1
|
Alpay EE, Zinani OQH, Hu X, Ay A, Özbudak EM. DeltaC and DeltaD ligands play different roles in the segmentation clock dynamics. Nat Commun 2025; 16:2413. [PMID: 40069165 PMCID: PMC11897328 DOI: 10.1038/s41467-025-57645-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 02/27/2025] [Indexed: 03/15/2025] Open
Abstract
The vertebrate segmentation clock drives periodic somite segmentation during embryonic development. Her1 and Her7 clock proteins generate oscillatory expression of their own genes as well as that of deltaC in zebrafish. In turn, DeltaC and DeltaD ligands activate Notch signaling, which then activates transcription of clock genes in neighboring cells. While DeltaC and DeltaD proteins form homo- and heterodimers, only DeltaC-containing oscillatory dimers were expected to be functional. To investigate the contributions of DeltaC and DeltaD proteins on the transcription of her1 and her7 segmentation clock genes, we counted their transcripts by performing single molecule fluorescent in situ hybridization imaging in different genetic backgrounds of zebrafish embryos. Surprisingly, we found that DeltaD homodimers are also functional. We further found that Notch signaling promotes transcription of both deltaC and deltaD genes, thereby creating a previously unnoticed positive feedback loop. Our computational model highlighted the intriguing differential roles of DeltaC and DeltaD dimers on the clock synchronization and transcript numbers, respectively. We anticipate that a mechanistic understanding of the Notch signaling pathway will not only shed light on the mechanism driving robust somite segmentation but also inspire similar quantitative studies in other tissues and organs.
Collapse
Affiliation(s)
- Eslim Esra Alpay
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Oriana Q H Zinani
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xiyan Hu
- Department of Mathematics, Colgate University, Hamilton, NY, USA
- Department of Computer Science, Colgate University, Hamilton, NY, USA
| | - Ahmet Ay
- Department of Mathematics, Colgate University, Hamilton, NY, USA
- Department of Biology, Colgate University, Hamilton, NY, USA
| | - Ertuğrul M Özbudak
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
2
|
Ulhaq ZS, You MS, Yabe T, Takada S, Chen JK, Ogino Y, Jiang YJ, Tse WKF. Fgf8 contributes to the pathogenesis of Nager syndrome. Int J Biol Macromol 2024; 280:135692. [PMID: 39288852 DOI: 10.1016/j.ijbiomac.2024.135692] [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: 11/17/2023] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/19/2024]
Abstract
Nager syndrome (NS, OMIM 154400) is a rare disease characterized by craniofacial and limb malformations due to variants in the gene encoding splicing factor 3B subunit 4 (SF3B4). Although various noncanonical functions of SF3B4 unrelated to splicing have been previously described, limited studies elucidate molecular mechanisms underlying NS pathogenesis. Here we showed that sf3b4-deficient fish displayed craniofacial and segmentation defects associated with suppression of fgf8 levels, which perturbed FGF signaling and neural crest cell (NCC) expression. Our finding also pointed out that oxidative stress-induced apoptosis was prominently detected in sf3b4-deficient fish and may further exaggerate the bone remodeling process. Notably, injection of exogenous FGF8 significantly rescued the demonstrated defects in sf3b4-deficient fish, which further supported the participation of Fgf8 in NS pathogenesis. Overall, our study provides valuable insights into the molecular mechanism underlying developmental abnormalities observed in NS and suggests future therapeutic strategies to protect against the pathogenesis of NS and possibilities for preventing severe outcomes.
Collapse
Affiliation(s)
- Zulvikar Syambani Ulhaq
- Laboratory of Developmental Disorders and Toxicology, Center for Promotion of International Education and Research, Faculty of Agriculture, Kyushu University, Fukuoka 819-0395, Japan; Research Center for Pre-clinical and Clinical Medicine, National Research and Innovation Agency Republic of Indonesia, Cibinong 16911, Indonesia.
| | - May-Su You
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli County 350, Taiwan
| | - Taijiro Yabe
- National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan; Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan; The Graduate University for Advanced Studies, SOKENDAI, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan
| | - Shinji Takada
- National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan; Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan; The Graduate University for Advanced Studies, SOKENDAI, 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan
| | - Jen-Kun Chen
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Miaoli County 350, Taiwan
| | - Yukiko Ogino
- Laboratory of Aquatic Molecular Developmental Biology, Center for Promotion of International Education and Research, Faculty of Agriculture, Kyushu University, Fukuoka 819-0395, Japan
| | - Yun-Jin Jiang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli County 350, Taiwan
| | - William Ka Fai Tse
- Laboratory of Developmental Disorders and Toxicology, Center for Promotion of International Education and Research, Faculty of Agriculture, Kyushu University, Fukuoka 819-0395, Japan.
| |
Collapse
|
3
|
Wopat S, Adhyapok P, Daga B, Crawford JM, Norman J, Bagwell J, Peskin B, Magre I, Fogerson SM, Levic DS, Di Talia S, Kiehart DP, Charbonneau P, Bagnat M. Notochord segmentation in zebrafish controlled by iterative mechanical signaling. Dev Cell 2024; 59:1860-1875.e5. [PMID: 38697108 PMCID: PMC11265980 DOI: 10.1016/j.devcel.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 01/25/2024] [Accepted: 04/12/2024] [Indexed: 05/04/2024]
Abstract
In bony fishes, patterning of the vertebral column, or spine, is guided by a metameric blueprint established in the notochord sheath. Notochord segmentation begins days after somitogenesis concludes and can occur in its absence. However, somite patterning defects lead to imprecise notochord segmentation, suggesting that these processes are linked. Here, we identify that interactions between the notochord and the axial musculature ensure precise spatiotemporal segmentation of the zebrafish spine. We demonstrate that myoseptum-notochord linkages drive notochord segment initiation by locally deforming the notochord extracellular matrix and recruiting focal adhesion machinery at these contact points. Irregular somite patterning alters this mechanical signaling, causing non-sequential and dysmorphic notochord segmentation, leading to altered spine development. Using a model that captures myoseptum-notochord interactions, we find that a fixed spatial interval is critical for driving sequential segment initiation. Thus, mechanical coupling of axial tissues facilitates spatiotemporal spine patterning.
Collapse
Affiliation(s)
- Susan Wopat
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Priyom Adhyapok
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Bijoy Daga
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | | | - James Norman
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Jennifer Bagwell
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Brianna Peskin
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Indrasen Magre
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | | | - Daniel S Levic
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Stefano Di Talia
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | | | - Patrick Charbonneau
- Department of Chemistry, Duke University, Durham, NC 27708, USA; Department of Physics, Duke University, Durham, NC 27708, USA.
| | - Michel Bagnat
- Department of Cell Biology, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
4
|
Maeda Y, Kageyama R. The significance of ultradian oscillations in development. Curr Opin Genet Dev 2024; 86:102180. [PMID: 38522266 DOI: 10.1016/j.gde.2024.102180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/10/2024] [Accepted: 03/01/2024] [Indexed: 03/26/2024]
Abstract
Genes regulating developmental processes have been identified, but the mechanisms underlying their expression with the correct timing are still under investigation. Several genes show oscillatory expression that regulates the timing of developmental processes, such as somitogenesis and neurogenesis. These oscillations are also important for other developmental processes, such as cell proliferation and differentiation. In this review, we discuss the significance of oscillatory gene expression in developmental time and other forms of regulation.
Collapse
Affiliation(s)
- Yuki Maeda
- RIKEN Center for Brain Science, Wako 351-0198, Japan
| | | |
Collapse
|
5
|
Chandel AS, Keseroglu K, Özbudak EM. Oscillatory control of embryonic development. Development 2024; 151:dev202191. [PMID: 38727565 PMCID: PMC11128281 DOI: 10.1242/dev.202191] [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] [Indexed: 05/28/2024]
Abstract
Proper embryonic development depends on the timely progression of a genetic program. One of the key mechanisms for achieving precise control of developmental timing is to use gene expression oscillations. In this Review, we examine how gene expression oscillations encode temporal information during vertebrate embryonic development by discussing the gene expression oscillations occurring during somitogenesis, neurogenesis, myogenesis and pancreas development. These oscillations play important but varied physiological functions in different contexts. Oscillations control the period of somite formation during somitogenesis, whereas they regulate the proliferation-to-differentiation switch of stem cells and progenitor cells during neurogenesis, myogenesis and pancreas development. We describe the similarities and differences of the expression pattern in space (i.e. whether oscillations are synchronous or asynchronous across neighboring cells) and in time (i.e. different time scales) of mammalian Hes/zebrafish Her genes and their targets in different tissues. We further summarize experimental evidence for the functional role of their oscillations. Finally, we discuss the outstanding questions for future research.
Collapse
Affiliation(s)
- Angad Singh Chandel
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Systems Biology and Physiology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Kemal Keseroglu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Ertuğrul M. Özbudak
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| |
Collapse
|
6
|
McMillen P, Levin M. Collective intelligence: A unifying concept for integrating biology across scales and substrates. Commun Biol 2024; 7:378. [PMID: 38548821 PMCID: PMC10978875 DOI: 10.1038/s42003-024-06037-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 03/11/2024] [Indexed: 04/01/2024] Open
Abstract
A defining feature of biology is the use of a multiscale architecture, ranging from molecular networks to cells, tissues, organs, whole bodies, and swarms. Crucially however, biology is not only nested structurally, but also functionally: each level is able to solve problems in distinct problem spaces, such as physiological, morphological, and behavioral state space. Percolating adaptive functionality from one level of competent subunits to a higher functional level of organization requires collective dynamics: multiple components must work together to achieve specific outcomes. Here we overview a number of biological examples at different scales which highlight the ability of cellular material to make decisions that implement cooperation toward specific homeodynamic endpoints, and implement collective intelligence by solving problems at the cell, tissue, and whole-organism levels. We explore the hypothesis that collective intelligence is not only the province of groups of animals, and that an important symmetry exists between the behavioral science of swarms and the competencies of cells and other biological systems at different scales. We then briefly outline the implications of this approach, and the possible impact of tools from the field of diverse intelligence for regenerative medicine and synthetic bioengineering.
Collapse
Affiliation(s)
- Patrick McMillen
- Department of Biology, Tufts University, Medford, MA, 02155, USA
- Allen Discovery Center at Tufts University, Medford, MA, 02155, USA
| | - Michael Levin
- Department of Biology, Tufts University, Medford, MA, 02155, USA.
- Allen Discovery Center at Tufts University, Medford, MA, 02155, USA.
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.
| |
Collapse
|
7
|
Ramesh PS, Chu LF. Species-specific roles of the Notch ligands, receptors, and targets orchestrating the signaling landscape of the segmentation clock. Front Cell Dev Biol 2024; 11:1327227. [PMID: 38348091 PMCID: PMC10859470 DOI: 10.3389/fcell.2023.1327227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/20/2023] [Indexed: 02/15/2024] Open
Abstract
Somitogenesis is a hallmark feature of all vertebrates and some invertebrate species that involves the periodic formation of block-like structures called somites. Somites are transient embryonic segments that eventually establish the entire vertebral column. A highly conserved molecular oscillator called the segmentation clock underlies this periodic event and the pace of this clock regulates the pace of somite formation. Although conserved signaling pathways govern the clock in most vertebrates, the mechanisms underlying the species-specific divergence in various clock characteristics remain elusive. For example, the segmentation clock in classical model species such as zebrafish, chick, and mouse embryos tick with a periodicity of ∼30, ∼90, and ∼120 min respectively. This enables them to form the species-specific number of vertebrae during their overall timespan of somitogenesis. Here, we perform a systematic review of the species-specific features of the segmentation clock with a keen focus on mouse embryos. We perform this review using three different perspectives: Notch-responsive clock genes, ligand-receptor dynamics, and synchronization between neighboring oscillators. We further review reports that use non-classical model organisms and in vitro model systems that complement our current understanding of the segmentation clock. Our review highlights the importance of comparative developmental biology to further our understanding of this essential developmental process.
Collapse
Affiliation(s)
- Pranav S. Ramesh
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Reproductive Biology and Regenerative Medicine Research Group, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Calgary, AB, Canada
| | - Li-Fang Chu
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Reproductive Biology and Regenerative Medicine Research Group, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Calgary, AB, Canada
| |
Collapse
|
8
|
Loureiro C, Venzin OF, Oates AC. Generation of patterns in the paraxial mesoderm. Curr Top Dev Biol 2023; 159:372-405. [PMID: 38729682 DOI: 10.1016/bs.ctdb.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
The Segmentation Clock is a tissue-level patterning system that enables the segmentation of the vertebral column precursors into transient multicellular blocks called somites. This patterning system comprises a set of elements that are essential for correct segmentation. Under the so-called "Clock and Wavefront" model, the system consists of two elements, a genetic oscillator that manifests itself as traveling waves of gene expression, and a regressing wavefront that transforms the temporally periodic signal encoded in the oscillations into a permanent spatially periodic pattern of somite boundaries. Over the last twenty years, every new discovery about the Segmentation Clock has been tightly linked to the nomenclature of the "Clock and Wavefront" model. This constrained allocation of discoveries into these two elements has generated long-standing debates in the field as what defines molecularly the wavefront and how and where the interaction between the two elements establishes the future somite boundaries. In this review, we propose an expansion of the "Clock and Wavefront" model into three elements, "Clock", "Wavefront" and signaling gradients. We first provide a detailed description of the components and regulatory mechanisms of each element, and we then examine how the spatiotemporal integration of the three elements leads to the establishment of the presumptive somite boundaries. To be as exhaustive as possible, we focus on the Segmentation Clock in zebrafish. Furthermore, we show how this three-element expansion of the model provides a better understanding of the somite formation process and we emphasize where our current understanding of this patterning system remains obscure.
Collapse
Affiliation(s)
- Cristina Loureiro
- Institute of Bioengineering, School of Life Sciences, Swiss Federal Institute of Technology Lausanne EPFL, Switzerland
| | - Olivier F Venzin
- Institute of Bioengineering, School of Life Sciences, Swiss Federal Institute of Technology Lausanne EPFL, Switzerland
| | - Andrew C Oates
- Institute of Bioengineering, School of Life Sciences, Swiss Federal Institute of Technology Lausanne EPFL, Switzerland.
| |
Collapse
|
9
|
Simsek MF, Özbudak EM. A design logic for sequential segmentation across organisms. FEBS J 2023; 290:5086-5093. [PMID: 37422856 PMCID: PMC10774455 DOI: 10.1111/febs.16899] [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: 04/19/2023] [Revised: 05/24/2023] [Accepted: 07/06/2023] [Indexed: 07/11/2023]
Abstract
Multitudes of organisms display metameric compartmentalization of their body plan. Segmentation of these compartments happens sequentially in diverse phyla. In several sequentially segmenting species, periodically active molecular clocks and signaling gradients have been found. The clocks are proposed to control the timing of segmentation, while the gradients are proposed to instruct the positions of segment boundaries. However, the identity of the clock and gradient molecules differs across species. Furthermore, sequential segmentation of a basal chordate, Amphioxus, continues at late stages when the small tail bud cell population cannot establish long-range signaling gradients. Thus, it remains to be explained how a conserved morphological trait (i.e., sequential segmentation) is achieved by using different molecules or molecules with different spatial profiles. Here, we first focus on sequential segmentation of somites in vertebrate embryos and then draw parallels with other species. Thereafter, we propose a candidate design principle that has the potential to answer this puzzling question.
Collapse
Affiliation(s)
- M Fethullah Simsek
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, OH, USA
| | - Ertuğrul M Özbudak
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, OH, USA
| |
Collapse
|
10
|
Yamanaka Y, Hamidi S, Yoshioka-Kobayashi K, Munira S, Sunadome K, Zhang Y, Kurokawa Y, Ericsson R, Mieda A, Thompson JL, Kerwin J, Lisgo S, Yamamoto T, Moris N, Martinez-Arias A, Tsujimura T, Alev C. Reconstituting human somitogenesis in vitro. Nature 2023; 614:509-520. [PMID: 36543322 DOI: 10.1038/s41586-022-05649-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
The segmented body plan of vertebrates is established during somitogenesis, a well-studied process in model organisms; however, the details of this process in humans remain largely unknown owing to ethical and technical limitations. Despite recent advances with pluripotent stem cell-based approaches1-5, models that robustly recapitulate human somitogenesis in both space and time remain scarce. Here we introduce a pluripotent stem cell-derived mesoderm-based 3D model of human segmentation and somitogenesis-which we termed 'axioloid'-that captures accurately the oscillatory dynamics of the segmentation clock and the morphological and molecular characteristics of sequential somite formation in vitro. Axioloids show proper rostrocaudal patterning of forming segments and robust anterior-posterior FGF-WNT signalling gradients and retinoic acid signalling components. We identify an unexpected critical role of retinoic acid signalling in the stabilization of forming segments, indicating distinct, but also synergistic effects of retinoic acid and extracellular matrix on the formation and epithelialization of somites. Comparative analysis demonstrates marked similarities of axioloids to the human embryo, further validated by the presence of a Hox code in axioloids. Finally, we demonstrate the utility of axioloids for studying the pathogenesis of human congenital spine diseases using induced pluripotent stem cells with mutations in HES7 and MESP2. Our results indicate that axioloids represent a promising platform for the study of axial development and disease in humans.
Collapse
Affiliation(s)
- Yoshihiro Yamanaka
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| | - Sofiane Hamidi
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| | | | - Sirajam Munira
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| | - Kazunori Sunadome
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Yi Zhang
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| | - Yuzuru Kurokawa
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| | - Rolf Ericsson
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| | - Ai Mieda
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| | | | - Janet Kerwin
- HDBR, Newcastle University, Newcastle Upon Tyne, UK
| | - Steven Lisgo
- HDBR, Newcastle University, Newcastle Upon Tyne, UK
| | - Takuya Yamamoto
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Medical-Risk Avoidance Based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| | | | - Alfonso Martinez-Arias
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra and ICREA, Barcelona, Spain
| | - Taro Tsujimura
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| | - Cantas Alev
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan.
| |
Collapse
|
11
|
Uriu K, Morelli LG. Orchestration of tissue shape changes and gene expression patterns in development. Semin Cell Dev Biol 2023; 147:24-33. [PMID: 36631335 DOI: 10.1016/j.semcdb.2022.12.009] [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: 10/28/2022] [Revised: 12/27/2022] [Accepted: 12/27/2022] [Indexed: 01/11/2023]
Abstract
In development, tissue shape changes and gene expression patterns give rise to morphogenesis. Understanding tissue shape changes requires the analysis of mechanical properties of the tissue such as tissue rigidity, cell influx from neighboring tissues, cell shape changes and cell proliferation. Local and global gene expression patterns can be influenced by neighbor exchange and tissue shape changes. Here we review recent studies on the mechanisms for tissue elongation and its influences on dynamic gene expression patterns by focusing on vertebrate somitogenesis. We first introduce mechanical and biochemical properties of the segmenting tissue that drive tissue elongation. Then, we discuss patterning in the presence of cell mixing, scaling of signaling gradients, and dynamic phase waves of rhythmic gene expression under tissue shape changes. We also highlight the importance of theoretical approaches to address the relation between tissue shape changes and patterning.
Collapse
Affiliation(s)
- Koichiro Uriu
- Graduate School of Natural Science and Technology, Kanazawa University, Kakuma-machi, Kanazawa 920-1192 Japan.
| | - Luis G Morelli
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Polo Científico Tecnológico, Godoy Cruz 2390, C1425FQD, Buenos Aires, Argentina; Departamento de Física, FCEyN UBA, Ciudad Universitaria, 1428 Buenos Aires, Argentina.
| |
Collapse
|
12
|
Blatnik MC, Gallagher TL, Amacher SL. Keeping development on time: Insights into post-transcriptional mechanisms driving oscillatory gene expression during vertebrate segmentation. WILEY INTERDISCIPLINARY REVIEWS. RNA 2023; 14:e1751. [PMID: 35851751 PMCID: PMC9840655 DOI: 10.1002/wrna.1751] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/13/2022] [Accepted: 06/20/2022] [Indexed: 01/31/2023]
Abstract
Biological time keeping, or the duration and tempo at which biological processes occur, is a phenomenon that drives dynamic molecular and morphological changes that manifest throughout many facets of life. In some cases, the molecular mechanisms regulating the timing of biological transitions are driven by genetic oscillations, or periodic increases and decreases in expression of genes described collectively as a "molecular clock." In vertebrate animals, molecular clocks play a crucial role in fundamental patterning and cell differentiation processes throughout development. For example, during early vertebrate embryogenesis, the segmentation clock regulates the patterning of the embryonic mesoderm into segmented blocks of tissue called somites, which later give rise to axial skeletal muscle and vertebrae. Segmentation clock oscillations are characterized by rapid cycles of mRNA and protein expression. For segmentation clock oscillations to persist, the transcript and protein molecules of clock genes must be short-lived. Faithful, rhythmic, genetic oscillations are sustained by precise regulation at many levels, including post-transcriptional regulation, and such mechanisms are essential for proper vertebrate development. This article is categorized under: RNA Export and Localization > RNA Localization RNA Turnover and Surveillance > Regulation of RNA Stability Translation > Regulation.
Collapse
Affiliation(s)
- Monica C. Blatnik
- The Ohio State University, Department of Molecular Genetics, Columbus, Ohio, 43210-1132, United States
| | - Thomas L. Gallagher
- The Ohio State University, Department of Molecular Genetics, Columbus, Ohio, 43210-1132, United States
| | - Sharon L. Amacher
- The Ohio State University, Department of Molecular Genetics, Columbus, Ohio, 43210-1132, United States
| |
Collapse
|
13
|
Periodic inhibition of Erk activity drives sequential somite segmentation. Nature 2023; 613:153-159. [PMID: 36517597 PMCID: PMC9846577 DOI: 10.1038/s41586-022-05527-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/04/2022] [Indexed: 12/23/2022]
Abstract
Sequential segmentation creates modular body plans of diverse metazoan embryos1-4. Somitogenesis establishes the segmental pattern of the vertebrate body axis. A molecular segmentation clock in the presomitic mesoderm sets the pace of somite formation4. However, how cells are primed to form a segment boundary at a specific location remains unclear. Here we developed precise reporters for the clock and double-phosphorylated Erk (ppErk) gradient in zebrafish. We show that the Her1-Her7 oscillator drives segmental commitment by periodically lowering ppErk, therefore projecting its oscillation onto the ppErk gradient. Pulsatile inhibition of the ppErk gradient can fully substitute for the role of the clock, and kinematic clock waves are dispensable for sequential segmentation. The clock functions upstream of ppErk, which in turn enables neighbouring cells to discretely establish somite boundaries in zebrafish5. Molecularly divergent clocks and morphogen gradients were identified in sequentially segmenting species3,4,6-8. Our findings imply that versatile clocks may establish sequential segmentation in diverse species provided that they inhibit gradients.
Collapse
|
14
|
McMillen P, Walker SI, Levin M. Information Theory as an Experimental Tool for Integrating Disparate Biophysical Signaling Modules. Int J Mol Sci 2022; 23:9580. [PMID: 36076979 PMCID: PMC9455895 DOI: 10.3390/ijms23179580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/13/2022] [Accepted: 08/14/2022] [Indexed: 11/16/2022] Open
Abstract
There is a growing appreciation in the fields of cell biology and developmental biology that cells collectively process information in time and space. While many powerful molecular tools exist to observe biophysical dynamics, biologists must find ways to quantitatively understand these phenomena at the systems level. Here, we present a guide for the application of well-established information theory metrics to biological datasets and explain these metrics using examples from cell, developmental and regenerative biology. We introduce a novel computational tool named after its intended purpose, calcium imaging, (CAIM) for simple, rigorous application of these metrics to time series datasets. Finally, we use CAIM to study calcium and cytoskeletal actin information flow patterns between Xenopus laevis embryonic animal cap stem cells. The tools that we present here should enable biologists to apply information theory to develop a systems-level understanding of information processing across a diverse array of experimental systems.
Collapse
Affiliation(s)
- Patrick McMillen
- Allen Discovery Center at Tufts University, Medford, MA 02155, USA
| | - Sara I. Walker
- Beyond Center for Fundamental Concepts in Science, Arizona State University, Tempe, AZ 85281, USA
- Santa Fe Institute, Santa Fe, NM 87501, USA
| | - Michael Levin
- Allen Discovery Center at Tufts University, Medford, MA 02155, USA
| |
Collapse
|
15
|
Carraco G, Martins-Jesus AP, Andrade RP. The vertebrate Embryo Clock: Common players dancing to a different beat. Front Cell Dev Biol 2022; 10:944016. [PMID: 36036002 PMCID: PMC9403190 DOI: 10.3389/fcell.2022.944016] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 07/11/2022] [Indexed: 11/25/2022] Open
Abstract
Vertebrate embryo somitogenesis is the earliest morphological manifestation of the characteristic patterned structure of the adult axial skeleton. Pairs of somites flanking the neural tube are formed periodically during early development, and the molecular mechanisms in temporal control of this early patterning event have been thoroughly studied. The discovery of a molecular Embryo Clock (EC) underlying the periodicity of somite formation shed light on the importance of gene expression dynamics for pattern formation. The EC is now known to be present in all vertebrate organisms studied and this mechanism was also described in limb development and stem cell differentiation. An outstanding question, however, remains unanswered: what sets the different EC paces observed in different organisms and tissues? This review aims to summarize the available knowledge regarding the pace of the EC, its regulation and experimental manipulation and to expose new questions that might help shed light on what is still to unveil.
Collapse
Affiliation(s)
- Gil Carraco
- ABC-RI, Algarve Biomedical Center Research Institute, Faro, Portugal
- Faculdade de Medicina e Ciências Biomédicas (FMCB), Universidade do Algarve, Campus de Gambelas, Faro, Portugal
| | | | - Raquel P. Andrade
- ABC-RI, Algarve Biomedical Center Research Institute, Faro, Portugal
- Faculdade de Medicina e Ciências Biomédicas (FMCB), Universidade do Algarve, Campus de Gambelas, Faro, Portugal
- Champalimaud Research Program, Champalimaud Center for the Unknown, Lisbon, Portugal
- *Correspondence: Raquel P. Andrade,
| |
Collapse
|
16
|
Nian FS, Hou PS. Evolving Roles of Notch Signaling in Cortical Development. Front Neurosci 2022; 16:844410. [PMID: 35422684 PMCID: PMC9001970 DOI: 10.3389/fnins.2022.844410] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/15/2022] [Indexed: 01/09/2023] Open
Abstract
Expansion of the neocortex is thought to pave the way toward acquisition of higher cognitive functions in mammals. The highly conserved Notch signaling pathway plays a crucial role in this process by regulating the size of the cortical progenitor pool, in part by controlling the balance between self-renewal and differentiation. In this review, we introduce the components of Notch signaling pathway as well as the different mode of molecular mechanisms, including trans- and cis-regulatory processes. We focused on the recent findings with regard to the expression pattern and levels in regulating neocortical formation in mammals and its interactions with other known signaling pathways, including Slit–Robo signaling and Shh signaling. Finally, we review the functions of Notch signaling pathway in different species as well as other developmental process, mainly somitogenesis, to discuss how modifications to the Notch signaling pathway can drive the evolution of the neocortex.
Collapse
Affiliation(s)
- Fang-Shin Nian
- Institute of Anatomy and Cell Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Brain Science, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Pei-Shan Hou
- Institute of Anatomy and Cell Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
- *Correspondence: Pei-Shan Hou,
| |
Collapse
|
17
|
Pourquié O. A brief history of the segmentation clock. Dev Biol 2022; 485:24-36. [DOI: 10.1016/j.ydbio.2022.02.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 11/16/2022]
|
18
|
Live imaging approach of dynamic multicellular responses in ERK signaling during vertebrate tissue development. Biochem J 2022; 479:129-143. [PMID: 35050327 PMCID: PMC8883488 DOI: 10.1042/bcj20210557] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/28/2021] [Accepted: 01/05/2022] [Indexed: 11/17/2022]
Abstract
The chemical and mechanical responses of cells via the exchange of information during growth and development result in the formation of biological tissues. Information processing within the cells through the signaling pathways and networks inherent to the constituent cells has been well-studied. However, the cell signaling mechanisms responsible for generating dynamic multicellular responses in developing tissues remain unclear. Here, I review the dynamic multicellular response systems during the development and growth of vertebrate tissues based on the extracellular signal-regulated kinase (ERK) pathway. First, an overview of the function of the ERK signaling network in cells is provided, followed by descriptions of biosensors essential for live imaging of the quantification of ERK activity in tissues. Then adducing four examples, I highlight the contribution of live imaging techniques for studying the involvement of spatio-temporal patterns of ERK activity change in tissue development and growth. In addition, theoretical implications of ERK signaling are also discussed from the viewpoint of dynamic systems. This review might help in understanding ERK-mediated dynamic multicellular responses and tissue morphogenesis.
Collapse
|
19
|
Chou KT, Lee DYD, Chiou JG, Galera-Laporta L, Ly S, Garcia-Ojalvo J, Süel GM. A segmentation clock patterns cellular differentiation in a bacterial biofilm. Cell 2022; 185:145-157.e13. [PMID: 34995513 PMCID: PMC8754390 DOI: 10.1016/j.cell.2021.12.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/13/2021] [Accepted: 11/30/2021] [Indexed: 01/09/2023]
Abstract
Contrary to multicellular organisms that display segmentation during development, communities of unicellular organisms are believed to be devoid of such sophisticated patterning. Unexpectedly, we find that the gene expression underlying the nitrogen stress response of a developing Bacillus subtilis biofilm becomes organized into a ring-like pattern. Mathematical modeling and genetic probing of the underlying circuit indicate that this patterning is generated by a clock and wavefront mechanism, similar to that driving vertebrate somitogenesis. We experimentally validated this hypothesis by showing that predicted nutrient conditions can even lead to multiple concentric rings, resembling segments. We additionally confirmed that this patterning mechanism is driven by cell-autonomous oscillations. Importantly, we show that the clock and wavefront process also spatially patterns sporulation within the biofilm. Together, these findings reveal a biofilm segmentation clock that organizes cellular differentiation in space and time, thereby challenging the paradigm that such patterning mechanisms are exclusive to plant and animal development.
Collapse
Affiliation(s)
- Kwang-Tao Chou
- Molecular Biology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Dong-Yeon D Lee
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Jian-Geng Chiou
- Molecular Biology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Leticia Galera-Laporta
- Molecular Biology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - San Ly
- Molecular Biology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Jordi Garcia-Ojalvo
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Gürol M Süel
- Molecular Biology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA; San Diego Center for Systems Biology, University of California San Diego, La Jolla, CA 92093-0380, USA; Center for Microbiome Innovation, University of California San Diego, La Jolla, CA 92093-0380, USA.
| |
Collapse
|
20
|
Venzin OF, Oates AC. What are you synching about? Emerging complexity of Notch signaling in the segmentation clock. Dev Biol 2020; 460:40-54. [DOI: 10.1016/j.ydbio.2019.06.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 06/30/2019] [Accepted: 06/30/2019] [Indexed: 10/26/2022]
|
21
|
Noise in the Vertebrate Segmentation Clock Is Boosted by Time Delays but Tamed by Notch Signaling. Cell Rep 2019; 23:2175-2185.e4. [PMID: 29768214 PMCID: PMC5989725 DOI: 10.1016/j.celrep.2018.04.069] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 03/05/2018] [Accepted: 04/16/2018] [Indexed: 02/04/2023] Open
Abstract
Taming cell-to-cell variability in gene expression is critical for precise pattern formation during embryonic development. To investigate the source and buffering mechanism of expression variability, we studied a biological clock, the vertebrate segmentation clock, controlling the precise spatiotemporal patterning of the vertebral column. By counting single transcripts of segmentation clock genes in zebrafish, we show that clock genes have low RNA amplitudes and expression variability is primarily driven by gene extrinsic sources, which is suppressed by Notch signaling. We further show that expression noise surprisingly increases from the posterior progenitor zone to the anterior segmentation and differentiation zone. Our computational model reproduces the spatial noise profile by incorporating spatially increasing time delays in gene expression. Our results, suggesting that expression variability is controlled by the balance of time delays and cell signaling in a vertebrate tissue, will shed light on the accuracy of natural clocks in multi-cellular systems and inspire engineering of robust synthetic oscillators.
Collapse
|
22
|
Abstract
Consistent asymmetries between the left and right sides of animal bodies are common. For example, the internal organs of vertebrates are left-right (L-R) asymmetric in a stereotyped fashion. Other structures, such as the skeleton and muscles, are largely symmetric. This Review considers how symmetries and asymmetries form alongside each other within the embryo, and how they are then maintained during growth. I describe how asymmetric signals are generated in the embryo. Using the limbs and somites as major examples, I then address mechanisms for protecting symmetrically forming tissues from asymmetrically acting signals. These examples reveal that symmetry should not be considered as an inherent background state, but instead must be actively maintained throughout multiple phases of embryonic patterning and organismal growth.
Collapse
Affiliation(s)
- Daniel T Grimes
- Institute of Molecular Biology, Department of Biology, University of Oregon, Eugene, OR 97403, USA
| |
Collapse
|
23
|
Notch-mediated inhibition of neurogenesis is required for zebrafish spinal cord morphogenesis. Sci Rep 2019; 9:9958. [PMID: 31292468 PMCID: PMC6620349 DOI: 10.1038/s41598-019-46067-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 06/18/2019] [Indexed: 12/26/2022] Open
Abstract
The morphogenesis of the nervous system requires coordinating the specification and differentiation of neural precursor cells, the establishment of neuroepithelial tissue architecture and the execution of specific cellular movements. How these aspects of neural development are linked is incompletely understood. Here we inactivate a major regulator of embryonic neurogenesis - the Delta/Notch pathway - and analyze the effect on zebrafish central nervous system morphogenesis. While some parts of the nervous system can establish neuroepithelial tissue architecture independently of Notch, Notch signaling is essential for spinal cord morphogenesis. In this tissue, Notch signaling is required to repress neuronal differentiation and allow thereby the emergence of neuroepithelial apico-basal polarity. Notch-mediated suppression of neurogenesis is also essential for the execution of specific morphogenetic movements of zebrafish spinal cord precursor cells. In the wild-type neural tube, cells divide at the organ midline to contribute one daughter cell to each organ half. Notch signaling deficient animals fail to display this behavior and therefore form a misproportioned spinal cord. Taken together, our findings show that Notch-mediated suppression of neurogenesis is required to allow the execution of morphogenetic programs that shape the zebrafish spinal cord.
Collapse
|
24
|
Characterization of paralogous uncx transcription factor encoding genes in zebrafish. Gene X 2019; 721S:100011. [PMID: 31193955 PMCID: PMC6543554 DOI: 10.1016/j.gene.2019.100011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 02/12/2019] [Accepted: 02/15/2019] [Indexed: 12/23/2022] Open
Abstract
The paired-type homeodomain transcription factor Uncx is involved in multiple processes of embryogenesis in vertebrates. Reasoning that zebrafish genes uncx4.1 and uncx are orthologs of mouse Uncx, we studied their genomic environment and developmental expression. Evolutionary analyses indicate the zebrafish uncx genes as being paralogs deriving from teleost-specific whole-genome duplication. Whole-mount in situ mRNA hybridization of uncx transcripts in zebrafish embryos reveals novel expression domains, confirms those previously known, and suggests sub-functionalization of paralogs. Using genetic mutants and pharmacological inhibitors, we investigate the role of signaling pathways on the expression of zebrafish uncx genes in developing somites. In identifying putative functional role(s) of zebrafish uncx genes, we hypothesized that they encode transcription factors that coordinate growth and innervation of somitic muscles. The Uncx4.1 and Uncx genes derive from the teleost-specific whole-genome duplication. Uncx genes are expressed during embryogenesis in unique and overlapping domains. Uncx gene expression during somite differentiation is regulated by FGF signaling. Synteny and expression profiles correlate Uncx genes with axon guidance.
Collapse
Key Words
- AP, antero-posterior
- Ace, acerebellar
- CAMP, conserved ancestral microsyntenic pairs
- CNE, conserved non-coding elements
- CRM, cis-regulatory module
- CS, Corpuscle of Stannius
- CaP, caudal primary motor neuron axons
- Ce, cerebellum
- Development
- Di, diencephalon
- Elfn1, Extracellular Leucine Rich Repeat And Fibronectin Type III Domain Containing 1
- Ey, eye
- FB, forebrain
- FGF, fibroblast growth factor
- Flh, floating head
- HB, hindbrain
- HM, hybridization mix
- Hy, hypothalamus
- MO, morpholino
- Mical, molecule interacting with CasL
- No, notochord
- OP, olfactory placode
- OT, optic tectum
- PA, pharyngeal arches
- PSM, presomitic mesoderm
- SC, spinal cord
- Shh, sonic hedgehog
- Signaling pathway
- So, somites
- Synteny
- TSGD
- TSGD, teleost-specific genome duplication
- Te, telencephalon
- Th, thalamus
- Uncx
- VLP, ventro-lateral-posterior
- WIHC, whole-mount immunohistochemistry
- WISH, whole-mount in situ hybridization
- YE, yolk extension
- Yo, yolk
- Zebrafish
- cyc, cyclops
- fss, fused-somites
- hpf, hours post fertilization
- ptc, patched
- smu, slow-muscle-omitted
- syu, sonic-you
- yot, you-too
Collapse
|
25
|
Pedone E, Marucci L. Role of β-Catenin Activation Levels and Fluctuations in Controlling Cell Fate. Genes (Basel) 2019; 10:genes10020176. [PMID: 30823613 PMCID: PMC6410200 DOI: 10.3390/genes10020176] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 02/18/2019] [Indexed: 12/12/2022] Open
Abstract
Cells have developed numerous adaptation mechanisms to external cues by controlling signaling-pathway activity, both qualitatively and quantitatively. The Wnt/β-catenin pathway is a highly conserved signaling pathway involved in many biological processes, including cell proliferation, differentiation, somatic cell reprogramming, development, and cancer. The activity of the Wnt/β-catenin pathway and the temporal dynamics of its effector β-catenin are tightly controlled by complex regulations. The latter encompass feedback loops within the pathway (e.g., a negative feedback loop involving Axin2, a β-catenin transcriptional target) and crosstalk interactions with other signaling pathways. Here, we provide a review shedding light on the coupling between Wnt/β-catenin activation levels and fluctuations across processes and cellular systems; in particular, we focus on development, in vitro pluripotency maintenance, and cancer. Possible mechanisms originating Wnt/β-catenin dynamic behaviors and consequently driving different cellular responses are also reviewed, and new avenues for future research are suggested.
Collapse
Affiliation(s)
- Elisa Pedone
- Department of Engineering Mathematics, University of Bristol, Bristol, BS8 1UB, UK.
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK.
| | - Lucia Marucci
- Department of Engineering Mathematics, University of Bristol, Bristol, BS8 1UB, UK.
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK.
- BrisSynBio, Bristol, BS8 1TQ, UK.
| |
Collapse
|
26
|
Fujino Y, Yamada K, Sugaya C, Ooka Y, Ovara H, Ban H, Akama K, Otosaka S, Kinoshita H, Yamasu K, Mishima Y, Kawamura A. Deadenylation by the CCR4-NOT complex contributes to the turnover of hairy-related mRNAs in the zebrafish segmentation clock. FEBS Lett 2018; 592:3388-3398. [PMID: 30281784 DOI: 10.1002/1873-3468.13261] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/10/2018] [Accepted: 09/23/2018] [Indexed: 01/09/2023]
Abstract
In the zebrafish segmentation clock, hairy/enhancer of split-related genes her1, her7, and hes6 encodes components of core oscillators. Since the expression of cyclic genes proceeds rapidly in the presomitic mesoderm (PSM), these hairy-related mRNAs are subject to strict post-transcriptional regulation. In this study, we demonstrate that inhibition of the CCR4-NOT deadenylase complex lengthens poly(A) tails of hairy-related mRNAs and increases the amount of these mRNAs, which is accompanied by defective somite segmentation. In transgenic embryos, we show that EGFP mRNAs with 3'UTRs of hairy-related genes exhibit turnover similar to endogenous mRNAs. Our results suggest that turnover rates of her1, her7, and hes6 mRNAs are differently regulated by the CCR4-NOT deadenylase complex possibly through their 3'UTRs in the zebrafish PSM.
Collapse
Affiliation(s)
- Yuuri Fujino
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Japan
| | - Kazuya Yamada
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Japan
| | - Chihiro Sugaya
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Japan
| | - Yuko Ooka
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Japan
| | - Hiroki Ovara
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Japan
| | - Hiroyuki Ban
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Japan
| | - Kagari Akama
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Japan
| | - Shiori Otosaka
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Japan
| | - Hirofumi Kinoshita
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Japan
| | - Kyo Yamasu
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Japan
| | - Yuichiro Mishima
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Japan
| | - Akinori Kawamura
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Japan
| |
Collapse
|
27
|
Pinto RA, Almeida-Santos J, Lourenço R, Saúde L. Identification of Dmrt2a downstream genes during zebrafish early development using a timely controlled approach. BMC DEVELOPMENTAL BIOLOGY 2018; 18:14. [PMID: 29914374 PMCID: PMC6006574 DOI: 10.1186/s12861-018-0173-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 05/25/2018] [Indexed: 11/10/2022]
Abstract
BACKGROUND Dmrt2a is a zinc finger like transcription factor with several roles during zebrafish early development: left-right asymmetry, synchronisation of the somite clock genes and fast muscle differentiation. Despite the described functions, Dmrt2a mechanism of action is unknown. Therefore, with this work, we propose to identify Dmrt2a downstream genes during zebrafish early development. RESULTS We generated and validated a heat-shock inducible transgenic line, to timely control dmrt2a overexpression, and dmrt2a mutant lines. We characterised dmrt2a overexpression phenotype and verified that it was very similar to the one described after knockdown of this gene, with left-right asymmetry defects and desynchronisation of somite clock genes. Additionally, we identified a new phenotype of somite border malformation. We generated several dmrt2a mutant lines, but we only detected a weak to negligible phenotype. As dmrt2a has a paralog gene, dmrt2b, with similar functions and expression pattern, we evaluated the possibility of redundancy. We found that dmrt2b does not seem to compensate the lack of dmrt2a. Furthermore, we took advantage of one of our mutant lines to confirm dmrt2a morpholino specificity, which was previously shown to be a robust knockdown tool in two independent studies. Using the described genetic tools to perform and validate a microarray, we were able to identify six genes downstream of Dmrt2a: foxj1b, pxdc1b, cxcl12b, etv2, foxc1b and cyp1a. CONCLUSIONS In this work, we generated and validated several genetic tools for dmrt2a and identified six genes downstream of this transcription factor. The identified genes will be crucial to the future understanding of Dmrt2a mechanism of action in zebrafish.
Collapse
Affiliation(s)
- Rita Alexandra Pinto
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - José Almeida-Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal.,Present address: Instituto Gulbenkian de Ciência, 2780-156, Oeiras, Portugal
| | - Raquel Lourenço
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal.,Present address: CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1150-190, Lisboa, Portugal
| | - Leonor Saúde
- Instituto de Medicina Molecular e Instituto de Histologia e Biologia do Desenvolvimento, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal.
| |
Collapse
|
28
|
Ishimatsu K, Hiscock TW, Collins ZM, Sari DWK, Lischer K, Richmond DL, Bessho Y, Matsui T, Megason SG. Size-reduced embryos reveal a gradient scaling-based mechanism for zebrafish somite formation. Development 2018; 145:dev.161257. [PMID: 29769221 DOI: 10.1242/dev.161257] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 05/09/2018] [Indexed: 12/29/2022]
Abstract
Little is known about how the sizes of animal tissues are controlled. A prominent example is somite size, which varies widely both within an individual and across species. Despite intense study of the segmentation clock governing the timing of somite generation, how it relates to somite size is poorly understood. Here, we examine somite scaling and find that somite size at specification scales with the length of the presomitic mesoderm (PSM) despite considerable variation in PSM length across developmental stages and in surgically size-reduced embryos. Measurement of clock period, axis elongation speed and clock gene expression patterns demonstrate that existing models fail to explain scaling. We posit a 'clock and scaled gradient' model, in which somite boundaries are set by a dynamically scaling signaling gradient across the PSM. Our model not only explains existing data, but also makes a unique prediction that we confirm experimentally - the formation of periodic 'echoes' in somite size following perturbation of the size of one somite. Our findings demonstrate that gradient scaling plays a central role in both progression and size control of somitogenesis.
Collapse
Affiliation(s)
- Kana Ishimatsu
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Tom W Hiscock
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Zach M Collins
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Dini Wahyu Kartika Sari
- Gene Regulation Research, Nara Institute of Science and Technology, Nara 630-0101, Japan.,Department of Fisheries, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Kenny Lischer
- Gene Regulation Research, Nara Institute of Science and Technology, Nara 630-0101, Japan
| | - David L Richmond
- Image and Data Analysis Core, Harvard Medical School, Boston, MA 02115, USA
| | - Yasumasa Bessho
- Gene Regulation Research, Nara Institute of Science and Technology, Nara 630-0101, Japan
| | - Takaaki Matsui
- Gene Regulation Research, Nara Institute of Science and Technology, Nara 630-0101, Japan
| | - Sean G Megason
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
29
|
Lleras Forero L, Narayanan R, Huitema LF, VanBergen M, Apschner A, Peterson-Maduro J, Logister I, Valentin G, Morelli LG, Oates AC, Schulte-Merker S. Segmentation of the zebrafish axial skeleton relies on notochord sheath cells and not on the segmentation clock. eLife 2018; 7:33843. [PMID: 29624170 PMCID: PMC5962341 DOI: 10.7554/elife.33843] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 04/04/2018] [Indexed: 12/12/2022] Open
Abstract
Segmentation of the axial skeleton in amniotes depends on the segmentation clock, which patterns the paraxial mesoderm and the sclerotome. While the segmentation clock clearly operates in teleosts, the role of the sclerotome in establishing the axial skeleton is unclear. We severely disrupt zebrafish paraxial segmentation, yet observe a largely normal segmentation process of the chordacentra. We demonstrate that axial entpd5+ notochord sheath cells are responsible for chordacentrum mineralization, and serve as a marker for axial segmentation. While autonomous within the notochord sheath, entpd5 expression and centrum formation show some plasticity and can respond to myotome pattern. These observations reveal for the first time the dynamics of notochord segmentation in a teleost, and are consistent with an autonomous patterning mechanism that is influenced, but not determined by adjacent paraxial mesoderm. This behavior is not consistent with a clock-type mechanism in the notochord.
Collapse
Affiliation(s)
- Laura Lleras Forero
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Münster, Germany.,CiM Cluster of Excellence (EXC-1003-CiM), Münster, Germany.,Hubrecht Institute-KNAW & UMC Utrecht, Utrecht, Netherlands
| | | | | | - Maaike VanBergen
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Münster, Germany
| | | | | | - Ive Logister
- Hubrecht Institute-KNAW & UMC Utrecht, Utrecht, Netherlands
| | | | - Luis G Morelli
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA), CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina.,Departamento de Fisica, FCEyN, UBA, Ciudad Universitaria, Buenos Aires, Argentina.,Department of Systemic Cell Biology, Max Planck Institute for Molecular Physiology, Dortmund, Germany
| | - Andrew C Oates
- The Francis Crick Institute, London, United Kingdom.,Department of Cell and Developmental Biology, University College London, London, United Kingdom.,Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Stefan Schulte-Merker
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Münster, Germany
| |
Collapse
|
30
|
Troll JV, Hamilton MK, Abel ML, Ganz J, Bates JM, Stephens WZ, Melancon E, van der Vaart M, Meijer AH, Distel M, Eisen JS, Guillemin K. Microbiota promote secretory cell determination in the intestinal epithelium by modulating host Notch signaling. Development 2018; 145:145/4/dev155317. [PMID: 29475973 DOI: 10.1242/dev.155317] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 01/19/2018] [Indexed: 12/15/2022]
Abstract
Resident microbes promote many aspects of host development, although the mechanisms by which microbiota influence host tissues remain unclear. We showed previously that the microbiota is required for allocation of appropriate numbers of secretory cells in the zebrafish intestinal epithelium. Because Notch signaling is crucial for secretory fate determination, we conducted epistasis experiments to establish whether the microbiota modulates host Notch signaling. We also investigated whether innate immune signaling transduces microbiota cues via the Myd88 adaptor protein. We provide the first evidence that microbiota-induced, Myd88-dependent signaling inhibits host Notch signaling in the intestinal epithelium, thereby promoting secretory cell fate determination. These results connect microbiota activity via innate immune signaling to the Notch pathway, which also plays crucial roles in intestinal homeostasis throughout life and when impaired can result in chronic inflammation and cancer.
Collapse
Affiliation(s)
- Joshua V Troll
- Institute of Molecular Biology, Department of Biology, 1229 University of Oregon, Eugene, OR 97403, USA
| | - M Kristina Hamilton
- Institute of Neuroscience, Department of Biology, 1254 University of Oregon, Eugene, OR 97403, USA
| | - Melissa L Abel
- Institute of Molecular Biology, Department of Biology, 1229 University of Oregon, Eugene, OR 97403, USA
| | - Julia Ganz
- Institute of Neuroscience, Department of Biology, 1254 University of Oregon, Eugene, OR 97403, USA
| | - Jennifer M Bates
- Institute of Molecular Biology, Department of Biology, 1229 University of Oregon, Eugene, OR 97403, USA
| | - W Zac Stephens
- Institute of Molecular Biology, Department of Biology, 1229 University of Oregon, Eugene, OR 97403, USA
| | - Ellie Melancon
- Institute of Neuroscience, Department of Biology, 1254 University of Oregon, Eugene, OR 97403, USA
| | | | - Annemarie H Meijer
- Institute of Biology, Leiden University, 2300 RA Leiden, The Netherlands
| | - Martin Distel
- Children's Cancer Research Institute, 1090 Vienna, Austria
| | - Judith S Eisen
- Institute of Neuroscience, Department of Biology, 1254 University of Oregon, Eugene, OR 97403, USA
| | - Karen Guillemin
- Institute of Molecular Biology, Department of Biology, 1229 University of Oregon, Eugene, OR 97403, USA .,Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, Ontario, Canada M5G 1Z8
| |
Collapse
|
31
|
Trivedi V, Choi HMT, Fraser SE, Pierce NA. Multidimensional quantitative analysis of mRNA expression within intact vertebrate embryos. Development 2018; 145:dev156869. [PMID: 29311262 PMCID: PMC5825878 DOI: 10.1242/dev.156869] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 11/23/2017] [Indexed: 12/29/2022]
Abstract
For decades, in situ hybridization methods have been essential tools for studies of vertebrate development and disease, as they enable qualitative analyses of mRNA expression in an anatomical context. Quantitative mRNA analyses typically sacrifice the anatomy, relying on embryo microdissection, dissociation, cell sorting and/or homogenization. Here, we eliminate the trade-off between quantitation and anatomical context, using quantitative in situ hybridization chain reaction (qHCR) to perform accurate and precise relative quantitation of mRNA expression with subcellular resolution within whole-mount vertebrate embryos. Gene expression can be queried in two directions: read-out from anatomical space to expression space reveals co-expression relationships in selected regions of the specimen; conversely, read-in from multidimensional expression space to anatomical space reveals those anatomical locations in which selected gene co-expression relationships occur. As we demonstrate by examining gene circuits underlying somitogenesis, quantitative read-out and read-in analyses provide the strengths of flow cytometry expression analyses, but by preserving subcellular anatomical context, they enable bi-directional queries that open a new era for in situ hybridization.
Collapse
Affiliation(s)
- Vikas Trivedi
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Translational Imaging Center, University of Southern California, Los Angeles, CA 90089, USA
- Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | - Harry M T Choi
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Scott E Fraser
- Translational Imaging Center, University of Southern California, Los Angeles, CA 90089, USA
- Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Niles A Pierce
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Division of Engineering & Applied Science, California Institute of Technology, Pasadena, CA 91125, USA
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| |
Collapse
|
32
|
Small molecule screen in embryonic zebrafish using modular variations to target segmentation. Nat Commun 2017; 8:1901. [PMID: 29196645 PMCID: PMC5711842 DOI: 10.1038/s41467-017-01469-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 09/19/2017] [Indexed: 01/19/2023] Open
Abstract
Small molecule in vivo phenotypic screening is used to identify drugs or biological activities by directly assessing effects in intact organisms. However, current screening designs may not exploit the full potential of chemical libraries due to false negatives. Here, we demonstrate a modular small molecule screen in embryonic zebrafish that varies concentration, genotype and timing to target segmentation disorders, birth defects that affect the spinal column. By testing each small molecule in multiple interrelated ways, this screen recovers compounds that a standard screening design would have missed, increasing the hit frequency from the chemical library three-fold. We identify molecular pathways and segmentation phenotypes, which we share in an open-access annotated database. These hits provide insight into human vertebral segmentation disorders and myopathies. This modular screening strategy is applicable to other developmental questions and disease models, highlighting the power of relatively small chemical libraries to accelerate gene discovery and disease study.
Collapse
|
33
|
Pnrc2 regulates 3'UTR-mediated decay of segmentation clock-associated transcripts during zebrafish segmentation. Dev Biol 2017. [PMID: 28648842 DOI: 10.1016/j.ydbio.2017.06.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Vertebrate segmentation is controlled by the segmentation clock, a molecular oscillator that regulates gene expression and cycles rapidly. The expression of many genes oscillates during segmentation, including hairy/Enhancer of split-related (her or Hes) genes, which encode transcriptional repressors that auto-inhibit their own expression, and deltaC (dlc), which encodes a Notch ligand. We previously identified the tortuga (tor) locus in a zebrafish forward genetic screen for genes involved in cyclic transcript regulation and showed that cyclic transcripts accumulate post-splicing in tor mutants. Here we show that cyclic mRNA accumulation in tor mutants is due to loss of pnrc2, which encodes a proline-rich nuclear receptor co-activator implicated in mRNA decay. Using an inducible in vivo reporter system to analyze transcript stability, we find that the her1 3'UTR confers Pnrc2-dependent instability to a heterologous transcript. her1 mRNA decay is Dicer-independent and likely employs a Pnrc2-Upf1-containing mRNA decay complex. Surprisingly, despite accumulation of cyclic transcripts in pnrc2-deficient embryos, we find that cyclic protein is expressed normally. Overall, we show that Pnrc2 promotes 3'UTR-mediated decay of developmentally-regulated segmentation clock transcripts and we uncover an additional post-transcriptional regulatory layer that ensures oscillatory protein expression in the absence of cyclic mRNA decay.
Collapse
|
34
|
Liao BK, Oates AC. Delta-Notch signalling in segmentation. ARTHROPOD STRUCTURE & DEVELOPMENT 2017; 46:429-447. [PMID: 27888167 PMCID: PMC5446262 DOI: 10.1016/j.asd.2016.11.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 11/20/2016] [Accepted: 11/21/2016] [Indexed: 06/06/2023]
Abstract
Modular body organization is found widely across multicellular organisms, and some of them form repetitive modular structures via the process of segmentation. It's vastly interesting to understand how these regularly repeated structures are robustly generated from the underlying noise in biomolecular interactions. Recent studies from arthropods reveal similarities in segmentation mechanisms with vertebrates, and raise the possibility that the three phylogenetic clades, annelids, arthropods and chordates, might share homology in this process from a bilaterian ancestor. Here, we discuss vertebrate segmentation with particular emphasis on the role of the Notch intercellular signalling pathway. We introduce vertebrate segmentation and Notch signalling, pointing out historical milestones, then describe existing models for the Notch pathway in the synchronization of noisy neighbouring oscillators, and a new role in the modulation of gene expression wave patterns. We ask what functions Notch signalling may have in arthropod segmentation and explore the relationship between Notch-mediated lateral inhibition and synchronization. Finally, we propose open questions and technical challenges to guide future investigations into Notch signalling in segmentation.
Collapse
Affiliation(s)
- Bo-Kai Liao
- Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, London NW7 1AA, UK
| | - Andrew C Oates
- Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, London NW7 1AA, UK; Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
35
|
Development Aspects of Zebrafish Myotendinous Junction: a Model System for Understanding Muscle Basement Membrane Formation and Failure. CURRENT PATHOBIOLOGY REPORTS 2017. [DOI: 10.1007/s40139-017-0140-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
36
|
Zheng MM, Shao B, Ouyang Q. Identifying network topologies that can generate turing pattern. J Theor Biol 2016; 408:88-96. [DOI: 10.1016/j.jtbi.2016.08.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 07/15/2016] [Accepted: 08/08/2016] [Indexed: 12/29/2022]
|
37
|
Liao BK, Jörg DJ, Oates AC. Faster embryonic segmentation through elevated Delta-Notch signalling. Nat Commun 2016; 7:11861. [PMID: 27302627 PMCID: PMC4912627 DOI: 10.1038/ncomms11861] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 05/06/2016] [Indexed: 12/21/2022] Open
Abstract
An important step in understanding biological rhythms is the control of period. A multicellular, rhythmic patterning system termed the segmentation clock is thought to govern the sequential production of the vertebrate embryo's body segments, the somites. Several genetic loss-of-function conditions, including the Delta-Notch intercellular signalling mutants, result in slower segmentation. Here, we generate DeltaD transgenic zebrafish lines with a range of copy numbers and correspondingly increased signalling levels, and observe faster segmentation. The highest-expressing line shows an altered oscillating gene expression wave pattern and shortened segmentation period, producing embryos with more, shorter body segments. Our results reveal surprising differences in how Notch signalling strength is quantitatively interpreted in different organ systems, and suggest a role for intercellular communication in regulating the output period of the segmentation clock by altering its spatial pattern. Rhythmic patterning governs the formation of somites in vertebrates, but how the period of such rhythms can be changed is unclear. Here, the authors generate a genetic model in zebrafish to increase DeltaD expression, which increases the range of Delta-Notch signalling, causing faster segmentation.
Collapse
Affiliation(s)
- Bo-Kai Liao
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstr. 108, Dresden 01037, Germany.,Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, London NW7 1AA, UK
| | - David J Jörg
- Max Planck Institute for the Physics of Complex Systems, Nöthnitzer Str. 38, Dresden 01187, Germany
| | - Andrew C Oates
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstr. 108, Dresden 01037, Germany.,Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, London NW7 1AA, UK.,Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
38
|
Webb AB, Lengyel IM, Jörg DJ, Valentin G, Jülicher F, Morelli LG, Oates AC. Persistence, period and precision of autonomous cellular oscillators from the zebrafish segmentation clock. eLife 2016; 5. [PMID: 26880542 PMCID: PMC4803185 DOI: 10.7554/elife.08438] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 02/11/2016] [Indexed: 12/11/2022] Open
Abstract
In vertebrate development, the sequential and rhythmic segmentation of the body axis
is regulated by a “segmentation clock”. This clock is comprised of a population of
coordinated oscillating cells that together produce rhythmic gene expression patterns
in the embryo. Whether individual cells autonomously maintain oscillations, or
whether oscillations depend on signals from neighboring cells is unknown. Using a
transgenic zebrafish reporter line for the cyclic transcription factor Her1, we
recorded single tailbud cells in vitro. We demonstrate that individual cells can
behave as autonomous cellular oscillators. We described the observed variability in
cell behavior using a theory of generic oscillators with correlated noise. Single
cells have longer periods and lower precision than the tissue, highlighting the role
of collective processes in the segmentation clock. Our work reveals a population of
cells from the zebrafish segmentation clock that behave as self-sustained, autonomous
oscillators with distinctive noisy dynamics. DOI:http://dx.doi.org/10.7554/eLife.08438.001 The timing and pattern of gene activity in cells can be very important. For example,
precise gene activity patterns in 24-hour circadian clocks help to set daily cycles
of rest and activity in organisms. In such scenarios, cells often communicate with
each other to coordinate the activity of their genes. To fully understand how the
behavior of the population emerges, scientists must first understand the gene
activity patterns in individual cells. Rhythmic gene activity is essential for the spinal column to form in fish and other
vertebrate embryos. A group of cells that switch genes on/off in a coordinated
pattern act like a clock to regulate the timing of the various steps in the process
of backbone formation. However, it is not clear if each cell is able to maintain a
rhythm of gene expression on their own, or whether they rely on messages from
neighboring cells to achieve it. Now, Webb et al. use time-lapse videos of individual cells isolated from the tail of
zebrafish embryos to show that each cell can maintain a pattern of rhythmic activity
in a gene called Her1. In the experiments, individual cells were
removed from zebrafish and placed under a microscope to record and track the activity
of Her1 over time using fluorescent proteins. These experiments show
that each cell is able to maintain a rhythmic pattern of Her1
expression on its own. Webb et al. then compared the Her1 activity patterns in individual
cells with the Her1 patterns present in a larger piece of zebrafish
tissue. The experiments showed that the rhythms in the individual cells are slower
and less precise in their timing than in the tissue. This suggests that groups of
cells must work together to create the synchronized rhythms of gene expression with
the right precision and timing needed for the spinal column to be patterned
correctly. In the future, further experiment with these cells will allow researchers to
investigate the genetic basis of the rhythms in single cells, and find out how
individual cells work together with their neighbors to allow tissues to work
properly. DOI:http://dx.doi.org/10.7554/eLife.08438.002
Collapse
Affiliation(s)
- Alexis B Webb
- MRC-National Institute for Medical Research, London, United Kingdom.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Iván M Lengyel
- Departamento de Física, FCEyN UBA and IFIBA, CONICET, Buenos Aires, Argentina
| | - David J Jörg
- Max Planck Institute for the Physics of Complex Systems, Dresden, Germany
| | - Guillaume Valentin
- MRC-National Institute for Medical Research, London, United Kingdom.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Frank Jülicher
- Max Planck Institute for the Physics of Complex Systems, Dresden, Germany
| | - Luis G Morelli
- Departamento de Física, FCEyN UBA and IFIBA, CONICET, Buenos Aires, Argentina
| | - Andrew C Oates
- MRC-National Institute for Medical Research, London, United Kingdom.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Department of Cell and Developmental Biology, University College London, London, United Kingdom
| |
Collapse
|
39
|
The many roles of Notch signaling during vertebrate somitogenesis. Semin Cell Dev Biol 2016; 49:68-75. [DOI: 10.1016/j.semcdb.2014.11.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 11/23/2014] [Accepted: 11/26/2014] [Indexed: 02/06/2023]
|
40
|
Posterior–anterior gradient of zebrafish hes6 expression in the presomitic mesoderm is established by the combinatorial functions of the downstream enhancer and 3′UTR. Dev Biol 2016; 409:543-54. [DOI: 10.1016/j.ydbio.2015.11.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 11/13/2015] [Accepted: 11/15/2015] [Indexed: 01/09/2023]
|
41
|
Martin BL. Factors that coordinate mesoderm specification from neuromesodermal progenitors with segmentation during vertebrate axial extension. Semin Cell Dev Biol 2016; 49:59-67. [DOI: 10.1016/j.semcdb.2015.11.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 11/25/2015] [Accepted: 11/26/2015] [Indexed: 12/15/2022]
|
42
|
Jenkins RP, Hanisch A, Soza-Ried C, Sahai E, Lewis J. Stochastic Regulation of her1/7 Gene Expression Is the Source of Noise in the Zebrafish Somite Clock Counteracted by Notch Signalling. PLoS Comput Biol 2015; 11:e1004459. [PMID: 26588097 PMCID: PMC4654481 DOI: 10.1371/journal.pcbi.1004459] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 07/09/2015] [Indexed: 12/30/2022] Open
Abstract
The somite segmentation clock is a robust oscillator used to generate regularly-sized segments during early vertebrate embryogenesis. It has been proposed that the clocks of neighbouring cells are synchronised via inter-cellular Notch signalling, in order to overcome the effects of noisy gene expression. When Notch-dependent communication between cells fails, the clocks of individual cells operate erratically and lose synchrony over a period of about 5 to 8 segmentation clock cycles (2-3 hours in the zebrafish). Here, we quantitatively investigate the effects of stochasticity on cell synchrony, using mathematical modelling, to investigate the likely source of such noise. We find that variations in the transcription, translation and degradation rate of key Notch signalling regulators do not explain the in vivo kinetics of desynchronisation. Rather, the analysis predicts that clock desynchronisation, in the absence of Notch signalling, is due to the stochastic dissociation of Her1/7 repressor proteins from the oscillating her1/7 autorepressed target genes. Using in situ hybridisation to visualise sites of active her1 transcription, we measure an average delay of approximately three minutes between the times of activation of the two her1 alleles in a cell. Our model shows that such a delay is sufficient to explain the in vivo rate of clock desynchronisation in Notch pathway mutant embryos and also that Notch-mediated synchronisation is sufficient to overcome this stochastic variation. This suggests that the stochastic nature of repressor/DNA dissociation is the major source of noise in the segmentation clock.
Collapse
Affiliation(s)
- Robert P. Jenkins
- Tumour Cell Biology Laboratory, The Francis Crick Institute Lincoln’s Inn Fields Laboratory, London, United Kingdom
- Vertebrate Development Laboratory, The Francis Crick Institute Lincoln’s Inn Fields Laboratory, London, United Kingdom
| | - Anja Hanisch
- Vertebrate Development Laboratory, The Francis Crick Institute Lincoln’s Inn Fields Laboratory, London, United Kingdom
| | - Cristian Soza-Ried
- Vertebrate Development Laboratory, The Francis Crick Institute Lincoln’s Inn Fields Laboratory, London, United Kingdom
| | - Erik Sahai
- Tumour Cell Biology Laboratory, The Francis Crick Institute Lincoln’s Inn Fields Laboratory, London, United Kingdom
| | - Julian Lewis
- Vertebrate Development Laboratory, The Francis Crick Institute Lincoln’s Inn Fields Laboratory, London, United Kingdom
| |
Collapse
|
43
|
A Local, Self-Organizing Reaction-Diffusion Model Can Explain Somite Patterning in Embryos. Cell Syst 2015; 1:257-69. [PMID: 27136055 DOI: 10.1016/j.cels.2015.10.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 09/11/2015] [Accepted: 10/07/2015] [Indexed: 11/22/2022]
Abstract
During somitogenesis in embryos, a posteriorly moving differentiation front arrests the oscillations of "segmentation clock" genes, leaving behind a frozen, periodic pattern of expression stripes. Both mathematical theories and experimental observations have invoked a "clock and wavefront" model to explain this phenomenon, in which long-range molecular gradients control the movement of the front and therefore the placement of the stripes in the embryo. Here, we develop a fundamentally different model-a progressive oscillatory reaction-diffusion (PORD) system driven by short-range interactions. In this model, posterior movement of the front is a local, emergent phenomenon that, in contrast to the clock and wavefront model, is not controlled by global positional information. The PORD model explains important features of somitogenesis, such as size regulation, that previous reaction-diffusion models could not explain. Moreover, the PORD and clock and wavefront models make different predictions about the results of FGF-inhibition and tissue-cutting experiments, and we demonstrate that the results of these experiments favor the PORD model.
Collapse
|
44
|
Shih NP, François P, Delaune EA, Amacher SL. Dynamics of the slowing segmentation clock reveal alternating two-segment periodicity. Development 2015; 142:1785-93. [PMID: 25968314 DOI: 10.1242/dev.119057] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The formation of reiterated somites along the vertebrate body axis is controlled by the segmentation clock, a molecular oscillator expressed within presomitic mesoderm (PSM) cells. Although PSM cells oscillate autonomously, they coordinate with neighboring cells to generate a sweeping wave of cyclic gene expression through the PSM that has a periodicity equal to that of somite formation. The velocity of each wave slows as it moves anteriorly through the PSM, although the dynamics of clock slowing have not been well characterized. Here, we investigate segmentation clock dynamics in the anterior PSM in developing zebrafish embryos using an in vivo clock reporter, her1:her1-venus. The her1:her1-venus reporter has single-cell resolution, allowing us to follow segmentation clock oscillations in individual cells in real-time. By retrospectively tracking oscillations of future somite boundary cells, we find that clock reporter signal increases in anterior PSM cells and that the periodicity of reporter oscillations slows to about ∼1.5 times the periodicity in posterior PSM cells. This gradual slowing of the clock in the anterior PSM creates peaks of clock expression that are separated at a two-segment periodicity both spatially and temporally, a phenomenon we observe in single cells and in tissue-wide analyses. These results differ from previous predictions that clock oscillations stop or are stabilized in the anterior PSM. Instead, PSM cells oscillate until they incorporate into somites. Our findings suggest that the segmentation clock may signal somite formation using a phase gradient with a two-somite periodicity.
Collapse
Affiliation(s)
- Nathan P Shih
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Paul François
- Department of Physics, McGill University, Montréal, Canada H3A 2T8
| | - Emilie A Delaune
- UMR 5305 CNRS/UCBL, 7 passage du Vercors, Lyon 69367, Cedex 07, France
| | - Sharon L Amacher
- Departments of Molecular Genetics and Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
45
|
Hsu CH, Lin JS, Po Lai K, Li JW, Chan TF, You MS, Tse WKF, Jiang YJ. A new mib allele with a chromosomal deletion covering foxc1a exhibits anterior somite specification defect. Sci Rep 2015; 5:10673. [PMID: 26039894 PMCID: PMC4454137 DOI: 10.1038/srep10673] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 04/23/2015] [Indexed: 12/20/2022] Open
Abstract
mibnn2002, found from an allele screen, showed early segmentation defect and severe cell death phenotypes, which are different from previously known mib mutants. Despite distinct morphological phenotypes, the typical mib molecular phenotypes: her4 down-regulation, neurogenic phenotype and cold sensitive dlc expression pattern, still remained. The linkage analysis also indicated that mibnn2002 is a new mib allele. Failure of specification in anterior 7-10 somites is likely due to lack of foxc1a expression in mibnn2002 homozygotes. Somites and somite markers gradually appeared after 7-10 somite stage, suggesting that foxc1a is only essential for the formation of anterior 7-10 somites. Apoptosis began around 16-somite stage with p53 up-regulation. To find the possible links of mib, foxc1a and apoptosis, transcriptome analysis was employed. About 140 genes, including wnt3a, foxc1a and mib, were not detected in the homozygotes. Overexpression of foxc1a mRNA in mibnn2002 homozygotes partially rescued the anterior somite specification. In the process of characterizing mibnn2002 mutation, we integrated the scaffolds containing mib locus into chromosome 2 (or linkage group 2, LG2) based on synteny comparison and transcriptome results. Genomic PCR analysis further supported the conclusion and showed that mibnn2002 has a chromosomal deletion with the size of about 9.6 Mbp.
Collapse
Affiliation(s)
- Chia-Hao Hsu
- 1] Institute of Molecular and Genomic Medicine, National Health Research Institutes, Taiwan [2] Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Taiwan
| | - Ji-Sheng Lin
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Taiwan
| | - Keng Po Lai
- School of Biological Sciences, The University of Hong Kong, Hong Kong
| | - Jing-Woei Li
- School of Life Sciences, Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong
| | - Ting-Fung Chan
- School of Life Sciences, Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong
| | - May-Su You
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Taiwan
| | | | - Yun-Jin Jiang
- 1] Institute of Molecular and Genomic Medicine, National Health Research Institutes, Taiwan [2] Biotechnology Center, National Chung Hsing University, Taiwan [3] Institute of Molecular and Cellular Biology, National Taiwan University, Taiwan
| |
Collapse
|
46
|
Della Noce I, Carra S, Brusegan C, Critelli R, Frassine A, De Lorenzo C, Giordano A, Bellipanni G, Villa E, Cotelli F, Pistocchi A, Schepis F. The Coiled-Coil Domain Containing 80 (ccdc80) gene regulates gadd45β2 expression in the developing somites of zebrafish as a new player of the hedgehog pathway. J Cell Physiol 2015; 230:821-30. [PMID: 25205658 DOI: 10.1002/jcp.24810] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Accepted: 09/05/2014] [Indexed: 11/10/2022]
Abstract
The Coiled-Coil Domain Containing 80 (CCDC80) gene has been identified as strongly induced in rat thyroid PC CL3 cells immortalized by the adenoviral E1A gene. In human, CCDC80 is a potential oncosoppressor due to its down-regulation in several tumor cell lines and tissues and it is expressed in almost all tissues. CCDC80 has homologous in mouse, chicken, and zebrafish. We cloned the zebrafish ccdc80 and analyzed its expression and function during embryonic development. The in-silico translated zebrafish protein shares high similarity with its mammalian homologous, with nuclear localization signals and a signal peptide. Gene expression analysis demonstrates that zebrafish ccdc80 is maternally and zygotically expressed throughout the development. In particular, ccdc80 is strongly expressed in the notochord and it is under the regulation of the Hedgehog pathway. In this work we investigated the functional effects of ccdc80-loss-of-function during embryonic development and verified its interaction with gadd45β2 in somitogenesis.
Collapse
Affiliation(s)
- Isabella Della Noce
- Department of Gastroenterology, University of Modena and Reggio Emilia, Modena, Italy; Parco Tecnologico Padano, via Einstein, Lodi, Italia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ay A, Holland J, Sperlea A, Devakanmalai GS, Knierer S, Sangervasi S, Stevenson A, Ozbudak EM. Spatial gradients of protein-level time delays set the pace of the traveling segmentation clock waves. Development 2014; 141:4158-67. [PMID: 25336742 DOI: 10.1242/dev.111930] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The vertebrate segmentation clock is a gene expression oscillator controlling rhythmic segmentation of the vertebral column during embryonic development. The period of oscillations becomes longer as cells are displaced along the posterior to anterior axis, which results in traveling waves of clock gene expression sweeping in the unsegmented tissue. Although various hypotheses necessitating the inclusion of additional regulatory genes into the core clock network at different spatial locations have been proposed, the mechanism underlying traveling waves has remained elusive. Here, we combined molecular-level computational modeling and quantitative experimentation to solve this puzzle. Our model predicts the existence of an increasing gradient of gene expression time delays along the posterior to anterior direction to recapitulate spatiotemporal profiles of the traveling segmentation clock waves in different genetic backgrounds in zebrafish. We validated this prediction by measuring an increased time delay of oscillatory Her1 protein production along the unsegmented tissue. Our results refuted the need for spatial expansion of the core feedback loop to explain the occurrence of traveling waves. Spatial regulation of gene expression time delays is a novel way of creating dynamic patterns; this is the first report demonstrating such a control mechanism in any tissue and future investigations will explore the presence of analogous examples in other biological systems.
Collapse
Affiliation(s)
- Ahmet Ay
- Department of Mathematics, Colgate University, Hamilton, NY 13346, USA Department of Biology, Colgate University, Hamilton, NY 13346, USA
| | - Jack Holland
- Department of Computer Science, Colgate University, Hamilton, NY 13346, USA
| | - Adriana Sperlea
- Department of Computer Science, Colgate University, Hamilton, NY 13346, USA
| | | | - Stephan Knierer
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | - Angel Stevenson
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ertuğrul M Ozbudak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
48
|
Rost F, Eugster C, Schröter C, Oates AC, Brusch L. Chevron formation of the zebrafish muscle segments. J Exp Biol 2014; 217:3870-82. [PMID: 25267843 PMCID: PMC4213178 DOI: 10.1242/jeb.102202] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 09/04/2014] [Indexed: 01/16/2023]
Abstract
The muscle segments of fish have a folded shape, termed a chevron, which is thought to be optimal for the undulating body movements of swimming. However, the mechanism shaping the chevron during embryogenesis is not understood. Here, we used time-lapse microscopy of developing zebrafish embryos spanning the entire somitogenesis period to quantify the dynamics of chevron shape development. By comparing such time courses with the start of movements in wildtype zebrafish and analysing immobile mutants, we show that the previously implicated body movements do not play a role in chevron formation. Further, the monotonic increase of chevron angle along the anteroposterior axis revealed by our data constrains or rules out possible contributions by previously proposed mechanisms. In particular, we found that muscle pioneers are not required for chevron formation. We put forward a tension-and-resistance mechanism involving interactions between intra-segmental tension and segment boundaries. To evaluate this mechanism, we derived and analysed a mechanical model of a chain of contractile and resisting elements. The predictions of this model were verified by comparison with experimental data. Altogether, our results support the notion that a simple physical mechanism suffices to self-organize the observed spatiotemporal pattern in chevron formation.
Collapse
Affiliation(s)
- Fabian Rost
- Center for Information Services and High-Performance Computing, Technische Universität Dresden, 01062 Dresden, Germany
| | - Christina Eugster
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Christian Schröter
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Andrew C Oates
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany MRC-National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Lutz Brusch
- Center for Information Services and High-Performance Computing, Technische Universität Dresden, 01062 Dresden, Germany
| |
Collapse
|
49
|
Maragh S, Miller RA, Bessling SL, Wang G, Hook PW, McCallion AS. Rbm24a and Rbm24b are required for normal somitogenesis. PLoS One 2014; 9:e105460. [PMID: 25170925 PMCID: PMC4149414 DOI: 10.1371/journal.pone.0105460] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 07/24/2014] [Indexed: 12/13/2022] Open
Abstract
We recently demonstrated that the gene encoding the RNA binding motif protein 24 (RBM24) is expressed during mouse cardiogenesis, and determined the developmental requirement for its zebrafish homologs Rbm24a and Rbm24b during cardiac development. We demonstrate here that both Rbm24a and Rbm24b are also required for normal somite and craniofacial development. Diminution of rbm24a or rbm24b gene products by morpholino knockdown resulted in significant disruption of somite formation. Detailed in situ hybridization-based analyses of a spectrum of somitogenesis-associated transcripts revealed reduced expression of the cyclic muscle pattering genes dlc and dld encoding Notch ligands, as well as their respective target genes her7, her1. By contrast expression of the Notch receptors notch1a and notch3 appears unchanged. Some RBM-family members have been implicated in pre-mRNA processing. Analysis of affected Notch-pathway mRNAs in rbm24a and rbm24b morpholino-injected embryos revealed aberrant transcript fragments of dlc and dld, but not her1 or her7, suggesting the reduction in transcription levels of Notch pathway components may result from aberrant processing of its ligands. These data imply a previously unknown requirement for Rbm24a and Rbm24b in somite and craniofacial development. Although we anticipate the influence of disrupting RBM24 homologs likely extends beyond the Notch pathway, our results suggest their perturbation may directly, or indirectly, compromise post-transcriptional processing, exemplified by imprecise processing of dlc and dld.
Collapse
Affiliation(s)
- Samantha Maragh
- Biochemical Science Division, National Institute of Standards and Technology, Gaithersburg, Maryland, United States of America
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Ronald A. Miller
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Seneca L. Bessling
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Guangliang Wang
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Paul W. Hook
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Andrew S. McCallion
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
50
|
Soza-Ried C, Öztürk E, Ish-Horowicz D, Lewis J. Pulses of Notch activation synchronise oscillating somite cells and entrain the zebrafish segmentation clock. Development 2014; 141:1780-8. [PMID: 24715465 DOI: 10.1242/dev.102111] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Formation of somites, the rudiments of vertebrate body segments, is an oscillatory process governed by a gene-expression oscillator, the segmentation clock. This operates in each cell of the presomitic mesoderm (PSM), but the individual cells drift out of synchrony when Delta/Notch signalling fails, causing gross anatomical defects. We and others have suggested that this is because synchrony is maintained by pulses of Notch activation, delivered cyclically by each cell to its neighbours, that serve to adjust or reset the phase of the intracellular oscillator. This, however, has never been proved. Here, we provide direct experimental evidence, using zebrafish containing a heat-shock-driven transgene that lets us deliver artificial pulses of expression of the Notch ligand DeltaC. In DeltaC-defective embryos, in which endogenous Notch signalling fails, the artificial pulses restore synchrony, thereby rescuing somite formation. The spacing of segment boundaries produced by repetitive heat-shocking varies according to the time interval between one heat-shock and the next. The induced synchrony is manifest both morphologically and at the level of the oscillations of her1, a core component of the intracellular oscillator. Thus, entrainment of intracellular clocks by periodic activation of the Notch pathway is indeed the mechanism maintaining cell synchrony during somitogenesis.
Collapse
Affiliation(s)
- Cristian Soza-Ried
- Vertebrate Development and Developmental Genetics Laboratories, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | | | | | | |
Collapse
|