1
|
Zhu M, Zhang K, Thomas EC, Xu R, Ciruna B, Hopyan S, Sun Y. Tissue stiffness mapping by light sheet elastography. SCIENCE ADVANCES 2025; 11:eadt7274. [PMID: 40085703 PMCID: PMC11908498 DOI: 10.1126/sciadv.adt7274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/06/2025] [Indexed: 03/16/2025]
Abstract
Tissue stiffness plays a crucial role in regulating morphogenesis. The ability to measure and monitor the dynamic progression of tissue stiffness is important for generating and testing mechanistic hypotheses. Methods to measure tissue properties in vivo have been emerging but present challenges with spatial and temporal resolution especially in 3D, by their reliance on highly specialized equipment, and/or due to their invasive nature. Here, we introduce light sheet elastography, a noninvasive method that couples low-frequency shear waves with light sheet fluorescence microscopy by adapting commercially available instruments. With this method, we achieved in toto stiffness mapping of organ-stage mouse and zebrafish embryos at cellular resolution. Versatility of the method enabled time-lapse stiffness mapping during tissue remodeling and of the beating embryonic heart. This method expands the spectrum of tools available to biologists and presents opportunities for uncovering the mechanical basis of morphogenesis.
Collapse
Affiliation(s)
- Min Zhu
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Kaiwen Zhang
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, M5S 3G8, Canada
| | - Evan C. Thomas
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Ran Xu
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Brian Ciruna
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Sevan Hopyan
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Division of Orthopaedic Surgery, The Hospital for Sick Children and University of Toronto, Toronto, ON, M5G 1X8, Canada
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, M5S 3G8, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON, M5S 3G4, Canada
| |
Collapse
|
2
|
Duan J, Li H, Zhang J, Xu H, Gao J, Cai M, Pan Y, Shi Y, Wang H. PIEZO1 Affects Cell Growth and Migration via Microfilament-Mediated YAP trans-Latitudinal Regulation. Anal Chem 2025; 97:147-156. [PMID: 39729436 DOI: 10.1021/acs.analchem.4c03420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2024]
Abstract
Environmental mechanical forces, such as cell membrane stress, cell extrusion, and stretch, have been proven to affect cell growth and migration. Piezo1, a mechanosensitive channel protein, responds directly to endogenous or exogenous mechanical stimuli. Here, we explored the Piezo1 distribution and microfilament morphological changes induced by mechanical forces in the tumor and normal cells. In addition, Piezo1 activation in tumor cells resulted in the nuclear accumulation of YAP, whereas nuclear export of YAP and microfilament depolymerization occurred with the prolonged activation, while a removal stimulation restored the YAP distribution and microfilament polymerization. Combining the morphological changes of the microfilament under Piezo1 activation and the function of YAP in regulating cell growth and development, we suggest that Piezo1 senses changes in environmental mechanical forces and regulates YAP distribution through the microfilament cytoskeleton network, which in turn affects the growth and migration more obviously in tumor cells rather than normal cells. Our results are essential for understanding the trans-latitudinal transmission of mechanical forces and exploring the role of environmental mechanical forces in tumor therapy.
Collapse
Affiliation(s)
- Jiawei Duan
- University of Science and Technology of China, Hefei, Anhui 230026, China
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Hongru Li
- University of Science and Technology of China, Hefei, Anhui 230026, China
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Jinrui Zhang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Haijiao Xu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Jing Gao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Mingjun Cai
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Yangang Pan
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Yan Shi
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Hongda Wang
- University of Science and Technology of China, Hefei, Anhui 230026, China
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| |
Collapse
|
3
|
Balaghi N, Fernandez-Gonzalez R. Waves of change: Dynamic actomyosin networks in embryonic development. Curr Opin Cell Biol 2024; 91:102435. [PMID: 39378575 DOI: 10.1016/j.ceb.2024.102435] [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: 07/06/2024] [Revised: 08/25/2024] [Accepted: 09/06/2024] [Indexed: 10/10/2024]
Abstract
As animals develop, molecules, cells, and cell ensembles move in beautifully orchestrated choreographies. Movement at each of these scales requires generation of mechanical force. In eukaryotic cells, the actomyosin cytoskeleton generates mechanical forces. Continuous advances in in vivo microscopy have enabled visualization and quantitative assessment of actomyosin dynamics and force generation, within and across cells, in living embryos. Recent studies reveal that actomyosin networks can form periodic waves in vivo. Here, we highlight contributions of actomyosin waves to molecular transport, cell movement, and cell coordination in developing embryos.
Collapse
Affiliation(s)
- Negar Balaghi
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, M5G 1M1, Canada. https://twitter.com/negberry
| | - Rodrigo Fernandez-Gonzalez
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, M5G 1M1, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada; Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada.
| |
Collapse
|
4
|
Liu J, Prahl LS, Huang AZ, Hughes AJ. Measurement of adhesion and traction of cells at high yield reveals an energetic ratchet operating during nephron condensation. Proc Natl Acad Sci U S A 2024; 121:e2404586121. [PMID: 39292750 PMCID: PMC11441508 DOI: 10.1073/pnas.2404586121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 08/21/2024] [Indexed: 09/20/2024] Open
Abstract
Developmental biology-inspired strategies for tissue-building have extraordinary promise for regenerative medicine, spurring interest in the relationship between cell biophysical properties and morphological transitions. However, mapping gene or protein expression data to cell biophysical properties to physical morphogenesis remains challenging with current techniques. Here, we present multiplexed adhesion and traction of cells at high yield (MATCHY). MATCHY advances the multiplexing and throughput capabilities of existing traction force and cell-cell adhesion assays using microfabrication and a semiautomated computation scheme with machine learning-driven cell segmentation. Both biophysical assays are coupled with serial downstream immunofluorescence to extract cell type/signaling state information. MATCHY is especially suited to complex primary tissue-, organoid-, or biopsy-derived cell mixtures since it does not rely on a priori knowledge of cell surface markers, cell sorting, or use of lineage-specific reporter animals. We first validate MATCHY on canine kidney epithelial cells engineered for rearranged during transfection (RET) tyrosine kinase expression and quantify a relationship between downstream signaling and cell traction. We then use MATCHY to create a biophysical atlas of mouse embryonic kidney primary cells and identify distinct biophysical states along the nephron differentiation trajectory. Our data complement expression-level knowledge of adhesion molecule changes that accompany nephron differentiation with quantitative biophysical information. These data reveal an "energetic ratchet" that accounts for spatial trends in nephron progenitor cell condensation as they differentiate into early nephron structures, which we validate through agent-based computational simulation. MATCHY offers semiautomated cell biophysical characterization at >10,000-cell throughput, an advance benefiting fundamental studies and new synthetic tissue strategies for regenerative medicine.
Collapse
Affiliation(s)
- Jiageng Liu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA19104
| | - Louis S. Prahl
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
- Center for Soft and Living Matter, University of Pennsylvania, Philadelphia, PA19104
| | - Aria Zheyuan Huang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA19104
| | - Alex J. Hughes
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA19104
- Center for Soft and Living Matter, University of Pennsylvania, Philadelphia, PA19104
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA19104
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, PA19104
- Center for Precision Engineering for Health, University of Pennsylvania, Philadelphia, PA19104
- Materials Research Science and Engineering Center, University of Pennsylvania, Philadelphia, PA19104
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA19104
| |
Collapse
|
5
|
Hu L, Cohen RI, Barroso M, Boustany NN. Comparison of vinculin tension in cellular monolayers and three-dimensional multicellular aggregates. BIOMEDICAL OPTICS EXPRESS 2024; 15:5199-5214. [PMID: 39296399 PMCID: PMC11407257 DOI: 10.1364/boe.529156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 09/21/2024]
Abstract
Confocal frequency-domain fluorescence lifetime and Förster resonance energy transfer (FRET) microscopy of Chinese hamster ovary (CHO-K1) cells expressing the vinculin tension sensor (VinTS) is used to compare vinculin tension in three-dimensional (3D) multicellular aggregates and 2D cellular monolayers. In both 2D and 3D cultures, the FRET efficiency of VinTS is 5-6% lower than that of VinTL (p < 0.05), a tail-less control which cannot bind actin or paxillin. The difference between VinTS and VinTL FRET efficiency can be mitigated by treatment with the Rho-associated kinase inhibitor Y-27632, demonstrating that VinTS is under tension in both 2D and 3D cultures. However, there is an overall decrease in FRET efficiency of both VinTS and VinTL in the 3D multicellular aggregates compared with the 2D monolayers. Expression of VinTS in 2D and 3D cultures exhibits puncta consistent with cellular adhesions. While paxillin is present at the sites of VinTS expression in the 2D monolayers, it is generally absent from VinTS puncta in the 3D aggregates. The results suggest that VinTS experiences a modified environment in 3D aggregates compared with 2D monolayers and provide a basis for further investigation of molecular tension sensors in 3D tissue models.
Collapse
Affiliation(s)
- Luni Hu
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA
| | - Rick I Cohen
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA
| | - Margarida Barroso
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Nada N Boustany
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
6
|
Zhu M, Gu B, Thomas EC, Huang Y, Kim YK, Tao H, Yung TM, Chen X, Zhang K, Woolaver EK, Nevin MR, Huang X, Winklbauer R, Rossant J, Sun Y, Hopyan S. A fibronectin gradient remodels mixed-phase mesoderm. SCIENCE ADVANCES 2024; 10:eadl6366. [PMID: 39028807 PMCID: PMC11259159 DOI: 10.1126/sciadv.adl6366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 06/14/2024] [Indexed: 07/21/2024]
Abstract
Physical processes ultimately shape tissue during development. Two emerging proposals are that cells migrate toward stiffer tissue (durotaxis) and that the extent of cell rearrangements reflects tissue phase, but it is unclear whether and how these concepts are related. Here, we identify fibronectin-dependent tissue stiffness as a control variable that underlies and unifies these phenomena in vivo. In murine limb bud mesoderm, cells are either caged, move directionally, or intercalate as a function of their location along a stiffness gradient. A modified Landau phase equation that incorporates tissue stiffness accurately predicts cell diffusivity upon loss or gain of fibronectin. Fibronectin is regulated by WNT5A-YAP feedback that controls cell movements, tissue shape, and skeletal pattern. The results identify a key determinant of phase transition and show how fibronectin-dependent directional cell movement emerges in a mixed-phase environment in vivo.
Collapse
Affiliation(s)
- Min Zhu
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Bin Gu
- Department of Obstetrics Gynecology and Reproductive Biology, and Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Evan C. Thomas
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Yunyun Huang
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Yun-Kyo Kim
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Hirotaka Tao
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Theodora M. Yung
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Xin Chen
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Kaiwen Zhang
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| | - Elizabeth K. Woolaver
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Mikaela R. Nevin
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Xi Huang
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Rudolph Winklbauer
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| | - Janet Rossant
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Yu Sun
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Sevan Hopyan
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Orthopaedic Surgery, The Hospital for Sick Children and University of Toronto, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
7
|
Alonso-Matilla R, Provenzano PP, Odde DJ. Biophysical modeling identifies an optimal hybrid amoeboid-mesenchymal phenotype for maximal T cell migration speeds. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.29.564655. [PMID: 39026744 PMCID: PMC11257493 DOI: 10.1101/2023.10.29.564655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Despite recent experimental progress in characterizing cell migration mechanics, our understanding of the mechanisms governing rapid cell movement remains limited. To effectively limit tumor growth, antitumoral T cells need to rapidly migrate to find and kill cancer cells. To investigate the upper limits of cell speed, we developed a new hybrid stochastic-mean field model of bleb-based cell motility. We first examined the potential for adhesion-free bleb-based migration and show that cells migrate inefficiently in the absence of adhesion-based forces, i.e., cell swimming. While no cortical contractility oscillations are needed for cells to swim in viscoelastic media, high-to-low cortical contractility oscillations are necessary for cell swimming in viscous media. This involves a high cortical contractility phase with multiple bleb nucleation events, followed by an intracellular pressure buildup recovery phase at low cortical tensions, resulting in modest net cell motion. However, our model suggests that cells can employ a hybrid bleb- and adhesion-based migration mechanism for rapid cell motility and identifies conditions for optimality. The model provides a momentum-conserving mechanism underlying rapid single-cell migration and identifies factors as design criteria for engineering T cell therapies to improve movement in mechanically complex environments.
Collapse
Affiliation(s)
- Roberto Alonso-Matilla
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
- University of Minnesota Physical Sciences in Oncology Center, Minneapolis, MN, USA
- University of Minnesota Center for Multiparametric Imaging of Tumor Immune Microenvironments, Minneapolis, MN, USA
| | - Paolo P. Provenzano
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
- University of Minnesota Physical Sciences in Oncology Center, Minneapolis, MN, USA
- University of Minnesota Center for Multiparametric Imaging of Tumor Immune Microenvironments, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, USA
- Department of Hematology, Oncology, and Transplantation, University of Minnesota, USA
- Stem Cell Institute, University of Minnesota, USA
| | - David J. Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
- University of Minnesota Physical Sciences in Oncology Center, Minneapolis, MN, USA
- University of Minnesota Center for Multiparametric Imaging of Tumor Immune Microenvironments, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, USA
| |
Collapse
|
8
|
Polsani N, Yung T, Thomas E, Phung-Rojas M, Gupta I, Denker J, Lau K, Feng X, Ibarra B, Hopyan S, Atit RP. Mesenchymal Wnts are required for morphogenetic movements of calvarial osteoblasts during apical expansion. Development 2024; 151:dev202596. [PMID: 38814743 PMCID: PMC11234264 DOI: 10.1242/dev.202596] [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: 12/08/2023] [Accepted: 05/13/2024] [Indexed: 06/01/2024]
Abstract
Apical expansion of calvarial osteoblast progenitors from the cranial mesenchyme (CM) above the eye is integral to calvarial growth and enclosure of the brain. The cellular behaviors and signals underlying the morphogenetic process of calvarial expansion are unknown. Time-lapse light-sheet imaging of mouse embryos revealed calvarial progenitors intercalate in 3D in the CM above the eye, and exhibit protrusive and crawling activity more apically. CM cells express non-canonical Wnt/planar cell polarity (PCP) core components and calvarial osteoblasts are bidirectionally polarized. We found non-canonical ligand Wnt5a-/- mutants have less dynamic cell rearrangements and protrusive activity. Loss of CM-restricted Wntless (CM-Wls), a gene required for secretion of all Wnt ligands, led to diminished apical expansion of Osx+ calvarial osteoblasts in the frontal bone primordia in a non-cell autonomous manner without perturbing proliferation or survival. Calvarial osteoblast polarization, progressive cell elongation and enrichment for actin along the baso-apical axis were dependent on CM-Wnts. Thus, CM-Wnts regulate cellular behaviors during calvarial morphogenesis for efficient apical expansion of calvarial osteoblasts. These findings also offer potential insights into the etiologies of calvarial dysplasias.
Collapse
Affiliation(s)
- Nikaya Polsani
- Department of Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Theodora Yung
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Evan Thomas
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Melissa Phung-Rojas
- Department of Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Isha Gupta
- Department of Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Julie Denker
- Department of Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Kimberly Lau
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Xiaotian Feng
- Department of Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Beatriz Ibarra
- Department of Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Sevan Hopyan
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Division of Orthopedics, The Hospital for Sick Children and Departments of Molecular Genetics and Surgery, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Radhika P. Atit
- Department of Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Dermatology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Genetics and Genome Sciences, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| |
Collapse
|
9
|
Stehbens SJ, Scarpa E, White MD. Perspectives in collective cell migration - moving forward. J Cell Sci 2024; 137:jcs261549. [PMID: 38904172 DOI: 10.1242/jcs.261549] [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: 06/22/2024] Open
Abstract
Collective cell migration, where cells move as a cohesive unit, is a vital process underlying morphogenesis and cancer metastasis. Thanks to recent advances in imaging and modelling, we are beginning to understand the intricate relationship between a cell and its microenvironment and how this shapes cell polarity, metabolism and modes of migration. The use of biophysical and mathematical models offers a fresh perspective on how cells migrate collectively, either flowing in a fluid-like state or transitioning to more static states. Continuing to unite researchers in biology, physics and mathematics will enable us to decode more complex biological behaviours that underly collective cell migration; only then can we understand how this coordinated movement of cells influences the formation and organisation of tissues and directs the spread of metastatic cancer. In this Perspective, we highlight exciting discoveries, emerging themes and common challenges that have arisen in recent years, and possible ways forward to bridge the gaps in our current understanding of collective cell migration.
Collapse
Affiliation(s)
- Samantha J Stehbens
- The University of Queensland, Australian Institute for Bioengineering and Nanotechnology, St Lucia, Brisbane, QLD 4072, Australia
- The University of Queensland, Institute for Molecular Bioscience, St Lucia, Brisbane, QLD 4072, Australia
| | - Elena Scarpa
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Site, Cambridge CB2 3DY, UK
| | - Melanie D White
- The University of Queensland, Institute for Molecular Bioscience, St Lucia, Brisbane, QLD 4072, Australia
- The University of Queensland, School of Biomedical Sciences, St Lucia, Brisbane, QLD 4072, Australia
| |
Collapse
|
10
|
Seto Y, Ogihara R, Takizawa K, Eiraku M. In vitro induction of patterned branchial arch-like aggregate from human pluripotent stem cells. Nat Commun 2024; 15:1351. [PMID: 38355589 PMCID: PMC10867012 DOI: 10.1038/s41467-024-45285-0] [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: 05/16/2023] [Accepted: 01/19/2024] [Indexed: 02/16/2024] Open
Abstract
Early patterning of neural crest cells (NCCs) in the craniofacial primordium is important for subsequent development of proper craniofacial structures. However, because of the complexity of the environment of developing tissues, surveying the early specification and patterning of NCCs is difficult. In this study, we develop a simplified in vitro 3D model using human pluripotent stem cells to analyze the early stages of facial development. In this model, cranial NCC-like cells spontaneously differentiate from neural plate border-like cells into maxillary arch-like mesenchyme after a long-term culture. Upon the addition of EDN1 and BMP4, these aggregates are converted into a mandibular arch-like state. Furthermore, temporary treatment with EDN1 and BMP4 induces the formation of spatially separated domains expressing mandibular and maxillary arch markers within a single aggregate. These results suggest that this in vitro model is useful for determining the mechanisms underlying cell fate specification and patterning during early facial development.
Collapse
Affiliation(s)
- Yusuke Seto
- Laboratory of Developmental Systems, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin Kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan.
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan.
| | - Ryoma Ogihara
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan
| | - Kaori Takizawa
- Laboratory of Developmental Systems, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin Kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Mototsugu Eiraku
- Laboratory of Developmental Systems, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin Kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan.
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan.
- Institute for Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan.
| |
Collapse
|
11
|
Liu J, Prahl LS, Huang A, Hughes AJ. Measurement of adhesion and traction of cells at high yield (MATCHY) reveals an energetic ratchet driving nephron condensation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.07.579368. [PMID: 38370771 PMCID: PMC10871361 DOI: 10.1101/2024.02.07.579368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Engineering of embryonic strategies for tissue-building has extraordinary promise for regenerative medicine. This has led to a resurgence in interest in the relationship between cell biophysical properties and morphological transitions. However, mapping gene or protein expression data to cell biophysical properties to physical morphogenesis remains challenging with current techniques. Here we present MATCHY (multiplexed adhesion and traction of cells at high yield). MATCHY advances the multiplexing and throughput capabilities of existing traction force and cell-cell adhesion assays using microfabrication and an automated computation scheme with machine learning-driven cell segmentation. Both biophysical assays are coupled with serial downstream immunofluorescence to extract cell type/signaling state information. MATCHY is especially suited to complex primary tissue-, organoid-, or biopsy-derived cell mixtures since it does not rely on a priori knowledge of cell surface markers, cell sorting, or use of lineage-specific reporter animals. We first validate MATCHY on canine kidney epithelial cells engineered for RET tyrosine kinase expression and quantify a relationship between downstream signaling and cell traction. We go on to create a biophysical atlas of primary cells dissociated from the mouse embryonic kidney and use MATCHY to identify distinct biophysical states along the nephron differentiation trajectory. Our data complement expression-level knowledge of adhesion molecule changes that accompany nephron differentiation with quantitative biophysical information. These data reveal an 'energetic ratchet' that explains spatial nephron progenitor cell condensation from the niche as they differentiate, which we validate through agent-based computational simulation. MATCHY offers automated cell biophysical characterization at >104-cell throughput, a highly enabling advance for fundamental studies and new synthetic tissue design strategies for regenerative medicine.
Collapse
Affiliation(s)
- Jiageng Liu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Louis S. Prahl
- Department of Bioengineering, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Aria Huang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Alex J. Hughes
- Department of Bioengineering, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Center for Soft and Living Matter, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, 19104, PA, USA
| |
Collapse
|
12
|
Du Y, Xu B, Li Q, Peng C, Yang K. The role of mechanically sensitive ion channel Piezo1 in bone remodeling. Front Bioeng Biotechnol 2024; 12:1342149. [PMID: 38390363 PMCID: PMC10882629 DOI: 10.3389/fbioe.2024.1342149] [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: 11/21/2023] [Accepted: 01/16/2024] [Indexed: 02/24/2024] Open
Abstract
Piezo1 (2010) was identified as a mechanically activated cation channel capable of sensing various physical forces, such as tension, osmotic pressure, and shear force. Piezo1 mediates mechanosensory transduction in different organs and tissues, including its role in maintaining bone homeostasis. This review aimed to summarize the function and possible mechanism of Piezo1 in the mechanical receptor cells in bone tissue. We found that it is a potential therapeutic target for the treatment of bone diseases.
Collapse
Affiliation(s)
| | | | | | | | - Kai Yang
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Pfeifer CR, Shyer AE, Rodrigues AR. Creative processes during vertebrate organ morphogenesis: Biophysical self-organization at the supracellular scale. Curr Opin Cell Biol 2024; 86:102305. [PMID: 38181658 DOI: 10.1016/j.ceb.2023.102305] [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: 07/26/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 01/07/2024]
Abstract
Here, we review recent developments in the literature that provide insight into self-organization at supracellular scales in vertebrate organ morphogenesis. We briefly present a historical and conceptual analysis of the term "self-organization." Based on this analysis, we suggest that self-organizing processes, at their root, possess a form of causal relationship, reciprocal causality, that is markedly distinct from linear causal chains. We survey the extent to which reciprocal causality can be used to interpret or clarify supracellular studies in development and disease. Finally, we explore how reciprocal causality can exist across length-scales, identifying situations where multiple scales require simultaneous analysis.
Collapse
Affiliation(s)
- Charlotte R Pfeifer
- Laboratory of Morphogenesis, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Amy E Shyer
- Laboratory of Morphogenesis, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Alan R Rodrigues
- Laboratory of Morphogenesis, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
14
|
Wang J, Maeda E, Tsujimura Y, Abe T, Kiyonari H, Kitaguchi T, Yokota H, Matsumoto T. In situ FRET measurement of cellular tension using conventional confocal laser microscopy in newly established reporter mice expressing actinin tension sensor. Sci Rep 2023; 13:22729. [PMID: 38123655 PMCID: PMC10733408 DOI: 10.1038/s41598-023-50142-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023] Open
Abstract
FRET-based sensors are utilized for real-time measurements of cellular tension. However, transfection of the sensor gene shows low efficacy and is only effective for a short period. Reporter mice expressing such sensors have been developed, but sensor fluorescence has not been measured successfully using conventional confocal microscopy. Therefore, methods for spatiotemporal measurement of cellular tension in vivo or ex vivo are still limited. We established a reporter mouse line expressing FRET-based actinin tension sensors consisting of EGFP as the donor and mCherry as the acceptor and whose FRET ratio change is observable with confocal microscopy. Tension-induced changes in FRET signals were monitored in the aorta and tail tendon fascicles, as well as aortic smooth muscle cells isolated from these mice. The pattern of FRET changes was distinctive, depending on tissue type. Indeed, aortic smooth muscle cells exhibit different sensitivity to macroscopic tensile strain in situ and in an isolated state. This mouse strain will enable novel types of biomechanical investigations of cell functions in important physiological events.
Collapse
Affiliation(s)
- Junfeng Wang
- Biomechanics Laboratory, Department of Mechanical Systems Engineering, Graduate School of Engineering, Nagoya University, Furo-Cho, Chikusa-Ku, Nagoya, Aichi, 464-8603, Japan
| | - Eijiro Maeda
- Biomechanics Laboratory, Department of Mechanical Systems Engineering, Graduate School of Engineering, Nagoya University, Furo-Cho, Chikusa-Ku, Nagoya, Aichi, 464-8603, Japan
| | - Yuki Tsujimura
- RIKEN Center for Advanced Photonics, RIKEN, Wako, Saitama, Japan
| | - Takaya Abe
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Tetsuya Kitaguchi
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Kanagawa, Japan
| | - Hideo Yokota
- RIKEN Center for Advanced Photonics, RIKEN, Wako, Saitama, Japan
| | - Takeo Matsumoto
- Biomechanics Laboratory, Department of Mechanical Systems Engineering, Graduate School of Engineering, Nagoya University, Furo-Cho, Chikusa-Ku, Nagoya, Aichi, 464-8603, Japan.
| |
Collapse
|
15
|
Du Y, Yang K. Role of mechanosensitive ion channel Piezo1 in tension-side orthodontic alveolar bone remodeling in rats. Arch Oral Biol 2023; 155:105798. [PMID: 37651768 DOI: 10.1016/j.archoralbio.2023.105798] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/02/2023]
Abstract
OBJECTIVE Orthodontic tooth movement (OTM) is based on alveolar bone remodeling under mechanical force. In 2010, Piezo1 was identified as a mechanosensitive ion channel that is involved in various physiological functions. We aimed to determine the role of Piezo1 in alveolar bone remodeling during OTM. DESIGN Twenty-five six-week-old male Sprague-Dawley rats were selected to establish OTM models and sacrificed in groups of five on days 0, 1, 3, 7, and 14. Stereomicroscopy measurements, hematoxylin and eosin staining, tartrate-resistant acid phosphatase staining, and immunohistochemical staining were performed to examine the tooth movement distance, periodontal tissue morphology, and number of multinucleated osteoclasts, and explore the levels of Piezo1, bone-related factors, and Wnt/Ca2+ signaling pathway at different time points in tension-side periodontal tissues during OTM. Furthermore, we injected equivalent grammostola mechanotoxin 4 (GsMTx4; GsMTx4 group, 25 rats) or saline (control group, 25 rats) to OTM rats and recorded the aforementioned measurement indices. RESULTS Piezo1, bone-related factors and Wnt/Ca2+ signaling pathway levels were elevated on the tension side by orthodontic force in the OTM model. GsMTX4 administration downregulated the aforementioned factors and reduced the tooth movement rate. CONCLUSIONS Piezo1 is essential for alveolar bone remodeling during OTM. The Wnt/Ca2+ signaling pathway might participate in Piezo1-mediated bone remodeling.
Collapse
Affiliation(s)
- Yugui Du
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Kai Yang
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China.
| |
Collapse
|
16
|
Yousafzai MS, Hammer JA. Using Biosensors to Study Organoids, Spheroids and Organs-on-a-Chip: A Mechanobiology Perspective. BIOSENSORS 2023; 13:905. [PMID: 37887098 PMCID: PMC10605946 DOI: 10.3390/bios13100905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/13/2023] [Accepted: 09/19/2023] [Indexed: 10/28/2023]
Abstract
The increasing popularity of 3D cell culture models is being driven by the demand for more in vivo-like conditions with which to study the biochemistry and biomechanics of numerous biological processes in health and disease. Spheroids and organoids are 3D culture platforms that self-assemble and regenerate from stem cells, tissue progenitor cells or cell lines, and that show great potential for studying tissue development and regeneration. Organ-on-a-chip approaches can be used to achieve spatiotemporal control over the biochemical and biomechanical signals that promote tissue growth and differentiation. These 3D model systems can be engineered to serve as disease models and used for drug screens. While culture methods have been developed to support these 3D structures, challenges remain to completely recapitulate the cell-cell and cell-matrix biomechanical interactions occurring in vivo. Understanding how forces influence the functions of cells in these 3D systems will require precise tools to measure such forces, as well as a better understanding of the mechanobiology of cell-cell and cell-matrix interactions. Biosensors will prove powerful for measuring forces in both of these contexts, thereby leading to a better understanding of how mechanical forces influence biological systems at the cellular and tissue levels. Here, we discussed how biosensors and mechanobiological research can be coupled to develop accurate, physiologically relevant 3D tissue models to study tissue development, function, malfunction in disease, and avenues for disease intervention.
Collapse
Affiliation(s)
- Muhammad Sulaiman Yousafzai
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - John A. Hammer
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
17
|
Lien JC, Wang YL. Cyclic stretching combined with cell-cell adhesion is sufficient for inducing cell intercalation. Biophys J 2023; 122:3146-3158. [PMID: 37408306 PMCID: PMC10432222 DOI: 10.1016/j.bpj.2023.06.019] [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: 04/13/2022] [Revised: 02/09/2023] [Accepted: 06/23/2023] [Indexed: 07/07/2023] Open
Abstract
Although the important role of cell intercalation within a collective has long been recognized particularly for morphogenesis, the underlying mechanism remains poorly understood. Here we investigate the possibility that cellular responses to cyclic stretching play a major role in this process. By applying synchronized imaging and cyclic stretching to epithelial cells cultured on micropatterned polyacrylamide (PAA) substrates, we discovered that uniaxial cyclic stretching induces cell intercalation along with cell shape change and cell-cell interfacial remodeling. The process involved intermediate steps as previously reported for cell intercalation during embryonic morphogenesis, including the appearance of cell vertices, anisotropic vertex resolution, and directional expansion of cell-cell interface. Using mathematical modeling, we further found that cell shape change in conjunction with dynamic cell-cell adhesions was sufficient to account for the observations. Further investigation with small-molecule inhibitors indicated that disruption of myosin II activities suppressed cyclic stretching-induced intercalation while inhibiting the appearance of oriented vertices. Inhibition of Wnt signaling did not suppress stretch-induced cell shape change but disrupted cell intercalation and vertex resolution. Our results suggest that cyclic stretching, by inducing cell shape change and reorientation in the presence of dynamic cell-cell adhesions, can induce at least some aspects of cell intercalation and that this process is dependent in distinct ways on myosin II activities and Wnt signaling.
Collapse
Affiliation(s)
- Jui-Chien Lien
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Yu-Li Wang
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania.
| |
Collapse
|
18
|
Kyomen S, Murillo-Rincón AP, Kaucká M. Evolutionary mechanisms modulating the mammalian skull development. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220080. [PMID: 37183900 PMCID: PMC10184257 DOI: 10.1098/rstb.2022.0080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023] Open
Abstract
Mammals possess impressive craniofacial variation that mirrors their adaptation to diverse ecological niches, feeding behaviour, physiology and overall lifestyle. The spectrum of craniofacial geometries is established mainly during embryonic development. The formation of the head represents a sequence of events regulated on genomic, molecular, cellular and tissue level, with each step taking place under tight spatio-temporal control. Even minor variations in timing, position or concentration of the molecular drivers and the resulting events can affect the final shape, size and position of the skeletal elements and the geometry of the head. Our knowledge of craniofacial development increased substantially in the last decades, mainly due to research using conventional vertebrate model organisms. However, how developmental differences in head formation arise specifically within mammals remains largely unexplored. This review highlights three evolutionary mechanisms acknowledged to modify ontogenesis: heterochrony, heterotopy and heterometry. We present recent research that links changes in developmental timing, spatial organization or gene expression levels to the acquisition of species-specific skull morphologies. We highlight how these evolutionary modifications occur on the level of the genes, molecules and cellular processes, and alter conserved developmental programmes to generate a broad spectrum of skull shapes characteristic of the class Mammalia. This article is part of the theme issue 'The mammalian skull: development, structure and function'.
Collapse
Affiliation(s)
- Stella Kyomen
- Max Planck Institute for Evolutionary Biology, August-Thienemann-Strasse 2, Plön 24306, Germany
| | - Andrea P Murillo-Rincón
- Max Planck Institute for Evolutionary Biology, August-Thienemann-Strasse 2, Plön 24306, Germany
| | - Markéta Kaucká
- Max Planck Institute for Evolutionary Biology, August-Thienemann-Strasse 2, Plön 24306, Germany
| |
Collapse
|
19
|
Li X, Han Y, Li G, Zhang Y, Wang J, Feng C. Role of Wnt signaling pathway in joint development and cartilage degeneration. Front Cell Dev Biol 2023; 11:1181619. [PMID: 37363728 PMCID: PMC10285172 DOI: 10.3389/fcell.2023.1181619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023] Open
Abstract
Osteoarthritis (OA) is a prevalent musculoskeletal disease that affects approximately 500 million people worldwide. Unfortunately, there is currently no effective treatment available to stop or delay the degenerative progression of joint disease. Wnt signaling pathways play fundamental roles in the regulation of growth, development, and homeostasis of articular cartilage. This review aims to summarize the role of Wnt pathways in joint development during embryonic stages and in cartilage maintenance throughout adult life. Specifically, we focus on aberrant mechanical loading and inflammation as major players in OA progression. Excessive mechanical load activates Wnt pathway in chondrocytes, resulting in chondrocyte apoptosis, matrix destruction and other osteoarthritis-related changes. Additionally, we discuss emerging Wnt-related modulators and present an overview of emerging treatments of OA targeting Wnt signaling. Ultimately, this review provides valuable insights towards discovering new drugs or gene therapies targeting Wnt signaling pathway for diagnosing and treating osteoarthritis and other degenerative joint diseases.
Collapse
Affiliation(s)
- Xinyan Li
- Orthopaedic Research Institution of Hebei Province, Shijiazhuang, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yuanyuan Han
- Orthopaedic Research Institution of Hebei Province, Shijiazhuang, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Guimiao Li
- Orthopaedic Research Institution of Hebei Province, Shijiazhuang, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yingze Zhang
- Orthopaedic Research Institution of Hebei Province, Shijiazhuang, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Juan Wang
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Joint Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chen Feng
- Orthopaedic Research Institution of Hebei Province, Shijiazhuang, China
- NHC Key Laboratory of Intelligent Orthopaedic Equipment, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Orthopedic Clinical Research Center, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
20
|
Konopelski Snavely SE, Srinivasan S, Dreyer CA, Tan J, Carraway KL, Ho HYH. Non-canonical WNT5A-ROR signaling: New perspectives on an ancient developmental pathway. Curr Top Dev Biol 2023; 153:195-227. [PMID: 36967195 PMCID: PMC11042798 DOI: 10.1016/bs.ctdb.2023.01.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023]
Abstract
Deciphering non-canonical WNT signaling has proven to be both fascinating and challenging. Discovered almost 30 years ago, non-canonical WNT ligands signal independently of the transcriptional co-activator β-catenin to regulate a wide range of morphogenetic processes during development. The molecular and cellular mechanisms that underlie non-canonical WNT function, however, remain nebulous. Recent results from various model systems have converged to define a core non-canonical WNT pathway consisting of the prototypic non-canonical WNT ligand, WNT5A, the receptor tyrosine kinase ROR, the seven transmembrane receptor Frizzled and the cytoplasmic scaffold protein Dishevelled. Importantly, mutations in each of these signaling components cause Robinow syndrome, a congenital disorder characterized by profound tissue morphogenetic abnormalities. Moreover, dysregulation of the pathway has also been linked to cancer metastasis. As new knowledge concerning the WNT5A-ROR pathway continues to grow, modeling these mutations will likely provide crucial insights into both the physiological regulation of the pathway and the etiology of WNT5A-ROR-driven diseases.
Collapse
Affiliation(s)
- Sara E Konopelski Snavely
- Department of Cell Biology and Human Anatomy, University of California Davis, School of Medicine, Davis, CA, United States
| | - Srisathya Srinivasan
- Department of Cell Biology and Human Anatomy, University of California Davis, School of Medicine, Davis, CA, United States
| | - Courtney A Dreyer
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, School of Medicine, Sacramento, CA, United States
| | - Jia Tan
- Department of Cell Biology and Human Anatomy, University of California Davis, School of Medicine, Davis, CA, United States
| | - Kermit L Carraway
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, School of Medicine, Sacramento, CA, United States
| | - Hsin-Yi Henry Ho
- Department of Cell Biology and Human Anatomy, University of California Davis, School of Medicine, Davis, CA, United States.
| |
Collapse
|
21
|
Kim YJ, Hyun J. Mechanosensitive ion channels in apoptosis and ferroptosis: focusing on the role of Piezo1. BMB Rep 2023; 56:145-152. [PMID: 36724905 PMCID: PMC10068349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/03/2023] Open
Abstract
Mechanosensitive ion channels sense mechanical stimuli applied directly to the cellular membranes or indirectly through their tethered components, provoking cellular mechanoresponses. Among others, Piezo1 mechanosensitive ion channel is a relatively novel Ca2+-permeable channel that is primarily present in non-sensory tissues. Recent studies have demonstrated that Piezo1 plays an important role in Ca2+-dependent cell death, including apoptosis and ferroptosis, in the presence of mechanical stimuli. It has also been proven that cancer cells are sensitive to mechanical stresses due to higher expression levels of Piezo1 compared to normal cells. In this review, we discuss Piezo1-mediated cell death mechanisms and therapeutic strategies to inhibit or induce cell death by modulating the activity of Piezo1 with pharmacological drugs or mechanical perturbations induced by stretch and ultrasound. [BMB Reports 2023; 56(3): 145-152].
Collapse
Affiliation(s)
- Yong-Jae Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | - Jeongeun Hyun
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
- Mechanobiology Dental Medicine Research Center, College of Dentistry, Dankook University, Cheonan 31116, Korea
| |
Collapse
|
22
|
Kim YJ, Hyun J. Mechanosensitive ion channels in apoptosis and ferroptosis: focusing on the role of Piezo1. BMB Rep 2023; 56:145-152. [PMID: 36724905 PMCID: PMC10068349 DOI: 10.5483/bmbrep.2023-0002] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 08/27/2023] Open
Abstract
Mechanosensitive ion channels sense mechanical stimuli applied directly to the cellular membranes or indirectly through their tethered components, provoking cellular mechanoresponses. Among others, Piezo1 mechanosensitive ion channel is a relatively novel Ca2+-permeable channel that is primarily present in non-sensory tissues. Recent studies have demonstrated that Piezo1 plays an important role in Ca2+-dependent cell death, including apoptosis and ferroptosis, in the presence of mechanical stimuli. It has also been proven that cancer cells are sensitive to mechanical stresses due to higher expression levels of Piezo1 compared to normal cells. In this review, we discuss Piezo1-mediated cell death mechanisms and therapeutic strategies to inhibit or induce cell death by modulating the activity of Piezo1 with pharmacological drugs or mechanical perturbations induced by stretch and ultrasound. [BMB Reports 2023; 56(3): 145-152].
Collapse
Affiliation(s)
- Yong-Jae Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| | - Jeongeun Hyun
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
- Mechanobiology Dental Medicine Research Center, College of Dentistry, Dankook University, Cheonan 31116, Korea
| |
Collapse
|
23
|
Windgasse L, Grashoff C. Multiplexed Molecular Tension Sensor Measurements Using PIE-FLIM. Methods Mol Biol 2023; 2600:221-237. [PMID: 36587101 DOI: 10.1007/978-1-0716-2851-5_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Genetically encoded Förster Resonance Energy Transfer (FRET)-based tension sensors were developed to enable the quantification of piconewton (pN)-scale forces that act across distinct proteins in living cells and organisms. An important extension of this technology is the multiplexing of tension sensors to monitor several independent FRET probes in parallel. Here we describe how pulsed interleaved excitation (PIE)-fluorescence lifetime imaging microscopy (FLIM) can be implemented to enable the analysis of two co-expressed tension sensor constructs. Our protocol covers all essential steps from biosensor expression and live cell PIE image acquisition to lifetime calculations.
Collapse
Affiliation(s)
- Lukas Windgasse
- Department of Quantitative Cell Biology, Institute of Integrative Cell Biology and Physiology, University of Münster, Münster, Germany
| | - Carsten Grashoff
- Department of Quantitative Cell Biology, Institute of Integrative Cell Biology and Physiology, University of Münster, Münster, Germany.
| |
Collapse
|
24
|
Fenelon KD, Thomas E, Samani M, Zhu M, Tao H, Sun Y, McNeill H, Hopyan S. Transgenic force sensors and software to measure force transmission across the mammalian nuclear envelope in vivo. Biol Open 2022; 11:bio059656. [PMID: 36350289 PMCID: PMC9672859 DOI: 10.1242/bio.059656] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 01/28/2025] Open
Abstract
Nuclear mechanotransduction is a growing field with exciting implications for the regulation of gene expression and cellular function. Mechanical signals may be transduced to the nuclear interior biochemically or physically through connections between the cell surface and chromatin. To define mechanical stresses upon the nucleus in physiological settings, we generated transgenic mouse strains that harbour FRET-based tension sensors or control constructs in the outer and inner aspects of the nuclear envelope. We knocked-in a published esprin-2G sensor to measure tensions across the LINC complex and generated a new sensor that links the inner nuclear membrane to chromatin. To mitigate challenges inherent to fluorescence lifetime analysis in vivo, we developed software (FLIMvivo) that markedly improves the fitting of fluorescence decay curves. In the mouse embryo, the sensors responded to cytoskeletal relaxation and stretch applied by micro-aspiration. They reported organ-specific differences and a spatiotemporal tension gradient along the proximodistal axis of the limb bud, raising the possibility that mechanical mechanisms coregulate pattern formation. These mouse strains and software are potentially valuable tools for testing and refining mechanotransduction hypotheses in vivo.
Collapse
Affiliation(s)
- Kelli D. Fenelon
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Evan Thomas
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Mohammad Samani
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Min Zhu
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| | - Hirotaka Tao
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| | - Helen McNeill
- Department of Developmental Biology, Washington University, St. Louis, MO 63110, USA
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON M5G 1X5, Canada
| | - Sevan Hopyan
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Orthopaedic Surgery, Hospital for Sick Children and University of Toronto, ON M5G 1X8, Canada
| |
Collapse
|
25
|
Chen J, Yao Y, Wang X, Wang Y, Li T, Du J. Chloroquine regulates the proliferation and apoptosis of palate development on mice embryo by activating P53 through blocking autophagy in vitro. In Vitro Cell Dev Biol Anim 2022; 58:558-570. [PMID: 35947289 DOI: 10.1007/s11626-022-00704-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 07/02/2022] [Indexed: 11/05/2022]
Abstract
Cleft lip and palate is one of the most frequent congenital developmental defects. Autophagy is a highly conserved process of cell self-degradation in eukaryotes, involving multiple biological processes in which chloroquine (CQ) is the most common inhibitor. However, whether CQ affects and how it affects palate development is unknown. Mouse embryonic palatal cells (MEPCs) were treated with CQ to observe cell viability, apoptosis, migration, osteogenic differentiation by cell proliferation assay, flow cytometric analysis, scratch assay, and alizarin red staining. PI staining was used to measure cell cycle distribution. Immunofluorescence (IF) assay and transmission electron microscopy were used to detect autophagosomes. The autophagy-related factors (LC3 and P62), apoptosis-related markers (P53, caspase-3 cleaved caspase-3, BAX, and BCL-2), and cell cycle-related proteins (P21, CDK2, CDK4, cyclin D1, and cyclin E) were all measured by western blot. CQ inhibited the proliferation of MEPCs by arresting the G0/G1 phase of the cell cycle in a concentration- and time-dependent manner with cell cycle-related proteins P21 upregulated and CDK2, CDK4, cyclin D1, and cyclin E downregulated. Then we detected CQ also induced cell apoptosis in a dose-dependent manner by decreasing the BCL-2/BAX ratio and increasing cleaved caspase-3. Next, it was investigated that migration and osteogenesis of MEPCs decreased with CQ treatment in a dose-dependent manner. Meanwhile, CQ blocked the autophagy pathway by upregulating LC3II and P62 expressions which activated the P53 pathway. CQ activates P53 which affects MEPC biological characteristics by changing the proliferation and apoptosis of MEPCs through inhibiting autophagy.
Collapse
Affiliation(s)
- Jing Chen
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Yaxia Yao
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Xiaotong Wang
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Yijia Wang
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Tianli Li
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Juan Du
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China.
| |
Collapse
|
26
|
Casanellas I, Lagunas A, Vida Y, Pérez-Inestrosa E, Rodríguez-Pereira C, Magalhaes J, Andrades JA, Becerra J, Samitier J. Nanoscale ligand density modulates gap junction intercellular communication of cell condensates during chondrogenesis. Nanomedicine (Lond) 2022; 17:775-791. [PMID: 35642556 DOI: 10.2217/nnm-2021-0399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To unveil the influence of cell-matrix adhesions in the establishment of gap junction intercellular communication (GJIC) during cell condensation in chondrogenesis. Materials & methods: Previously developed nanopatterns of the cell adhesive ligand arginine-glycine-aspartic acid were used as cell culture substrates to control cell adhesion at the nanoscale. In vitro chondrogenesis of mesenchymal stem cells was conducted on the nanopatterns. Cohesion and GJIC were evaluated in cell condensates. Results: Mechanical stability and GJIC are enhanced by a nanopattern configuration in which 90% of the surface area presents adhesion sites separated less than 70 nm, thus providing an onset for cell signaling. Conclusion: Cell-matrix adhesions regulate GJIC of mesenchymal cell condensates during in vitro chondrogenesis from a threshold configuration at the nanoscale.
Collapse
Affiliation(s)
- Ignasi Casanellas
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science &Technology (BIST). c/Baldiri Reixac, 10-12, Barcelona, 08028, Spain.,Department of Electronics & Biomedical Engineering, University of Barcelona (UB). c/Martí i Franquès, 1, 08028, Barcelona, Spain.,Biomedical Research Networking Center in Bioengineering,Biomaterials & Nanomedicine (CIBER-BBN). Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
| | - Anna Lagunas
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science &Technology (BIST). c/Baldiri Reixac, 10-12, Barcelona, 08028, Spain.,Biomedical Research Networking Center in Bioengineering,Biomaterials & Nanomedicine (CIBER-BBN). Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
| | - Yolanda Vida
- Universidad de Málaga-IBIMA, Dpto. Química Orgánica. Campus de Teatinos s/n, Málaga, 29071, Spain.,Centro Andaluz de Nanomedicina y Biotecnología-BIONAND. Parque Tecnológico de Andalucía, c/Severo Ochoa 35, C,ampanillas, Málaga, 29590, Spain
| | - Ezequiel Pérez-Inestrosa
- Universidad de Málaga-IBIMA, Dpto. Química Orgánica. Campus de Teatinos s/n, Málaga, 29071, Spain.,Centro Andaluz de Nanomedicina y Biotecnología-BIONAND. Parque Tecnológico de Andalucía, c/Severo Ochoa 35, C,ampanillas, Málaga, 29590, Spain
| | - Cristina Rodríguez-Pereira
- Unidad de Medicina Regenerativa, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC). c/Xubias de Arriba, 84, A Coruña, 15006, Spain
| | - Joana Magalhaes
- Biomedical Research Networking Center in Bioengineering,Biomaterials & Nanomedicine (CIBER-BBN). Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain.,Unidad de Medicina Regenerativa, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC). c/Xubias de Arriba, 84, A Coruña, 15006, Spain
| | - José A Andrades
- Biomedical Research Networking Center in Bioengineering,Biomaterials & Nanomedicine (CIBER-BBN). Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain.,Centro Andaluz de Nanomedicina y Biotecnología-BIONAND. Parque Tecnológico de Andalucía, c/Severo Ochoa 35, C,ampanillas, Málaga, 29590, Spain.,Department of Cell Biology, Genetics & Physiology, Universidad de Málaga (UMA), Instituto de Investigación Biomédica de Málaga (IBIMA). Av. Cervantes, 2, Málaga, 29071, Spain
| | - José Becerra
- Biomedical Research Networking Center in Bioengineering,Biomaterials & Nanomedicine (CIBER-BBN). Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain.,Centro Andaluz de Nanomedicina y Biotecnología-BIONAND. Parque Tecnológico de Andalucía, c/Severo Ochoa 35, C,ampanillas, Málaga, 29590, Spain.,Department of Cell Biology, Genetics & Physiology, Universidad de Málaga (UMA), Instituto de Investigación Biomédica de Málaga (IBIMA). Av. Cervantes, 2, Málaga, 29071, Spain
| | - Josep Samitier
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science &Technology (BIST). c/Baldiri Reixac, 10-12, Barcelona, 08028, Spain.,Department of Electronics & Biomedical Engineering, University of Barcelona (UB). c/Martí i Franquès, 1, 08028, Barcelona, Spain.,Biomedical Research Networking Center in Bioengineering,Biomaterials & Nanomedicine (CIBER-BBN). Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
| |
Collapse
|
27
|
Huebner RJ, Weng S, Lee C, Sarıkaya S, Papoulas O, Cox RM, Marcotte EM, Wallingford JB. ARVCF catenin controls force production during vertebrate convergent extension. Dev Cell 2022; 57:1119-1131.e5. [PMID: 35476939 DOI: 10.1016/j.devcel.2022.04.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 02/01/2022] [Accepted: 04/01/2022] [Indexed: 11/03/2022]
Abstract
The design of an animal's body plan is encoded in the genome, and the execution of this program is a mechanical progression involving coordinated movement of proteins, cells, and whole tissues. Thus, a challenge to understanding morphogenesis is connecting events that occur across various length scales. Here, we describe how a poorly characterized adhesion effector, Arvcf catenin, controls Xenopus head-to-tail axis extension. We find that Arvcf is required for axis extension within the intact organism but not within isolated tissues. We show that the organism-scale phenotype results from a defect in tissue-scale force production. Finally, we determine that the force defect results from the dampening of the pulsatile recruitment of cell adhesion and cytoskeletal proteins to membranes. These results provide a comprehensive understanding of Arvcf function during axis extension and produce an insight into how a cellular-scale defect in adhesion results in an organism-scale failure of development.
Collapse
Affiliation(s)
- Robert J Huebner
- Department of Molecular Biosciences, University of Texas, Austin, TX 78712, USA
| | - Shinuo Weng
- Department of Molecular Biosciences, University of Texas, Austin, TX 78712, USA
| | - Chanjae Lee
- Department of Molecular Biosciences, University of Texas, Austin, TX 78712, USA
| | - Sena Sarıkaya
- Department of Molecular Biosciences, University of Texas, Austin, TX 78712, USA
| | - Ophelia Papoulas
- Department of Molecular Biosciences, University of Texas, Austin, TX 78712, USA
| | - Rachael M Cox
- Department of Molecular Biosciences, University of Texas, Austin, TX 78712, USA
| | - Edward M Marcotte
- Department of Molecular Biosciences, University of Texas, Austin, TX 78712, USA
| | - John B Wallingford
- Department of Molecular Biosciences, University of Texas, Austin, TX 78712, USA.
| |
Collapse
|
28
|
Vignes H, Vagena-Pantoula C, Vermot J. Mechanical control of tissue shape: Cell-extrinsic and -intrinsic mechanisms join forces to regulate morphogenesis. Semin Cell Dev Biol 2022; 130:45-55. [PMID: 35367121 DOI: 10.1016/j.semcdb.2022.03.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 11/30/2022]
Abstract
During vertebrate development, cells must proliferate, move, and differentiate to form complex shapes. Elucidating the mechanisms underlying the molecular and cellular processes involved in tissue morphogenesis is essential to understanding developmental programmes. Mechanical stimuli act as a major contributor of morphogenetic processes and impact on cell behaviours to regulate tissue shape and size. Specifically, cell extrinsic physical forces are translated into biochemical signals within cells, through the process of mechanotransduction, activating multiple mechanosensitive pathways and defining cell behaviours. Physical forces generated by tissue mechanics and the extracellular matrix are crucial to orchestrate tissue patterning and cell fate specification. At the cell scale, the actomyosin network generates the cellular tension behind the tissue mechanics involved in building tissue. Thus, understanding the role of physical forces during morphogenetic processes requires the consideration of the contribution of cell intrinsic and cell extrinsic influences. The recent development of multidisciplinary approaches, as well as major advances in genetics, microscopy, and force-probing tools, have been key to push this field forward. With this review, we aim to discuss recent work on how tissue shape can be controlled by mechanical forces by focusing specifically on vertebrate organogenesis. We consider the influences of mechanical forces by discussing the cell-intrinsic forces (such as cell tension and proliferation) and cell-extrinsic forces (such as substrate stiffness and flow forces). We review recently described processes supporting the role of intratissue force generation and propagation in the context of shape emergence. Lastly, we discuss the emerging role of tissue-scale changes in tissue material properties, extrinsic forces, and shear stress on shape establishment.
Collapse
Affiliation(s)
- Hélène Vignes
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique UMR7104, Institut National de la Santé et de la Recherche Médicale U1258 and Université de Strasbourg, Illkirch, France
| | | | - Julien Vermot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique UMR7104, Institut National de la Santé et de la Recherche Médicale U1258 and Université de Strasbourg, Illkirch, France; Department of Bioengineering, Imperial College London, London, United Kingdom.
| |
Collapse
|
29
|
Integrating mechanical signals into cellular identity. Trends Cell Biol 2022; 32:669-680. [PMID: 35337714 PMCID: PMC9288541 DOI: 10.1016/j.tcb.2022.02.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/19/2022] [Accepted: 02/23/2022] [Indexed: 11/24/2022]
Abstract
The large arrays of cell types in a multicellular organism are defined by their stereotypic size and/or morphology, and, for cells in vivo, by their anatomic positions. Historically, this identity-structure-function correlation was conceptualized as arising from distinct gene expression programs that dictate how cells appear and behave. However, a growing number of studies suggest that a cell's mechanical state is also an important determinant of its identity, both in lineage-committed cells and in pluripotent stem cells. Defining the mechanism by which mechanical inputs influence complex cellular programs remains an area of ongoing investigation. Here, we discuss how the cytoskeleton actively participates in instructing the response of the nucleus and genome to integrate mechanical and biochemical inputs, with a primary focus on the role of the actomyosin-LINC (linker of nucleoskeleton and cytoskeleton) complex axis.
Collapse
|
30
|
Goodwin K, Jaslove JM, Tao H, Zhu M, Hopyan S, Nelson CM. Patterning the embryonic pulmonary mesenchyme. iScience 2022; 25:103838. [PMID: 35252804 PMCID: PMC8889149 DOI: 10.1016/j.isci.2022.103838] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/13/2021] [Accepted: 01/25/2022] [Indexed: 12/31/2022] Open
Abstract
Smooth muscle guides the morphogenesis of several epithelia during organogenesis, including the mammalian airways. However, it remains unclear how airway smooth muscle differentiation is spatiotemporally patterned and whether it originates from transcriptionally distinct mesenchymal progenitors. Using single-cell RNA-sequencing of embryonic mouse lungs, we show that the pulmonary mesenchyme contains a continuum of cell identities, but no transcriptionally distinct progenitors. Transcriptional variability correlates with spatially distinct sub-epithelial and sub-mesothelial mesenchymal compartments that are regulated by Wnt signaling. Live-imaging and tension-sensors reveal compartment-specific migratory behaviors and cortical forces and show that sub-epithelial mesenchyme contributes to airway smooth muscle. Reconstructing differentiation trajectories reveals early activation of cytoskeletal and Wnt signaling genes. Consistently, Wnt activation induces the earliest stages of smooth muscle differentiation and local accumulation of mesenchymal F-actin, which influences epithelial morphology. Our single-cell approach uncovers the principles of pulmonary mesenchymal patterning and identifies a morphogenetically active mesenchymal layer that sculpts the airway epithelium. The embryonic lung mesenchyme is organized into spatially distinct compartments Migratory behaviors and cortical forces differ between compartments Diffusion analysis recapitulates airway smooth muscle differentiation The early stages of smooth muscle differentiation influence airway branching
Collapse
Affiliation(s)
- Katharine Goodwin
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Jacob M. Jaslove
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
- Graduate School of Biomedical Sciences, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Hirotaka Tao
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Min Zhu
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto M5S 3G8, Canada
| | - Sevan Hopyan
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto M5S 1A8, Canada
- Division of Orthopaedic Surgery, Hospital for Sick Children and University of Toronto, Toronto M5G 1X8, Canada
| | - Celeste M. Nelson
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
- Corresponding author
| |
Collapse
|
31
|
Mechanical regulation of bone remodeling. Bone Res 2022; 10:16. [PMID: 35181672 PMCID: PMC8857305 DOI: 10.1038/s41413-022-00190-4] [Citation(s) in RCA: 215] [Impact Index Per Article: 71.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/04/2021] [Accepted: 12/13/2021] [Indexed: 12/17/2022] Open
Abstract
Bone remodeling is a lifelong process that gives rise to a mature, dynamic bone structure via a balance between bone formation by osteoblasts and resorption by osteoclasts. These opposite processes allow the accommodation of bones to dynamic mechanical forces, altering bone mass in response to changing conditions. Mechanical forces are indispensable for bone homeostasis; skeletal formation, resorption, and adaptation are dependent on mechanical signals, and loss of mechanical stimulation can therefore significantly weaken the bone structure, causing disuse osteoporosis and increasing the risk of fracture. The exact mechanisms by which the body senses and transduces mechanical forces to regulate bone remodeling have long been an active area of study among researchers and clinicians. Such research will lead to a deeper understanding of bone disorders and identify new strategies for skeletal rejuvenation. Here, we will discuss the mechanical properties, mechanosensitive cell populations, and mechanotransducive signaling pathways of the skeletal system.
Collapse
|
32
|
Gu B, Bradshaw B, Zhu M, Sun Y, Hopyan S, Rossant J. Live imaging YAP signalling in mouse embryo development. Open Biol 2022; 12:210335. [PMID: 35042406 PMCID: PMC8767199 DOI: 10.1098/rsob.210335] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
YAP protein is a critical regulator of mammalian embryonic development. By generating a near-infrared fusion YAP reporter mouse line, we have achieved high-resolution live imaging of YAP localization during mouse embryonic development. We have validated the reporter by demonstrating its predicted responses to blocking LATS kinase activity or blocking cell polarity. By time lapse imaging preimplantation embryos, we revealed a mitotic reset behaviour of YAP nuclear localization. We also demonstrated deep tissue live imaging in post-implantation embryos and revealed an intriguing nuclear YAP pattern in migrating cells. The YAP fusion reporter mice and imaging methods will open new opportunities for understanding dynamic YAP signalling in vivo in many different situations.
Collapse
Affiliation(s)
- Bin Gu
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada M5G 0A4
| | - Brian Bradshaw
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada M5G 0A4
| | - Min Zhu
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada M5G 0A4.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada M5S 1A8.,Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario, Canada M5S 3G8
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario, Canada M5S 3G8.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada M5S 3G9.,Department of Electrical and Computer Engineering, University of Toronto, Toronto, Ontario, Canada M5S 3G4
| | - Sevan Hopyan
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada M5G 0A4.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada M5S 1A8.,Division of Orthopaedic Surgery, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada M5G 1X8
| | - Janet Rossant
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada M5G 0A4.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| |
Collapse
|
33
|
Xie Z, Hou L, Shen S, Wu Y, Wang J, Jie Z, Zhao X, Li X, Zhang X, Chen J, Xu W, Ning L, Ma Q, Wang S, Wang H, Yuan P, Fang X, Qin A, Fan S. Mechanical force promotes dimethylarginine dimethylaminohydrolase 1-mediated hydrolysis of the metabolite asymmetric dimethylarginine to enhance bone formation. Nat Commun 2022; 13:50. [PMID: 35013196 PMCID: PMC8748781 DOI: 10.1038/s41467-021-27629-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 11/30/2021] [Indexed: 12/30/2022] Open
Abstract
Mechanical force is critical for the development and remodeling of bone. Here we report that mechanical force regulates the production of the metabolite asymmetric dimethylarginine (ADMA) via regulating the hydrolytic enzyme dimethylarginine dimethylaminohydrolase 1 (Ddah1) expression in osteoblasts. The presence of -394 4 N del/ins polymorphism of Ddah1 and higher serum ADMA concentration are negatively associated with bone mineral density. Global or osteoblast-specific deletion of Ddah1 leads to increased ADMA level but reduced bone formation. Further molecular study unveils that mechanical stimulation enhances TAZ/SMAD4-induced Ddah1 transcription. Deletion of Ddah1 in osteoblast-lineage cells fails to respond to mechanical stimulus-associated bone formation. Taken together, the study reveals mechanical force is capable of down-regulating ADMA to enhance bone formation.
Collapse
Affiliation(s)
- Ziang Xie
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Lei Hou
- Department of Cardiology, Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shuying Shen
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Yizheng Wu
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Jian Wang
- Department of Orthopaedics, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Zhiwei Jie
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xiangde Zhao
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xiang Li
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xuyang Zhang
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Junxin Chen
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Wenbin Xu
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Lei Ning
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Qingliang Ma
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Shiyu Wang
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Haoming Wang
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Putao Yuan
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xiangqian Fang
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| | - An Qin
- Department of Orthopaedics, Shanghai Key Laboratory of Orthopaedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Shunwu Fan
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
34
|
Trubuil E, D'Angelo A, Solon J. Tissue mechanics in morphogenesis: Active control of tissue material properties to shape living organisms. Cells Dev 2021; 168:203777. [DOI: 10.1016/j.cdev.2022.203777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 03/01/2022] [Accepted: 03/29/2022] [Indexed: 11/17/2022]
|
35
|
Bosutti A, Giniatullin A, Odnoshivkina Y, Giudice L, Malm T, Sciancalepore M, Giniatullin R, D'Andrea P, Lorenzon P, Bernareggi A. "Time window" effect of Yoda1-evoked Piezo1 channel activity during mouse skeletal muscle differentiation. Acta Physiol (Oxf) 2021; 233:e13702. [PMID: 34097801 PMCID: PMC9286833 DOI: 10.1111/apha.13702] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/04/2021] [Accepted: 06/04/2021] [Indexed: 12/12/2022]
Abstract
Aim Mechanosensitive Piezo1 ion channels emerged recently as important contributors to various vital functions including modulation of the blood supply to skeletal muscles. The specific Piezo1 channel agonist Yoda1 was shown to regulate the tone of blood vessels similarly to physical exercise. However, the direct role of Piezo1 channels in muscle function has been little studied so far. We therefore investigated the action of Yoda1 on the functional state of skeletal muscle precursors (satellite cells and myotubes) and on adult muscle fibres. Methods Immunostaining, electrophysiological intracellular recordings and Ca2+ imaging experiments were performed to localize and assess the effect of the chemical activation of Piezo1 channels with Yoda1, on myogenic precursors, adult myofibres and at the adult neuromuscular junction. Results Piezo1 channels were detected by immunostaining in satellite cells (SCs) and myotubes as well as in adult myofibres. In the skeletal muscle precursors, Yoda1 treatment stimulated the differentiation and cell fusion rather than the proliferation of SCs. Moreover, in myotubes, Yoda1 induced significant [Ca2+]i transients, without detectable [Ca2+]i response in adult myofibres. Furthermore, although expression of Piezo1 channels was detected around the muscle endplate region, Yoda1 application did not alter either the nerve‐evoked or spontaneous synaptic activity or muscle contractions in adult myofibres. Conclusion Our data indicate that the chemical activation of Piezo1 channels specifically enhances the differentiation of skeletal muscle precursors, suggesting a possible new strategy to promote muscle regeneration.
Collapse
Affiliation(s)
| | - Arthur Giniatullin
- Department of Physiology Kazan State Medical University Kazan Russia
- Laboratory of Biophysics of Synaptic Processes Kazan Institute of Biochemistry and BiophysicsFederal Research Center “Kazan Scientific Center of RAS” Kazan Russia
| | | | - Luca Giudice
- A.I. Virtanen Institute for Molecular Sciences University of Eastern Finland Kuopio Finland
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences University of Eastern Finland Kuopio Finland
| | - Marina Sciancalepore
- Department of Life Sciences University of Trieste Trieste Italy
- B.R.A.I.N., University of Trieste Centre for Neuroscience Trieste Italy
| | - Rashid Giniatullin
- A.I. Virtanen Institute for Molecular Sciences University of Eastern Finland Kuopio Finland
- Institute of Fundamental Medicine and Biology Federal University Kazan Russia
| | - Paola D'Andrea
- Department of Life Sciences University of Trieste Trieste Italy
| | - Paola Lorenzon
- Department of Life Sciences University of Trieste Trieste Italy
- B.R.A.I.N., University of Trieste Centre for Neuroscience Trieste Italy
| | - Annalisa Bernareggi
- Department of Life Sciences University of Trieste Trieste Italy
- B.R.A.I.N., University of Trieste Centre for Neuroscience Trieste Italy
| |
Collapse
|
36
|
Gómez-Gálvez P, Anbari S, Escudero LM, Buceta J. Mechanics and self-organization in tissue development. Semin Cell Dev Biol 2021; 120:147-159. [PMID: 34417092 DOI: 10.1016/j.semcdb.2021.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/25/2021] [Accepted: 07/01/2021] [Indexed: 01/01/2023]
Abstract
Self-organization is an all-important feature of living systems that provides the means to achieve specialization and functionality at distinct spatio-temporal scales. Herein, we review this concept by addressing the packing organization of cells, the sorting/compartmentalization phenomenon of cell populations, and the propagation of organizing cues at the tissue level through traveling waves. We elaborate on how different theoretical models and tools from Topology, Physics, and Dynamical Systems have improved the understanding of self-organization by shedding light on the role played by mechanics as a driver of morphogenesis. Altogether, by providing a historical perspective, we show how ideas and hypotheses in the field have been revisited, developed, and/or rejected and what are the open questions that need to be tackled by future research.
Collapse
Affiliation(s)
- Pedro Gómez-Gálvez
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla and Departamento de Biologia Celular, Universidad de Sevilla, 41013 Seville, Spain; Biomedical Network Research Centre on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
| | - Samira Anbari
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Luis M Escudero
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla and Departamento de Biologia Celular, Universidad de Sevilla, 41013 Seville, Spain; Biomedical Network Research Centre on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
| | - Javier Buceta
- Institute for Integrative Systems Biology (I2SysBio), CSIC-UV, Paterna, 46980 Valencia, Spain.
| |
Collapse
|
37
|
Danescu A, Rens EG, Rehki J, Woo J, Akazawa T, Fu K, Edelstein-Keshet L, Richman JM. Symmetry and fluctuation of cell movements in neural crest-derived facial mesenchyme. Development 2021; 148:dev.193755. [PMID: 33757991 PMCID: PMC8126411 DOI: 10.1242/dev.193755] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 03/05/2021] [Indexed: 12/13/2022]
Abstract
In the face, symmetry is established when bilateral streams of neural crest cells leave the neural tube at the same time, follow identical migration routes and then give rise to the facial prominences. However, developmental instability exists, particularly surrounding the steps of lip fusion. The causes of instability are unknown but inability to cope with developmental fluctuations are a likely cause of congenital malformations, such as non-syndromic orofacial clefts. Here, we tracked cell movements over time in the frontonasal mass, which forms the facial midline and participates in lip fusion, using live-cell imaging of chick embryos. Our mathematical examination of cell velocity vectors uncovered temporal fluctuations in several parameters, including order/disorder, symmetry/asymmetry and divergence/convergence. We found that treatment with a Rho GTPase inhibitor completely disrupted the temporal fluctuations in all measures and blocked morphogenesis. Thus, we discovered that genetic control of symmetry extends to mesenchymal cell movements and that these movements are of the type that could be perturbed in asymmetrical malformations, such as non-syndromic cleft lip. This article has an associated ‘The people behind the papers’ interview. Highlighted Article: Live imaging of the chick embryo face followed by mathematical analysis of mesenchymal cell tracks captures novel fluctuations between states of order/disorder as well as symmetry/asymmetry, revealing developmental instabilities that are part of normal morphogenesis.
Collapse
Affiliation(s)
- Adrian Danescu
- Life Sciences Institute, 2350 Health Sciences Mall, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Elisabeth G Rens
- Department of Mathematics, University of British Columbia, 1986 Mathematics Road, Vancouver, V6T 1Z2, Canada
| | - Jaspreet Rehki
- Life Sciences Institute, 2350 Health Sciences Mall, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Johnathan Woo
- Life Sciences Institute, 2350 Health Sciences Mall, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Takashi Akazawa
- Life Sciences Institute, 2350 Health Sciences Mall, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Katherine Fu
- Life Sciences Institute, 2350 Health Sciences Mall, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Leah Edelstein-Keshet
- Department of Mathematics, University of British Columbia, 1986 Mathematics Road, Vancouver, V6T 1Z2, Canada
| | - Joy M Richman
- Life Sciences Institute, 2350 Health Sciences Mall, University of British Columbia, Vancouver, V6T 1Z3, Canada
| |
Collapse
|
38
|
Fischer LS, Rangarajan S, Sadhanasatish T, Grashoff C. Molecular Force Measurement with Tension Sensors. Annu Rev Biophys 2021; 50:595-616. [PMID: 33710908 DOI: 10.1146/annurev-biophys-101920-064756] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The ability of cells to generate mechanical forces, but also to sense, adapt to, and respond to mechanical signals, is crucial for many developmental, postnatal homeostatic, and pathophysiological processes. However, the molecular mechanisms underlying cellular mechanotransduction have remained elusive for many decades, as techniques to visualize and quantify molecular forces across individual proteins in cells were missing. The development of genetically encoded molecular tension sensors now allows the quantification of piconewton-scale forces that act upon distinct molecules in living cells and even whole organisms. In this review, we discuss the physical principles, advantages, and limitations of this increasingly popular method. By highlighting current examples from the literature, we demonstrate how molecular tension sensors can be utilized to obtain access to previously unappreciated biophysical parameters that define the propagation of mechanical forces on molecular scales. We discuss how the methodology can be further developed and provide a perspective on how the technique could be applied to uncover entirely novel aspects of mechanobiology in the future.
Collapse
Affiliation(s)
- Lisa S Fischer
- Department of Quantitative Cell Biology, Institute of Molecular Cell Biology, University of Münster, Münster D-48149, Germany;
| | - Srishti Rangarajan
- Department of Quantitative Cell Biology, Institute of Molecular Cell Biology, University of Münster, Münster D-48149, Germany;
| | - Tanmay Sadhanasatish
- Department of Quantitative Cell Biology, Institute of Molecular Cell Biology, University of Münster, Münster D-48149, Germany;
| | - Carsten Grashoff
- Department of Quantitative Cell Biology, Institute of Molecular Cell Biology, University of Münster, Münster D-48149, Germany;
| |
Collapse
|
39
|
Du W, Bhojwani A, Hu JK. FACEts of mechanical regulation in the morphogenesis of craniofacial structures. Int J Oral Sci 2021; 13:4. [PMID: 33547271 PMCID: PMC7865003 DOI: 10.1038/s41368-020-00110-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023] Open
Abstract
During embryonic development, organs undergo distinct and programmed morphological changes as they develop into their functional forms. While genetics and biochemical signals are well recognized regulators of morphogenesis, mechanical forces and the physical properties of tissues are now emerging as integral parts of this process as well. These physical factors drive coordinated cell movements and reorganizations, shape and size changes, proliferation and differentiation, as well as gene expression changes, and ultimately sculpt any developing structure by guiding correct cellular architectures and compositions. In this review we focus on several craniofacial structures, including the tooth, the mandible, the palate, and the cranium. We discuss the spatiotemporal regulation of different mechanical cues at both the cellular and tissue scales during craniofacial development and examine how tissue mechanics control various aspects of cell biology and signaling to shape a developing craniofacial organ.
Collapse
Affiliation(s)
- Wei Du
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Arshia Bhojwani
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Jimmy K Hu
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
40
|
Murillo-Rincón AP, Kaucka M. Insights Into the Complexity of Craniofacial Development From a Cellular Perspective. Front Cell Dev Biol 2020; 8:620735. [PMID: 33392208 PMCID: PMC7775397 DOI: 10.3389/fcell.2020.620735] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/02/2020] [Indexed: 12/13/2022] Open
Abstract
The head represents the most complex part of the body and a distinctive feature of the vertebrate body plan. This intricate structure is assembled during embryonic development in the four-dimensional process of morphogenesis. The head integrates components of the central and peripheral nervous system, sensory organs, muscles, joints, glands, and other specialized tissues in the framework of a complexly shaped skull. The anterior part of the head is referred to as the face, and a broad spectrum of facial shapes across vertebrate species enables different feeding strategies, communication styles, and diverse specialized functions. The face formation starts early during embryonic development and is an enormously complex, multi-step process regulated on a genomic, molecular, and cellular level. In this review, we will discuss recent discoveries that revealed new aspects of facial morphogenesis from the time of the neural crest cell emergence till the formation of the chondrocranium, the primary design of the individual facial shape. We will focus on molecular mechanisms of cell fate specification, the role of individual and collective cell migration, the importance of dynamic and continuous cellular interactions, responses of cells and tissues to generated physical forces, and their morphogenetic outcomes. In the end, we will examine the spatiotemporal activity of signaling centers tightly regulating the release of signals inducing the formation of craniofacial skeletal elements. The existence of these centers and their regulation by enhancers represent one of the core morphogenetic mechanisms and might lay the foundations for intra- and inter-species facial variability.
Collapse
Affiliation(s)
| | - Marketa Kaucka
- Max Planck Research Group Craniofacial Biology, Max Planck Institute for Evolutionary Biology, Plön, Germany
| |
Collapse
|
41
|
Goodwin K, Nelson CM. Mechanics of Development. Dev Cell 2020; 56:240-250. [PMID: 33321105 DOI: 10.1016/j.devcel.2020.11.025] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/07/2020] [Accepted: 11/24/2020] [Indexed: 01/06/2023]
Abstract
Mechanical forces are integral to development-from the earliest stages of embryogenesis to the construction and differentiation of complex organs. Advances in imaging and biophysical tools have allowed us to delve into the developmental mechanobiology of increasingly complex organs and organisms. Here, we focus on recent work that highlights the diversity and importance of mechanical influences during morphogenesis. Developing tissues experience intrinsic mechanical signals from active forces and changes to tissue mechanical properties as well as extrinsic mechanical signals, including constraint and compression, pressure, and shear forces. Finally, we suggest promising avenues for future work in this rapidly expanding field.
Collapse
Affiliation(s)
- Katharine Goodwin
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Celeste M Nelson
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
42
|
Shellard A, Mayor R. Durotaxis: The Hard Path from In Vitro to In Vivo. Dev Cell 2020; 56:227-239. [PMID: 33290722 DOI: 10.1016/j.devcel.2020.11.019] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/21/2020] [Accepted: 11/17/2020] [Indexed: 01/21/2023]
Abstract
Durotaxis, the process by which cells follow gradients of extracellular mechanical stiffness, has been proposed as a mechanism driving directed migration. Despite the lack of evidence for its existence in vivo, durotaxis has become an active field of research, focusing on the mechanism by which cells respond to mechanical stimuli from the environment. In this review, we describe the technical and conceptual advances in the study of durotaxis in vitro, discuss to what extent the evidence suggests durotaxis may occur in vivo, and emphasize the urgent need for in vivo demonstration of durotaxis.
Collapse
Affiliation(s)
- Adam Shellard
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
43
|
Goodwin K, Nelson CM. Uncovering cellular networks in branching morphogenesis using single-cell transcriptomics. Curr Top Dev Biol 2020; 143:239-280. [PMID: 33820623 DOI: 10.1016/bs.ctdb.2020.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Single-cell RNA-sequencing (scRNA-seq) and related technologies to identify cell types and measure gene expression in space, in time, and within lineages have multiplied rapidly in recent years. As these techniques proliferate, we are seeing an increase in their application to the study of developing tissues. Here, we focus on single-cell investigations of branching morphogenesis. Branched organs are highly complex but typically develop recursively, such that a given developmental stage theoretically contains the entire spectrum of cell identities from progenitor to terminally differentiated. Therefore, branched organs are a highly attractive system for study by scRNA-seq. First, we provide an update on advances in the field of scRNA-seq analysis, focusing on spatial transcriptomics, computational reconstruction of differentiation trajectories, and integration of scRNA-seq with lineage tracing. In addition, we discuss the possibilities and limitations for applying these techniques to studying branched organs. We then discuss exciting advances made using scRNA-seq in the study of branching morphogenesis and differentiation in mammalian organs, with emphasis on the lung, kidney, and mammary gland. We propose ways that scRNA-seq could be used to address outstanding questions in each organ. Finally, we highlight the importance of physical and mechanical signals in branching morphogenesis and speculate about how scRNA-seq and related techniques could be applied to study tissue morphogenesis beyond just differentiation.
Collapse
Affiliation(s)
- Katharine Goodwin
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, United States
| | - Celeste M Nelson
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, United States; Department of Molecular Biology, Princeton University, Princeton, NJ, United States.
| |
Collapse
|
44
|
Tao H, Lambert JP, Yung TM, Zhu M, Hahn NA, Li D, Lau K, Sturgeon K, Puviindran V, Zhang X, Gong W, Chen XX, Anderson G, Garry DJ, Henkelman RM, Sun Y, Iulianella A, Kawakami Y, Gingras AC, Hui CC, Hopyan S. IRX3/5 regulate mitotic chromatid segregation and limb bud shape. Development 2020; 147:dev.180042. [PMID: 32907847 DOI: 10.1242/dev.180042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/25/2020] [Indexed: 01/19/2023]
Abstract
Pattern formation is influenced by transcriptional regulation as well as by morphogenetic mechanisms that shape organ primordia, although factors that link these processes remain under-appreciated. Here we show that, apart from their established transcriptional roles in pattern formation, IRX3/5 help to shape the limb bud primordium by promoting the separation and intercalation of dividing mesodermal cells. Surprisingly, IRX3/5 are required for appropriate cell cycle progression and chromatid segregation during mitosis, possibly in a nontranscriptional manner. IRX3/5 associate with, promote the abundance of, and share overlapping functions with co-regulators of cell division such as the cohesin subunits SMC1, SMC3, NIPBL and CUX1. The findings imply that IRX3/5 coordinate early limb bud morphogenesis with skeletal pattern formation.
Collapse
Affiliation(s)
- Hirotaka Tao
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Jean-Philippe Lambert
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Theodora M Yung
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Min Zhu
- Department of Mechanical and Industrial Engineering, University of Toronto, ON M5S 3G8, Canada
| | - Noah A Hahn
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Danyi Li
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Kimberly Lau
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Kendra Sturgeon
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Vijitha Puviindran
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Xiaoyun Zhang
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Wuming Gong
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Xiao Xiao Chen
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Gregory Anderson
- Mouse Imaging Centre, Hospital for Sick Children, Toronto Centre for Phenogenomics, Department of Medical Biophysics, University of Toronto, Toronto, ON M5T 3H7, Canada
| | - Daniel J Garry
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - R Mark Henkelman
- Mouse Imaging Centre, Hospital for Sick Children, Toronto Centre for Phenogenomics, Department of Medical Biophysics, University of Toronto, Toronto, ON M5T 3H7, Canada
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, ON M5S 3G8, Canada
| | - Angelo Iulianella
- Department of Medical Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Yasuhiko Kawakami
- Department of Genetics, Cell Biology and Development, Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON M5G 1X5, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Chi-Chung Hui
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada .,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sevan Hopyan
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada .,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.,Division of Orthopaedic Surgery, Hospital for Sick Children and University of Toronto, Toronto M5G 1X8, Canada
| |
Collapse
|
45
|
Kindberg A, Hu JK, Bush JO. Forced to communicate: Integration of mechanical and biochemical signaling in morphogenesis. Curr Opin Cell Biol 2020; 66:59-68. [PMID: 32569947 PMCID: PMC7577940 DOI: 10.1016/j.ceb.2020.05.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/06/2020] [Accepted: 05/05/2020] [Indexed: 01/05/2023]
Abstract
Morphogenesis is a physical process that requires the generation of mechanical forces to achieve dynamic changes in cell position, tissue shape, and size as well as biochemical signals to coordinate these events. Mechanical forces are also used by the embryo to transmit detailed information across space and detected by target cells, leading to downstream changes in cellular properties and behaviors. Indeed, forces provide signaling information of complementary quality that can both synergize and diversify the functional outputs of biochemical signaling. Here, we discuss recent findings that reveal how mechanical signaling and biochemical signaling are integrated during morphogenesis and the possible context-specific advantages conferred by the interactions between these signaling mechanisms.
Collapse
Affiliation(s)
- Abigail Kindberg
- Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA, USA; Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA; Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
| | - Jimmy K Hu
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA.
| | - Jeffrey O Bush
- Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA, USA; Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA; Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
46
|
Chen X, Yuan W, Li Y, Luo J, Hou N. Role of Hippo-YAP1/TAZ pathway and its crosstalk in cardiac biology. Int J Biol Sci 2020; 16:2454-2463. [PMID: 32760212 PMCID: PMC7378646 DOI: 10.7150/ijbs.47142] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 06/11/2020] [Indexed: 12/14/2022] Open
Abstract
The Hippo pathway undertakes a pivotal role in organ size control and the process of physiology and pathology in tissue. Its downstream effectors YAP1 and TAZ receive upstream stimuli and exert transcription activity to produce biological output. Studies have demonstrated that the Hippo pathway contributes to maintenance of cardiac homeostasis and occurrence of cardiac disease. And these cardiac biological events are affected by crosstalk among Hippo-YAP1/TAZ, Wnt/β-catenin, Bone morphogenetic protein (BMP) and G-protein-coupled receptor (GPCR) signaling, which provides new insights into the Hippo pathway in heart. This review delineates the interaction among Hippo, Wnt, BMP and GPCR pathways and discusses the effects of these pathways in cardiac biology.
Collapse
Affiliation(s)
- Xiaoqing Chen
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Wenchang Yuan
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou 511436, China
| | - Yilang Li
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Jiandong Luo
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Ning Hou
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
47
|
Hannezo E, Heisenberg CP. Mechanochemical Feedback Loops in Development and Disease. Cell 2020; 178:12-25. [PMID: 31251912 DOI: 10.1016/j.cell.2019.05.052] [Citation(s) in RCA: 205] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/17/2019] [Accepted: 05/24/2019] [Indexed: 12/31/2022]
Abstract
There is increasing evidence that both mechanical and biochemical signals play important roles in development and disease. The development of complex organisms, in particular, has been proposed to rely on the feedback between mechanical and biochemical patterning events. This feedback occurs at the molecular level via mechanosensation but can also arise as an emergent property of the system at the cellular and tissue level. In recent years, dynamic changes in tissue geometry, flow, rheology, and cell fate specification have emerged as key platforms of mechanochemical feedback loops in multiple processes. Here, we review recent experimental and theoretical advances in understanding how these feedbacks function in development and disease.
Collapse
Affiliation(s)
- Edouard Hannezo
- Institute of Science and Technology Austria, Klosterneuburg, Austria.
| | | |
Collapse
|
48
|
Spatial mapping of tissue properties in vivo reveals a 3D stiffness gradient in the mouse limb bud. Proc Natl Acad Sci U S A 2020; 117:4781-4791. [PMID: 32071242 DOI: 10.1073/pnas.1912656117] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Numerous hypotheses invoke tissue stiffness as a key parameter that regulates morphogenesis and disease progression. However, current methods are insufficient to test hypotheses that concern physical properties deep in living tissues. Here we introduce, validate, and apply a magnetic device that generates a uniform magnetic field gradient within a space that is sufficient to accommodate an organ-stage mouse embryo under live conditions. The method allows rapid, nontoxic measurement of the three-dimensional (3D) spatial distribution of viscoelastic properties within mesenchyme and epithelia. Using the device, we identify an anteriorly biased mesodermal stiffness gradient along which cells move to shape the early limb bud. The stiffness gradient corresponds to a Wnt5a-dependent domain of fibronectin expression, raising the possibility that durotaxis underlies cell movements. Three-dimensional stiffness mapping enables the generation of hypotheses and potentially the rigorous testing of mechanisms of development and disease.
Collapse
|
49
|
Wang L, You X, Lotinun S, Zhang L, Wu N, Zou W. Mechanical sensing protein PIEZO1 regulates bone homeostasis via osteoblast-osteoclast crosstalk. Nat Commun 2020; 11:282. [PMID: 31941964 PMCID: PMC6962448 DOI: 10.1038/s41467-019-14146-6] [Citation(s) in RCA: 299] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 12/09/2019] [Indexed: 12/15/2022] Open
Abstract
Wolff’s law and the Utah Paradigm of skeletal physiology state that bone architecture adapts to mechanical loads. These models predict the existence of a mechanostat that links strain induced by mechanical forces to skeletal remodeling. However, how the mechanostat influences bone remodeling remains elusive. Here, we find that Piezo1 deficiency in osteoblastic cells leads to loss of bone mass and spontaneous fractures with increased bone resorption. Furthermore, Piezo1-deficient mice are resistant to further bone loss and bone resorption induced by hind limb unloading, demonstrating that PIEZO1 can affect osteoblast-osteoclast crosstalk in response to mechanical forces. At the mechanistic level, in response to mechanical loads, PIEZO1 in osteoblastic cells controls the YAP-dependent expression of type II and IX collagens. In turn, these collagen isoforms regulate osteoclast differentiation. Taken together, our data identify PIEZO1 as the major skeletal mechanosensor that tunes bone homeostasis. Mechanical forces induce bone remodeling, but how bone cells sense mechanical signaling is unclear. Here, the authors show that loss of the mechanotransduction channel Piezo1 in osteoblastic cells impairs osteoclast activity via YAP signaling and collagen expression, leading to reduced bone mass and spontaneous fractures.
Collapse
Affiliation(s)
- Lijun Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xiuling You
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Sutada Lotinun
- Department of Physiology and Skeletal Disorders Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Lingli Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Nan Wu
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China. .,Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| |
Collapse
|
50
|
Ibarra BA, Atit R. What Do Animal Models Teach Us About Congenital Craniofacial Defects? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1236:137-155. [PMID: 32304072 PMCID: PMC7394376 DOI: 10.1007/978-981-15-2389-2_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The formation of the head and face is a complex process which involves many different signaling cues regulating the migration, differentiation, and proliferation of the neural crest. This highly complex process is very error-prone, resulting in craniofacial defects in nearly 10,000 births in the United States annually. Due to the highly conserved mechanisms of craniofacial development, animal models are widely used to understand the pathogenesis of various human diseases and assist in the diagnosis and generation of preventative therapies and treatments. Here, we provide a brief background of craniofacial development and discuss several rare diseases affecting craniofacial bone development. We focus on rare congenital diseases of the cranial bone, facial jaw bones, and two classes of diseases, ciliopathies and RASopathies. Studying the animal models of these rare diseases sheds light not only on the etiology and pathology of each disease, but also provides meaningful insights towards the mechanisms which regulate normal development of the head and face.
Collapse
Affiliation(s)
- Beatriz A Ibarra
- Department of Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Radhika Atit
- Department of Biology, Case Western Reserve University, Cleveland, OH, USA.
- Department of Genetics, Case Western Reserve University, Cleveland, OH, USA.
- Department of Dermatology, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|