Skip to main content

EDITORIAL article

Front. Cell Dev. Biol., 30 January 2024
Sec. Cell Adhesion and Migration
This article is part of the Research Topic Mechanobiology of Organoid Systems View all 6 articles

Editorial: Mechanobiology of organoid systems

  • 1Clinical and Translational Epidemiology Unit, Department of Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, MA, United States
  • 2Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, Leipzig University, Leipzig, Germany
  • 3Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, MA, United States
  • 4Kilachand Center for Life Sciences and Engineering, Department of Biomedical Engineering, Boston University, Boston, MA, United States
  • 5Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States

Editorial on the Research Topic
Mechanobiology of organoid systems

Mechanobiology and diseases

Seminal studies in the mid 2000s reported that extracellular matrix (ECM) stiffness (Engler et al., 2006), cell shape and cellular contractility (McBeath et al., 2004) can direct mesenchymal stem cell differentiation. During the subsequent 20 years, the field of mechanobiology has been significant inspired and substantially developed. These studies helped to better understand how mechanical forces regulate complex cell behaviors and tissue functions (Han et al., 2018; Li et al., 2021a; Li et al., 2021b), and influence homeostasis and disease development (Chowdhury et al., 2021).

Mechanical forces play a pivotal role in regulating cellular biochemical signaling pathways, with reciprocal interactions influencing both cellular activities and mechanical properties in response to environmental cues (Han et al., 2018; Yang et al., 2023). These mutual interactions between mechanical forces and biochemical signaling pathways are critical to human health and disease development. In general, stiffening of the ECM during inflammatory diseases, fibrotic diseases or tumor development can regulate cellular signaling pathways, such as YAP/TAZ, via increasing cellular tractions on ECM, contributing to pathogenesis and exacerbating disease outcomes (Ingber, 2003; He et al., 2022; He et al., 2023). In the realm of glaucoma research, Du et al. uncovered a role for cellular senescence in disrupting the mechanoresponses of trabecular mesh cells (TMCs). Senescent TMCs, subjected to fluid shear stress, exhibited diminished F-actin formation, poor realignment of F-actin fibers, reduced cellular stiffness, and abnormal expression of ECM remodeling-related genes, compared to their non-senescent counterparts. In another study by Chi et al., deficiency in Integrin β4 expression led to increased lung tissue stiffness and elevated ECM components, such as collagen and elastin. Furthermore, Integrin β4 deficiency hindered the adaptation of bronchial epithelial cells to the ECM stiffening due to decreased cytoskeletal stabilization and impaired RhoA activity, ultimately contributing to the development of lung dysplasia.

In addition to affecting cytoskeletal proteins, mechanical forces can activate mechanosensitive Piezo proteins, which serve as pore-forming subunits of ion channels at the cell membrane. In response to mechanical stimuli, such as pressure, shear, and stretch, Piezo ion channels open and allow positively charged ions to flow into the cell, including calcium (Wu et al., 2017). As reviewed by George and Bates in this Research Topic, calcium oscillations occur in almost all cell types and tissues, playing a crucial role in morphogenesis and tissue development. Disruptions of these oscillations can lead to developmental abnormalities and pathogenesis. However, the underlying mechanisms by which mechanical stimuli impact bioelectrical signals and associated pathophysiological functions remains unclear.

Mechanobiology in organoid systems

The role of mechanical forces in cell proliferation, differentiation, and migration have been extensively studied (He et al., 2014; He et al., 2015; Guo et al., 2017; He et al., 2019; He et al., 2023). However, due to the complexity of living organisms, it is challenging to interpret how these mechano-biochemical coupling signaling pathways impact complex organ-level functions. The emerging technique of organoid culture provides a feasible platform recapitulating in vivo organ anatomy and functions for researchers to connect the cellular level mechanisms with the organ-level behaviors, including organoids of brain, lung, kidney, and gut, among others. In this Research Topic, Nauryzgaliyeva et al. presented a comprehensive reviews wherein they introduced the cutting-edge human pluripotent stem cells (hPSCs)- kidney organoid culture which faithfully captures in vivo kidney development and diseases. As they pointed out, the mechanical cues have largely been unexplored within hPSCs-derived organoid cultures. These studies in the future will help to better understand their impact on organ development and disease pathogenesis. They comprehensively reviewed the state-of-the-art techniques to interrogate organoid mechanobiology, including mimicking the extraembryonic microenvironment, using natural or synthetic substrates, combining with microfluidic devices, manipulating mechanosensing and mechanotransduction machineries, and measuring forces in complex organoids.

Regarding quantification of mechanical forces in 3D system, like organoids, Tian et al. reported a novel strategy in this Research Topic to analyze E-cadherin mediated intercellular forces using a series of DNA-hairpin molecular probes which they have developed for 2D cell models (Zhao et al., 2017; Zhao et al., 2020; Kes et al., 2021). Excitingly, after 1–2 h of incubation, these small molecule probes can penetrate a dosage-dependent depth of 50–200 µm of various 3D spheroids, including embryonic stem cell-derived embryoid bodies with strong cell-cell junctions. Combined with confocal microscopy or potentially more advanced imaging tools such as light sheet microscopy, this advanced technology will facilitate the quantification of complex intercellular mechanical interactions within 3D organoids.

Organoids and mechanomedicine

Throughout daily life, cells, composing living organisms, experience various mechanical forces, such as stretch, shear and pressure, as well as encounter different material properties, including varying stiffness, viscosity, surface roughness, and geometries. These constitutive/inherent mechanical cues can regulate cellular signaling pathways and reshape functions of muscles, bones, heart, and other organs, ultimately impacting human health as aforementioned. Targeting mechanosensing pathways is indispensable to tackle diseases and improve human health. Organoid-based systems bridge the cellular level signaling pathways with organ level functions in basic research and clinical studies, which are guaranteed to provide a powerful system for the fields of mechanobiology and mechanomedicine.

Author contributions

SH: Conceptualization, Funding acquisition, Writing–original draft, Writing–review and editing. CM: Conceptualization, Writing–original draft, Writing–review and editing. YS: Funding acquisition, Writing–original draft, Writing–review and editing. JE: Funding acquisition, Writing–original draft, Writing–review and editing. MG: Writing–original draft, Writing–review and editing.

Funding

The author(s) declare financial support was received for the research, authorship, and/or publication of this article. We acknowledge the support from the National Institutes of Health–National Cancer Institute (5R35CA253185-04) and Harvard Medical School Eleanor and Miles Shore Faculty Development Fellowship awards for SH; National Science Foundation (CMMI 1846866) for YS and (CMMI 2311640) for JE

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Chowdhury, F., Huang, B., and Wang, N. (2021). Cytoskeletal prestress: the cellular hallmark in mechanobiology and mechanomedicine. Cytoskeleton 78 (6), 249–276. doi:10.1002/cm.21658

PubMed Abstract | CrossRef Full Text | Google Scholar

Engler, A. J., Sen, S., Sweeney, H. L., and Discher, D. E. (2006). Matrix elasticity directs stem cell Lineage specification. Cell 126 (4), 677–689. doi:10.1016/j.cell.2006.06.044

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, M., Pegoraro, A. F., Mao, A., Zhou, E. H., Arany, P. R., Han, Y., et al. (2017). Cell volume change through water efflux impacts cell stiffness and stem cell fate. Proc. Natl. Acad. Sci. 114 (41), E8618–E27. doi:10.1073/pnas.1705179114

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, Y. L., Ronceray, P., Xu, G., Malandrino, A., Kamm, R. D., Lenz, M., et al. (2018). Cell contraction induces long-ranged stress stiffening in the extracellular matrix. Proc. Natl. Acad. Sci. 115 (16), 4075–4080. doi:10.1073/pnas.1722619115

PubMed Abstract | CrossRef Full Text | Google Scholar

He, S., Azar, D. A., Esfahani, F. N., Azar, G. A., Shazly, T., and Saeidi, N. (2022). Mechanoscopy: a novel device and procedure for in vivo detection of chronic colitis in mice. Inflamm. Bowel Dis. 28 (8), 1143–1150. doi:10.1093/ibd/izac046

PubMed Abstract | CrossRef Full Text | Google Scholar

He, S., Carman, C. V., Lee, J. H., Lan, B., Koehler, S., Atia, L., et al. (2019). The tumor suppressor p53 can promote collective cellular migration. PloS one 14 (2), e0202065. doi:10.1371/journal.pone.0202065

PubMed Abstract | CrossRef Full Text | Google Scholar

He, S., Lei, P., Kang, W., Cheung, P., Xu, T., Mana, M., et al. (2023). Stiffness restricts the stemness of the intestinal stem cells and skews their differentiation toward goblet cells. Gastroenterology 164, 1137–1151. doi:10.1053/j.gastro.2023.02.030

PubMed Abstract | CrossRef Full Text | Google Scholar

He, S., Liu, C., Li, X., Ma, S., Huo, B., and Ji, B. (2015). Dissecting collective cell behavior in polarization and alignment on micropatterned substrates. Biophysical J. 109 (3), 489–500. doi:10.1016/j.bpj.2015.06.058

PubMed Abstract | CrossRef Full Text | Google Scholar

He, S., Su, Y., Ji, B., and Gao, H. (2014). Some basic questions on mechanosensing in cell-substrate interaction. J. Mech. Phys. Solids 70, 116–135. doi:10.1016/j.jmps.2014.05.016

CrossRef Full Text | Google Scholar

Ingber, D. (2003). Mechanobiology and diseases of mechanotransduction. Ann. Med. 35 (8), 564–577. doi:10.1080/07853890310016333

PubMed Abstract | CrossRef Full Text | Google Scholar

Keshri, P., Zhao, B., Xie, T., Bagheri, Y., Chambers, J., Sun, Y., et al. (2021). Quantitative and multiplexed fluorescence lifetime imaging of intercellular tensile forces. Angew. Chem. Int. Ed. Engl. 60 (28), 15548–15555. doi:10.1002/anie.202103986

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Y., Chen, M., Hu, J., Sheng, R., Lin, Q., He, X., et al. (2021b). Volumetric compression induces intracellular crowding to control intestinal organoid growth via wnt/β-catenin signaling. Cell Stem Cell 28 (1), 63–78.e7. doi:10.1016/j.stem.2020.09.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Y., Tang, W., and Guo, M. (2021a). The cell as matter: connecting molecular biology to cellular functions. Matter 4 (6), 1863–1891. doi:10.1016/j.matt.2021.03.013

PubMed Abstract | CrossRef Full Text | Google Scholar

McBeath, R., Pirone, D. M., Nelson, C. M., Bhadriraju, K., and Chen, C. S. (2004). Cell shape, cytoskeletal tension, and RhoA regulate stem cell lineage commitment. Dev. Cell 6 (4), 483–495. doi:10.1016/s1534-5807(04)00075-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, J., Lewis, A. H., and Grandl, J. (2017). Touch, tension, and transduction–the function and regulation of Piezo ion channels. Trends Biochem. Sci. 42 (1), 57–71. doi:10.1016/j.tibs.2016.09.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, H., Berthier, E., Li, C., Ronceray, P., Han, Y. L., Broedersz, C. P., et al. (2023). Local response and emerging nonlinear elastic length scale in biopolymer matrices. Proc. Natl. Acad. Sci. 120 (23), e2304666120. doi:10.1073/pnas.2304666120

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhao, B., Li, N., Xie, T., Bagheri, Y., Liang, C., Keshri, P., et al. (2020). Quantifying tensile forces at cell–cell junctions with a DNA-based fluorescent probe. Chem. Sci. 11 (32), 8558–8566. doi:10.1039/d0sc01455a

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhao, B., O’Brien, C., Mudiyanselage, APKKK, Li, N., Bagheri, Y., Wu, R., et al. (2017). Visualizing intercellular tensile forces by DNA-based membrane molecular probes. J. Am. Chem. Soc. 139 (50), 18182–18185. doi:10.1021/jacs.7b11176

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: mechanobiology, organoid, lung stiffening, senescence, 3D force quantification

Citation: He S, Mierke CT, Sun Y, Eyckmans J and Guo M (2024) Editorial: Mechanobiology of organoid systems. Front. Cell Dev. Biol. 12:1369713. doi: 10.3389/fcell.2024.1369713

Received: 12 January 2024; Accepted: 16 January 2024;
Published: 30 January 2024.

Edited and reviewed by:

Akihiko Ito, Kindai University, Japan

Copyright © 2024 He, Mierke, Sun, Eyckmans and Guo. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Shijie He, she9@mgh.harvard.edu; Claudia Tanja Mierke, claudia.mierke@uni-leipzig.de; Yubing Sun, ybsun@umass.edu; Jeroen Eyckmans, eyckmans@bu.edu; Ming Guo, guom@mit.edu

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.