accompanied by rescue of the delay in blastopore closure and cell migration. These results indicate that XTSC-22 is required for cell movement during gastrulation though cell cycle regulation.
Collapse
Affiliation(s)
- Akiko Hashiguchi
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
| | | | | |
Collapse
|
35
|
Walter BE, Henry JJ. Embryonic expression of pre-initiation DNA replication factors in Xenopus laevis. Gene Expr Patterns 2005; 5:81-9. [PMID: 15533822 DOI: 10.1016/j.modgep.2004.06.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2004] [Revised: 06/07/2004] [Accepted: 06/11/2004] [Indexed: 12/01/2022]
Abstract
We examined the expression of various DNA replication factors, including: cdc45, the factors of the GINS heterotetramer (Sld5, Psf1, Psf2, Psf3), and PCNA, in Xenopus laevis during embryonic development via whole mount in situ hybridization. For the most part, these factors were expressed in similar patterns, with some subtle variations, throughout development within the anterior CNS, pharyngeal arches, and various placodes. More significant variations were also observed, including expression of only Psf1 and Psf2 in the pronephros and unique Psf2 expression in the somitic mesoderm. Overall, these results suggest that common regulatory mechanisms are involved in the transcriptional deployment of these factors. Significantly, expression of these factors does not directly coincide with elevated patterns of DNA replication, suggesting that different replication factors are utilized in different developmental regions.
Collapse
Affiliation(s)
- Brian E Walter
- Department of Cell and Structural Biology, University of Illinois, 601 South Goodwin Avenue, Urbana, IL 61801, USA
| | | |
Collapse
|
36
|
Gestri G, Carl M, Appolloni I, Wilson SW, Barsacchi G, Andreazzoli M. Six3 functions in anterior neural plate specification by promoting cell proliferation and inhibiting Bmp4 expression. Development 2005; 132:2401-13. [PMID: 15843413 PMCID: PMC2789257 DOI: 10.1242/dev.01814] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Although it is well established that Six3 is a crucial regulator of vertebrate eye and forebrain development, it is unknown whether this homeodomain protein has a role in the initial specification of the anterior neural plate. In this study, we show that exogenous Six3 can expand the anterior neural plate in both Xenopus and zebrafish, and that this occurs in part through Six3-dependent transcriptional regulation of the cell cycle regulators cyclinD1 and p27Xic1, as well as the anti-neurogenic genes Zic2 and Xhairy2. However, Six3 can still expand the neural plate in the presence of cell cycle inhibitors and we show that this is likely to be due to its ability to repress the expression of Bmp4 in ectoderm adjacent to the anterior neural plate. Furthermore, exogenous Six3 is able to restore the size of the anterior neural plate in chordino mutant zebrafish, indicating that it has the ability to promote anterior neural development by antagonising the activity of the BMP pathway. On its own, Six3 is unable to induce neural tissue in animal caps, but it can do so in combination with Otx2. These results suggest a very early role for Six3 in specification of the anterior neural plate, through the regulation of cell proliferation and the inhibition of BMP signalling.
Collapse
Affiliation(s)
- Gaia Gestri
- Sezione di Biologia Cellulare e dello Sviluppo, Dipartimento di Fisiologia, e Biochimica, Universita' degli Studi di Pisa, Via Carducci 13, 56010 Ghezzano (Pisa), Italy
- AMBISEN Center, High Technology Center for the Study of the Environmental Damage of the Endocrine and Nervous Systems, Universita' degli Studi di Pisa, Italy
- Department of Anatomy and Developmental Biology, University College of London, Gower Street, London WC1E 6BT, UK
| | - Matthias Carl
- Department of Anatomy and Developmental Biology, University College of London, Gower Street, London WC1E 6BT, UK
| | - Irene Appolloni
- Sezione di Biologia Cellulare e dello Sviluppo, Dipartimento di Fisiologia, e Biochimica, Universita' degli Studi di Pisa, Via Carducci 13, 56010 Ghezzano (Pisa), Italy
- AMBISEN Center, High Technology Center for the Study of the Environmental Damage of the Endocrine and Nervous Systems, Universita' degli Studi di Pisa, Italy
| | - Stephen W. Wilson
- Department of Anatomy and Developmental Biology, University College of London, Gower Street, London WC1E 6BT, UK
| | - Giuseppina Barsacchi
- Sezione di Biologia Cellulare e dello Sviluppo, Dipartimento di Fisiologia, e Biochimica, Universita' degli Studi di Pisa, Via Carducci 13, 56010 Ghezzano (Pisa), Italy
- AMBISEN Center, High Technology Center for the Study of the Environmental Damage of the Endocrine and Nervous Systems, Universita' degli Studi di Pisa, Italy
| | - Massimiliano Andreazzoli
- Sezione di Biologia Cellulare e dello Sviluppo, Dipartimento di Fisiologia, e Biochimica, Universita' degli Studi di Pisa, Via Carducci 13, 56010 Ghezzano (Pisa), Italy
- AMBISEN Center, High Technology Center for the Study of the Environmental Damage of the Endocrine and Nervous Systems, Universita' degli Studi di Pisa, Italy
| |
Collapse
|
37
|
Shin Y, Kitayama A, Koide T, Peiffer DA, Mochii M, Liao A, Ueno N, Cho KWY. Identification of neural genes usingXenopus DNA microarrays. Dev Dyn 2005; 232:432-44. [PMID: 15614765 DOI: 10.1002/dvdy.20229] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To isolate novel genes regulating neural induction, we used a DNA microarray approach. As neural induction is thought to occur by means of the inhibition of bone morphogenetic protein (BMP) signaling, BMP signaling was inhibited in ectodermal cells by overexpression of a dominant-negative receptor. RNAs were isolated from control animal cap explants and from dominant-negative BMP receptor expressing animal caps and subjected to a microarray experiment using newly generated high-density Xenopus DNA microarray chips representing over 17,000 unigenes. We have identified 77 genes that are induced in animal caps after inhibition of BMP signaling, and all of these genes were subjected to whole-mount in situ hybridization analysis. Thirty-two genes showed specific expression in neural tissues. Of the 32, 14 genes have never been linked to neural induction. Two genes that are highly induced by BMP inhibition are inhibitors of Wnt signaling, suggesting that a key step in neural induction is to produce Wnt antagonists to promote anterior neural plate development. Our current analysis also proves that a microarray approach is useful in identifying novel candidate factors involved in neural induction and patterning.
Collapse
Affiliation(s)
- Yongchol Shin
- Department of Developmental and Cell Biology, University of California, Irvine, California, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Richard-Parpaillon L, Cosgrove RA, Devine C, Vernon AE, Philpott A. G1/S phase cyclin-dependent kinase overexpression perturbs early development and delays tissue-specific differentiation in Xenopus. Development 2004; 131:2577-86. [PMID: 15115752 DOI: 10.1242/dev.01121] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Cell division and differentiation are largely incompatible but the molecular links between the two processes are poorly understood. Here, we overexpress G1/S phase cyclins and cyclin-dependent kinases in Xenopus embryos to determine their effect on early development and differentiation. Overexpression of cyclin E prior to the midblastula transition (MBT), with or without cdk2, results in a loss of nuclear DNA and subsequent apoptosis at early gastrula stages. By contrast, overexpressed cyclin A2 protein does not affect early development and, when stabilised by binding to cdk2, persists to tailbud stages. Overexpression of cyclin A2/cdk2 in post-MBT embryos results in increased proliferation specifically in the epidermis with concomitant disruption of skin architecture and delay in differentiation. Moreover, ectopic cyclin A2/cdk2 also inhibits differentiation of primary neurons but does not affect muscle. Thus, overexpression of a single G1/S phase cyclin/cdk pair disrupts the balance between division and differentiation in the early vertebrate embryo in a tissue-specific manner.
Collapse
Affiliation(s)
- Laurent Richard-Parpaillon
- Department of Oncology, Cambridge University, Hutchison/MRC Research Centre, Addenbrookes Hospital, Hills Road, Cambridge CB2 2XZ, UK.
| | | | | | | | | |
Collapse
|
39
|
Andreazzoli M, Gestri G, Cremisi F, Casarosa S, Dawid IB, Barsacchi G. Xrx1 controls proliferation and neurogenesis in Xenopus anterior neural plate. Development 2003; 130:5143-54. [PMID: 12975341 DOI: 10.1242/dev.00665] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In Xenopus neuroectoderm, posterior cells start differentiating at the end of gastrulation, while anterior cells display an extended proliferative period and undergo neurogenesis only at tailbud stage. Recent studies have identified several important components of the molecular pathways controlling posterior neurogenesis, but little is known about those controlling the timing and positioning of anterior neurogenesis. We investigate the role of Xrx1, a homeobox gene required for eye and anterior brain development, in the control of proliferation and neurogenesis of the anterior neural plate. Xrx1 is expressed in the entire proliferative region of the anterior neural plate delimited by cells expressing the neuronal determination gene X-ngnr-1, the neurogenic gene X-Delta-1, and the cell cycle inhibitor p27Xic1. Positive and negative signals position Xrx1 expression to this region. Xrx1 is activated by chordin and Hedgehog gene signaling, which induce anterior and proliferative fate, and is repressed by the differentiation-promoting activity of neurogenin and retinoic acid. Xrx1 is required for anterior neural plate proliferation and, when overexpressed, induces proliferation, inhibits X-ngnr-1, X-Delta-1 and N-tubulin and counteracts X-ngnr-1- and retinoic acid-mediated differentiation. We find that Xrx1 does not act by increasing lateral inhibition but by inducing the antineurogenic transcriptional repressors Xhairy2 and Zic2, and by repressing p27Xic1. The effects of Xrx1 on proliferation, neurogenesis and gene expression are restricted to the most rostral region of the embryo, implicating this gene as an anterior regulator of neurogenesis.
Collapse
Affiliation(s)
- Massimiliano Andreazzoli
- Dipartimento di Fisiologia e Biochimica, Università degli Studi di Pisa, Via Carducci 13, 56010 Ghezzano (Pisa), Italy.
| | | | | | | | | | | |
Collapse
|