1
|
ten Tusscher KH. Computational modeling of plant root development: the art and the science. THE NEW PHYTOLOGIST 2025; 246:2446-2461. [PMID: 40269551 PMCID: PMC12095987 DOI: 10.1111/nph.70164] [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] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 03/25/2025] [Indexed: 04/25/2025]
Abstract
Plant root development, like any developmental process, arises from the interplay between processes like gene expression, cell-cell signaling, cell growth and division, and tissue mechanics, which unfold over a wide range of temporal and spatial scales. Computational models are uniquely suited to integrate these different processes and spatio-temporal scales to investigate how their interplay determines developmental outcomes and have become part of mainstream plant developmental research. Still, for non-modeling experts, it often remains unclear how models are built, why a particular modeling approach was chosen, and how to interpret and value model outcomes. This review attempts to explain the science behind the art of model building, illustrating the simplifications that are often made to keep models simple to understand and when these are and are not justified. Similarly, it discusses when it is safe to ignore certain processes like growth or tissue mechanics and when it is not. Additionally, this review discusses a range of major breakthrough modeling articles. Their approaches are linked to classical concepts and models in developmental biology like the French flag positional information gradient of Lewis Wolpert and the repetitive patterning mechanism proposed by Turing, in addition to highlighting the lessons they taught us on plant root development.
Collapse
Affiliation(s)
- Kirsten H. ten Tusscher
- Experimental and Computational Plant Development, IEB, Department of BiologyUtrecht UniversityWinthontlaan 30C3526 KVUtrechtthe Netherlands
- Theoretical Biology, IBB, Department of BiologyUtrecht UniversityWinthontlaan 30C3526 KVUtrechtthe Netherlands
| |
Collapse
|
2
|
Duboule D, Rekaik H. Comments on the Hox timer and related issues. Cells Dev 2024:203991. [PMID: 39734021 DOI: 10.1016/j.cdev.2024.203991] [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: 11/23/2024] [Revised: 12/26/2024] [Accepted: 12/26/2024] [Indexed: 12/31/2024]
Abstract
2024 not only marked the 100th anniversary of the discovery of the organizer by Hilde Pröscholdt-Mangold and Hans Spemann, but also the 40th anniversary of the discovery of the homeobox, a DNA region encoding a DNA binding peptide present in several transcription factors of critical importance for the gastrulating embryo. In particular, this sequence is found in the 39 members of the amniote Hox gene family, a series of genes activated in mid-gastrulation and involved in organizing morphologies along the extending anterior to posterior (AP) body axis. Over the past 30 years, the study of their coordinated regulation in various contexts has progressively revealed their surprising regulatory strategies, based on mechanisms acting in-cis, which can translate a linear distribution of series of genes along the chromatin fiber into the proper sequences of morphologies observed along our various body axes. The first regulatory layer is controlled by the Hox timer, a mechanism implementing a time-sequenced activation of these genes following their chromosomal order. Here, we discuss various aspects of this mechanism, emphasizing some of its singularities.
Collapse
Affiliation(s)
- Denis Duboule
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France; School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland.
| | - Hocine Rekaik
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France; School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|
3
|
Yadav US, Biswas T, Singh PN, Gupta P, Chakraborty S, Delgado I, Zafar H, Capellini TD, Torres M, Bandyopadhyay A. Molecular mechanism of synovial joint site specification and induction in developing vertebrate limbs. Development 2023; 150:dev201335. [PMID: 37272420 PMCID: PMC10323242 DOI: 10.1242/dev.201335] [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: 09/30/2022] [Accepted: 05/25/2023] [Indexed: 06/06/2023]
Abstract
The vertebrate appendage comprises three primary segments, the stylopod, zeugopod and autopod, each separated by joints. The molecular mechanisms governing the specification of joint sites, which define segment lengths and thereby limb architecture, remain largely unknown. Existing literature suggests that reciprocal gradients of retinoic acid (RA) and fibroblast growth factor (FGF) signaling define the expression domains of the putative segment markers Meis1, Hoxa11 and Hoxa13. Barx1 is expressed in the presumptive joint sites. Our data demonstrate that RA-FGF signaling gradients define the expression domain of Barx1 in the first presumptive joint site. When misexpressed, Barx1 induces ectopic interzone-like structures, and its loss of function partially blocks interzone development. Simultaneous perturbations of RA-FGF signaling gradients result in predictable shifts of Barx1 expression domains along the proximo-distal axis and, consequently, in the formation of repositioned joints. Our data suggest that during early limb bud development in chick, Meis1 and Hoxa11 expression domains are overlapping, whereas the Barx1 expression domain resides within the Hoxa11 expression domain. However, once the interzone is formed, the expression domains are refined and the Barx1 expression domain becomes congruent with the border of these two putative segment markers.
Collapse
Affiliation(s)
- Upendra S. Yadav
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - Tathagata Biswas
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - Pratik N. Singh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - Pankaj Gupta
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - Soura Chakraborty
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - Irene Delgado
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares, CNIC, 28029 Madrid, Spain
| | - Hamim Zafar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
- Department of Computer Science and Engineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - Terence D. Capellini
- Department of Human Evolutionary Biology, Harvard University, 11 Divinity Avenue, Cambridge, MA 02138, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02138, USA
| | - Miguel Torres
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares, CNIC, 28029 Madrid, Spain
| | - Amitabha Bandyopadhyay
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| |
Collapse
|
4
|
Saito S, Mizumoto S, Yonekura T, Yamashita R, Nakano K, Okubo T, Yamada S, Okamura T, Furuichi T. Mice lacking nucleotide sugar transporter SLC35A3 exhibit lethal chondrodysplasia with vertebral anomalies and impaired glycosaminoglycan biosynthesis. PLoS One 2023; 18:e0284292. [PMID: 37053259 PMCID: PMC10101523 DOI: 10.1371/journal.pone.0284292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
SLC35A3 is considered an uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) transporter in mammals and regulates the branching of N-glycans. A missense mutation in SLC35A3 causes complex vertebral malformation (CVM) in cattle. However, the biological functions of SLC35A3 have not been fully clarified. To address these issues, we have established Slc35a3-/-mice using CRISPR/Cas9 genome editing system. The generated mutant mice were perinatal lethal and exhibited chondrodysplasia recapitulating CVM-like vertebral anomalies. During embryogenesis, Slc35a3 mRNA was expressed in the presomitic mesoderm of wild-type mice, suggesting that SLC35A3 transports UDP-GlcNAc used for the sugar modification that is essential for somite formation. In the growth plate cartilage of Slc35a3-/-embryos, extracellular space was drastically reduced, and many flat proliferative chondrocytes were reshaped. Proliferation, apoptosis and differentiation were not affected in the chondrocytes of Slc35a3-/-mice, suggesting that the chondrodysplasia phenotypes were mainly caused by the abnormal extracellular matrix quality. Because these histological abnormalities were similar to those observed in several mutant mice accompanying the impaired glycosaminoglycan (GAG) biosynthesis, GAG levels were measured in the spine and limbs of Slc35a3-/-mice using disaccharide composition analysis. Compared with control mice, the amounts of heparan sulfate, keratan sulfate, and chondroitin sulfate/dermatan sulfate, were significantly decreased in Slc35a3-/-mice. These findings suggest that SLC35A3 regulates GAG biosynthesis and the chondrodysplasia phenotypes were partially caused by the decreased GAG synthesis. Hence, Slc35a3-/- mice would be a useful model for investigating the in vivo roles of SLC35A3 and the pathological mechanisms of SLC35A3-associated diseases.
Collapse
Affiliation(s)
- Soichiro Saito
- Laboratory of Laboratory Animal Science and Medicine, Co-Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, Iwate, Japan
| | - Shuji Mizumoto
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, Nagoya, Aichi, Japan
| | - Tsukasa Yonekura
- Laboratory of Laboratory Animal Science and Medicine, Co-Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, Iwate, Japan
| | - Rina Yamashita
- Laboratory of Laboratory Animal Science and Medicine, Co-Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, Iwate, Japan
| | - Kenta Nakano
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine (NCGM), Shinjuku-ku, Tokyo, Japan
| | - Tadashi Okubo
- Department of Laboratory Animal Science, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Shuhei Yamada
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, Nagoya, Aichi, Japan
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine (NCGM), Shinjuku-ku, Tokyo, Japan
| | - Tatsuya Furuichi
- Laboratory of Laboratory Animal Science and Medicine, Co-Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, Iwate, Japan
- Laboratory of Laboratory Animal Science and Medicine, Graduate School of Veterinary Sciences, Iwate University, Morioka, Iwate, Japan
| |
Collapse
|
5
|
Kent MR, Calderon D, Silvius KM, Kucinski JP, LaVigne CA, Cannon MV, Kendall GC. Zebrafish her3 knockout impacts developmental and cancer-related gene signatures. Dev Biol 2023; 496:1-14. [PMID: 36696714 PMCID: PMC10054701 DOI: 10.1016/j.ydbio.2023.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 01/04/2023] [Accepted: 01/07/2023] [Indexed: 01/24/2023]
Abstract
HES3 is a basic helix-loop-helix transcription factor that regulates neural stem cell renewal during development. HES3 overexpression is predictive of reduced overall survival in patients with fusion-positive rhabdomyosarcoma, a pediatric cancer that resembles immature and undifferentiated skeletal muscle. However, the mechanisms of HES3 cooperation in fusion-positive rhabdomyosarcoma are unclear and are likely related to her3/HES3's role in neurogenesis. To investigate HES3's function during development, we generated a zebrafish CRISPR/Cas9 null mutation of her3, the zebrafish ortholog of HES3. Loss of her3 is not embryonic lethal and adults exhibit expected Mendelian ratios. Embryonic her3 zebrafish mutants exhibit dysregulated neurog1 expression, a her3 target gene, and the mutant her3 fails to bind the neurog1 promoter sequence. Further, her3 mutants are significantly smaller than wildtype and a subset present with lens defects as adults. Transcriptomic analysis of her3 mutant embryos indicates that genes involved in organ development, such as pctp and grinab, are significantly downregulated. Further, differentially expressed genes in her3 null mutant embryos are enriched for Hox and Sox10 motifs. Several cancer-related gene pathways are impacted, including the inhibition of matrix metalloproteinases. Altogether, this new model is a powerful system to study her3/HES3-mediated neural development and its misappropriation in cancer contexts.
Collapse
Affiliation(s)
- Matthew R Kent
- Center for Childhood Cancer & Blood Diseases, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | - Delia Calderon
- Center for Childhood Cancer & Blood Diseases, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, 43205, USA; Molecular, Cellular, and Developmental Biology Ph.D. Program, The Ohio State University, Columbus, OH, 43210, USA
| | - Katherine M Silvius
- Center for Childhood Cancer & Blood Diseases, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | - Jack P Kucinski
- Center for Childhood Cancer & Blood Diseases, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, 43205, USA; Molecular, Cellular, and Developmental Biology Ph.D. Program, The Ohio State University, Columbus, OH, 43210, USA
| | - Collette A LaVigne
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Matthew V Cannon
- Center for Childhood Cancer & Blood Diseases, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | - Genevieve C Kendall
- Center for Childhood Cancer & Blood Diseases, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, 43205, USA; Molecular, Cellular, and Developmental Biology Ph.D. Program, The Ohio State University, Columbus, OH, 43210, USA; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, 43205, USA.
| |
Collapse
|
6
|
Özelçi E, Mailand E, Rüegg M, Oates AC, Sakar MS. Deconstructing body axis morphogenesis in zebrafish embryos using robot-assisted tissue micromanipulation. Nat Commun 2022; 13:7934. [PMID: 36566327 PMCID: PMC9789989 DOI: 10.1038/s41467-022-35632-4] [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: 04/25/2022] [Accepted: 12/14/2022] [Indexed: 12/25/2022] Open
Abstract
Classic microsurgical techniques, such as those used in the early 1900s by Mangold and Spemann, have been instrumental in advancing our understanding of embryonic development. However, these techniques are highly specialized, leading to issues of inter-operator variability. Here we introduce a user-friendly robotic microsurgery platform that allows precise mechanical manipulation of soft tissues in zebrafish embryos. Using our platform, we reproducibly targeted precise regions of tail explants, and quantified the response in real-time by following notochord and presomitic mesoderm (PSM) morphogenesis and segmentation clock dynamics during vertebrate anteroposterior axis elongation. We find an extension force generated through the posterior notochord that is strong enough to buckle the structure. Our data suggest that this force generates a unidirectional notochord extension towards the tailbud because PSM tissue around the posterior notochord does not let it slide anteriorly. These results complement existing biomechanical models of axis elongation, revealing a critical coupling between the posterior notochord, the tailbud, and the PSM, and show that somite patterning is robust against structural perturbations.
Collapse
Affiliation(s)
- Ece Özelçi
- Institute of Mechanical Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
- Institute of Bioengineering, EPFL, 1015, Lausanne, Switzerland
| | - Erik Mailand
- Institute of Mechanical Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Matthias Rüegg
- Institute of Mechanical Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Andrew C Oates
- Institute of Bioengineering, EPFL, 1015, Lausanne, Switzerland.
| | - Mahmut Selman Sakar
- Institute of Mechanical Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland.
- Institute of Bioengineering, EPFL, 1015, Lausanne, Switzerland.
| |
Collapse
|
7
|
Samarasinghe KTG, An E, Genuth MA, Chu L, Holley SA, Crews CM. OligoTRAFTACs: A generalizable method for transcription factor degradation. RSC Chem Biol 2022; 3:1144-1153. [PMID: 36128504 PMCID: PMC9428672 DOI: 10.1039/d2cb00138a] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 07/24/2022] [Indexed: 11/21/2022] Open
Abstract
Targeted transcription factor degradation using oligonucleotide-based transcription factor targeting chimeras (TRAFTACs).
Collapse
Affiliation(s)
- Kusal T. G. Samarasinghe
- Department of Molecular, Cellular & Developmental Biology, Yale University, New Haven, CT, 06511, USA
| | - Elvira An
- Department of Pharmacology, Yale University, New Haven, CT, 06511, USA
| | - Miriam A. Genuth
- Department of Molecular, Cellular & Developmental Biology, Yale University, New Haven, CT, 06511, USA
| | - Ling Chu
- Department of Molecular, Cellular & Developmental Biology, Yale University, New Haven, CT, 06511, USA
| | - Scott A. Holley
- Department of Molecular, Cellular & Developmental Biology, Yale University, New Haven, CT, 06511, USA
| | - Craig M. Crews
- Department of Molecular, Cellular & Developmental Biology, Yale University, New Haven, CT, 06511, USA
- Department of Pharmacology, Yale University, New Haven, CT, 06511, USA
- Department of Chemistry, Yale University, New Haven, CT, 06511, USA
| |
Collapse
|
8
|
Thomson L, Muresan L, Steventon B. The zebrafish presomitic mesoderm elongates through compaction-extension. Cells Dev 2021. [PMID: 34597846 DOI: 10.1101/2021.03.11.434927] [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: 05/07/2023]
Abstract
In vertebrate embryos the presomitic mesoderm becomes progressively segmented into somites at the anterior end while extending along the anterior-posterior axis. A commonly adopted model to explain how this tissue elongates is that of posterior growth, driven in part by the addition of new cells from uncommitted progenitor populations in the tailbud. However, in zebrafish, much of somitogenesis is associated with an absence of overall volume increase, and posterior progenitors do not contribute new cells until the final stages of somitogenesis. Here, we perform a comprehensive 3D morphometric analysis of the paraxial mesoderm and reveal that extension is linked to a volumetric decrease and an increase in cell density. We also find that individual cells decrease in volume over successive somite stages. Live cell tracking confirms that much of this tissue deformation occurs within the presomitic mesoderm progenitor zone and is associated with non-directional rearrangement. Taken together, we propose a compaction-extension mechanism of tissue elongation that highlights the need to better understand the role tissue intrinsic and extrinsic forces in regulating morphogenesis.
Collapse
Affiliation(s)
- Lewis Thomson
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | - Leila Muresan
- Cambridge Advanced Imaging Centre, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
9
|
Kuyyamudi C, Menon SN, Sinha S. Morphogen-regulated contact-mediated signaling between cells can drive the transitions underlying body segmentation in vertebrates. Phys Biol 2021; 19. [PMID: 34670199 DOI: 10.1088/1478-3975/ac31a3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 10/20/2021] [Indexed: 11/12/2022]
Abstract
We propose a unified mechanism that reproduces the sequence of dynamical transitions observed during somitogenesis, the process of body segmentation during embryonic development, that is invariant across all vertebrate species. This is achieved by combining inter-cellular interactions mediated via receptor-ligand coupling with global spatial heterogeneity introduced through a morphogen gradient known to occur along the anteroposterior axis. Our model reproduces synchronized oscillations in the gene expression in cells at the anterior of the presomitic mesoderm as it grows by adding new cells at its posterior, followed by travelling waves and subsequent arrest of activity, with the eventual appearance of somite-like patterns. This framework integrates a boundary-organized pattern formation mechanism, which uses positional information provided by a morphogen gradient, with the coupling-mediated self-organized emergence of collective dynamics, to explain the processes that lead to segmentation.
Collapse
Affiliation(s)
- Chandrashekar Kuyyamudi
- The Institute of Mathematical Sciences, CIT Campus, Taramani, Chennai 600113, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai 400 094, India
| | - Shakti N Menon
- The Institute of Mathematical Sciences, CIT Campus, Taramani, Chennai 600113, India
| | - Sitabhra Sinha
- The Institute of Mathematical Sciences, CIT Campus, Taramani, Chennai 600113, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai 400 094, India
| |
Collapse
|
10
|
The zebrafish presomitic mesoderm elongates through compaction-extension. Cells Dev 2021; 168:203748. [PMID: 34597846 PMCID: PMC7612712 DOI: 10.1016/j.cdev.2021.203748] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 09/21/2021] [Accepted: 09/21/2021] [Indexed: 11/21/2022]
Abstract
In vertebrate embryos the presomitic mesoderm becomes progressively segmented into somites at the anterior end while extending along the anterior-posterior axis. A commonly adopted model to explain how this tissue elongates is that of posterior growth, driven in part by the addition of new cells from uncommitted progenitor populations in the tailbud. However, in zebrafish, much of somitogenesis is associated with an absence of overall volume increase, and posterior progenitors do not contribute new cells until the final stages of somitogenesis. Here, we perform a comprehensive 3D morphometric analysis of the paraxial mesoderm and reveal that extension is linked to a volumetric decrease and an increase in cell density. We also find that individual cells decrease in volume over successive somite stages. Live cell tracking confirms that much of this tissue deformation occurs within the presomitic mesoderm progenitor zone and is associated with non-directional rearrangement. Taken together, we propose a compaction-extension mechanism of tissue elongation that highlights the need to better understand the role tissue intrinsic and extrinsic forces in regulating morphogenesis.
Collapse
|
11
|
Abstract
Arthropod segmentation and vertebrate somitogenesis are leading fields in the experimental and theoretical interrogation of developmental patterning. However, despite the sophistication of current research, basic conceptual issues remain unresolved. These include: (i) the mechanistic origins of spatial organization within the segment addition zone (SAZ); (ii) the mechanistic origins of segment polarization; (iii) the mechanistic origins of axial variation; and (iv) the evolutionary origins of simultaneous patterning. Here, I explore these problems using coarse-grained models of cross-regulating dynamical processes. In the morphogenetic framework of a row of cells undergoing axial elongation, I simulate interactions between an 'oscillator', a 'switch' and up to three 'timers', successfully reproducing essential patterning behaviours of segmenting systems. By comparing the output of these largely cell-autonomous models to variants that incorporate positional information, I find that scaling relationships, wave patterns and patterning dynamics all depend on whether the SAZ is regulated by temporal or spatial information. I also identify three mechanisms for polarizing oscillator output, all of which functionally implicate the oscillator frequency profile. Finally, I demonstrate significant dynamical and regulatory continuity between sequential and simultaneous modes of segmentation. I discuss these results in the context of the experimental literature.
Collapse
Affiliation(s)
- Erik Clark
- Department of Systems Biology, Harvard Medical School, 210 Longwood Ave, Boston, MA 02115, USA
- Trinity College Cambridge, University of Cambridge, Trinity Street, Cambridge CB2 1TQ, UK
| |
Collapse
|
12
|
A "Numerical Evo-Devo" Synthesis for the Identification of Pattern-Forming Factors. Cells 2020; 9:cells9081840. [PMID: 32764501 PMCID: PMC7463486 DOI: 10.3390/cells9081840] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 12/22/2022] Open
Abstract
Animals display extensive diversity in motifs adorning their coat, yet these patterns have reproducible orientation and periodicity within species or groups. Morphological variation has been traditionally used to dissect the genetic basis of evolutionary change, while pattern conservation and stability in both mathematical and organismal models has served to identify core developmental events. Two patterning theories, namely instruction and self-organisation, emerged from this work. Combined, they provide an appealing explanation for how natural patterns form and evolve, but in vivo factors underlying these mechanisms remain elusive. By bridging developmental biology and mathematics, novel frameworks recently allowed breakthroughs in our understanding of pattern establishment, unveiling how patterning strategies combine in space and time, or the importance of tissue morphogenesis in generating positional information. Adding results from surveys of natural variation to these empirical-modelling dialogues improves model inference, analysis, and in vivo testing. In this evo-devo-numerical synthesis, mathematical models have to reproduce not only given stable patterns but also the dynamics of their emergence, and the extent of inter-species variation in these dynamics through minimal parameter change. This integrative approach can help in disentangling molecular, cellular and mechanical interaction during pattern establishment.
Collapse
|
13
|
Grall E, Tschopp P. A sense of place, many times over ‐ pattern formation and evolution of repetitive morphological structures. Dev Dyn 2019; 249:313-327. [DOI: 10.1002/dvdy.131] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/30/2019] [Accepted: 11/04/2019] [Indexed: 12/14/2022] Open
|
14
|
Mongera A, Michaut A, Guillot C, Xiong F, Pourquié O. Mechanics of Anteroposterior Axis Formation in Vertebrates. Annu Rev Cell Dev Biol 2019; 35:259-283. [PMID: 31412208 PMCID: PMC7394480 DOI: 10.1146/annurev-cellbio-100818-125436] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The vertebrate anteroposterior axis forms through elongation of multiple tissues during embryogenesis. This process is based on tissue-autonomous mechanisms of force generation and intertissue mechanical coupling whose failure leads to severe developmental anomalies such as body truncation and spina bifida. Similar to other morphogenetic modules, anteroposterior body extension requires both the rearrangement of existing materials-such as cells and extracellular matrix-and the local addition of new materials, i.e., anisotropic growth, through cell proliferation, cell growth, and matrix deposition. Numerous signaling pathways coordinate body axis formation via regulation of cell behavior during tissue rearrangements and/or volumetric growth. From a physical perspective, morphogenesis depends on both cell-generated forces and tissue material properties. As the spatiotemporal variation of these mechanical parameters has recently been explored in the context of vertebrate body elongation, the study of this process is likely to shed light on the cross talk between signaling and mechanics during morphogenesis.
Collapse
Affiliation(s)
- Alessandro Mongera
- Department of Genetics, Harvard Medical School, and Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA;
| | - Arthur Michaut
- Department of Genetics, Harvard Medical School, and Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA;
| | - Charlène Guillot
- Department of Genetics, Harvard Medical School, and Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA;
| | - Fengzhu Xiong
- Department of Genetics, Harvard Medical School, and Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA;
| | - Olivier Pourquié
- Department of Genetics, Harvard Medical School, and Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA;
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts 02138, USA
| |
Collapse
|
15
|
Marchini M, Silva Hernandez E, Rolian C. Morphology and development of a novel murine skeletal dysplasia. PeerJ 2019; 7:e7180. [PMID: 31308998 PMCID: PMC6612423 DOI: 10.7717/peerj.7180] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 05/23/2019] [Indexed: 12/11/2022] Open
Abstract
Background Limb bones develop and grow by endochondral ossification, which is regulated by specific cell and molecular pathways. Changes in one or more of these pathways can have severe effects on normal skeletal development, leading to skeletal dysplasias. Many skeletal dysplasias are known to result from mis-expression of major genes involved in skeletal development, but the etiology of many skeletal dysplasias remains unknown. We investigated the morphology and development of a mouse line with an uncharacterized mutation exhibiting a skeletal dysplasia-like phenotype (Nabo). Methods We used µCT scanning and histology to comprehensively characterize the phenotype and its development, and to determine the developmental stage when this phenotype first appears. Results Nabo mice have shorter limb elements compared to wildtype mice, while clavicles and dermal bones of the skull are not affected. Nabo embryos at embryonic stage E14 show shorter limb cartilage condensations. The tibial growth plate in Nabo mice is wider than in wildtype, particularly in the proliferative zone, however proliferative chondrocytes show less activity than wildtype mice. Cell proliferation assays and immunohistochemistry against the chondrogenic marker Sox9 suggest relatively lower, spatially-restricted, chondrocyte proliferation activity in Nabo. Bone volume and trabecular thickness in Nabo tibiae are also decreased compared to wildtype. Discussion Our data suggest that the Nabo mutation affects endochondral ossification only, with the strongest effects manifesting in more proximal limb structures. The phenotype appears before embryonic stage E14, suggesting that outgrowth and patterning processes may be affected. Nabo mice present a combination of skeletal dysplasia-like characteristics not present in any known skeletal dysplasia. Further genomic and molecular analysis will help to identify the genetic basis and precise developmental pathways involved in this unique skeletal dysplasia.
Collapse
Affiliation(s)
- Marta Marchini
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Canada.,McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Canada
| | - Elizabeth Silva Hernandez
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| | - Campbell Rolian
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Canada.,Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada
| |
Collapse
|
16
|
Kho M, Shi H, Nie S. Cdc42 Effector Protein 3 Interacts With Cdc42 in Regulating Xenopus Somite Segmentation. Front Physiol 2019; 10:542. [PMID: 31133876 PMCID: PMC6514426 DOI: 10.3389/fphys.2019.00542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 04/17/2019] [Indexed: 11/15/2022] Open
Abstract
Somitogenesis is a critical process during vertebrate development that establishes the segmented body plan and gives rise to the vertebra, skeletal muscles, and dermis. While segmentation clock and wave front mechanisms have been elucidated to control the size and time of somite formation, regulation of the segmentation process that physically separates somites is not understood in detail. Here, we identified a cytoskeletal player, Cdc42 effector protein 3 (Cdc42ep3, CEP3) that is required for somite segmentation in Xenopus embryos. CEP3 is specifically expressed in somite tissue during somite segmentation. Loss-of-function experiments showed that CEP3 is not required for the specification of paraxial mesoderm, nor the differentiation of muscle cells, but is required for the segmentation process. Live imaging analysis further revealed that CEP3 is required for cell shape changes and alignment during somitogenesis. When CEP3 was knocked down, somitic cells did not elongate efficiently along the mediolateral axis and failed to undertake the 90° rotation. As a result, cells remained in a continuous sheet without an apparent segmentation cleft. CEP3 likely interacts with Cdc42 during this process, and both increased and decreased Cdc42 activity led to defective somite segmentation. Segmentation defects caused by Cdc42 knockdown can be partially rescued by the overexpression of CEP3. Conversely, loss of CEP3 resulted in the maintenance of high levels of Cdc42 activity at the cell membrane, which is normally reduced during and after somite segmentation. These results suggest that there is a feedback regulation between Cdc42 and CEP3 during somite segmentation and the activity of Cdc42 needs to be fine-tuned to control the coordinated cell shape changes and movement required for somite segmentation.
Collapse
Affiliation(s)
- Mary Kho
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States
| | - Hongyu Shi
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States
| | - Shuyi Nie
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States.,Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States.,Integrated Cancer Research Center, Georgia Institute of Technology, Atlanta, GA, United States
| |
Collapse
|
17
|
Shukrun N, Shabtai Y, Pillemer G, Fainsod A. Retinoic acid signaling reduction recapitulates the effects of alcohol on embryo size. Genesis 2019; 57:e23284. [PMID: 30672660 DOI: 10.1002/dvg.23284] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/13/2019] [Accepted: 01/21/2019] [Indexed: 12/16/2022]
Abstract
Intrauterine growth restriction (IUGR) is commonly observed in human pregnancies and can result in severe clinical outcomes. IUGR is observed in Fetal Alcohol Syndrome (FAS) fetuses as a result of alcohol (ethanol) exposure during pregnancy. To further understand FAS, the severe form of Fetal Alcohol Spectrum Disorder, we performed an extensive quantitative analysis of the effects of ethanol on embryo size utilizing our Xenopus model. Ethanol-treated embryos exhibited size reduction along the anterior-posterior axis. This effect was evident primarily from the hindbrain caudally, while rostral regions appeared refractive to ethanol-induced size changes, also known as asymmetric IUGR. Interestingly, some embryo batches in addition to shortening from the hindbrain caudally also exhibited an alcohol-dependent reduction of the anterior head domain, known as symmetric IUGR. To study the connection between ethanol exposure and reduced retinoic acid levels we treated embryos with the retinaldehyde dehydrogenase inhibitors, DEAB and citral. Inhibition of retinoic acid biosynthesis recapitulated the growth defects induced by ethanol affecting mainly axial elongation from the hindbrain caudally. To study the competition between ethanol clearance and retinoic acid biosynthesis we demonstrated that, co-exposure to alcohol reduces the teratogenic effects of treatment with retinol (vitamin A), the retinoic acid precursor. These results further support the role of retinoic acid in the regulation of axial elongation.
Collapse
Affiliation(s)
- Natalie Shukrun
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yehuda Shabtai
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Graciela Pillemer
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Abraham Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
18
|
Peli1b governs the brain patterning via ERK signaling pathways in zebrafish embryos. Gene 2019; 694:1-6. [PMID: 30716445 DOI: 10.1016/j.gene.2018.12.078] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/18/2018] [Accepted: 12/30/2018] [Indexed: 12/21/2022]
Abstract
Pellino proteins are associated with immune and stress responses through their effects on NF-κB signaling and B-cell development, and through their role as a scaffold in TLR/IL-1R signaling pathways. However, their function during embryonic development is unclear. Here, we report the developmental expression patterns and functions of peli1b, which encodes a zebrafish ortholog of human Pellino1. Maternal peli1b transcripts were present in zebrafish embryos at the 1-cell stage and zygotic transcripts appeared in the shield area at 6 hours post fertilization (hpf), particularly in the neural plate of the dorsal region. peli1b transcripts were concentrated in the somites, lens, myogenic cells, lateral plate mesoderm, and presomitic mesoderm at 12 hpf, but expression shifted to the telencephalon, diencephalon, hindbrain, and rhombomeres (r1-7) at 24 hpf. Distribution of peli1b transcripts was further restricted to the telencephalon, diencephalon, hindbrain, eyes, and pectoral fins at 48 hpf. Knock-down of peli1b with a peli1b antisense morpholino resulted in significant developmental defects and a reduction in size of the telencephalon, diencephalon, rhombomeres (r1-7), and spinal cord at 24 hpf. When peli1b-knock-down embryos were analyzed for zic3, a marker associated with the central nervous system, we found lower levels of zic3 transcripts in the shield area at 6 hpf and in the posterior diencephalon, dorsal neural plate, midbrain, and hindbrain at 14 hpf. Finally, the ERK3/4 inhibitor SB203580 also induced a significant reduction in the level of zic3 transcripts in the neural plate at 6 hpf and in the posterior diencephalon, dorsal neural plate, midbrain, hindbrain, segmental plate, dorsal spinal cord, and dorsal posterior neural plate at 14 hpf. It is thus likely that the association between Peli1b and brain development in zebrafish embryos occurs via ERK3/4 pathways.
Collapse
|
19
|
Geminin Orchestrates Somite Formation by Regulating Fgf8 and Notch Signaling. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6543196. [PMID: 29984243 PMCID: PMC6011172 DOI: 10.1155/2018/6543196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/24/2018] [Accepted: 05/08/2018] [Indexed: 11/20/2022]
Abstract
During somitogenesis, Fgf8 maintains the predifferentiation stage of presomitic mesoderm (PSM) cells and its retraction gives a cue for somite formation. Delta/Notch initiates the expression of oscillation genes in the tail bud and subsequently contributes to somite formation in a periodic way. Whether there exists a critical factor coordinating Fgf8 and Notch signaling pathways is largely unknown. Here, we demonstrate that the loss of function of geminin gave rise to narrower somites as a result of derepressed Fgf8 gradient in the PSM and tail bud. Furthermore, in geminin morphants, the somite boundary could not form properly but the oscillation of cyclic genes was normal, displaying the blurry somitic boundary and disturbed somite polarity along the AP axis. In mechanism, these manifestations were mediated by the disrupted association of the geminin/Brg1 complex with intron 3 of mib1. The latter interaction was found to positively regulate mib1 transcription, Notch activity, and sequential somite segmentation during somitogenesis. In addition, geminin was also shown to regulate the expression of deltaD in mib1-independent way. Collectively, our data for the first time demonstrate that geminin regulates Fgf8 and Notch signaling to regulate somite segmentation during somitogenesis.
Collapse
|
20
|
Double abdomen in a short-germ insect: Zygotic control of axis formation revealed in the beetle Tribolium castaneum. Proc Natl Acad Sci U S A 2018; 115:1819-1824. [PMID: 29432152 PMCID: PMC5828605 DOI: 10.1073/pnas.1716512115] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
One of the first crucial steps of animal development is to distinguish the anterior versus the posterior pole of the embryo, i.e., the AP axis. If this process fails, embryos may develop two mirror image tails or heads. In the fly Drosophila, the mother provides the signals required for AP axis formation, while in vertebrates, gene activity of the embryo is required as well. We identified two genes whose knockdown leads to double-tail phenotypes in the beetle Tribolium, representing the insect-typical short-germ embryogenesis. Intriguingly, embryo polarity depends on zygotic gene activities and Wnt signaling. Hence, short-germ insect axis formation is more similar to vertebrates than the mechanism employed by Drosophila. The distinction of anterior versus posterior is a crucial first step in animal embryogenesis. In the fly Drosophila, this axis is established by morphogenetic gradients contributed by the mother that regulate zygotic target genes. This principle has been considered to hold true for insects in general but is fundamentally different from vertebrates, where zygotic genes and Wnt signaling are required. We investigated symmetry breaking in the beetle Tribolium castaneum, which among insects represents the more ancestral short-germ embryogenesis. We found that maternal Tc-germ cell-less is required for anterior localization of maternal Tc-axin, which represses Wnt signaling and promotes expression of anterior zygotic genes. Both RNAi targeting Tc-germ cell-less or double RNAi knocking down the zygotic genes Tc-homeobrain and Tc-zen1 led to the formation of a second growth zone at the anterior, which resulted in double-abdomen phenotypes. Conversely, interfering with two posterior factors, Tc-caudal and Wnt, caused double-anterior phenotypes. These findings reveal that maternal and zygotic mechanisms, including Wnt signaling, are required for establishing embryo polarity and induce the segmentation clock in a short-germ insect.
Collapse
|
21
|
Abashev TM, Metzler MA, Wright DM, Sandell LL. Retinoic acid signaling regulates Krt5 and Krt14 independently of stem cell markers in submandibular salivary gland epithelium. Dev Dyn 2018; 246:135-147. [PMID: 27884045 DOI: 10.1002/dvdy.24476] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Revised: 11/15/2016] [Accepted: 11/16/2016] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Retinoic acid (RA), the active metabolite of vitamin A, has been demonstrated to be important for growth and branching morphogenesis of mammalian embryonic salivary gland epithelium. However, it is not known whether RA functions directly within epithelial cells or in associated tissues that influence morphogenesis of salivary epithelium. Moreover, downstream targets of RA regulation have not been identified. RESULTS Here, we show that canonical RA signaling occurs in multiple tissues of embryonic mouse salivary glands, including epithelium, associated parasympathetic ganglion neurons, and nonneuronal mesenchyme. By culturing epithelium explants in isolation from other tissues, we demonstrate that RA influences epithelium morphogenesis by direct action in that tissue. Moreover, we demonstrate that inhibition of RA signaling represses cell proliferation and expression of FGF10 signaling targets, and upregulates expression of basal epithelial keratins Krt5 and Krt14. Importantly, we show that the stem cell gene Kit is regulated inversely from Krt5/Krt14 by RA signaling. CONCLUSIONS RA regulates Krt5 and Krt14 expression independently of stem cell character in developing salivary epithelium. RA, or chemical inhibitors of RA signaling, could potentially be used for modulating growth and differentiation of epithelial stem cells for the purpose of re-populating damaged glands or generating bioengineered organs. Developmental Dynamics 246:135-147, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Timur M Abashev
- University of Louisville, School of Dentistry, Department of Molecular, Cellular and Craniofacial Biology, Louisville, Kentucky
| | - Melissa A Metzler
- University of Louisville, School of Dentistry, Department of Molecular, Cellular and Craniofacial Biology, Louisville, Kentucky
| | - Diana M Wright
- University of Louisville, School of Dentistry, Department of Molecular, Cellular and Craniofacial Biology, Louisville, Kentucky
| | - Lisa L Sandell
- University of Louisville, School of Dentistry, Department of Molecular, Cellular and Craniofacial Biology, Louisville, Kentucky
| |
Collapse
|
22
|
Abstract
Our understanding of satellite cells, now known to be the obligate stem cells of skeletal muscle, has increased dramatically in recent years due to the introduction of new molecular, genetic, and technical resources. In addition to their role in acute repair of damaged muscle, satellite cells are of interest in the fields of aging, exercise, neuromuscular disease, and stem cell therapy, and all of these applications have driven a dramatic increase in our understanding of the activity and potential of satellite cells. However, many fundamental questions of satellite cell biology remain to be answered, including their emergence as a specific lineage, the degree and significance of heterogeneity within the satellite cell population, the roles of their interactions with other resident and infiltrating cell types during homeostasis and regeneration, and the relative roles of intrinsic vs extrinsic factors that may contribute to satellite cell dysfunction in the context of aging or disease. This review will address the current state of these open questions in satellite cell biology.
Collapse
Affiliation(s)
- Ddw Cornelison
- University of Missouri, Columbia, MO, United States; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States.
| |
Collapse
|
23
|
Brinegar AE, Xia Z, Loehr JA, Li W, Rodney GG, Cooper TA. Extensive alternative splicing transitions during postnatal skeletal muscle development are required for calcium handling functions. eLife 2017; 6:27192. [PMID: 28826478 PMCID: PMC5577920 DOI: 10.7554/elife.27192] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 08/04/2017] [Indexed: 01/08/2023] Open
Abstract
Postnatal development of skeletal muscle is a highly dynamic period of tissue remodeling. Here, we used RNA-seq to identify transcriptome changes from late embryonic to adult mouse muscle and demonstrate that alternative splicing developmental transitions impact muscle physiology. The first 2 weeks after birth are particularly dynamic for differential gene expression and alternative splicing transitions, and calcium-handling functions are significantly enriched among genes that undergo alternative splicing. We focused on the postnatal splicing transitions of the three calcineurin A genes, calcium-dependent phosphatases that regulate multiple aspects of muscle biology. Redirected splicing of calcineurin A to the fetal isoforms in adult muscle and in differentiated C2C12 slows the timing of muscle relaxation, promotes nuclear localization of calcineurin target Nfatc3, and/or affects expression of Nfatc transcription targets. The results demonstrate a previously unknown specificity of calcineurin isoforms as well as the broader impact of alternative splicing during muscle postnatal development.
Collapse
Affiliation(s)
- Amy E Brinegar
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, United States.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, United States
| | - Zheng Xia
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, United States.,Division of Biostatistics, Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, United States
| | - James Anthony Loehr
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, United States
| | - Wei Li
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, United States.,Division of Biostatistics, Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, United States
| | - George Gerald Rodney
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, United States
| | - Thomas A Cooper
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, United States.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, United States.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, United States
| |
Collapse
|
24
|
Laganà AS, Vitale SG, Salmeri FM, Triolo O, Ban Frangež H, Vrtačnik-Bokal E, Stojanovska L, Apostolopoulos V, Granese R, Sofo V. Unus pro omnibus, omnes pro uno: A novel, evidence-based, unifying theory for the pathogenesis of endometriosis. Med Hypotheses 2017; 103:10-20. [DOI: 10.1016/j.mehy.2017.03.032] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Accepted: 03/21/2017] [Indexed: 01/17/2023]
|
25
|
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
|
26
|
Sudheer S, Liu J, Marks M, Koch F, Anurin A, Scholze M, Senft AD, Wittler L, Macura K, Grote P, Herrmann BG. Different Concentrations of FGF Ligands, FGF2 or FGF8 Determine Distinct States of WNT-Induced Presomitic Mesoderm. Stem Cells 2016; 34:1790-800. [DOI: 10.1002/stem.2371] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 02/07/2016] [Accepted: 03/03/2016] [Indexed: 12/25/2022]
Affiliation(s)
- Smita Sudheer
- Department of Developmental Genetics; Max Planck Institute for Molecular Genetics; Berlin Germany
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, The University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU; United Kingdom
| | - Jinhua Liu
- Department of Developmental Genetics; Max Planck Institute for Molecular Genetics; Berlin Germany
| | - Matthias Marks
- Department of Developmental Genetics; Max Planck Institute for Molecular Genetics; Berlin Germany
| | - Frederic Koch
- Department of Developmental Genetics; Max Planck Institute for Molecular Genetics; Berlin Germany
| | - Anna Anurin
- Department of Developmental Genetics; Max Planck Institute for Molecular Genetics; Berlin Germany
- Department of Biology; Chemistry and Pharmacy, Free University Berlin; Berlin Germany
| | - Manuela Scholze
- Department of Developmental Genetics; Max Planck Institute for Molecular Genetics; Berlin Germany
| | - Anna Dorothea Senft
- Department of Developmental Genetics; Max Planck Institute for Molecular Genetics; Berlin Germany
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE; United Kingdom
| | - Lars Wittler
- Department of Developmental Genetics; Max Planck Institute for Molecular Genetics; Berlin Germany
| | - Karol Macura
- Department of Developmental Genetics; Max Planck Institute for Molecular Genetics; Berlin Germany
| | - Phillip Grote
- Department of Developmental Genetics; Max Planck Institute for Molecular Genetics; Berlin Germany
- Institute of Cardiovascular Regeneration, Center for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main; Germany
| | - Bernhard G. Herrmann
- Department of Developmental Genetics; Max Planck Institute for Molecular Genetics; Berlin Germany
| |
Collapse
|
27
|
Kalcheim C. Epithelial-Mesenchymal Transitions during Neural Crest and Somite Development. J Clin Med 2015; 5:jcm5010001. [PMID: 26712793 PMCID: PMC4730126 DOI: 10.3390/jcm5010001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 12/09/2015] [Accepted: 12/14/2015] [Indexed: 01/14/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a central process during embryonic development that affects selected progenitor cells of all three germ layers. In addition to driving the onset of cellular migrations and subsequent tissue morphogenesis, the dynamic conversions of epithelium into mesenchyme and vice-versa are intimately associated with the segregation of homogeneous precursors into distinct fates. The neural crest and somites, progenitors of the peripheral nervous system and of skeletal tissues, respectively, beautifully illustrate the significance of EMT to the above processes. Ongoing studies progressively elucidate the gene networks underlying EMT in each system, highlighting the similarities and differences between them. Knowledge of the mechanistic logic of this normal ontogenetic process should provide important insights to the understanding of pathological conditions such as cancer metastasis, which shares some common molecular themes.
Collapse
Affiliation(s)
- Chaya Kalcheim
- Edmond and Lili Safra Center for Brain Sciences (ELSC), Department of Medical Neurobiology, Institute for Medical Research Israel-Canada (IMRIC), Hebrew University of Jerusalem-Hadassah Medical School, P.O. Box 12272, Jerusalem 9112102, Israel.
| |
Collapse
|
28
|
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
|
29
|
Tijchon E, van Ingen Schenau D, van Opzeeland F, Tirone F, Hoogerbrugge PM, Van Leeuwen FN, Scheijen B. Targeted Deletion of Btg1 and Btg2 Results in Homeotic Transformation of the Axial Skeleton. PLoS One 2015. [PMID: 26218146 PMCID: PMC4517811 DOI: 10.1371/journal.pone.0131481] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Btg1 and Btg2 encode highly homologous proteins that are broadly expressed in different cell lineages, and have been implicated in different types of cancer. Btg1 and Btg2 have been shown to modulate the function of different transcriptional regulators, including Hox and Smad transcription factors. In this study, we examined the in vivo role of the mouse Btg1 and Btg2 genes in specifying the regional identity of the axial skeleton. Therefore, we examined the phenotype of Btg1 and Btg2 single knockout mice, as well as novel generated Btg1-/-;Btg2-/- double knockout mice, which were viable, but displayed a non-mendelian inheritance and smaller litter size. We observed both unique and overlapping phenotypes reminiscent of homeotic transformation along the anterior-posterior axis in the single and combined Btg1 and Btg2 knockout animals. Both Btg1-/- and Btg2-/- mice displayed partial posterior transformation of the seventh cervical vertebra, which was more pronounced in Btg1-/-;Btg2-/- mice, demonstrating that Btg1 and Btg2 act in synergy. Loss of Btg2, but not Btg1, was sufficient for complete posterior transformation of the thirteenth thoracic vertebra to the first lumbar vertebra. Moreover, Btg2-/- animals displayed complete posterior transformation of the sixth lumbar vertebra to the first sacral vertebra, which was only partially present at a low frequency in Btg1-/- mice. The Btg1-/-;Btg2-/- animals showed an even stronger phenotype, with L5 to S1 transformation. Together, these data show that both Btg1 and Btg2 are required for normal vertebral patterning of the axial skeleton, but each gene contributes differently in specifying the identity along the anterior-posterior axis of the skeleton.
Collapse
Affiliation(s)
- Esther Tijchon
- Laboratory of Pediatric Oncology, Radboud university medical center, Nijmegen, The Netherlands
| | | | - Fred van Opzeeland
- Laboratory of Pediatric Infectious Diseases, Radboud university medical center, Nijmegen, The Netherlands
| | - Felice Tirone
- Institute of Cell Biology and Neurobiology, National Research Council, Fondazione Santa Lucia 00143, Rome, Italy
| | | | - Frank N. Van Leeuwen
- Laboratory of Pediatric Oncology, Radboud university medical center, Nijmegen, The Netherlands
| | - Blanca Scheijen
- Laboratory of Pediatric Oncology, Radboud university medical center, Nijmegen, The Netherlands
- * E-mail:
| |
Collapse
|
30
|
Ruf-Zamojski F, Trivedi V, Fraser SE, Trinh LA. Spatio-Temporal Differences in Dystrophin Dynamics at mRNA and Protein Levels Revealed by a Novel FlipTrap Line. PLoS One 2015; 10:e0128944. [PMID: 26083378 PMCID: PMC4471274 DOI: 10.1371/journal.pone.0128944] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 05/01/2015] [Indexed: 11/18/2022] Open
Abstract
Dystrophin (Dmd) is a structural protein that links the extracellular matrix to actin filaments in muscle fibers and is required for the maintenance of muscles integrity. Mutations in Dmd lead to muscular dystrophies in humans and other vertebrates. Here, we report the characterization of a zebrafish gene trap line that fluorescently labels the endogenous Dmd protein (Dmd-citrine, Gt(dmd-citrine) ct90a). We show that the Dmd-citrine line recapitulates endogenous dmd transcript expression and Dmd protein localization. Using this Dmd-citrine line, we follow Dmd localization to the myosepta in real-time using time-lapse microscopy, and find that the accumulation of Dmd protein at the transverse myosepta coincides with the onset of myotome formation, a critical stage in muscle maturation. We observed that Dmd protein localizes specifically to the myosepta prior to dmd mRNA localization. Additionally, we demonstrate that the Dmd-citrine line can be used to assess muscular dystrophy following both genetic and physical disruptions of the muscle.
Collapse
Affiliation(s)
- Frederique Ruf-Zamojski
- California Institute of Technology, Biological Imaging Center, Beckman Institute, Division of Biology, Pasadena, California 91125, United States of America
| | - Vikas Trivedi
- California Institute of Technology, Department of Bioengineering, Pasadena, California 91125, United States of America
| | - Scott E. Fraser
- California Institute of Technology, Biological Imaging Center, Beckman Institute, Division of Biology, Pasadena, California 91125, United States of America
| | - Le A. Trinh
- California Institute of Technology, Biological Imaging Center, Beckman Institute, Division of Biology, Pasadena, California 91125, United States of America
- * E-mail:
| |
Collapse
|
31
|
Gouti M, Metzis V, Briscoe J. The route to spinal cord cell types: a tale of signals and switches. Trends Genet 2015; 31:282-9. [PMID: 25823696 DOI: 10.1016/j.tig.2015.03.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 02/28/2015] [Accepted: 03/02/2015] [Indexed: 01/20/2023]
Abstract
Understanding the mechanisms that control induction and elaboration of the vertebrate central nervous system (CNS) requires an analysis of the extrinsic signals and downstream transcriptional networks that assign cell fates in the correct space and time. We focus on the generation and patterning of the spinal cord. We summarize evidence that the origin of the spinal cord is distinct from the anterior regions of the CNS. We discuss how this affects the gene regulatory networks and cell state transitions that specify spinal cord cell subtypes, and we highlight how the timing of extracellular signals and dynamic control of transcriptional networks contribute to the correct spatiotemporal generation of different neural cell types.
Collapse
Affiliation(s)
- Mina Gouti
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, NW7 1AA, UK
| | - Vicki Metzis
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, NW7 1AA, UK
| | - James Briscoe
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, NW7 1AA, UK.
| |
Collapse
|
32
|
Papaioannou VE. The T-box gene family: emerging roles in development, stem cells and cancer. Development 2014; 141:3819-33. [PMID: 25294936 DOI: 10.1242/dev.104471] [Citation(s) in RCA: 224] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The T-box family of transcription factors exhibits widespread involvement throughout development in all metazoans. T-box proteins are characterized by a DNA-binding motif known as the T-domain that binds DNA in a sequence-specific manner. In humans, mutations in many of the genes within the T-box family result in developmental syndromes, and there is increasing evidence to support a role for these factors in certain cancers. In addition, although early studies focused on the role of T-box factors in early embryogenesis, recent studies in mice have uncovered additional roles in unsuspected places, for example in adult stem cell populations. Here, I provide an overview of the key features of T-box transcription factors and highlight their roles and mechanisms of action during various stages of development and in stem/progenitor cell populations.
Collapse
Affiliation(s)
- Virginia E Papaioannou
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
33
|
Keyte AL, Smith KK. Heterochrony and developmental timing mechanisms: changing ontogenies in evolution. Semin Cell Dev Biol 2014; 34:99-107. [PMID: 24994599 DOI: 10.1016/j.semcdb.2014.06.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 06/14/2014] [Accepted: 06/23/2014] [Indexed: 01/20/2023]
Abstract
Heterochrony, or a change in developmental timing, is an important mechanism of evolutionary change. Historically the concept of heterochrony has focused alternatively on changes in size and shape or changes in developmental sequence, but most have focused on the pattern of change. Few studies have examined changes in the mechanisms that embryos use to actually measure time during development. Recently, evolutionary studies focused on changes in distinct timekeeping mechanisms have appeared, and this review examines two such case studies: the evolution of increased segment number in snakes and the extreme rostral to caudal gradient of developmental maturation in marsupials. In both examples, heterochronic modifications of the somite clock have been important drivers of evolutionary change.
Collapse
Affiliation(s)
- Anna L Keyte
- Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | - Kathleen K Smith
- Department of Biology, Duke University, Durham, NC, United States.
| |
Collapse
|
34
|
Muehsam D, Ventura C. Life rhythm as a symphony of oscillatory patterns: electromagnetic energy and sound vibration modulates gene expression for biological signaling and healing. Glob Adv Health Med 2014; 3:40-55. [PMID: 24808981 PMCID: PMC4010966 DOI: 10.7453/gahmj.2014.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- David Muehsam
- Visual Institute of Developmental Sciences, Bologna, Italy (Dr Muehsam)
| | - Carlo Ventura
- National Institute of Biostructures and Biosystems, Visual Institute of Developmental Sciences, Bologna; Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna (Dr Ventura), Italy
| |
Collapse
|
35
|
Rosenberg MI, Brent AE, Payre F, Desplan C. Dual mode of embryonic development is highlighted by expression and function of Nasonia pair-rule genes. eLife 2014; 3:e01440. [PMID: 24599282 PMCID: PMC3941026 DOI: 10.7554/elife.01440] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Embryonic anterior-posterior patterning is well understood in Drosophila, which uses 'long germ' embryogenesis, in which all segments are patterned before cellularization. In contrast, most insects use 'short germ' embryogenesis, wherein only head and thorax are patterned in a syncytial environment while the remainder of the embryo is generated after cellularization. We use the wasp Nasonia (Nv) to address how the transition from short to long germ embryogenesis occurred. Maternal and gap gene expression in Nasonia suggest long germ embryogenesis. However, the Nasonia pair-rule genes even-skipped, odd-skipped, runt and hairy are all expressed as early blastoderm pair-rule stripes and late-forming posterior stripes. Knockdown of Nv eve, odd or h causes loss of alternate segments at the anterior and complete loss of abdominal segments. We propose that Nasonia uses a mixed mode of segmentation wherein pair-rule genes pattern the embryo in a manner resembling Drosophila at the anterior and ancestral Tribolium at the posterior. DOI: http://dx.doi.org/10.7554/eLife.01440.001.
Collapse
Affiliation(s)
- Miriam I Rosenberg
- Center for Developmental Genetics, Department of Biology, New York University, New York, United States
| | | | | | | |
Collapse
|
36
|
Fongang B, Kudlicki A. The precise timeline of transcriptional regulation reveals causation in mouse somitogenesis network. BMC DEVELOPMENTAL BIOLOGY 2013; 13:42. [PMID: 24304493 PMCID: PMC4235037 DOI: 10.1186/1471-213x-13-42] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 11/15/2013] [Indexed: 11/23/2022]
Abstract
Background In vertebrate development, the segmental pattern of the body axis is established as somites, masses of mesoderm distributed along the two sides of the neural tube, are formed sequentially in the anterior-posterior axis. This mechanism depends on waves of gene expression associated with the Notch, Fgf and Wnt pathways. The underlying transcriptional regulation has been studied by whole-transcriptome mRNA profiling; however, interpretation of the results is limited by poor resolution, noisy data, small sample size and by the absence of a wall clock to assign exact time for recorded points. Results We present a method of Maximum Entropy deconvolution in both space and time and apply it to extract, from microarray timecourse data, the full spatiotemporal expression profiles of genes involved in mouse somitogenesis. For regulated genes, we have reconstructed the temporal profiles and determined the timing of expression peaks along the somite cycle to a single-minute resolution. Our results also indicate the presence of a new class of genes (including Raf1 and Hes7) with two peaks of activity in two distinct phases of the somite cycle. We demonstrate that the timeline of gene expression precisely reflects their functions in the biochemical pathways and the direction of causation in the regulatory networks. Conclusions By applying a novel framework for data analysis, we have shown a striking correspondence between gene expression times and their interactions and regulations during somitogenesis. These results prove the key role of finely tuned transcriptional regulation in the process. The presented method can be readily applied to studying somite formation in other datasets and species, and to other spatiotemporal processes.
Collapse
Affiliation(s)
| | - Andrzej Kudlicki
- Department of Biochemistry and Molecular Biology, Sealy Center for Molecular Medicine, Institute for Translational Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555, USA.
| |
Collapse
|
37
|
Franzosa JA, Bugel SM, Tal TL, La Du JK, Tilton SC, Waters KM, Tanguay RL. Retinoic acid-dependent regulation of miR-19 expression elicits vertebrate axis defects. FASEB J 2013; 27:4866-76. [PMID: 23975936 DOI: 10.1096/fj.12-225524] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Retinoic acid (RA) is involved in multifarious and complex functions necessary for vertebrate development. RA signaling is reliant on strict enzymatic regulation of RA synthesis and metabolism. Improper spatiotemporal expression of RA during development can result in vertebrate axis defects. microRNAs (miRNAs) are also pivotal in orchestrating developmental processes. While mechanistic links between miRNAs and axial development are established, the role of miRNAs in regulating metabolic enzymes responsible for RA abundance during axis formation has yet to be elucidated. Our results uncovered a role of miR-19 family members in controlling RA metabolism through the regulation of CYP26A1 during vertebrate axis formation. Global miRNA expression profiling showed that developmental RA exposure suppressed the expression of miR-19 family members during zebrafish somitogenesis. A reporter assay confirmed that cyp26a1 is a bona fide target of miR-19 in vivo. Transient knockdown of miR-19 phenocopied axis defects caused by RA exposure. Exogenous miR-19 rescued the axis defects induced by RA exposure. Taken together, these results indicate that the teratogenic effects of RA exposure result, in part, from repression of miR-19 expression and subsequent misregulation of cyp26a1. This highlights a previously unidentified role of miR-19 in facilitating vertebrate axis development via regulation of RA signaling.
Collapse
Affiliation(s)
- Jill A Franzosa
- 3Department of Environmental and Molecular Toxicology, Oregon State University, 28645 East HWY 34. Corvallis, OR 97333, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
Casaca A, Santos AC, Mallo M. Controlling Hox gene expression and activity to build the vertebrate axial skeleton. Dev Dyn 2013; 243:24-36. [DOI: 10.1002/dvdy.24007] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 06/18/2013] [Accepted: 06/21/2013] [Indexed: 11/10/2022] Open
Affiliation(s)
- Ana Casaca
- Instituto Gulbenkian de Ciência; Oeiras Portugal
| | | | - Moisés Mallo
- Instituto Gulbenkian de Ciência; Oeiras Portugal
| |
Collapse
|
39
|
Main steps of skeletal muscle development in the human. HANDBOOK OF CLINICAL NEUROLOGY 2013; 113:1299-310. [DOI: 10.1016/b978-0-444-59565-2.00002-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
40
|
Neurogenin2 expression together with NeuroM regulates GDNF family neurotrophic factor receptor α1 (GFRα1) expression in the embryonic spinal cord. Dev Biol 2012; 370:250-63. [DOI: 10.1016/j.ydbio.2012.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 08/01/2012] [Accepted: 08/02/2012] [Indexed: 10/28/2022]
|
41
|
Pasini A, Manenti R, Rothbächer U, Lemaire P. Antagonizing retinoic acid and FGF/MAPK pathways control posterior body patterning in the invertebrate chordate Ciona intestinalis. PLoS One 2012; 7:e46193. [PMID: 23049976 PMCID: PMC3458022 DOI: 10.1371/journal.pone.0046193] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 08/28/2012] [Indexed: 11/18/2022] Open
Abstract
Vertebrate embryos exploit the mutual inhibition between the RA and FGF signalling pathways to coordinate the proliferative elongation of the main body axis with the progressive patterning and differentiation of its neuroectodermal and paraxial mesodermal structures. The evolutionary history of this patterning system is still poorly understood. Here, we investigate the role played by the RA and FGF/MAPK signals during the development of the tail structures in the tunicate Ciona intestinalis, an invertebrate chordate belonging to the sister clade of vertebrates, in which the prototypical chordate body plan is established through very derived morphogenetic processes. Ciona embryos are constituted of few cells and develop according to a fixed lineage; elongation of the tail occurs largely by rearrangement of postmitotic cells; mesoderm segmentation and somitogenesis are absent. We show that in the Ciona embryo, the antagonism of the RA and FGF/MAPK signals is required to control the anteroposterior patterning of the tail epidermis. We also demonstrate that the RA, FGF/MAPK and canonical Wnt pathways control the anteroposterior patterning of the tail peripheral nervous system, and reveal the existence of distinct subpopulations of caudal epidermal neurons with different responsiveness to the RA, FGF/MAPK and canonical Wnt signals. Our data provide the first demonstration that the use of the antagonism between the RA and FGF signals to pattern the main body axis predates the emergence of vertebrates and highlight the evolutionary plasticity of this patterning strategy, showing that in different chordates it can be used to pattern different tissues within the same homologous body region.
Collapse
Affiliation(s)
- Andrea Pasini
- Institut de Biologie du Développement de Marseille-Luminy (IBDML), UMR7288, CNRS/Université Aix-Marseille, Marseille, France.
| | | | | | | |
Collapse
|
42
|
Ten Broek CMA, Bakker AJ, Varela-Lasheras I, Bugiani M, Van Dongen S, Galis F. Evo-Devo of the Human Vertebral Column: On Homeotic Transformations, Pathologies and Prenatal Selection. Evol Biol 2012; 39:456-471. [PMID: 23226903 PMCID: PMC3514701 DOI: 10.1007/s11692-012-9196-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 07/11/2012] [Indexed: 12/21/2022]
Abstract
Homeotic transformations of vertebrae are particularly common in humans and tend to come associated with malformations in a wide variety of organ systems. In a dataset of 1,389 deceased human foetuses and infants a majority had cervical ribs and approximately half of these individuals also had missing twelfth ribs or lumbar ribs. In ~10 % of all cases there was an additional shift of the lumbo-sacral boundary and, hence, homeotic transformations resulted in shifts of at least three vertebral boundaries. We found a strong coupling between the abnormality of the vertebral patterns and the amount and strength of associated malformations, i.e., the longer the disturbance of the vertebral patterning has lasted, the more associated malformations have developed and the more organ systems are affected. The germ layer of origin of the malformations was not significantly associated with the frequency of vertebral patterns. In contrast, we find significant associations with the different developmental mechanisms that are involved in the causation of the malformations, that is, segmentation, neural crest development, left-right patterning, etc. Our results, thus, suggest that locally perceived developmental signals are more important for the developmental outcome than the origin of the cells. The low robustness of vertebral A-P patterning apparent from the large number of homeotic transformations is probably caused by the strong interactivity of developmental processes and the low redundancy of involved morphogens during early organogenesis. Additionally, the early irreversibility of the specification of the A-P identity of vertebrae probably adds to the vulnerability of the process by limiting the possibility for recovery from developmental disturbances. The low developmental robustness of vertebral A-P patterning contrasts with a high robustness of the A-P patterning of the vertebral regions. Not only the order is invariable, also the variation in the number of vertebrae per region is small. This robustness is in agreement with the evolutionary stability of vertebral regions in tetrapods. Finally, we propose a new hypothesis regarding the constancy of the presacral number of vertebrae in mammals.
Collapse
Affiliation(s)
- Clara M A Ten Broek
- Group of Evolutionary Ecology, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium ; Naturalis Biodiversity Center, Darwinweg 2, 2333 CR Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
43
|
Rifes P, Thorsteinsdóttir S. Extracellular matrix assembly and 3D organization during paraxial mesoderm development in the chick embryo. Dev Biol 2012; 368:370-81. [DOI: 10.1016/j.ydbio.2012.06.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 06/01/2012] [Accepted: 06/02/2012] [Indexed: 10/28/2022]
|
44
|
Zhu H, Zhao J, Zhou W, Li H, Zhou R, Zhang L, Zhao H, Cao J, Zhu X, Hu H, Ma G, He L, Yao Z, Yao L, Guo X. Ndrg2 regulates vertebral specification in differentiating somites. Dev Biol 2012; 369:308-18. [PMID: 22819676 DOI: 10.1016/j.ydbio.2012.07.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 06/29/2012] [Accepted: 07/02/2012] [Indexed: 12/17/2022]
Abstract
It is generally thought that vertebral patterning and identity are globally determined prior to somite formation. Relatively little is known about the regulators of vertebral specification after somite segmentation. Here, we demonstrated that Ndrg2, a tumor suppressor gene, was dynamically expressed in the presomitic mesoderm (PSM) and at early stage of differentiating somites. Loss of Ndrg2 in mice resulted in vertebral homeotic transformations in thoracic/lumbar and lumbar/sacral transitional regions in a dose-dependent manner. Interestingly, the inactivation of Ndrg2 in osteoblasts or chondrocytes caused defects resembling those observed in Ndrg2(-/-) mice, with a lower penetrance. In addition, forced overexpression of Ndrg2 in osteoblasts or chondrocytes also conferred vertebral defects, which were distinct from those in Ndrg2(-/-) mice. These genetic analyses revealed that Ndrg2 modulates vertebral identity in segmented somites rather than in the PSM. At the molecular level, combinatory alterations of the amount of Hoxc8-11 gene transcripts were detected in the differentiating somites of Ndrg2(-/-) embryos, which may partially account for the vertebral defects in Ndrg2 mutants. Nevertheless, Bmp/Smad signaling activity was elevated in the differentiating somites of Ndrg2(-/-) embryos. Collectively, our findings unveiled Ndrg2 as a novel regulator of vertebral specification in differentiating somites.
Collapse
Affiliation(s)
- Huang Zhu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Rnf11-like is a novel component of NF-κB signaling, governing the posterior patterning in the zebrafish embryos. Biochem Biophys Res Commun 2012; 422:602-6. [PMID: 22609198 DOI: 10.1016/j.bbrc.2012.05.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2012] [Accepted: 05/09/2012] [Indexed: 11/21/2022]
Abstract
RING finger protein 11 (RNF11) is a novel regulator of immunity and cell survival via ubiquitination process in mammalian cells whereas its vertebrate embryonic roles are undefined. Here, we are reporting the isolation, expression and functional roles of an RNF11 orthologue, Rnf11-like in zebrafish embryos. Zebrafish Rnf11-like is composed of 154 amino acids containing RING-H2-finger domain in the C-terminal region and PY-motif. Spatiotemporal expression patterns of rnf11-like indicate that rnf11-like is expressed maternally and zygotically throughout embryogenesis. However, rnf11-like transcripts are present specifically in the presomatic mesoderm (PSM), and later in the brain and retina. Knock-down of Rnf11-like using rnf11-like-specific morpholino causes cell death and developmental defects in the posterior somites, elevating transcripts of NF-κB target gene, ikk1, a negative regulator of NF-κB signaling. All these findings indicate that Rnf11-like is an essential component of NF-κB signaling pathway for specification of the posterior somites in zebrafish embryos.
Collapse
|
46
|
Nowotschin S, Ferrer-Vaquer A, Concepcion D, Papaioannou VE, Hadjantonakis AK. Interaction of Wnt3a, Msgn1 and Tbx6 in neural versus paraxial mesoderm lineage commitment and paraxial mesoderm differentiation in the mouse embryo. Dev Biol 2012; 367:1-14. [PMID: 22546692 DOI: 10.1016/j.ydbio.2012.04.012] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2011] [Revised: 03/27/2012] [Accepted: 04/03/2012] [Indexed: 01/28/2023]
Abstract
Paraxial mesoderm is the tissue which gives rise to the skeletal muscles and vertebral column of the body. A gene regulatory network operating in the formation of paraxial mesoderm has been described. This network hinges on three key factors, Wnt3a, Msgn1 and Tbx6, each of which is critical for paraxial mesoderm formation, since absence of any one of these factors results in complete absence of posterior somites. In this study we determined and compared the spatial and temporal patterns of expression of Wnt3a, Msgn1 and Tbx6 at a time when paraxial mesoderm is being formed. Then, we performed a comparative characterization of mutants in Wnt3a, Msgn1 and Tbx6. To determine the epistatic relationship between these three genes, and begin to decipher the complex interplay between them, we analyzed double mutant embryos and compared their phenotypes to the single mutants. Through the analysis of molecular markers in mutants, our data support the bipotential nature of the progenitor cells for paraxial mesoderm and establish regulatory relationships between genes involved in the choice between neural and mesoderm fates.
Collapse
Affiliation(s)
- Sonja Nowotschin
- Developmental Biology Program, Sloan-Kettering Institute, 1275 York Ave, New York, NY 10065, USA
| | | | | | | | | |
Collapse
|
47
|
Eckalbar WL, Fisher RE, Rawls A, Kusumi K. Scoliosis and segmentation defects of the vertebrae. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2012; 1:401-23. [PMID: 23801490 DOI: 10.1002/wdev.34] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The vertebral column derives from somites, which are transient paired segments of mesoderm that surround the neural tube in the early embryo. Somites are formed by a genetic mechanism that is regulated by cyclical expression of genes in the Notch, Wnt, and fibroblast growth factor (FGF) signaling pathways. These oscillators together with signaling gradients within the presomitic mesoderm help to set somitic boundaries and rostral-caudal polarity that are essential for the precise patterning of the vertebral column. Disruption of this mechanism has been identified as the cause of severe segmentation defects of the vertebrae in humans. These segmentation defects are part of a spectrum of spinal disorders affecting the skeletal elements and musculature of the spine, resulting in curvatures such as scoliosis, kyphosis, and lordosis. While the etiology of most disorders with spinal curvatures is still unknown, genetic and developmental studies of somitogenesis and patterning of the axial skeleton and musculature are yielding insights into the causes of these diseases.
Collapse
|
48
|
Rolland-Lagan AG, Paquette M, Tweedle V, Akimenko MA. Morphogen-based simulation model of ray growth and joint patterning during fin development and regeneration. Development 2012; 139:1188-97. [PMID: 22318227 DOI: 10.1242/dev.073452] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The fact that some organisms are able to regenerate organs of the correct shape and size following amputation is particularly fascinating, but the mechanism by which this occurs remains poorly understood. The zebrafish (Danio rerio) caudal fin has emerged as a model system for the study of bone development and regeneration. The fin comprises 16 to 18 bony rays, each containing multiple joints along its proximodistal axis that give rise to segments. Experimental observations on fin ray growth, regeneration and joint formation have been described, but no unified theory has yet been put forward to explain how growth and joint patterns are controlled. We present a model for the control of fin ray growth during development and regeneration, integrated with a model for joint pattern formation, which is in agreement with published, as well as new, experimental data. We propose that fin ray growth and joint patterning are coordinated through the interaction of three morphogens. When the model is extended to incorporate multiple rays across the fin, it also accounts for how the caudal fin acquires its shape during development, and regains its correct size and shape following amputation.
Collapse
|
49
|
Samarut E, Rochette-Egly C. Nuclear retinoic acid receptors: conductors of the retinoic acid symphony during development. Mol Cell Endocrinol 2012; 348:348-60. [PMID: 21504779 DOI: 10.1016/j.mce.2011.03.025] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 03/21/2011] [Accepted: 03/31/2011] [Indexed: 11/18/2022]
Abstract
The vitamin A derivative, retinoic acid (RA), is essential for embryonic development through the activation of cognate nuclear receptors, RARs, which work as ligand dependent regulators of transcription. In vitro studies revealed how RARs control gene expression at the molecular level and now it appears that it is fine-tuned by a phosphorylation code. In addition, several genetic approaches provided valuable insights on the functions of RARs during development and on the influence of other actors such as the enzymes involved in RA synthesis and degradation and other signaling pathways. It appears that RARs are the conductors of the RA signaling symphony through controlling the dynamics and the coordination of the different players and development steps.
Collapse
Affiliation(s)
- Eric Samarut
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM, U596; CNRS, UMR7104; Université de Strasbourg, 1 rue Laurent Fries, BP 10142, 67404 Illkirch Cedex, France.
| | | |
Collapse
|
50
|
Keyte A, Smith KK. Heterochrony in somitogenesis rate in a model marsupial,Monodelphis domestica. Evol Dev 2012; 14:93-103. [DOI: 10.1111/j.1525-142x.2011.00524.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Anna Keyte
- Duke University; Department of Biology; Durham NC 27708 USA
| | | |
Collapse
|