1
|
Levin M. The Multiscale Wisdom of the Body: Collective Intelligence as a Tractable Interface for Next-Generation Biomedicine. Bioessays 2025; 47:e202400196. [PMID: 39623868 PMCID: PMC11848127 DOI: 10.1002/bies.202400196] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 02/25/2025]
Abstract
The dominant paradigm in biomedicine focuses on genetically-specified components of cells and their biochemical dynamics, emphasizing bottom-up emergence of complexity. Here, I explore the biomedical implications of a complementary emerging field: diverse intelligence. Using tools from behavioral science and multiscale neuroscience, we can study development, regenerative repair, and cancer suppression as behaviors of a collective intelligence of cells navigating the spaces of possible morphologies and transcriptional and physiological states. A focus on the competencies of living material-from molecular to organismal scales-reveals a new landscape for interventions. Such top-down approaches take advantage of the memories and homeodynamic goal-seeking behavior of cells and tissues, offering the same massive advantages in biomedicine and bioengineering that reprogrammable hardware has provided information technologies. The bioelectric networks that bind individual cells toward large-scale anatomical goals are an especially tractable interface to organ-level plasticity, and tools to modulate them already exist. This suggests a research program to understand and tame the software of life for therapeutic gain by understanding the many examples of basal cognition that operate throughout living bodies.
Collapse
Affiliation(s)
- Michael Levin
- Biology DepartmentAllen Discovery Center at Tufts UniversityMedfordMassachusettsUSA
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMassachusettsUSA
| |
Collapse
|
2
|
Michaut A, Mongera A, Gupta A, Tarazona OA, Serra M, Kefala GM, Rigoni P, Lee JG, Rivas F, Hall AR, Mahadevan L, Guevorkian K, Pourquié O. Extracellular volume expansion drives vertebrate axis elongation. Curr Biol 2025; 35:843-853.e6. [PMID: 39879975 DOI: 10.1016/j.cub.2024.12.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 10/15/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025]
Abstract
The vertebrate bauplan is primarily established via the formation of embryonic tissues in a head-to-tail progression. The mechanics of this elongation, which requires the presomitic mesoderm (PSM), remain poorly understood. Here, we find that avian PSM explants can elongate autonomously when physically confined in vitro, producing a pushing force promoting posterior elongation of the embryo. This tissue elongation is caused by volumetric expansion, which results from an increase in the extracellular fraction accompanied by graded cellular motility. We show that fibroblast growth factor (FGF) signaling promotes glycolysis-dependent production of hyaluronic acid (HA), which is required for expansion of the posterior PSM. Our findings link body axis elongation to tissue expansion through the metabolic control of extracellular matrix production downstream of FGF signaling.
Collapse
Affiliation(s)
- Arthur Michaut
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Alessandro Mongera
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Anupam Gupta
- Department of Physics, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana, 502285, India
| | - Oscar A Tarazona
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Mattia Serra
- Department of Physics, University of California at San Diego, San Diego, CA 92093, USA
| | - Georgia-Maria Kefala
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Physics of Cells and Cancer, 75005 Paris, France
| | - Pietro Rigoni
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Jong Gwan Lee
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Felipe Rivas
- Virginia Tech, Wake Forest School of Biomedical Engineering and Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Adam R Hall
- Virginia Tech, Wake Forest School of Biomedical Engineering and Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - L Mahadevan
- Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Department of Physics, Harvard University, Cambridge, MA 02138, USA.
| | - Karine Guevorkian
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Physics of Cells and Cancer, 75005 Paris, France.
| | - Olivier Pourquié
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
3
|
Stooke-Vaughan GA, Kim S, Yen ST, Son K, Banavar SP, Giammona J, Kimelman D, Campàs O. The physical roles of different posterior tissues in zebrafish axis elongation. Nat Commun 2025; 16:1839. [PMID: 39984461 PMCID: PMC11845790 DOI: 10.1038/s41467-025-56334-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 01/16/2025] [Indexed: 02/23/2025] Open
Abstract
Shaping embryonic tissues requires spatiotemporal changes in genetic and signaling activity as well as in tissue mechanics. Studies linking specific molecular perturbations to changes in the tissue physical state remain sparse. Here we study how specific genetic perturbations affecting different posterior tissues during zebrafish body axis elongation change their physical state, the resulting large-scale tissue flows, and posterior elongation. Using a custom analysis software to reveal spatiotemporal variations in tissue fluidity, we show that dorsal tissues are most fluid at the posterior end, rigidify anterior of this region, and become more fluid again yet further anteriorly. In the absence of notochord (noto mutants) or when the presomitic mesoderm is substantially reduced (tbx16 mutants), dorsal tissues elongate normally. Perturbations of posterior-directed morphogenetic flows in dorsal tissues (vangl2 mutants) strongly affect the speed of elongation, highlighting the essential role of dorsal cell flows in delivering the necessary material to elongate the axis.
Collapse
Affiliation(s)
| | - Sangwoo Kim
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA
- Institute of Mechanical Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Shuo-Ting Yen
- Cluster of Excellence Physics of Life, TU Dresden, Dresden, Germany
| | - Kevin Son
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA
| | - Samhita P Banavar
- Department of Physics, University of California, Santa Barbara, CA, USA
- Department of Chemical and Biological Engineering, Princeton University, New Jersey, NJ, USA
| | - James Giammona
- Department of Physics, University of California, Santa Barbara, CA, USA
| | - David Kimelman
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Otger Campàs
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA.
- Cluster of Excellence Physics of Life, TU Dresden, Dresden, Germany.
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
- Center for Systems Biology Dresden, Dresden, Germany.
| |
Collapse
|
4
|
Uriu K, Morelli LG. Statistical description of mobile oscillators in embryonic pattern formation. Phys Rev E 2025; 111:024407. [PMID: 40103159 DOI: 10.1103/physreve.111.024407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 01/27/2025] [Indexed: 03/20/2025]
Abstract
Synchronization of mobile oscillators occurs in numerous contexts, including physical, chemical, biological, and engineered systems. In vertebrate embryonic development, a segmental body structure is generated by a population of mobile oscillators. Cells in this population produce autonomous gene expression rhythms and interact with their neighbors through local signaling. These cells form an extended tissue where frequency and cell mobility gradients coexist. Gene expression kinematic waves travel through this tissue and pattern the segment boundaries. It has been shown that oscillator mobility promotes global synchronization. However, in vertebrate segment formation, mobility may also introduce local fluctuations in kinematic waves and impair segment boundaries. Here, we derive a general framework for mobile oscillators that relates local mobility fluctuations to synchronization dynamics and pattern robustness. We formulate a statistical description of mobile phase oscillators in terms of probability density. We obtain and solve diffusion equations for the average phase and variance, revealing the relationship between local fluctuations and global synchronization in a homogeneous population of oscillators. Analysis of the probability density for large mobility identifies a mean-field onset, where locally coupled oscillators start behaving as if each oscillator was coupled with all the others. We extend the statistical description to inhomogeneous systems to address the gradients present in the vertebrate segmenting tissue. The theory relates pattern stability to mobility, coupling, and pattern wavelength. The general approach of the statistical description may be applied to mobile oscillators in other contexts, as well as to other patterning systems where mobility is present.
Collapse
Affiliation(s)
- Koichiro Uriu
- School of Life Science and Technology, Institute of Science Tokyo, 2-12-1, Ookayama, Meguro-ku Tokyo 152-8550, Japan
- Kanazawa University, Graduate School of Natural Science and Technology, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Luis G Morelli
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET/Partner Institute of the Max Planck Society, Polo Científico Tecnológico, Godoy Cruz 2390, Buenos Aires C1425FQD, Argentina
| |
Collapse
|
5
|
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
|
6
|
Romanos M, Salisbury T, Stephan S, Lansford R, Degond P, Trescases A, Bénazéraf B. Differential proliferation regulates multi-tissue morphogenesis during embryonic axial extension: integrating viscous modeling and experimental approaches. Development 2024; 151:dev202836. [PMID: 38856082 DOI: 10.1242/dev.202836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/28/2024] [Indexed: 06/11/2024]
Abstract
A major challenge in biology is to understand how mechanical interactions and cellular behavior affect the shapes of tissues and embryo morphology. The extension of the neural tube and paraxial mesoderm, which form the spinal cord and musculoskeletal system, respectively, results in the elongated shape of the vertebrate embryonic body. Despite our understanding of how each of these tissues elongates independently of the others, the morphogenetic consequences of their simultaneous growth and mechanical interactions are still unclear. Our study investigates how differential growth, tissue biophysical properties and mechanical interactions affect embryonic morphogenesis during axial extension using a 2D multi-tissue continuum-based mathematical model. Our model captures the dynamics observed in vivo by time-lapse imaging of bird embryos, and reveals the underestimated influence of differential tissue proliferation rates. We confirmed this prediction in quail embryos by showing that decreasing the rate of cell proliferation in the paraxial mesoderm affects long-term tissue dynamics, and shaping of both the paraxial mesoderm and the neighboring neural tube. Overall, our work provides a new theoretical platform upon which to consider the long-term consequences of tissue differential growth and mechanical interactions on morphogenesis.
Collapse
Affiliation(s)
- Michèle Romanos
- Molecular, Cellular and Developmental Biology Unit (MCD, UMR 5077), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
- Institut de Mathématiques de Toulouse UMR 5219, Université de Toulouse, CNRS, 31062 Toulouse Cedex 9, France
- Université Claude Bernard Lyon 1, CNRS, Ecole Centrale de Lyon, INSA Lyon, Université Jean Monnet, ICJ UMR5208, 69622 Villeurbanne, France
| | - Tasha Salisbury
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- University of Southern California, Los Angeles, CA 90089, USA
| | - Samuel Stephan
- Molecular, Cellular and Developmental Biology Unit (MCD, UMR 5077), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Rusty Lansford
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- University of Southern California, Los Angeles, CA 90089, USA
| | - Pierre Degond
- Institut de Mathématiques de Toulouse UMR 5219, Université de Toulouse, CNRS, 31062 Toulouse Cedex 9, France
| | - Ariane Trescases
- Institut de Mathématiques de Toulouse UMR 5219, Université de Toulouse, CNRS, 31062 Toulouse Cedex 9, France
| | - Bertrand Bénazéraf
- Molecular, Cellular and Developmental Biology Unit (MCD, UMR 5077), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
| |
Collapse
|
7
|
Boutillon A, Banavar SP, Campàs O. Conserved physical mechanisms of cell and tissue elongation. Development 2024; 151:dev202687. [PMID: 38767601 PMCID: PMC11190436 DOI: 10.1242/dev.202687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Living organisms have the ability to self-shape into complex structures appropriate for their function. The genetic and molecular mechanisms that enable cells to do this have been extensively studied in several model and non-model organisms. In contrast, the physical mechanisms that shape cells and tissues have only recently started to emerge, in part thanks to new quantitative in vivo measurements of the physical quantities guiding morphogenesis. These data, combined with indirect inferences of physical characteristics, are starting to reveal similarities in the physical mechanisms underlying morphogenesis across different organisms. Here, we review how physics contributes to shape cells and tissues in a simple, yet ubiquitous, morphogenetic transformation: elongation. Drawing from observed similarities across species, we propose the existence of conserved physical mechanisms of morphogenesis.
Collapse
Affiliation(s)
- Arthur Boutillon
- Cluster of Excellence Physics of Life, TU Dresden, 01062 Dresden, Germany
| | - Samhita P. Banavar
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08540, USA
| | - Otger Campàs
- Cluster of Excellence Physics of Life, TU Dresden, 01062 Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
- Center for Systems Biology Dresden, 01307 Dresden, Germany
| |
Collapse
|
8
|
McDaniel C, Simsek MF, Chandel AS, Özbudak EM. Spatiotemporal control of pattern formation during somitogenesis. SCIENCE ADVANCES 2024; 10:eadk8937. [PMID: 38277458 PMCID: PMC10816718 DOI: 10.1126/sciadv.adk8937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 12/27/2023] [Indexed: 01/28/2024]
Abstract
Spatiotemporal patterns widely occur in biological, chemical, and physical systems. Particularly, embryonic development displays a diverse gamut of repetitive patterns established in many tissues and organs. Branching treelike structures in lungs, kidneys, livers, pancreases, and mammary glands as well as digits and bones in appendages, teeth, and palates are just a few examples. A fascinating instance of repetitive patterning is the sequential segmentation of the primary body axis, which is conserved in all vertebrates and many arthropods and annelids. In these species, the body axis elongates at the posterior end of the embryo containing an unsegmented tissue. Meanwhile, segments sequentially bud off from the anterior end of the unsegmented tissue, laying down an exquisite repetitive pattern and creating a segmented body plan. In vertebrates, the paraxial mesoderm is sequentially divided into somites. In this review, we will discuss the most prominent models, the most puzzling experimental data, and outstanding questions in vertebrate somite segmentation.
Collapse
Affiliation(s)
- Cassandra McDaniel
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Systems Biology and Physiology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - M. Fethullah Simsek
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Angad Singh Chandel
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Systems Biology and Physiology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Ertuğrul M. Özbudak
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| |
Collapse
|
9
|
Cumplido N, Arratia G, Desvignes T, Muñoz-Sánchez S, Postlethwait JH, Allende ML. Hox genes control homocercal caudal fin development and evolution. SCIENCE ADVANCES 2024; 10:eadj5991. [PMID: 38241378 PMCID: PMC10798566 DOI: 10.1126/sciadv.adj5991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 12/19/2023] [Indexed: 01/21/2024]
Abstract
Ancient bony fishes had heterocercal tails, like modern sharks and sturgeons, with asymmetric caudal fins and a vertebral column extending into an elongated upper lobe. Teleost fishes, in contrast, developed a homocercal tail characterized by two separate equal-sized fin lobes and the body axis not extending into the caudal fin. A similar heterocercal-to-homocercal transition occurs during teleost ontogeny, although the underlying genetic and developmental mechanisms for either transition remain unresolved. Here, we investigated the role of hox13 genes in caudal fin formation as these genes control posterior identity in animals. Analysis of expression profiles of zebrafish hox13 paralogs and phenotypes of CRISPR/Cas9-induced mutants showed that double hoxb13a and hoxc13a mutants fail to form a caudal fin. Furthermore, single mutants display heterocercal-like morphologies not seen since Mesozoic fossil teleosteomorphs. Relaxation of functional constraints after the teleost genome duplication may have allowed hox13 duplicates to neo- or subfunctionalize, ultimately contributing to the evolution of a homocercal tail in teleost fishes.
Collapse
Affiliation(s)
- Nicolás Cumplido
- Millennium Institute Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Gloria Arratia
- University of Kansas, Department of Ecology and Evolutionary Biology and Biodiversity Institute, Lawrence, KS, USA
| | - Thomas Desvignes
- Institute of Neuroscience, University of Oregon, Eugene, OR, USA
| | - Salomé Muñoz-Sánchez
- Millennium Institute Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | | | - Miguel L. Allende
- Millennium Institute Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| |
Collapse
|
10
|
Abstract
Organismal development requires the reproducible unfolding of an ordered sequence of discrete steps (cell fate determination, migration, tissue folding, etc.) in both time and space. Here, we review the mechanisms that grant temporal specificity to developmental steps, including molecular clocks and timers. Individual timing mechanisms must be coordinated with each other to maintain the overall developmental sequence. However, phenotypic novelties can also arise through the modification of temporal patterns over the course of evolution. Two main types of variation in temporal patterning characterize interspecies differences in developmental time: allochrony, where the overall developmental sequence is either accelerated or slowed down while maintaining the relative duration of individual steps, and heterochrony, where the duration of specific developmental steps is altered relative to the rest. New advances in in vitro modeling of mammalian development using stem cells have recently enabled the revival of mechanistic studies of allochrony and heterochrony. In both cases, differences in the rate of basic cellular functions such as splicing, translation, protein degradation, and metabolism seem to underlie differences in developmental time. In the coming years, these studies should identify the genetic differences that drive divergence in developmental time between species.
Collapse
Affiliation(s)
- Margarete Diaz-Cuadros
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts, USA;
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA;
| | - Olivier Pourquié
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA;
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
11
|
Yang Y, Xu X, He B, Chang J, Zheng Y, Li Y. The role of miRNA-26a-5p and target gene socs1a in flutolanil induced hepatotoxicity of zebrafish at environmental relevant levels. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 335:122322. [PMID: 37544405 DOI: 10.1016/j.envpol.2023.122322] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 07/11/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
Flutolanil has been detected worldwide in aquatic environment and fish, which has become an undeniable stressor on ecosystem and human health. Flutolanil has been reported to be toxic to aquatic organisms. However, the pathophysiological and molecular mechanism behind the detrimental effects remains obscure. Here we reported hepatotoxicity induced by flutolanil in HepG2 cells and zebrafish, as revealed by toxicokinetic, HE staining, miRNAs-mRNAs sequencing, molecular dynamic simulations and dual luciferase reporter assays. Collectively, our results indicated that flutolanil could be absorbed by and accumulated in the liver of zebrafish, causing hepatic vacuolar degeneration, steatosis and nuclear condensation and abnormal liver function, where its exposure at environmental levels disrupted the expressions of miRNA-26a-5p and its target gene socs1a by mediating JAK-STAT signaling pathway, which was partially responsible for hepatotoxicity, correlated with oxidative stress, cell apoptosis, inflammation, cell cycle disorder and mitochondrial dysfunction. These findings suggest that miRNA-26a-5p/socs1a can serve as potential biomarkers of hepatotoxicity in zebrafish following exposure to flutolanil. This uncovered route will provide a new tool for the risk assessment of flutolanil and a guide to proper use of flutolanil and environmental remedy, and open up a new horizon for liver disease assessment.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory for Biology of Plant Disease and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, People's Republic of China
| | - Xiyan Xu
- State Key Laboratory for Biology of Plant Disease and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, People's Republic of China; College of Plant Health and Medicine, Shandong Engineering Research Center for Environment-Friendly Agricultural Pest Management, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Bin He
- Institute of Animal Husbandry and Veterinary, Wuhan Academy of Agricultural Sciences, Wuhan, 430070, People's Republic of China
| | - Jinhe Chang
- State Key Laboratory for Biology of Plant Disease and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, People's Republic of China
| | - Yongquan Zheng
- College of Plant Health and Medicine, Shandong Engineering Research Center for Environment-Friendly Agricultural Pest Management, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Yuanbo Li
- State Key Laboratory for Biology of Plant Disease and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, People's Republic of China.
| |
Collapse
|
12
|
Riesco MF, Valcarce DG, Sellés-Egea A, Esteve-Codina A, Herráez MP, Robles V. miR-29a Is Downregulated in Progenies Derived from Chronically Stressed Males. Int J Mol Sci 2023; 24:14107. [PMID: 37762407 PMCID: PMC10531283 DOI: 10.3390/ijms241814107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/24/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Recent research has provided compelling evidence demonstrating that paternal exposure to different stressors can influence their offspring's phenotypes. We hypothesized that paternal stress can negatively impact the progeny, altering different miRs and triggering different physiological alterations that could compromise offspring development. To investigate this, we exposed zebrafish male siblings to a chronic stress protocol for 21 days. We performed RNA-sequencing (RNA-seq) analyses to identify differentially expressed small noncoding RNAs in 7-day postfertilization (dpf) larvae derived from paternally stressed males crossed with control females compared with the control progeny. We found a single miRNA differentially expressed-miR-29a-which was validated in larva and was also tested in the sperm, testicles, and brain of the stressed progenitors. We observed a vertical transmission of chronic stress to the unexposed larvae, reporting novel consequences of paternally inherited chronic stress at a molecular level. The deregulation of mi-R29a in those larvae could affect relevant biological processes affecting development, morphogenesis, or neurogenesis, among others. Additionally, these disruptions were associated with reduced rates of survival and hatching in the affected offspring.
Collapse
Affiliation(s)
- Marta F. Riesco
- INDEGSAL, Cell Biology Area, Molecular Biology Department, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - David G. Valcarce
- INDEGSAL, Cell Biology Area, Molecular Biology Department, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Alba Sellés-Egea
- INDEGSAL, Cell Biology Area, Molecular Biology Department, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Anna Esteve-Codina
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Department of Medicine and Health Sciences, Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain
| | - María Paz Herráez
- INDEGSAL, Cell Biology Area, Molecular Biology Department, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Vanesa Robles
- INDEGSAL, Cell Biology Area, Molecular Biology Department, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| |
Collapse
|
13
|
Grodstein J, McMillen P, Levin M. Closing the loop on morphogenesis: a mathematical model of morphogenesis by closed-loop reaction-diffusion. Front Cell Dev Biol 2023; 11:1087650. [PMID: 37645245 PMCID: PMC10461482 DOI: 10.3389/fcell.2023.1087650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 07/31/2023] [Indexed: 08/31/2023] Open
Abstract
Morphogenesis, the establishment and repair of emergent complex anatomy by groups of cells, is a fascinating and biomedically-relevant problem. One of its most fascinating aspects is that a developing embryo can reliably recover from disturbances, such as splitting into twins. While this reliability implies some type of goal-seeking error minimization over a morphogenic field, there are many gaps with respect to detailed, constructive models of such a process. A common way to achieve reliability is negative feedback, which requires characterizing the existing body shape to create an error signal-but measuring properties of a shape may not be simple. We show how cells communicating in a wave-like pattern could analyze properties of the current body shape. We then describe a closed-loop negative-feedback system for creating reaction-diffusion (RD) patterns with high reliability. Specifically, we use a wave to count the number of peaks in a RD pattern, letting us use a negative-feedback controller to create a pattern with N repetitions, where N can be altered over a wide range. Furthermore, the individual repetitions of the RD pattern can be easily stretched or shrunk under genetic control to create, e.g., some morphological features larger than others. This work contributes to the exciting effort of understanding design principles of morphological computation, which can be used to understand evolved developmental mechanisms, manipulate them in regenerative-medicine settings, or engineer novel synthetic morphology constructs with desired robust behavior.
Collapse
Affiliation(s)
- Joel Grodstein
- Department of Electrical and Computer Engineering, Tufts University, Medford, MA, United States
| | - Patrick McMillen
- Allen Discovery Center at Tufts University, Medford, MA, United States
| | - Michael Levin
- Allen Discovery Center at Tufts University, Medford, MA, United States
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, United States
| |
Collapse
|
14
|
Kukreja K, Patel N, Megason SG, Klein AM. Global decoupling of cell differentiation from cell division in early embryo development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.29.551123. [PMID: 37546736 PMCID: PMC10402169 DOI: 10.1101/2023.07.29.551123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
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. 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 during initial cell type differentiation from early gastrulation to the end of segmentation. In the absence of cell division, differentiation slows down in some cell types, and cells exhibit global stress responses. While differentiation is robust to blocking cell division, the proportions of cells across cell states are not. This work simplifies 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
| | - Nikit Patel
- Department of Systems Biology, Harvard Medical School, 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
|
15
|
Levin M. Darwin's agential materials: evolutionary implications of multiscale competency in developmental biology. Cell Mol Life Sci 2023; 80:142. [PMID: 37156924 PMCID: PMC10167196 DOI: 10.1007/s00018-023-04790-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/24/2023] [Accepted: 04/27/2023] [Indexed: 05/10/2023]
Abstract
A critical aspect of evolution is the layer of developmental physiology that operates between the genotype and the anatomical phenotype. While much work has addressed the evolution of developmental mechanisms and the evolvability of specific genetic architectures with emergent complexity, one aspect has not been sufficiently explored: the implications of morphogenetic problem-solving competencies for the evolutionary process itself. The cells that evolution works with are not passive components: rather, they have numerous capabilities for behavior because they derive from ancestral unicellular organisms with rich repertoires. In multicellular organisms, these capabilities must be tamed, and can be exploited, by the evolutionary process. Specifically, biological structures have a multiscale competency architecture where cells, tissues, and organs exhibit regulative plasticity-the ability to adjust to perturbations such as external injury or internal modifications and still accomplish specific adaptive tasks across metabolic, transcriptional, physiological, and anatomical problem spaces. Here, I review examples illustrating how physiological circuits guiding cellular collective behavior impart computational properties to the agential material that serves as substrate for the evolutionary process. I then explore the ways in which the collective intelligence of cells during morphogenesis affect evolution, providing a new perspective on the evolutionary search process. This key feature of the physiological software of life helps explain the remarkable speed and robustness of biological evolution, and sheds new light on the relationship between genomes and functional anatomical phenotypes.
Collapse
Affiliation(s)
- Michael Levin
- Allen Discovery Center at Tufts University, 200 Boston Ave. 334 Research East, Medford, MA, 02155, USA.
- Wyss Institute for Biologically Inspired Engineering at Harvard University, 3 Blackfan St., Boston, MA, 02115, USA.
| |
Collapse
|
16
|
Uriu K, Morelli LG. Orchestration of tissue shape changes and gene expression patterns in development. Semin Cell Dev Biol 2023; 147:24-33. [PMID: 36631335 DOI: 10.1016/j.semcdb.2022.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/27/2022] [Accepted: 12/27/2022] [Indexed: 01/11/2023]
Abstract
In development, tissue shape changes and gene expression patterns give rise to morphogenesis. Understanding tissue shape changes requires the analysis of mechanical properties of the tissue such as tissue rigidity, cell influx from neighboring tissues, cell shape changes and cell proliferation. Local and global gene expression patterns can be influenced by neighbor exchange and tissue shape changes. Here we review recent studies on the mechanisms for tissue elongation and its influences on dynamic gene expression patterns by focusing on vertebrate somitogenesis. We first introduce mechanical and biochemical properties of the segmenting tissue that drive tissue elongation. Then, we discuss patterning in the presence of cell mixing, scaling of signaling gradients, and dynamic phase waves of rhythmic gene expression under tissue shape changes. We also highlight the importance of theoretical approaches to address the relation between tissue shape changes and patterning.
Collapse
Affiliation(s)
- Koichiro Uriu
- Graduate School of Natural Science and Technology, Kanazawa University, Kakuma-machi, Kanazawa 920-1192 Japan.
| | - Luis G Morelli
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET-Partner Institute of the Max Planck Society, Polo Científico Tecnológico, Godoy Cruz 2390, C1425FQD, Buenos Aires, Argentina; Departamento de Física, FCEyN UBA, Ciudad Universitaria, 1428 Buenos Aires, Argentina.
| |
Collapse
|
17
|
Toh K, Saunders D, Verd B, Steventon B. Zebrafish neuromesodermal progenitors undergo a critical state transition in vivo. iScience 2022; 25:105216. [PMID: 36274939 PMCID: PMC9579027 DOI: 10.1016/j.isci.2022.105216] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 08/05/2022] [Accepted: 09/22/2022] [Indexed: 11/30/2022] Open
Abstract
The transition state model of cell differentiation proposes that a transient window of gene expression stochasticity precedes entry into a differentiated state. Here, we assess this theoretical model in zebrafish neuromesodermal progenitors (NMps) in vivo during late somitogenesis stages. We observed an increase in gene expression variability at the 24 somite stage (24ss) before their differentiation into spinal cord and paraxial mesoderm. Analysis of a published 18ss scRNA-seq dataset showed that the NMp population is noisier than its derivatives. By building in silico composite gene expression maps from image data, we assigned an 'NM index' to in silico NMps based on the expression of neural and mesodermal markers and demonstrated that cell population heterogeneity peaked at 24ss. Further examination revealed cells with gene expression profiles incongruent with their prospective fate. Taken together, our work supports the transition state model within an endogenous cell fate decision making event.
Collapse
Affiliation(s)
- Kane Toh
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | - Dillan Saunders
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | - Berta Verd
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
- Department of Zoology, University of Oxford, Oxford OX1 3SZ, UK
| | | |
Collapse
|
18
|
Martin BL, Steventon B. A fishy tail: Insights into the cell and molecular biology of neuromesodermal cells from zebrafish embryos. Dev Biol 2022; 487:67-73. [PMID: 35525020 DOI: 10.1016/j.ydbio.2022.04.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/29/2022] [Accepted: 04/26/2022] [Indexed: 11/03/2022]
Abstract
Vertebrate embryos establish their primary body axis in a conserved progressive fashion from the anterior to the posterior. During this process, a posteriorly localized neuromesodermal cell population called neuromesodermal progenitors (NMps) plays a critical role in contributing new cells to the spinal cord and mesoderm as the embryo elongates. Defects in neuromesodermal population development can cause severe disruptions to the formation of the body posterior to the head. Given their importance during development and their potential, some of which has already been realized, for revealing new methods of in vitro tissue generation, there is great interest in better understanding NMp biology. The zebrafish model system has been instrumental in advancing our understanding of the molecular and cellular attributes of the NM cell population and its derivatives. In this review, we focus on our current understanding of the zebrafish NM population and its contribution to body axis formation, with particular emphasis on the lineage potency, morphogenesis, and niche factors that promote or inhibit differentiation.
Collapse
Affiliation(s)
- Benjamin L Martin
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, 11794-5215, USA.
| | - Benjamin Steventon
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
19
|
Thomson L, Muresan L, Steventon B. The zebrafish presomitic mesoderm elongates through compaction-extension. Cells Dev 2021. [PMID: 34597846 DOI: 10.1101/2021.03.11.434927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
In vertebrate embryos the presomitic mesoderm becomes progressively segmented into somites at the anterior end while extending along the anterior-posterior axis. A commonly adopted model to explain how this tissue elongates is that of posterior growth, driven in part by the addition of new cells from uncommitted progenitor populations in the tailbud. However, in zebrafish, much of somitogenesis is associated with an absence of overall volume increase, and posterior progenitors do not contribute new cells until the final stages of somitogenesis. Here, we perform a comprehensive 3D morphometric analysis of the paraxial mesoderm and reveal that extension is linked to a volumetric decrease and an increase in cell density. We also find that individual cells decrease in volume over successive somite stages. Live cell tracking confirms that much of this tissue deformation occurs within the presomitic mesoderm progenitor zone and is associated with non-directional rearrangement. Taken together, we propose a compaction-extension mechanism of tissue elongation that highlights the need to better understand the role tissue intrinsic and extrinsic forces in regulating morphogenesis.
Collapse
Affiliation(s)
- Lewis Thomson
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | - Leila Muresan
- Cambridge Advanced Imaging Centre, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
20
|
Cerveny KL, Bronstein H, Hagen O, Lamb DB, Martin G, Tower I, Van Duzer A, Welch E, Varga M. Mutations linked to loss of cell cycle control can render cells responsive to local differentiation cues. MICROPUBLICATION BIOLOGY 2021; 2021. [PMID: 34723143 PMCID: PMC8553407 DOI: 10.17912/micropub.biology.000481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 11/22/2022]
Abstract
Cell behaviors such as survival, proliferation, and death are governed by a multitude of cues, both intrinsic and extrinsic. To test whether a wild-type environment could encourage the survival and/or differentiation of neuronal progenitor cells with impaired cell cycle progression, we transplanted cells from cdk1, dtl, slbp, fbxo5, ahctf1, gins2, hdac1, mcm5, ssrp1a, and rbbp6 mutant zebrafish embryos into wild-type embryos, creating chimeric zebrafish with mutant cells in the developing eye. We found that when cells from cdk1, dtl, slbp, gins2, mcm5, or rbbp6 mutants were transplanted into wild-type hosts, survival and/or differentiation was almost always compromised in a manner consistent with cell-autonomous cell death. Interestingly, we observed that fbxo5, ahctf1, hdac1, or ssrp1a mutant cells survived and sometimes exhibited signs of differentiation when grafted into wild-type eyes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Máté Varga
- Department of Genetics, ELTE Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
21
|
Mofatteh M, Echegaray-Iturra F, Alamban A, Dalla Ricca F, Bakshi A, Aydogan MG. Autonomous clocks that regulate organelle biogenesis, cytoskeletal organization, and intracellular dynamics. eLife 2021; 10:e72104. [PMID: 34586070 PMCID: PMC8480978 DOI: 10.7554/elife.72104] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/14/2021] [Indexed: 12/27/2022] Open
Abstract
How do cells perceive time? Do cells use temporal information to regulate the production/degradation of their enzymes, membranes, and organelles? Does controlling biological time influence cytoskeletal organization and cellular architecture in ways that confer evolutionary and physiological advantages? Potential answers to these fundamental questions of cell biology have historically revolved around the discussion of 'master' temporal programs, such as the principal cyclin-dependent kinase/cyclin cell division oscillator and the circadian clock. In this review, we provide an overview of the recent evidence supporting an emerging concept of 'autonomous clocks,' which under normal conditions can be entrained by the cell cycle and/or the circadian clock to run at their pace, but can also run independently to serve their functions if/when these major temporal programs are halted/abrupted. We begin the discussion by introducing recent developments in the study of such clocks and their roles at different scales and complexities. We then use current advances to elucidate the logic and molecular architecture of temporal networks that comprise autonomous clocks, providing important clues as to how these clocks may have evolved to run independently and, sometimes at the cost of redundancy, have strongly coupled to run under the full command of the cell cycle and/or the circadian clock. Next, we review a list of important recent findings that have shed new light onto potential hallmarks of autonomous clocks, suggestive of prospective theoretical and experimental approaches to further accelerate their discovery. Finally, we discuss their roles in health and disease, as well as possible therapeutic opportunities that targeting the autonomous clocks may offer.
Collapse
Affiliation(s)
- Mohammad Mofatteh
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Fabio Echegaray-Iturra
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Andrew Alamban
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Francesco Dalla Ricca
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Anand Bakshi
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Mustafa G Aydogan
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
22
|
The zebrafish presomitic mesoderm elongates through compaction-extension. Cells Dev 2021; 168:203748. [PMID: 34597846 PMCID: PMC7612712 DOI: 10.1016/j.cdev.2021.203748] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 09/21/2021] [Accepted: 09/21/2021] [Indexed: 11/21/2022]
Abstract
In vertebrate embryos the presomitic mesoderm becomes progressively segmented into somites at the anterior end while extending along the anterior-posterior axis. A commonly adopted model to explain how this tissue elongates is that of posterior growth, driven in part by the addition of new cells from uncommitted progenitor populations in the tailbud. However, in zebrafish, much of somitogenesis is associated with an absence of overall volume increase, and posterior progenitors do not contribute new cells until the final stages of somitogenesis. Here, we perform a comprehensive 3D morphometric analysis of the paraxial mesoderm and reveal that extension is linked to a volumetric decrease and an increase in cell density. We also find that individual cells decrease in volume over successive somite stages. Live cell tracking confirms that much of this tissue deformation occurs within the presomitic mesoderm progenitor zone and is associated with non-directional rearrangement. Taken together, we propose a compaction-extension mechanism of tissue elongation that highlights the need to better understand the role tissue intrinsic and extrinsic forces in regulating morphogenesis.
Collapse
|
23
|
Abstract
Arthropod segmentation and vertebrate somitogenesis are leading fields in the experimental and theoretical interrogation of developmental patterning. However, despite the sophistication of current research, basic conceptual issues remain unresolved. These include: (i) the mechanistic origins of spatial organization within the segment addition zone (SAZ); (ii) the mechanistic origins of segment polarization; (iii) the mechanistic origins of axial variation; and (iv) the evolutionary origins of simultaneous patterning. Here, I explore these problems using coarse-grained models of cross-regulating dynamical processes. In the morphogenetic framework of a row of cells undergoing axial elongation, I simulate interactions between an 'oscillator', a 'switch' and up to three 'timers', successfully reproducing essential patterning behaviours of segmenting systems. By comparing the output of these largely cell-autonomous models to variants that incorporate positional information, I find that scaling relationships, wave patterns and patterning dynamics all depend on whether the SAZ is regulated by temporal or spatial information. I also identify three mechanisms for polarizing oscillator output, all of which functionally implicate the oscillator frequency profile. Finally, I demonstrate significant dynamical and regulatory continuity between sequential and simultaneous modes of segmentation. I discuss these results in the context of the experimental literature.
Collapse
Affiliation(s)
- Erik Clark
- Department of Systems Biology, Harvard Medical School, 210 Longwood Ave, Boston, MA 02115, USA
- Trinity College Cambridge, University of Cambridge, Trinity Street, Cambridge CB2 1TQ, UK
| |
Collapse
|
24
|
Banavar SP, Carn EK, Rowghanian P, Stooke-Vaughan G, Kim S, Campàs O. Mechanical control of tissue shape and morphogenetic flows during vertebrate body axis elongation. Sci Rep 2021; 11:8591. [PMID: 33883563 PMCID: PMC8060277 DOI: 10.1038/s41598-021-87672-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 03/30/2021] [Indexed: 02/02/2023] Open
Abstract
Shaping embryonic tissues into their functional morphologies requires cells to control the physical state of the tissue in space and time. While regional variations in cellular forces or cell proliferation have been typically assumed to be the main physical factors controlling tissue morphogenesis, recent experiments have revealed that spatial variations in the tissue physical (fluid/solid) state play a key role in shaping embryonic tissues. Here we theoretically study how the regional control of fluid and solid tissue states guides morphogenetic flows to shape the extending vertebrate body axis. Our results show that both the existence of a fluid-to-solid tissue transition along the anteroposterior axis and the tissue surface tension determine the shape of the tissue and its ability to elongate unidirectionally, with large tissue tensions preventing unidirectional elongation and promoting blob-like tissue expansions. We predict both the tissue morphogenetic flows and stresses that enable unidirectional axis elongation. Our results show the existence of a sharp transition in the structure of morphogenetic flows, from a flow with no vortices to a flow with two counter-rotating vortices, caused by a transition in the number and location of topological defects in the flow field. Finally, comparing the theoretical predictions to quantitative measurements of both tissue flows and shape during zebrafish body axis elongation, we show that the observed morphogenetic events can be explained by the existence of a fluid-to-solid tissue transition along the anteroposterior axis. These results highlight the role of spatiotemporally-controlled fluid-to-solid transitions in the tissue state as a physical mechanism of embryonic morphogenesis.
Collapse
Affiliation(s)
- Samhita P Banavar
- Department of Physics, University of California, Santa Barbara, CA, 93106, USA
- California NanoSystems Institute, University of California, Santa Barbara, CA, 93106, USA
- Stanford University, Stanford, CA, USA
| | - Emmet K Carn
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, 93106, USA
- California NanoSystems Institute, University of California, Santa Barbara, CA, 93106, USA
| | - Payam Rowghanian
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, 93106, USA
- California NanoSystems Institute, University of California, Santa Barbara, CA, 93106, USA
| | - Georgina Stooke-Vaughan
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, 93106, USA
- California NanoSystems Institute, University of California, Santa Barbara, CA, 93106, USA
| | - Sangwoo Kim
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, 93106, USA
- California NanoSystems Institute, University of California, Santa Barbara, CA, 93106, USA
| | - Otger Campàs
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, 93106, USA.
- California NanoSystems Institute, University of California, Santa Barbara, CA, 93106, USA.
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Barbara, CA, 93106, USA.
- Center for Bioengineering, University of California, Santa Barbara, CA, 93106, USA.
- Cluster of Excellence Physics of Life, TU Dresden, 01062, Dresden, Germany.
| |
Collapse
|
25
|
Uriu K, Liao BK, Oates AC, Morelli LG. From local resynchronization to global pattern recovery in the zebrafish segmentation clock. eLife 2021; 10:61358. [PMID: 33587039 PMCID: PMC7984840 DOI: 10.7554/elife.61358] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 01/27/2021] [Indexed: 01/26/2023] Open
Abstract
Integrity of rhythmic spatial gene expression patterns in the vertebrate segmentation clock requires local synchronization between neighboring cells by Delta-Notch signaling and its inhibition causes defective segment boundaries. Whether deformation of the oscillating tissue complements local synchronization during patterning and segment formation is not understood. We combine theory and experiment to investigate this question in the zebrafish segmentation clock. We remove a Notch inhibitor, allowing resynchronization, and analyze embryonic segment recovery. We observe unexpected intermingling of normal and defective segments, and capture this with a new model combining coupled oscillators and tissue mechanics. Intermingled segments are explained in the theory by advection of persistent phase vortices of oscillators. Experimentally observed changes in recovery patterns are predicted in the theory by temporal changes in tissue length and cell advection pattern. Thus, segmental pattern recovery occurs at two length and time scales: rapid local synchronization between neighboring cells, and the slower transport of the resulting patterns across the tissue through morphogenesis.
Collapse
Affiliation(s)
- Koichiro Uriu
- Graduate School of Natural Science and Technology, Kanazawa University, Kakuma-machi, Kanazawa, Japan
| | - Bo-Kai Liao
- Department of Aquaculture, National Taiwan Ocean University, Keelung, Taiwan.,Department of Cell and Developmental Biology, University College London, Gower Street, London, United Kingdom.,The Francis Crick Institute, London, United Kingdom.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Andrew C Oates
- Department of Cell and Developmental Biology, University College London, Gower Street, London, United Kingdom.,The Francis Crick Institute, London, United Kingdom.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Institute of Bioengineering, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Luis G Morelli
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Polo Científico Tecnológico, Buenos Aires, Argentina.,Departamento de Física, FCEyN UBA, Ciudad Universitaria, Buenos Aires, Argentina.,Max Planck Institute for Molecular Physiology, Department of Systemic Cell Biology, Dortmund, Germany
| |
Collapse
|
26
|
Sambasivan R, Steventon B. Neuromesodermal Progenitors: A Basis for Robust Axial Patterning in Development and Evolution. Front Cell Dev Biol 2021; 8:607516. [PMID: 33520989 PMCID: PMC7843932 DOI: 10.3389/fcell.2020.607516] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/14/2020] [Indexed: 12/24/2022] Open
Abstract
During early development the vertebrate embryo elongates through a combination of tissue shape change, growth and progenitor cell expansion across multiple regions of the body axis. How these events are coordinated across the length of the embryo to generate a well-proportioned body axis is unknown. Understanding the multi-tissue interplay of morphogenesis, growth and cell fate specification is essential for us to gain a complete understanding how diverse body plans have evolved in a robust manner. Within the posterior region of the embryo, a population of bipotent neuromesodermal progenitors generate both spinal cord and paraxial mesoderm derivatives during the elongation of the vertebrate body. Here we summarize recent data comparing neuromesodermal lineage and their underlying gene-regulatory networks between species and through development. We find that the common characteristic underlying this population is a competence to generate posterior neural and paraxial mesoderm cells, with a conserved Wnt/FGF and Sox2/T/Tbx6 regulatory network. We propose the hypothesis that by maintaining a population of multi-germ layer competent progenitors at the posterior aspect of the embryo, a flexible pool of progenitors is maintained whose contribution to the elongating body axis varies as a consequence of the relative growth rates occurring within anterior and posterior regions of the body axis. We discuss how this capacity for variation in the proportions and rates of NM specification might have been important allowing for alterations in the timing of embryo growth during evolution.
Collapse
Affiliation(s)
- Ramkumar Sambasivan
- Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, India
| | - Benjamin Steventon
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
27
|
Dynamic optima in cell sizes during early development enable normal gastrulation in zebrafish embryos. Dev Biol 2020; 468:26-40. [DOI: 10.1016/j.ydbio.2020.09.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 11/21/2022]
|
28
|
Nerli E, Rocha-Martins M, Norden C. Asymmetric neurogenic commitment of retinal progenitors involves Notch through the endocytic pathway. eLife 2020; 9:e60462. [PMID: 33141024 PMCID: PMC7679139 DOI: 10.7554/elife.60462] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 11/02/2020] [Indexed: 01/07/2023] Open
Abstract
During brain development, progenitor cells need to balanceproliferation and differentiation in order to generate different neurons in the correct numbers and proportions. Currently, the patterns of multipotent progenitor divisions that lead to neurogenic entry and the factors that regulate them are not fully understood. We here use the zebrafish retina to address this gap, exploiting its suitability for quantitative live-imaging. We show that early neurogenic progenitors arise from asymmetric divisions. Notch regulates this asymmetry, as when inhibited, symmetric divisions producing two neurogenic progenitors occur. Surprisingly however, Notch does not act through an apicobasal activity gradient as previously suggested, but through asymmetric inheritance of Sara-positive endosomes. Further, the resulting neurogenic progenitors show cell biological features different from multipotent progenitors, raising the possibility that an intermediate progenitor state exists in the retina. Our study thus reveals new insights into the regulation of proliferative and differentiative events during central nervous system development.
Collapse
Affiliation(s)
- Elisa Nerli
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Instituto Gulbenkian de CiênciaOeirasPortugal
| | - Mauricio Rocha-Martins
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Instituto Gulbenkian de CiênciaOeirasPortugal
| | - Caren Norden
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Instituto Gulbenkian de CiênciaOeirasPortugal
| |
Collapse
|
29
|
Levin M. The Biophysics of Regenerative Repair Suggests New Perspectives on Biological Causation. Bioessays 2020; 42:e1900146. [PMID: 31994772 DOI: 10.1002/bies.201900146] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/03/2019] [Indexed: 12/13/2022]
Abstract
Evolution exploits the physics of non-neural bioelectricity to implement anatomical homeostasis: a process in which embryonic patterning, remodeling, and regeneration achieve invariant anatomical outcomes despite external interventions. Linear "developmental pathways" are often inadequate explanations for dynamic large-scale pattern regulation, even when they accurately capture relationships between molecular components. Biophysical and computational aspects of collective cell activity toward a target morphology reveal interesting aspects of causation in biology. This is critical not only for unraveling evolutionary and developmental events, but also for the design of effective strategies for biomedical intervention. Bioelectrical controls of growth and form, including stochastic behavior in such circuits, highlight the need for the formulation of nuanced views of pathways, drivers of system-level outcomes, and modularity, borrowing from concepts in related disciplines such as cybernetics, control theory, computational neuroscience, and information theory. This approach has numerous practical implications for basic research and for applications in regenerative medicine and synthetic bioengineering.
Collapse
Affiliation(s)
- Michael Levin
- Allen Discovery Center at Tufts University, Medford, MA, 02155, USA.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| |
Collapse
|
30
|
Gawne R, McKenna KZ, Levin M. Competitive and Coordinative Interactions between Body Parts Produce Adaptive Developmental Outcomes. Bioessays 2020; 42:e1900245. [DOI: 10.1002/bies.201900245] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Indexed: 01/25/2023]
Affiliation(s)
- Richard Gawne
- Allen Discovery Center at Tufts University Medford MA 02155
| | - Kenneth Z. McKenna
- Division of Biological SciencesSection of Cellular and Developmental BiologyUniversity of California San Diego La Jolla CA 92093
| | - Michael Levin
- Allen Discovery Center at Tufts University Medford MA 02155
| |
Collapse
|
31
|
Abstract
Gastrulation is a critical early morphogenetic process of animal development, during which the three germ layers; mesoderm, endoderm and ectoderm, are rearranged by internalization movements. Concurrent epiboly movements spread and thin the germ layers while convergence and extension movements shape them into an anteroposteriorly elongated body with head, trunk, tail and organ rudiments. In zebrafish, gastrulation follows the proliferative and inductive events that establish the embryonic and extraembryonic tissues and the embryonic axis. Specification of these tissues and embryonic axes are controlled by the maternal gene products deposited in the egg. These early maternally controlled processes need to generate sufficient cell numbers and establish the embryonic polarity to ensure normal gastrulation. Subsequently, after activation of the zygotic genome, the zygotic gene products govern mesoderm and endoderm induction and germ layer patterning. Gastrulation is initiated during the maternal-to-zygotic transition, a process that entails both activation of the zygotic genome and downregulation of the maternal transcripts. Genomic studies indicate that gastrulation is largely controlled by the zygotic genome. Nonetheless, genetic studies that investigate the relative contributions of maternal and zygotic gene function by comparing zygotic, maternal and maternal zygotic mutant phenotypes, reveal significant contribution of maternal gene products, transcripts and/or proteins, that persist through gastrulation, to the control of gastrulation movements. Therefore, in zebrafish, the maternally expressed gene products not only set the stage for, but they also actively participate in gastrulation morphogenesis.
Collapse
Affiliation(s)
- Lilianna Solnica-Krezel
- Department of Developmental Biology and Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, United States.
| |
Collapse
|
32
|
Schauer A, Pinheiro D, Hauschild R, Heisenberg CP. Zebrafish embryonic explants undergo genetically encoded self-assembly. eLife 2020; 9:55190. [PMID: 32250246 PMCID: PMC7190352 DOI: 10.7554/elife.55190] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/05/2020] [Indexed: 12/20/2022] Open
Abstract
Embryonic stem cell cultures are thought to self-organize into embryoid bodies, able to undergo symmetry-breaking, germ layer specification and even morphogenesis. Yet, it is unclear how to reconcile this remarkable self-organization capacity with classical experiments demonstrating key roles for extrinsic biases by maternal factors and/or extraembryonic tissues in embryogenesis. Here, we show that zebrafish embryonic tissue explants, prepared prior to germ layer induction and lacking extraembryonic tissues, can specify all germ layers and form a seemingly complete mesendoderm anlage. Importantly, explant organization requires polarized inheritance of maternal factors from dorsal-marginal regions of the blastoderm. Moreover, induction of endoderm and head-mesoderm, which require peak Nodal-signaling levels, is highly variable in explants, reminiscent of embryos with reduced Nodal signals from the extraembryonic tissues. Together, these data suggest that zebrafish explants do not undergo bona fide self-organization, but rather display features of genetically encoded self-assembly, where intrinsic genetic programs control the emergence of order.
Collapse
|
33
|
Liu B, Großhans J. The role of dNTP metabolites in control of the embryonic cell cycle. Cell Cycle 2019; 18:2817-2827. [PMID: 31544596 PMCID: PMC6791698 DOI: 10.1080/15384101.2019.1665948] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/03/2019] [Accepted: 09/06/2019] [Indexed: 01/06/2023] Open
Abstract
Deoxyribonucleotide metabolites (dNTPs) are the substrates for DNA synthesis. It has been proposed that their availability influences the progression of the cell cycle during development and pathological situations such as tumor growth. The mechanism has remained unclear for the link between cell cycle and dNTP levels beyond their role as substrates. Here, we review recent studies concerned with the dynamics of dNTP levels in early embryos and the role of DNA replication checkpoint as a sensor of dNTP levels.
Collapse
Affiliation(s)
- Boyang Liu
- Institut für Entwicklungsbiochemie, Universitätsmedizin, Georg-August-Universität, Göttingen, Germany
| | - Jörg Großhans
- Institut für Entwicklungsbiochemie, Universitätsmedizin, Georg-August-Universität, Göttingen, Germany
- Entwicklungsgenetik, Fachbereich Biologie, Philipps-Universität, Marburg, Germany
| |
Collapse
|
34
|
Mongera A, Michaut A, Guillot C, Xiong F, Pourquié O. Mechanics of Anteroposterior Axis Formation in Vertebrates. Annu Rev Cell Dev Biol 2019; 35:259-283. [PMID: 31412208 PMCID: PMC7394480 DOI: 10.1146/annurev-cellbio-100818-125436] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The vertebrate anteroposterior axis forms through elongation of multiple tissues during embryogenesis. This process is based on tissue-autonomous mechanisms of force generation and intertissue mechanical coupling whose failure leads to severe developmental anomalies such as body truncation and spina bifida. Similar to other morphogenetic modules, anteroposterior body extension requires both the rearrangement of existing materials-such as cells and extracellular matrix-and the local addition of new materials, i.e., anisotropic growth, through cell proliferation, cell growth, and matrix deposition. Numerous signaling pathways coordinate body axis formation via regulation of cell behavior during tissue rearrangements and/or volumetric growth. From a physical perspective, morphogenesis depends on both cell-generated forces and tissue material properties. As the spatiotemporal variation of these mechanical parameters has recently been explored in the context of vertebrate body elongation, the study of this process is likely to shed light on the cross talk between signaling and mechanics during morphogenesis.
Collapse
Affiliation(s)
- Alessandro Mongera
- Department of Genetics, Harvard Medical School, and Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA;
| | - Arthur Michaut
- Department of Genetics, Harvard Medical School, and Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA;
| | - Charlène Guillot
- Department of Genetics, Harvard Medical School, and Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA;
| | - Fengzhu Xiong
- Department of Genetics, Harvard Medical School, and Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA;
| | - Olivier Pourquié
- Department of Genetics, Harvard Medical School, and Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA;
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts 02138, USA
| |
Collapse
|
35
|
Hsu AY, Wang D, Liu S, Lu J, Syahirah R, Bennin DA, Huttenlocher A, Umulis DM, Wan J, Deng Q. Phenotypical microRNA screen reveals a noncanonical role of CDK2 in regulating neutrophil migration. Proc Natl Acad Sci U S A 2019; 116:18561-18570. [PMID: 31451657 PMCID: PMC6744913 DOI: 10.1073/pnas.1905221116] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Neutrophil migration is essential for inflammatory responses to kill pathogens; however, excessive neutrophilic inflammation also leads to tissue injury and adverse effects. To discover novel therapeutic targets that modulate neutrophil migration, we performed a neutrophil-specific microRNA (miRNA) overexpression screen in zebrafish and identified 8 miRNAs as potent suppressors of neutrophil migration. Among those, miR-199 decreases neutrophil chemotaxis in zebrafish and human neutrophil-like cells. Intriguingly, in terminally differentiated neutrophils, miR-199 alters the cell cycle-related pathways and directly suppresses cyclin-dependent kinase 2 (Cdk2), whose known activity is restricted to cell cycle progression and cell differentiation. Inhibiting Cdk2, but not DNA replication, disrupts cell polarity and chemotaxis of zebrafish neutrophils without inducing cell death. Human neutrophil-like cells deficient in CDK2 fail to polarize and display altered signaling downstream of the formyl peptide receptor. Chemotaxis of primary human neutrophils is also reduced upon CDK2 inhibition. Furthermore, miR-199 overexpression or CDK2 inhibition significantly improves the outcome of lethal systemic inflammation challenges in zebrafish. Our results therefore reveal previously unknown functions of miR-199 and CDK2 in regulating neutrophil migration and provide directions in alleviating systemic inflammation.
Collapse
Affiliation(s)
- Alan Y Hsu
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Decheng Wang
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
- The Institute of Infection and Inflammation, Medical College of China Three Gorges University, 443002 Yichang, Hubei, People's Republic of China
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202
- Collaborative Core for Cancer Bioinformatics, Indiana University Simon Cancer Center, Indianapolis, IN 46202
| | - Justice Lu
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Ramizah Syahirah
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - David A Bennin
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53706
| | - David M Umulis
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN 47907
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202
- Collaborative Core for Cancer Bioinformatics, Indiana University Simon Cancer Center, Indianapolis, IN 46202
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Qing Deng
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907;
- Purdue Institute for Inflammation, Immunology, & Infectious Disease, Purdue University, West Lafayette, IN 47907
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907
| |
Collapse
|
36
|
Casar Tena T, Maerz LD, Szafranski K, Groth M, Blätte TJ, Donow C, Matysik S, Walther P, Jeggo PA, Burkhalter MD, Philipp M. Resting cells rely on the DNA helicase component MCM2 to build cilia. Nucleic Acids Res 2019; 47:134-151. [PMID: 30329080 PMCID: PMC6326816 DOI: 10.1093/nar/gky945] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 10/04/2018] [Indexed: 12/24/2022] Open
Abstract
Minichromosome maintenance (MCM) proteins facilitate replication by licensing origins and unwinding the DNA double strand. Interestingly, the number of MCM hexamers greatly exceeds the number of firing origins suggesting additional roles of MCMs. Here we show a hitherto unanticipated function of MCM2 in cilia formation in human cells and zebrafish that is uncoupled from replication. Zebrafish depleted of MCM2 develop ciliopathy-phenotypes including microcephaly and aberrant heart looping due to malformed cilia. In non-cycling human fibroblasts, loss of MCM2 promotes transcription of a subset of genes, which cause cilia shortening and centriole overduplication. Chromatin immunoprecipitation experiments show that MCM2 binds to transcription start sites of cilia inhibiting genes. We propose that such binding may block RNA polymerase II-mediated transcription. Depletion of a second MCM (MCM7), which functions in complex with MCM2 during its canonical functions, reveals an overlapping cilia-deficiency phenotype likely unconnected to replication, although MCM7 appears to regulate a distinct subset of genes and pathways. Our data suggests that MCM2 and 7 exert a role in ciliogenesis in post-mitotic tissues.
Collapse
Affiliation(s)
- Teresa Casar Tena
- Institute of Biochemistry and Molecular Biology, Ulm University, 89081 Ulm, Germany
| | - Lars D Maerz
- Institute of Biochemistry and Molecular Biology, Ulm University, 89081 Ulm, Germany
| | - Karol Szafranski
- Leibniz Institute on Aging, Fritz Lipmann Institute, 07745 Jena, Germany
| | - Marco Groth
- Leibniz Institute on Aging, Fritz Lipmann Institute, 07745 Jena, Germany
| | - Tamara J Blätte
- Institute of Biochemistry and Molecular Biology, Ulm University, 89081 Ulm, Germany
| | - Cornelia Donow
- Institute of Biochemistry and Molecular Biology, Ulm University, 89081 Ulm, Germany
| | - Sabrina Matysik
- Institute of Biochemistry and Molecular Biology, Ulm University, 89081 Ulm, Germany
| | - Paul Walther
- Central Facility for Electron Microscopy, Ulm University, 89081 Ulm, Germany
| | - Penelope A Jeggo
- Genome Damage and Stability Centre, University of Sussex, Brighton BN1 9RQ, UK
| | - Martin D Burkhalter
- Institute of Biochemistry and Molecular Biology, Ulm University, 89081 Ulm, Germany
| | - Melanie Philipp
- Institute of Biochemistry and Molecular Biology, Ulm University, 89081 Ulm, Germany
| |
Collapse
|
37
|
Petrachkova T, Wortinger LA, Bard AJ, Singh J, Warga RM, Kane DA. Lack of Cyclin B1 in zebrafish causes lengthening of G2 and M phases. Dev Biol 2019; 451:167-179. [PMID: 30930047 DOI: 10.1016/j.ydbio.2019.03.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 03/20/2019] [Accepted: 03/22/2019] [Indexed: 12/23/2022]
Abstract
An essential part of the Mitosis Promoting Factor, Cyclin B1 is indispensable for cells to enter mitosis. We report here that the zebrafish early arrest mutant specter is a loss-of-function mutation in the сyclin B1 gene. cyclin B1 is maternally transcribed in zebrafish, and the zygotic phenotype is apparent by early segmentation. Lack of zygotic Cyclin B1 does not stop cells from dividing, rather it causes an abnormal and elongated progression through the G2 and M phases of the cell cycle. Many mutant cells show signs of chromosomal instability or enter apoptosis. Using CRISPR-mediated gene editing, we produced a more severe gain-of-function mutation confirming that specter is the result of nonfunctional Cyclin B1. Although also a recessive phenotype, this new mutation produces an alternative splice-form of cyclin B1 mRNA, whose product lacks several key components for Cyclin B1, but not the Cdk1-binding domain. This mutant form of Cyclin B1 completely prevents cell division. We conclude that, although Cyclin B1 is critical for cells to enter mitosis, another cell cycle protein may be cooperating with Cdk1 at the G2/M checkpoint to sustain a partly functional Mitosis Promoting Factor.
Collapse
Affiliation(s)
- Tetiana Petrachkova
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA.
| | - Laura A Wortinger
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA
| | - Amber J Bard
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA
| | - Jyotika Singh
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA
| | - Rachel M Warga
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA
| | - Donald A Kane
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA
| |
Collapse
|
38
|
Robinton DA, Chal J, Lummertz da Rocha E, Han A, Yermalovich AV, Oginuma M, Schlaeger TM, Sousa P, Rodriguez A, Urbach A, Pourquié O, Daley GQ. The Lin28/let-7 Pathway Regulates the Mammalian Caudal Body Axis Elongation Program. Dev Cell 2019; 48:396-405.e3. [DOI: 10.1016/j.devcel.2018.12.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 08/13/2018] [Accepted: 12/17/2018] [Indexed: 02/09/2023]
|
39
|
Maerz LD, Casar Tena T, Gerhards J, Donow C, Jeggo PA, Philipp M. Analysis of cilia dysfunction phenotypes in zebrafish embryos depleted of Origin recognition complex factors. Eur J Hum Genet 2019; 27:772-782. [PMID: 30696958 DOI: 10.1038/s41431-019-0338-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 12/26/2018] [Accepted: 01/04/2019] [Indexed: 12/15/2022] Open
Abstract
Meier-Gorlin syndrome (MGS) is a rare, congenital primordial microcephalic dwarfism disorder. MGS is caused by genetic variants of components of the origin recognition complex (ORC) consisting of ORC1-6 and the pre-replication complex, which together enable origin firing and hence genome replication. In addition, ORC1 has previously been shown to play a role in ciliogenesis. Here, we extend this work and investigate the function of ORC1 and two other members of the complex on cilia at an organismal level. Knockdown experiments in zebrafish confirmed the impact of ORC1 on cilia. ORC1-deficiency confers defects anticipated to arise from impaired cilia function such as formation of oedema, kidney cysts, curved bodies and left-right asymmetry defects. We found ORC1 furthermore required for cilium formation in zebrafish and demonstrate that ciliopathy phenotypes in ORC1-depleted zebrafish could not be rescued by reconstitution with ORC1 bearing a genetic variant previously identified in MGS patients. Loss-of-function of Orc4 and Orc6, respectively, conferred similar ciliopathy phenotypes and cilium shortening in zebrafish, suggesting that several, if not all, components of the ORC regulate ciliogenesis downstream to or in addition to their canonical function in replication initiation. This study presents the first in vivo evidence of an influence of the MGS genes of the ORC family on cilia, and consolidates the possibility that cilia dysfunction could contribute to the clinical manifestation of ORC-deficient MGS.
Collapse
Affiliation(s)
- Lars D Maerz
- Institute of Biochemistry and Molecular Biology, Ulm University, 89081, Ulm, Germany
| | - Teresa Casar Tena
- Institute of Biochemistry and Molecular Biology, Ulm University, 89081, Ulm, Germany
| | - Julian Gerhards
- Institute of Biochemistry and Molecular Biology, Ulm University, 89081, Ulm, Germany
| | - Cornelia Donow
- Institute of Biochemistry and Molecular Biology, Ulm University, 89081, Ulm, Germany
| | - Penelope A Jeggo
- Genome Damage and Stability Centre, University of Sussex, Brighton, BN1 9RQ, UK
| | - Melanie Philipp
- Institute of Biochemistry and Molecular Biology, Ulm University, 89081, Ulm, Germany.
| |
Collapse
|
40
|
Antúnez-Mojica M, Rojas-Sepúlveda AM, Mendieta-Serrano MA, Gonzalez-Maya L, Marquina S, Salas-Vidal E, Alvarez L. Lignans from Bursera fagaroides Affect In Vivo Cell Behavior by Disturbing the Tubulin Cytoskeleton in Zebrafish Embryos. Molecules 2018; 24:molecules24010008. [PMID: 30577489 PMCID: PMC6337621 DOI: 10.3390/molecules24010008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 12/13/2018] [Accepted: 12/17/2018] [Indexed: 12/04/2022] Open
Abstract
By using a zebrafish embryo model to guide the chromatographic fractionation of antimitotic secondary metabolites, seven podophyllotoxin-type lignans were isolated from a hydroalcoholic extract obtained from the steam bark of Bursera fagaroides. The compounds were identified as podophyllotoxin (1), β-peltatin-A-methylether (2), 5′-desmethoxy-β-peltatin-A-methylether (3), desmethoxy-yatein (4), desoxypodophyllotoxin (5), burseranin (6), and acetyl podophyllotoxin (7). The biological effects on mitosis, cell migration, and microtubule cytoskeleton remodeling of lignans 1–7 were further evaluated in zebrafish embryos by whole-mount immunolocalization of the mitotic marker phospho-histone H3 and by a tubulin antibody. We found that lignans 1, 2, 4, and 7 induced mitotic arrest, delayed cell migration, and disrupted the microtubule cytoskeleton in zebrafish embryos. Furthermore, microtubule cytoskeleton destabilization was observed also in PC3 cells, except for 7. Therefore, these results demonstrate that the cytotoxic activity of 1, 2, and 4 is mediated by their microtubule-destabilizing activity. In general, the in vivo and in vitro models here used displayed equivalent mitotic effects, which allows us to conclude that the zebrafish model can be a fast and cheap in vivo model that can be used to identify antimitotic natural products through bioassay-guided fractionation.
Collapse
Affiliation(s)
- Mayra Antúnez-Mojica
- CONACYT-Centro de Investigaciones Químicas-IICBA, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Morelos, Mexico.
| | | | - Mario A Mendieta-Serrano
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 62210, Morelos, Mexico.
| | - Leticia Gonzalez-Maya
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Morelos, Mexico.
| | - Silvia Marquina
- Centro de Investigaciones Químicas-IICBA, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Morelos, Mexico.
| | - Enrique Salas-Vidal
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 62210, Morelos, Mexico.
| | - Laura Alvarez
- Centro de Investigaciones Químicas-IICBA, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Morelos, Mexico.
| |
Collapse
|
41
|
Mongera A, Rowghanian P, Gustafson HJ, Shelton E, Kealhofer DA, Carn EK, Serwane F, Lucio AA, Giammona J, Campàs O. A fluid-to-solid jamming transition underlies vertebrate body axis elongation. Nature 2018; 561:401-405. [PMID: 30185907 PMCID: PMC6148385 DOI: 10.1038/s41586-018-0479-2] [Citation(s) in RCA: 409] [Impact Index Per Article: 58.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 08/03/2018] [Indexed: 12/19/2022]
Abstract
Just as in clay molding or glass blowing, sculpting biological structures requires the constituent material to locally flow like a fluid while maintaining overall mechanical integrity like a solid. Disordered soft materials, such as foams, emulsions and colloidal suspensions, switch from fluid-like to solid-like behaviors at a jamming transition1–4. Similarly, cell collectives have been shown to display glassy dynamics in 2D and 3D5,6 and jamming in cultured epithelial monolayers7,8, behaviors recently predicted theoretically9–11 and proposed to influence asthma pathobiology8 and tumor progression12. However, it is unknown if these seemingly universal behaviors occur in vivo and, specifically, if they play any functional role during embryonic morphogenesis. By combining direct in vivo measurements of tissue mechanics with analysis of cellular dynamics, we show that during vertebrate body axis elongation, posterior tissues undergo a jamming transition from a fluid-like behavior at the extending end, the mesodermal progenitor zone (MPZ), to a solid-like behavior in the presomitic mesoderm (PSM). We uncover an anteroposterior, N-cadherin-dependent gradient in yield stress that provides increasing mechanical integrity to the PSM, consistent with the tissue transiting from a wetter to a dryer foam-like architecture. Our results show that cell-scale stresses fluctuate rapidly (~1 min), enabling cell rearrangements and effectively ‘melting’ the tissue at the growing end. Persistent (>0.5 h) stresses at supracellular scales, rather than cell-scale stresses, guide morphogenetic flows in fluid-like tissue regions. Unidirectional axis extension is sustained by the reported PSM rigidification, which mechanically supports posterior, fluid-like tissues during remodeling prior to their maturation. The spatiotemporal control of fluid-like and solid-like tissue states may represent a generic physical mechanism of embryonic morphogenesis.
Collapse
Affiliation(s)
- Alessandro Mongera
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA.,California NanoSystems Institute, University of California, Santa Barbara, CA, USA.,European Molecular Biology Laboratory, Heidelberg, Germany
| | - Payam Rowghanian
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA.,California NanoSystems Institute, University of California, Santa Barbara, CA, USA
| | - Hannah J Gustafson
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA.,California NanoSystems Institute, University of California, Santa Barbara, CA, USA.,Biomolecular Science and Engineering Program, University of California, Santa Barbara, CA, USA
| | - Elijah Shelton
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA.,California NanoSystems Institute, University of California, Santa Barbara, CA, USA
| | - David A Kealhofer
- Department of Physics, University of California, Santa Barbara, CA, USA
| | - Emmet K Carn
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA
| | - Friedhelm Serwane
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA.,California NanoSystems Institute, University of California, Santa Barbara, CA, USA.,Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Adam A Lucio
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA.,California NanoSystems Institute, University of California, Santa Barbara, CA, USA
| | - James Giammona
- California NanoSystems Institute, University of California, Santa Barbara, CA, USA.,Department of Physics, University of California, Santa Barbara, CA, USA
| | - Otger Campàs
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA. .,California NanoSystems Institute, University of California, Santa Barbara, CA, USA. .,Department of Molecular, Cell and Developmental Biology, University of California, Santa Barbara, CA, USA. .,Center for Bioengineering, University of California, Santa Barbara, CA, USA.
| |
Collapse
|
42
|
Steventon B, Martinez Arias A. Evo-engineering and the cellular and molecular origins of the vertebrate spinal cord. Dev Biol 2017; 432:3-13. [DOI: 10.1016/j.ydbio.2017.01.021] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 01/03/2017] [Accepted: 01/31/2017] [Indexed: 12/31/2022]
|
43
|
Cepeda RE, Pardo RV, Macaya CC, Sarrazin AF. Contribution of cell proliferation to axial elongation in the red flour beetle Tribolium castaneum. PLoS One 2017; 12:e0186159. [PMID: 29016664 PMCID: PMC5633189 DOI: 10.1371/journal.pone.0186159] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 09/26/2017] [Indexed: 11/18/2022] Open
Abstract
Most arthropods generate their posterior bodies by adding segments periodically, as the embryo grows, from a posteriorly located region called the segment addition zone. This mode of segmentation is shared with vertebrates and relies on oscillatory mechanisms, where the temporal periodicity of a clock is translated into repetitive spatial patterns. This ordered anterior-to-posterior pattern is achieved at the same time as the tissue elongates, opening the question of the functional coordination between the mechanisms of segmental patterning and posterior growth. The study of these processes in different arthropods has played an important role in unravelling some of the molecular mechanisms of segment formation. However, the behavior of cells during elongation and how cellular processes affect this segmental patterning has been poorly studied. Cell proliferation together with cell rearrangements are presumed to be the major forces driving axis elongation in the red flour beetle Tribolium castaneum. However, there still no strong evidence about the role and distribution of cell proliferation within the embryo. In this study, we propose to address these questions by using whole embryo cultures and pharmacological manipulation. We show that considerable cell proliferation occurs during germband elongation, measured by incorporation of the nucleoside analog of thymidine 5-Ethynyl-2’-deoxyuridine, EdU. Moreover, proliferating cells appeared to be spread along the elongating embryo with a posterior bias at early segmentation. In addition, when we blocked cell division, treated germbands were always shorter than controls and in some cases not able to fully elongate, even when control embryos already started to retract and leg buds are evident. Finally, we found that the absence of cell proliferation has no apparent effect on segmental patterning, as evidenced by Tc-engrailed (Tc-en) gene expression.
Collapse
Affiliation(s)
- Rodrigo E. Cepeda
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Renato V. Pardo
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Constanza C. Macaya
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Andres F. Sarrazin
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
- * E-mail:
| |
Collapse
|
44
|
Das D, Chatti V, Emonet T, Holley SA. Patterned Disordered Cell Motion Ensures Vertebral Column Symmetry. Dev Cell 2017; 42:170-180.e5. [PMID: 28743003 DOI: 10.1016/j.devcel.2017.06.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 03/30/2017] [Accepted: 06/23/2017] [Indexed: 12/24/2022]
Abstract
The biomechanics of posterior embryonic growth must be dynamically regulated to ensure bilateral symmetry of the spinal column. Throughout vertebrate trunk elongation, motile mesodermal progenitors undergo an order-to-disorder transition via an epithelial-to-mesenchymal transition and sort symmetrically into the left and right paraxial mesoderm. We combine theoretical modeling of cell migration in a tail-bud-like geometry with experimental data analysis to assess the importance of ordered and disordered cell motion. We find that increasing order in cell motion causes a phase transition from symmetric to asymmetric body elongation. In silico and in vivo, overly ordered cell motion converts normal anisotropic fluxes into stable vortices near the posterior tail bud, contributing to asymmetric cell sorting. Thus, disorder is a physical mechanism that ensures the bilateral symmetry of the spinal column. These physical properties of the tissue connect across scales such that patterned disorder at the cellular level leads to the emergence of organism-level order.
Collapse
Affiliation(s)
- Dipjyoti Das
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Veena Chatti
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Thierry Emonet
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA; Department of Physics, Yale University, New Haven, CT, USA.
| | - Scott A Holley
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA.
| |
Collapse
|
45
|
Williams TA, Nagy LM. Linking gene regulation to cell behaviors in the posterior growth zone of sequentially segmenting arthropods. ARTHROPOD STRUCTURE & DEVELOPMENT 2017; 46:380-394. [PMID: 27720841 DOI: 10.1016/j.asd.2016.10.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 10/03/2016] [Indexed: 06/06/2023]
Abstract
Virtually all arthropods all arthropods add their body segments sequentially, one by one in an anterior to posterior progression. That process requires not only segment specification but typically growth and elongation. Here we review the functions of some of the key genes that regulate segmentation: Wnt, caudal, Notch pathway, and pair-rule genes, and discuss what can be inferred about their evolution. We focus on how these regulatory factors are integrated with growth and elongation and discuss the importance and challenges of baseline measures of growth and elongation. We emphasize a perspective that integrates the genetic regulation of segment patterning with the cellular mechanisms of growth and elongation.
Collapse
Affiliation(s)
| | - Lisa M Nagy
- Department of Molecular and Cellular Biology, The University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
46
|
Naylor RW, Han HI, Hukriede NA, Davidson AJ. Wnt8a expands the pool of embryonic kidney progenitors in zebrafish. Dev Biol 2017; 425:130-141. [PMID: 28359809 DOI: 10.1016/j.ydbio.2017.03.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 03/24/2017] [Accepted: 03/25/2017] [Indexed: 01/15/2023]
Abstract
During zebrafish embryogenesis the pronephric kidney arises from a small population of posterior mesoderm cells that then undergo expansion during early stages of renal organogenesis. While wnt8 is required for posterior mesoderm formation during gastrulation, it is also transiently expressed in the post-gastrula embryo in the intermediate mesoderm, the precursor to the pronephros and some blood/vascular lineages. Here, we show that knockdown of wnt8a, using a low dose of morpholino that does not disrupt early mesoderm patterning, reduces the number of kidney and blood cells. For the kidney, wnt8a deficiency decreases renal progenitor growth during early somitogenesis, as detected by EdU incorporation, but has no effect on apoptosis. The depletion of the renal progenitor pool in wnt8a knockdown embryos leads to cellular deficits in the pronephros at 24 hpf that are characterised by a shortened distal-most segment and stretched proximal tubule cells. A pulse of the canonical Wnt pathway agonist BIO during early somitogenesis is sufficient to rescue the size of the renal progenitor pool while longer treatment expands the number of kidney cells. Taken together, these observations indicate that Wnt8, in addition to its well-established role in posterior mesoderm patterning, also plays a later role as a factor that expands the renal progenitor pool prior to kidney morphogenesis.
Collapse
Affiliation(s)
- Richard W Naylor
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland 1142, New Zealand.
| | - Hwa In Han
- Department of Developmental Biology, Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | - Neil A Hukriede
- Department of Developmental Biology, Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | - Alan J Davidson
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland 1142, New Zealand.
| |
Collapse
|
47
|
Chun C, Wu Y, Lee SH, Williamson EA, Reinert BL, Jaiswal AS, Nickoloff JA, Hromas RA. The homologous recombination component EEPD1 is required for genome stability in response to developmental stress of vertebrate embryogenesis. Cell Cycle 2017; 15:957-62. [PMID: 26900729 PMCID: PMC4889227 DOI: 10.1080/15384101.2016.1151585] [Citation(s) in RCA: 16] [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] Open
Abstract
Stressed replication forks can be conservatively repaired and restarted using homologous recombination (HR), initiated by nuclease cleavage of branched structures at stalled forks. We previously reported that the 5' nuclease EEPD1 is recruited to stressed replication forks, where it plays critical early roles in HR initiation by promoting fork cleavage and end resection. HR repair of stressed replication forks prevents their repair by non-homologous end-joining (NHEJ), which would cause genome instability. Rapid cell division during vertebrate embryonic development generates enormous pressure to maintain replication speed and accuracy. To determine the role of EEPD1 in maintaining replication fork integrity and genome stability during rapid cell division in embryonic development, we assessed the role of EEPD1 during zebrafish embryogenesis. We show here that when EEPD1 is depleted, zebrafish embryos fail to develop normally and have a marked increase in death rate. Zebrafish embryos depleted of EEPD1 are far more sensitive to replication stress caused by nucleotide depletion. We hypothesized that the HR defect with EEPD1 depletion would shift repair of stressed replication forks to unopposed NHEJ, causing chromosome abnormalities. Consistent with this, EEPD1 depletion results in nuclear defects including anaphase bridges and micronuclei in stressed zebrafish embryos, similar to BRCA1 deficiency. These results demonstrate that the newly characterized HR protein EEPD1 maintains genome stability during embryonic replication stress. These data also imply that the rapid cell cycle transit seen during embryonic development produces replication stress that requires HR to resolve.
Collapse
Affiliation(s)
- Changzoon Chun
- a Division of Hematology/Oncology , Department of Medicine, University of Florida Health , Gainesville , FL , USA
| | - Yuehan Wu
- a Division of Hematology/Oncology , Department of Medicine, University of Florida Health , Gainesville , FL , USA
| | - Suk-Hee Lee
- b Department of Biochemistry and Molecular Biology , Indiana University School of Medicine , Indianapolis , IN , USA
| | - Elizabeth A Williamson
- a Division of Hematology/Oncology , Department of Medicine, University of Florida Health , Gainesville , FL , USA
| | - Brian L Reinert
- a Division of Hematology/Oncology , Department of Medicine, University of Florida Health , Gainesville , FL , USA
| | - Aruna Shanker Jaiswal
- a Division of Hematology/Oncology , Department of Medicine, University of Florida Health , Gainesville , FL , USA
| | - Jac A Nickoloff
- c Department of Environmental and Radiological Health Sciences , Colorado State University , Fort Collins , CO , USA
| | - Robert A Hromas
- a Division of Hematology/Oncology , Department of Medicine, University of Florida Health , Gainesville , FL , USA
| |
Collapse
|
48
|
Liu Y, Sepich DS, Solnica-Krezel L. Stat3/Cdc25a-dependent cell proliferation promotes embryonic axis extension during zebrafish gastrulation. PLoS Genet 2017; 13:e1006564. [PMID: 28222105 PMCID: PMC5319674 DOI: 10.1371/journal.pgen.1006564] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/04/2017] [Indexed: 11/29/2022] Open
Abstract
Cell proliferation has generally been considered dispensable for anteroposterior extension of embryonic axis during vertebrate gastrulation. Signal transducer and activator of transcription 3 (Stat3), a conserved controller of cell proliferation, survival and regeneration, is associated with human scoliosis, cancer and Hyper IgE Syndrome. Zebrafish Stat3 was proposed to govern convergence and extension gastrulation movements in part by promoting Wnt/Planar Cell Polarity (PCP) signaling, a conserved regulator of mediolaterally polarized cell behaviors. Here, using zebrafish stat3 null mutants and pharmacological tools, we demonstrate that cell proliferation contributes to anteroposterior embryonic axis extension. Zebrafish embryos lacking maternal and zygotic Stat3 expression exhibit normal convergence movements and planar cell polarity signaling, but transient axis elongation defect due to insufficient number of cells resulting largely from reduced cell proliferation and increased apoptosis. Pharmacologic inhibition of cell proliferation during gastrulation phenocopied axis elongation defects. Stat3 regulates cell proliferation and axis extension in part via upregulation of Cdc25a expression during oogenesis. Accordingly, restoring Cdc25a expression in stat3 mutants partially suppressed cell proliferation and gastrulation defects. During later development, stat3 mutant zebrafish exhibit stunted growth, scoliosis, excessive inflammation, and fail to thrive, affording a genetic tool to study Stat3 function in vertebrate development, regeneration, and disease. During vertebrate embryogenesis, cell proliferation, fate specification and cell movements are key processes that transform a fertilized egg into an embryo with head, trunk and tail. Cell proliferation is orchestrated by maternal and zygotic functions of conserved regulators including Cdc25a, and has generally been considered dispensable for embryonic axis elongation. Stat3 transcriptional factor, a known promoter of cell proliferation, is associated with human scoliosis, inflammation and cancer. Based on morpholino-mediated downregulation of Stat3 during zebrafish embryogenesis, Stat3 was previously proposed to regulate convergence and extension cell movements that narrow the embryonic body and elongate it from head to tail partially through planar cell polarity signaling and unknown transcriptional targets. Here, we report that zebrafish mutants lacking maternal and zygotic Stat3 expression exhibit normal convergence movements and planar cell polarity signaling, but transient axis elongation defect due to insufficient number of cells resulting largely from reduced cell proliferation and increased cell death. Accordingly, pharmacologic inhibition of cell proliferation also hinders axis elongation. Further experiments indicate that Stat3 promotes head- to -tail axis elongation by stimulating cell proliferation in part via upregulation of Cdc25a expression during oogenesis. During later development, zebrafish stat3 mutants exhibit scoliosis and inflammation, potentially affording a new tool to study related human diseases.
Collapse
Affiliation(s)
- Yinzi Liu
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Diane S. Sepich
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Lilianna Solnica-Krezel
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
49
|
Unveiling a rhythmic regulatory mode hidden in developmental tissue growth by fluorescence live imaging-based mathematical modeling. J Oral Biosci 2017. [DOI: 10.1016/j.job.2016.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
50
|
Warga RM, Wicklund A, Webster SE, Kane DA. Progressive loss of RacGAP1/ ogre activity has sequential effects on cytokinesis and zebrafish development. Dev Biol 2016; 418:307-22. [PMID: 27339293 DOI: 10.1016/j.ydbio.2016.06.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 05/27/2016] [Accepted: 06/16/2016] [Indexed: 12/20/2022]
|