1
|
Kukreja K, Jia BZ, McGeary SE, Patel N, Megason SG, Klein AM. Cell state transitions are decoupled from cell division during early embryo development. Nat Cell Biol 2024; 26:2035-2045. [PMID: 39516639 DOI: 10.1038/s41556-024-01546-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 09/23/2024] [Indexed: 11/16/2024]
Abstract
As tissues develop, cells divide and differentiate concurrently. Conflicting evidence shows that cell division is either dispensable or required for formation of cell types. Here, to determine the role of cell division in differentiation, we arrested the cell cycle in zebrafish embryos using two independent approaches and profiled them at single-cell resolution. We show that cell division is dispensable for differentiation of all embryonic tissues from early gastrulation to the end of segmentation. However, arresting cell division does slow down differentiation in some cell types, and it induces global stress responses. While differentiation is robust to blocking cell division, the proportions of cells across cell states are not, but show evidence of partial compensation. This work clarifies our understanding of the role of cell division in development and showcases the utility of combining embryo-wide perturbations with single-cell RNA sequencing to uncover the role of common biological processes across multiple tissues.
Collapse
Affiliation(s)
- Kalki Kukreja
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | - Bill Z Jia
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Sean E McGeary
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Nikit Patel
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sean G Megason
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Allon M Klein
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Herre M. Immunostaining Whole-Mount Olfactory Tissues of Mosquitoes. Cold Spring Harb Protoc 2023; 2023:55-60. [PMID: 35940641 DOI: 10.1101/pdb.prot107915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Mosquito olfactory tissues are covered in fine hair-like sensory structures called sensilla that house olfactory sensory neurons. This protocol for immunostaining whole-mounted mosquito antennae and maxillary palps enables the visualization of these neurons.
Collapse
Affiliation(s)
- Margaret Herre
- The Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, New York 10065, USA
| |
Collapse
|
3
|
Azuma Y, Okada H, Onami S. Systematic analysis of cell morphodynamics in C. elegans early embryogenesis. FRONTIERS IN BIOINFORMATICS 2023; 3:1082531. [PMID: 37026092 PMCID: PMC10070942 DOI: 10.3389/fbinf.2023.1082531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 03/07/2023] [Indexed: 04/08/2023] Open
Abstract
The invariant cell lineage of Caenorhabditis elegans allows unambiguous assignment of the identity for each cell, which offers a unique opportunity to study developmental dynamics such as the timing of cell division, dynamics of gene expression, and cell fate decisions at single-cell resolution. However, little is known about cell morphodynamics, including the extent to which they are variable between individuals, mainly due to the lack of sufficient amount and quality of quantified data. In this study, we systematically quantified the cell morphodynamics in 52 C. elegans embryos from the two-cell stage to mid-gastrulation at the high spatiotemporal resolution, 0.5 μm thickness of optical sections, and 30-second intervals of recordings. Our data allowed systematic analyses of the morphological features. We analyzed sphericity dynamics and found a significant increase at the end of metaphase in every cell, indicating the universality of the mitotic cell rounding. Concomitant with the rounding, the volume also increased in most but not all cells, suggesting less universality of the mitotic swelling. Combining all features showed that cell morphodynamics was unique for each cell type. The cells before the onset of gastrulation could be distinguished from all the other cell types. Quantification of reproducibility in cell-cell contact revealed that variability in division timings and cell arrangements produced variability in contacts between the embryos. However, the area of such contacts occupied less than 5% of the total area, suggesting the high reproducibility of spatial occupancies and adjacency relationships of the cells. By comparing the morphodynamics of identical cells between the embryos, we observed diversity in the variability between cells and found it was determined by multiple factors, including cell lineage, cell generation, and cell-cell contact. We compared the variabilities of cell morphodynamics and cell-cell contacts with those in ascidian Phallusia mammillata embryos. The variabilities were larger in C. elegans, despite smaller differences in embryo size and number of cells at each developmental stage.
Collapse
|
4
|
Wong MK, Ho VWS, Huang X, Chan LY, Xie D, Li R, Ren X, Guan G, Ma Y, Hu B, Yan H, Zhao Z. Initial characterization of gap phase introduction in every cell cycle of C. elegans embryogenesis. Front Cell Dev Biol 2022; 10:978962. [PMID: 36393848 PMCID: PMC9641140 DOI: 10.3389/fcell.2022.978962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/20/2022] [Indexed: 11/28/2022] Open
Abstract
Early embryonic cell cycles usually alternate between S and M phases without any gap phase. When the gap phases are developmentally introduced in various cell types remains poorly defined especially during embryogenesis. To establish the cell-specific introduction of gap phases in embryo, we generate multiple fluorescence ubiquitin cell cycle indicators (FUCCI) in C. elegans. Time-lapse 3D imaging followed by lineal expression profiling reveals sharp and differential accumulation of the FUCCI reporters, allowing the systematic demarcation of cell cycle phases throughout embryogenesis. Accumulation of the reporters reliably identifies both G1 and G2 phases only in two embryonic cells with an extended cell cycle length, suggesting that the remaining cells divide either without a G1 phase, or with a brief G1 phase that is too short to be picked up by our reporters. In summary, we provide an initial picture of gap phase introduction in a metazoan embryo. The newly developed FUCCI reporters pave the way for further characterization of developmental control of cell cycle progression.
Collapse
Affiliation(s)
- Ming-Kin Wong
- Department of Biology, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Vincy Wing Sze Ho
- Department of Biology, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Xiaotai Huang
- School of Computer Science and Technology, Xidian University, Xi’an, China
- Department of Electronic Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Lu-Yan Chan
- Department of Biology, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Dongying Xie
- Department of Biology, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Runsheng Li
- Department of Biology, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Xiaoliang Ren
- Department of Biology, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Guoye Guan
- Center for Quantitative Biology, Peking University, Beijing, China
| | - Yiming Ma
- Department of Biology, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Boyi Hu
- Department of Biology, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Hong Yan
- Department of Electronic Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Zhongying Zhao
- Department of Biology, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
- *Correspondence: Zhongying Zhao,
| |
Collapse
|
5
|
Xiao L, Fan D, Qi H, Cong Y, Du Z. Defect-buffering cellular plasticity increases robustness of metazoan embryogenesis. Cell Syst 2022; 13:615-630.e9. [PMID: 35882226 DOI: 10.1016/j.cels.2022.07.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 04/14/2022] [Accepted: 06/30/2022] [Indexed: 01/26/2023]
Abstract
Developmental processes are intrinsically robust so as to preserve a normal-like state in response to genetic and environmental fluctuations. However, the robustness and potential phenotypic plasticity of individual developing cells under genetic perturbations remain to be systematically evaluated. Using large-scale gene perturbation, live imaging, lineage tracing, and single-cell phenomics, we quantified the phenotypic landscape of C. elegans embryogenesis in >2,000 embryos following individual knockdown of over 750 conserved genes. We observed that cellular genetic systems are not sufficiently robust to single-gene perturbations across all cells; rather, gene knockdowns frequently induced cellular defects. Dynamic phenotypic analyses revealed many cellular defects to be transient, with cells exhibiting phenotypic plasticity that serves to alleviate, correct, and accommodate the defects. Moreover, potential developmentally related cell modules may buffer the phenotypic effects of individual cell position changes. Our findings reveal non-negligible contributions of cellular plasticity and multicellularity as compensatory strategies to increase developmental robustness.
Collapse
Affiliation(s)
- Long Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Duchangjiang Fan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huan Qi
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yulin Cong
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhuo Du
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
6
|
Yesbolatova AK, Arai R, Sakaue T, Kimura A. Formulation of Chromatin Mobility as a Function of Nuclear Size during C. elegans Embryogenesis Using Polymer Physics Theories. PHYSICAL REVIEW LETTERS 2022; 128:178101. [PMID: 35570447 DOI: 10.1103/physrevlett.128.178101] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 03/21/2022] [Indexed: 06/15/2023]
Abstract
During early embryogenesis of the nematode, Caenorhabditis elegans, the chromatin motion markedly decreases. Despite its biological implications, the underlying mechanism for this transition was unclear. By combining theory and experiment, we analyze the mean-square displacement (MSD) of the chromatin loci, and demonstrate that MSD-vs-time relationships in various nuclei collapse into a single master curve by normalizing them with the mesh size and the corresponding time scale. This enables us to identify the onset of the entangled dynamics with the size of tube diameter of chromatin polymer in the C. elegans embryo. Our dynamical scaling analysis predicts the transition between unentangled and entangled dynamics of chromatin polymers, the quantitative formula for MSD as a function of nuclear size and timescale, and provides testable hypotheses on chromatin mobility in other cell types and species.
Collapse
Affiliation(s)
- Aiya K Yesbolatova
- Department of Genetics, School of Life Science, The Graduate University for Advanced Studies, SOKENDAI, Mishima 411-8540, Japan
- Cell Architecture Laboratory, Department of Chromosome Science, National Institute of Genetics, Mishima 411-8540, Japan
| | - Ritsuko Arai
- Cell Architecture Laboratory, Department of Chromosome Science, National Institute of Genetics, Mishima 411-8540, Japan
| | - Takahiro Sakaue
- Department of Physical Sciences, Aoyama Gakuin University, 5-10-1 Fuchinobe, Chuo-ku, Sagamihara, Kanagawa 252-5258, Japan
| | - Akatsuki Kimura
- Department of Genetics, School of Life Science, The Graduate University for Advanced Studies, SOKENDAI, Mishima 411-8540, Japan
- Cell Architecture Laboratory, Department of Chromosome Science, National Institute of Genetics, Mishima 411-8540, Japan
| |
Collapse
|
7
|
Adikes RC, Kohrman AQ, Martinez MAQ, Palmisano NJ, Smith JJ, Medwig-Kinney TN, Min M, Sallee MD, Ahmed OB, Kim N, Liu S, Morabito RD, Weeks N, Zhao Q, Zhang W, Feldman JL, Barkoulas M, Pani AM, Spencer SL, Martin BL, Matus DQ. Visualizing the metazoan proliferation-quiescence decision in vivo. eLife 2020; 9:e63265. [PMID: 33350383 PMCID: PMC7880687 DOI: 10.7554/elife.63265] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/21/2020] [Indexed: 12/15/2022] Open
Abstract
Cell proliferation and quiescence are intimately coordinated during metazoan development. Here, we adapt a cyclin-dependent kinase (CDK) sensor to uncouple these key events of the cell cycle in Caenorhabditis elegans and zebrafish through live-cell imaging. The CDK sensor consists of a fluorescently tagged CDK substrate that steadily translocates from the nucleus to the cytoplasm in response to increasing CDK activity and consequent sensor phosphorylation. We show that the CDK sensor can distinguish cycling cells in G1 from quiescent cells in G0, revealing a possible commitment point and a cryptic stochasticity in an otherwise invariant C. elegans cell lineage. Finally, we derive a predictive model of future proliferation behavior in C. elegans based on a snapshot of CDK activity in newly born cells. Thus, we introduce a live-cell imaging tool to facilitate in vivo studies of cell-cycle control in a wide-range of developmental contexts.
Collapse
Affiliation(s)
- Rebecca C Adikes
- Department of Biochemistry and Cell Biology, Stony Brook UniversityStony BrookUnited States
| | - Abraham Q Kohrman
- Department of Biochemistry and Cell Biology, Stony Brook UniversityStony BrookUnited States
| | - Michael A Q Martinez
- Department of Biochemistry and Cell Biology, Stony Brook UniversityStony BrookUnited States
| | - Nicholas J Palmisano
- Department of Biochemistry and Cell Biology, Stony Brook UniversityStony BrookUnited States
| | - Jayson J Smith
- Department of Biochemistry and Cell Biology, Stony Brook UniversityStony BrookUnited States
| | - Taylor N Medwig-Kinney
- Department of Biochemistry and Cell Biology, Stony Brook UniversityStony BrookUnited States
| | - Mingwei Min
- Department of Biochemistry and BioFrontiers Institute, University of Colorado BoulderBoulderUnited States
| | - Maria D Sallee
- Department of Biology, Stanford UniversityStanfordUnited States
| | - Ononnah B Ahmed
- Department of Biochemistry and Cell Biology, Stony Brook UniversityStony BrookUnited States
| | - Nuri Kim
- Department of Biochemistry and Cell Biology, Stony Brook UniversityStony BrookUnited States
| | - Simeiyun Liu
- Department of Biochemistry and Cell Biology, Stony Brook UniversityStony BrookUnited States
| | - Robert D Morabito
- Department of Biochemistry and Cell Biology, Stony Brook UniversityStony BrookUnited States
| | - Nicholas Weeks
- Department of Biochemistry and Cell Biology, Stony Brook UniversityStony BrookUnited States
| | - Qinyun Zhao
- Department of Biochemistry and Cell Biology, Stony Brook UniversityStony BrookUnited States
| | - Wan Zhang
- Department of Biochemistry and Cell Biology, Stony Brook UniversityStony BrookUnited States
| | | | | | - Ariel M Pani
- Department of Biology, University of VirginiaCharlottesvilleUnited States
| | - Sabrina L Spencer
- Department of Biochemistry and BioFrontiers Institute, University of Colorado BoulderBoulderUnited States
| | - Benjamin L Martin
- Department of Biochemistry and Cell Biology, Stony Brook UniversityStony BrookUnited States
| | - David Q Matus
- Department of Biochemistry and Cell Biology, Stony Brook UniversityStony BrookUnited States
| |
Collapse
|
8
|
Gaggioli V, Kieninger MR, Klucnika A, Butler R, Zegerman P. Identification of the critical replication targets of CDK reveals direct regulation of replication initiation factors by the embryo polarity machinery in C. elegans. PLoS Genet 2020; 16:e1008948. [PMID: 33320862 PMCID: PMC7771872 DOI: 10.1371/journal.pgen.1008948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 12/29/2020] [Accepted: 11/02/2020] [Indexed: 12/26/2022] Open
Abstract
During metazoan development, the cell cycle is remodelled to coordinate proliferation with differentiation. Developmental cues cause dramatic changes in the number and timing of replication initiation events, but the mechanisms and physiological importance of such changes are poorly understood. Cyclin-dependent kinases (CDKs) are important for regulating S-phase length in many metazoa, and here we show in the nematode Caenorhabditis elegans that an essential function of CDKs during early embryogenesis is to regulate the interactions between three replication initiation factors SLD-3, SLD-2 and MUS-101 (Dpb11/TopBP1). Mutations that bypass the requirement for CDKs to generate interactions between these factors is partly sufficient for viability in the absence of Cyclin E, demonstrating that this is a critical embryonic function of this Cyclin. Both SLD-2 and SLD-3 are asymmetrically localised in the early embryo and the levels of these proteins inversely correlate with S-phase length. We also show that SLD-2 asymmetry is determined by direct interaction with the polarity protein PKC-3. This study explains an essential function of CDKs for replication initiation in a metazoan and provides the first direct molecular mechanism through which polarization of the embryo is coordinated with DNA replication initiation factors. How and when a cell divides changes as the cell assumes different fates. How these changes in cell division are brought about are poorly understood, but are critical to ensure that cells do not over-proliferate leading to cancer. The nematode C. elegans is an excellent system to study the role of cell cycle changes during animal development. Here we show that two factors SLD-2 and SLD-3 are critical to control the decision to begin genome duplication. We show that these factors are differently distributed to different cell lineages in the early embryo, which may be a key event in determining the cell cycle rate in these cells. For the first time we show that, PKC-3, a key component of the machinery that determines the front (anterior) from the back (posterior) of the embryo directly controls SLD-2 distribution, which might explain how the polarisation of the embryo causes changes in the proliferation of different cell lineages. As PKC-3 is frequently mutated in human cancers, how this factor controls cell proliferation may be important to understand tumour progression.
Collapse
Affiliation(s)
- Vincent Gaggioli
- Wellcome Trust/Cancer Research UK Gurdon Institute, The Henry Wellcome Building of Cancer and Developmental Biology, University of Cambridge, Cambridge, United Kingdom
| | - Manuela R. Kieninger
- Wellcome Trust/Cancer Research UK Gurdon Institute, The Henry Wellcome Building of Cancer and Developmental Biology, University of Cambridge, Cambridge, United Kingdom
| | - Anna Klucnika
- Wellcome Trust/Cancer Research UK Gurdon Institute, The Henry Wellcome Building of Cancer and Developmental Biology, University of Cambridge, Cambridge, United Kingdom
- Department of Genetics, University of Cambridge, United Kingdom
| | - Richard Butler
- Wellcome Trust/Cancer Research UK Gurdon Institute, The Henry Wellcome Building of Cancer and Developmental Biology, University of Cambridge, Cambridge, United Kingdom
| | - Philip Zegerman
- Wellcome Trust/Cancer Research UK Gurdon Institute, The Henry Wellcome Building of Cancer and Developmental Biology, University of Cambridge, Cambridge, United Kingdom
- Department of Biochemistry, University of Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
9
|
Cohen JD, Sundaram MV. C. elegans Apical Extracellular Matrices Shape Epithelia. J Dev Biol 2020; 8:E23. [PMID: 33036165 PMCID: PMC7712855 DOI: 10.3390/jdb8040023] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 02/07/2023] Open
Abstract
Apical extracellular matrices (aECMs) coat exposed surfaces of epithelia to shape developing tissues and protect them from environmental insults. Despite their widespread importance for human health, aECMs are poorly understood compared to basal and stromal ECMs. The nematode Caenorhabditis elegans contains a variety of distinct aECMs, some of which share many of the same types of components (lipids, lipoproteins, collagens, zona pellucida domain proteins, chondroitin glycosaminoglycans and proteoglycans) with mammalian aECMs. These aECMs include the eggshell, a glycocalyx-like pre-cuticle, both collagenous and chitin-based cuticles, and other understudied aECMs of internal epithelia. C. elegans allows rapid genetic manipulations and live imaging of fluorescently-tagged aECM components, and is therefore providing new insights into aECM structure, trafficking, assembly, and functions in tissue shaping.
Collapse
Affiliation(s)
| | - Meera V. Sundaram
- Department of Genetics, University of Pennsylvania Perelman School of Medicine 415 Curie Blvd, Philadelphia, PA 19104-6145, USA;
| |
Collapse
|
10
|
Hwang Y, Hidalgo D, Socolovsky M. The shifting shape and functional specializations of the cell cycle during lineage development. WIREs Mech Dis 2020; 13:e1504. [PMID: 32916032 DOI: 10.1002/wsbm.1504] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 06/29/2020] [Accepted: 07/08/2020] [Indexed: 12/12/2022]
Abstract
Essentially all cell cycling in multicellular organisms in vivo takes place in the context of lineage differentiation. This notwithstanding, the regulation of the cell cycle is often assumed to be generic, independent of tissue or developmental stage. Here we review developmental-stage-specific cell cycle adaptations that may influence developmental decisions, in mammalian erythropoiesis and in other lineages. The length of the cell cycle influences the balance between self-renewal and differentiation in multiple tissues, and may determine lineage fate. Shorter cycles contribute to the efficiency of reprogramming somatic cells into induced pluripotency stem cells and help maintain the pluripotent state. While the plasticity of G1 length is well established, the speed of S phase is emerging as a novel regulated parameter that may influence cell fate transitions in the erythroid lineage, in neural tissue and in embryonic stem cells. A slow S phase may stabilize the self-renewal state, whereas S phase shortening may favor a cell fate change. In the erythroid lineage, functional approaches and single-cell RNA-sequencing show that a key transcriptional switch, at the transition from self-renewal to differentiation, is synchronized with and dependent on S phase. This specific S phase is shorter, as a result of a genome-wide increase in the speed of replication forks. Furthermore, there is progressive shortening in G1 in the period preceding this switch. Together these studies suggest an integrated regulatory landscape of the cycle and differentiation programs, where cell cycle adaptations are controlled by, and in turn feed back on, the propagation of developmental trajectories. This article is categorized under: Biological Mechanisms > Cell Fates Developmental Biology > Stem Cell Biology and Regeneration Developmental Biology > Lineages.
Collapse
Affiliation(s)
- Yung Hwang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Daniel Hidalgo
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Merav Socolovsky
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
11
|
Goldstein B, Nance J. Caenorhabditis elegans Gastrulation: A Model for Understanding How Cells Polarize, Change Shape, and Journey Toward the Center of an Embryo. Genetics 2020; 214:265-277. [PMID: 32029580 PMCID: PMC7017025 DOI: 10.1534/genetics.119.300240] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 12/02/2019] [Indexed: 11/18/2022] Open
Abstract
Gastrulation is fundamental to the development of multicellular animals. Along with neurulation, gastrulation is one of the major processes of morphogenesis in which cells or whole tissues move from the surface of an embryo to its interior. Cell internalization mechanisms that have been discovered to date in Caenorhabditis elegans gastrulation bear some similarity to internalization mechanisms of other systems including Drosophila, Xenopus, and mouse, suggesting that ancient and conserved mechanisms internalize cells in diverse organisms. C. elegans gastrulation occurs at an early stage, beginning when the embryo is composed of just 26 cells, suggesting some promise for connecting the rich array of developmental mechanisms that establish polarity and pattern in embryos to the force-producing mechanisms that change cell shapes and move cells interiorly. Here, we review our current understanding of C. elegans gastrulation mechanisms. We address how cells determine which direction is the interior and polarize with respect to that direction, how cells change shape by apical constriction and internalize, and how the embryo specifies which cells will internalize and when. We summarize future prospects for using this system to discover some of the general principles by which animal cells change shape and internalize during development.
Collapse
Affiliation(s)
- Bob Goldstein
- Department of Biology and
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, North Carolina 27599 and
| | - Jeremy Nance
- Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine and
- Department of Cell Biology, New York University School of Medicine, New York 10016
| |
Collapse
|
12
|
Rothman J, Jarriault S. Developmental Plasticity and Cellular Reprogramming in Caenorhabditis elegans. Genetics 2019; 213:723-757. [PMID: 31685551 PMCID: PMC6827377 DOI: 10.1534/genetics.119.302333] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/25/2019] [Indexed: 12/28/2022] Open
Abstract
While Caenorhabditis elegans was originally regarded as a model for investigating determinate developmental programs, landmark studies have subsequently shown that the largely invariant pattern of development in the animal does not reflect irreversibility in rigidly fixed cell fates. Rather, cells at all stages of development, in both the soma and germline, have been shown to be capable of changing their fates through mutation or forced expression of fate-determining factors, as well as during the normal course of development. In this chapter, we review the basis for natural and induced cellular plasticity in C. elegans We describe the events that progressively restrict cellular differentiation during embryogenesis, starting with the multipotency-to-commitment transition (MCT) and subsequently through postembryonic development of the animal, and consider the range of molecular processes, including transcriptional and translational control systems, that contribute to cellular plasticity. These findings in the worm are discussed in the context of both classical and recent studies of cellular plasticity in vertebrate systems.
Collapse
Affiliation(s)
- Joel Rothman
- Department of MCD Biology and Neuroscience Research Institute, University of California, Santa Barbara, California 93111, and
| | - Sophie Jarriault
- IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire), Department of Development and Stem Cells, CNRS UMR7104, Inserm U1258, Université de Strasbourg, 67404 Illkirch CU Strasbourg, France
| |
Collapse
|
13
|
Hu X, Eastman AE, Guo S. Cell cycle dynamics in the reprogramming of cellular identity. FEBS Lett 2019; 593:2840-2852. [PMID: 31562821 DOI: 10.1002/1873-3468.13625] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 12/13/2022]
Abstract
Reprogramming of cellular identity is fundamentally at odds with replication of the genome: cell fate reprogramming requires complex multidimensional epigenomic changes, whereas genome replication demands fidelity. In this review, we discuss how the pace of the genome's replication and cell cycle influences the way daughter cells take on their identity. We highlight several biochemical processes that are pertinent to cell fate control, whose propagation into the daughter cells should be governed by more complex mechanisms than simple templated replication. With this mindset, we summarize multiple scenarios where rapid cell cycle could interfere with cell fate copying and promote cell fate reprogramming. Prominent examples of cell fate regulation by specific cell cycle phases are also discussed. Overall, there is much to be learned regarding the relationship between cell fate reprogramming and cell cycle control. Harnessing cell cycle dynamics could greatly facilitate the derivation of desired cell types.
Collapse
Affiliation(s)
- Xiao Hu
- Department of Cell Biology, Yale University, New Haven, CT, USA.,Yale Stem Cell Center, Yale University, New Haven, CT, USA
| | - Anna E Eastman
- Department of Cell Biology, Yale University, New Haven, CT, USA.,Yale Stem Cell Center, Yale University, New Haven, CT, USA
| | - Shangqin Guo
- Department of Cell Biology, Yale University, New Haven, CT, USA.,Yale Stem Cell Center, Yale University, New Haven, CT, USA
| |
Collapse
|
14
|
Torfeh E, Simon M, Muggiolu G, Devès G, Vianna F, Bourret S, Incerti S, Barberet P, Seznec H. Monte-Carlo dosimetry and real-time imaging of targeted irradiation consequences in 2-cell stage Caenorhabditis elegans embryo. Sci Rep 2019; 9:10568. [PMID: 31332255 PMCID: PMC6646656 DOI: 10.1038/s41598-019-47122-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 07/11/2019] [Indexed: 12/26/2022] Open
Abstract
Charged-particle microbeams (CPMs) provide a unique opportunity to investigate the effects of ionizing radiation on living biological specimens with a precise control of the delivered dose, i.e. the number of particles per cell. We describe a methodology to manipulate and micro-irradiate early stage C. elegans embryos at a specific phase of the cell division and with a controlled dose using a CPM. To validate this approach, we observe the radiation-induced damage, such as reduced cell mobility, incomplete cell division and the appearance of chromatin bridges during embryo development, in different strains expressing GFP-tagged proteins in situ after irradiation. In addition, as the dosimetry of such experiments cannot be extrapolated from random irradiations of cell populations, realistic three-dimensional models of 2 cell-stage embryo were imported into the Geant4 Monte-Carlo simulation toolkit. Using this method, we investigate the energy deposit in various chromatin condensation states during the cell division phases. The experimental approach coupled to Monte-Carlo simulations provides a way to selectively irradiate a single cell in a rapidly dividing multicellular model with a reproducible dose. This method opens the way to dose-effect investigations following targeted irradiation.
Collapse
Affiliation(s)
- Eva Torfeh
- Université de Bordeaux, Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Chemin du Solarium, 33175, Gradignan, France.,CNRS, UMR5797, Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Chemin du Solarium, 33175, Gradignan, France
| | - Marina Simon
- Université de Bordeaux, Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Chemin du Solarium, 33175, Gradignan, France.,CNRS, UMR5797, Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Chemin du Solarium, 33175, Gradignan, France
| | - Giovanna Muggiolu
- Université de Bordeaux, Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Chemin du Solarium, 33175, Gradignan, France.,CNRS, UMR5797, Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Chemin du Solarium, 33175, Gradignan, France
| | - Guillaume Devès
- Université de Bordeaux, Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Chemin du Solarium, 33175, Gradignan, France.,CNRS, UMR5797, Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Chemin du Solarium, 33175, Gradignan, France
| | - François Vianna
- Université de Bordeaux, Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Chemin du Solarium, 33175, Gradignan, France.,CNRS, UMR5797, Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Chemin du Solarium, 33175, Gradignan, France.,François Vianna: Institut de Radioprotection et de Sûreté Nucléaire, Bat.159, BP3, 13115, St-Paul-Lez-Durance, Cedex, France
| | - Stéphane Bourret
- Université de Bordeaux, Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Chemin du Solarium, 33175, Gradignan, France.,CNRS, UMR5797, Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Chemin du Solarium, 33175, Gradignan, France
| | - Sébastien Incerti
- Université de Bordeaux, Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Chemin du Solarium, 33175, Gradignan, France.,CNRS, UMR5797, Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Chemin du Solarium, 33175, Gradignan, France
| | - Philippe Barberet
- Université de Bordeaux, Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Chemin du Solarium, 33175, Gradignan, France. .,CNRS, UMR5797, Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Chemin du Solarium, 33175, Gradignan, France.
| | - Hervé Seznec
- Université de Bordeaux, Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Chemin du Solarium, 33175, Gradignan, France. .,CNRS, UMR5797, Centre d'Etudes Nucléaires Bordeaux Gradignan (CENBG), Chemin du Solarium, 33175, Gradignan, France.
| |
Collapse
|
15
|
Youssef K, Tandon A, Rezai P. Studying Parkinson’s disease using Caenorhabditis elegans models in microfluidic devices. Integr Biol (Camb) 2019; 11:186-207. [DOI: 10.1093/intbio/zyz017] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 04/30/2019] [Accepted: 05/16/2019] [Indexed: 12/21/2022]
Abstract
Abstract
Parkinson’s disease (PD) is a progressive neurological disorder associated with the loss of dopaminergic neurons (DNs) in the substantia nigra and the widespread accumulation of α-synuclein (α-syn) protein, leading to motor impairments and eventual cognitive dysfunction. In-vitro cell cultures and in-vivo animal models have provided the opportunity to investigate the PD pathological hallmarks and identify different therapeutic compounds. However, PD pathogenesis and causes are still not well understood, and effective inhibitory drugs for PD are yet to be discovered. Biologically simple but pathologically relevant disease models and advanced screening technologies are needed to reveal the mechanisms underpinning protein aggregation and PD progression. For instance, Caenorhabditis elegans (C. elegans) offers many advantages for fundamental PD neurobehavioral studies including a simple, well-mapped, and accessible neuronal system, genetic homology to humans, body transparency and amenability to genetic manipulation. Several transgenic worm strains that exhibit multiple PD-related phenotypes have been developed to perform neuronal and behavioral assays and drug screening. However, in conventional worm-based assays, the commonly used techniques are equipment-intensive, slow and low in throughput. Over the past two decades, microfluidics technology has contributed significantly to automation and control of C. elegans assays. In this review, we focus on C. elegans PD models and the recent advancements in microfluidic platforms used for manipulation, handling and neurobehavioral screening of these models. Moreover, we highlight the potential of C. elegans to elucidate the in-vivo mechanisms of neuron-to-neuron protein transfer that may underlie spreading Lewy pathology in PD, and its suitability for in-vitro studies. Given the advantages of C. elegans and microfluidics technology, their integration has the potential to facilitate the investigation of disease pathology and discovery of potential chemical leads for PD.
Collapse
Affiliation(s)
- Khaled Youssef
- Department of Mechanical Engineering, York University, Toronto, ON, Canada
| | - Anurag Tandon
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Pouya Rezai
- Department of Mechanical Engineering, York University, Toronto, ON, Canada
| |
Collapse
|
16
|
Developmental Control of the Cell Cycle: Insights from Caenorhabditis elegans. Genetics 2019; 211:797-829. [PMID: 30846544 PMCID: PMC6404260 DOI: 10.1534/genetics.118.301643] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 10/10/2018] [Indexed: 12/11/2022] Open
Abstract
During animal development, a single fertilized egg forms a complete organism with tens to trillions of cells that encompass a large variety of cell types. Cell cycle regulation is therefore at the center of development and needs to be carried out in close coordination with cell differentiation, migration, and death, as well as tissue formation, morphogenesis, and homeostasis. The timing and frequency of cell divisions are controlled by complex combinations of external and cell-intrinsic signals that vary throughout development. Insight into how such controls determine in vivo cell division patterns has come from studies in various genetic model systems. The nematode Caenorhabditis elegans has only about 1000 somatic cells and approximately twice as many germ cells in the adult hermaphrodite. Despite the relatively small number of cells, C. elegans has diverse tissues, including intestine, nerves, striated and smooth muscle, and skin. C. elegans is unique as a model organism for studies of the cell cycle because the somatic cell lineage is invariant. Somatic cells divide at set times during development to produce daughter cells that adopt reproducible developmental fates. Studies in C. elegans have allowed the identification of conserved cell cycle regulators and provided insights into how cell cycle regulation varies between tissues. In this review, we focus on the regulation of the cell cycle in the context of C. elegans development, with reference to other systems, with the goal of better understanding how cell cycle regulation is linked to animal development in general.
Collapse
|
17
|
Artiles KL, Fire AZ, Frøkjær-Jensen C. Assessment and Maintenance of Unigametic Germline Inheritance for C. elegans. Dev Cell 2019; 48:827-839.e9. [PMID: 30799227 PMCID: PMC6435406 DOI: 10.1016/j.devcel.2019.01.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 11/06/2018] [Accepted: 01/21/2019] [Indexed: 12/22/2022]
Abstract
The recent work of Besseling and Bringmann (2016) identified a molecular intervention for C. elegans in which premature segregation of maternal and paternal chromosomes in the fertilized oocyte can produce viable animals exhibiting a non-Mendelian inheritance pattern. Overexpression in embryos of a single protein regulating chromosome segregation (GPR-1) provides a germline derived clonally from a single parental gamete. We present a collection of strains and cytological assays to consistently generate and track non-Mendelian inheritance. These tools allow reproducible and high-frequency (>80%) production of non-Mendelian inheritance, the facile and simultaneous homozygosis for all nuclear chromosomes in a single generation, the precise exchange of nuclear and mitochondrial genomes between strains, and the assessments of non-canonical mitosis events. We show the utility of these strains by demonstrating a rapid assessment of cell lineage requirements (AB versus P1) for a set of genes (lin-2, lin-3, lin-12, and lin-31) with roles in C. elegans vulval development.
Collapse
Affiliation(s)
- Karen L Artiles
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Andrew Z Fire
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Christian Frøkjær-Jensen
- King Abdullah University of Science and Technology, Biological and Environmental Science and Engineering Division, KAUST Environmental Epigenetics Program, Thuwal 23955-6900, Saudi Arabia.
| |
Collapse
|
18
|
Smith L, Maddox P. Embryo timelapses can be compiled and quantified to understand canonical histone dynamics across multiple cell cycles. Cytoskeleton (Hoboken) 2018; 75:522-530. [PMID: 30203569 DOI: 10.1002/cm.21493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 07/20/2018] [Accepted: 07/25/2018] [Indexed: 11/09/2022]
Abstract
In the last decade, computational processing of big datasets has facilitated the analyses of unprecedented quantities of biological data. Thus, automation and big data analysis have been revolutionary in detecting and quantifying subtle phenotypes in cell biological contexts. Analyzing similar quantities of data in larger and more complicated biological systems such as developing embryos has been more challenging due to experimental limitations on both ensemble data collection and analysis. These challenges include photosensitivity of living samples and of the fluorescently tagged proteins under study, collectively limiting the number of images that can be acquired in a single timelapse series. Here we present a streamlined workflow to quantify dynamics of fluorescently labeled proteins over the course of several cell cycles in early embryos, taking advantage of the stereotypical nature of early development that is inherent for many organisms. We benchmark this pipeline studying a fluorescently labeled histone during early embryonic development of the nematode Caenorhabditis elegans. Our strategy allowed us to overcome biological and experimental variation among our timelapse series and quantify nuclear accumulation rate, chromatin incorporation, and turnover/stability of canonical histones. We find that histone proteins are broadly stable in early C. elegans development. Thus, changes in genome regulation occurring in early development do not manifest in gross changes in histone metabolism. Our method enabling characterization of cumulative protein dynamics over several cell cycles of developmental time with high temporal resolution can be applied to expand our understanding of diverse cellular and developmental processes.
Collapse
Affiliation(s)
- Lydia Smith
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, North Carolina
| | - Paul Maddox
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, North Carolina
- Department of Biology, University of North Carolina at Chapel Hill, North Carolina
| |
Collapse
|
19
|
Sallee MD, Zonka JC, Skokan TD, Raftrey BC, Feldman JL. Tissue-specific degradation of essential centrosome components reveals distinct microtubule populations at microtubule organizing centers. PLoS Biol 2018; 16:e2005189. [PMID: 30080857 PMCID: PMC6103517 DOI: 10.1371/journal.pbio.2005189] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 08/21/2018] [Accepted: 07/20/2018] [Indexed: 11/19/2022] Open
Abstract
Non-centrosomal microtubule organizing centers (ncMTOCs) are found in most differentiated cells, but how these structures regulate microtubule organization and dynamics is largely unknown. We optimized a tissue-specific degradation system to test the role of the essential centrosomal microtubule nucleators γ-tubulin ring complex (γ-TuRC) and AIR-1/Aurora A at the apical ncMTOC, where they both localize in Caenorhabditis elegans embryonic intestinal epithelial cells. As at the centrosome, the core γ-TuRC component GIP-1/GCP3 is required to recruit other γ-TuRC components to the apical ncMTOC, including MZT-1/MZT1, characterized here for the first time in animal development. In contrast, AIR-1 and MZT-1 were specifically required to recruit γ-TuRC to the centrosome, but not to centrioles or to the apical ncMTOC. Surprisingly, microtubules remain robustly organized at the apical ncMTOC upon γ-TuRC and AIR-1 co-depletion, and upon depletion of other known microtubule regulators, including TPXL-1/TPX2, ZYG-9/ch-TOG, PTRN-1/CAMSAP, and NOCA-1/Ninein. However, loss of GIP-1 removed a subset of dynamic EBP-2/EB1-marked microtubules, and the remaining dynamic microtubules grew faster. Together, these results suggest that different microtubule organizing centers (MTOCs) use discrete proteins for their function, and that the apical ncMTOC is composed of distinct populations of γ-TuRC-dependent and -independent microtubules that compete for a limited pool of resources.
Collapse
Affiliation(s)
- Maria D. Sallee
- Department of Biology, Stanford University, Stanford, California, United States of America
| | - Jennifer C. Zonka
- Department of Biology, Stanford University, Stanford, California, United States of America
| | - Taylor D. Skokan
- Department of Biology, Stanford University, Stanford, California, United States of America
| | - Brian C. Raftrey
- Department of Biology, Stanford University, Stanford, California, United States of America
| | - Jessica L. Feldman
- Department of Biology, Stanford University, Stanford, California, United States of America
| |
Collapse
|
20
|
Furuta T, Joo HJ, Trimmer KA, Chen SY, Arur S. GSK-3 promotes S-phase entry and progression in C. elegans germline stem cells to maintain tissue output. Development 2018; 145:dev.161042. [PMID: 29695611 DOI: 10.1242/dev.161042] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 04/17/2018] [Indexed: 12/26/2022]
Abstract
Adult C. elegans germline stem cells (GSCs) and mouse embryonic stem cells (mESCs) exhibit a non-canonical cell cycle structure with an abbreviated G1 phase and phase-independent expression of Cdk2 and cyclin E. Mechanisms that promote the abbreviated cell cycle remain unknown, as do the consequences of not maintaining an abbreviated cell cycle in these tissues. In GSCs, we discovered that loss of gsk-3 results in reduced GSC proliferation without changes in differentiation or responsiveness to GLP-1/Notch signaling. We find that DPL-1 transcriptional activity inhibits CDK-2 mRNA accumulation in GSCs, which leads to slower S-phase entry and progression. Inhibition of dpl-1 or transgenic expression of CDK-2 via a heterologous germline promoter rescues the S-phase entry and progression defects of the gsk-3 mutants, demonstrating that transcriptional regulation rather than post-translational control of CDK-2 establishes the abbreviated cell cycle structure in GSCs. This highlights an inhibitory cascade wherein GSK-3 inhibits DPL-1 and DPL-1 inhibits cdk-2 transcription. Constitutive GSK-3 activity through this cascade maintains an abbreviated cell cycle structure to permit the efficient proliferation of GSCs necessary for continuous tissue output.
Collapse
Affiliation(s)
- Tokiko Furuta
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hyoe-Jin Joo
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kenneth A Trimmer
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Genes and Development Graduate Program, MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Shin-Yu Chen
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Swathi Arur
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA .,Genes and Development Graduate Program, MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
21
|
Penfield L, Wysolmerski B, Mauro M, Farhadifar R, Martinez MA, Biggs R, Wu HY, Broberg C, Needleman D, Bahmanyar S. Dynein pulling forces counteract lamin-mediated nuclear stability during nuclear envelope repair. Mol Biol Cell 2018; 29:852-868. [PMID: 29386297 PMCID: PMC5905298 DOI: 10.1091/mbc.e17-06-0374] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Transient nuclear envelope (NE) ruptures in the Caenorhabditis elegans zygote are caused by a weakened nuclear lamina during nuclear positioning. Dynein-pulling forces enhance the severity of ruptures, while lamin restricts nucleocytoplasmic mixing and allows stable NE repair. This work is the first mechanistic analysis of NE rupture and repair in an organism. Recent work done exclusively in tissue culture cells revealed that the nuclear envelope (NE) ruptures and repairs in interphase. The duration of NE ruptures depends on lamins; however, the underlying mechanisms and relevance to in vivo events are not known. Here, we use the Caenorhabditis elegans zygote to analyze lamin’s role in NE rupture and repair in vivo. Transient NE ruptures and subsequent NE collapse are induced by weaknesses in the nuclear lamina caused by expression of an engineered hypomorphic C. elegans lamin allele. Dynein-generated forces that position nuclei enhance the severity of transient NE ruptures and cause NE collapse. Reduction of dynein forces allows the weakened lamin network to restrict nucleo–cytoplasmic mixing and support stable NE recovery. Surprisingly, the high incidence of transient NE ruptures does not contribute to embryonic lethality, which is instead correlated with stochastic chromosome scattering resulting from premature NE collapse, suggesting that C. elegans tolerates transient losses of NE compartmentalization during early embryogenesis. In sum, we demonstrate that lamin counteracts dynein forces to promote stable NE repair and prevent catastrophic NE collapse, and thus provide the first mechanistic analysis of NE rupture and repair in an organismal context.
Collapse
Affiliation(s)
- Lauren Penfield
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520
| | - Brian Wysolmerski
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520
| | - Michael Mauro
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520
| | - Reza Farhadifar
- Department of Molecular and Cellular Biology, School of Engineering and Applied Sciences, FAS Center for Systems Biology, Harvard University, Cambridge, MA 02138
| | - Michael A Martinez
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520
| | - Ronald Biggs
- Department of Cellular & Molecular Medicine, Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093
| | - Hai-Yin Wu
- Department of Molecular and Cellular Biology, School of Engineering and Applied Sciences, FAS Center for Systems Biology, Harvard University, Cambridge, MA 02138
| | - Curtis Broberg
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520
| | - Daniel Needleman
- Department of Molecular and Cellular Biology, School of Engineering and Applied Sciences, FAS Center for Systems Biology, Harvard University, Cambridge, MA 02138
| | - Shirin Bahmanyar
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520
| |
Collapse
|
22
|
Michael WM. Cyclin CYB-3 controls both S-phase and mitosis and is asymmetrically distributed in the early C. elegans embryo. Development 2017; 143:3119-27. [PMID: 27578178 DOI: 10.1242/dev.141226] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 07/21/2016] [Indexed: 12/20/2022]
Abstract
In early C. elegans embryos the timing of cell division is both invariant and developmentally regulated, yet how the cell cycle is controlled in the embryo and how cell cycle timing impacts early development remain important, unanswered questions. Here, I focus on the cyclin B3 ortholog CYB-3, and show that this cyclin has the unusual property of controlling both the timely progression through S-phase and mitotic entry, suggesting that CYB-3 is both an S-phase-promoting and mitosis-promoting factor. Furthermore, I find that CYB-3 is asymmetrically distributed in the two-cell embryo, such that the somatic precursor AB cell contains ∼2.5-fold more CYB-3 than its sister cell, the germline progenitor P1 CYB-3 is not only physically limited in P1 but also functionally limited, and this asymmetry is controlled by the par polarity network. These findings highlight the importance of the CYB-3 B3-type cyclin in cell cycle regulation in the early embryo and suggest that CYB-3 asymmetry helps establish the well-documented cell cycle asynchrony that occurs during cell division within the P-lineage.
Collapse
Affiliation(s)
- W Matthew Michael
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
23
|
Fickentscher R, Weiss M. Physical determinants of asymmetric cell divisions in the early development of Caenorhabditis elegans. Sci Rep 2017; 7:9369. [PMID: 28839200 PMCID: PMC5571195 DOI: 10.1038/s41598-017-09690-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 07/25/2017] [Indexed: 01/06/2023] Open
Abstract
Asymmetric cell divisions are of fundamental importance for the development of multicellular organisms, e.g. for the generation of founder cells. Prime examples are asymmetric cell divisions in germline precursors during the early embryogenesis of the transparent roundworm Caenorhabditis elegans, one of the major developmental model organisms. However, due to a lack of quantitative data it has remained unclear how frequent unequal daughter cell sizes emerge in the worm’s early embryogenesis, and whether these originate from sterical or biochemical cues. Using quantitative light-sheet microscopy, we have found that about 40% of all cell divisions in C. elegans until gastrulation generate daughter cells with significantly different volumes. Removing the embryo’s rigid eggshell revealed asymmetric divisions in somatic cells to be primarily induced by steric effects. Division asymmetries in the germline remained unaltered and were correctly reproduced by a model based on a cell-size independent, eccentric displacement of the metaphase plate. Our data suggest that asymmetric cell divisions, imposed by physical determinants, are essential for establishing important cell-cell interactions that eventually fuel a successful embryogenesis.
Collapse
Affiliation(s)
- Rolf Fickentscher
- Experimental Physics I, University of Bayreuth, D-95440, Bayreuth, Germany
| | - Matthias Weiss
- Experimental Physics I, University of Bayreuth, D-95440, Bayreuth, Germany.
| |
Collapse
|
24
|
Ogura Y, Sasakura Y. Emerging mechanisms regulating mitotic synchrony during animal embryogenesis. Dev Growth Differ 2017; 59:565-579. [DOI: 10.1111/dgd.12391] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 07/20/2017] [Accepted: 07/23/2017] [Indexed: 01/21/2023]
Affiliation(s)
- Yosuke Ogura
- Laboratory for Morphogenetic Signaling; RIKEN Center for Developmental Biology; Kobe Japan
| | - Yasunori Sasakura
- Shimoda Marine Research Center; University of Tsukuba; Shizuoka Japan
| |
Collapse
|
25
|
Hwang Y, Futran M, Hidalgo D, Pop R, Iyer DR, Scully R, Rhind N, Socolovsky M. Global increase in replication fork speed during a p57 KIP2-regulated erythroid cell fate switch. SCIENCE ADVANCES 2017; 3:e1700298. [PMID: 28560351 PMCID: PMC5446218 DOI: 10.1126/sciadv.1700298] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 03/28/2017] [Indexed: 06/07/2023]
Abstract
Cell cycle regulators are increasingly implicated in cell fate decisions, such as the acquisition or loss of pluripotency and self-renewal potential. The cell cycle mechanisms that regulate these cell fate decisions are largely unknown. We studied an S phase-dependent cell fate switch, in which murine early erythroid progenitors transition in vivo from a self-renewal state into a phase of active erythroid gene transcription and concurrent maturational cell divisions. We found that progenitors are dependent on p57KIP2-mediated slowing of replication forks for self-renewal, a novel function for cyclin-dependent kinase inhibitors. The switch to differentiation entails rapid down-regulation of p57KIP2 with a consequent global increase in replication fork speed and an abruptly shorter S phase. Our work suggests that cell cycles with specialized global DNA replication dynamics are integral to the maintenance of specific cell states and to cell fate decisions.
Collapse
Affiliation(s)
- Yung Hwang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Melinda Futran
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Daniel Hidalgo
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Ramona Pop
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Divya Ramalingam Iyer
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Ralph Scully
- Division of Hematology-Oncology, Department of Medicine, and Cancer Research Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| | - Nicholas Rhind
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Merav Socolovsky
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|
26
|
Rodríguez-Martínez M, Pinzón N, Ghommidh C, Beyne E, Seitz H, Cayrou C, Méchali M. The gastrula transition reorganizes replication-origin selection in Caenorhabditis elegans. Nat Struct Mol Biol 2017; 24:290-299. [PMID: 28112731 DOI: 10.1038/nsmb.3363] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 12/13/2016] [Indexed: 01/09/2023]
Abstract
Although some features underlying replication-origin activation in metazoan cells have been determined, little is known about their regulation during metazoan development. Using the nascent-strand purification method, here we identified replication origins throughout Caenorhabditis elegans embryonic development and found that the origin repertoire is thoroughly reorganized after gastrulation onset. During the pluripotent embryonic stages (pregastrula), potential cruciform structures and open chromatin are determining factors that establish replication origins. The observed enrichment of replication origins in transcription factor-binding sites and their presence in promoters of highly transcribed genes, particularly operons, suggest that transcriptional activity contributes to replication initiation before gastrulation. After the gastrula transition, when embryonic differentiation programs are set, new origins are selected at enhancers, close to CpG-island-like sequences, and at noncoding genes. Our findings suggest that origin selection coordinates replication initiation with transcriptional programs during metazoan development.
Collapse
Affiliation(s)
| | | | - Charles Ghommidh
- Agropolymer Engineering and Emerging Technologies, University of Montpellier, Montpellier, France
| | | | - Hervé Seitz
- Institute of Human Genetics, CNRS, Montpellier, France
| | | | | |
Collapse
|
27
|
Pourkarimi E, Bellush JM, Whitehouse I. Spatiotemporal coupling and decoupling of gene transcription with DNA replication origins during embryogenesis in C. elegans. eLife 2016; 5:21728. [PMID: 28009254 PMCID: PMC5222557 DOI: 10.7554/elife.21728] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 12/22/2016] [Indexed: 12/13/2022] Open
Abstract
The primary task of developing embryos is genome replication, yet how DNA replication is integrated with the profound cellular changes that occur through development is largely unknown. Using an approach to map DNA replication at high resolution in C. elegans, we show that replication origins are marked with specific histone modifications that define gene enhancers. We demonstrate that the level of enhancer associated modifications scale with the efficiency at which the origin is utilized. By mapping replication origins at different developmental stages, we show that the positions and activity of origins is largely invariant through embryogenesis. Contrary to expectation, we find that replication origins are specified prior to the broad onset of zygotic transcription, yet when transcription initiates it does so in close proximity to the pre-defined replication origins. Transcription and DNA replication origins are correlated, but the association breaks down when embryonic cell division ceases. Collectively, our data indicate that replication origins are fundamental organizers and regulators of gene activity through embryonic development.
Collapse
Affiliation(s)
- Ehsan Pourkarimi
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, United States
| | - James M Bellush
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Iestyn Whitehouse
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, United States
| |
Collapse
|
28
|
Stevens H, Williams AB, Michael WM. Cell-Type Specific Responses to DNA Replication Stress in Early C. elegans Embryos. PLoS One 2016; 11:e0164601. [PMID: 27727303 PMCID: PMC5058509 DOI: 10.1371/journal.pone.0164601] [Citation(s) in RCA: 12] [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: 08/28/2016] [Accepted: 09/27/2016] [Indexed: 12/29/2022] Open
Abstract
To better understand how the cellular response to DNA replication stress is regulated during embryonic development, we and others have established the early C. elegans embryo as a model system to study this important problem. As is the case in most eukaryotic cell types, the replication stress response is controlled by the ATR kinase in early worm embryos. In this report we use RNAi to systematically characterize ATR pathway components for roles in promoting cell cycle delay during a replication stress response, and we find that these genetic requirements vary, depending on the source of stress. We also examine how individual cell types within the embryo respond to replication stress, and we find that the strength of the response, as defined by duration of cell cycle delay, varies dramatically within blastomeres of the early embryo. Our studies shed light on how the replication stress response is managed in the context of embryonic development and show that this pathway is subject to developmental regulation.
Collapse
Affiliation(s)
- Holly Stevens
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, United States of America
| | - Ashley B. Williams
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, United States of America
| | - W. Matthew Michael
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, United States of America
- * E-mail:
| |
Collapse
|
29
|
Tintori SC, Osborne Nishimura E, Golden P, Lieb JD, Goldstein B. A Transcriptional Lineage of the Early C. elegans Embryo. Dev Cell 2016; 38:430-44. [PMID: 27554860 PMCID: PMC4999266 DOI: 10.1016/j.devcel.2016.07.025] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 05/19/2016] [Accepted: 07/27/2016] [Indexed: 12/30/2022]
Abstract
During embryonic development, cells must establish fates, morphologies, and behaviors in coordination with one another to form a functional body. A prevalent hypothesis for how this coordination is achieved is that each cell's fate and behavior is determined by a defined mixture of RNAs. Only recently has it become possible to measure the full suite of transcripts in a single cell. Here we quantify genome-wide mRNA abundance in each cell of the Caenorhabditis elegans embryo up to the 16-cell stage. We describe spatially dynamic expression, quantify cell-specific differential activation of the zygotic genome, and identify genes that were previously unappreciated as being critical for development. We present an interactive data visualization tool that allows broad access to our dataset. This genome-wide single-cell map of mRNA abundance, alongside the well-studied life history and fate of each cell, describes at a cellular resolution the mRNA landscape that guides development.
Collapse
Affiliation(s)
- Sophia C Tintori
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Erin Osborne Nishimura
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Patrick Golden
- School of Information and Library Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jason D Lieb
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bob Goldstein
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
30
|
Wong MK, Guan D, Ng KHC, Ho VWS, An X, Li R, Ren X, Zhao Z. Timing of Tissue-specific Cell Division Requires a Differential Onset of Zygotic Transcription during Metazoan Embryogenesis. J Biol Chem 2016; 291:12501-12513. [PMID: 27056332 DOI: 10.1074/jbc.m115.705426] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Indexed: 12/20/2022] Open
Abstract
Metazoan development demands not only precise cell fate differentiation but also accurate timing of cell division to ensure proper development. How cell divisions are temporally coordinated during development is poorly understood. Caenorhabditis elegans embryogenesis provides an excellent opportunity to study this coordination due to its invariant development and widespread division asynchronies. One of the most pronounced asynchronies is a significant delay of cell division in two endoderm progenitor cells, Ea and Ep, hereafter referred to as E2, relative to its cousins that mainly develop into mesoderm organs and tissues. To unravel the genetic control over the endoderm-specific E2 division timing, a total of 822 essential and conserved genes were knocked down using RNAi followed by quantification of cell cycle lengths using in toto imaging of C. elegans embryogenesis and automated lineage. Intriguingly, knockdown of numerous genes encoding the components of general transcription pathway or its regulatory factors leads to a significant reduction in the E2 cell cycle length but an increase in cell cycle length of the remaining cells, indicating a differential requirement of transcription for division timing between the two. Analysis of lineage-specific RNA-seq data demonstrates an earlier onset of transcription in endoderm than in other germ layers, the timing of which coincides with the birth of E2, supporting the notion that the endoderm-specific delay in E2 division timing demands robust zygotic transcription. The reduction in E2 cell cycle length is frequently associated with cell migration defect and gastrulation failure. The results suggest that a tissue-specific transcriptional activation is required to coordinate fate differentiation, division timing, and cell migration to ensure proper development.
Collapse
Affiliation(s)
- Ming-Kin Wong
- Department of Biology, Hong Kong Baptist University, Hong Kong Baptist University, Hong Kong, China
| | - Daogang Guan
- Department of Biology, Hong Kong Baptist University, Hong Kong Baptist University, Hong Kong, China
| | - Kaoru Hon Chun Ng
- Department of Biology, Hong Kong Baptist University, Hong Kong Baptist University, Hong Kong, China
| | - Vincy Wing Sze Ho
- Department of Biology, Hong Kong Baptist University, Hong Kong Baptist University, Hong Kong, China
| | - Xiaomeng An
- Department of Biology, Hong Kong Baptist University, Hong Kong Baptist University, Hong Kong, China
| | - Runsheng Li
- Department of Biology, Hong Kong Baptist University, Hong Kong Baptist University, Hong Kong, China
| | - Xiaoliang Ren
- Department of Biology, Hong Kong Baptist University, Hong Kong Baptist University, Hong Kong, China
| | - Zhongying Zhao
- Department of Biology, Hong Kong Baptist University, Hong Kong Baptist University, Hong Kong, China; State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, China.
| |
Collapse
|
31
|
Zacharias AL, Murray JI. Combinatorial decoding of the invariant C. elegans embryonic lineage in space and time. Genesis 2016; 54:182-97. [PMID: 26915329 PMCID: PMC4840027 DOI: 10.1002/dvg.22928] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 02/18/2016] [Accepted: 02/22/2016] [Indexed: 12/19/2022]
Abstract
Understanding how a single cell, the zygote, can divide and differentiate to produce the diverse animal cell types is a central goal of developmental biology research. The model organism Caenorhabditis elegans provides a system that enables a truly comprehensive understanding of this process across all cells. Its invariant cell lineage makes it possible to identify all of the cells in each individual and compare them across organisms. Recently developed methods automate the process of cell identification, allowing high-throughput gene expression characterization and phenotyping at single cell resolution. In this Review, we summarize the sequences of events that pattern the lineage including establishment of founder cell identity, the signaling pathways that diversify embryonic fate, and the regulators involved in patterning within these founder lineages before cells adopt their terminal fates. We focus on insights that have emerged from automated approaches to lineage tracking, including insights into mechanisms of robustness, context-specific regulation of gene expression, and temporal coordination of differentiation. We suggest a model by which lineage history produces a combinatorial code of transcription factors that act, often redundantly, to ensure terminal fate.
Collapse
Affiliation(s)
- Amanda L. Zacharias
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - John Isaac Murray
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104 USA
| |
Collapse
|
32
|
Identifying Regulators of Morphogenesis Common to Vertebrate Neural Tube Closure and Caenorhabditis elegans Gastrulation. Genetics 2015; 202:123-39. [PMID: 26434722 DOI: 10.1534/genetics.115.183137] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 09/29/2015] [Indexed: 12/18/2022] Open
Abstract
Neural tube defects including spina bifida are common and severe congenital disorders. In mice, mutations in more than 200 genes can result in neural tube defects. We hypothesized that this large gene set might include genes whose homologs contribute to morphogenesis in diverse animals. To test this hypothesis, we screened a set of Caenorhabditis elegans homologs for roles in gastrulation, a topologically similar process to vertebrate neural tube closure. Both C. elegans gastrulation and vertebrate neural tube closure involve the internalization of surface cells, requiring tissue-specific gene regulation, actomyosin-driven apical constriction, and establishment and maintenance of adhesions between specific cells. Our screen identified several neural tube defect gene homologs that are required for gastrulation in C. elegans, including the transcription factor sptf-3. Disruption of sptf-3 in C. elegans reduced the expression of early endodermally expressed genes as well as genes expressed in other early cell lineages, establishing sptf-3 as a key contributor to multiple well-studied C. elegans cell fate specification pathways. We also identified members of the actin regulatory WAVE complex (wve-1, gex-2, gex-3, abi-1, and nuo-3a). Disruption of WAVE complex members reduced the narrowing of endodermal cells' apical surfaces. Although WAVE complex members are expressed broadly in C. elegans, we found that expression of a vertebrate WAVE complex member, nckap1, is enriched in the developing neural tube of Xenopus. We show that nckap1 contributes to neural tube closure in Xenopus. This work identifies in vivo roles for homologs of mammalian neural tube defect genes in two manipulable genetic model systems.
Collapse
|
33
|
Sonneville R, Craig G, Labib K, Gartner A, Blow JJ. Both Chromosome Decondensation and Condensation Are Dependent on DNA Replication in C. elegans Embryos. Cell Rep 2015; 12:405-17. [PMID: 26166571 PMCID: PMC4521082 DOI: 10.1016/j.celrep.2015.06.046] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 05/22/2015] [Accepted: 06/13/2015] [Indexed: 01/19/2023] Open
Abstract
During cell division, chromatin alternates between a condensed state to facilitate chromosome segregation and a decondensed form when DNA replicates. In most tissues, S phase and mitosis are separated by defined G1 and G2 gap phases, but early embryogenesis involves rapid oscillations between replication and mitosis. Using Caenorhabditis elegans embryos as a model system, we show that chromosome condensation and condensin II concentration on chromosomal axes require replicated DNA. In addition, we found that, during late telophase, replication initiates on condensed chromosomes and promotes the rapid decondensation of the chromatin. Upon replication initiation, the CDC-45-MCM-GINS (CMG) DNA helicase drives the release of condensin I complexes from chromatin and the activation or displacement of inactive MCM-2-7 complexes, which together with the nucleoporin MEL-28/ELYS tethers condensed chromatin to the nuclear envelope, thereby promoting chromatin decondensation. Our results show how, in an early embryo, the chromosome-condensation cycle is functionally linked with DNA replication.
Collapse
Affiliation(s)
- Remi Sonneville
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dundee DD1 5EH, UK; MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Gillian Craig
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Karim Labib
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Anton Gartner
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dundee DD1 5EH, UK.
| | - J Julian Blow
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dundee DD1 5EH, UK.
| |
Collapse
|
34
|
Yang R, Feldman JL. SPD-2/CEP192 and CDK Are Limiting for Microtubule-Organizing Center Function at the Centrosome. Curr Biol 2015; 25:1924-31. [PMID: 26119750 DOI: 10.1016/j.cub.2015.06.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 05/18/2015] [Accepted: 06/01/2015] [Indexed: 10/23/2022]
Abstract
The centrosome acts as the microtubule-organizing center (MTOC) during mitosis in animal cells. Microtubules are nucleated and anchored by γ-tubulin ring complexes (γ-TuRCs) embedded within the centrosome's pericentriolar material (PCM). The PCM is required for the localization of γ-TuRCs, and both are steadily recruited to the centrosome, culminating in a peak in MTOC function in metaphase. In differentiated cells, the centrosome is often attenuated as an MTOC and MTOC function is reassigned to non-centrosomal sites such as the apical membrane in epithelial cells, the nuclear envelope in skeletal muscle, and down the lengths of axons and dendrites in neurons. Hyperactive MTOC function at the centrosome is associated with epithelial cancers and with invasive behavior in tumor cells. Little is known about the mechanisms that limit MTOC activation at the centrosome. Here, we find that MTOC function at the centrosome is completely inactivated during cell differentiation in C. elegans embryonic intestinal cells and MTOC function is reassigned to the apical membrane. In cells that divide after differentiation, the cellular MTOC state switches between the membrane and the centrosome. Using cell fusion experiments in live embryos, we find that the centrosome MTOC state is dominant and that the inactive MTOC state of the centrosome is malleable; fusion of a mitotic cell to a differentiated or interphase cell results in rapid reactivation of the centrosome MTOC. We show that conversion of MTOC state involves the conserved centrosome protein SPD-2/CEP192 and CDK activity from the mitotic cell.
Collapse
Affiliation(s)
- Renzhi Yang
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Jessica L Feldman
- Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
35
|
Kowalski MP, Baylis HA, Krude T. Non-coding stem-bulge RNAs are required for cell proliferation and embryonic development in C. elegans. J Cell Sci 2015; 128:2118-29. [PMID: 25908866 PMCID: PMC4450293 DOI: 10.1242/jcs.166744] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 04/06/2015] [Indexed: 12/21/2022] Open
Abstract
Stem bulge RNAs (sbRNAs) are a family of small non-coding stem-loop RNAs present in Caenorhabditis elegans and other nematodes, the function of which is unknown. Here, we report the first functional characterisation of nematode sbRNAs. We demonstrate that sbRNAs from a range of nematode species are able to reconstitute the initiation of chromosomal DNA replication in the presence of replication proteins in vitro, and that conserved nucleotide sequence motifs are essential for this function. By functionally inactivating sbRNAs with antisense morpholino oligonucleotides, we show that sbRNAs are required for S phase progression, early embryonic development and the viability of C. elegans in vivo. Thus, we demonstrate a new and essential role for sbRNAs during the early development of C. elegans. sbRNAs show limited nucleotide sequence similarity to vertebrate Y RNAs, which are also essential for the initiation of DNA replication. Our results therefore establish that the essential function of small non-coding stem-loop RNAs during DNA replication extends beyond vertebrates.
Collapse
Affiliation(s)
- Madzia P Kowalski
- Department of Zoology, University of Cambridge, Downing Street, Cambridge, CB2 3EJ, UK
| | - Howard A Baylis
- Department of Zoology, University of Cambridge, Downing Street, Cambridge, CB2 3EJ, UK
| | - Torsten Krude
- Department of Zoology, University of Cambridge, Downing Street, Cambridge, CB2 3EJ, UK
| |
Collapse
|
36
|
Arata Y, Takagi H, Sako Y, Sawa H. Power law relationship between cell cycle duration and cell volume in the early embryonic development of Caenorhabditis elegans. Front Physiol 2015; 5:529. [PMID: 25674063 PMCID: PMC4309120 DOI: 10.3389/fphys.2014.00529] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/27/2014] [Indexed: 11/13/2022] Open
Abstract
Cell size is a critical factor for cell cycle regulation. In Xenopus embryos after midblastula transition (MBT), the cell cycle duration elongates in a power law relationship with the cell radius squared. This correlation has been explained by the model that cell surface area is a candidate to determine cell cycle duration. However, it remains unknown whether this second power law is conserved in other animal embryos. Here, we found that the relationship between cell cycle duration and cell size in Caenorhabditis elegans embryos exhibited a power law distribution. Interestingly, the powers of the time-size relationship could be grouped into at least three classes: highly size-correlated, moderately size-correlated, and potentially a size-non-correlated class according to C. elegans founder cell lineages (1.2, 0.81, and <0.39 in radius, respectively). Thus, the power law relationship is conserved in Xenopus and C. elegans, while the absolute powers in C. elegans were different from that in Xenopus. Furthermore, we found that the volume ratio between the nucleus and cell exhibited a power law relationship in the size-correlated classes. The power of the volume relationship was closest to that of the time-size relationship in the highly size-correlated class. This correlation raised the possibility that the time-size relationship, at least in the highly size-correlated class, is explained by the volume ratio of nuclear size and cell size. Thus, our quantitative measurements shed a light on the possibility that early embryonic C. elegans cell cycle duration is coordinated with cell size as a result of geometric constraints between intracellular structures.
Collapse
Affiliation(s)
- Yukinobu Arata
- Laboratory for Cell Fate Decision, Center for Developmental Biology, RIKEN Hyogo, Japan ; Cellular Informatics Laboratory, RIKEN Saitama, Japan
| | - Hiroaki Takagi
- Department of Physics, School of Medicine, Nara Medical University Nara, Japan
| | - Yasushi Sako
- Cellular Informatics Laboratory, RIKEN Saitama, Japan
| | - Hitoshi Sawa
- Laboratory for Cell Fate Decision, Center for Developmental Biology, RIKEN Hyogo, Japan ; Multicellular Organization Laboratory, National Institute of Genetics Shizuoka, Japan
| |
Collapse
|
37
|
Samson M, Jow MM, Wong CCL, Fitzpatrick C, Aslanian A, Saucedo I, Estrada R, Ito T, Park SKR, Yates JR, Chu DS. The specification and global reprogramming of histone epigenetic marks during gamete formation and early embryo development in C. elegans. PLoS Genet 2014; 10:e1004588. [PMID: 25299455 PMCID: PMC4191889 DOI: 10.1371/journal.pgen.1004588] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 07/09/2014] [Indexed: 11/18/2022] Open
Abstract
In addition to the DNA contributed by sperm and oocytes, embryos receive parent-specific epigenetic information that can include histone variants, histone post-translational modifications (PTMs), and DNA methylation. However, a global view of how such marks are erased or retained during gamete formation and reprogrammed after fertilization is lacking. To focus on features conveyed by histones, we conducted a large-scale proteomic identification of histone variants and PTMs in sperm and mixed-stage embryo chromatin from C. elegans, a species that lacks conserved DNA methylation pathways. The fate of these histone marks was then tracked using immunostaining. Proteomic analysis found that sperm harbor ∼2.4 fold lower levels of histone PTMs than embryos and revealed differences in classes of PTMs between sperm and embryos. Sperm chromatin repackaging involves the incorporation of the sperm-specific histone H2A variant HTAS-1, a widespread erasure of histone acetylation, and the retention of histone methylation at sites that mark the transcriptional history of chromatin domains during spermatogenesis. After fertilization, we show HTAS-1 and 6 histone PTM marks distinguish sperm and oocyte chromatin in the new embryo and characterize distinct paternal and maternal histone remodeling events during the oocyte-to-embryo transition. These include the exchange of histone H2A that is marked by ubiquitination, retention of HTAS-1, removal of the H2A variant HTZ-1, and differential reprogramming of histone PTMs. This work identifies novel and conserved features of paternal chromatin that are specified during spermatogenesis and processed in the embryo. Furthermore, our results show that different species, even those with diverged DNA packaging and imprinting strategies, use conserved histone modification and removal mechanisms to reprogram epigenetic information.
Collapse
Affiliation(s)
- Mark Samson
- Department of Biology, San Francisco State University, San Francisco, California, United States of America
| | - Margaret M. Jow
- Department of Biology, San Francisco State University, San Francisco, California, United States of America
| | - Catherine C. L. Wong
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, United States of America
- Mass Spectrometry Division, National Center for Protein Science Shanghai, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Science, Shanghai, China
| | - Colin Fitzpatrick
- Department of Biology, San Francisco State University, San Francisco, California, United States of America
| | - Aaron Aslanian
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Israel Saucedo
- Department of Biology, San Francisco State University, San Francisco, California, United States of America
| | - Rodrigo Estrada
- Department of Biology, San Francisco State University, San Francisco, California, United States of America
| | - Takashi Ito
- Department of Biochemistry, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Sung-kyu Robin Park
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, United States of America
| | - John R. Yates
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Diana S. Chu
- Department of Biology, San Francisco State University, San Francisco, California, United States of America
| |
Collapse
|
38
|
Xu C, Su Z. Identification of genes driving lineage divergence from single-cell gene expression data in C. elegans. Dev Biol 2014; 393:236-244. [PMID: 25050933 DOI: 10.1016/j.ydbio.2014.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 07/09/2014] [Accepted: 07/11/2014] [Indexed: 11/25/2022]
Abstract
The nematode Caenorhabditis elegans (C. elegans) is an ideal model organism to study the cell fate specification mechanisms during embryogenesis. It is generally believed that cell fate specification in C. elegans is mainly mediated by lineage-based mechanisms, where the specification paths are driven forward by a succession of asymmetric cell divisions. However, little is known about how each binary decision is made by gene regulatory programs. In this study, we endeavor to obtain a global understanding of cell lineage/fate divergence processes during the early embryogenesis of C. elegans. We reanalyzed the EPIC data set, which traced the expression level of reporter genes at single-cell resolution on a nearly continuous time scale up to the 350-cell stage in C. elegans embryos. We examined the expression patterns for a total of 131 genes from 287 embryos with high quality image recordings, among which 86 genes have replicate embryos. Our results reveal that during early embryogenesis, divergence between sister lineages could be largely explained by a few genes. We predicted genes driving lineage divergence and explored their expression patterns in sister lineages. Moreover, we found that divisions leading to fate divergence are associated with a large number of genes being differentially expressed between sister lineages. Interestingly, we found that the developmental paths of lineages could be differentiated by a small set of genes. Therefore, our results support the notion that the cell fate patterns in C. elegans are achieved through stepwise binary decisions punctuated by cell divisions. Our predicted genes driving lineage divergence provide good starting points for future detailed characterization of their roles in the embryogenesis in this important model organism.
Collapse
Affiliation(s)
- Chen Xu
- Department of Bioinformatics and Genomics, The University of North Carolina at Charlotte, 351 Bioinformatics Building, 9201 University City Blvd, Charlotte, NC 28223, USA.
| | - Zhengchang Su
- Department of Bioinformatics and Genomics, The University of North Carolina at Charlotte, 351 Bioinformatics Building, 9201 University City Blvd, Charlotte, NC 28223, USA.
| |
Collapse
|
39
|
Benkemoun L, Descoteaux C, Chartier NT, Pintard L, Labbé JC. PAR-4/LKB1 regulates DNA replication during asynchronous division of the early C. elegans embryo. ACTA ACUST UNITED AC 2014; 205:447-55. [PMID: 24841566 PMCID: PMC4033775 DOI: 10.1083/jcb.201312029] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
DNA replication is asymmetrically regulated in the two-cell stage C. elegans embryo by the PAR-4 and PAR-1 polarity proteins, which function independently of known regulators of cell cycle timing to dampen DNA replication dynamics specifically in the posterior blastomere. Regulation of cell cycle duration is critical during development, yet the underlying molecular mechanisms are still poorly understood. The two-cell stage Caenorhabditis elegans embryo divides asynchronously and thus provides a powerful context in which to study regulation of cell cycle timing during development. Using genetic analysis and high-resolution imaging, we found that deoxyribonucleic acid (DNA) replication is asymmetrically regulated in the two-cell stage embryo and that the PAR-4 and PAR-1 polarity proteins dampen DNA replication dynamics specifically in the posterior blastomere, independently of regulators previously implicated in the control of cell cycle timing. Our results demonstrate that accurate control of DNA replication is crucial during C. elegans early embryonic development and further provide a novel mechanism by which PAR proteins control cell cycle progression during asynchronous cell division.
Collapse
Affiliation(s)
- Laura Benkemoun
- Cell Division and Differentiation Laboratory, Institute of Research in Immunology and Cancer, and Department of Pathology and Cell Biology, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Catherine Descoteaux
- Cell Division and Differentiation Laboratory, Institute of Research in Immunology and Cancer, and Department of Pathology and Cell Biology, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Nicolas T Chartier
- Cell Division and Differentiation Laboratory, Institute of Research in Immunology and Cancer, and Department of Pathology and Cell Biology, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Lionel Pintard
- Institut Jacques Monod, Centre National de la Recherche Scientifique and Université Paris Diderot, F-75013 Paris, France
| | - Jean-Claude Labbé
- Cell Division and Differentiation Laboratory, Institute of Research in Immunology and Cancer, and Department of Pathology and Cell Biology, Université de Montréal, Montréal, Québec H3T 1J4, CanadaCell Division and Differentiation Laboratory, Institute of Research in Immunology and Cancer, and Department of Pathology and Cell Biology, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| |
Collapse
|
40
|
Fischer AHL, Pang K, Henry JQ, Martindale MQ. A cleavage clock regulates features of lineage-specific differentiation in the development of a basal branching metazoan, the ctenophore Mnemiopsis leidyi. EvoDevo 2014; 5:4. [PMID: 24485336 PMCID: PMC3909359 DOI: 10.1186/2041-9139-5-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 11/20/2013] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND An important question in experimental embryology is to understand how the developmental potential responsible for the generation of distinct cell types is spatially segregated over developmental time. Classical embryological work showed that ctenophores, a group of gelatinous marine invertebrates that arose early in animal evolution, display a highly stereotyped pattern of early development and a precocious specification of blastomere fates. Here we investigate the role of autonomous cell specification and the developmental timing of two distinct ctenophore cell types (motile compound comb-plate-like cilia and light-emitting photocytes) in embryos of the lobate ctenophore, Mnemiopsis leidyi. RESULTS In Mnemiopsis, 9 h after fertilization, comb plate cilia differentiate into derivatives of the E lineage, while the bioluminescent capability begins in derivatives of the M lineage. Arresting cleavage with cytochalasin B at the 1-, 2- or 4-cell stage does not result in blastomere death; however, no visible differentiation of the comb-plate-like cilia or bioluminescence was observed. Cleavage arrest at the 8- or 16-cell stage, in contrast, results in the expression of both differentiation products. Fate-mapping experiments indicate that only the lineages of cells that normally express these markers in an autonomous fashion during normal development express these traits in cleavage-arrested 8- and 16-cell stage embryos. Lineages that form comb plates in a non-autonomous fashion (derivatives of the M lineage) do not. Timed actinomycin D and puromycin treatments show that transcription and translation are required for comb formation and suggest that the segregated material might be necessary for activation of the appropriate genes. Interestingly, even in the absence of cytokinesis, differentiation markers appear to be activated at the correct times. Treatments with a DNA synthesis inhibitor, aphidicolin, show that the number of nuclear divisions, and perhaps the DNA to cytoplasmic ratio, are critical for the appearance of lineage-specific differentiation. CONCLUSION Our work corroborates previous studies demonstrating that the cleavage program is causally involved in the spatial segregation and/or activation of factors that give rise to distinct cell types in ctenophore development. These factors are segregated independently to the appropriate lineage at the 8- and the 16-cell stages and have features of a clock, such that comb-plate-like cilia and light-emitting photoproteins appear at roughly the same developmental time in cleavage-arrested embryos as they do in untreated embryos. Nuclear division, which possibly affects DNA-cytoplasmic ratios, appears to be important in the timing of differentiation markers. Evidence suggests that the 60-cell stage, just prior to gastrulation, is the time of zygotic gene activation. Such cleavage-clock-regulated phenomena appear to be widespread amongst the Metazoa and these cellular and molecular developmental mechanisms probably evolved early in metazoan evolution.
Collapse
Affiliation(s)
- Antje HL Fischer
- Developmental Biology Unit, European Molecular Biology Laboratory Heidelberg, Meyerhof Strasse 1, Heidelberg 69117, Germany
- current address: Molecular and Cell Biology Department, Harvard University, 16 Divinity Avenue, Cambridge, MA 02138, USA
| | - Kevin Pang
- Kewalo Marine Laboratory, Pacific Biosciences Research Center, University of Hawaii at Manoa, Honolulu, HI, USA
- current address: Sars International Centre for Marine Molecular Biology, Thormøhlensgt. 55, Bergen N-5008, Norway
| | - Jonathan Q Henry
- Department of Cell and Structural Biology, University of Illinois, 601 S. Goodwin Ave, Urbana, IL 61801, USA
| | - Mark Q Martindale
- Whitney Lab for Marine Bioscience, Univ. Florida, 9505 Oceanshore Blvd, St, Augustine, FL 32080, USA
| |
Collapse
|
41
|
Wang J, Garrey J, Davis RE. Transcription in pronuclei and one- to four-cell embryos drives early development in a nematode. Curr Biol 2013; 24:124-133. [PMID: 24374308 DOI: 10.1016/j.cub.2013.11.045] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 11/18/2013] [Accepted: 11/21/2013] [Indexed: 10/25/2022]
Abstract
BACKGROUND A long-standing view of development is that transcription is silenced in the oocyte until early divisions in the embryo. The point at which major transcription is reactivated varies between organisms, but is usually after the two-cell stage. However, this model may not be universal. RESULTS We used RNA-seq and exploited the protracted development of the parasitic nematode Ascaris suum to provide a comprehensive time course of mRNA expression, degradation, and translation during early development. Surprisingly, we find that ∼4,000 genes are transcribed prior to pronuclear fusion and in the one- to four-cell embryos. Intriguingly, we do not detect maternal contribution of many orthologs of maternal C. elegans mRNAs, but instead find that these are newly transcribed in the A. suum zygote prior to pronuclear fusion. Ribosome profiling demonstrates that, in general, early embryonic mRNAs are not stored for subsequent translation, but are directly translated after their synthesis. The role of maternally contributed and zygotically transcribed genes differs between the nematodes A. suum and C. elegans despite the fact that the two nematodes appear to exhibit highly similar morphological patterns during early development. CONCLUSIONS Our study indicates that major transcription can occur immediately after fertilization and prior to pronuclear fusion in metazoa, suggesting that newly transcribed genes appear to drive A. suum early development. Furthermore, the mechanisms used for controlling the timing of the expression of key conserved genes has been altered between the two nematodes, illustrating significant plasticity in the regulatory networks that play important roles in developmental outcomes in nematodes.
Collapse
Affiliation(s)
- Jianbin Wang
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Julianne Garrey
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Richard E Davis
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| |
Collapse
|
42
|
Nair G, Walton T, Murray JI, Raj A. Gene transcription is coordinated with, but not dependent on, cell divisions during C. elegans embryonic fate specification. Development 2013; 140:3385-94. [PMID: 23863485 DOI: 10.1242/dev.098012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cell differentiation and proliferation are coordinated during animal development, but the link between them remains uncharacterized. To examine this relationship, we combined single-molecule RNA imaging with time-lapse microscopy to generate high-resolution measurements of transcriptional dynamics in Caenorhabditis elegans embryogenesis. We found that globally slowing the overall development rate of the embryo by altering temperature or by mutation resulted in cell proliferation and transcription slowing, but maintaining, their relative timings, suggesting that cell division may directly control transcription. However, using mutants with specific defects in cell cycle pathways that lead to abnormal lineages, we found that the order between cell divisions and expression onset can switch, showing that expression of developmental regulators is not strictly dependent on cell division. Delaying cell divisions resulted in only slight changes in absolute expression time, suggesting that expression and proliferation are independently entrained to a separate clock-like process. These changes in relative timing can change the number of cells expressing a gene at a given time, suggesting that timing may help determine which cells adopt particular transcriptional patterns. Our results place limits on the types of mechanisms that are used during normal development to ensure that division timing and fate specification occur at appropriate times.
Collapse
Affiliation(s)
- Gautham Nair
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104-6321, USA
| | | | | | | |
Collapse
|
43
|
Johnson NM, Lemmens BBLG, Tijsterman M. A role for the malignant brain tumour (MBT) domain protein LIN-61 in DNA double-strand break repair by homologous recombination. PLoS Genet 2013; 9:e1003339. [PMID: 23505385 PMCID: PMC3591299 DOI: 10.1371/journal.pgen.1003339] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 01/08/2013] [Indexed: 11/18/2022] Open
Abstract
Malignant brain tumour (MBT) domain proteins are transcriptional repressors that function within Polycomb complexes. Some MBT genes are tumour suppressors, but how they prevent tumourigenesis is unknown. The Caenorhabditis elegans MBT protein LIN-61 is a member of the synMuvB chromatin-remodelling proteins that control vulval development. Here we report a new role for LIN-61: it protects the genome by promoting homologous recombination (HR) for the repair of DNA double-strand breaks (DSBs). lin-61 mutants manifest numerous problems associated with defective HR in germ and somatic cells but remain proficient in meiotic recombination. They are hypersensitive to ionizing radiation and interstrand crosslinks but not UV light. Using a novel reporter system that monitors repair of a defined DSB in C. elegans somatic cells, we show that LIN-61 contributes to HR. The involvement of this MBT protein in HR raises the possibility that MBT–deficient tumours may also have defective DSB repair. The genome is continually under threat from exogenous sources of DNA damage, as well as from sources that originate within the cell. DNA double-strand breaks (DSBs) are arguably the most problematic type of damage as they can cause dangerous chromosome rearrangements, which can lead to cancer, as well as mutation at the break site and/or cell death. A complex network of molecular pathways, collectively referred to as the DNA damage response (DDR), have evolved to protect the cell from these threats. We have discovered a new DDR factor, LIN-61, that promotes the repair of DSBs. This is a novel and unexpected role for LIN-61, which was previously known to act as a regulator of gene transcription during development.
Collapse
Affiliation(s)
- Nicholas M. Johnson
- Department of Toxicogenetics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Marcel Tijsterman
- Department of Toxicogenetics, Leiden University Medical Center, Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
44
|
Richards JL, Zacharias AL, Walton T, Burdick JT, Murray JI. A quantitative model of normal Caenorhabditis elegans embryogenesis and its disruption after stress. Dev Biol 2012; 374:12-23. [PMID: 23220655 DOI: 10.1016/j.ydbio.2012.11.034] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 11/12/2012] [Accepted: 11/29/2012] [Indexed: 10/27/2022]
Abstract
The invariant lineage of Caenorhabditis elegans has powerful potential for quantifying developmental variability in normal and stressed embryos. Previous studies of division timing by automated lineage tracing suggested that variability in cell cycle timing is low in younger embryos, but manual lineage tracing of specific lineages suggested that variability may increase for later divisions. We developed improved automated lineage tracing methods that allowroutine lineage tracing through the last round of embryonic cell divisions and we applied these methods to trace the lineage of 18 wild-type embryos. Cell cycle lengths, division axes and cell positions are remarkably consistent among these embryos at all stages, with only slight increase in variability later in development. The resulting quantitative 4-dimensional model of embryogenesis provides a powerful reference dataset to identify defects in mutants or in embryos that have experienced environmental perturbations. We also traced the lineages of embryos imaged at higher temperatures to quantify the decay in developmental robustness under temperature stress. Developmental variability increases modestly at 25°C compared with 22°C and dramatically at 26°C, and we identify homeotic transformations in a subset of embryos grown at 26°C. The deep lineage tracing methods provide a powerful tool for analysis of normal development, gene expression and mutants and we provide a graphical user interface to allow other researchers to explore the average behavior of arbitrary cells in a reference embryo.
Collapse
Affiliation(s)
- Julia L Richards
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, Philadelphia, PA 19104, USA.
| | | | | | | | | |
Collapse
|
45
|
McGhee JD. TheCaenorhabditis elegansintestine. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2012; 2:347-67. [DOI: 10.1002/wdev.93] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
46
|
A broad requirement for TLS polymerases η and κ, and interacting sumoylation and nuclear pore proteins, in lesion bypass during C. elegans embryogenesis. PLoS Genet 2012; 8:e1002800. [PMID: 22761594 PMCID: PMC3386174 DOI: 10.1371/journal.pgen.1002800] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 05/16/2012] [Indexed: 12/04/2022] Open
Abstract
Translesion synthesis (TLS) polymerases are specialized DNA polymerases capable of inserting nucleotides opposite DNA lesions that escape removal by dedicated DNA repair pathways. TLS polymerases allow cells to complete DNA replication in the presence of damage, thereby preventing checkpoint activation, genome instability, and cell death. Here, we characterize functional knockouts for polh-1 and polk-1, encoding the Caenorhabditis elegans homologs of the Y-family TLS polymerases η and κ. POLH-1 acts at many different DNA lesions as it protects cells against a wide range of DNA damaging agents, including UV, γ-irradiation, cisplatin, and methyl methane sulphonate (MMS). POLK-1 acts specifically but redundantly with POLH-1 in protection against methylation damage. Importantly, both polymerases play a prominent role early in embryonic development to allow fast replication of damaged genomes. Contrary to observations in mammalian cells, we show that neither POLH-1 nor POLK-1 is required for homologous recombination (HR) repair of DNA double-strand breaks. A genome-wide RNAi screen for genes that protect the C. elegans genome against MMS–induced DNA damage identified novel components in DNA damage bypass in the early embryo. Our data suggest SUMO-mediated regulation of both POLH-1 and POLK-1, and point towards a previously unrecognized role of the nuclear pore in regulating TLS. Unrepaired DNA damage on the template strand poses a problem for the progression of the replication fork. Specialized translesion synthesis (TLS) polymerases are capable of bypassing DNA lesions without repairing them. Here, we use the nematode C. elegans, to show that there is modulation of the choice between repair and bypass during development. We show that during gametogenesis and later development repair dominates, while there is a short phase during embryonic development where resistance to damage depends heavily on TLS polymerases. The rapid divisions at this stage do not allow for delay in which repair processes can occur. Furthermore, we identify new factors that may play a role in the regulation of TLS during early embryogenesis.
Collapse
|
47
|
Sonneville R, Querenet M, Craig A, Gartner A, Blow JJ. The dynamics of replication licensing in live Caenorhabditis elegans embryos. J Cell Biol 2012; 196:233-46. [PMID: 22249291 PMCID: PMC3265957 DOI: 10.1083/jcb.201110080] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 12/13/2011] [Indexed: 01/07/2023] Open
Abstract
Accurate DNA replication requires proper regulation of replication licensing, which entails loading MCM-2-7 onto replication origins. In this paper, we provide the first comprehensive view of replication licensing in vivo, using video microscopy of Caenorhabditis elegans embryos. As expected, MCM-2-7 loading in late M phase depended on the prereplicative complex (pre-RC) proteins: origin recognition complex (ORC), CDC-6, and CDT-1. However, many features we observed have not been described before: GFP-ORC-1 bound chromatin independently of ORC-2-5, and CDC-6 bound chromatin independently of ORC, whereas CDT-1 and MCM-2-7 DNA binding was interdependent. MCM-3 chromatin loading was irreversible, but CDC-6 and ORC turned over rapidly, consistent with ORC/CDC-6 loading multiple MCM-2-7 complexes. MCM-2-7 chromatin loading further reduced ORC and CDC-6 DNA binding. This dynamic behavior creates a feedback loop allowing ORC/CDC-6 to repeatedly load MCM-2-7 and distribute licensed origins along chromosomal DNA. During S phase, ORC and CDC-6 were excluded from nuclei, and DNA was overreplicated in export-defective cells. Thus, nucleocytoplasmic compartmentalization of licensing factors ensures that DNA replication occurs only once.
Collapse
Affiliation(s)
- Remi Sonneville
- Wellcome Trust Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, UK
| | | | | | | | | |
Collapse
|
48
|
Abstract
Caenorhabditis elegans is an important system for the study of cell cycle regulation in the context of animal development. One of the most powerful features of C. elegans is the invariant cell lineage in which somatic cells initiate cell division at specific times within the developmental program. The cell lineage is fully known and provides the foundation for the analysis of cell cycle progression at single-cell resolution in a multicellular organism. In this chapter, we describe the different types of cell cycles observed in C. elegans, and provide methods and reagents for the analysis of cell cycle progression as well as specific cell cycle phases. We also provide strategies for the analysis and proper interpretation of cell cycle and checkpoint mutants.
Collapse
|
49
|
Abstract
The direct manipulation of embryonic cells is an important tool for addressing key questions in cell and developmental biology. C. elegans is relatively unique among genetic model systems in being amenable to manipulation of embryonic cells. Embryonic cell manipulation has allowed the identification of cell interactions by direct means, and it has been an important technique for dissecting mechanisms by which cell fates are specified, cell divisions are oriented, and morphogenesis is accomplished. Here, we present detailed methods for isolating, manipulating and culturing embryonic cells of C. elegans.
Collapse
Affiliation(s)
- Lois G Edgar
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Colorado, USA
| | | |
Collapse
|
50
|
Zhang S, Banerjee D, Kuhn JR. Isolation and culture of larval cells from C. elegans. PLoS One 2011; 6:e19505. [PMID: 21559335 PMCID: PMC3084877 DOI: 10.1371/journal.pone.0019505] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Accepted: 03/30/2011] [Indexed: 12/20/2022] Open
Abstract
Cell culture is an essential tool to study cell function. In C. elegans the ability to isolate and culture cells has been limited to embryonically derived cells. However, cells or blastomeres isolated from mixed stage embryos terminally differentiate within 24 hours of culture, thus precluding post-embryonic stage cell culture. We have developed an efficient and technically simple method for large-scale isolation and primary culture of larval-stage cells. We have optimized the treatment to maximize cell number and minimize cell death for each of the four larval stages. We obtained up to 7.8×10(4) cells per microliter of packed larvae, and up to 97% of adherent cells isolated by this method were viable for at least 16 hours. Cultured larval cells showed stage-specific increases in both cell size and multinuclearity and expressed lineage- and cell type-specific reporters. The majority (81%) of larval cells isolated by our method were muscle cells that exhibited stage-specific phenotypes. L1 muscle cells developed 1 to 2 wide cytoplasmic processes, while L4 muscle cells developed 4 to 14 processes of various thicknesses. L4 muscle cells developed bands of myosin heavy chain A thick filaments at the cell center and spontaneously contracted ex vivo. Neurons constituted less than 10% of the isolated cells and the majority of neurons developed one or more long, microtubule-rich protrusions that terminated in actin-rich growth cones. In addition to cells such as muscle and neuron that are high abundance in vivo, we were also able to isolate M-lineage cells that constitute less than 0.2% of cells in vivo. Our novel method of cell isolation extends C. elegans cell culture to larval developmental stages, and allows use of the wealth of cell culture tools, such as cell sorting, electrophysiology, co-culture, and high-resolution imaging of subcellular dynamics, in investigation of post-embryonic development and physiology.
Collapse
Affiliation(s)
- Sihui Zhang
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Diya Banerjee
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Jeffrey R. Kuhn
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, United States of America
- * E-mail:
| |
Collapse
|