1
|
Gao J, Martin L, Loffet EA, Bertin R, Durel JF, Oikonomou P, Nerurkar NL. Material properties of the embryonic small intestine during buckling morphogenesis. Acta Biomater 2025; 198:257-266. [PMID: 40180004 PMCID: PMC12065659 DOI: 10.1016/j.actbio.2025.03.055] [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: 11/13/2024] [Revised: 03/20/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
During embryonic development, tissues undergo dramatic deformations as functional morphologies are stereotypically sculpted from simple rudiments. Formation of healthy, functional organs therefore requires tight control over the material properties of embryonic tissues during development, yet the biological basis of embryonic tissue mechanics is poorly understood. The present study investigates the mechanics of the embryonic small intestine, a tissue that is compactly organized in the body cavity by a mechanical instability during development, wherein differential elongation rates between the intestinal tube and its attached mesentery create compressive forces that buckle the tube into loops. The wavelength and curvature of these loops are tightly conserved for a given species. Focusing on the intestinal tube, we combined micromechanical testing with histologic analyses and enzymatic degradation experiments to conclude that elastic fibers closely associated with intestinal smooth muscle layers are responsible for the bending stiffness of the tube, and for establishing its pronounced mechanical anisotropy. These findings provide insights into the developmental role of elastic fibers in controlling tissue stiffness, and raise new questions on the physiologic function of elastic fibers in the intestine during adulthood. STATEMENT OF SIGNIFICANCE: The functional form of adult organs is established during embryogenesis through the action of physical forces on tissues with precise material properties. Despite this, however, biological control of material properties during embryogenesis is poorly understood. Focusing on the small intestine, we identified elastic fibers - rather than oriented smooth muscle - as defining bending stiffness, prescribing the lengthy intestine to be buckled precisely into compact loops for proper placement within the body cavity. We revealed a role for elastin in storing elastic energy during cell contraction, highlighting a potential role for elastin in gut motility through the ability to resist cyclic deformations associated with peristalsis. These results provide insights into intestinal development and adult function, and highlight elastin's diverse roles during organogenesis.
Collapse
Affiliation(s)
- Jenny Gao
- Department of Biomedical Engineering, Columbia University, New York 10027
| | - Lucia Martin
- Department of Biomedical Engineering, Columbia University, New York 10027
| | - Elise A Loffet
- Department of Biomedical Engineering, Columbia University, New York 10027
| | - Raphael Bertin
- Department of Biomedical Engineering, Columbia University, New York 10027
| | - John F Durel
- Department of Biomedical Engineering, Columbia University, New York 10027
| | | | - Nandan L Nerurkar
- Department of Biomedical Engineering, Columbia University, New York 10027.
| |
Collapse
|
2
|
Tavano S, Brückner DB, Tasciyan S, Tong X, Kardos R, Schauer A, Hauschild R, Heisenberg CP. BMP-dependent patterning of ectoderm tissue material properties modulates lateral mesendoderm cell migration during early zebrafish gastrulation. Cell Rep 2025; 44:115387. [PMID: 40057955 DOI: 10.1016/j.celrep.2025.115387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/24/2024] [Accepted: 02/11/2025] [Indexed: 03/29/2025] Open
Abstract
Cell migration is a fundamental process during embryonic development. Most studies in vivo have focused on the migration of cells using the extracellular matrix (ECM) as their substrate for migration. In contrast, much less is known about how cells migrate on other cells, as found in early embryos when the ECM has not yet formed. Here, we show that lateral mesendoderm (LME) cells in the early zebrafish gastrula use the ectoderm as their substrate for migration. We show that the lateral ectoderm is permissive for the animal-pole-directed migration of LME cells, while the ectoderm at the animal pole halts it. These differences in permissiveness depend on the lateral ectoderm being more cohesive than the animal ectoderm, a property controlled by bone morphogenetic protein (BMP) signaling within the ectoderm. Collectively, these findings identify ectoderm tissue cohesion as one critical factor in regulating LME migration during zebrafish gastrulation.
Collapse
Affiliation(s)
- Stefania Tavano
- Institute of Science and Technology Austria, Klosterneuburg, Austria.
| | - David B Brückner
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Saren Tasciyan
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Xin Tong
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Roland Kardos
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Alexandra Schauer
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Robert Hauschild
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | | |
Collapse
|
3
|
Zaher M, Yelin R, Arraf AA, Jadon J, Asleh MA, Goltzman S, Shaulov L, Reinhardt DP, Schultheiss TM. Stored elastic bending tension as a mediator of embryonic body folding. Cell Rep 2025; 44:115200. [PMID: 39798089 DOI: 10.1016/j.celrep.2024.115200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 11/18/2024] [Accepted: 12/20/2024] [Indexed: 01/15/2025] Open
Abstract
During development, amniote vertebrate embryos transform from a flat sheet into a three-dimensional cylindrical form through ventral folding of the lateral sides of the sheet (the lateral plate [LP]) and their fusion in the ventral midline. Using a chick embryo slice system, we find that the flat stage is actually a poised balance of opposing dorsal and ventral elastic bending tensions. An intact extracellular matrix (ECM) is required for generating tension, as localized digestion of ECM dissipates tension, while removal of endoderm or ectoderm layers has no significant effect. As development proceeds, dorsal bending tension dissipates coincident with epithelial-mesenchymal transition in the dorsal LP while ventral tension is maintained, changing the balance of forces to promote ventral folding. Interference with the elastic ECM component fibrillin reduces ventral bending tension and perturbs body folding in vivo. A model is presented for the accumulation and harnessing of LP bending tension to drive body folding.
Collapse
Affiliation(s)
- Mira Zaher
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Ronit Yelin
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Alaa A Arraf
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Julian Jadon
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Manar Abboud Asleh
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Sivan Goltzman
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Lihi Shaulov
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Dieter P Reinhardt
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada
| | - Thomas M Schultheiss
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel.
| |
Collapse
|
4
|
Gou Y, Lin F, Dan L, Zhang D. Exposure to toluene diisocyanate induces dysbiosis of gut-lung homeostasis: Involvement of gut microbiota. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 363:125119. [PMID: 39414067 DOI: 10.1016/j.envpol.2024.125119] [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: 05/19/2024] [Revised: 09/29/2024] [Accepted: 10/12/2024] [Indexed: 10/18/2024]
Abstract
Toluene diisocyanate (TDI) is a major industrial compound that induces occupational asthma with steroid-resistant properties. Recent studies suggest that the gastrointestinal tract may be an effective target for the treatment of respiratory diseases. However, the alterations of the gut-lung axis in TDI-induced asthma remain unexplored. Therefore, in this study, a model of stable occupational asthma caused by TDI exposure was established to detect the alteration of the gut-lung axis. Exposure to TDI resulted in dysbiosis of the gut microbiome, with significant decreases in Barnesiella_intestinihominis, Faecalicoccus_pleomorphus, Lactobacillus_apodemi, and Lactobacillus_intestinalis, but increases in Alistipes_shahii and Odoribacter_laneus. The largest change in abundance was in Barnesiella_intestinihominis, which decreased from 12.14 per cent to 6.18 per cent. The histopathological abnormalities, including shorter length of intestinal villi, thinner thickness of muscularis, reduced number of goblet cells and inflammatory cell infiltration, were found in TDI-treated mice compared to control mice. In addition, increased permeability (evidenced by significantly reduced levels of ZO-1, Occludin and Claudin-1) and activation of TLR4/NF-κB signaling were observed in the intestine of these TDI-exposed mice. Concurrently, exposure to TDI resulted in airway hyperresponsiveness, overt cytokine production (e.g., IL-4, IL-5, IL-13, IL-25, and IL-33), and elevated IgE level within the respiratory tract. The expression of tight junction proteins is reduced and TLR4/NF-κB signaling is activated in the lung following TDI treatment. In addition, correlation analyses showed that changes in the gut microbiota were correlated with TDI exposure-induced airway inflammation. In conclusion, the present study suggests that the immune gut-lung axis may be involved in the development of TDI-induced asthma, which may have implications for potential interventions against steroid-resistant asthma.
Collapse
Affiliation(s)
- Yuxuan Gou
- Clinical Medical School, Guizhou Medical University, Guiyang, Guizhou, 561113, China.
| | - Fu Lin
- Clinical Medical School, Guizhou Medical University, Guiyang, Guizhou, 561113, China
| | - Li Dan
- Clinical Medical School, Guizhou Medical University, Guiyang, Guizhou, 561113, China
| | - Dianyu Zhang
- Clinical Medical School, Guizhou Medical University, Guiyang, Guizhou, 561113, China
| |
Collapse
|
5
|
Oikonomou P, Calvary L, Cirne HC, Welch AE, Durel JF, Powell O, Kim K, Nerurkar NL. Application of tissue-scale tension to avian epithelia in vivo to study multiscale mechanics and inter-germ layer coupling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.04.588089. [PMID: 38617324 PMCID: PMC11014599 DOI: 10.1101/2024.04.04.588089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
As cross-disciplinary approaches drawing from physics and mechanics have increasingly influenced our understanding of morphogenesis, the tools available to measure and perturb physical aspects of embryonic development have expanded as well. However, it remains a challenge to measure mechanical properties and apply exogenous tissue-scale forces in vivo, particularly for epithelia. Exploiting the size and accessibility of the developing chick embryo, here we describe a simple technique to quantitatively apply exogenous forces on the order of ~1-100 μN to the endodermal epithelium. To demonstrate the utility of this approach, we performed a series of proof-of-concept experiments that reveal fundamental and unexpected mechanical behaviors in the early chick embryo, including mechanotype heterogeneity among cells of the midgut endoderm, complex non-cell autonomous effects of actin disruption, and a high degree of mechanical coupling between the endoderm and adjacent paraxial mesoderm. To illustrate the broader utility of this method, we determined that forces on the order of ~ 10 μN are sufficient to unzip the neural tube during primary neurulation. Together, these findings provide basic insights into the mechanics of embryonic epithelia in vivo in the early avian embryo, and provide a useful tool for future investigations of how morphogenesis is influenced by mechanical factors.
Collapse
Affiliation(s)
| | | | - Helena C. Cirne
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - Andreas E. Welch
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - John F. Durel
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - Olivia Powell
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - Kwantae Kim
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - Nandan L. Nerurkar
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| |
Collapse
|
6
|
Powell O, Garcia E, Sriram V, Qu Y, Nerurkar NL. Elongation of the nascent avian foregut requires coordination of intrinsic and extrinsic cell behaviors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.31.621372. [PMID: 39554178 PMCID: PMC11565921 DOI: 10.1101/2024.10.31.621372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
The foregut tube gives rise to the lungs and upper gastrointestinal tract, enabling vital functions of respiration and digestion. How the foregut tube forms during embryonic development has historically received considerable attention, but over the past few decades this question has primarily been addressed indirectly through studies on morphogenesis of the primitive heart tube, a closely related process. As a result, many aspects of foregut development remain unresolved. Here, we exploit the accessibility of the chick embryo to study the initial formation of the foregut tube, combining embryology with fate mapping, live imaging, and biomechanical analyses. The present study reveals that the foregut forms and elongates over a narrower time window than previously thought, and displays marked dorso-ventral and left-right asymmetries early in its development. Through tissue-specific ablation of endoderm along the anterior intestinal portal, we confirm its central biomechanical role in driving foregut morphogenesis, despite not directly contributing cells to the elongating tube. We further confirm the important role of this cell population in formation of the heart tube, with evidence that this role extends to later stages of cardiac looping as well. Together, these data reveal the need for an intricate balance between intrinsic cell behaviors and extrinsic forces for normal foregut elongation, and set the stage for future studies aimed at understanding the underlying molecular cues that coordinate this balance.
Collapse
Affiliation(s)
- Olivia Powell
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - Emily Garcia
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - Vanshika Sriram
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - Yi Qu
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - Nandan L Nerurkar
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| |
Collapse
|
7
|
Zhu M, Catta-Preta R, Lee C, Tabin C. Shifts in embryonic oxygen levels cue heterochrony in limb initiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620348. [PMID: 39484532 PMCID: PMC11527133 DOI: 10.1101/2024.10.25.620348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Heterochrony, or the alteration of developmental timing, is an important mechanism of evolutionary change. Avian species display synchronized growth of the forelimbs and hindlimbs, while mammalian species show delayed hindlimb development. We find that mammalian limb heterochrony is evident from the start of limb bud formation, and is associated with heterochronic expression of T-box transcription factors. This heterochronic shift is not due to changes in cis-regulatory sequences controlling T-box gene expression, but unexpectedly, is dependent upon differential oxygen levels to which avian and mammalian embryos are exposed prior to limb initiation, mediated, at least partially, by an NFKB transcription factor, cRel. Together, these results provide mechanistic understanding of an important example of developmental heterochrony and exemplify how the maternal environment regulates timing during embryonic development.
Collapse
|
8
|
Miyazaki K, Hoshino D, Kasajima R, Koizume S, Koshikawa N, Miyagi Y. Oncofetal morphogenesis similar to embryonic gut formation by a subpopulation of DLD-1 human colon cancer cells. Exp Cell Res 2024; 442:114188. [PMID: 39128553 DOI: 10.1016/j.yexcr.2024.114188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 06/25/2024] [Accepted: 07/25/2024] [Indexed: 08/13/2024]
Abstract
Cancer stem cells (CSC) are thought to be responsible for cancer phenotypes and cellular heterogeneity. Here we demonstrate that the human colon cancer cell line DLD1 contains two types of CSC-like cells that undergo distinct morphogenesis in the reconstituted basement membrane gel Matrigel. In our method with cancer cell spheroids, the parent cell line (DLD1-P) developed grape-like budding structures, whereas the other (DLD1-Wm) and its single-cell clones dynamically developed worm-like ones. Gene expression analysis suggested that the former mimicked intestinal crypt-villus morphogenesis, while the latter mimicked embryonic hindgut development. The organoids of DLD1-Wm cells rapidly extended in two opposite directions by expressing dipolar proteolytic activity. The invasive morphogenesis required the expression of MMP-2 and CD133 genes and ROCK activity. These cells also exhibited gastrula-like morphogenesis even in two-dimensional cultures without Matrigel. Moreover, the two DLD1 cell lines showed clear differences in cellular growth, tumor growth and susceptibility to paclitaxel. This study also provides a simple organoid culture method for human cancer cell lines. HT-29 and other cancer cell lines underwent characteristic morphogenesis in direct contact with normal fibroblasts. Such organoid cultures would be useful for investigating the nature of CSCs and for screening anti-cancer drugs. Our results lead to the hypothesis that CSC-like cells with both invasive activity and a fetal phenotype, i. e. oncofetal CSCs, are generated in some types of colon cancers.
Collapse
Affiliation(s)
- Kaoru Miyazaki
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-8515, Japan.
| | - Daisuke Hoshino
- Cancer Biology Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-8515, Japan.
| | - Rika Kasajima
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-8515, Japan.
| | - Shiro Koizume
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-8515, Japan.
| | - Naohiko Koshikawa
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuda-cho, Midori-ku, Yokohama, 226-8501, Japan.
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-8515, Japan.
| |
Collapse
|
9
|
Zhao W, Qiang L, Zhang C, Li S, Liu Y, Wang C, Ma X, Wang J, Bao Y. Near-Infrared Stimuli-Responsive Hydrogel Promotes Cell Migration for Accelerated Diabetic Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:50175-50187. [PMID: 39269914 DOI: 10.1021/acsami.4c05852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Diabetic wound healing including diabetic foot ulcers is a major clinical challenge, which could bring an increased level of mortality and morbidity. However, conventional wound dressings exhibit limited healing efficacy due to their lack of active modulation for the healing process. Here, a near-infrared (NIR) stimuli-responsive composite hydrogel dressing with the synergistic effect of both mechanical contraction and epithelial-mesenchymal transition (EMT) was developed to facilitate cell migration and vascularization for diabetic wound healing. In the methacrylated gelatin-based composite hydrogel, N-isopropylacrylamide and polydopamine nanoparticles were incorporated to endow the composite hydrogel with thermosensitive and photothermal properties. Linagliptin (LIN) was loaded into the composite hydrogel, and the drug release rate could be controlled by NIR laser irradiation. NIR-triggered on-demand active contraction of wound area and LIN release for biological stimulation were potentially realized in this responsive system due to the thermally induced sol-gel transition of the composite hydrogel. The release of loaded LIN could effectively promote cell migration by activating EMT and enhancing angiogenesis. In the full-thickness skin defect model, the LIN-loaded composite hydrogel with NIR laser irradiation had the highest wound closure rate as compared with the pure hydrogel and LIN-loaded hydrogel groups. Therefore, this composite hydrogel can serve as an excellent platform for promoting wound healing and will find more practical value in clinical treatment.
Collapse
Affiliation(s)
- Weijing Zhao
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai 200233, China
| | - Lei Qiang
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Changru Zhang
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Shuai Li
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yihao Liu
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Chengwei Wang
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Xiaojun Ma
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Jinwu Wang
- Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yuqian Bao
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai 200233, China
| |
Collapse
|
10
|
Alvarez YD, van der Spuy M, Wang JX, Noordstra I, Tan SZ, Carroll M, Yap AS, Serralbo O, White MD. A Lifeact-EGFP quail for studying actin dynamics in vivo. J Cell Biol 2024; 223:e202404066. [PMID: 38913324 PMCID: PMC11194674 DOI: 10.1083/jcb.202404066] [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: 04/12/2024] [Revised: 05/15/2024] [Accepted: 06/03/2024] [Indexed: 06/25/2024] Open
Abstract
Here, we report the generation of a transgenic Lifeact-EGFP quail line for the investigation of actin organization and dynamics during morphogenesis in vivo. This transgenic avian line allows for the high-resolution visualization of actin structures within the living embryo, from the subcellular filaments that guide cell shape to the supracellular assemblies that coordinate movements across tissues. The unique suitability of avian embryos to live imaging facilitates the investigation of previously intractable processes during embryogenesis. Using high-resolution live imaging approaches, we present the dynamic behaviors and morphologies of cellular protrusions in different tissue contexts. Furthermore, through the integration of live imaging with computational segmentation, we visualize cells undergoing apical constriction and large-scale actin structures such as multicellular rosettes within the neuroepithelium. These findings not only enhance our understanding of tissue morphogenesis but also demonstrate the utility of the Lifeact-EGFP transgenic quail as a new model system for live in vivo investigations of the actin cytoskeleton.
Collapse
Affiliation(s)
- Yanina D. Alvarez
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Marise van der Spuy
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Jian Xiong Wang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Ivar Noordstra
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Siew Zhuan Tan
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Murron Carroll
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Alpha S. Yap
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Olivier Serralbo
- Commonwealth Scientific and Industrial Research (CSIRO) Health and Biosecurity, Geelong, Australia
| | - Melanie D. White
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| |
Collapse
|
11
|
Gao J, Martin L, Loffet EA, Durel JF, Oikonomou P, Nerurkar NL. MATERIAL PROPERTIES OF THE EMBRYONIC SMALL INTESTINE DURING BUCKLING MORPHOGENESIS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.07.606927. [PMID: 39149332 PMCID: PMC11326276 DOI: 10.1101/2024.08.07.606927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
During embryonic development, tissues undergo dramatic deformations as functional morphologies are stereotypically sculpted from simple rudiments. Formation of healthy, functional organs therefore requires tight control over the material properties of embryonic tissues during development, yet the biological basis of embryonic tissue mechanics is poorly understood. The present study investigates the mechanics of the embryonic small intestine, a tissue that is compactly organized in the body cavity by a mechanical instability during development, wherein differential elongation rates between the intestinal tube and its attached mesentery create compressive forces that buckle the tube into loops with wavelength and curvature that are tightly conserved for a given species. Focusing on the intestinal tube, we combined micromechanical testing with histologic analyses and enzymatic degradation experiments to conclude that elastic fibers closely associated with intestinal smooth muscle layers are responsible for the bending stiffness of the tube, and for establishing its pronounced mechanical anisotropy. These findings provide insights into the developmental role of elastic fibers in controlling tissue stiffness, and raise new questions on the physiologic function of elastic fibers in the intestine during adulthood.
Collapse
Affiliation(s)
- Jenny Gao
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - Lucia Martin
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - Elise A. Loffet
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - John F. Durel
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | | | - Nandan L. Nerurkar
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| |
Collapse
|
12
|
Gill HK, Yin S, Lawlor JC, Huycke TR, Nerurkar NL, Tabin CJ, Mahadevan L. The developmental mechanics of divergent buckling patterns in the chick gut. Proc Natl Acad Sci U S A 2024; 121:e2310992121. [PMID: 38968105 PMCID: PMC11252809 DOI: 10.1073/pnas.2310992121] [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: 06/30/2023] [Accepted: 05/06/2024] [Indexed: 07/07/2024] Open
Abstract
Tissue buckling is an increasingly appreciated mode of morphogenesis in the embryo, but it is often unclear how geometric and material parameters are molecularly determined in native developmental contexts to generate diverse functional patterns. Here, we study the link between differential mechanical properties and the morphogenesis of distinct anteroposterior compartments in the intestinal tract-the esophagus, small intestine, and large intestine. These regions originate from a simple, common tube but adopt unique forms. Using measured data from the developing chick gut coupled with a minimal theory and simulations of differential growth, we investigate divergent lumen morphologies along the entire early gut and demonstrate that spatiotemporal geometries, moduli, and growth rates control the segment-specific patterns of mucosal buckling. Primary buckling into wrinkles, folds, and creases along the gut, as well as secondary buckling phenomena, including period-doubling in the foregut and multiscale creasing-wrinkling in the hindgut, are captured and well explained by mechanical models. This study advances our existing knowledge of how identity leads to form in these regions, laying the foundation for future work uncovering the relationship between molecules and mechanics in gut morphological regionalization.
Collapse
Affiliation(s)
- Hasreet K. Gill
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Sifan Yin
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
| | - John C. Lawlor
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Tyler R. Huycke
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA02115
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA94158
| | - Nandan L. Nerurkar
- The Fu Foundation School of Engineering and Applied Science, Department of Biomedical Engineering, Columbia University, New York, NY10027
| | - Clifford J. Tabin
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - L. Mahadevan
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
- Department of Physics, Harvard University, Cambridge, MA02138
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA02138
| |
Collapse
|
13
|
Sugiyama Y, Reed DA, Herrmann D, Lovicu FJ, Robinson ML, Timpson P, Masai I. Fibroblast growth factor-induced lens fiber cell elongation is driven by the stepwise activity of Rho and Rac. Development 2024; 151:dev202123. [PMID: 38240393 PMCID: PMC10911273 DOI: 10.1242/dev.202123] [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: 06/27/2023] [Accepted: 01/09/2024] [Indexed: 01/30/2024]
Abstract
The spheroidal shape of the eye lens is crucial for precise light focusing onto the retina. This shape is determined by concentrically aligned, convexly elongated lens fiber cells along the anterior and posterior axis of the lens. Upon differentiation at the lens equator, the fiber cells increase in height as their apical and basal tips migrate towards the anterior and posterior poles, respectively. The forces driving this elongation and migration remain unclear. We found that, in the mouse lens, membrane protrusions or lamellipodia are observed only in the maturing fibers undergoing cell curve conversion, indicating that lamellipodium formation is not the primary driver of earlier fiber migration. We demonstrated that elevated levels of fibroblast growth factor (FGF) suppressed the extension of Rac-dependent protrusions, suggesting changes in the activity of FGF controlling Rac activity, switching to lamellipodium-driven migration. Inhibitors of ROCK, myosin and actin reduced the height of both early and later fibers, indicating that elongation of these fibers relies on actomyosin contractility. Consistent with this, active RhoA was detected throughout these fibers. Given that FGF promotes fiber elongation, we propose that it does so through regulation of Rho activity.
Collapse
Affiliation(s)
- Yuki Sugiyama
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa 904-0495, Japan
- Save Sight Institute, The University of Sydney, Sydney, NSW 2000, Australia
| | - Daniel A. Reed
- Cancer Ecosystems Program, The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - David Herrmann
- Cancer Ecosystems Program, The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - Frank J. Lovicu
- Save Sight Institute, The University of Sydney, Sydney, NSW 2000, Australia
- Molecular and Cellular Biomedicine, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Michael L. Robinson
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH 45056, USA
| | - Paul Timpson
- Cancer Ecosystems Program, The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - Ichiro Masai
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa 904-0495, Japan
| |
Collapse
|
14
|
Loffet EA, Durel JF, Nerurkar NL. Evo-Devo Mechanobiology: The Missing Link. Integr Comp Biol 2023; 63:1455-1473. [PMID: 37193661 DOI: 10.1093/icb/icad033] [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: 03/16/2023] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 05/18/2023] Open
Abstract
While the modern framework of evolutionary development (evo-devo) has been decidedly genetic, historic analyses have also considered the importance of mechanics in the evolution of form. With the aid of recent technological advancements in both quantifying and perturbing changes in the molecular and mechanical effectors of organismal shape, how molecular and genetic cues regulate the biophysical aspects of morphogenesis is becoming increasingly well studied. As a result, this is an opportune time to consider how the tissue-scale mechanics that underlie morphogenesis are acted upon through evolution to establish morphological diversity. Such a focus will enable a field of evo-devo mechanobiology that will serve to better elucidate the opaque relations between genes and forms by articulating intermediary physical mechanisms. Here, we review how the evolution of shape is measured and related to genetics, how recent strides have been made in the dissection of developmental tissue mechanics, and how we expect these areas to coalesce in evo-devo studies in the future.
Collapse
Affiliation(s)
- Elise A Loffet
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA
| | - John F Durel
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA
| | - Nandan L Nerurkar
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA
| |
Collapse
|
15
|
Sugiyama Y, Reed DA, Herrmann D, Lovicu FJ, Robinson ML, Timpson P, Masai I. Fibroblast Growth Factor-induced lens fiber cell elongation is driven by the stepwise activity of Rho and Rac. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.03.569812. [PMID: 38106159 PMCID: PMC10723307 DOI: 10.1101/2023.12.03.569812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
The spheroidal shape of the eye lens is critical for precise light focusing onto the retina. This shape is determined by concentrically aligned, convexly elongated lens fiber cells along the anterior and posterior axis of the lens. Upon differentiation at the lens equator, the fiber cells increase in height as their apical and basal tips migrate towards the anterior and posterior poles, respectively. The forces driving this elongation and migration remain unclear. We found that membrane protrusions or lamellipodia are observed only in the maturing fibers undergoing cell curve conversion, indicating lamellipodium is not the primary driver of earlier fiber migration. We demonstrated that elevated levels of fibroblast growth factor (FGF) suppressed the extension of Rac-dependent protrusions, suggesting changes in the activity of FGF controling Rac activity, switching to lamellipodium-driven migration. Inhibitors of ROCK, myosin, and actin reduced the height of both early and later fibers, indicating elongation of these fibers relies on actomyosin contractility. Consistently, active RhoA was detected throughout these fibers. Given that FGF promotes fiber elongation, we propose it to do so through regulation of Rho activity.
Collapse
Affiliation(s)
- Yuki Sugiyama
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa 904-0495, Japan
- Save Sight Institute, The University of Sydney, Sydney, NSW 2000, Australia
| | - Daniel A. Reed
- Cancer Ecosystems Program, The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, St Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - David Herrmann
- Cancer Ecosystems Program, The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, St Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - Frank J. Lovicu
- Save Sight Institute, The University of Sydney, Sydney, NSW 2000, Australia
- Molecular and Cellular Biomedicine, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| | - Michael L. Robinson
- Department of Biology and Center for Visual Sciences, Miami University, Oxford, OH, USA
| | - Paul Timpson
- Cancer Ecosystems Program, The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, St Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - Ichiro Masai
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa 904-0495, Japan
| |
Collapse
|
16
|
Loffet EA, Durel JF, Gao J, Kam R, Lim H, Nerurkar NL. Elastic fibers define embryonic tissue stiffness to enable buckling morphogenesis of the small intestine. Biomaterials 2023; 303:122405. [PMID: 38000151 PMCID: PMC10842730 DOI: 10.1016/j.biomaterials.2023.122405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/22/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023]
Abstract
During embryonic development, tissues must possess precise material properties to ensure that cell-generated forces give rise to the stereotyped morphologies of developing organs. However, the question of how material properties are established and regulated during development remains understudied. Here, we aim to address these broader questions through the study of intestinal looping, a process by which the initially straight intestinal tube buckles into loops, permitting ordered packing within the body cavity. Looping results from elongation of the tube against the constraint of an attached tissue, the dorsal mesentery, which is elastically stretched by the elongating tube to nearly triple its length. This elastic energy storage allows the mesentery to provide stable compressive forces that ultimately buckle the tube into loops. Beginning with a transcriptomic analysis of the mesentery, we identified widespread upregulation of extracellular matrix related genes during looping, including genes related to elastic fiber deposition. Combining molecular and mechanical analyses, we conclude that elastin confers tensile stiffness to the mesentery, enabling its mechanical role in organizing the developing small intestine. These results shed light on the role of elastin as a driver of morphogenesis that extends beyond its more established role in resisting cyclic deformation in adult tissues.
Collapse
Affiliation(s)
- Elise A Loffet
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - John F Durel
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Jenny Gao
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Richard Kam
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Hyunjee Lim
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Nandan L Nerurkar
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA.
| |
Collapse
|
17
|
Oikonomou P, Cirne HC, Nerurkar NL. A chemo-mechanical model of endoderm movements driving elongation of the amniote hindgut. Development 2023; 150:dev202010. [PMID: 37840469 PMCID: PMC10690059 DOI: 10.1242/dev.202010] [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: 05/22/2023] [Accepted: 10/09/2023] [Indexed: 10/17/2023]
Abstract
Although mechanical and biochemical descriptions of development are each essential, integration of upstream morphogenic cues with downstream tissue mechanics remains understudied during vertebrate morphogenesis. Here, we developed a two-dimensional chemo-mechanical model to investigate how mechanical properties of the endoderm and transport properties of fibroblast growth factor (FGF) regulate avian hindgut morphogenesis in a coordinated manner. Posterior endoderm cells convert a gradient of FGF ligands into a contractile force gradient, leading to a force imbalance that drives collective cell movements that elongate the forming hindgut tube. We formulated a 2D reaction-diffusion-advection model describing the formation of an FGF protein gradient as a result of posterior displacement of cells transcribing unstable Fgf8 mRNA during axis elongation, coupled with translation, diffusion and degradation of FGF protein. The endoderm was modeled as an active viscous fluid that generates contractile stresses in proportion to FGF concentration. With parameter values constrained by experimental data, the model replicates key aspects of hindgut morphogenesis, suggests that graded isotropic contraction is sufficient to generate large anisotropic cell movements, and provides new insight into how chemo-mechanical coupling across the mesoderm and endoderm coordinates hindgut elongation with axis elongation.
Collapse
Affiliation(s)
- Panagiotis Oikonomou
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA
| | - Helena C. Cirne
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA
| | - Nandan L. Nerurkar
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA
| |
Collapse
|
18
|
Colin A, Orhant-Prioux M, Guérin C, Savinov M, Cao W, Vianay B, Scarfone I, Roux A, De La Cruz EM, Mogilner A, Théry M, Blanchoin L. Friction patterns guide actin network contraction. Proc Natl Acad Sci U S A 2023; 120:e2300416120. [PMID: 37725653 PMCID: PMC10523593 DOI: 10.1073/pnas.2300416120] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 08/09/2023] [Indexed: 09/21/2023] Open
Abstract
The shape of cells is the outcome of the balance of inner forces produced by the actomyosin network and the resistive forces produced by cell adhesion to their environment. The specific contributions of contractile, anchoring and friction forces to network deformation rate and orientation are difficult to disentangle in living cells where they influence each other. Here, we reconstituted contractile actomyosin networks in vitro to study specifically the role of the friction forces between the network and its anchoring substrate. To modulate the magnitude and spatial distribution of friction forces, we used glass or lipids surface micropatterning to control the initial shape of the network. We adapted the concentration of Nucleating Promoting Factor on each surface to induce the assembly of actin networks of similar densities and compare the deformation of the network toward the centroid of the pattern shape upon myosin-induced contraction. We found that actin network deformation was faster and more coordinated on lipid bilayers than on glass, showing the resistance of friction to network contraction. To further study the role of the spatial distribution of these friction forces, we designed heterogeneous micropatterns made of glass and lipids. The deformation upon contraction was no longer symmetric but biased toward the region of higher friction. Furthermore, we showed that the pattern of friction could robustly drive network contraction and dominate the contribution of asymmetric distributions of myosins. Therefore, we demonstrate that during contraction, both the active and resistive forces are essential to direct the actin network deformation.
Collapse
Affiliation(s)
- Alexandra Colin
- Université Grenoble-Alpes, CEA, CNRS, UMR5168, Interdisciplinary Research Institute of Grenoble, CytoMorpho Lab, Grenoble38054, France
| | - Magali Orhant-Prioux
- Université Grenoble-Alpes, CEA, CNRS, UMR5168, Interdisciplinary Research Institute of Grenoble, CytoMorpho Lab, Grenoble38054, France
| | - Christophe Guérin
- Université Grenoble-Alpes, CEA, CNRS, UMR5168, Interdisciplinary Research Institute of Grenoble, CytoMorpho Lab, Grenoble38054, France
| | - Mariya Savinov
- Courant Institute of Mathematical Sciences, New York University, New York, NY10012
| | - Wenxiang Cao
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT06520-8114
| | - Benoit Vianay
- University of Paris, INSERM, Commissariat à l'énergie atomique et aux énergies alternatives, UMRS1160, Institut de Recherche Saint Louis, CytoMorpho Lab, Hôpital Saint Louis, Paris75010, France
| | - Ilaria Scarfone
- Université Grenoble-Alpes, CEA, CNRS, UMR5168, Interdisciplinary Research Institute of Grenoble, CytoMorpho Lab, Grenoble38054, France
| | - Aurélien Roux
- Department of Biochemistry, University of Geneva, CH-1211Geneva, Switzerland
| | - Enrique M. De La Cruz
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT06520-8114
| | - Alex Mogilner
- Courant Institute of Mathematical Sciences, New York University, New York, NY10012
| | - Manuel Théry
- Université Grenoble-Alpes, CEA, CNRS, UMR5168, Interdisciplinary Research Institute of Grenoble, CytoMorpho Lab, Grenoble38054, France
- University of Paris, INSERM, Commissariat à l'énergie atomique et aux énergies alternatives, UMRS1160, Institut de Recherche Saint Louis, CytoMorpho Lab, Hôpital Saint Louis, Paris75010, France
| | - Laurent Blanchoin
- Université Grenoble-Alpes, CEA, CNRS, UMR5168, Interdisciplinary Research Institute of Grenoble, CytoMorpho Lab, Grenoble38054, France
- University of Paris, INSERM, Commissariat à l'énergie atomique et aux énergies alternatives, UMRS1160, Institut de Recherche Saint Louis, CytoMorpho Lab, Hôpital Saint Louis, Paris75010, France
| |
Collapse
|
19
|
Loffet EA, Durel JF, Kam R, Lim H, Nerurkar NL. ELASTIC FIBERS DEFINE EMBRYONIC TISSUE STIFFNESS TO ENABLE BUCKLING MORPHOGENESIS OF THE SMALL INTESTINE. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.18.549562. [PMID: 37502968 PMCID: PMC10370103 DOI: 10.1101/2023.07.18.549562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
During embryonic development, tissues must possess precise material properties to ensure that cell-generated forces give rise to the stereotyped morphologies of developing organs. However, the question of how material properties are established and regulated during development remains understudied. Here, we aim to address these broader questions through the study of intestinal looping, a process by which the initially straight intestinal tube buckles into loops, permitting ordered packing within the body cavity. Looping results from elongation of the tube against the constraint of an attached tissue, the dorsal mesentery, which is elastically stretched by the elongating tube to nearly triple its length. This elastic energy storage allows the mesentery to provide stable compressive forces that ultimately buckle the tube into loops. Beginning with a transcriptomic analysis of the mesentery, we identified widespread upregulation of extracellular matrix related genes during looping, including genes related to elastic fiber deposition. Combining molecular and mechanical analyses, we conclude that elastin confers tensile stiffness to the mesentery, enabling its mechanical role in organizing the developing small intestine. These results shed light on the role of elastin as a driver of morphogenesis that extends beyond its more established role in resisting cyclic deformation in adult tissues.
Collapse
Affiliation(s)
- Elise A. Loffet
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - John F. Durel
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - Richard Kam
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - Hyunjee Lim
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - Nandan L. Nerurkar
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| |
Collapse
|
20
|
Oikonomou P, Cirne HC, Nerurkar NL. A chemo-mechanical model of endoderm movements driving elongation of the amniote hindgut. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.18.541363. [PMID: 37292966 PMCID: PMC10245718 DOI: 10.1101/2023.05.18.541363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
While mechanical and biochemical descriptions of development are each essential, integration of upstream morphogenic cues with downstream tissue mechanics remains understudied in many contexts during vertebrate morphogenesis. A posterior gradient of Fibroblast Growth Factor (FGF) ligands generates a contractile force gradient in the definitive endoderm, driving collective cell movements to form the hindgut. Here, we developed a two-dimensional chemo-mechanical model to investigate how mechanical properties of the endoderm and transport properties of FGF coordinately regulate this process. We began by formulating a 2-D reaction-diffusion-advection model that describes the formation of an FGF protein gradient due to posterior displacement of cells transcribing unstable Fgf8 mRNA during axis elongation, coupled with translation, diffusion, and degradation of FGF protein. This was used together with experimental measurements of FGF activity in the chick endoderm to inform a continuum model of definitive endoderm as an active viscous fluid that generates contractile stresses in proportion to FGF concentration. The model replicated key aspects of hindgut morphogenesis, confirms that heterogeneous - but isotropic - contraction is sufficient to generate large anisotropic cell movements, and provides new insight into how chemo-mechanical coupling across the mesoderm and endoderm coordinates hindgut elongation with outgrowth of the tailbud.
Collapse
Affiliation(s)
| | - Helena C. Cirne
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - Nandan L. Nerurkar
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| |
Collapse
|
21
|
Lemma LM, Varghese M, Ross TD, Thomson M, Baskaran A, Dogic Z. Spatio-temporal patterning of extensile active stresses in microtubule-based active fluids. PNAS NEXUS 2023; 2:pgad130. [PMID: 37168671 PMCID: PMC10165807 DOI: 10.1093/pnasnexus/pgad130] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 02/27/2023] [Accepted: 04/03/2023] [Indexed: 05/13/2023]
Abstract
Microtubule-based active fluids exhibit turbulent-like autonomous flows, which are driven by the molecular motor powered motion of filamentous constituents. Controlling active stresses in space and time is an essential prerequisite for controlling the intrinsically chaotic dynamics of extensile active fluids. We design single-headed kinesin molecular motors that exhibit optically enhanced clustering and thus enable precise and repeatable spatial and temporal control of extensile active stresses. Such motors enable rapid, reversible switching between flowing and quiescent states. In turn, spatio-temporal patterning of the active stress controls the evolution of the ubiquitous bend instability of extensile active fluids and determines its critical length dependence. Combining optically controlled clusters with conventional kinesin motors enables one-time switching from contractile to extensile active stresses. These results open a path towards real-time control of the autonomous flows generated by active fluids.
Collapse
Affiliation(s)
- Linnea M Lemma
- Department of Physics, Brandeis University, 415 South St., Waltham, 02453 MA, USA
- Department of Physics, University of California, Santa Barbara, 93106 CA, USA
| | - Minu Varghese
- Department of Physics, Brandeis University, 415 South St., Waltham, 02453 MA, USA
| | - Tyler D Ross
- Department of Computing and Mathematical Sciences, California Institute of Technology, 1200 E California Blvd. Pasadena, 91125 CA, USA
| | - Matt Thomson
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 E California Blvd., Pasadena, 91125 CA, USA
| | - Aparna Baskaran
- Department of Physics, Brandeis University, 415 South St., Waltham, 02453 MA, USA
| | | |
Collapse
|
22
|
Zhang Z, Li GY, Jiang Y, Zheng Y, Gower AL, Destrade M, Cao Y. Noninvasive measurement of local stress inside soft materials with programmed shear waves. SCIENCE ADVANCES 2023; 9:eadd4082. [PMID: 36888699 PMCID: PMC9995030 DOI: 10.1126/sciadv.add4082] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 02/07/2023] [Indexed: 06/18/2023]
Abstract
Mechanical stresses across different length scales play a fundamental role in understanding biological systems' functions and engineering soft machines and devices. However, it is challenging to noninvasively probe local mechanical stresses in situ, particularly when the mechanical properties are unknown. We propose an acoustoelastic imaging-based method to infer the local stresses in soft materials by measuring the speeds of shear waves induced by custom-programmed acoustic radiation force. Using an ultrasound transducer to excite and track the shear waves remotely, we demonstrate the application of the method by imaging uniaxial and bending stresses in an isotropic hydrogel and the passive uniaxial stress in a skeletal muscle. These measurements were all done without the knowledge of the constitutive parameters of the materials. The experiments indicate that our method will find broad applications, ranging from health monitoring of soft structures and machines to diagnosing diseases that alter stresses in soft tissues.
Collapse
Affiliation(s)
- Zhaoyi Zhang
- Institute of Biomechanics and Medical Engineering, AML, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, P.R. China
| | - Guo-Yang Li
- Harvard Medical School and Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02139, USA
| | - Yuxuan Jiang
- Institute of Biomechanics and Medical Engineering, AML, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, P.R. China
| | - Yang Zheng
- Institute of Biomechanics and Medical Engineering, AML, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, P.R. China
| | - Artur L. Gower
- Department of Mechanical Engineering, University of Sheffield, Sheffield, UK
| | - Michel Destrade
- School of Mathematical and Statistical Sciences, University of Galway, Galway, Ireland
- Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province and Department of Engineering Mechanics, Zhejiang University, Hangzhou 310027, Zhejiang Province, P.R. China
| | - Yanping Cao
- Institute of Biomechanics and Medical Engineering, AML, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, P.R. China
| |
Collapse
|
23
|
Hack JM, Anwar NZ, Jackson JG, Furth ME, Varner VD. Quantifying endodermal strains during heart tube formation in the developing chicken embryo. J Biomech 2023; 149:111481. [PMID: 36787674 PMCID: PMC10163833 DOI: 10.1016/j.jbiomech.2023.111481] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 12/17/2022] [Accepted: 02/01/2023] [Indexed: 02/09/2023]
Abstract
In the early avian embryo, the developing heart forms when bilateral fields of cardiac progenitor cells, which reside in the lateral plate mesoderm, move toward the embryonic midline, and fuse above the anterior intestinal portal (AIP) to form a straight, muscle-wrapped tube. During this process, the precardiac mesoderm remains in close contact with the underlying endoderm. Previous work has shown that the endoderm around the AIP actively contracts to pull the cardiac progenitors toward the midline. The morphogenetic deformations associated with this endodermal convergence, however, remain unclear, as do the signaling pathways that might regulate this process. Here, we fluorescently labeled populations of endodermal cells in early chicken embryos and tracked their motion during heart tube formation to compute time-varying strains along the anterior endoderm. We then determined how the computed endodermal strain distributions are affected by the pharmacological inhibition of either myosin II or fibroblast growth factor (FGF) signaling. Our data indicate that a mediolateral gradient in endodermal shortening is present around the AIP, as well as substantial convergence and extension movements both anterior and lateral to the AIP. These active endodermal deformations are disrupted if either actomyosin contractility or FGF signaling are inhibited pharmacologically. Taken together, these results demonstrate how active deformations along the anterior endoderm contribute to heart tube formation within the developing embryo.
Collapse
Affiliation(s)
- Joshua M Hack
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
| | - Nareen Z Anwar
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
| | - John G Jackson
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
| | - Meagan E Furth
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
| | - Victor D Varner
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States; Department of Surgery, UT Southwestern Medical Center, Dallas, TX, United States.
| |
Collapse
|
24
|
Chevalier NR. Physical organogenesis of the gut. Development 2022; 149:276365. [DOI: 10.1242/dev.200765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
ABSTRACT
The gut has been a central subject of organogenesis since Caspar Friedrich Wolff’s seminal 1769 work ‘De Formatione Intestinorum’. Today, we are moving from a purely genetic understanding of cell specification to a model in which genetics codes for layers of physical–mechanical and electrical properties that drive organogenesis such that organ function and morphogenesis are deeply intertwined. This Review provides an up-to-date survey of the extrinsic and intrinsic mechanical forces acting on the embryonic vertebrate gut during development and of their role in all aspects of intestinal morphogenesis: enteric nervous system formation, epithelium structuring, muscle orientation and differentiation, anisotropic growth and the development of myogenic and neurogenic motility. I outline numerous implications of this biomechanical perspective in the etiology and treatment of pathologies, such as short bowel syndrome, dysmotility, interstitial cells of Cajal-related disorders and Hirschsprung disease.
Collapse
Affiliation(s)
- Nicolas R. Chevalier
- Laboratoire Matière et Systèmes Complexes, Université Paris Cité, CNRS UMR 7057 , 10 rue Alice Domon et Léonie Duquet, 75013 Paris , France
| |
Collapse
|
25
|
Ohtsuka D, Kida N, Lee SW, Kawahira N, Morishita Y. Cell disorientation by loss of SHH-dependent mechanosensation causes cyclopia. SCIENCE ADVANCES 2022; 8:eabn2330. [PMID: 35857502 PMCID: PMC9278851 DOI: 10.1126/sciadv.abn2330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
The physical causes of organ malformation remain largely unclear in most cases due to a lack of information on tissue/cell dynamics. Here, we address this issue by considering onset of cyclopia in sonic hedgehog (SHH)-inhibited chick embryos. We show that ventral forebrain-specific self-organization ability driven by SHH-dependent polarized patterns in cell shape, phosphorylated myosin localization, and collective cell motion promotes optic vesicle elongation during normal development. Stress loading tests revealed that these polarized dynamics result from mechanical responses. In particular, stress and active tissue deformation satisfy orthogonality, defining an SHH-regulated morphogenetic law. Without SHH signaling, cells cannot detect the direction of stress and move randomly, leading to insufficient optic vesicle elongation and consequently a cyclopia phenotype. Since polarized tissue/cell dynamics are common in organogenesis, cell disorientation caused by loss of mechanosensation could be a pathogenic mechanism for other malformations.
Collapse
Affiliation(s)
- Daisuke Ohtsuka
- Laboratory for Developmental Morphogeometry, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
- Corresponding author. (Y.M.); (D.O.)
| | - Naoki Kida
- Laboratory for Developmental Morphogeometry, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Sang-Woo Lee
- Laboratory for Developmental Morphogeometry, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Naofumi Kawahira
- Laboratory for Developmental Morphogeometry, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
- Department of Molecular Cell Developmental Biology, School of Life Science, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Yoshihiro Morishita
- Laboratory for Developmental Morphogeometry, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
- Corresponding author. (Y.M.); (D.O.)
| |
Collapse
|
26
|
Mitchell NP, Cislo DJ, Shankar S, Lin Y, Shraiman BI, Streichan SJ. Visceral organ morphogenesis via calcium-patterned muscle constrictions. eLife 2022; 11:e77355. [PMID: 35593701 PMCID: PMC9275821 DOI: 10.7554/elife.77355] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 05/08/2022] [Indexed: 11/24/2022] Open
Abstract
Organ architecture is often composed of multiple laminar tissues arranged in concentric layers. During morphogenesis, the initial geometry of visceral organs undergoes a sequence of folding, adopting a complex shape that is vital for function. Genetic signals are known to impact form, yet the dynamic and mechanical interplay of tissue layers giving rise to organs' complex shapes remains elusive. Here, we trace the dynamics and mechanical interactions of a developing visceral organ across tissue layers, from subcellular to organ scale in vivo. Combining deep tissue light-sheet microscopy for in toto live visualization with a novel computational framework for multilayer analysis of evolving complex shapes, we find a dynamic mechanism for organ folding using the embryonic midgut of Drosophila as a model visceral organ. Hox genes, known regulators of organ shape, control the emergence of high-frequency calcium pulses. Spatiotemporally patterned calcium pulses trigger muscle contractions via myosin light chain kinase. Muscle contractions, in turn, induce cell shape change in the adjacent tissue layer. This cell shape change collectively drives a convergent extension pattern. Through tissue incompressibility and initial organ geometry, this in-plane shape change is linked to out-of-plane organ folding. Our analysis follows tissue dynamics during organ shape change in vivo, tracing organ-scale folding to a high-frequency molecular mechanism. These findings offer a mechanical route for gene expression to induce organ shape change: genetic patterning in one layer triggers a physical process in the adjacent layer - revealing post-translational mechanisms that govern shape change.
Collapse
Affiliation(s)
- Noah P Mitchell
- Kavli Institute for Theoretical Physics, University of California, Santa BarbaraSanta BarbaraUnited States
- Department of Physics, University of California, Santa BarbaraSanta BarbaraUnited States
| | - Dillon J Cislo
- Department of Physics, University of California, Santa BarbaraSanta BarbaraUnited States
| | - Suraj Shankar
- Kavli Institute for Theoretical Physics, University of California, Santa BarbaraSanta BarbaraUnited States
- Department of Physics, Harvard UniversityCambridgeUnited States
| | - Yuzheng Lin
- Department of Physics, University of California, Santa BarbaraSanta BarbaraUnited States
| | - Boris I Shraiman
- Kavli Institute for Theoretical Physics, University of California, Santa BarbaraSanta BarbaraUnited States
| | - Sebastian J Streichan
- Department of Physics, University of California, Santa BarbaraSanta BarbaraUnited States
- Biomolecular Science and Engineering, University of California, Santa BarbaraSanta BarbaraUnited States
| |
Collapse
|
27
|
Mechanosignaling in vertebrate development. Dev Biol 2022; 488:54-67. [DOI: 10.1016/j.ydbio.2022.05.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/06/2022] [Accepted: 05/07/2022] [Indexed: 12/13/2022]
|
28
|
Regev I, Guevorkian K, Gupta A, Pourquié O, Mahadevan L. Rectified random cell motility as a mechanism for embryo elongation. Development 2022; 149:274852. [PMID: 35344041 PMCID: PMC9017234 DOI: 10.1242/dev.199423] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 01/25/2022] [Indexed: 12/24/2022]
Abstract
ABSTRACT
The body of vertebrate embryos forms by posterior elongation from a terminal growth zone called the tail bud. The tail bud is a source of highly motile cells that eventually constitute the presomitic mesoderm (PSM), a tissue that plays an important role in elongation movements. PSM cells establish an anterior-posterior cell motility gradient that parallels a gradient associated with the degradation of a specific cellular signal (FGF) known to be implicated in cell motility. Here, we combine the electroporation of fluorescent reporters in the PSM with time-lapse imaging in the chicken embryo to quantify cell diffusive movements along the motility gradient. We show that a simple microscopic model for random cell motility induced by FGF activity along with geometric confinement leads to rectified tissue elongation consistent with our observations. A continuum analog of the microscopic model leads to a macroscopic mechano-chemical model for tissue extension that couples FGF activity-induced cell motility and tissue rheology, and is consistent with the experimentally observed speed and extent of elongation. Together, our experimental observations and theoretical models explain how the continuous addition of cells at the tail bud combined with lateral confinement can be converted into oriented movement and drive body elongation.
Collapse
Affiliation(s)
- Ido Regev
- Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
- Department of Solar Energy and Environmental Physics, Jacob Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Sede Boqer Campus 84990, Israel
| | - Karine Guevorkian
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, CNRS, Inserm, Illkirch, France
- Harvard Medical School, Department of Genetics, Brigham and Women's Hospital, Department of Pathology, Boston, MA 02115, USA
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico-Chimie Curie, 75005 Paris, France
| | - Anupam Gupta
- Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Indian Institute of Technology Hyderabad, Telangana 502285, India
| | - Olivier Pourquié
- Harvard Medical School, Department of Genetics, Brigham and Women's Hospital, Department of Pathology, Boston, MA 02115, USA
| | - L. Mahadevan
- Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
- Department of Physics and Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
29
|
Leng M, Peng Y, Wang H. [Research advances on the biomechanical micro- environment facilitated wound repair through the regulation of cell migration]. ZHONGHUA SHAO SHANG YU CHUANG MIAN XIU FU ZA ZHI 2022; 38:90-94. [PMID: 35152690 DOI: 10.3760/cma.j.cn501120-20200921-00419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Biomechanical microenvironment refers to a variety of mechanical signals in the extracellular mechanical microenvironment, which will change correspondingly with time and space. It plays an important role in histological changes such as cell migration, proliferation, and differentiation, and can further affect wound healing. Wound healing is a complex pathophysiological process, and one of the important factors that affects wound healing is whether the cells can efficiently and quickly migrate to the wound center or not. Previous studies have shown that biomechanical microenvironment can not only induce the directional migration of cells, but also improve the migration rate of cells. In the complex natural environment, cells adopt various migration patterns and are dominated by special patterns such as local myosin contractility and extracellular microenvironment. In addition to overcoming the extracellular barrier, cells also need to interact with neighboring cells and tissue through local physical and mechanical forces and signals to complete migration and thus accelerate wound healing. Therefore, in recent years, scholars at home and abroad have been actively developing biological materials based on improving biomechanical microenvironment in order to further promote cell migration and thus accelerate wound healing. This paper reviews the recent research advances on the role of biomechanical environment in wound healing promotion via the regulating of cell migration and the development of related biomaterials.
Collapse
Affiliation(s)
- M Leng
- Institute of Burns, the Second Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Y Peng
- Institute of Burns, the Second Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - H Wang
- Institute of Burns, the Second Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| |
Collapse
|
30
|
Suong DNA, Imamura K, Inoue I, Kabai R, Sakamoto S, Okumura T, Kato Y, Kondo T, Yada Y, Klein WL, Watanabe A, Inoue H. Induction of inverted morphology in brain organoids by vertical-mixing bioreactors. Commun Biol 2021; 4:1213. [PMID: 34686776 PMCID: PMC8536773 DOI: 10.1038/s42003-021-02719-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 09/28/2021] [Indexed: 12/19/2022] Open
Abstract
Organoid technology provides an opportunity to generate brain-like structures by recapitulating developmental steps in the manner of self-organization. Here we examined the vertical-mixing effect on brain organoid structures using bioreactors and established inverted brain organoids. The organoids generated by vertical mixing showed neurons that migrated from the outer periphery to the inner core of organoids, in contrast to orbital mixing. Computational analysis of flow dynamics clarified that, by comparison with orbital mixing, vertical mixing maintained the high turbulent energy around organoids, and continuously kept inter-organoid distances by dispersing and adding uniform rheological force on organoids. To uncover the mechanisms of the inverted structure, we investigated the direction of primary cilia, a cellular mechanosensor. Primary cilia of neural progenitors by vertical mixing were aligned in a multidirectional manner, and those by orbital mixing in a bidirectional manner. Single-cell RNA sequencing revealed that neurons of inverted brain organoids presented a GABAergic character of the ventral forebrain. These results suggest that controlling fluid dynamics by biomechanical engineering can direct stem cell differentiation of brain organoids, and that inverted brain organoids will be applicable for studying human brain development and disorders in the future. Dang Ngoc Anh Suong et al find that vertical mixing generates iPSC-derived brain organoids displaying an inverted structure with neurons localising at the centre and neural progenitors at the outside. This study illustrates the influence of fluid mechanics relevant to the direction of primary cilia on stem cell differentiation.
Collapse
Affiliation(s)
- Dang Ngoc Anh Suong
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.,iPSC-based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan
| | - Keiko Imamura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.,iPSC-based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan.,Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| | - Ikuyo Inoue
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.,Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| | - Ryotaro Kabai
- Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | - Yoshikazu Kato
- Mixing Technology Laboratory, SATAKE Chemical Equipment Manufacturing Ltd., Saitama, Japan
| | - Takayuki Kondo
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.,iPSC-based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan.,Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| | - Yuichiro Yada
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.,iPSC-based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan
| | - William L Klein
- Department of Neurobiology, Northwestern University, Evanston, IL, 60208, USA
| | - Akira Watanabe
- Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Haruhisa Inoue
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan. .,iPSC-based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan. .,Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan. .,Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan.
| |
Collapse
|
31
|
Zhu Y, Deng S, Zhao X, Xia G, Zhao R, Chan HF. Deciphering and engineering tissue folding: A mechanical perspective. Acta Biomater 2021; 134:32-42. [PMID: 34325076 DOI: 10.1016/j.actbio.2021.07.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 07/16/2021] [Accepted: 07/21/2021] [Indexed: 12/19/2022]
Abstract
The folding of tissues/organs into complex shapes is a common phenomenon that occurs in organisms such as animals and plants, and is both structurally and functionally important. Deciphering the process of tissue folding and applying this knowledge to engineer folded systems would significantly advance the field of tissue engineering. Although early studies focused on investigating the biochemical signaling events that occur during the folding process, the physical or mechanical aspects of the process have received increasing attention in recent years. In this review, we will summarize recent findings on the mechanical aspects of folding and introduce strategies by which folding can be controlled in vitro. Emphasis will be placed on the folding events triggered by mechanical effects at the cellular and tissue levels and on the different cell- and biomaterial-based approaches used to recapitulate folding. Finally, we will provide a perspective on the development of engineering tissue folding toward preclinical and clinical translation. STATEMENT OF SIGNIFICANCE: Tissue folding is a common phenomenon in a variety of organisms including human, and has been shown to serve important structural and functional roles. Understanding how folding forms and applying the concept in tissue engineering would represent an advance of the research field. Recently, the physical or mechanical aspect of tissue folding has gained increasing attention. In this review, we will cover recent findings of the mechanical aspect of folding mechanisms, and introduce strategies to control the folding process in vitro. We will also provide a perspective on the future development of the field towards preclinical and clinical translation of various bio fabrication technologies.
Collapse
Affiliation(s)
- Yanlun Zhu
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Shuai Deng
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Xiaoyu Zhao
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; Shun Hing Institute of Advanced Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Guanggai Xia
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Rd, Shanghai 200233, China
| | - Ruike Zhao
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; Shun Hing Institute of Advanced Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China; Hong Kong Branch of CAS Center for Excellence in Animal Evolution and Genetics, Hong Kong SAR, China.
| |
Collapse
|
32
|
Blackley DG, Cooper JH, Pokorska P, Ratheesh A. Mechanics of developmental migration. Semin Cell Dev Biol 2021; 120:66-74. [PMID: 34275746 DOI: 10.1016/j.semcdb.2021.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 02/01/2023]
Abstract
The ability to migrate is a fundamental property of animal cells which is essential for development, homeostasis and disease progression. Migrating cells sense and respond to biochemical and mechanical cues by rapidly modifying their intrinsic repertoire of signalling molecules and by altering their force generating and transducing machinery. We have a wealth of information about the chemical cues and signalling responses that cells use during migration. Our understanding of the role of forces in cell migration is rapidly evolving but is still best understood in the context of cells migrating in 2D and 3D environments in vitro. Advances in live imaging of developing embryos combined with the use of experimental and theoretical tools to quantify and analyse forces in vivo, has begun to shed light on the role of mechanics in driving embryonic cell migration. In this review, we focus on the recent studies uncovering the physical basis of embryonic cell migration in vivo. We look at the physical basis of the classical steps of cell migration such as protrusion formation and cell body translocation and review the recent research on how these processes work in the complex 3D microenvironment of a developing organism.
Collapse
Affiliation(s)
- Deannah G Blackley
- Warwick Medical School and Centre for Mechanochemical Cell Biology, Gibbet Hill Campus, University of Warwick, Coventry CV4 7AL, UK
| | - Jack H Cooper
- Warwick Medical School and Centre for Mechanochemical Cell Biology, Gibbet Hill Campus, University of Warwick, Coventry CV4 7AL, UK
| | - Paulina Pokorska
- Warwick Medical School and Centre for Mechanochemical Cell Biology, Gibbet Hill Campus, University of Warwick, Coventry CV4 7AL, UK
| | - Aparna Ratheesh
- Warwick Medical School and Centre for Mechanochemical Cell Biology, Gibbet Hill Campus, University of Warwick, Coventry CV4 7AL, UK.
| |
Collapse
|
33
|
Al-Izzi SC, Morris RG. Active flows and deformable surfaces in development. Semin Cell Dev Biol 2021; 120:44-52. [PMID: 34266757 DOI: 10.1016/j.semcdb.2021.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 12/15/2022]
Abstract
We review progress in active hydrodynamic descriptions of flowing media on curved and deformable manifolds: the state-of-the-art in continuum descriptions of single-layers of epithelial and/or other tissues during development. First, after a brief overview of activity, flows and hydrodynamic descriptions, we highlight the generic challenge of identifying the dependence on dynamical variables of so-called active kinetic coefficients- active counterparts to dissipative Onsager coefficients. We go on to describe some of the subtleties concerning how curvature and active flows interact, and the issues that arise when surfaces are deformable. We finish with a broad discussion around the utility of such theories in developmental biology. This includes limitations to analytical techniques, challenges associated with numerical integration, fitting-to-data and inference, and potential tools for the future, such as discrete differential geometry.
Collapse
Affiliation(s)
- Sami C Al-Izzi
- School of Physics and EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales - Sydney, 2052, Australia
| | - Richard G Morris
- School of Physics and EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales - Sydney, 2052, Australia.
| |
Collapse
|
34
|
Abstract
Morphogenesis is one of the most remarkable examples of biological pattern formation. Despite substantial progress in the field, we still do not understand the organizational principles responsible for the robust convergence of the morphogenesis process across scales to form viable organisms under variable conditions. Achieving large-scale coordination requires feedback between mechanical and biochemical processes, spanning all levels of organization and relating the emerging patterns with the mechanisms driving their formation. In this review, we highlight the role of mechanics in the patterning process, emphasizing the active and synergistic manner in which mechanical processes participate in developmental patterning rather than merely following a program set by biochemical signals. We discuss the value of applying a coarse-grained approach toward understanding this complex interplay, which considers the large-scale dynamics and feedback as well as complementing the reductionist approach focused on molecular detail. A central challenge in this approach is identifying relevant coarse-grained variables and developing effective theories that can serve as a basis for an integrated framework for understanding this remarkable pattern-formation process. Expected final online publication date for the Annual Review of Cell and Developmental Biology, Volume 37 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Yonit Maroudas-Sacks
- Department of Physics, Technion-Israel Institute of Technology, Haifa 32000, Israel;
| | - Kinneret Keren
- Department of Physics, Technion-Israel Institute of Technology, Haifa 32000, Israel; .,Network Biology Research Laboratories and The Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 32000, Israel
| |
Collapse
|
35
|
Rezalotfi A, Vrynas AV, Dehghanian M, Rezaei N. Lessons from the Embryo: an Unrejected Transplant and a Benign Tumor. Stem Cell Rev Rep 2021; 17:850-861. [PMID: 33225425 DOI: 10.1007/s12015-020-10088-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2020] [Indexed: 10/22/2022]
Abstract
Embryogenesis is regarded the 'miracle of life', yet numerous aspects of this process are not fully understood. As the embryo grows in the mother's womb, immune components, stem cells and microenvironmental cues cooperate among others to promote embryonic development. Evidently, these key players are frequently associated with transplantation failure and tumor growth. While the fields of transplantation and cancer biology do not overlap, both can be viewed from the perspective of an embryo. As an 'unrejected transplant' and a 'benign tumor', lessons from embryonic development may reveal features of transplants and tumors that have been overlooked. Therefore, eavesdropping at these natural complex events during pregnancy may inspire more durable approaches to arrest transplant rejection or cancer progression.
Collapse
Affiliation(s)
- Alaleh Rezalotfi
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | | | - Maryam Dehghanian
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Children's Medical Center Hospital, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran.
| |
Collapse
|
36
|
Abstract
The generation of organismal form - morphogenesis - arises from forces produced at the cellular level. In animal cells, much of this force is produced by the actin cytoskeleton. Here, we review how mechanisms of actin-based force generation are deployed during animal morphogenesis to sculpt organs and organisms. Furthermore, we consider how cytoskeletal forces are coupled through cell adhesions to propagate across tissues, and discuss cases where cytoskeletal force or adhesion is patterned across a tissue to direct shape changes. Together, our review provides a conceptual framework that reflects our current understanding of animal morphogenesis and gives perspectives on future opportunities for study.
Collapse
Affiliation(s)
- D Nathaniel Clarke
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Adam C Martin
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| |
Collapse
|
37
|
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
|
38
|
Durel JF, Nerurkar NL. Mechanobiology of vertebrate gut morphogenesis. Curr Opin Genet Dev 2020; 63:45-52. [PMID: 32413823 DOI: 10.1016/j.gde.2020.04.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/09/2020] [Indexed: 01/15/2023]
Abstract
Approximately a century after D'Arcy Thompson's On Growth and Form, there continues to be widespread interest in the biophysical and mathematical basis of morphogenesis. Particularly over the past 20 years, this interest has led to great advances in our understanding of a broad range of processes in embryonic development through a quantitative, mechanically driven framework. Nowhere in vertebrate development is this more apparent than the development of endodermally derived organs. Here, we discuss recent advances in the study of gut development that have emerged primarily from mechanobiology-motivated approaches that span from gut tube morphogenesis and later organogenesis of the respiratory and gastrointestinal systems.
Collapse
Affiliation(s)
- John F Durel
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States
| | - Nandan L Nerurkar
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States; Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, United States.
| |
Collapse
|
39
|
Garriock RJ, Chalamalasetty RB, Zhu J, Kennedy MW, Kumar A, Mackem S, Yamaguchi TP. A dorsal-ventral gradient of Wnt3a/β-catenin signals controls mouse hindgut extension and colon formation. Development 2020; 147:dev.185108. [PMID: 32156757 DOI: 10.1242/dev.185108] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 02/19/2020] [Indexed: 12/20/2022]
Abstract
Despite the importance of Wnt signaling for adult intestinal stem cell homeostasis and colorectal cancer, relatively little is known about its role in colon formation during embryogenesis. The development of the colon starts with the formation and extension of the hindgut. We show that Wnt3a is expressed in the caudal embryo in a dorsal-ventral (DV) gradient across all three germ layers, including the hindgut. Using genetic and lineage-tracing approaches, we describe novel dorsal and ventral hindgut domains, and show that ventrolateral hindgut cells populate the majority of the colonic epithelium. A Wnt3a-β-catenin-Sp5/8 pathway, which is active in the dorsal hindgut endoderm, is required for hindgut extension and colon formation. Interestingly, the absence of Wnt activity in the ventral hindgut is crucial for proper hindgut morphogenesis, as ectopic stabilization of β-catenin in the ventral hindgut via gain- or loss-of-function mutations in Ctnnb1 or Apc, respectively, leads to severe colonic hyperplasia. Thus, the DV Wnt gradient is required to coordinate growth between dorsal and ventral hindgut domains to regulate the extension of the hindgut that leads to colon formation.
Collapse
Affiliation(s)
- Robert J Garriock
- Center for Cancer Research, Cancer and Developmental Biology Laboratory, Cell Signaling in Vertebrate Development Section, NCI-Frederick, NIH, Frederick, MD 21702, USA
| | - Ravindra B Chalamalasetty
- Center for Cancer Research, Cancer and Developmental Biology Laboratory, Cell Signaling in Vertebrate Development Section, NCI-Frederick, NIH, Frederick, MD 21702, USA
| | - JianJian Zhu
- Center for Cancer Research, Cancer and Developmental Biology Laboratory, Cell Signaling in Vertebrate Development Section, NCI-Frederick, NIH, Frederick, MD 21702, USA
| | - Mark W Kennedy
- Center for Cancer Research, Cancer and Developmental Biology Laboratory, Cell Signaling in Vertebrate Development Section, NCI-Frederick, NIH, Frederick, MD 21702, USA
| | - Amit Kumar
- Center for Cancer Research, Cancer and Developmental Biology Laboratory, Cell Signaling in Vertebrate Development Section, NCI-Frederick, NIH, Frederick, MD 21702, USA
| | - Susan Mackem
- Center for Cancer Research, Cancer and Developmental Biology Laboratory, Cell Signaling in Vertebrate Development Section, NCI-Frederick, NIH, Frederick, MD 21702, USA
| | - Terry P Yamaguchi
- Center for Cancer Research, Cancer and Developmental Biology Laboratory, Cell Signaling in Vertebrate Development Section, NCI-Frederick, NIH, Frederick, MD 21702, USA
| |
Collapse
|
40
|
Abstract
Cell migration is essential for physiological processes as diverse as development, immune defence and wound healing. It is also a hallmark of cancer malignancy. Thousands of publications have elucidated detailed molecular and biophysical mechanisms of cultured cells migrating on flat, 2D substrates of glass and plastic. However, much less is known about how cells successfully navigate the complex 3D environments of living tissues. In these more complex, native environments, cells use multiple modes of migration, including mesenchymal, amoeboid, lobopodial and collective, and these are governed by the local extracellular microenvironment, specific modalities of Rho GTPase signalling and non-muscle myosin contractility. Migration through 3D environments is challenging because it requires the cell to squeeze through complex or dense extracellular structures. Doing so requires specific cellular adaptations to mechanical features of the extracellular matrix (ECM) or its remodelling. In addition, besides navigating through diverse ECM environments and overcoming extracellular barriers, cells often interact with neighbouring cells and tissues through physical and signalling interactions. Accordingly, cells need to call on an impressively wide diversity of mechanisms to meet these challenges. This Review examines how cells use both classical and novel mechanisms of locomotion as they traverse challenging 3D matrices and cellular environments. It focuses on principles rather than details of migratory mechanisms and draws comparisons between 1D, 2D and 3D migration.
Collapse
Affiliation(s)
- Kenneth M Yamada
- Cell Biology Section, Division of Intramural Research, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA.
| | - Michael Sixt
- Institute of Science and Technology Austria, Klosterneuburg, Austria.
| |
Collapse
|
41
|
Abstract
Organoids are multicellular structures that can be derived from adult organs or pluripotent stem cells. Early versions of organoids range from simple epithelial structures to complex, disorganized tissues with large cellular diversity. The current challenge is to engineer cellular complexity into organoids in a controlled manner that results in organized assembly and acquisition of tissue function. These efforts have relied on studies of organ assembly during embryonic development and have resulted in the development of organoids with multilayer tissue complexity and higher-order functions. We discuss how the next generation of organoids can be designed by means of an engineering-based narrative design to control patterning, assembly, morphogenesis, growth, and function.
Collapse
Affiliation(s)
- Takanori Takebe
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Institute of Research, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - James M Wells
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
42
|
Verger S, Liu M, Hamant O. Mechanical Conflicts in Twisting Growth Revealed by Cell-Cell Adhesion Defects. FRONTIERS IN PLANT SCIENCE 2019; 10:173. [PMID: 30858857 PMCID: PMC6397936 DOI: 10.3389/fpls.2019.00173] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 02/01/2019] [Indexed: 05/31/2023]
Abstract
Many plants grow organs and tissues with twisted shapes. Arabidopsis mutants with impaired microtubule dynamics exhibit such a phenotype constitutively. Although the activity of the corresponding microtubule regulators is better understood at the molecular level, how large-scale twisting can emerge in the mutants remains largely unknown. Classically, oblique cortical microtubules would constrain the deposition of cellulose microfibrils in cells, and such conflicts at the cell level would be relaxed at the tissue scale by supracellular torsion. This model implicitly assumes that cell-cell adhesion is a key step to transpose local mechanical conflicts into a macroscopic twisting phenotype. Here we tested this prediction using the quasimodo1 mutant, which displays cell-cell adhesion defects. Using the spriral2/tortifolia1 mutant with hypocotyl helical growth, we found that qua1-induced cell-cell adhesion defects restore straight growth in qua1-1 spr2-2. Detached cells in qua1-1 spr2-2 displayed helical growth, confirming that straight growth results from the lack of mechanical coupling between cells rather than a restoration of SPR2 activity in the qua1 mutant. Because adhesion defects in qua1 depend on tension in the outer wall, we also showed that hypocotyl twisting in qua1-1 spr2-2 could be restored when decreasing the matrix potential of the growth medium, i.e., by reducing the magnitude of the pulling force between adjacent cells, in the double mutant. Interestingly, the induction of straight growth in qua1-1 spr2-2 could be achieved beyond hypocotyls, as leaves also displayed a flat phenotype in the double mutant. Altogether, these results provide formal experimental support for a scenario in which twisted growth in spr2 mutant would result from the relaxation of local mechanical conflicts between adjacent cells via global organ torsion.
Collapse
|