1
|
Liao W, Huang Y, Wang X, Hu Z, Zhao C, Wang G. Multidimensional excavation of the current status and trends of mechanobiology in cardiovascular homeostasis and remodeling within 20 years. MECHANOBIOLOGY IN MEDICINE 2025; 3:100127. [PMID: 40395770 PMCID: PMC12067904 DOI: 10.1016/j.mbm.2025.100127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 02/12/2025] [Accepted: 03/09/2025] [Indexed: 05/22/2025]
Abstract
Mechanobiology is essential for cardiovascular structure and function and regulates the normal physiological and pathological processes of the cardiovascular system. Cells in the cardiovascular system are extremely sensitive to their mechanical environment, and once mechanical stimulation is abnormal, the homeostasis mechanism is damaged or lost, leading to the occurrence of pathological remodeling diseases. In the past 20 years, many articles concerning the mechanobiology of cardiovascular homeostasis and remodeling have been published. To better understand the current development status, research hotspots and future development trends in the field, this paper uses CiteSpace software for bibliometric analysis, quantifies and visualizes the articles published in this field in the past 20 years, and reviews the research hotspots and emerging trends. The regulatory effects of mechanical stimulation on the biological behavior of endothelial cells, smooth muscle cells and the extracellular matrix, as well as the mechanical-related remodeling mechanism in heart failure, have always been research hotspots in this field. This paper reviews the research advances of these research hotspots in detail. This paper also introduces the research status of emerging hotspots, such as those related to cardiac fibrosis, homeostasis, mechanosensitive transcription factors and mechanosensitive ion channels. We hope to provide a systematic framework and new ideas for follow-up research on mechanobiology in the field of cardiovascular homeostasis and remodeling and promote the discovery of more therapeutic targets and novel markers of mechanobiology in the cardiovascular system.
Collapse
Affiliation(s)
- Wei Liao
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044, China
| | - Yuxi Huang
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044, China
| | | | - Ziqiu Hu
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044, China
| | - Chuanrong Zhao
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044, China
- JinFeng Laboratory, Chongqing, 401329, China
| | - Guixue Wang
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044, China
- JinFeng Laboratory, Chongqing, 401329, China
| |
Collapse
|
2
|
Hwangbo H, Koo Y, Nacionales F, Kim J, Chae S, Kim GH. Stimulus-assisted in situ bioprinting: advancing direct bench-to-bedside delivery. Trends Biotechnol 2025; 43:1015-1030. [PMID: 39643527 DOI: 10.1016/j.tibtech.2024.11.001] [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: 02/28/2024] [Revised: 10/04/2024] [Accepted: 11/04/2024] [Indexed: 12/09/2024]
Abstract
The fabrication of 3D bioconstructs using bioprinters will advance the field of regenerative medicine owing to its ability to facilitate clinical treatments. Additional stimulations have been applied to the bioconstructs to guide cells laden in the bioconstructs. However, the conventional bench-to-bedside delivery based on separate bioprinting and biostimulating processes may increase the risks of contamination and shape discordance owing to the considerably long process involved. In situ bioprinting is aimed at eliminating these risks, but stimulation strategies implied during in situ printing have not yet been extensively reviewed. Here, we present the concept of stimulus-assisted in situ bioprinting, which integrates the printing and biostimulation processes by directly applying stimuli to the bioink during fabrication.
Collapse
Affiliation(s)
- Hanjun Hwangbo
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon 16419, Republic of Korea; Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - YoungWon Koo
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon 16419, Republic of Korea
| | - Francis Nacionales
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon 16419, Republic of Korea
| | - JuYeon Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon 16419, Republic of Korea
| | - SooJung Chae
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon 16419, Republic of Korea
| | - Geun Hyung Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon 16419, Republic of Korea; Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Republic of Korea; Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
3
|
Muthumalage T, Sarles E, Wang Q, Hensel E, Hill T, Rahman I, Robinson R, Stroup AM, Thongphanh K, Miller LA. In Vitro assessments of ENDS toxicity in the respiratory tract: Are we there yet? NAM JOURNAL 2025; 1:100016. [PMID: 40264558 PMCID: PMC12013380 DOI: 10.1016/j.namjnl.2025.100016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Approximately 4.6 % of U.S. adults over the age of 18 use e-cigarettes, which are a type of electronic nicotine delivery system (ENDS). Over 2.5 million U.S. middle and high school students also use both disposable and/or flavored ENDS products. The health impacts of ENDS use by adults and adolescents are considered a controversial topic in the social media partially due to misperceptions surrounding ENDS toxicity compared to that of combustible cigarettes. There is growing evidence that ENDS, particularly their product composition and design, individual and combined ingredients, and produced aerosols, are toxic to human health. Animal studies have been critical for defining the pathophysiologic outcomes resulting from ENDS use. However, in vitro approaches using human cells can measure the potential toxicity of ENDS e-liquids and aerosols on a shorter timeline and are in keeping with recent statements to replace, reduce and refine the use of animals in biomedical research and regulatory decision making. This review examines current research related to cell culture models of the respiratory tract and exposure methodologies for ENDS use and compares known in vivo parameters of injury and inflammation associated with ENDS to different in vitro systems developed to replicate the inhaled toxicant outcomes. The design and interpretation of exposure methodologies and technological gaps in the evaluation of ENDS aerosols are also discussed. Given the ongoing evolution and popularity of ENDS products, in vitro assessments for measuring respiratory tract injury and inflammation resulting from ENDS use provide a critical scientific platform for rapid evaluation of potential inhalation toxicity in tobacco regulatory science.
Collapse
Affiliation(s)
| | - Emma Sarles
- Department of Mechanical Engineering, Rochester Institute of Technology, Rochester, NY 14623, USA
| | - Qixin Wang
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Edward Hensel
- Department of Mechanical Engineering, Rochester Institute of Technology, Rochester, NY 14623, USA
| | - Thomas Hill
- Office of Science, Center for Tobacco Products, US Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Risa Robinson
- Department of Mechanical Engineering, Rochester Institute of Technology, Rochester, NY 14623, USA
| | - Andrea M. Stroup
- Behavioral Health and Health Policy Practice, Westat, Rockville, MD, 20850, USA
| | - Krista Thongphanh
- California National Primate Research Center, Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California, Davis 95616, USA
| | - Lisa A. Miller
- California National Primate Research Center, Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California, Davis 95616, USA
| |
Collapse
|
4
|
Shyti R, Nays P, Vargiolu R, Zahouani H. An innovative wearable device for sensing mechanoreceptor activation during touch. Sci Rep 2025; 15:9986. [PMID: 40121223 PMCID: PMC11929929 DOI: 10.1038/s41598-025-92379-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 02/27/2025] [Indexed: 03/25/2025] Open
Abstract
Combined studies on the biophysical properties and friction-induced vibrations measured from the fingerpad during touch have yielded promising results. Thus, the ensuing investigation focuses on the underlying mechanism of how surface's roughness affects tactile perception by analysing the finger friction-induced vibrations signals so as to identify the activation of skin mechanoreceptors. The test consists in measuring the skin friction-induced vibrations through the finger sliding against different surfaces' roughness by dint of a wearable augmented finger. The test illustrates that the magnitude of the vibration acceleration signal increases the rougher the surface is, which follows the trends observed in the scientific literature. A notable correlation was identified between amplitude and spacing parameters, validating the robustness of the measurements. To ensure data comparability, parameters such as normal force and sliding direction were meticulously controlled for each participant. Furthermore, the advanced signal processing strategy uncovered additional parameters related to the activation of skin mechanoreceptors, providing deeper insights into what can be perceived by the finger during tactile exploration. Correlations between these signal parameters and the topographical characteristics of the surfaces were explored, demonstrating that the magnitude of several parameters increased with surface roughness. These findings highlight the capabilities of the innovative device to discriminate different touch.
Collapse
Affiliation(s)
- Rexhina Shyti
- Laboratoire de tribologie et dynamique des systèmes, Ecole centrale de Lyon, Ecully, France.
- Renault Group, Technocentre Renault, Guyancourt, France.
| | - Pascale Nays
- Renault Group, Technocentre Renault, Guyancourt, France
| | - Roberto Vargiolu
- Laboratoire de tribologie et dynamique des systèmes, Ecole centrale de Lyon, Ecully, France
| | - Hassan Zahouani
- Laboratoire de tribologie et dynamique des systèmes, Ecole centrale de Lyon, Ecully, France
| |
Collapse
|
5
|
Gmitrov J. Vascular mechanoreceptor magnetic activation, hemodynamic evidence and potential clinical outcomes. Electromagn Biol Med 2025; 44:228-249. [PMID: 40029020 DOI: 10.1080/15368378.2025.2468248] [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: 09/15/2024] [Accepted: 02/10/2025] [Indexed: 03/05/2025]
Abstract
There is sufficient proof that static magnetic fields (SMFs) of different parameters have a significant effect on the cardiovascular system. The sometimes contradictory, opposite-directional nature of SMF's hemodynamic effect generates uncertainty; therefore, an explanation of the underlying mechanisms is required. Following SMF selective carotid baroreceptors or microvascular net exposure, both high and low blood pressure (BP)/vascular tone starting conditions showed a return to normal. Beyond the previous descriptions of SMF's simple hemodynamic results, the current study aims to clarify the physiology of the SMF BP/vascular tone normalizing effects. The examination of available literature and hemodynamic tracings provided strong evidence that mechanoreceptor magnetic activation is concealed behind SMF vascular tone adjustment (increasing or decreasing as needed), filling in the knowledge gap regarding SMF opposite directional vascular tone normalizing outcomes. It has been proposed that cytoskeletal actin filament rearrangement, mechanically-gated Ca2+ influx, and nitric oxide (NO) activity may strengthen SMF's vascular mechanoreceptor sensing/regulation ability, modifying BP and vascular tone features in a hemodynamic normalizing pattern. It is suggested that baro/mechanoreceptor magnetic activation physiology is a unique mechanism of the magneto-cardiovascular interaction with substantial potential for cardiovascular protection.
Collapse
Affiliation(s)
- Juraj Gmitrov
- Hospital Agel Krompachy Inc, Diabetology Clinic, Krompachy, Slovakia
| |
Collapse
|
6
|
Albrecht FB, Schick A, Klatt A, Schmidt FF, Nellinger S, Kluger PJ. Exploring Morphological and Molecular Properties of Different Adipose Cell Models: Monolayer, Spheroids, Gellan Gum-Based Hydrogels, and Explants. Macromol Biosci 2025; 25:e2400320. [PMID: 39450850 PMCID: PMC11904394 DOI: 10.1002/mabi.202400320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/04/2024] [Indexed: 10/26/2024]
Abstract
White adipose tissue (WAT) plays a crucial role in energy homeostasis and secretes numerous adipokines with far-reaching effects. WAT is linked to diseases such as diabetes, cardiovascular disease, and cancer. There is a high demand for suitable in vitro models to study diseases and tissue metabolism. Most of these models are covered by 2D-monolayer cultures. This study aims to evaluate the performance of different WAT models to better derive potential applications. The stability of adipocyte characteristics in spheroids and two 3D gellan gum hydrogels with ex situ lobules and 2D-monolayer culture is analyzed. First, the differentiation to achieve adipocyte-like characteristics is determined. Second, to evaluate the maintenance of differentiated ASC-based models, an adipocyte-based model, and explants over 3 weeks, viability, intracellular lipid content, perilipin A expression, adipokine, and gene expression are analyzed. Several advantages are supported using each of the models. Including, but not limited to, the strong differentiation in 2D-monolayers, the self-assembling within spheroids, the long-term stability of the stem cell-containing hydrogels, and the mature phenotype within adipocyte-containing hydrogels and the lobules. This study highlights the advantages of 3D models due to their more in vivo-like behavior and provides an overview of the different adipose cell models.
Collapse
Affiliation(s)
- Franziska B. Albrecht
- Reutlingen Research InstituteReutlingen UniversityAlteburgstraße 15072762ReutlingenGermany
- Faculty of Natural ScienceUniversity of HohenheimSchloss Hohenheim 170599StuttgartGermany
| | - Ann‐Kathrin Schick
- Faculty of ScienceEnergy and Building ServicesEsslingen UniversityKanalstraße 3373728EsslingenGermany
| | - Annemarie Klatt
- Reutlingen Research InstituteReutlingen UniversityAlteburgstraße 15072762ReutlingenGermany
| | - Freia F. Schmidt
- Reutlingen Research InstituteReutlingen UniversityAlteburgstraße 15072762ReutlingenGermany
| | - Svenja Nellinger
- Reutlingen Research InstituteReutlingen UniversityAlteburgstraße 15072762ReutlingenGermany
| | - Petra J. Kluger
- School of Life SciencesReutlingen UniversityAlteburgstraße 15072762ReutlingenGermany
| |
Collapse
|
7
|
Pattaroni C, Marsland BJ, Harris NL. Early-Life Host-Microbial Interactions and Asthma Development: A Lifelong Impact? Immunol Rev 2025; 330:e70019. [PMID: 40099971 PMCID: PMC11917194 DOI: 10.1111/imr.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 02/19/2025] [Accepted: 02/28/2025] [Indexed: 03/20/2025]
Abstract
Childhood is a multifactorial disease, and recent research highlights the influence of early-life microbial communities in shaping disease risk. This review explores the roles of the gut and respiratory microbiota in asthma development, emphasizing the importance of early microbial exposure. The gut microbiota has been particularly well studied, with certain taxa like Faecalibacterium and Bifidobacterium linked to asthma protection, whereas short-chain fatty acids produced by gut microbes support immune tolerance through the gut-lung axis. In contrast, the respiratory microbiota, though low in biomass, shows consistent associations between early bacterial colonization by Streptococcus, Moraxella, and Haemophilus and increased asthma risk. The review also addresses the emerging roles of the skin microbiota and environmental fungi in asthma, though findings remain inconsistent. Timing is a critical factor, with early-life disruptions, such as antibiotic use, potentially leading to increased asthma risk. Despite significant advances, there are still unresolved questions about the long-term consequences of early microbial perturbations, particularly regarding whether microbial dysbiosis is a cause or consequence of asthma. This review integrates current findings, highlighting the need for deeper investigation into cross-organ interactions and early microbial exposures to understand childhood asthma pathophysiology.
Collapse
Affiliation(s)
- Céline Pattaroni
- Department of Immunology, School of Translational MedicineMonash UniversityMelbourneVictoriaAustralia
| | - Benjamin J. Marsland
- Department of Immunology, School of Translational MedicineMonash UniversityMelbourneVictoriaAustralia
| | - Nicola L. Harris
- Department of Immunology, School of Translational MedicineMonash UniversityMelbourneVictoriaAustralia
| |
Collapse
|
8
|
Mishchenko O, Volchykhina K, Maksymov D, Manukhina O, Pogorielov M, Pavlenko M, Iatsunskyi I. Advanced Strategies for Enhancing the Biocompatibility and Antibacterial Properties of Implantable Structures. MATERIALS (BASEL, SWITZERLAND) 2025; 18:822. [PMID: 40004345 PMCID: PMC11857362 DOI: 10.3390/ma18040822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025]
Abstract
This review explores the latest advancements in enhancing the biocompatibility and antibacterial properties of implantable structures, with a focus on titanium (Ti) and its alloys. Titanium implants, widely used in dental and orthopedic applications, demonstrate excellent mechanical strength and biocompatibility, yet face challenges such as peri-implantitis, a bacterial infection that can lead to implant failure. To address these issues, both passive and active surface modification strategies have been developed. Passive modifications, such as altering surface texture and chemistry, aim to prevent bacterial adhesion, while active approaches incorporate antimicrobial agents for sustained infection control. Nanotechnology has emerged as a transformative tool, enabling the creation of nanoscale materials and coatings like TiO2 and ZnO that promote osseointegration and inhibit biofilm formation. Techniques such as plasma spraying, ion implantation, and plasma electrolytic oxidation (PEO) show promising results in improving implant integration and durability. Despite significant progress, further research is needed to refine these technologies, optimize surface properties, and address the clinical challenges associated with implant longevity and safety. This review highlights the intersection of surface engineering, nanotechnology, and biomedical innovation, paving the way for the next generation of implantable devices.
Collapse
Affiliation(s)
- Oleg Mishchenko
- Department of Dentistry of Postgraduate Education, Zaporizhzhia State Medical and Pharmaceutical University, 26 Marii Prymachenko Blvd., 69035 Zaporizhzhia, Ukraine; (K.V.); (D.M.); (O.M.)
| | - Kristina Volchykhina
- Department of Dentistry of Postgraduate Education, Zaporizhzhia State Medical and Pharmaceutical University, 26 Marii Prymachenko Blvd., 69035 Zaporizhzhia, Ukraine; (K.V.); (D.M.); (O.M.)
| | - Denis Maksymov
- Department of Dentistry of Postgraduate Education, Zaporizhzhia State Medical and Pharmaceutical University, 26 Marii Prymachenko Blvd., 69035 Zaporizhzhia, Ukraine; (K.V.); (D.M.); (O.M.)
| | - Olesia Manukhina
- Department of Dentistry of Postgraduate Education, Zaporizhzhia State Medical and Pharmaceutical University, 26 Marii Prymachenko Blvd., 69035 Zaporizhzhia, Ukraine; (K.V.); (D.M.); (O.M.)
| | - Maksym Pogorielov
- Insitute of Atomic Physics and Spectroscopy, University of Latvia, 3 Jelgavas Str., LV-1004 Riga, Latvia;
| | - Mykola Pavlenko
- NanoBioMedical Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej Str. 3, 61-614 Poznan, Poland;
| | - Igor Iatsunskyi
- NanoBioMedical Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej Str. 3, 61-614 Poznan, Poland;
| |
Collapse
|
9
|
Fukui H, Chow RWY, Yap CH, Vermot J. Rhythmic forces shaping the zebrafish cardiac system. Trends Cell Biol 2025; 35:166-176. [PMID: 39665884 DOI: 10.1016/j.tcb.2024.10.012] [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: 03/06/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 12/13/2024]
Abstract
The structural development of the heart depends heavily on mechanical forces, and rhythmic contractions generate essential physical stimuli during morphogenesis. Cardiac cells play a critical role in coordinating this process by sensing and responding to these mechanical forces. In vivo, cells experience rhythmic spatial and temporal variations in deformation-related stresses throughout heart development. What impact do these mechanical forces have on heart morphogenesis? Recent work in zebrafish (Danio rerio) offers important insights into this question. This review focuses on endocardial (EdCs) and myocardial cells (cardiomyocytes, CMs), key cell types in the heart, and provides a comprehensive overview of forces and tissue mechanics in zebrafish and their direct influence on cardiac cell identity.
Collapse
Affiliation(s)
- Hajime Fukui
- Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan; Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Renee Wei-Yan Chow
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Choon Hwai Yap
- Department of Bioengineering, Imperial College London, London, UK
| | - Julien Vermot
- Department of Bioengineering, Imperial College London, London, UK.
| |
Collapse
|
10
|
McNicol GR, Dalby MJ, Stewart PS. A theoretical model for focal adhesion and cytoskeleton formation in non-motile cells. J Theor Biol 2025; 596:111965. [PMID: 39442686 DOI: 10.1016/j.jtbi.2024.111965] [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: 02/05/2024] [Revised: 07/25/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024]
Abstract
To function and survive cells need to be able to sense and respond to their local environment through mechanotransduction. Crucially, mechanical and biochemical perturbations initiate cell signalling cascades, which can induce responses such as growth, apoptosis, proliferation and differentiation. At the heart of this process are actomyosin stress fibres (SFs), which form part of the cell cytoskeleton, and focal adhesions (FAs), which bind this cytoskeleton to the extra-cellular matrix (ECM). The formation and maturation of these structures (connected by a positive feedback loop) is pivotal in non-motile cells, where SFs are generally of ventral type, interconnecting FAs and producing isometric tension. In this study we formulate a one-dimensional bio-chemo-mechanical continuum model to describe the coupled formation and maturation of ventral SFs and FAs. We use a set of reaction-diffusion-advection equations to describe three sets of biochemical events: the polymerisation of actin and subsequent bundling into activated SFs; the formation and maturation of cell-substrate adhesions; and the activation of signalling proteins in response to FA and SF formation. The evolution of these key proteins is coupled to a Kelvin-Voigt viscoelastic description of the cell cytoplasm and the ECM. We employ this model to understand how cells respond to external and intracellular cues in vitro and are able to reproduce experimentally observed phenomena including non-uniform cell striation and cells forming weaker SFs and FAs on softer substrates.
Collapse
Affiliation(s)
- Gordon R McNicol
- School of Mathematics and Statistics, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Matthew J Dalby
- Centre for the Cellular Microenvironment, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow, G11 6EW, United Kingdom
| | - Peter S Stewart
- School of Mathematics and Statistics, University of Glasgow, Glasgow, G12 8QQ, United Kingdom.
| |
Collapse
|
11
|
Sinha NR, Hofmann AC, Suleiman LA, Jeffrey MT, Jeffrey WC, Kumar R, Tripathi R, Mohan RR. Mustard Gas Induced Corneal Injury Involves Ferroptosis and p38 MAPK Signaling. Invest Ophthalmol Vis Sci 2025; 66:23. [PMID: 39792076 PMCID: PMC11730948 DOI: 10.1167/iovs.66.1.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025] Open
Abstract
Purpose Sulfur mustard gas (SM) exposure to eyes causes multiple corneal injuries including stromal cell loss in vivo. However, mechanisms mediating stromal cell loss/death remains elusive. This study sought to test the novel hypothesis that SM-induced toxicity to human corneal stromal fibroblasts involves ferroptosis mechanism via p38 MAPK signaling. Methods New Zealand white rabbit corneas, naïve and SM exposed (200 mg-min/m3 for eight minutes and collected after three days) were used to examine the levels of cell death and reactive oxygen species (ROS) for in vivo studies. Donor human corneas were used to generate primary human corneal stromal fibroblasts (hCSF) for in vitro studies. The hCSFs were exposed to nitrogen mustard (NM; SM analogue) at various timepoints (30 minutes, eight hours, and 24 hours). A p38 MAPK specific inhibitor, SB202190, was also used. Quantitative reverse transcription polymerase chain reaction, Western blotting, reactive oxygen species (ROS), lipid peroxidation, live/dead assay, and RNASeq were used in various investigations. Results SM caused a significant increase in cell death and ROS production three days after SM exposure in rabbit corneas. NM exposure to hCSF demonstrated a significant increase in ROS, lipid peroxidation, and ferroptosis biomarkers ACSL4 (inducer) and significant decrease in reducer (SLC7A11 and GPX4) compared to controls in a time-dependent manner. The inhibition of p38 MAPK promoted cell survival and reduced ROS production following mustard gas exposure. Conclusions The results of in vivo and in vitro investigations uncovered a novel mechanism that mustard gas toxicity to the cornea involves ferroptosis pathway and p38 MAPK activation.
Collapse
Affiliation(s)
- Nishant R. Sinha
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri, United States
- Departments of Veterinary Medicine & Surgery and Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Alexandria C. Hofmann
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri, United States
- Departments of Veterinary Medicine & Surgery and Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Laila A. Suleiman
- Departments of Veterinary Medicine & Surgery and Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - Maxwell T. Jeffrey
- Departments of Veterinary Medicine & Surgery and Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - William C. Jeffrey
- Departments of Veterinary Medicine & Surgery and Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - Rajnish Kumar
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri, United States
- Departments of Veterinary Medicine & Surgery and Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - Ratnakar Tripathi
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri, United States
- Departments of Veterinary Medicine & Surgery and Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - Rajiv R. Mohan
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri, United States
- Departments of Veterinary Medicine & Surgery and Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
12
|
Ozan VB, Wang H, Akshay A, Anand D, Hibaoui Y, Feki A, Gote-Schniering J, Gheinani AH, Heller M, Uldry AC, Lagache SB, Gazdhar A, Geiser T. Influence of Microenvironmental Orchestration on Multicellular Lung Alveolar Organoid Development from Human Induced Pluripotent Stem Cells. Stem Cell Rev Rep 2025; 21:254-275. [PMID: 39417930 PMCID: PMC11762634 DOI: 10.1007/s12015-024-10789-1] [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] [Accepted: 09/15/2024] [Indexed: 10/19/2024]
Abstract
Induced pluripotent stem cells (iPSCs) have emerged as promising in vitro tools, providing a robust system for disease modelling and facilitating drug screening. Human iPSCs have been successfully differentiated into lung cells and three-dimensional lung spheroids or organoids. The lung is a multicellular complex organ that develops under the symphonic influence of the microenvironment. Here, we hypothesize that the generation of lung organoids in a controlled microenvironment (cmO) (oxygen and pressure) yields multicellular organoids with architectural complexity resembling the lung alveoli. iPSCs were differentiated into mature lung organoids following a stepwise protocol in an oxygen and pressure-controlled microenvironment. The organoids developed in the controlled microenvironment displayed complex alveolar architecture and stained for SFTPC, PDPN, and KRT5, indicating the presence of alveolar epithelial type II and type I cells, as well as basal cells. Moreover, gene and protein expression levels were also increased in the cmO. Furthermore, pathway analysis of proteomics revealed upregulation of lung development-specific pathways in the cmO compared to those growing in normal culture conditions. In summary, by using a controlled microenvironment, we established a complex multicellular lung organoid derived from iPSCs as a novel cellular model to study lung alveolar biology in both lung health and disease.
Collapse
Affiliation(s)
- Vedat Burak Ozan
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland
| | - Huijuan Wang
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Rheumatology and Immunology Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Akshay Akshay
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland
- Functional Urology Research Group, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Deepika Anand
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Youssef Hibaoui
- Department of Gynecology and Obstetrics, Cantonal Hospital Fribourg, Fribourg, Switzerland
| | - Anis Feki
- Department of Gynecology and Obstetrics, Cantonal Hospital Fribourg, Fribourg, Switzerland
| | - Janine Gote-Schniering
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Rheumatology and Immunology Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ali Hashemi Gheinani
- Functional Urology Research Group, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Urological Diseases Research Center, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Manfred Heller
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Anne-Christine Uldry
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Sophie Braga Lagache
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Amiq Gazdhar
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Thomas Geiser
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| |
Collapse
|
13
|
Zhou K, Chung M, Cheng J, Powell JT, Yan Q, Liu J, Xiong Y, Schwartz MA, Lin C. DNA nanodevice for analysis of force-activated protein extension and interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620262. [PMID: 39554144 PMCID: PMC11565787 DOI: 10.1101/2024.10.25.620262] [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
Force-induced changes in protein structure and function mediate cellular responses to mechanical stresses. Existing methods to study protein conformation under mechanical force are incompatible with biochemical and structural analysis. Taking advantage of DNA nanotechnology, including the well-defined geometry of DNA origami and programmable mechanics of DNA hairpins, we built a nanodevice to apply controlled forces to proteins. This device was used to study the R1-R2 segment of the talin1 rod domain as a model protein, which comprises two alpha-helical bundles that reversibly unfold under tension to expose binding sites for the cytoskeletal protein vinculin. Electron microscopy confirmed tension-dependent protein extension while biochemical analysis demonstrated enhanced vinculin binding under tension. The device could also be used in pull down assays with cell lysates, which identified filamins as novel tension-dependent talin binders. The DNA nanodevice is thus a valuable addition to the molecular toolbox for studying mechanosensitive proteins.
Collapse
Affiliation(s)
- Kun Zhou
- Department of Cell Biology, Yale School of Medicine, USA
- Nanobiology Institute, Yale University, USA
| | - Minhwan Chung
- Department of Cell Biology, Yale School of Medicine, USA
- Yale Cardiovascular Research Center, Yale School of Medicine, USA
| | - Jing Cheng
- Department of Biophysics and Biochemistry, Yale University, USA
| | - John T Powell
- Department of Cell Biology, Yale School of Medicine, USA
- Nanobiology Institute, Yale University, USA
| | - Qi Yan
- Department of Cell Biology, Yale School of Medicine, USA
- Nanobiology Institute, Yale University, USA
| | - Jun Liu
- Department of Cell Biology, Yale School of Medicine, USA
- Department of Microbial Pathogenesis, Yale School of Medicine, USA
- Microbial Sciences Institute, Yale University, USA
| | - Yong Xiong
- Department of Biophysics and Biochemistry, Yale University, USA
| | - Martin A Schwartz
- Department of Cell Biology, Yale School of Medicine, USA
- Yale Cardiovascular Research Center, Yale School of Medicine, USA
- Department of Biomedical Engineering, Yale University, USA
| | - Chenxiang Lin
- Department of Cell Biology, Yale School of Medicine, USA
- Nanobiology Institute, Yale University, USA
- Department of Biomedical Engineering, Yale University, USA
| |
Collapse
|
14
|
Kao CY, Ho CT, Huang TH, Kao CT. The Effects of Photobiomodulation Therapy on Tensile-Cultured Cementoblasts Cells. Photobiomodul Photomed Laser Surg 2024; 42:754-759. [PMID: 39330936 DOI: 10.1089/photob.2024.0076] [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] [Indexed: 09/28/2024] Open
Abstract
Background: Studies show that photobiomodulation therapy (PBMT) boosts cellular ATP production and cell growth and reduces inflammation. Additionally, mechanical tension affects gene expression, impacting cellular functions like proliferation and migration. Objective: We investigated the impact of PBMT on OCCM-30 cementoblast cells under tensile stress, focusing on cell survival, differentiation, and inflammatory responses, particularly relating to orthodontic tooth movement and root resorption. Methods: Cultured OCCM-30 cells under negative pressure received PBMT with a 10.6 μm wavelength in continuous mode at 1.0 W power for 3, 5, or 10 sec, corresponding to energy densities of 3, 5, or 10 J/cm2. We assessed cell viability with the Presto Blue assay and inflammatory markers Interleukin 6 (IL-6), inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2) through western blots at 1, 12, 24 h, and 7 days post-irradiation. Results: PBMT improved cell viability over time while maintaining levels of inflammatory markers. alkaline phosphatase levels dropped initially but increased after 7 days, suggesting enhanced cementoblast differentiation. IL-6 levels rose gradually, with 3J and 5J treatments showing significantly higher levels than the control. iNOS levels spiked within the first 24 h, then declined by day 7. COX-2 levels consistently rose, with the 5J treatment showing greater increases. Conclusions: PBMT appears to support cementoblast survival and differentiation while managing inflammation, potentially aiding root repair during orthodontic treatments and reducing inflammatory root resorption.
Collapse
Affiliation(s)
- Chuan-Yi Kao
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Psychiatry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chun-Te Ho
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Tsui-Hsein Huang
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chia-Tze Kao
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
15
|
Lima JT, Ferreira JG. Mechanobiology of the nucleus during the G2-M transition. Nucleus 2024; 15:2330947. [PMID: 38533923 DOI: 10.1080/19491034.2024.2330947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/09/2024] [Indexed: 03/28/2024] Open
Abstract
Cellular behavior is continuously influenced by mechanical forces. These forces span the cytoskeleton and reach the nucleus, where they trigger mechanotransduction pathways that regulate downstream biochemical events. Therefore, the nucleus has emerged as a regulator of cellular response to mechanical stimuli. Cell cycle progression is regulated by cyclin-CDK complexes. Recent studies demonstrated these biochemical pathways are influenced by mechanical signals, highlighting the interdependence of cellular mechanics and cell cycle regulation. In particular, the transition from G2 to mitosis (G2-M) shows significant changes in nuclear structure and organization, ranging from nuclear pore complex (NPC) and nuclear lamina disassembly to chromosome condensation. The remodeling of these mechanically active nuclear components indicates that mitotic entry is particularly sensitive to forces. Here, we address how mechanical forces crosstalk with the nucleus to determine the timing and efficiency of the G2-M transition. Finally, we discuss how the deregulation of nuclear mechanics has consequences for mitosis.
Collapse
Affiliation(s)
- Joana T Lima
- Epithelial Polarity and Cell Division Laboratory, Instituto de Investigação e Inovação em Saúde (i3S), Porto, Portugal
- Departamento de Biomedicina, Unidade de Biologia Experimental, Faculdade de Medicina do Porto, Porto, Portugal
- Programa Doutoral em Biomedicina, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Jorge G Ferreira
- Epithelial Polarity and Cell Division Laboratory, Instituto de Investigação e Inovação em Saúde (i3S), Porto, Portugal
- Departamento de Biomedicina, Unidade de Biologia Experimental, Faculdade de Medicina do Porto, Porto, Portugal
| |
Collapse
|
16
|
Sreedasyam R, Wilson BG, Ferrandez PR, Botvinick EL, Venugopalan V. An optical system for cellular mechanostimulation in 3D hydrogels. Acta Biomater 2024; 189:439-448. [PMID: 39368720 DOI: 10.1016/j.actbio.2024.09.050] [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: 01/04/2023] [Revised: 09/20/2024] [Accepted: 09/26/2024] [Indexed: 10/07/2024]
Abstract
We introduce a method utilizing single laser-generated cavitation bubbles to stimulate cellular mechanotransduction in dermal fibroblasts embedded within 3D hydrogels. We demonstrate that fibroblasts embedded in either amorphous or fibrillar hydrogels engage in Ca2+ signaling following exposure to an impulsive mechanical stimulus provided by a single 250 µm diameter laser-generated cavitation bubble. We find that the spatial extent of the cellular signaling is larger for cells embedded within a fibrous collagen hydrogel as compared to those embedded within an amorphous polyvinyl alcohol polymer (SLO-PVA) hydrogel. Additionally, for fibroblasts embedded in collagen, we find an increased range of cellular mechanosensitivity for cells that are polarized relative to the radial axis as compared to the circumferential axis. By contrast, fibroblasts embedded within SLO-PVA did not display orientation-dependent mechanosensitivity. Fibroblasts embedded in hydrogels and cultured in calcium-free media did not show cavitation-induced mechanotransduction; implicating calcium signaling based on transmembrane Ca2+ transport. This study demonstrates the utility of single laser-generated cavitation bubbles to provide local non-invasive impulsive mechanical stimuli within 3D hydrogel tissue models with concurrent imaging using optical microscopy. STATEMENT OF SIGNIFICANCE: Currently, there are limited methods for the non-invasive real-time assessment of cellular sensitivity to mechanical stimuli within 3D tissue scaffolds. We describe an original approach that utilizes a pulsed laser microbeam within a standard laser scanning microscope system to generate single cavitation bubbles to provide impulsive mechanostimulation to cells within 3D fibrillar and amorphous hydrogels. Using this technique, we measure the cellular mechanosensitivity of primary human dermal fibroblasts embedded in amorphous and fibrillar hydrogels, thereby providing a useful method to examine cellular mechanotransduction in 3D biomaterials. Moreover, the implementation of our method within a standard optical microscope makes it suitable for broad adoption by cellular mechanotransduction researchers and opens the possibility of high-throughput evaluation of biomaterials with respect to cellular mechanosignaling.
Collapse
Affiliation(s)
- Rahul Sreedasyam
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697-2715, United States
| | - Bryce G Wilson
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA 92697-2580, United States
| | - Patricia R Ferrandez
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697-2715, United States
| | - Elliot L Botvinick
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697-2715, United States; Beckman Laser Institute, University of California Irvine, Irvine, CA 92697-3010, United States.
| | - Vasan Venugopalan
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697-2715, United States; Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA 92697-2580, United States; Beckman Laser Institute, University of California Irvine, Irvine, CA 92697-3010, United States.
| |
Collapse
|
17
|
Manning ML. Rigidity in mechanical biological networks. Curr Biol 2024; 34:R1024-R1030. [PMID: 39437721 DOI: 10.1016/j.cub.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Multicellular organisms generate complex morphologies required for their function. Organisms control these morphologies by tuning active forces and by altering the emergent 'material properties' of a tissue, i.e. the rheology of the tissue. In many cases, organisms take advantage of dramatic changes in the rheology that occur when the material undergoes a rigidity transition from a fluid-like or floppy state to a solid-like or rigid state. This transition in turn depends on internal parameters at the scale of cells and molecules. This review highlights recent theoretical work identifying the mechanisms that drive such transitions, so that biologists can look for these mechanisms in in vivo or in vitro systems. We discuss two main types of transition: a first-order rigidity transition that depends on the connectivity of small-scale structures, such as the number of contacts between cells or the number of branch points in a biopolymer network; and a second-order rigidity transition that depends on the geometry of small-scale structures, such as the shape of cells or the distance between crosslinks in a polymer network. We provide examples of each type of transition in model organisms and discuss methods for distinguishing between the mechanisms in future experiments.
Collapse
Affiliation(s)
- M Lisa Manning
- Department of Physics and BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA.
| |
Collapse
|
18
|
Ohashi K, Kunitomi A, Chiba S, Mizuno K. Roles of the Dbl family of RhoGEFs in mechanotransduction - a review. Front Cell Dev Biol 2024; 12:1485725. [PMID: 39479515 PMCID: PMC11521908 DOI: 10.3389/fcell.2024.1485725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024] Open
Abstract
Rho guanine nucleotide exchange factors (RhoGEFs) comprise a wide range of proteins with a common domain responsible for the activation of the Rho family of small GTPases and various domains in other regions. The evolutionary divergence of RhoGEFs enables actin cytoskeletal reorganization, leading to complex cellular responses in higher organisms. In this review, we address the involvement of RhoGEFs in the mechanical stress response of mammalian cells. The cellular mechanical stress response is essential for the proper and orderly regulation of cell populations, including the maintenance of homeostasis, tissue morphogenesis, and adaptation to the mechanical environment. In particular, this review focuses on the recent findings regarding the Dbl family of RhoGEFs involved in mechanical stress responses at the cell-cell and cell-substrate adhesion sites, and their molecular mechanisms underlying actin cytoskeleton remodeling and signal transduction.
Collapse
Affiliation(s)
- Kazumasa Ohashi
- Department of Molecular and Chemical Life Sciences, Laboratory of Molecular and Cellular Biology, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
| | | | | | | |
Collapse
|
19
|
Chung M, Zhou K, Powell JT, Lin C, Schwartz MA. DNA-Based Molecular Clamp for Probing Protein Interactions and Structure under Force. ACS NANO 2024; 18:27590-27596. [PMID: 39344156 PMCID: PMC11518680 DOI: 10.1021/acsnano.4c08663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Cellular mechanotransduction, a process central to cell biology, embryogenesis, adult physiology, and multiple diseases, is thought to be mediated by force-driven changes in protein conformation that control protein function. However, methods to study proteins under defined mechanical loads on a biochemical scale are lacking. We report the development of a DNA-based device in which the transition between single- and double-stranded DNA applies tension to an attached protein. Using a fragment of the talin rod domain as a test case, negative-stain electron microscopy reveals programmable extension, while pull down assays show tension-induced binding to two ligands, ARPC5L and vinculin, known to bind to cryptic sites inside the talin structure. These results demonstrate the utility of the DNA clamp for biochemical studies and potential structural analysis.
Collapse
Affiliation(s)
- Minhwan Chung
- Yale Cardiovascular Research Center, Department of Internal Medicine (Cardiology), Yale University School of Medicine, New Haven, Connecticut 06511, United States
| | | | | | | | - Martin A Schwartz
- Yale Cardiovascular Research Center, Department of Internal Medicine (Cardiology), Yale University School of Medicine, New Haven, Connecticut 06511, United States
| |
Collapse
|
20
|
Rashad A, Gomez A, Gangrade A, Zehtabi F, Mandal K, Maity S, Ma C, Li B, Khademhosseini A, Roberto de Barros N. Effect of viscosity of gelatin methacryloyl-based bioinks on bone cells. Biofabrication 2024; 16:10.1088/1758-5090/ad6d91. [PMID: 39121892 PMCID: PMC11491941 DOI: 10.1088/1758-5090/ad6d91] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/09/2024] [Indexed: 08/12/2024]
Abstract
The viscosity of gelatin methacryloyl (GelMA)-based bioinks generates shear stresses throughout the printing process that can affect cell integrity, reduce cell viability, cause morphological changes, and alter cell functionality. This study systematically investigated the impact of the viscosity of GelMA-gelatin bioinks on osteoblast-like cells in 2D and 3D culture conditions. Three bioinks with low, medium, and high viscosity prepared by supplementing a 5% GelMA solution with different concentrations of gelatin were evaluated. Cell responses were studied in a 2D environment after printing and incubation in non-cross-linked bioinks that caused the gelatin and GelMA to dissolve and release cells for attachment to tissue culture plates. The increased viscosity of the bioinks significantly affected cell area and aspect ratio. Cells printed using the bioink with medium viscosity exhibited greater metabolic activity and proliferation rate than those printed using the high viscosity bioink and even the unprinted control cells. Additionally, cells printed using the bioink with high viscosity demonstrated notably elevated expression levels of alkaline phosphatase and bone morphogenetic protein-2 genes. In the 3D condition, the printed cell-laden hydrogels were photo-cross-linked prior to incubation. The medium viscosity bioink supported greater cell proliferation compared to the high viscosity bioink. However, there were no significant differences in the expression of osteogenic markers between the medium and high viscosity bioinks. Therefore, the choice between medium and high viscosity bioinks should be based on the desired outcomes and objectives of the bone tissue engineering application. Furthermore, the bioprinting procedure with the medium viscosity bioink was used as an automated technique for efficiently seeding cells onto 3D printed porous titanium scaffolds for bone tissue engineering purposes.
Collapse
Affiliation(s)
- Ahmad Rashad
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 91367
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556, USA
- Department of Clinical Dentistry, University of Bergen, Bergen 5009, Norway
| | - Alejandro Gomez
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 91367
- Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324
| | - Ankit Gangrade
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 91367
| | - Fatemeh Zehtabi
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 91367
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 91367
| | - Surjendu Maity
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 91367
| | - Changyu Ma
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 91367
- Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324
| | - Bingbing Li
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 91367
- Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324
| | | | - Natan Roberto de Barros
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 91367
- National Laboratory of Bioscience (LNBio), National Center of Research in Energy and Materials (CNPEM), Campinas, Brazil
| |
Collapse
|
21
|
Latypova AA, Yaremenko AV, Pechnikova NA, Minin AS, Zubarev IV. Magnetogenetics as a promising tool for controlling cellular signaling pathways. J Nanobiotechnology 2024; 22:327. [PMID: 38858689 PMCID: PMC11163773 DOI: 10.1186/s12951-024-02616-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024] Open
Abstract
Magnetogenetics emerges as a transformative approach for modulating cellular signaling pathways through the strategic application of magnetic fields and nanoparticles. This technique leverages the unique properties of magnetic nanoparticles (MNPs) to induce mechanical or thermal stimuli within cells, facilitating the activation of mechano- and thermosensitive proteins without the need for traditional ligand-receptor interactions. Unlike traditional modalities that often require invasive interventions and lack precision in targeting specific cellular functions, magnetogenetics offers a non-invasive alternative with the capacity for deep tissue penetration and the potential for targeting a broad spectrum of cellular processes. This review underscores magnetogenetics' broad applicability, from steering stem cell differentiation to manipulating neuronal activity and immune responses, highlighting its potential in regenerative medicine, neuroscience, and cancer therapy. Furthermore, the review explores the challenges and future directions of magnetogenetics, including the development of genetically programmed magnetic nanoparticles and the integration of magnetic field-sensitive cells for in vivo applications. Magnetogenetics stands at the forefront of cellular manipulation technologies, offering novel insights into cellular signaling and opening new avenues for therapeutic interventions.
Collapse
Affiliation(s)
- Anastasiia A Latypova
- Institute of Future Biophysics, Dolgoprudny, 141701, Russia
- Moscow Center for Advanced Studies, Moscow, 123592, Russia
| | - Alexey V Yaremenko
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997, Russia.
| | - Nadezhda A Pechnikova
- Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
- Saint Petersburg Pasteur Institute, Saint Petersburg, 197101, Russia
| | - Artem S Minin
- M.N. Mikheev Institute of Metal Physics of the Ural Branch of the Russian Academy of Sciences, Yekaterinburg, 620108, Russia
| | - Ilya V Zubarev
- Institute of Future Biophysics, Dolgoprudny, 141701, Russia.
| |
Collapse
|
22
|
Chung M, Zhou K, Powell J, Lin C, Schwartz MA. A DNA-based molecular clamp for probing protein interactions and structure under force. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597759. [PMID: 38895381 PMCID: PMC11185747 DOI: 10.1101/2024.06.06.597759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Cellular mechanotransduction, a process central to cell biology, embryogenesis, adult physiology and multiple diseases, is thought to be mediated by force-driven changes in protein conformation that control protein function. However, methods to study proteins under defined mechanical loads on a biochemical scale are lacking. We report the development of a DNA based device in which the transition between single-stranded and double-stranded DNA applies tension to an attached protein. Using a fragment of the talin rod domain as a test case, negative-stain electron microscopy reveals programmable extension while pull down assays show tension-induced binding to two ligands, ARPC5L and vinculin, known to bind to cryptic sites inside the talin structure. These results demonstrate the utility of the DNA clamp for biochemical studies and potential structural analysis.
Collapse
Affiliation(s)
- Minhwan Chung
- Yale Cardiovascular Research Center, Department of Internal Medicine (Cardiology) 300 George St., New Haven CT, 06511
| | - Kun Zhou
- Depart of Cell Biology, Yale School of Medicine
- Nanobiology Institute, Yale University
| | - John Powell
- Depart of Cell Biology, Yale School of Medicine
- Nanobiology Institute, Yale University
| | - Chenxiang Lin
- Depart of Cell Biology, Yale School of Medicine
- Nanobiology Institute, Yale University
- Department of Biomedical Engineering, Yale University
| | - Martin A Schwartz
- Yale Cardiovascular Research Center, Department of Internal Medicine (Cardiology) 300 George St., New Haven CT, 06511
- Depart of Cell Biology, Yale School of Medicine
- Department of Biomedical Engineering, Yale University
| |
Collapse
|
23
|
Song Y, Zhao Z, Xu L, Huang P, Gao J, Li J, Wang X, Zhou Y, Wang J, Zhao W, Wang L, Zheng C, Gao B, Jiang L, Liu K, Guo Y, Yao X, Duan L. Using an ER-specific optogenetic mechanostimulator to understand the mechanosensitivity of the endoplasmic reticulum. Dev Cell 2024; 59:1396-1409.e5. [PMID: 38569547 DOI: 10.1016/j.devcel.2024.03.014] [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: 01/06/2023] [Revised: 12/21/2023] [Accepted: 03/08/2024] [Indexed: 04/05/2024]
Abstract
The ability of cells to perceive and respond to mechanical cues is essential for numerous biological activities. Emerging evidence indicates important contributions of organelles to cellular mechanosensitivity and mechanotransduction. However, whether and how the endoplasmic reticulum (ER) senses and reacts to mechanical forces remains elusive. To fill the knowledge gap, after developing a light-inducible ER-specific mechanostimulator (LIMER), we identify that mechanostimulation of ER elicits a transient, rapid efflux of Ca2+ from ER in monkey kidney COS-7 cells, which is dependent on the cation channels transient receptor potential cation channel, subfamily V, member 1 (TRPV1) and polycystin-2 (PKD2) in an additive manner. This ER Ca2+ release can be repeatedly stimulated and tuned by varying the intensity and duration of force application. Moreover, ER-specific mechanostimulation inhibits ER-to-Golgi trafficking. Sustained mechanostimuli increase the levels of binding-immunoglobulin protein (BiP) expression and phosphorylated eIF2α, two markers for ER stress. Our results provide direct evidence for ER mechanosensitivity and tight mechanoregulation of ER functions, placing ER as an important player on the intricate map of cellular mechanotransduction.
Collapse
Affiliation(s)
- Yutong Song
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR 999077, China
| | - Zhihao Zhao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR 999077, China
| | - Linyu Xu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR 999077, China
| | - Peiyuan Huang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR 999077, China
| | - Jiayang Gao
- School of Life Sciences, Centre for Cell & Developmental Biology and State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR 999077, China
| | - Jingxuan Li
- School of Biomedical Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR 999077, China
| | - Xuejie Wang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR 999077, China
| | - Yiren Zhou
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR 999077, China
| | - Jinhui Wang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR 999077, China
| | - Wenting Zhao
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637457, Singapore
| | - Likun Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Chaogu Zheng
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Pok Fu Lam, Hong Kong SAR 999077, China
| | - Bo Gao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR 999077, China
| | - Liwen Jiang
- School of Life Sciences, Centre for Cell & Developmental Biology and State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR 999077, China
| | - Kai Liu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR 999077, China; Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR 999077, China
| | - Yusong Guo
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR 999077, China
| | - Xiaoqiang Yao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR 999077, China
| | - Liting Duan
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR 999077, China.
| |
Collapse
|
24
|
Na Nan D, Klincumhom N, Trachoo V, Everts V, Ferreira JN, Osathanon T, Pavasant P. Periostin-integrin interaction regulates force-induced TGF-β1 and α-SMA expression by hPDLSCs. Oral Dis 2024; 30:2570-2579. [PMID: 37466141 DOI: 10.1111/odi.14691] [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: 01/07/2023] [Revised: 06/15/2023] [Accepted: 07/10/2023] [Indexed: 07/20/2023]
Abstract
OBJECTIVE Periostin (PN), a major matricellular periodontal ligament (PDL) protein, modulates the remodeling of the PDL and bone, especially under mechanical stress. This study investigated the requirement of PN-integrin signaling in force-induced expression of transforming growth factor-beta 1 (TGF-β1) and alpha-smooth muscle actin (α-SMA) in human PDL stem cells (hPDLSCs). METHODS Cells were stimulated with intermittent compressive force (ICF) using computerized controlled apparatus. Cell migration was examined using in vitro scratch assay. The mRNA expression was examined using real-time polymerase chain reaction. The protein expression was determined using immunofluorescent staining and western blot analysis. RESULTS Stimulation with ICF for 24 h increased the expression of PN, TGF-β1, and α-SMA, along with increased SMAD2/3 phosphorylation. Knockdown of POSTN (PN gene) decreased the protein levels of TGF-β1 and pSMAD2/3 upon force stimulation. POSTN knockdown of hPDLSCs resulted in delayed cell migration, as determined by a scratch assay. However, migration improved after seeding these knockdown cells on pre-PN-coated surfaces. Further, the knockdown of αVβ5 significantly attenuated the force-induced TGF-β1 expression. CONCLUSION Our findings indicate the importance of PN-αVβ5 interactions in ICF-induced TGF-β1 signaling and the expression of α-SMA. Findings support the critical role of PN in maintaining the PDL's tissue integrity and homeostasis.
Collapse
Affiliation(s)
- Daneeya Na Nan
- Center of Excellence in Regenerative Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Nuttha Klincumhom
- Center of Excellence in Regenerative Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Vorapat Trachoo
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Vincent Everts
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Department of Oral Cell Biology, Faculty of Dentistry, University of Amsterdam and Vrije Universiteit, Amsterdam, The Netherlands
| | - Joao N Ferreira
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Thanaphum Osathanon
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Prasit Pavasant
- Center of Excellence in Regenerative Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
25
|
Emma Sarles S, Hensel EC, Terry J, Nuss C, Robinson RJ. Flow Rate and Wall Shear Stress Characterization of a Biomimetic Aerosol Exposure System. J Biomech Eng 2024; 146:045001. [PMID: 38270928 PMCID: PMC10983703 DOI: 10.1115/1.4064549] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 01/26/2024]
Abstract
Current in vitro emissions and exposure systems lack biomimicry, use unrealistic flow conditions, produce unrealistic dose, and provide inaccurate biomechanical cues to cell cultures, limiting ability to correlate in vitro outcomes with in vivo health effects. A biomimetic in vitro system capable of puffing aerosol and clean air inhalation may empower researchers to investigate complex questions related to lung injury and disease. A biomimetic aerosol exposure system (BAES), including an electronic cigarette adapter, oral cavity module (OCM), and bifurcated exposure chamber (BEC) was designed and manufactured. The fraction of aerosol deposited in transit to a filter pad or lost as volatiles was 0.116±0.021 in a traditional emissions setup versus 0.098 ± 0.015 with the adapter. The observed flowrate was within 5% of programed flowrate for puffing (25 mL/s), puff-associated respiration (450 mL/s), and tidal inhalation (350 mL/s). The maximum flowrate observed in the fabricated BAES was 450 mL/s, exceeding the lower target nominal wall shear stress of 0.025 Pa upstream of the bifurcation and fell below the target of 0.02 Pa downstream. This in vitro system addresses several gaps observed in commercially available systems and may be used to study many inhaled aerosols. The current work illustrates how in silico models may be used to correlate results of an in vitro study to in vivo conditions, rather than attempting to design an in vitro system that performs exactly as the human respiratory tract.
Collapse
Affiliation(s)
- S. Emma Sarles
- Department of Mechanical Engineering, Rochester Institute of Technology, 77 Lomb Memorial Drive, Rochester, NY 14623
| | - Edward C. Hensel
- Department of Mechanical Engineering, Rochester Institute of Technology, 77 Lomb Memorial Drive, Rochester, NY 14623
| | - Janessa Terry
- Department of Mechanical Engineering, Rochester Institute of Technology, 77 Lomb Memorial Drive, Rochester, NY 14623
| | - Caleb Nuss
- Department of Mechanical Engineering, Rochester Institute of Technology, 77 Lomb Memorial Drive, Rochester, NY 14623
| | - Risa J. Robinson
- Department of Mechanical Engineering, Rochester Institute of Technology, 77 Lomb Memorial Drive, Rochester, NY 14623
| |
Collapse
|
26
|
Hughes L, Centner C. Idiosyncratic bone responses to blood flow restriction exercise: new insights and future directions. J Appl Physiol (1985) 2024; 136:283-297. [PMID: 37994414 PMCID: PMC11212818 DOI: 10.1152/japplphysiol.00723.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 11/14/2023] [Accepted: 11/21/2023] [Indexed: 11/24/2023] Open
Abstract
Applying blood flow restriction (BFR) during low-load exercise induces beneficial adaptations of the myotendinous and neuromuscular systems. Despite the low mechanical tension, BFR exercise facilitates a localized hypoxic environment and increase in metabolic stress, widely regarded as the primary stimulus for tissue adaptations. First evidence indicates that low-load BFR exercise is effective in promoting an osteogenic response in bone, although this has previously been postulated to adapt primarily during high-impact weight-bearing exercise. Besides studies investigating the acute response of bone biomarkers following BFR exercise, first long-term trials demonstrate beneficial adaptations in bone in both healthy and clinical populations. Despite the increasing number of studies, the physiological mechanisms are largely unknown. Moreover, heterogeneity in methodological approaches such as biomarkers of bone metabolism measured, participant and study characteristics, and time course of measurement renders it difficult to formulate accurate conclusions. Furthermore, incongruity in the methods of BFR application (e.g., cuff pressure) limits the comparability of datasets and thus hinders generalizability of study findings. Appropriate use of biomarkers, effective BFR application, and befitting study design have the potential to progress knowledge on the acute and chronic response of bone to BFR exercise and contribute toward the development of a novel strategy to protect or enhance bone health. Therefore, the purpose of the present synthesis review is to 1) evaluate current mechanistic evidence; 2) discuss and offer explanations for similar and contrasting data findings; and 3) create a methodological framework for future mechanistic and applied research.
Collapse
Affiliation(s)
- Luke Hughes
- Department of Sport Exercise & Rehabilitation, Northumbria University, Newcastle upon Tyne, United Kingdom
| | - Christoph Centner
- Department of Sport and Sport Science, University of Freiburg, Freiburg, Germany
- Praxisklinik Rennbahn, Muttenz, Switzerland
| |
Collapse
|
27
|
He Q, Xu S, He F, Wu Z, Wu F, Zhou R, Zhou B, Li F, Yang X. Combined Proteomic and Phosphoproteomic Characterization of the Molecular Regulators and Functional Modules During Pancreatic Progenitor Cell Development. J Proteome Res 2024; 23:40-51. [PMID: 37993262 DOI: 10.1021/acs.jproteome.3c00309] [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] [Indexed: 11/24/2023]
Abstract
Differentiated multipotent pancreatic progenitors have major advantages for both modeling pancreas development and preventing or treating diabetes. Despite significant advancements in inducing the differentiation of human pluripotent stem cells into insulin-producing cells, the complete mechanism governing proliferation and differentiation remains poorly understood. This study used large-scale mass spectrometry to characterize molecular processes at various stages of human embryonic stem cell (hESC) differentiation toward pancreatic progenitors. hESCs were induced into pancreatic progenitor cells in a five-stage differentiation protocol. A high-performance liquid chromatography-mass spectrometry platform was used to undertake comprehensive proteome and phosphoproteome profiling of cells at different stages. A series of bioinformatic explorations, including coregulated modules, gene regulatory networks, and phosphosite enrichment analysis, were then conducted. A total of 27,077 unique phosphorylated sites and 8122 proteins were detected, including several cyclin-dependent kinases at the initial stage of cell differentiation. Furthermore, we discovered that ERK1, a member of the MAPK cascade, contributed to proliferation at an early stage. Finally, Western blotting confirmed that the phosphosites from SIRT1 and CHEK1 could inhibit the corresponding substrate abundance in the late stage. Thus, this study extends our understanding of the molecular mechanism during pancreatic cell development.
Collapse
Affiliation(s)
- Qian He
- Translational Medicine Collaborative Innovation Center, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; the Second Clinical Medical College of Jinan University), Shenzhen 518055, China
- School of Food and Drug, Shenzhen Polytechnic, Shenzhen 518055, China
- Guangdong Engineering Technology Research Center of Stem Cell and Cell Therapy, Shenzhen Key Laboratory of Stem Cell Research and Clinical Transformation, Shenzhen Immune Cell Therapy Public Service Platform, Shenzhen 518020, China
- Institute of Health Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Shaohang Xu
- Deepxomics Co., Ltd., Shenzhen 518000, China
| | - Fei He
- Translational Medicine Collaborative Innovation Center, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; the Second Clinical Medical College of Jinan University), Shenzhen 518055, China
- Guangdong Engineering Technology Research Center of Stem Cell and Cell Therapy, Shenzhen Key Laboratory of Stem Cell Research and Clinical Transformation, Shenzhen Immune Cell Therapy Public Service Platform, Shenzhen 518020, China
- Institute of Health Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zubiao Wu
- Translational Medicine Collaborative Innovation Center, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; the Second Clinical Medical College of Jinan University), Shenzhen 518055, China
- Guangdong Engineering Technology Research Center of Stem Cell and Cell Therapy, Shenzhen Key Laboratory of Stem Cell Research and Clinical Transformation, Shenzhen Immune Cell Therapy Public Service Platform, Shenzhen 518020, China
- Institute of Health Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Fujian Wu
- Translational Medicine Collaborative Innovation Center, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; the Second Clinical Medical College of Jinan University), Shenzhen 518055, China
- Guangdong Engineering Technology Research Center of Stem Cell and Cell Therapy, Shenzhen Key Laboratory of Stem Cell Research and Clinical Transformation, Shenzhen Immune Cell Therapy Public Service Platform, Shenzhen 518020, China
- Post-doctoral Scientific Research Station of Basic Medicine, Jinan University, Guangzhou 510632, China
- Institute of Health Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Ruo Zhou
- Deepxomics Co., Ltd., Shenzhen 518000, China
| | - Baojin Zhou
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Furong Li
- Translational Medicine Collaborative Innovation Center, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; the Second Clinical Medical College of Jinan University), Shenzhen 518055, China
- Guangdong Engineering Technology Research Center of Stem Cell and Cell Therapy, Shenzhen Key Laboratory of Stem Cell Research and Clinical Transformation, Shenzhen Immune Cell Therapy Public Service Platform, Shenzhen 518020, China
- Institute of Health Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xiaofei Yang
- Translational Medicine Collaborative Innovation Center, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; the Second Clinical Medical College of Jinan University), Shenzhen 518055, China
- Guangdong Engineering Technology Research Center of Stem Cell and Cell Therapy, Shenzhen Key Laboratory of Stem Cell Research and Clinical Transformation, Shenzhen Immune Cell Therapy Public Service Platform, Shenzhen 518020, China
- Institute of Health Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
28
|
Southern BD, Li H, Mao H, Crish JF, Grove LM, Scheraga RG, Mansoor S, Reinhardt A, Abraham S, Deshpande G, Loui A, Ivanov AI, Rosenfeld SS, Bresnick AR, Olman MA. A novel mechanoeffector role of fibroblast S100A4 in myofibroblast transdifferentiation and fibrosis. J Biol Chem 2024; 300:105530. [PMID: 38072048 PMCID: PMC10789633 DOI: 10.1016/j.jbc.2023.105530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/25/2023] [Accepted: 10/28/2023] [Indexed: 12/23/2023] Open
Abstract
Fibroblast to myofibroblast transdifferentiation mediates numerous fibrotic disorders, such as idiopathic pulmonary fibrosis (IPF). We have previously demonstrated that non-muscle myosin II (NMII) is activated in response to fibrotic lung extracellular matrix, thereby mediating myofibroblast transdifferentiation. NMII-A is known to interact with the calcium-binding protein S100A4, but the mechanism by which S100A4 regulates fibrotic disorders is unclear. In this study, we show that fibroblast S100A4 is a calcium-dependent, mechanoeffector protein that is uniquely sensitive to pathophysiologic-range lung stiffness (8-25 kPa) and thereby mediates myofibroblast transdifferentiation. Re-expression of endogenous fibroblast S100A4 rescues the myofibroblastic phenotype in S100A4 KO fibroblasts. Analysis of NMII-A/actin dynamics reveals that S100A4 mediates the unraveling and redistribution of peripheral actomyosin to a central location, resulting in a contractile myofibroblast. Furthermore, S100A4 loss protects against murine in vivo pulmonary fibrosis, and S100A4 expression is dysregulated in IPF. Our data reveal a novel mechanosensor/effector role for endogenous fibroblast S100A4 in inducing cytoskeletal redistribution in fibrotic disorders such as IPF.
Collapse
Affiliation(s)
- Brian D Southern
- Lerner Research Institute Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Haiyan Li
- Lerner Research Institute Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Hongxia Mao
- Lerner Research Institute Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - James F Crish
- Lerner Research Institute Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Lisa M Grove
- Lerner Research Institute Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Rachel G Scheraga
- Lerner Research Institute Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Sanaa Mansoor
- Lerner Research Institute Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Amanda Reinhardt
- Lerner Research Institute Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Susamma Abraham
- Lerner Research Institute Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Gauravi Deshpande
- Lerner Research Institute Imaging Core, Cleveland Clinic, Cleveland, Ohio, USA
| | - Alicia Loui
- University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Andrei I Ivanov
- Lerner Research Institute Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Steven S Rosenfeld
- Division of Hematology/Oncology, Mayo Clinic Jacksonville, Jacksonville, Florida, USA
| | - Anne R Bresnick
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Mitchell A Olman
- Lerner Research Institute Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA; Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, USA.
| |
Collapse
|
29
|
Davidson CD, Midekssa FS, DePalma SJ, Kamen JL, Wang WY, Jayco DKP, Wieger ME, Baker BM. Mechanical Intercellular Communication via Matrix-Borne Cell Force Transmission During Vascular Network Formation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306210. [PMID: 37997199 PMCID: PMC10797481 DOI: 10.1002/advs.202306210] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Indexed: 11/25/2023]
Abstract
Intercellular communication is critical to the formation and homeostatic function of all tissues. Previous work has shown that cells can communicate mechanically via the transmission of cell-generated forces through their surrounding extracellular matrix, but this process is not well understood. Here, mechanically defined, synthetic electrospun fibrous matrices are utilized in conjunction with a microfabrication-based cell patterning approach to examine mechanical intercellular communication (MIC) between endothelial cells (ECs) during their assembly into interconnected multicellular networks. It is found that cell force-mediated matrix displacements in deformable fibrous matrices underly directional extension and migration of neighboring ECs toward each other prior to the formation of stable cell-cell connections enriched with vascular endothelial cadherin (VE-cadherin). A critical role is also identified for calcium signaling mediated by focal adhesion kinase and mechanosensitive ion channels in MIC that extends to multicellular assembly of 3D vessel-like networks when ECs are embedded within fibrin hydrogels. These results illustrate a role for cell-generated forces and ECM mechanical properties in multicellular assembly of capillary-like EC networks and motivates the design of biomaterials that promote MIC for vascular tissue engineering.
Collapse
Affiliation(s)
| | - Firaol S. Midekssa
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMI48109USA
| | - Samuel J. DePalma
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMI48109USA
| | - Jordan L. Kamen
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMI48109USA
| | - William Y. Wang
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMI48109USA
| | | | - Megan E. Wieger
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMI48109USA
| | - Brendon M. Baker
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMI48109USA
- Department of Chemical EngineeringUniversity of MichiganAnn ArborMI48109USA
| |
Collapse
|
30
|
Boulingre M, Portillo-Lara R, Green RA. Biohybrid neural interfaces: improving the biological integration of neural implants. Chem Commun (Camb) 2023; 59:14745-14758. [PMID: 37991846 PMCID: PMC10720954 DOI: 10.1039/d3cc05006h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 11/10/2023] [Indexed: 11/24/2023]
Abstract
Implantable neural interfaces (NIs) have emerged in the clinic as outstanding tools for the management of a variety of neurological conditions caused by trauma or disease. However, the foreign body reaction triggered upon implantation remains one of the major challenges hindering the safety and longevity of NIs. The integration of tools and principles from biomaterial design and tissue engineering has been investigated as a promising strategy to develop NIs with enhanced functionality and performance. In this Feature Article, we highlight the main bioengineering approaches for the development of biohybrid NIs with an emphasis on relevant device design criteria. Technical and scientific challenges associated with the fabrication and functional assessment of technologies composed of both artificial and biological components are discussed. Lastly, we provide future perspectives related to engineering, regulatory, and neuroethical challenges to be addressed towards the realisation of the promise of biohybrid neurotechnology.
Collapse
Affiliation(s)
- Marjolaine Boulingre
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2AZ, UK
| | - Roberto Portillo-Lara
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2AZ, UK
| | - Rylie A Green
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2AZ, UK
| |
Collapse
|
31
|
Zhu C, Lu Y, Peng W, Gao H, Cao X, Su M, Wu Z, Huo X, Yu C. Stretchable Sponge-Based Electrochemical Biosensor for Real-Time Sensing of Cells in Three-Dimensional Culture. Anal Chem 2023; 95:16885-16891. [PMID: 37937709 DOI: 10.1021/acs.analchem.3c02676] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
For the study of cell biology, real-time information on cell physiological processes will be more accurate and closer to the in vivo condition in a three-dimensional (3D) culture system. Although most reported 3D cell culture scaffolds can better mimic the in vivo dynamic microenvironment, the real-time analysis technique is deficient or lacking. Herein, a stretchable and conductive 3D scaffold is developed to construct an electrochemical biosensor for real-time monitoring of cell release in 3D culture under stimulation of drug stimulant and mechanical force. In our design, the polyurethane sponge (PU) dipped with conductive carbon ink (CC/PU) was used as a conductive scaffold, and gold nanoparticles (nano-Au) were electrodeposited on the CC/PU (nano-Au CC/PU) to improve the electrochemical sensing performance. The prepared nano-Au CC/PU scaffold exhibits a good electrocatalytic ability to H2O2 with a linear range from 20 nM to 43 μM. Due to the great biocompatibility, HeLa cells can be cultured directly on the nano-Au CC/PU and the in situ and real-time tracking of H2O2 secretion from cells was achieved. The results demonstrate that both the drug stimulant and mechanical force can rapidly activate the release of reactive oxygen species. This study indicates that the stretchable 3D sensing scaffold has good potential for cell biology research in an in vivo-like microenvironment and can be extensively used in the fields of tissue engineering, drug screening, and pathological research.
Collapse
Affiliation(s)
- Cailing Zhu
- School of Public Health, Nantong University, Nantong 226019, P. R. China
| | - Yanling Lu
- Qidong Hospital of Traditional Chinese Medicine, Qidong, Jiangsu 226200, China
| | - Wenjing Peng
- School of Public Health, Nantong University, Nantong 226019, P. R. China
| | - Hui Gao
- School of Public Health, Nantong University, Nantong 226019, P. R. China
| | - Xiaoqing Cao
- School of Public Health, Nantong University, Nantong 226019, P. R. China
| | - Mengjie Su
- School of Public Health, Nantong University, Nantong 226019, P. R. China
| | - Zengqiang Wu
- School of Public Health, Nantong University, Nantong 226019, P. R. China
| | - Xiaolei Huo
- School of Public Health, Nantong University, Nantong 226019, P. R. China
| | - Chunmei Yu
- School of Public Health, Nantong University, Nantong 226019, P. R. China
| |
Collapse
|
32
|
Duan Y, Szlam F, Hu Y, Chen W, Li R, Ke Y, Sniecinski R, Salaita K. Detection of cellular traction forces via the force-triggered Cas12a-mediated catalytic cleavage of a fluorogenic reporter strand. Nat Biomed Eng 2023; 7:1404-1418. [PMID: 37957275 PMCID: PMC11289779 DOI: 10.1038/s41551-023-01114-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 09/26/2023] [Indexed: 11/15/2023]
Abstract
Molecular forces generated by cell receptors are infrequent and transient, and hence difficult to detect. Here we report an assay that leverages the CRISPR-associated protein 12a (Cas12a) to amplify the detection of cellular traction forces generated by as few as 50 adherent cells. The assay involves the immobilization of a DNA duplex modified with a ligand specific for a cell receptor. Traction forces of tens of piconewtons trigger the dehybridization of the duplex, exposing a cryptic Cas12-activating strand that sets off the indiscriminate Cas12-mediated cleavage of a fluorogenic reporter strand. We used the assay to perform hundreds of force measurements using human platelets from a single blood draw to extract individualized dose-response curves and half-maximal inhibitory concentrations for a panel of antiplatelet drugs. For seven patients who had undergone cardiopulmonary bypass, platelet dysfunction strongly correlated with the need for platelet transfusion to limit bleeding. The Cas12a-mediated detection of cellular traction forces may be used to assess cell state, and to screen for genes, cell-adhesion ligands, drugs or metabolites that modulate cell mechanics.
Collapse
Affiliation(s)
- Yuxin Duan
- Department of Chemistry, Emory University, Atlanta, GA, USA
| | - Fania Szlam
- Department of Anesthesiology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Yuesong Hu
- Department of Chemistry, Emory University, Atlanta, GA, USA
| | - Wenchun Chen
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Departments of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Renhao Li
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Departments of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Yonggang Ke
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Roman Sniecinski
- Department of Anesthesiology, School of Medicine, Emory University, Atlanta, GA, USA.
| | - Khalid Salaita
- Department of Chemistry, Emory University, Atlanta, GA, USA.
| |
Collapse
|
33
|
Li C, Schramma N, Wang Z, Qari NF, Jalaal M, Latz MI, Cai S. Ultrasensitive and robust mechanoluminescent living composites. SCIENCE ADVANCES 2023; 9:eadi8643. [PMID: 37862415 PMCID: PMC10588950 DOI: 10.1126/sciadv.adi8643] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/19/2023] [Indexed: 10/22/2023]
Abstract
Mechanosensing, the transduction of extracellular mechanical stimuli into intracellular biochemical signals, is a fundamental property of living cells. However, endowing synthetic materials with mechanosensing capabilities comparable to biological levels is challenging. Here, we developed ultrasensitive and robust mechanoluminescent living composites using hydrogels embedded with dinoflagellates, unicellular microalgae with a near-instantaneous and ultrasensitive bioluminescent response to mechanical stress. Not only did embedded dinoflagellates retain their intrinsic mechanoluminescence, but with hydrophobic coatings, living composites had a lifetime of ~5 months under harsh conditions with minimal maintenance. We 3D-printed living composites into large-scale mechanoluminescent structures with high spatial resolution, and we also enhanced their mechanical properties with double-network hydrogels. We propose a counterpart mathematical model that captured experimental mechanoluminescent observations to predict mechanoluminescence based on deformation and applied stress. We also demonstrated the use of the mechanosensing composites for biomimetic soft actuators that emitted colored light upon magnetic actuation. These mechanosensing composites have substantial potential in biohybrid sensors and robotics.
Collapse
Affiliation(s)
- Chenghai Li
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nico Schramma
- Van der Waals-Zeeman Institute, Institute of Physics, University of Amsterdam, Science Park 904, Amsterdam 1098XH, Netherlands
| | - Zijun Wang
- Materials Science and Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nada F. Qari
- Materials Science and Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Maziyar Jalaal
- Van der Waals-Zeeman Institute, Institute of Physics, University of Amsterdam, Science Park 904, Amsterdam 1098XH, Netherlands
| | - Michael I. Latz
- Scripps Institution of Oceanography, University of California, San Diego, La Jolla, CA 92093, USA
| | - Shengqiang Cai
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA 92093, USA
- Materials Science and Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
34
|
Gulati K, Adachi T. Profiling to Probing: Atomic force microscopy to characterize nano-engineered implants. Acta Biomater 2023; 170:15-38. [PMID: 37562516 DOI: 10.1016/j.actbio.2023.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 07/26/2023] [Accepted: 08/03/2023] [Indexed: 08/12/2023]
Abstract
Surface modification of implants in the nanoscale or implant nano-engineering has been recognized as a strategy for augmenting implant bioactivity and achieving long-term implant success. Characterizing and optimizing implant characteristics is crucial to achieving desirable effects post-implantation. Modified implant enables tailored, guided and accelerated tissue integration; however, our understanding is limited to multicellular (bulk) interactions. Finding the nanoscale forces experienced by a single cell on nano-engineered implants will aid in predicting implants' bioactivity and engineering the next generation of bioactive implants. Atomic force microscope (AFM) is a unique tool that enables surface characterization and understanding of the interactions between implant surface and biological tissues. The characterization of surface topography using AFM to gauge nano-engineered implants' characteristics (topographical, mechanical, chemical, electrical and magnetic) and bioactivity (adhesion of cells) is presented. A special focus of the review is to discuss the use of single-cell force spectroscopy (SCFS) employing AFM to investigate the minute forces involved with the adhesion of a single cell (resident tissue cell or bacterium) to the surface of nano-engineered implants. Finally, the research gaps and future perspectives relating to AFM-characterized current and emerging nano-engineered implants are discussed towards achieving desirable bioactivity performances. This review highlights the use of advanced AFM-based characterization of nano-engineered implant surfaces via profiling (investigating implant topography) or probing (using a single cell as a probe to study precise adhesive forces with the implant surface). STATEMENT OF SIGNIFICANCE: Nano-engineering is emerging as a surface modification platform for implants to augment their bioactivity and achieve favourable treatment outcomes. In this extensive review, we closely examine the use of Atomic Force Microscopy (AFM) to characterize the properties of nano-engineered implant surfaces (topography, mechanical, chemical, electrical and magnetic). Next, we discuss Single-Cell Force Spectroscopy (SCFS) via AFM towards precise force quantification encompassing a single cell's interaction with the implant surface. This interdisciplinary review will appeal to researchers from the broader scientific community interested in implants and cell adhesion to implants and provide an improved understanding of the surface characterization of nano-engineered implants.
Collapse
Affiliation(s)
- Karan Gulati
- Institute for Life and Medical Sciences, Kyoto University, Sakyo, Kyoto 606-8507, Japan; The University of Queensland, School of Dentistry, Herston QLD 4006, Australia.
| | - Taiji Adachi
- Institute for Life and Medical Sciences, Kyoto University, Sakyo, Kyoto 606-8507, Japan
| |
Collapse
|
35
|
Yousafzai MS, Hammer JA. Using Biosensors to Study Organoids, Spheroids and Organs-on-a-Chip: A Mechanobiology Perspective. BIOSENSORS 2023; 13:905. [PMID: 37887098 PMCID: PMC10605946 DOI: 10.3390/bios13100905] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/13/2023] [Accepted: 09/19/2023] [Indexed: 10/28/2023]
Abstract
The increasing popularity of 3D cell culture models is being driven by the demand for more in vivo-like conditions with which to study the biochemistry and biomechanics of numerous biological processes in health and disease. Spheroids and organoids are 3D culture platforms that self-assemble and regenerate from stem cells, tissue progenitor cells or cell lines, and that show great potential for studying tissue development and regeneration. Organ-on-a-chip approaches can be used to achieve spatiotemporal control over the biochemical and biomechanical signals that promote tissue growth and differentiation. These 3D model systems can be engineered to serve as disease models and used for drug screens. While culture methods have been developed to support these 3D structures, challenges remain to completely recapitulate the cell-cell and cell-matrix biomechanical interactions occurring in vivo. Understanding how forces influence the functions of cells in these 3D systems will require precise tools to measure such forces, as well as a better understanding of the mechanobiology of cell-cell and cell-matrix interactions. Biosensors will prove powerful for measuring forces in both of these contexts, thereby leading to a better understanding of how mechanical forces influence biological systems at the cellular and tissue levels. Here, we discussed how biosensors and mechanobiological research can be coupled to develop accurate, physiologically relevant 3D tissue models to study tissue development, function, malfunction in disease, and avenues for disease intervention.
Collapse
Affiliation(s)
- Muhammad Sulaiman Yousafzai
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - John A. Hammer
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
36
|
Su É, Villard C, Manneville JB. Mitochondria: At the crossroads between mechanobiology and cell metabolism. Biol Cell 2023; 115:e2300010. [PMID: 37326132 DOI: 10.1111/boc.202300010] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 06/11/2023] [Accepted: 06/13/2023] [Indexed: 06/17/2023]
Abstract
Metabolism and mechanics are two key facets of structural and functional processes in cells, such as growth, proliferation, homeostasis and regeneration. Their reciprocal regulation has been increasingly acknowledged in recent years: external physical and mechanical cues entail metabolic changes, which in return regulate cell mechanosensing and mechanotransduction. Since mitochondria are pivotal regulators of metabolism, we review here the reciprocal links between mitochondrial morphodynamics, mechanics and metabolism. Mitochondria are highly dynamic organelles which sense and integrate mechanical, physical and metabolic cues to adapt their morphology, the organization of their network and their metabolic functions. While some of the links between mitochondrial morphodynamics, mechanics and metabolism are already well established, others are still poorly documented and open new fields of research. First, cell metabolism is known to correlate with mitochondrial morphodynamics. For instance, mitochondrial fission, fusion and cristae remodeling allow the cell to fine-tune its energy production through the contribution of mitochondrial oxidative phosphorylation and cytosolic glycolysis. Second, mechanical cues and alterations in mitochondrial mechanical properties reshape and reorganize the mitochondrial network. Mitochondrial membrane tension emerges as a decisive physical property which regulates mitochondrial morphodynamics. However, the converse link hypothesizing a contribution of morphodynamics to mitochondria mechanics and/or mechanosensitivity has not yet been demonstrated. Third, we highlight that mitochondrial mechanics and metabolism are reciprocally regulated, although little is known about the mechanical adaptation of mitochondria in response to metabolic cues. Deciphering the links between mitochondrial morphodynamics, mechanics and metabolism still presents significant technical and conceptual challenges but is crucial both for a better understanding of mechanobiology and for potential novel therapeutic approaches in diseases such as cancer.
Collapse
Affiliation(s)
- Émilie Su
- Laboratoire Matière et Systèmes Complexes (MSC), Université Paris Cité - CNRS, UMR 7057, Paris, France
- Laboratoire Interdisciplinaire des Énergies de Demain (LIED), Université Paris Cité - CNRS, UMR 8236, Paris, France
| | - Catherine Villard
- Laboratoire Interdisciplinaire des Énergies de Demain (LIED), Université Paris Cité - CNRS, UMR 8236, Paris, France
| | - Jean-Baptiste Manneville
- Laboratoire Matière et Systèmes Complexes (MSC), Université Paris Cité - CNRS, UMR 7057, Paris, France
| |
Collapse
|
37
|
Yadav S, Singha P, Nguyen NK, Ooi CH, Kashaninejad N, Nguyen NT. Uniaxial Cyclic Cell Stretching Device for Accelerating Cellular Studies. MICROMACHINES 2023; 14:1537. [PMID: 37630073 PMCID: PMC10456305 DOI: 10.3390/mi14081537] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/24/2023] [Accepted: 07/29/2023] [Indexed: 08/27/2023]
Abstract
Cellular response to mechanical stimuli is a crucial factor for maintaining cell homeostasis. The interaction between the extracellular matrix and mechanical stress plays a significant role in organizing the cytoskeleton and aligning cells. Tools that apply mechanical forces to cells and tissues, as well as those capable of measuring the mechanical properties of biological cells, have greatly contributed to our understanding of fundamental mechanobiology. These tools have been extensively employed to unveil the substantial influence of mechanical cues on the development and progression of various diseases. In this report, we present an economical and high-performance uniaxial cell stretching device. This paper reports the detailed operation concept of the device, experimental design, and characterization. The device was tested with MDA-MB-231 breast cancer cells. The experimental results agree well with previously documented morphological changes resulting from stretching forces on cancer cells. Remarkably, our new device demonstrates comparable cellular changes within 30 min compared with the previous 2 h stretching duration. This third-generation device significantly improved the stretching capabilities compared with its previous counterparts, resulting in a remarkable reduction in stretching time and a substantial increase in overall efficiency. Moreover, the device design incorporates an open-source software interface, facilitating convenient parameter adjustments such as strain, stretching speed, frequency, and duration. Its versatility enables seamless integration with various optical microscopes, thereby yielding novel insights into the realm of mechanobiology.
Collapse
Affiliation(s)
| | | | | | | | | | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan, QLD 4111, Australia; (S.Y.); (P.S.); (N.-K.N.); (C.H.O.); (N.K.)
| |
Collapse
|
38
|
Radstake WE, Gautam K, Miranda S, Van Rompay C, Vermeesen R, Tabury K, Verslegers M, Dowson A, Gorissen J, van Loon JJWA, Savage NDL, Baatout S, Baselet B. Gravitational effects on fibroblasts' function in relation to wound healing. NPJ Microgravity 2023; 9:48. [PMID: 37344509 DOI: 10.1038/s41526-023-00286-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 05/25/2023] [Indexed: 06/23/2023] Open
Abstract
The spaceflight environment imposes risks for maintaining a healthy skin function as the observed delayed wound healing can contribute to increased risks of infection. To counteract delayed wound healing in space, a better understanding of the fibroblasts' reaction to altered gravity levels is needed. In this paper, we describe experiments that were carried out at the Large Diameter Centrifuge located in ESA-ESTEC as part of the ESA Academy 2021 Spin Your Thesis! Campaign. We exposed dermal fibroblasts to a set of altered gravity levels, including transitions between simulated microgravity and hypergravity. The addition of the stress hormone cortisol to the cell culture medium was done to account for possible interaction effects of gravity and cortisol exposure. Results show a main impact of cortisol on the secretion of pro-inflammatory cytokines as well as extracellular matrix proteins. Altered gravity mostly induced a delay in cellular migration and changes in mechanosensitive cell structures. Furthermore, 20 × g hypergravity transitions induced changes in nuclear morphology. These findings provide insights into the effect of gravity transitions on the fibroblasts' function related to wound healing, which may be useful for the development of countermeasures.
Collapse
Affiliation(s)
- Wilhelmina E Radstake
- Radiobiology Unit, SCK CEN, Belgian Nuclear Research Centre, 2400, Mol, Belgium
- Department of Molecular Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000, Ghent, Belgium
| | - Kiran Gautam
- Radiobiology Unit, SCK CEN, Belgian Nuclear Research Centre, 2400, Mol, Belgium
| | - Silvana Miranda
- Radiobiology Unit, SCK CEN, Belgian Nuclear Research Centre, 2400, Mol, Belgium
- Department of Molecular Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000, Ghent, Belgium
| | - Cynthia Van Rompay
- Radiobiology Unit, SCK CEN, Belgian Nuclear Research Centre, 2400, Mol, Belgium
| | - Randy Vermeesen
- Radiobiology Unit, SCK CEN, Belgian Nuclear Research Centre, 2400, Mol, Belgium
| | - Kevin Tabury
- Radiobiology Unit, SCK CEN, Belgian Nuclear Research Centre, 2400, Mol, Belgium
- Department of Biomedical Engineering, University of South Carolina, Columbia, SC, 29208, USA
| | - Mieke Verslegers
- Radiobiology Unit, SCK CEN, Belgian Nuclear Research Centre, 2400, Mol, Belgium
| | - Alan Dowson
- Serco Nederland for the European Space Agency (ESA), European Space Research and Technology Centre (ESTEC), TEC-MMG, Keplerlaan 1, 2201, AZ, Noordwijk, the Netherlands
| | | | - Jack J W A van Loon
- ESA/ESTEC, Keplerlaan 1, 2200, AZ, Noordwijk, The Netherlands
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam Movement Sciences & Amsterdam Bone Center (ABC), Amsterdam UMC location Vrije Universiteit Amsterdam & Academic Center for Dentistry Amsterdam (ACTA), Gustav Mahlerlaan 3004, 1081, LA, Amsterdam, The Netherlands
| | - Nigel D L Savage
- HE Space Operations for the European Space Agency, ESA/ESTEC, Keplerlaan 1, 2200, AZ, Noordwijk, The Netherlands
| | - Sarah Baatout
- Radiobiology Unit, SCK CEN, Belgian Nuclear Research Centre, 2400, Mol, Belgium
- Department of Molecular Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000, Ghent, Belgium
| | - Bjorn Baselet
- Radiobiology Unit, SCK CEN, Belgian Nuclear Research Centre, 2400, Mol, Belgium.
| |
Collapse
|
39
|
Davis MJ, Earley S, Li YS, Chien S. Vascular mechanotransduction. Physiol Rev 2023; 103:1247-1421. [PMID: 36603156 PMCID: PMC9942936 DOI: 10.1152/physrev.00053.2021] [Citation(s) in RCA: 97] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno, Nevada
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
- Department of Medicine, University of California, San Diego, California
| |
Collapse
|
40
|
Melo-Fonseca F, Carvalho O, Gasik M, Miranda G, Silva FS. Mechanical stimulation devices for mechanobiology studies: a market, literature, and patents review. Biodes Manuf 2023. [DOI: 10.1007/s42242-023-00232-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
AbstractSignificant advancements in various research and technological fields have contributed to remarkable findings on the physiological dynamics of the human body. To more closely mimic the complex physiological environment, research has moved from two-dimensional (2D) culture systems to more sophisticated three-dimensional (3D) dynamic cultures. Unlike bioreactors or microfluidic-based culture models, cells are typically seeded on polymeric substrates or incorporated into 3D constructs which are mechanically stimulated to investigate cell response to mechanical stresses, such as tensile or compressive. This review focuses on the working principles of mechanical stimulation devices currently available on the market or custom-built by research groups or protected by patents and highlights the main features still open to improvement. These are the features which could be focused on to perform, in the future, more reliable and accurate mechanobiology studies.
Graphic abstract
Collapse
|
41
|
Elayan H, Eckford AW, Adve R. Selectivity of Protein Interactions Stimulated by Terahertz Signals. IEEE Trans Nanobioscience 2023; 22:318-328. [PMID: 35797314 DOI: 10.1109/tnb.2022.3189549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
It has been established that Terahertz (THz) band signals can interact with biomolecules through resonant modes. Specifically, of interest here, protein activation. Our research goal is to show how directing the mechanical signaling inside protein molecules using THz signals can control changes in their structure and activate associated biochemical and biomechanical events. To establish that, we formulate a selectivity metric that quantifies the system performance and captures the capability of the nanoantenna to induce a conformational change in the desired protein molecule/population. The metric provides a score between -1 and 1 that indicates the degree of control we have over the system to achieve targeted protein interactions. To develop the selectivity measure, we first use the Langevin stochastic equation driven by an external force to model the protein behavior. We then determine the probability of protein folding by computing the steady-state energy of the driven protein and then generalize our model to account for protein populations. Our numerical analysis results indicate that a maximum selectivity score is attained when only the targeted population experiences a folding behavior due to the impinging THz signal. From the achieved selectivity values, we conclude that the system response not only depends on the resonant frequency but also on the system controlling parameters namely, the nanoantenna force, the damping constant, and the abundance of each protein population. Based on the selectivity metric, the nanoantenna must be tuned to a frequency that is not necessarily the resonant frequency of the protein. The presented work sheds light on the potential associated with the electromagnetic-based control of protein networks, which could lead to a plethora of applications in the medical field ranging from bio-sensing to targeted therapy.
Collapse
|
42
|
Liao X, Li X, Liu R. Extracellular-matrix mechanics regulate cellular metabolism: A ninja warrior behind mechano-chemo signaling crosstalk. Rev Endocr Metab Disord 2023; 24:207-220. [PMID: 36385696 DOI: 10.1007/s11154-022-09768-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/21/2022] [Indexed: 11/18/2022]
Abstract
Mechanical forces are the indispensable constituent of environmental cues, such as gravity, barometric pressure, vibration, and contact with bodies, which are involved in pattern and organogenesis, providing mechanical input to tissues and determining the ultimate fate of cells. Extracellular matrix (ECM) stiffness, the slow elastic force, carries the external physical force load onto the cell or outputs the internal force exerted by the cell and its neighbors into the environment. Accumulating evidence illustrates the pivotal role of ECM stiffness in the regulation of organogenesis, maintenance of tissue homeostasis, and the development of multiple diseases, which is largely fulfilled through its systematical impact on cellular metabolism. This review summarizes the establishment and regulation of ECM stiffness, the mechanisms underlying how ECM stiffness is sensed by cells and signals to modulate diverse cell metabolic pathways, and the physiological and pathological significance of the ECM stiffness-cell metabolism axis.
Collapse
Affiliation(s)
- Xiaoyu Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, 14, Section 3, Renminnan Road, Chengdu, 610041, Sichuan, China
| | - Xin Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, 14, Section 3, Renminnan Road, Chengdu, 610041, Sichuan, China
| | - Rui Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, 14, Section 3, Renminnan Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
43
|
Shao CG, Sinha NR, Mohan RR, Webel AD. Novel Therapies for the Prevention of Fibrosis in Glaucoma Filtration Surgery. Biomedicines 2023; 11:657. [PMID: 36979636 PMCID: PMC10045591 DOI: 10.3390/biomedicines11030657] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/07/2023] [Accepted: 02/15/2023] [Indexed: 02/24/2023] Open
Abstract
Conjunctival fibrosis remains the major impediment to the success of glaucoma filtration surgery. Anti-metabolites remain the gold standard for mitigating post-surgical fibrosis, but they are associated with high complication rates and surgical failure rates. Establishing a more targeted approach to attenuate conjunctival fibrosis may revolutionize the surgical approach to glaucoma. A new strategy is needed to prevent progressive tissue remodeling and formation of a fibrotic scar, subsequently increasing surgical success and reducing the prevalence of glaucoma-related vision loss. Advancements in our understanding of molecular signaling and biomechanical cues in the conjunctival tissue architecture are broadening the horizon for new therapies and biomaterials for the mitigation of fibrosis. This review aims to highlight the strategies and current state of promising future approaches for targeting fibrosis in glaucoma filtration surgery.
Collapse
Affiliation(s)
| | - Nishant R. Sinha
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65212, USA
- One-Health Vision Research Program, Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Rajiv R. Mohan
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 65212, USA
- One-Health Vision Research Program, Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO 65212, USA
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Aaron D. Webel
- Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
44
|
Hu J, Chen Q, Zhu H, Hou L, Liu W, Yang Q, Shen H, Chai G, Zhang B, Chen S, Cai Z, Wu C, Hong F, Li H, Chen S, Xiao N, Wang ZX, Zhang X, Wang B, Zhang L, Mo W. Microglial Piezo1 senses Aβ fibril stiffness to restrict Alzheimer's disease. Neuron 2023; 111:15-29.e8. [PMID: 36368316 DOI: 10.1016/j.neuron.2022.10.021] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/15/2022] [Accepted: 10/12/2022] [Indexed: 11/12/2022]
Abstract
The pathology of Alzheimer's disease (AD) is featured with extracellular amyloid-β (Aβ) plaques, whose impact on the mechanical properties of the surrounding brain tissues is unclear. Microglia sense and integrate biochemical cues of the microenvironment. However, whether the microglial mechanosensing pathways influence AD pathogenesis is unknown. Here, we surveyed the elevated stiffness of Aβ-plaque-associated tissues and observed the selective upregulation of the mechanosensitive ion channel Piezo1 in Aβ-plaque-associated microglia. Piezo1 sensed the stiffness stimuli of Aβ fibrils and subsequently induced Ca2+ influx for microglial clustering, phagocytosis, and compacting of Aβ plaques. Microglia lacking Piezo1 led to the exacerbation of Aβ pathology and cognitive decline, whereas pharmacological activation of microglial Piezo1 ameliorated brain Aβ burden and cognitive impairment in 5 × FAD mice. Together, our results reveal that Piezo1, a mechanosensor of Aβ fibril stiffness in microglia, represents a potential therapeutic target for AD.
Collapse
Affiliation(s)
- Jin Hu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China.
| | - Qiang Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Hongrui Zhu
- Department of Anesthesiology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Lichao Hou
- Department of Anesthesiology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Wei Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Qihua Yang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Huidan Shen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China; Department of Anesthesiology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Guolin Chai
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Boxin Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Shaoxuan Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Zhiyu Cai
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Chongxin Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Fan Hong
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Hongda Li
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Sifang Chen
- Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, the First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Naian Xiao
- Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, the First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Zhan-Xiang Wang
- Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, the First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Xueqin Zhang
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Bo Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Liang Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China.
| | - Wei Mo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Department of Neuroscience, the First Affiliated Hospital, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China.
| |
Collapse
|
45
|
Kopecny LR, Lee BWH, Coroneo MT. A systematic review on the effects of ROCK inhibitors on proliferation and/or differentiation in human somatic stem cells: A hypothesis that ROCK inhibitors support corneal endothelial healing via acting on the limbal stem cell niche. Ocul Surf 2023; 27:16-29. [PMID: 36586668 DOI: 10.1016/j.jtos.2022.12.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/18/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022]
Abstract
Rho kinase inhibitors (ROCKi) have attracted growing multidisciplinary interest, particularly in Ophthalmology where the question as to how they promote corneal endothelial healing remains unresolved. Concurrently, stem cell biology has rapidly progressed in unravelling drivers of stem cell (SC) proliferation and differentiation, where mechanical niche factors and the actin cytoskeleton are increasingly recognized as key players. There is mounting evidence from the study of the peripheral corneal endothelium that supports the likelihood of an internal limbal stem cell niche. The possibility that ROCKi stimulate the endothelial SC niche has not been addressed. Furthermore, there is currently a paucity of data that directly evaluates whether ROCKi promotes corneal endothelial healing by acting on this limbal SC niche located near the transition zone. Therefore, we performed a systematic review examining the effects ROCKi on the proliferation and differentiation of human somatic SC, to provide insight into its effects on various human SC populations. An appraisal of electronic searches of four databases identified 1 in vivo and 58 in vitro studies (36 evaluated proliferation while 53 examined differentiation). Types of SC studied included mesenchymal (n = 32), epithelial (n = 11), epidermal (n = 8), hematopoietic and other (n = 8). The ROCK 1/2 selective inhibitor Y-27632 was used in almost all studies (n = 58), while several studies evaluated ≥2 ROCKi (n = 4) including fasudil, H-1152, and KD025. ROCKi significantly influenced human somatic SC proliferation in 81% of studies (29/36) and SC differentiation in 94% of studies (50/53). The present systemic review highlights that ROCKi are influential in regulating human SC proliferation and differentiation, and provides evidence to support the hypothesis that ROCKi promotes corneal endothelial division and maintenance via acting on the inner limbal SC niche.
Collapse
Affiliation(s)
- Lloyd R Kopecny
- School of Clinical Medicine, University of New South Wales, Sydney, Australia.
| | - Brendon W H Lee
- Department of Ophthalmology, School of Clinical Medicine, University of New South Wales, Level 2 South Wing, Edmund Blacket Building, Prince of Wales Hospital, Randwick, NSW, 2031, Australia
| | - Minas T Coroneo
- Department of Ophthalmology, Prince of Wales Hospital, Sydney, Australia
| |
Collapse
|
46
|
Abstract
Immune responses are governed by signals from the tissue microenvironment, and in addition to biochemical signals, mechanical cues and forces arising from the tissue, its extracellular matrix and its constituent cells shape immune cell function. Indeed, changes in biophysical properties of tissue alter the mechanical signals experienced by cells in many disease conditions, in inflammatory states and in the context of ageing. These mechanical cues are converted into biochemical signals through the process of mechanotransduction, and multiple pathways of mechanotransduction have been identified in immune cells. Such pathways impact important cellular functions including cell activation, cytokine production, metabolism, proliferation and trafficking. Changes in tissue mechanics may also represent a new form of 'danger signal' that alerts the innate and adaptive immune systems to the possibility of injury or infection. Tissue mechanics can change temporally during an infection or inflammatory response, offering a novel layer of dynamic immune regulation. Here, we review the emerging field of mechanoimmunology, focusing on how mechanical cues at the scale of the tissue environment regulate immune cell behaviours to initiate, propagate and resolve the immune response.
Collapse
|
47
|
Spiaggia G, Taladriz-Blanco P, Hengsberger S, Septiadi D, Geers C, Lee A, Rothen-Rutishauser B, Petri-Fink A. A Near-Infrared Mechanically Switchable Elastomeric Film as a Dynamic Cell Culture Substrate. Biomedicines 2022; 11:biomedicines11010030. [PMID: 36672538 PMCID: PMC9855853 DOI: 10.3390/biomedicines11010030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/09/2022] [Accepted: 12/17/2022] [Indexed: 12/25/2022] Open
Abstract
Commercial static cell culture substrates can usually not change their physical properties over time, resulting in a limited representation of the variation in biomechanical cues in vivo. To overcome this limitation, approaches incorporating gold nanoparticles to act as transducers to external stimuli have been employed. In this work, gold nanorods were embedded in an elastomeric matrix and used as photothermal transducers to fabricate biocompatible light-responsive substrates. The nanocomposite films analysed by lock-in thermography and nanoindentation show a homogeneous heat distribution and a greater stiffness when irradiated with NIR light. After irradiation, the initial stiffness values were recovered. In vitro experiments performed during NIR irradiation with NIH-3T3 fibroblasts demonstrated that these films were biocompatible and cells remained viable. Cells cultured on the light stiffened nanocomposite exhibited a greater proliferation rate and stronger focal adhesion clustering, indicating increased cell-surface binding strength.
Collapse
Affiliation(s)
- Giovanni Spiaggia
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Patricia Taladriz-Blanco
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
- International Iberian Nanotechnology Laboratory (INL), Water Quality Group, Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal
- Correspondence: (P.T.-B.); (A.P.-F.)
| | - Stefan Hengsberger
- School of Engineering and Architecture (HEIA-FR), HES-SO, University of Applied Science and Arts in Western Switzerland, Boulevard de Pérolles 80, 1700 Fribourg, Switzerland
| | - Dedy Septiadi
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Christoph Geers
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Aaron Lee
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | | | - Alke Petri-Fink
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
- Department of Chemistry, University of Fribourg, Chemin du Musée 9, 1700 Fribourg, Switzerland
- Correspondence: (P.T.-B.); (A.P.-F.)
| |
Collapse
|
48
|
Ghuloum FI, Johnson CA, Riobo-Del Galdo NA, Amer MH. From mesenchymal niches to engineered in vitro model systems: Exploring and exploiting biomechanical regulation of vertebrate hedgehog signalling. Mater Today Bio 2022; 17:100502. [PMID: 36457847 PMCID: PMC9707069 DOI: 10.1016/j.mtbio.2022.100502] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/08/2022] [Accepted: 11/20/2022] [Indexed: 11/23/2022] Open
Abstract
Tissue patterning is the result of complex interactions between transcriptional programs and various mechanical cues that modulate cell behaviour and drive morphogenesis. Vertebrate Hedgehog signalling plays key roles in embryogenesis and adult tissue homeostasis, and is central to skeletal development and the osteogenic differentiation of mesenchymal stem cells. The expression of several components of the Hedgehog signalling pathway have been reported to be mechanically regulated in mesodermal tissue patterning and osteogenic differentiation in response to external stimulation. Since a number of bone developmental defects and skeletal diseases, such as osteoporosis, are directly linked to aberrant Hedgehog signalling, a better knowledge of the regulation of Hedgehog signalling in the mechanosensitive bone marrow-residing mesenchymal stromal cells will present novel avenues for modelling these diseases and uncover novel opportunities for extracellular matrix-targeted therapies. In this review, we present a brief overview of the key molecular players involved in Hedgehog signalling and the basic concepts of mechanobiology, with a focus on bone development and regeneration. We also highlight the correlation between the activation of the Hedgehog signalling pathway in response to mechanical cues and osteogenesis in bone marrow-derived mesenchymal stromal cells. Finally, we propose different tissue engineering strategies to apply the expanding knowledge of 3D material-cell interactions in the modulation of Hedgehog signalling in vitro for fundamental and translational research applications.
Collapse
Affiliation(s)
- Fatmah I. Ghuloum
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait City, Kuwait
| | - Colin A. Johnson
- Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Natalia A. Riobo-Del Galdo
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, UK
| | - Mahetab H. Amer
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
49
|
Zhang H, Zeng H, Eklund A, Guo H, Priimagi A, Ikkala O. Feedback-controlled hydrogels with homeostatic oscillations and dissipative signal transduction. NATURE NANOTECHNOLOGY 2022; 17:1303-1310. [PMID: 36443600 PMCID: PMC9747616 DOI: 10.1038/s41565-022-01241-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 09/14/2022] [Indexed: 05/06/2023]
Abstract
Driving systems out of equilibrium under feedback control is characteristic for living systems, where homeostasis and dissipative signal transduction facilitate complex responses. This feature not only inspires dissipative dynamic functionalities in synthetic systems but also poses great challenges in designing novel pathways. Here we report feedback-controlled systems comprising two coupled hydrogels driven by constant light, where the system can be tuned to undergo stable homeostatic self-oscillations or damped steady states of temperature. We demonstrate that stable temperature oscillations can be utilized for dynamic colours and cargo transport, whereas damped steady states enable signal transduction pathways. Here mechanical triggers cause temperature changes that lead to responses such as bending motions inspired by the single-touch mechanoresponse in Mimosa pudica and the frequency-gated snapping motion inspired by the plant arithmetic in the Venus flytrap. The proposed concepts suggest generalizable feedback pathways for dissipative dynamic materials and interactive soft robotics.
Collapse
Affiliation(s)
- Hang Zhang
- Department of Applied Physics, Aalto University, Espoo, Finland
| | - Hao Zeng
- Smart Photonic Materials, Faculty of Engineering and Natural Sciences, Tampere University, Tampere, Finland.
| | - Amanda Eklund
- Department of Applied Physics, Aalto University, Espoo, Finland
| | - Hongshuang Guo
- Smart Photonic Materials, Faculty of Engineering and Natural Sciences, Tampere University, Tampere, Finland
| | - Arri Priimagi
- Smart Photonic Materials, Faculty of Engineering and Natural Sciences, Tampere University, Tampere, Finland.
| | - Olli Ikkala
- Department of Applied Physics, Aalto University, Espoo, Finland.
| |
Collapse
|
50
|
Alshehri S, Pavlovič T, Farsinejad S, Behboodi P, Quan L, Centeno D, Kung D, Rezler M, Lee W, Jasiński P, Dziabaszewska E, Nowak-Markwitz E, Kalyon D, Zaborowski MP, Iwanicki M. Extracellular Matrix Modulates Outgrowth Dynamics in Ovarian Cancer. Adv Biol (Weinh) 2022; 6:e2200197. [PMID: 36084257 PMCID: PMC9772079 DOI: 10.1002/adbi.202200197] [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: 07/15/2022] [Revised: 08/18/2022] [Indexed: 01/28/2023]
Abstract
Ovarian carcinoma (OC) forms outgrowths that extend from the outer surface of an afflicted organ into the peritoneum. OC outgrowth formation is poorly understood due to the limited availability of cell culture models examining the behavior of cells that form outgrowths. Prompted by immunochemical evaluation of extracellular matrix (ECM) components in human tissues, laminin and collagen-rich ECM-reconstituted cell culture models amenable to studies of cell clusters that can form outgrowths are developed. It is demonstrated that ECM promotes outgrowth formation in fallopian tube non-ciliated epithelial cells (FNE) expressing mutant p53 and various OC cell lines. Outgrowths are initiated by cells that underwent outward translocation and retained the ability to intercalate into mesothelial cell monolayers. Electron microscopy, optical coherence tomography, and small amplitude oscillatory shear experiments reveal that increased ECM levels led to increased fibrous network thickness and high shear elasticity of the microenvironment. These physical characteristics are associated with outgrowth suppression. The low ECM microenvironment mimicks the viscoelasticity of malignant peritoneal fluid (ascites) and supports cell proliferation, cell translocation, and outgrowth formation. These results highlight the importance of the ECM microenvironment in modulating OC growth and can provide additional insights into the mode of dissemination of primary and recurrent ovarian tumors.
Collapse
Affiliation(s)
- Sarah Alshehri
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Tonja Pavlovič
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Sadaf Farsinejad
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Panteha Behboodi
- Department of Chemical Engineering, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Li Quan
- Department of Chemical Engineering, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Daniel Centeno
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Douglas Kung
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Marta Rezler
- Poznań University of Medical Sciences, Collegium Maius, Fredry 10, Poznań, 61-701, Poland
| | - Woo Lee
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
- Department of Chemical Engineering, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Piotr Jasiński
- Department of Pathology, Poznań University of Medical Sciences, Polna 33, Poznań, 60-535, Poland
| | - Elżbieta Dziabaszewska
- Department of Pathology, Poznań University of Medical Sciences, Polna 33, Poznań, 60-535, Poland
| | - Ewa Nowak-Markwitz
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Poznań University of Medical Sciences, Polna 33, Poznań, 60-535, Poland
| | - Dilhan Kalyon
- Department of Chemical Engineering, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Mikołaj P Zaborowski
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Poznań University of Medical Sciences, Polna 33, Poznań, 60-535, Poland
| | - Marcin Iwanicki
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| |
Collapse
|