1
|
Zhou EFM, Wong AYL, Zheng YP, Lam KHS, Fu SN. Reliability of Ultrasound Shear Wave Elastography for Evaluating Psoas Major and Quadratus Lumborum Stiffness: Gender and Physical Activity Effects. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:564-570. [PMID: 38272743 DOI: 10.1016/j.ultrasmedbio.2023.12.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/04/2023] [Accepted: 12/22/2023] [Indexed: 01/27/2024]
Abstract
OBJECTIVE We aimed to assess the reliability of quantifying psoas major (PM) and quadratus lumborum (QL) stiffness with ultrasound shear wave elastography (SWE), and to explore the effects of gender and physical activity on muscle stiffness. METHODS Fifty-two healthy participants (18-32 y) were recruited. To determine reliability, 29 of them underwent repeated SWE measurements of PM and QL stiffness by an operator on the same day. The intra-class correlation coefficients (ICC3,1), standard error of measurement (SEM) and minimal detectable change with 95% confidence interval (MDC95) were calculated. The rest participants underwent a single measurement. Two-way MANCOVA was conducted for the effects of gender and physical activity on muscle stiffness. RESULTS The observed reliability for PM (ICC3,1 = 0.89-0.92) and QL (ICC3,1 = 0.79-0.82) were good-to-excellent and good, respectively. The SEM (kPa) was 0.79-1.03 and 1.23-1.28, and the MDC95 (kPa) was 2.20-2.85 and 3.41-3.56 for PM and QL, respectively. After BMI adjustment, both gender (PM: F = 10.15, p = 0.003; QL: F = 18.07, p < 0.001) and activity level (PM: F = 5.90, p = 0.005; QL: F = 6.33, p = 0.004) influenced muscle stiffness. The female and inactive groups exhibited higher stiffness in both muscles. CONCLUSION SWE is reliable for quantifying the stiffness of PM and QL. Female and physical inactivity may elevate PM and QL stiffness, underscoring the importance of accounting for these factors in muscle stiffness investigations. Larger prospective studies are needed to further elucidate their effects.
Collapse
Affiliation(s)
- Emma Feng Ming Zhou
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, China
| | - Arnold Yu Lok Wong
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, China
| | - Yong Ping Zheng
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China; Research Institute for Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, China
| | - King Hei Stanley Lam
- The Hong Kong Institute of Musculoskeletal Medicine, Hong Kong, Hong Kong, China; Department of Family Medicine, The Chinese University of Hong Kong, Hong Kong, China; Department of Family Medicine, The University of Hong Kong, Hong Kong, China.
| | - Siu Ngor Fu
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, China
| |
Collapse
|
2
|
Nguyen J, Wang L, Lei W, Hu Y, Gulati N, Chavez-Madero C, Ahn H, Ginsberg HJ, Krawetz R, Brandt M, Betz T, Gilbert PM. Culture substrate stiffness impacts human myoblast contractility-dependent proliferation and nuclear envelope wrinkling. J Cell Sci 2024; 137:jcs261666. [PMID: 38345101 PMCID: PMC11033523 DOI: 10.1242/jcs.261666] [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: 09/27/2023] [Accepted: 02/04/2024] [Indexed: 03/28/2024] Open
Abstract
Understanding how biophysical and biochemical microenvironmental cues together influence the regenerative activities of muscle stem cells and their progeny is crucial in strategizing remedies for pathological dysregulation of these cues in aging and disease. In this study, we investigated the cell-level influences of extracellular matrix (ECM) ligands and culture substrate stiffness on primary human myoblast contractility and proliferation within 16 h of plating and found that tethered fibronectin led to stronger stiffness-dependent responses compared to laminin and collagen. A proteome-wide analysis further uncovered cell metabolism, cytoskeletal and nuclear component regulation distinctions between cells cultured on soft and stiff substrates. Interestingly, we found that softer substrates increased the incidence of myoblasts with a wrinkled nucleus, and that the extent of wrinkling could predict Ki67 (also known as MKI67) expression. Nuclear wrinkling and Ki67 expression could be controlled by pharmacological manipulation of cellular contractility, offering a potential cellular mechanism. These results provide new insights into the regulation of human myoblast stiffness-dependent contractility response by ECM ligands and highlight a link between myoblast contractility and proliferation.
Collapse
Affiliation(s)
- Jo Nguyen
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3E2, Canada
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Lu Wang
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Wen Lei
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Yechen Hu
- Department of Chemistry, University of Toronto, Toronto, ON, M5S 3H6, Canada
| | - Nitya Gulati
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3E2, Canada
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Carolina Chavez-Madero
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3E2, Canada
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Henry Ahn
- Department of Surgery, University of Toronto, Toronto, ON, M5G 2C4, Canada
- Li Ka Shing Knowledge Institute, Saint Michael's Hospital, Toronto, ON, M5B 1W8, Canada
| | - Howard J. Ginsberg
- Department of Surgery, University of Toronto, Toronto, ON, M5G 2C4, Canada
- Li Ka Shing Knowledge Institute, Saint Michael's Hospital, Toronto, ON, M5B 1W8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Roman Krawetz
- McCaig Institute, University of Calgary, Calgary, AB, T2N 4Z6, Canada
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Matthias Brandt
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University Münster, 48149 Münster, Germany
| | - Timo Betz
- Third Institute of Physics – Biophysics, Georg August University Göttingen, 37077 Göttingen, Germany
| | - Penney M. Gilbert
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3E2, Canada
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada
| |
Collapse
|
3
|
Nguyen J, Gilbert PM. Decoding the forces that shape muscle stem cell function. Curr Top Dev Biol 2024; 158:279-306. [PMID: 38670710 DOI: 10.1016/bs.ctdb.2024.02.009] [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: 04/28/2024]
Abstract
Skeletal muscle is a force-producing organ composed of muscle tissues, connective tissues, blood vessels, and nerves, all working in synergy to enable movement and provide support to the body. While robust biomechanical descriptions of skeletal muscle force production at the body or tissue level exist, little is known about force application on microstructures within the muscles, such as cells. Among various cell types, skeletal muscle stem cells reside in the muscle tissue environment and play a crucial role in driving the self-repair process when muscle damage occurs. Early evidence indicates that the fate and function of skeletal muscle stem cells are controlled by both biophysical and biochemical factors in their microenvironments, but much remains to accomplish in quantitatively describing the biophysical muscle stem cell microenvironment. This book chapter aims to review current knowledge on the influence of biophysical stresses and landscape properties on muscle stem cells in heath, aging, and diseases.
Collapse
Affiliation(s)
- Jo Nguyen
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Penney M Gilbert
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Donnelly Centre, University of Toronto, Toronto, ON, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
4
|
Li DCW, Rudloff S, Langer HT, Norman K, Herpich C. Age-Associated Differences in Recovery from Exercise-Induced Muscle Damage. Cells 2024; 13:255. [PMID: 38334647 PMCID: PMC10854791 DOI: 10.3390/cells13030255] [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: 12/31/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/10/2024] Open
Abstract
Understanding the intricate mechanisms governing the cellular response to resistance exercise is paramount for promoting healthy aging. This narrative review explored the age-related alterations in recovery from resistance exercise, focusing on the nuanced aspects of exercise-induced muscle damage in older adults. Due to the limited number of studies in older adults that attempt to delineate age differences in muscle discovery, we delve into the multifaceted cellular influences of chronic low-grade inflammation, modifications in the extracellular matrix, and the role of lipid mediators in shaping the recovery landscape in aging skeletal muscle. From our literature search, it is evident that aged muscle displays delayed, prolonged, and inefficient recovery. These changes can be attributed to anabolic resistance, the stiffening of the extracellular matrix, mitochondrial dysfunction, and unresolved inflammation as well as alterations in satellite cell function. Collectively, these age-related impairments may impact subsequent adaptations to resistance exercise. Insights gleaned from this exploration may inform targeted interventions aimed at enhancing the efficacy of resistance training programs tailored to the specific needs of older adults, ultimately fostering healthy aging and preserving functional independence.
Collapse
Affiliation(s)
- Donna Ching Wah Li
- Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany
| | - Stefan Rudloff
- Department of Geriatrics and Medical Gerontology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13347 Berlin, Germany
| | | | - Kristina Norman
- Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany
- Department of Geriatrics and Medical Gerontology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13347 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10785 Berlin, Germany
| | - Catrin Herpich
- Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558 Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany
- Department of Geriatrics and Medical Gerontology, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13347 Berlin, Germany
| |
Collapse
|
5
|
Nakajima T, Imai A, Ishii C, Tsuruyama K, Yamanaka R, Tomooka Y, Saito S, Adachi N, Kohno S, Sato T. SMAD2/3 signaling regulates initiation of mouse Wolffian ducts and proximal differentiation in Müllerian ducts. FEBS Open Bio 2024; 14:37-50. [PMID: 37953493 PMCID: PMC10761927 DOI: 10.1002/2211-5463.13729] [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: 09/03/2023] [Revised: 10/24/2023] [Accepted: 10/24/2023] [Indexed: 11/14/2023] Open
Abstract
Male and female reproductive tracts develop from anterior intermediate mesoderm with similar differentiation processes. The anterior intermediate mesoderm develops into the mesonephros, and the Wolffian duct initiates by epithelialization in the mesonephros. The Müllerian duct invaginates from the coelomic epithelium of the cranial mesonephros for ductal formation and is then regionalized into proximal to caudal female reproductive tracts. In this study, we focused on the epithelialization of the Wolffian duct, initiation of the Müllerian duct, and the regionalization step of the Müllerian ducts as a continuous process. By using intermediate mesodermal cells from mouse pluripotent stem cells, we identified that inhibition of SMAD2/3 signaling might be involved in the differentiation into mesenchymal cells, after which mesonephric cells might be then epithelialized during differentiation of the Wolffian duct. Aggregation of coelomic epithelial cells might be related to initiation of the Müllerian duct. Transcriptomic analysis predicted that consensus sequences of SMAD3/4 were enriched among highly expressed genes in the proximal Müllerian duct. SMAD2/3 signaling to regulate differentiation of the Wolffian duct was continuously activated in the proximal Müllerian duct and was involved in proximal and oviductal regionalization. Therefore, SMAD2/3 signaling may be finely tuned to regulate differentiation from initiation to regionalization steps.
Collapse
Affiliation(s)
- Tadaaki Nakajima
- Department of Biological Science and Technology, Faculty of Industrial Science and TechnologyTokyo University of ScienceJapan
- Department of ScienceYokohama City UniversityJapan
- Graduate School of NanobioscienceYokohama City UniversityJapan
| | - Akihiro Imai
- Department of Biological Science and Technology, Faculty of Industrial Science and TechnologyTokyo University of ScienceJapan
| | - Chihiro Ishii
- Department of Biological Science and Technology, Faculty of Industrial Science and TechnologyTokyo University of ScienceJapan
| | - Kota Tsuruyama
- Department of Biological Science and Technology, Faculty of Industrial Science and TechnologyTokyo University of ScienceJapan
| | - Risa Yamanaka
- Department of Biological Science and Technology, Faculty of Industrial Science and TechnologyTokyo University of ScienceJapan
| | - Yasuhiro Tomooka
- Department of Biological Science and Technology, Faculty of Industrial Science and TechnologyTokyo University of ScienceJapan
| | - Shinta Saito
- Department of ScienceYokohama City UniversityJapan
- Graduate School of NanobioscienceYokohama City UniversityJapan
| | - Noritaka Adachi
- Department of ScienceYokohama City UniversityJapan
- Graduate School of NanobioscienceYokohama City UniversityJapan
| | - Satomi Kohno
- Department of Biological SciencesSt. Cloud State UniversityMNUSA
| | - Tomomi Sato
- Department of ScienceYokohama City UniversityJapan
- Graduate School of NanobioscienceYokohama City UniversityJapan
| |
Collapse
|
6
|
Krajnik A, Nimmer E, Brazzo JA, Biber JC, Drewes R, Tumenbayar BI, Sullivan A, Pham K, Krug A, Heo Y, Kolega J, Heo SJ, Lee K, Weil BR, Kim DH, Gupte SA, Bae Y. Survivin regulates intracellular stiffness and extracellular matrix production in vascular smooth muscle cells. APL Bioeng 2023; 7:046104. [PMID: 37868708 PMCID: PMC10590228 DOI: 10.1063/5.0157549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/05/2023] [Indexed: 10/24/2023] Open
Abstract
Vascular dysfunction is a common cause of cardiovascular diseases characterized by the narrowing and stiffening of arteries, such as atherosclerosis, restenosis, and hypertension. Arterial narrowing results from the aberrant proliferation of vascular smooth muscle cells (VSMCs) and their increased synthesis and deposition of extracellular matrix (ECM) proteins. These, in turn, are modulated by arterial stiffness, but the mechanism for this is not fully understood. We found that survivin is an important regulator of stiffness-mediated ECM synthesis and intracellular stiffness in VSMCs. Whole-transcriptome analysis and cell culture experiments showed that survivin expression is upregulated in injured femoral arteries in mice and in human VSMCs cultured on stiff fibronectin-coated hydrogels. Suppressed expression of survivin in human VSMCs significantly decreased the stiffness-mediated expression of ECM components related to arterial stiffening, such as collagen-I, fibronectin, and lysyl oxidase. By contrast, expression of these ECM proteins was rescued by ectopic expression of survivin in human VSMCs cultured on soft hydrogels. Interestingly, atomic force microscopy analysis showed that suppressed or ectopic expression of survivin decreases or increases intracellular stiffness, respectively. Furthermore, we observed that inhibiting Rac and Rho reduces survivin expression, elucidating a mechanical pathway connecting intracellular tension, mediated by Rac and Rho, to survivin induction. Finally, we found that survivin inhibition decreases FAK phosphorylation, indicating that survivin-dependent intracellular tension feeds back to maintain signaling through FAK. These findings suggest a novel mechanism by which survivin potentially modulates arterial stiffness.
Collapse
Affiliation(s)
- Amanda Krajnik
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Erik Nimmer
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, Buffalo, New York 14260, USA
| | - Joseph A. Brazzo
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - John C. Biber
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Rhonda Drewes
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Bat-Ider Tumenbayar
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Andra Sullivan
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, Buffalo, New York 14260, USA
| | - Khanh Pham
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Alanna Krug
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, Buffalo, New York 14260, USA
| | | | - John Kolega
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Su-Jin Heo
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | - Brian R. Weil
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | - Sachin A. Gupte
- Department of Pharmacology, New York Medical College, Valhalla, New York 10595, USA
| | - Yongho Bae
- Author to whom correspondence should be addressed:
| |
Collapse
|
7
|
Ohashi A, Terai S, Furukawa S, Yamamoto S, Kashimoto R, Satoh A. Tenascin-C-enriched regeneration-specific extracellular matrix guarantees superior muscle regeneration in Ambystoma mexicanum. Dev Biol 2023; 504:98-112. [PMID: 37778717 DOI: 10.1016/j.ydbio.2023.09.012] [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: 03/20/2023] [Revised: 09/11/2023] [Accepted: 09/28/2023] [Indexed: 10/03/2023]
Abstract
Severe muscle injury causes distress and difficulty in humans. Studying the high regenerative ability of the axolotls may provide hints for the development of an effective treatment for severe injuries to muscle tissue. Here, we examined the regenerative process in response to a muscle injury in axolotls. We found that axolotls are capable of complete regeneration in response to a partial muscle resection called volumetric muscle loss (VML), which mammals cannot perfectly regenerate. We investigated the mechanisms underlying this high regenerative capacity in response to VML, focusing on the migration of muscle satellite cells and the extracellular matrix (ECM) formed during VML injury. Axolotls form tenascin-C (TN-C)-enriched ECM after VML injury. This TN-C-enriched ECM promotes the satellite cell migration. We confirmed the importance of TN-C in successful axolotl muscle regeneration by creating TN-C mutant animals. Our results suggest that the maintenance of a TN-C-enriched ECM environment after muscle injury promotes the release of muscle satellite cells and supports eventually high muscle regenerative capacity. In the future, better muscle regeneration may be achieved in mammals through the maintenance of TN-C expression.
Collapse
Affiliation(s)
- Ayaka Ohashi
- Graduate School of Environment, Life, Natural Science and Technology, Okayama University, Japan
| | - Suzuno Terai
- Okayama University, Faculty of Science, Department of Biological Sciences, Okayama, Japan
| | - Saya Furukawa
- Graduate School of Environment, Life, Natural Science and Technology, Okayama University, Japan
| | - Sakiya Yamamoto
- Graduate School of Environment, Life, Natural Science and Technology, Okayama University, Japan
| | - Rena Kashimoto
- Graduate School of Environment, Life, Natural Science and Technology, Okayama University, Japan
| | - Akira Satoh
- Graduate School of Environment, Life, Natural Science and Technology, Okayama University, Japan; Research Core for Interdisciplinary Sciences (RCIS), Okayama University, Okayama, Japan.
| |
Collapse
|
8
|
Szabó L, Telek A, Fodor J, Dobrosi N, Dócs K, Hegyi Z, Gönczi M, Csernoch L, Dienes B. Reduced Expression of Septin7 Hinders Skeletal Muscle Regeneration. Int J Mol Sci 2023; 24:13536. [PMID: 37686339 PMCID: PMC10487768 DOI: 10.3390/ijms241713536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/28/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
Septins are considered the fourth component of the cytoskeleton with the septin7 isoform playing a critical role in the formation of diffusion barriers in phospholipid bilayers and intra- and extracellular scaffolds. While its importance has already been confirmed in different intracellular processes, very little is known about its role in skeletal muscle. Muscle regeneration was studied in a Sept7 conditional knock-down mouse model to prove the possible role of septin7 in this process. Sterile inflammation in skeletal muscle was induced which was followed by regeneration resulting in the upregulation of septin7 expression. Partial knock-down of Sept7 resulted in an increased number of inflammatory cells and myofibers containing central nuclei. Taken together, our data suggest that partial knock-down of Sept7 hinders the kinetics of muscle regeneration, indicating its crucial role in skeletal muscle functions.
Collapse
Affiliation(s)
- László Szabó
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Doctoral School of Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Cell Physiology Research Group, University of Debrecen, 4032 Debrecen, Hungary
| | - Andrea Telek
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - János Fodor
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Nóra Dobrosi
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Cell Physiology Research Group, University of Debrecen, 4032 Debrecen, Hungary
| | - Klaudia Dócs
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Zoltán Hegyi
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Mónika Gönczi
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Cell Physiology Research Group, University of Debrecen, 4032 Debrecen, Hungary
| | - László Csernoch
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Cell Physiology Research Group, University of Debrecen, 4032 Debrecen, Hungary
| | - Beatrix Dienes
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
9
|
Özdemir C, Akçay D, Yöyen-Ermiş D, Taşkıran EZ, Soylu-Kucharz R, Esendağlı G, Kocaefe YÇ. Pro-fibrogenic and adipogenic aspects of chronic muscle degeneration are contributed by distinct stromal cell subpopulations. PLoS One 2023; 18:e0288800. [PMID: 37463149 DOI: 10.1371/journal.pone.0288800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 07/03/2023] [Indexed: 07/20/2023] Open
Abstract
Chronic skeletal muscle degeneration is characterized by fiber atrophy accompanied by deposition of extracellular matrix (ECM) components and fatty infiltration. Excessive accumulation of ECM leads to fibrosis via the contribution of fibro-adipogenic precursors (FAPs). Fibrosis also accompanies disuse atrophy and sarcopenia without significant inflammation. The present study aimed to comparatively analyze heterogeneous population of FAPs during acute injury and immobilization (tenotomy and denervation). The comparative analysis was accomplished based on the following 3 stromal cell subpopulations: i) CD140a(+)/Sca1(+); ii) CD140a(+)/Sca1(-); iii) CD140a(-)/Sca1(+). RNASeq analysis was employed to characterize and compare their quiescent and activated states. Whereas CD140a(-)/Sca1(+) was the most predominant activated subpopulation in tenotomy, denervation stimulated the CD140a(+)/Sca1(+) subpopulation. Immobilization models lacked myofiber damage and exhibited a minute increase in CD45(+) cells, as compared to acute injury. Transcriptome analysis showed common and discordant regulation of ECM components, without profound proliferative activation. Herein, we suggest unique surface markers for further identification of the investigated cell subpopulations. FAP subpopulations show similar activation kinetics in an inflammatory environment but the present findings highlight the fact that inflammation may not be a prerequisite for FAP activation. Delayed proliferation kinetics indicate that signals beyond inflammation might trigger FAP activation, leading to irreversible stromal changes.
Collapse
Affiliation(s)
- Cansu Özdemir
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Hacettepe University, Ankara, Turkey
- Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey
| | - Duygu Akçay
- Department of Medical Biology, School of Medicine, Hacettepe University, Ankara, Turkey
| | - Diğdem Yöyen-Ermiş
- Department of Basic Oncology, Cancer Institute, Hacettepe University, Ankara, Turkey
- Department of Immunology, School of Medicine, Uludağ University, Bursa, Turkey
| | - Ekim Zihni Taşkıran
- Department of Medical Genetics, School of Medicine, Hacettepe University, Ankara, Turkey
| | - Rana Soylu-Kucharz
- Department of Medical Biology, School of Medicine, Hacettepe University, Ankara, Turkey
| | - Güneş Esendağlı
- Department of Basic Oncology, Cancer Institute, Hacettepe University, Ankara, Turkey
| | - Yusuf Çetin Kocaefe
- Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey
- Department of Medical Biology, School of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
10
|
Kodama Y, Masuda S, Ohmori T, Kanamaru A, Tanaka M, Sakaguchi T, Nakagawa M. Response to Mechanical Properties and Physiological Challenges of Fascia: Diagnosis and Rehabilitative Therapeutic Intervention for Myofascial System Disorders. Bioengineering (Basel) 2023; 10:bioengineering10040474. [PMID: 37106661 PMCID: PMC10135675 DOI: 10.3390/bioengineering10040474] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Damage to the fascia can cause significant performance deficits in high-performance sports and recreational exercise and may contribute to the development of musculoskeletal disorders and persistent potential pain. The fascia is widely distributed from head to toe, encompassing muscles, bones, blood vessels, nerves, and internal organs and comprising various layers of different depths, indicating the complexity of its pathogenesis. It is a connective tissue composed of irregularly arranged collagen fibers, distinctly different from the regularly arranged collagen fibers found in tendons, ligaments, or periosteum, and mechanical changes in the fascia (stiffness or tension) can produce changes in its connective tissue that can cause pain. While these mechanical changes induce inflammation associated with mechanical loading, they are also affected by biochemical influences such as aging, sex hormones, and obesity. Therefore, this paper will review the current state of knowledge on the molecular level response to the mechanical properties of the fascia and its response to other physiological challenges, including mechanical changes, innervation, injury, and aging; imaging techniques available to study the fascial system; and therapeutic interventions targeting fascial tissue in sports medicine. This article aims to summarize contemporary views.
Collapse
Affiliation(s)
- Yuya Kodama
- Department of Orthopaedic Surgery, Okayama Rosai Hospital, 1-10-25 Midorimachi, Minamiku, Okayama 702-8055, Japan
| | - Shin Masuda
- Department of Orthopaedic Surgery, Okayama Rosai Hospital, 1-10-25 Midorimachi, Minamiku, Okayama 702-8055, Japan
| | - Toshinori Ohmori
- Department of Orthopaedic Surgery, Okayama Rosai Hospital, 1-10-25 Midorimachi, Minamiku, Okayama 702-8055, Japan
| | - Akihiro Kanamaru
- Department of Orthopaedic Surgery, Okayama Rosai Hospital, 1-10-25 Midorimachi, Minamiku, Okayama 702-8055, Japan
| | - Masato Tanaka
- Department of Orthopaedic Surgery, Okayama Rosai Hospital, 1-10-25 Midorimachi, Minamiku, Okayama 702-8055, Japan
| | - Tomoyoshi Sakaguchi
- Department of Central Rehabilitation, Okayama Rosai Hospital, 1-10-25 Midorimachi, Minamiku, Okayama 702-8055, Japan
| | - Masami Nakagawa
- Department of Central Rehabilitation, Okayama Rosai Hospital, 1-10-25 Midorimachi, Minamiku, Okayama 702-8055, Japan
| |
Collapse
|
11
|
Fede C, Fan C, Pirri C, Petrelli L, Biz C, Porzionato A, Macchi V, De Caro R, Stecco C. The Effects of Aging on the Intramuscular Connective Tissue. Int J Mol Sci 2022; 23:ijms231911061. [PMID: 36232366 PMCID: PMC9569538 DOI: 10.3390/ijms231911061] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/01/2022] [Accepted: 09/18/2022] [Indexed: 11/24/2022] Open
Abstract
The intramuscular connective tissue plays a critical role in maintaining the structural integrity of the muscle and in providing mechanical support. The current study investigates age-related changes that may contribute to passive stiffness and functional impairment of skeletal muscles. Variations in the extracellular matrix in human quadriceps femoris muscles in 10 young men, 12 elderly males and 16 elderly females, and in the hindlimb muscles of 6 week old, 8 month old and 2 year old C57BL/6J male mice, were evaluated. Picrosirius red, Alcian blue and Weigert Van Gieson stainings were performed to evaluate collagen, glycosamynoglycans and elastic fibers. Immunohistochemistry analyses were carried out to assess collagen I, collagen III and hyaluronan. The percentage area of collagen was significantly higher with aging (p < 0.01 in humans, p < 0.001 in mice), mainly due to an increase in collagen I, with no differences in collagen III (p > 0.05). The percentage area of elastic fibers in the perimysium was significantly lower (p < 0.01) in elderly men, together with a significant decrease in hyaluronan content both in humans and in mice. No significant differences were detected according to gender. The accumulation of collagen I and the lower levels of hyaluronan and elastic fibers with aging could cause a stiffening of the muscles and a reduction of their adaptability.
Collapse
Affiliation(s)
- Caterina Fede
- Department of Neurosciences, Institute of Human Anatomy, University of Padua, 35121 Padua, Italy
- Correspondence:
| | - Chenglei Fan
- Department of Neurosciences, Institute of Human Anatomy, University of Padua, 35121 Padua, Italy
| | - Carmelo Pirri
- Department of Neurosciences, Institute of Human Anatomy, University of Padua, 35121 Padua, Italy
| | - Lucia Petrelli
- Department of Neurosciences, Institute of Human Anatomy, University of Padua, 35121 Padua, Italy
| | - Carlo Biz
- Department of Surgery, Oncology and Gastroenterology, Orthopedic Clinic, University of Padua, 35128 Padua, Italy
| | - Andrea Porzionato
- Department of Neurosciences, Institute of Human Anatomy, University of Padua, 35121 Padua, Italy
| | - Veronica Macchi
- Department of Neurosciences, Institute of Human Anatomy, University of Padua, 35121 Padua, Italy
| | - Raffaele De Caro
- Department of Neurosciences, Institute of Human Anatomy, University of Padua, 35121 Padua, Italy
| | - Carla Stecco
- Department of Neurosciences, Institute of Human Anatomy, University of Padua, 35121 Padua, Italy
| |
Collapse
|
12
|
Ngai D, Mohabeer AL, Mao A, Lino M, Bendeck MP. Stiffness-Responsive Feedback Autoregulation of DDR1 Expression is Mediated by a DDR1-YAP/TAZ Axis. Matrix Biol 2022; 110:129-140. [PMID: 35562016 DOI: 10.1016/j.matbio.2022.05.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 05/02/2022] [Accepted: 05/05/2022] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Increased matrix stiffness is sensed by the collagen-binding receptor tyrosine kinase discoidin domain receptor 1 (DDR1). We have previously shown that DDR1 stimulates a positive feedback loop to increase its own expression in vascular smooth muscle cells (VSMCs). The transcriptional co-factors YAP/TAZ are stiffness sensing molecules that have not previously been investigated in DDR1 signaling. Here, we test the hypothesis that DDR1 signals through YAP/TAZ to auto-regulate its own expression. APPROACH AND RESULTS We used vascular smooth muscle cells (VSMCs) from wild-type and DDR1 knockout mice stimulated with collagen and/or substrates of different stiffness. We show that DDR1 controls YAP/TAZ nuclear localization and activity, whereas knockdown of YAP/TAZ attenuates DDR1 expression. In response to increased substrate stiffness, collagen stimulation, or RhoA activation, YAP/TAZ translocate to the nucleus and bind to chromatin. Finally, collagen stimulation promotes increased YAP/TAZ association with the Ddr1 promoter. CONCLUSIONS These findings reveal the mechanism by which DDR1 regulates YAP/TAZ activity which can then mediate positive feedback regulation of DDR1 expression by promoting transcription of the DDR1 gene.
Collapse
Affiliation(s)
- David Ngai
- Department of Laboratory Medicine and Pathobiology, University of Toronto; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto
| | - Amanda L Mohabeer
- Department of Laboratory Medicine and Pathobiology, University of Toronto; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto
| | - Amanda Mao
- Department of Laboratory Medicine and Pathobiology, University of Toronto; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto
| | - Marsel Lino
- Department of Laboratory Medicine and Pathobiology, University of Toronto; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto
| | - Michelle P Bendeck
- Department of Laboratory Medicine and Pathobiology, University of Toronto; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto.
| |
Collapse
|
13
|
(-) - Epicatechin improves Tibialis anterior muscle repair in CD1 mice with BaCl2-induced damage. J Nutr Biochem 2022; 107:109069. [DOI: 10.1016/j.jnutbio.2022.109069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 02/05/2022] [Accepted: 04/26/2022] [Indexed: 11/23/2022]
|
14
|
Geyer M, Schönfeld C, Schreiyäck C, Susanto S, Michel C, Looso M, Braun T, Borchardt T, Neumann E, Müller-Ladner U. Comparative transcriptional profiling of regenerating damaged knee joints in two animal models of the newt Notophthalmus viridescens strengthens the role of candidate genes involved in osteoarthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2022; 4:100273. [DOI: 10.1016/j.ocarto.2022.100273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 10/18/2022] Open
|
15
|
Seifert AW, Temple-Smith P. A remarkable rodent: Regeneration and reproduction in spiny mice (Acomys). Curr Top Dev Biol 2022; 147:659-707. [PMID: 35337466 DOI: 10.1016/bs.ctdb.2021.12.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Although certain organisms are chosen and employed to better understand a specific problem in biology (so-called model organisms), sometimes an animal model reveals its' biomedical importance by happenstance. In many ways, the advent of spiny mice (Acomys) as an emerging model to study regeneration and menstruation stands as a case study in scientific pseudoserendipity (Diaz de Chumaceiro, 1995). As we recount in this chapter, the discovery of these phenotypes, while not entirely accidental, was nonetheless unexpected. In addition to recounting how we uncovered these unusual mammalian traits, we outline recent work by our groups and others that has begun to outline the cellular and genetic mechanisms underlying bonafide mammalian tissue regeneration and a human-like mode of reproduction in spiny mice.
Collapse
Affiliation(s)
- Ashley W Seifert
- Department of Biology, University of Kentucky, Lexington, KY, United States; Department of Veterinary Anatomy and Physiology, University of Nairobi, Nairobi, Kenya.
| | - Peter Temple-Smith
- Department of Obstetrics & Gynecology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
16
|
Preclinical Development of Bioengineered Allografts Derived from Decellularized Human Diaphragm. Biomedicines 2022; 10:biomedicines10040739. [PMID: 35453490 PMCID: PMC9031975 DOI: 10.3390/biomedicines10040739] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/01/2022] [Accepted: 03/17/2022] [Indexed: 11/16/2022] Open
Abstract
Volumetric muscle loss (VML) is the traumatic/surgical loss of skeletal muscle, causing aesthetic damage and functional impairment. Suboptimal current surgical treatments are driving research towards the development of optimised regenerative therapies. The grafting of bioengineered scaffolds derived from decellularized skeletal muscle may be a valid option to promote structural and functional healing. In this work, a cellular human diaphragm was considered as a scaffold material for VML treatment. Decellularization occurred through four detergent-enzymatic protocols involving (1) sodium dodecyl sulfate (SDS), (2) SDS + TergitolTM, (3) sodium deoxycholate, and (4) TergitolTM. After decellularization, cells, DNA (≤50 ng/mg of tissue), and muscle fibres were efficiently removed, with the preservation of collagen/elastin and 60%–70% of the glycosaminoglycan component. The detergent-enzymatic treatments did not affect the expression of specific extracellular matrix markers (Collagen I and IV, Laminin), while causing the loss of HLA-DR expression to produce non-immunogenic grafts. Adipose-derived stem cells grown by indirect co-culture with decellularized samples maintained 80%–90% viability, demonstrating the biosafety of the scaffolds. Overall, the tested protocols were quite equivalent, with the patches treated by SDS + TergitolTM showing better collagen preservation. After subcutaneous implant in Balb/c mice, these acellular diaphragmatic grafts did not elicit a severe immune reaction, integrating with the host tissue.
Collapse
|
17
|
Regulation of Epicardial Cell Fate during Cardiac Development and Disease: An Overview. Int J Mol Sci 2022; 23:ijms23063220. [PMID: 35328640 PMCID: PMC8950551 DOI: 10.3390/ijms23063220] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 01/27/2023] Open
Abstract
The epicardium is the outermost cell layer in the vertebrate heart that originates during development from mesothelial precursors located in the proepicardium and septum transversum. The epicardial layer plays a key role during cardiogenesis since a subset of epicardial-derived cells (EPDCs) undergo an epithelial–mesenchymal transition (EMT); migrate into the myocardium; and differentiate into distinct cell types, such as coronary vascular smooth muscle cells, cardiac fibroblasts, endothelial cells, and presumably a subpopulation of cardiomyocytes, thus contributing to complete heart formation. Furthermore, the epicardium is a source of paracrine factors that support cardiac growth at the last stages of cardiogenesis. Although several lineage trace studies have provided some evidence about epicardial cell fate determination, the molecular mechanisms underlying epicardial cell heterogeneity remain not fully understood. Interestingly, seminal works during the last decade have pointed out that the adult epicardium is reactivated after heart damage, re-expressing some embryonic genes and contributing to cardiac remodeling. Therefore, the epicardium has been proposed as a potential target in the treatment of cardiovascular disease. In this review, we summarize the previous knowledge regarding the regulation of epicardial cell contribution during development and the control of epicardial reactivation in cardiac repair after damage.
Collapse
|
18
|
Growth and mechanobiology of the tendon-bone enthesis. Semin Cell Dev Biol 2022; 123:64-73. [PMID: 34362655 PMCID: PMC8810906 DOI: 10.1016/j.semcdb.2021.07.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/17/2021] [Accepted: 07/20/2021] [Indexed: 12/15/2022]
Abstract
Tendons are cable-like connective tissues that transfer both active and passive forces generated by skeletal muscle to bone. In the mature skeleton, the tendon-bone enthesis is an interfacial zone of transitional tissue located between two mechanically dissimilar tissues: compliant, fibrous tendon to rigid, dense mineralized bone. In this review, we focus on emerging areas in enthesis development related to its structure, function, and mechanobiology, as well as highlight established and emerging signaling pathways and physiological processes that influence the formation and adaptation of this important transitional tissue.
Collapse
|
19
|
Guan L, Fan P, Liu X, Liu R, Liu Y, Bai H. Migration of Human Renal Tubular Epithelial Cells in Response to Physiological Electric Signals. Front Cell Dev Biol 2021; 9:724012. [PMID: 34595174 PMCID: PMC8476913 DOI: 10.3389/fcell.2021.724012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/27/2021] [Indexed: 02/05/2023] Open
Abstract
Restoration of proximal tubular cell integrity and function after ischemic injury involves cell migration and proliferation. Endogenous fields are present during embryonic development and wound healing. Electric field (EF)-induced effects on cell migration have been observed in many cell types. This study investigated the effect of physiological direct current EF (dc EF) on the motility of renal epithelial cells. Human renal tubular epithelial (HK-2) and human-derived renal epithelial (HEK-293) cells were exposed to dc EF at physiological magnitude. Cell images were recorded and analyzed using an image analyzer. Cell lysates were used to detect protein expression by western blot. Scratch wounds were created in monolayers of HK-2 cells, and wound areas of cells were measured in response to EF exposure. Cells migrated significantly faster in the presence of an EF and toward the cathode. Application of an EF led to activation of the Erk1/2, p38 MAPK, and Akt signaling pathways. Pharmacological inhibition of Erk1/2, p38 MAPK, and Akt impaired EF-induced migratory responses, such as motility rate and directedness. In addition, exposure of the monolayers to EF enhanced EF-induced HK-2 wound healing. Our results suggest that EFs augment the rate of single renal epithelium migration and induce cell cathodal migration through activation of Erk1/2, p38 MAPK, and Akt signaling. Moreover, exposure of the renal epithelium to EF facilitated closure of in vitro small wounds by enhancing cell migration.
Collapse
Affiliation(s)
- Linbo Guan
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Ping Fan
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xinghui Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Rui Liu
- Division of Peptides Related with Human Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Liu
- Department of Biochemistry and Molecular Biology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, China
| | - Huai Bai
- Laboratory of Genetic Disease and Perinatal Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Tonti OR, Larson H, Lipp SN, Luetkemeyer CM, Makam M, Vargas D, Wilcox SM, Calve S. Tissue-specific parameters for the design of ECM-mimetic biomaterials. Acta Biomater 2021; 132:83-102. [PMID: 33878474 PMCID: PMC8434955 DOI: 10.1016/j.actbio.2021.04.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 03/18/2021] [Accepted: 04/08/2021] [Indexed: 02/06/2023]
Abstract
The extracellular matrix (ECM) is a complex network of biomolecules that mechanically and biochemically directs cell behavior and is crucial for maintaining tissue function and health. The heterogeneous organization and composition of the ECM varies within and between tissue types, directing mechanics, aiding in cell-cell communication, and facilitating tissue assembly and reassembly during development, injury and disease. As technologies like 3D printing rapidly advance, researchers are better able to recapitulate in vivo tissue properties in vitro; however, tissue-specific variations in ECM composition and organization are not given enough consideration. This is in part due to a lack of information regarding how the ECM of many tissues varies in both homeostatic and diseased states. To address this gap, we describe the components and organization of the ECM, and provide examples for different tissues at various states of disease. While many aspects of ECM biology remain unknown, our goal is to highlight the complexity of various tissues and inspire engineers to incorporate unique components of the native ECM into in vitro platform design and fabrication. Ultimately, we anticipate that the use of biomaterials that incorporate key tissue-specific ECM will lead to in vitro models that better emulate human pathologies. STATEMENT OF SIGNIFICANCE: Biomaterial development primarily emphasizes the engineering of new materials and therapies at the expense of identifying key parameters of the tissue that is being emulated. This can be partially attributed to the difficulty in defining the 3D composition, organization, and mechanics of the ECM within different tissues and how these material properties vary as a function of homeostasis and disease. In this review, we highlight a range of tissues throughout the body and describe how ECM content, cell diversity, and mechanical properties change in diseased tissues and influence cellular behavior. Accurately mimicking the tissue of interest in vitro by using ECM specific to the appropriate state of homeostasis or pathology in vivo will yield results more translatable to humans.
Collapse
Affiliation(s)
- Olivia R Tonti
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Hannah Larson
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Sarah N Lipp
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Callan M Luetkemeyer
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Megan Makam
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Diego Vargas
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Sean M Wilcox
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Sarah Calve
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States.
| |
Collapse
|
21
|
Bassat E, Tanaka EM. The cellular and signaling dynamics of salamander limb regeneration. Curr Opin Cell Biol 2021; 73:117-123. [PMID: 34521022 DOI: 10.1016/j.ceb.2021.07.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/19/2021] [Accepted: 07/27/2021] [Indexed: 12/16/2022]
Abstract
Limb amputation in salamanders yields a wound response that ultimately leads to replacement of the missing part. This unique-among-tetrapod trait involves the migration and recruitment of multiple cell types including epithelium, immune cells, axonal growth cones, and connective tissue cells to build the blastema which contains the proliferating stem and progenitor cells to rebuild the limb tissues. A number of the signaling and cell biological events have been defined. They point to the intimate coordination of physical events such as osmotic pressure, cell migration, and cell-cell communication with changes in cell identity such as dedifferentiation into embryonic-like epithelial and mesenchymal cells.
Collapse
Affiliation(s)
- Elad Bassat
- Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Campus Vienna Biocenter, 1030, Vienna, Austria
| | - Elly M Tanaka
- Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Campus Vienna Biocenter, 1030, Vienna, Austria.
| |
Collapse
|
22
|
Extracellular matrix: an important regulator of cell functions and skeletal muscle development. Cell Biosci 2021; 11:65. [PMID: 33789727 PMCID: PMC8011170 DOI: 10.1186/s13578-021-00579-4] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 03/23/2021] [Indexed: 12/15/2022] Open
Abstract
Extracellular matrix (ECM) is a kind of connective tissue in the cell microenvironment, which is of great significance to tissue development. ECM in muscle fiber niche consists of three layers: the epimysium, the perimysium, and the endomysium (basal lamina). These three layers of connective tissue structure can not only maintain the morphology of skeletal muscle, but also play an important role in the physiological functions of muscle cells, such as the transmission of mechanical force, the regeneration of muscle fiber, and the formation of neuromuscular junction. In this paper, detailed discussions are made for the structure and key components of ECM in skeletal muscle tissue, the role of ECM in skeletal muscle development, and the application of ECM in biomedical engineering. This review will provide the reader with a comprehensive overview of ECM, as well as a comprehensive understanding of the structure, physiological function, and application of ECM in skeletal muscle tissue.
Collapse
|
23
|
Escalante T, Saravia-Otten P, Gastaldello S, Hernández R, Marín A, García G, García L, Estrada E, Rucavado A, Gutiérrez JM. Changes in basement membrane components in an experimental model of skeletal muscle degeneration and regeneration induced by snake venom and myotoxic phospholipase A 2. Toxicon 2021; 192:46-56. [PMID: 33460638 DOI: 10.1016/j.toxicon.2021.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/02/2020] [Accepted: 01/10/2021] [Indexed: 12/27/2022]
Abstract
Skeletal muscle regeneration is impaired after myonecrosis induced by viperid snake venoms, but the mechanisms behind such poor regenerative outcome are not fully understood. This study compared the changes in basement membrane (BM) components in mouse skeletal muscle in two different scenarios of muscle injury: (a) injection of Bothrops asper venom, as a model of poor regeneration, and (b) injection of a myotoxic fraction (Mtx) isolated from this venom, as a model of successful regeneration. The degradation and reposition of laminin, type IV collagen and fibronectin were assessed over time by a combination of immunohistochemistry, Western blot, and real time polymerase chain reaction. Both treatments induced degradation of laminin and type IV collagen in areas of muscle necrosis since day one, however, there were differences in the pattern of degradation and reposition of these proteins along time. Overall, Mtx induced a higher synthesis of fibronectin and higher degradation of laminin at intermediate time points, together with higher levels of transcripts for the chains of the three proteins. Instead, venom induced a higher degradation of laminin and type IV collagen at early time intervals, followed by a reduced recovery of type IV collagen by 15 days. These differences in extracellular matrix degradation and remodeling between the two models could be associated to the poor muscle regeneration after myonecrosis induced by B. asper venom.
Collapse
Affiliation(s)
- Teresa Escalante
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica.
| | - Patricia Saravia-Otten
- Departamento de Bioquímica, Facultad de Ciencias Químicas y Farmacia, Universidad de San Carlos de Guatemala, Guatemala
| | - Stefano Gastaldello
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Precision Medicine Research Center, School of Pharmacy, Binzhou Medical University, Laishan District, Guanhai Road 346, Yantai, Shandong Province, 264003, China
| | - Rosario Hernández
- Departamento de Bioquímica, Facultad de Ciencias Químicas y Farmacia, Universidad de San Carlos de Guatemala, Guatemala
| | - Alexa Marín
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Gabriela García
- Departamento de Bioquímica, Facultad de Ciencias Químicas y Farmacia, Universidad de San Carlos de Guatemala, Guatemala
| | - Lourdes García
- Departamento de Bioquímica, Facultad de Ciencias Químicas y Farmacia, Universidad de San Carlos de Guatemala, Guatemala
| | - Erick Estrada
- Departamento de Bioquímica, Facultad de Ciencias Químicas y Farmacia, Universidad de San Carlos de Guatemala, Guatemala
| | - Alexandra Rucavado
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - José María Gutiérrez
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica.
| |
Collapse
|
24
|
Ma SKY, Chan ASF, Rubab A, Chan WCW, Chan D. Extracellular Matrix and Cellular Plasticity in Musculoskeletal Development. Front Cell Dev Biol 2020; 8:781. [PMID: 32984311 PMCID: PMC7477050 DOI: 10.3389/fcell.2020.00781] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022] Open
Abstract
Cellular plasticity refers to the ability of cell fates to be reprogrammed given the proper signals, allowing for dedifferentiation or transdifferentiation into different cell fates. In vitro, this can be induced through direct activation of gene expression, however this process does not naturally occur in vivo. Instead, the microenvironment consisting of the extracellular matrix (ECM) and signaling factors, directs the signals presented to cells. Often the ECM is involved in regulating both biochemical and mechanical signals. In stem cell populations, this niche is necessary for maintenance and proper function of the stem cell pool. However, recent studies have demonstrated that differentiated or lineage restricted cells can exit their current state and transform into another state under different situations during development and regeneration. This may be achieved through (1) cells responding to a changing niche; (2) cells migrating and encountering a new niche; and (3) formation of a transitional niche followed by restoration of the homeostatic niche to sequentially guide cells along the regenerative process. This review focuses on examples in musculoskeletal biology, with the concept of ECM regulating cells and stem cells in development and regeneration, extending beyond the conventional concept of small population of progenitor cells, but under the right circumstances even “lineage-restricted” or differentiated cells can be reprogrammed to enter into a different fate.
Collapse
Affiliation(s)
- Sophia Ka Yan Ma
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | | | - Aqsa Rubab
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Wilson Cheuk Wing Chan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China.,Department of Orthopedics Surgery and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.,The University of Hong Kong Shenzhen Institute of Research and Innovation (HKU-SIRI), Shenzhen, China
| | - Danny Chan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China.,The University of Hong Kong Shenzhen Institute of Research and Innovation (HKU-SIRI), Shenzhen, China
| |
Collapse
|
25
|
Ngai D, Lino M, Rothenberg KE, Simmons CA, Fernandez-Gonzalez R, Bendeck MP. DDR1 (Discoidin Domain Receptor-1)-RhoA (Ras Homolog Family Member A) Axis Senses Matrix Stiffness to Promote Vascular Calcification. Arterioscler Thromb Vasc Biol 2020; 40:1763-1776. [PMID: 32493168 PMCID: PMC7310304 DOI: 10.1161/atvbaha.120.314697] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Supplemental Digital Content is available in the text. Objective: Vascular calcification is a pathology characterized by arterial mineralization, which is a common late-term complication of atherosclerosis that independently increases the risk of adverse cardiovascular events by fourfold. A major source of calcifying cells is transdifferentiating vascular smooth muscle cells (VSMCs). Previous studies showed that deletion of the collagen-binding receptor, DDR1 (discoidin domain receptor-1), attenuated VSMC calcification. Increased matrix stiffness drives osteogenesis, and DDR1 has been implicated in stiffness sensing in other cell types; however, the role of DDR1 as a mechanosensor in VSMCs has not been investigated. Here, we test the hypothesis that DDR1 senses increased matrix stiffness and promotes VSMC transdifferentiation and calcification. Approach and Results: Primary VSMCs isolated from Ddr1+/+ (wild-type) and Ddr1−/− (knockout) mice were studied on collagen-I–coated silicon substrates of varying stiffness, culturing in normal or calcifying medium. DDR1 expression and phosphorylation increased with increasing stiffness, as did in vitro calcification, nuclear localization of Runx2 (Runt-related transcription factor 2), and expression of other osteochondrocytic markers. By contrast, DDR1 deficient VSMCs were not responsive to stiffness and did not undergo transdifferentiation. DDR1 regulated stress fiber formation and RhoA (ras homolog family member A) activation through the RhoGEF (rho guanine nucleotide exchange factor), Vav2. Inhibition of actomyosin contractility reduced Runx2 activation and attenuated in vitro calcification in wild-type VSMCs. Finally, a novel positive feedforward loop was uncovered between DDR1 and actomyosin contractility, important in regulating DDR1 expression, clustering, and activation. Conclusions: This study provides mechanistic insights into DDR1 mechanosignaling and shows that DDR1 activity and actomyosin contractility are interdependent in mediating stiffness-dependent increases in VSMC calcification.
Collapse
Affiliation(s)
- David Ngai
- From the Department of Laboratory Medicine and Pathobiology (D.N., M.L., M.P.B.), University of Toronto, Canada.,Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research (D.N. M.L., K.E.R., C.A.S., R.F.-G., M.P.B.), University of Toronto, Canada
| | - Marsel Lino
- From the Department of Laboratory Medicine and Pathobiology (D.N., M.L., M.P.B.), University of Toronto, Canada.,Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research (D.N. M.L., K.E.R., C.A.S., R.F.-G., M.P.B.), University of Toronto, Canada
| | - Katheryn E Rothenberg
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research (D.N. M.L., K.E.R., C.A.S., R.F.-G., M.P.B.), University of Toronto, Canada.,Institute of Biomaterials and Biomedical Engineering (K.E.R., C.A.S., R.F.-G.), University of Toronto, Canada
| | - Craig A Simmons
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research (D.N. M.L., K.E.R., C.A.S., R.F.-G., M.P.B.), University of Toronto, Canada.,Institute of Biomaterials and Biomedical Engineering (K.E.R., C.A.S., R.F.-G.), University of Toronto, Canada.,Department of Mechanical and Industrial Engineering (C.A.S.), University of Toronto, Canada
| | - Rodrigo Fernandez-Gonzalez
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research (D.N. M.L., K.E.R., C.A.S., R.F.-G., M.P.B.), University of Toronto, Canada.,Institute of Biomaterials and Biomedical Engineering (K.E.R., C.A.S., R.F.-G.), University of Toronto, Canada.,Department of Cell and Systems Biology (R.F.-G.), University of Toronto, Canada
| | - Michelle P Bendeck
- From the Department of Laboratory Medicine and Pathobiology (D.N., M.L., M.P.B.), University of Toronto, Canada.,Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research (D.N. M.L., K.E.R., C.A.S., R.F.-G., M.P.B.), University of Toronto, Canada
| |
Collapse
|
26
|
McCreery KP, Calve S, Neu CP. Ontogeny informs regeneration: explant models to investigate the role of the extracellular matrix in cartilage tissue assembly and development. Connect Tissue Res 2020; 61:278-291. [PMID: 32186210 PMCID: PMC7190409 DOI: 10.1080/03008207.2019.1698556] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 11/22/2019] [Indexed: 02/03/2023]
Abstract
Osteoarthritis (OA) is typically managed in late stages by replacement of the articular cartilage surface with a prosthesis as an effective, though undesirable outcome. As an alternative, hydrogel implants or growth factor treatments are currently of great interest in the tissue engineering community, and scaffold materials are often designed to emulate the mechanical and chemical composition of mature extracellular matrix (ECM) tissue. However, scaffolds frequently fail to capture the structure and organization of cartilage. Additionally, many current scaffold designs do not mimic processes by which structurally sound cartilage is formed during musculoskeletal development. The objective of this review is to highlight methods that investigate cartilage ontogenesis with native and model systems in the context of regenerative medicine. Specific emphasis is placed on the use of cartilage explant cultures that provide a physiologically relevant microenvironment to study tissue assembly and development. Ex vivo cartilage has proven to be a cost-effective and accessible model system that allows researchers to control the culture conditions and stimuli and perform proteomics and imaging studies that are not easily possible using in vivo experiments, while preserving native cell-matrix interactions. We anticipate our review will promote a developmental biology approach using explanted tissues to guide cartilage tissue engineering and inform new treatment methods for OA and joint damage.
Collapse
Affiliation(s)
| | - Sarah Calve
- Department of Mechanical Engineering, University of Colorado, Boulder, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, USA
| | - Corey P. Neu
- Department of Mechanical Engineering, University of Colorado, Boulder, USA
| |
Collapse
|
27
|
Ireland RG, Kibschull M, Audet J, Ezzo M, Hinz B, Lye SJ, Simmons CA. Combinatorial extracellular matrix microarray identifies novel bioengineered substrates for xeno-free culture of human pluripotent stem cells. Biomaterials 2020; 248:120017. [PMID: 32283392 DOI: 10.1016/j.biomaterials.2020.120017] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 03/09/2020] [Accepted: 03/27/2020] [Indexed: 11/24/2022]
Abstract
Stem cells in their microenvironment are exposed to a plethora of biochemical signals and biophysical forces. Interrogating the role of each factor in the cell microenvironment, however, remains difficult due to the inability to study microenvironmental cues and tease apart their interactions in high throughput. To address this need, we developed an extracellular matrix (ECM) microarray screening platform capable of tightly controlling substrate stiffness and ECM protein composition to screen the effects of these cues and their interactions on cell fate. We combined this platform with a design of experiments screening strategy to identify optimal conditions that can maintain human pluripotent stem cell (hPSC) pluripotency in chemically defined, xeno-free conditions. Combinations of ECM proteins (fibronectin, vitronectin, laminin-521, and collagen IV) were deposited on polydimethylsiloxane substrates with elastic moduli ranging from ~1 to 60 kPa using a high throughput protein plotter. Through our screening approach, we identified several non-intuitive protein-protein and protein-stiffness interactions and developed three novel culture substrates. hPSCs grown on these novel culture substrates displayed higher proliferation rates and pluripotency marker expression than current gold-standard culture substrates Geltrex- and vitronectin-coated plastic. This ECM microarray and screening approach is not limited to the factors studied here and can be broadly applied to other cell types to systematically screen microenvironmental conditions to optimally guide cell phenotype.
Collapse
Affiliation(s)
- Ronald G Ireland
- Institute of Biomaterials & Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada
| | - Mark Kibschull
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Julie Audet
- Institute of Biomaterials & Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Maya Ezzo
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Boris Hinz
- Institute of Biomaterials & Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Stephen J Lye
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada; Departments of Obstetrics & Gynaecology, Physiology, and Medicine, University of Toronto, Toronto, ON, Canada; Alliance for Human Development, Sinai Health System, Toronto, ON, Canada
| | - Craig A Simmons
- Institute of Biomaterials & Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
28
|
Iberite F, Salerno M, Canale C, Rosa A, Ricotti L. Influence of substrate stiffness on human induced pluripotent stem cells: preliminary results. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2019:1039-1043. [PMID: 31946071 DOI: 10.1109/embc.2019.8857397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Skeletal muscle differentiation was proven to be influenced by changes in the substrate stiffness. However, a lack of knowledge features this field, concerning skeletal muscle tissues obtained from human induced pluripotent stem cells. Here we report the fabrication of polydimethylsiloxane-based substrates in a range of stiffness values from 3.5 to 141 kPa and the response of human induced pluripotent stem cells cultured on them for 5 days. The substrates were able to sustain cell adhesion and proliferation throughout the whole period. An inversely proportional relationship (although not significant) was found between the proliferation rate and the substrate stiffness. Initial analyses of iPSCs skeletal muscle differentiation shown no influences on markers of the early stages. These results lay the foundations for further studies on the influence of extrinsic mechanical stimuli on induced pluripotent stem cells-derived skeletal muscle tissues.
Collapse
|
29
|
Acuna A, Sofronici SH, Goergen CJ, Calve S. In situ measurement of native extracellular matrix strain. EXPERIMENTAL MECHANICS 2019; 59:1307-1321. [PMID: 31762478 PMCID: PMC6874413 DOI: 10.1007/s11340-019-00499-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 03/05/2019] [Indexed: 06/10/2023]
Abstract
Cells directly interact with the extracellular matrix (ECM) in their microenvironment; however, the mechanical properties of the networks at this scale are not well defined. This work describes a method to quantify ECM network strain in situ after the application of a known load. Visualization of the ECM in the native 3D organization is facilitated using murine embryos and a novel decellularization method. During embryonic development, the ECM architecture is less dense making it easier to visualize and manipulate. Briefly, embryonic day (E)14.5 forelimbs were harvested and incubated in an acrylamide-based hydrogel mixture to maintain the 3D architecture of the ECM during decellularization. After decellularization, forelimbs were stained for fibrillin-2 and proteoglycans to visualize different networks. Samples were imaged, before and after the application of a static load, using confocal microscopy. A MATLAB-based fast iterative digital volume correlation algorithm was used to quantify network displacement fields by comparing the reference and compressed z-stacks. We observed that the amount of Green-Lagrange strain experienced by different proteins was dependent on whether the sub-region analyzed was located within cartilage or the adjacent connective tissue. The combination of these experimental and computational methods will enable the development of constitutive equations that describe the material behavior of ECM networks. In the future, this information has the potential to improve the fabrication of physiologically relevant scaffolds by establishing mechanical guidelines for microenvironments that support beneficial cell-ECM interactions.
Collapse
Affiliation(s)
- A Acuna
- Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive, West Lafayette, IN 47907
| | - S H Sofronici
- Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive, West Lafayette, IN 47907
| | - C J Goergen
- Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive, West Lafayette, IN 47907
| | - S Calve
- Weldon School of Biomedical Engineering, Purdue University, 206 South Martin Jischke Drive, West Lafayette, IN 47907
| |
Collapse
|
30
|
Carmen L, Maria V, Morales-Medina JC, Vallelunga A, Palmieri B, Iannitti T. Role of proteoglycans and glycosaminoglycans in Duchenne muscular dystrophy. Glycobiology 2019; 29:110-123. [PMID: 29924302 DOI: 10.1093/glycob/cwy058] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 06/18/2018] [Indexed: 12/25/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an inherited fatal X-linked myogenic disorder with a prevalence of 1 in 3500 male live births. It affects voluntary muscles, and heart and breathing muscles. DMD is characterized by continuous degeneration and regeneration cycles resulting in extensive fibrosis and a progressive reduction in muscle mass. Since the identification of a reduction in dystrophin protein as the cause of this disorder, numerous innovative and experimental therapies, focusing on increasing the levels of dystrophin, have been proposed, but the clinical improvement has been unsatisfactory. Dystrophin forms the dystrophin-associated glycoprotein complex and its proteins have been studied as a promising novel therapeutic target to treat DMD. Among these proteins, cell surface glycosaminoglycans (GAGs) are found almost ubiquitously on the surface and in the extracellular matrix (ECM) of mammalian cells. These macromolecules interact with numerous ligands, including ECM constituents, adhesion molecules and growth factors that play a crucial role in muscle development and maintenance. In this article, we have reviewed in vitro, in vivo and clinical studies focused on the functional role of GAGs in the pathophysiology of DMD with the final aim of summarizing the state of the art of GAG dysregulation within the ECM in DMD and discussing future therapeutic perspectives.
Collapse
Affiliation(s)
- Laurino Carmen
- Department of General Surgery and Surgical Specialties, University of Modena and Reggio Emilia Medical School, Surgical Clinic, Modena, Italy
| | - Vadala' Maria
- Department of General Surgery and Surgical Specialties, University of Modena and Reggio Emilia Medical School, Surgical Clinic, Modena, Italy
| | - Julio Cesar Morales-Medina
- Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, CP, AP 62, Mexico
| | - Annamaria Vallelunga
- Department of Medicine and Surgery, Centre for Neurodegenerative Diseases (CEMAND), University of Salerno, Salerno, Italy
| | - Beniamino Palmieri
- Department of General Surgery and Surgical Specialties, University of Modena and Reggio Emilia Medical School, Surgical Clinic, Modena, Italy
| | | |
Collapse
|
31
|
Filip S, Mokrý J, Forostyak O, Dayanithi G. The extracellular matrix and Ca(2+)signaling mechanisms. Physiol Res 2019; 68:161-170. [DOI: 10.33549/physiolres.934081] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The extracellular matrix (ECM) consists of proteins, glycosaminoglycans and glycoproteins, that support the dynamic interactions between cells, including intercellular communication, cell attachment, cell differentiation, cell growth and migration. As such, the ECM represents an essential and very sensitive system within the tissue microenvironment that is involved in processes such as tissue regeneration and carcinogenesis. The aim of the present review is to evaluate its diversity through Ca(2+) signaling and its role in muscle cell function. Here, we discuss some methodological approaches dissecting Ca(2+) handling mechanisms in myogenic and non-myogenic cells, e.g. the importance of Ca(2+) and calpains in muscle dystrophy. We also consider the reconstruction of skeletal muscle by colonization of decellularized ECM with muscle-derived cells isolated from skeletal muscle. Therefore, it is necessary to establish new methodological procedures based on Ca(2+) signaling in skeletal muscle cells and their effect on ECM homeostasis, allowing the monitoring of skeletal muscle reconstruction and organ repair.
Collapse
Affiliation(s)
- S. Filip
- Charles University, Faculty of Medicine, Dept. of Oncology and Radiotherapy, Hradec Králové, Czech Republic.
| | | | | | | |
Collapse
|
32
|
Petrosino JM, Leask A, Accornero F. Genetic manipulation of CCN2/CTGF unveils cell-specific ECM-remodeling effects in injured skeletal muscle. FASEB J 2019; 33:2047-2057. [PMID: 30216109 PMCID: PMC6338641 DOI: 10.1096/fj.201800622rr] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 08/20/2018] [Indexed: 01/03/2023]
Abstract
In skeletal muscle, extracellular matrix (ECM) remodeling can either support the complete regeneration of injured muscle or facilitate pathologic fibrosis and muscle degeneration. Muscular dystrophy (MD) is a group of genetic disorders that results in a progressive decline in muscle function and is characterized by the abundant deposition of fibrotic tissue. Unlike acute injury, where ECM remodeling is acute and transient, in MD, remodeling persists until fibrosis obstructs the regenerative efforts of diseased muscles. Thus, understanding how ECM is deposited and organized is critical in the context of muscle repair. Connective tissue growth factor (CTGF or CCN2) is a matricellular protein expressed by multiple cell types in response to tissue injury. Although used as a general marker of fibrosis, the cell type-dependent role of CTGF in dystrophic muscle has not been elucidated. To address this question, a conditional Ctgf myofiber and fibroblast-knockout mouse lines were generated and crossed to a dystrophic background. Only myofiber-selective inhibition of CTGF protected δ-sarcoglycan-null ( Sgcd-/-) mice from the dystrophic phenotype, and it did so by affecting collagen organization in a way that allowed for improvements in dystrophic muscle regeneration and function. To confirm that muscle-specific CTGF functions to mediate collagen organization, we generated mice with transgenic muscle-specific overexpression of CTGF. Again, genetic modulation of CTGF in muscle was not sufficient to drive fibrosis, but altered collagen content and organization after injury. Our results show that the myofibers are critical mediators of the deleterious effects associated with CTGF in MD and acutely injured skeletal muscle.-Petrosino, J. M., Leask, A., Accornero, F. Genetic manipulation of CCN2/CTGF unveils cell-specific ECM-remodeling effects in injured skeletal muscle.
Collapse
Affiliation(s)
- Jennifer M. Petrosino
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| | - Andrew Leask
- Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Federica Accornero
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
33
|
Silva Garcia JM, Panitch A, Calve S. Functionalization of hyaluronic acid hydrogels with ECM-derived peptides to control myoblast behavior. Acta Biomater 2019; 84:169-179. [PMID: 30508655 DOI: 10.1016/j.actbio.2018.11.030] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/31/2018] [Accepted: 11/19/2018] [Indexed: 01/07/2023]
Abstract
Volumetric muscle loss (VML) occurs when skeletal muscle injury is too large for the body to fully self-repair. Typically, fibrotic tissue fills the void, which reduces muscle functionality and limb movement. Although a wide variety of natural and synthetic scaffolds have been studied with the purpose of providing the appropriate structural support, to date no scaffold has significantly restored muscle functionality after VML. Satellite cells, adult stem cells within the muscle capable of restoring smaller injuries, are sensitive to the stiffness and composition of the surrounding environment. Scaffolds that only address structural support are not sufficient to restore functionality and instead need to be designed to both promote satellite cell activation and prevent excessive fibroblast recruitment. The objective of this study was to design a scaffold that mimicked the regenerative environment and determine how the biomechanical properties differentially influence myogenic precursor and connective tissue cells. One of the main extracellular matrix (ECM) molecules upregulated during regeneration is hyaluronic acid (HA). Therefore, thiol-modified HA and poly(ethylene glycol) diacrylate hydrogels were generated and functionalized with peptides based on ECM known to influence regeneration, including fibronectin, laminin and tenascin-C. Scaffolds with different stiffness were created by varying HA content. The influence of HA stiffness and peptide functionalization on myogenic precursor and connective tissue cell proliferation, migration and gene expression was quantified. Our results indicated that HA hydrogels functionalized with the laminin peptide, IKVAV, show potential due to the enhanced promotion of myogenic cell behaviors including migration, proliferation and an increase in relevant transcription factors. STATEMENT OF SIGNIFICANCE: The goal of this study was to identify hyaluronic acid (HA) hydrogels with peptide and stiffness combinations that will direct muscle-derived cells towards regenerating phenotypes. While the interaction of skeletal muscle with RGD-functionalized HA hydrogels has been investigated, none of the other peptides described in this study had been used in the context of HA-based scaffolds and skeletal muscle-derived cells. Notably, the response of cells to variations in mechanics was dependent on ECM coating and lineage. The 3% HA functionalized with the laminin peptide, IKVAV, showed the most promise for future in vivo studies, as these hydrogels best promoted myoblast cell proliferation, attachment and spreading, enhanced migration over connective tissue cells and upregulated transcription factors associated with activated satellite cells.
Collapse
|
34
|
Zügel M, Maganaris CN, Wilke J, Jurkat-Rott K, Klingler W, Wearing SC, Findley T, Barbe MF, Steinacker JM, Vleeming A, Bloch W, Schleip R, Hodges PW. Fascial tissue research in sports medicine: from molecules to tissue adaptation, injury and diagnostics: consensus statement. Br J Sports Med 2018; 52:1497. [PMID: 30072398 PMCID: PMC6241620 DOI: 10.1136/bjsports-2018-099308] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2018] [Indexed: 01/10/2023]
Abstract
The fascial system builds a three-dimensional continuum of soft, collagen-containing, loose and dense fibrous connective tissue that permeates the body and enables all body systems to operate in an integrated manner. Injuries to the fascial system cause a significant loss of performance in recreational exercise as well as high-performance sports, and could have a potential role in the development and perpetuation of musculoskeletal disorders, including lower back pain. Fascial tissues deserve more detailed attention in the field of sports medicine. A better understanding of their adaptation dynamics to mechanical loading as well as to biochemical conditions promises valuable improvements in terms of injury prevention, athletic performance and sports-related rehabilitation. This consensus statement reflects the state of knowledge regarding the role of fascial tissues in the discipline of sports medicine. It aims to (1) provide an overview of the contemporary state of knowledge regarding the fascial system from the microlevel (molecular and cellular responses) to the macrolevel (mechanical properties), (2) summarise the responses of the fascial system to altered loading (physical exercise), to injury and other physiological challenges including ageing, (3) outline the methods available to study the fascial system, and (4) highlight the contemporary view of interventions that target fascial tissue in sport and exercise medicine. Advancing this field will require a coordinated effort of researchers and clinicians combining mechanobiology, exercise physiology and improved assessment technologies.
Collapse
Affiliation(s)
- Martina Zügel
- Division of Sports Medicine, Ulm University, Ulm, Germany
| | - Constantinos N Maganaris
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | - Jan Wilke
- Department of Sports Medicine, Goethe University, Frankfurt, Germany
| | | | - Werner Klingler
- Department of Anesthesiology, BKH Günzburg, Günzburg, Germany
| | - Scott C Wearing
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Thomas Findley
- Department of Physical Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Mary F Barbe
- Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Andry Vleeming
- Department of Rehabilitation Sciences and Physiotherapy, Faculty of Medicine and Health Sciences, Medical University Ghent, Ghent, Belgium
| | - Wilhelm Bloch
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne, Cologne, Germany
| | - Robert Schleip
- Fascia Research Group, Experimental Anesthesiology, Ulm University, Ulm, Germany
| | - Paul William Hodges
- Centre of Clinical Research Excellence in Spinal Pain, Injury and Health, School of Health and Rehabilitation Sciences, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
35
|
McClure MJ, Cohen DJ, Ramey AN, Bivens CB, Mallu S, Isaacs JE, Imming E, Huang YC, Sunwoo M, Schwartz Z, Boyan BD. Decellularized Muscle Supports New Muscle Fibers and Improves Function Following Volumetric Injury. Tissue Eng Part A 2018; 24:1228-1241. [DOI: 10.1089/ten.tea.2017.0386] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Michael J. McClure
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - David J. Cohen
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Allison N. Ramey
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Caroline B. Bivens
- Department of School of Art, Virginia Commonwealth University, Richmond, Virginia
| | - Satya Mallu
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, Virginia
| | - Jonathan E. Isaacs
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, Virginia
| | - Emily Imming
- MTF Biologics, Musculoskeletal Transplant Foundation, Edison, New Jersey
| | - Yen-Chen Huang
- MTF Biologics, Musculoskeletal Transplant Foundation, Edison, New Jersey
| | - MoonHae Sunwoo
- MTF Biologics, Musculoskeletal Transplant Foundation, Edison, New Jersey
| | - Zvi Schwartz
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Barbara D. Boyan
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|
36
|
The Roles of Matrix Stiffness and ß-Catenin Signaling in Endothelial-to-Mesenchymal Transition of Aortic Valve Endothelial Cells. Cardiovasc Eng Technol 2018; 9:158-167. [PMID: 29761409 DOI: 10.1007/s13239-018-0363-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 05/09/2018] [Indexed: 12/19/2022]
Abstract
Valve stiffening is a hallmark of aortic valve stenosis caused by excess extracellular matrix accumulation by myofibroblasts. We aimed to elucidate whether matrix stiffness regulates endothelial-to-mesenchymal transition (EndMT) of adult valvular endothelial cells (VECs) to myofibroblasts as a mechanism to further promote valve fibrosis. In addition, we specifically examined the role of the Wnt/β-catenin signaling pathway in the development of myofibroblasts during EndMT, as Wnt/β-catenin signaling has been implicated in EndMT during heart development, is reactivated in valve disease, and is required for mechanically-regulated myofibrogenesis of valve interstitial cells. Clonally derived porcine VECs were cultured on soft (5 kPa) or stiff (50 kPa) silicone Sylgard 527 substrates and treated with transforming growth factor (TGF)-β1 to induce EndMT. Immunofluorescent staining revealed that TGF-β1 preferentially promoted EndMT in VECs on stiffer substrates, evidenced by a decrease in the endothelial marker VE-cadherin and an increase in the myofibroblast marker α-smooth muscle actin (α-SMA). These changes were accompanied by β-catenin nuclear localization both in vitro and in vivo, assessed by immunostaining. Degradation of β-catenin with endostatin reduced VEC myofibroblast transition, as indicated by decreased α-SMA fiber expression. We conclude that TGF-β1-induced EndMT in aortic VECs is dependent on matrix stiffness and Wnt/β-catenin signaling promotes myofibrogenesis during EndMT.
Collapse
|
37
|
Abstract
The extracellular matrix (ECM) has central roles in tissue integrity and remodeling throughout the life span of animals. While collagens are the most abundant structural components of ECM in most tissues, tissue-specific molecular complexity is contributed by ECM glycoproteins. The matricellular glycoproteins are categorized primarily according to functional criteria and represented predominantly by the thrombospondin, tenascin, SPARC/osteonectin, and CCN families. These proteins do not self-assemble into ECM fibrils; nevertheless, they shape ECM properties through interactions with structural ECM proteins, growth factors, and cells. Matricellular proteins also promote cell migration or morphological changes through adhesion-modulating or counter-adhesive actions on cell-ECM adhesions, intracellular signaling, and the actin cytoskeleton. Typically, matricellular proteins are most highly expressed during embryonic development. In adult tissues, expression is more limited unless activated by cues for dynamic tissue remodeling and cell motility, such as occur during inflammatory response and wound repair. Many insights in the complex roles of matricellular proteins have been obtained from studies of gene knockout mice. However, with the exception of chordate-specific tenascins, these are highly conserved proteins that are encoded in many animal phyla. This review will consider the increasing body of research on matricellular proteins in nonmammalian animal models. These models provide better access to the very earliest stages of embryonic development and opportunities to study biological processes such as limb and organ regeneration. In aggregate, this research is expanding concepts of the functions and mechanisms of action of matricellular proteins.
Collapse
Affiliation(s)
- Josephine C Adams
- School of Biochemistry, University of Bristol, Bristol, United Kingdom.
| |
Collapse
|
38
|
The blastema and epimorphic regeneration in mammals. Dev Biol 2017; 433:190-199. [PMID: 29291973 DOI: 10.1016/j.ydbio.2017.08.007] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/28/2017] [Accepted: 08/04/2017] [Indexed: 01/02/2023]
Abstract
Studying regeneration in animals where and when it occurs is inherently interesting and a challenging research topic within developmental biology. Historically, vertebrate regeneration has been investigated in animals that display enhanced regenerative abilities and we have learned much from studying organ regeneration in amphibians and fish. From an applied perspective, while regeneration biologists will undoubtedly continue to study poikilothermic animals (i.e., amphibians and fish), studies focused on homeotherms (i.e., mammals and birds) are also necessary to advance regeneration biology. Emerging mammalian models of epimorphic regeneration are poised to help link regenerative biology and regenerative medicine. The regenerating rodent digit tip, which parallels human fingertip regeneration, and the regeneration of large circular defects through the ear pinna in spiny mice and rabbits, provide tractable, experimental systems where complex tissue structures are regrown through blastema formation and morphogenesis. Using these models as examples, we detail similarities and differences between the mammalian blastema and its classical counterpart to arrive at a broad working definition of a vertebrate regeneration blastema. This comparison leads us to conclude that regenerative failure is not related to the availability of regeneration-competent progenitor cells, but is most likely a function of the cellular response to the microenvironment that forms following traumatic injury. Recent studies demonstrating that targeted modification of this microenvironment can restrict or enhance regenerative capabilities in mammals helps provide a roadmap for eventually pushing the limits of human regeneration.
Collapse
|
39
|
Glucocorticoids Improve Myogenic Differentiation In Vitro by Suppressing the Synthesis of Versican, a Transitional Matrix Protein Overexpressed in Dystrophic Skeletal Muscles. Int J Mol Sci 2017; 18:ijms18122629. [PMID: 29211034 PMCID: PMC5751232 DOI: 10.3390/ijms18122629] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 11/24/2017] [Accepted: 11/27/2017] [Indexed: 12/17/2022] Open
Abstract
In Duchenne muscular dystrophy (DMD), a dysregulated extracellular matrix (ECM) directly exacerbates pathology. Glucocorticoids are beneficial therapeutics in DMD, and have pleiotropic effects on the composition and processing of ECM proteins in other biological contexts. The synthesis and remodelling of a transitional versican-rich matrix is necessary for myogenesis; whether glucocorticoids modulate this transitional matrix is not known. Here, versican expression and processing were examined in hindlimb and diaphragm muscles from mdx dystrophin-deficient mice and C57BL/10 wild type mice. V0/V1 versican (Vcan) mRNA transcripts and protein levels were upregulated in dystrophic compared to wild type muscles, especially in the more severely affected mdx diaphragm. Processed versican (versikine) was detected in wild type and dystrophic muscles, and immunoreactivity was highly associated with newly regenerated myofibres. Glucocorticoids enhanced C2C12 myoblast fusion by modulating the expression of genes regulating transitional matrix synthesis and processing. Specifically, Tgfβ1, Vcan and hyaluronan synthase-2 (Has2) mRNA transcripts were decreased by 50% and Adamts1 mRNA transcripts were increased three-fold by glucocorticoid treatment. The addition of exogenous versican impaired myoblast fusion, whilst glucocorticoids alleviated this inhibition in fusion. In dystrophic mdx muscles, versican upregulation correlated with pathology. We propose that versican is a novel and relevant target gene in DMD, given its suppression by glucocorticoids and that in excess it impairs myoblast fusion, a process key for muscle regeneration.
Collapse
|
40
|
Patra C, Boccaccini A, Engel F. Vascularisation for cardiac tissue engineering: the extracellular matrix. Thromb Haemost 2017; 113:532-47. [DOI: 10.1160/th14-05-0480] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 09/03/2014] [Indexed: 02/07/2023]
Abstract
SummaryCardiovascular diseases present a major socio-economic burden. One major problem underlying most cardiovascular and congenital heart diseases is the irreversible loss of contractile heart muscle cells, the cardiomyocytes. To reverse damage incurred by myocardial infarction or by surgical correction of cardiac malformations, the loss of cardiac tissue with a thickness of a few millimetres needs to be compensated. A promising approach to this issue is cardiac tissue engineering. In this review we focus on the problem of in vitro vascularisation as implantation of cardiac patches consisting of more than three layers of cardiomyocytes (> 100 μm thick) already results in necrosis. We explain the need for vascularisation and elaborate on the importance to include non-myocytes in order to generate functional vascularised cardiac tissue. We discuss the potential of extracellular matrix molecules in promoting vascularisation and introduce nephronectin as an example of a new promising candidate. Finally, we discuss current biomaterial- based approaches including micropatterning, electrospinning, 3D micro-manufacturing technology and porogens. Collectively, the current literature supports the notion that cardiac tissue engineering is a realistic option for future treatment of paediatric and adult patients with cardiac disease.
Collapse
|
41
|
Hartman CD, Isenberg BC, Chua SG, Wong JY. Extracellular matrix type modulates cell migration on mechanical gradients. Exp Cell Res 2017; 359:361-366. [PMID: 28821395 PMCID: PMC5603420 DOI: 10.1016/j.yexcr.2017.08.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/21/2017] [Accepted: 08/12/2017] [Indexed: 11/25/2022]
Abstract
Extracellular matrix composition and stiffness are known to be critical determinants of cell behavior, modulating processes including differentiation, traction generation, and migration. Recent studies have demonstrated that the ECM composition can modulate how cells migrate in response to gradients in environmental stiffness, altering a cell's ability to undergo durotaxis. These observations were limited to single varieties of extracellular matrix, but typically cells are exposed to environments containing complex mixtures of extracellular matrix proteins. Here, we investigate migration of NIH 3T3 fibroblasts on mechanical gradients coated with one or more type of extracellular matrix protein. Our results show that NIH 3T3 fibroblasts exhibit durotaxis on fibronectin-coated mechanical gradients but not on those coated with laminin, demonstrating that extracellular matrix type can act as a regulator of cell response to mechanical gradients. Interestingly, NIH 3T3 fibroblasts were also observed to migrate randomly on gradients coated with a mixture of both fibronectin and laminin, suggesting that there may be a complex interplay in the cellular response to mechanical gradients in the presence of multiple extracellular matrix signals. These findings indicate that specific composition of available adhesion ligands is a critical determinant of a cell's migratory response to mechanical gradients.
Collapse
Affiliation(s)
- Christopher D Hartman
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
| | - Brett C Isenberg
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
| | - Samantha G Chua
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
| | - Joyce Y Wong
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States.
| |
Collapse
|
42
|
Dettman RW, Simon HG. Rebooting the collagen gel: Artificial hydrogels for the study of epithelial mesenchymal transformation. Dev Dyn 2017; 247:332-339. [PMID: 28786157 DOI: 10.1002/dvdy.24560] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/28/2017] [Accepted: 07/31/2017] [Indexed: 12/11/2022] Open
Abstract
The collagen gel has been used to study epithelial-mesenchymal transformation (EMT) for over 30 years. With advances in the field of materials sciences, new options are available to design optically clear, three-dimensional nature-inspired matrix mimetics to study EMT. Here, we review the history of the collagen gel assay, discuss its current use and how newer artificial matrices can be built to simulate in vivo extracellular environments and investigate important current questions in the EMT field. We suggest that further collaborations between materials scientists and biologists will be critical to move the field of EMT forward. Developmental Dynamics 247:332-339, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Robert W Dettman
- Department of Urology, Feinberg School of Medicine, Northwestern University and Stanley Manne Children's Research Institute, Chicago, Illinois
| | - Hans-Georg Simon
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University and Stanley Manne Children's Research Institute, Chicago, Illinois
| |
Collapse
|
43
|
Stricker S, Knaus P, Simon HG. Putting Cells into Context. Front Cell Dev Biol 2017; 5:32. [PMID: 28424772 PMCID: PMC5380720 DOI: 10.3389/fcell.2017.00032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 03/21/2017] [Indexed: 01/05/2023] Open
Affiliation(s)
- Sigmar Stricker
- Musculoskeletal Development and Regeneration Group, Institute for Chemistry and Biochemistry, Freie Universität BerlinBerlin, Germany
| | - Petra Knaus
- Cell Signaling and Regeneration Group, Institute for Chemistry and Biochemistry, Freie Universität BerlinBerlin, Germany
| | - Hans-Georg Simon
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University and Stanley Manne Children's Research InstituteChicago, IL, USA
| |
Collapse
|
44
|
Simões IN, Vale P, Soker S, Atala A, Keller D, Noiva R, Carvalho S, Peleteiro C, Cabral JMS, Eberli D, da Silva CL, Baptista PM. Acellular Urethra Bioscaffold: Decellularization of Whole Urethras for Tissue Engineering Applications. Sci Rep 2017; 7:41934. [PMID: 28165009 PMCID: PMC5292742 DOI: 10.1038/srep41934] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 01/03/2017] [Indexed: 11/16/2022] Open
Abstract
Patients with stress urinary incontinence mainly suffer from malfunction of the urethra closure mechanism. We established the decellularization of porcine urethras to produce acellular urethra bioscaffolds for future tissue engineering applications, using bioscaffolds or bioscaffold-derived soluble products. Cellular removal was evaluated by H&E, DAPI and DNA quantification. The presence of specific ECM proteins was assessed through immunofluorescence staining and colorimetric assay kits. Human skeletal muscle myoblasts, muscle progenitor cells and adipose-derived stromal vascular fractions were used to evaluate the recellularization of the acellular urethra bioscaffolds. The mechanochemical decellularization system removed ~93% of tissue's DNA, generally preserving ECM's components and microarchitecture. Recellularization was achieved, though methodological advances are required regarding cell seeding strategies and functional assessment. Through microdissection and partial digestion, different urethra ECM-derived coating substrates were formulated (i.e. containing smooth or skeletal muscle ECM) and used to culture MPCs in vitro. The skeletal muscle ECM substrates enhanced fiber formation leading to the expression of the main skeletal muscle-related proteins and genes, as confirmed by immunofluorescence and RT-qPCR. The described methodology produced a urethra bioscaffold that retained vital ECM proteins and was liable to cell repopulation, a crucial first step towards the generation of urethra bioscaffold-based Tissue Engineering products.
Collapse
Affiliation(s)
- Irina N. Simões
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Laboratory for Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Paulo Vale
- Serviço Urologia, Hospital Garcia de Orta, Almada, Portugal
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
| | - Daniel Keller
- Laboratory for Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Rute Noiva
- Faculdade de Medicina Veterinária, The Interdisciplinary Centre of Research in Animal Health (CIISA), Universidade de Lisboa, Lisboa, Portugal
| | - Sandra Carvalho
- Faculdade de Medicina Veterinária, The Interdisciplinary Centre of Research in Animal Health (CIISA), Universidade de Lisboa, Lisboa, Portugal
| | - Conceição Peleteiro
- Faculdade de Medicina Veterinária, The Interdisciplinary Centre of Research in Animal Health (CIISA), Universidade de Lisboa, Lisboa, Portugal
| | - Joaquim M. S. Cabral
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Daniel Eberli
- Laboratory for Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Cláudia L. da Silva
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Pedro M. Baptista
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Instituto de Investigacion Sanitaria de Aragón (IIS Aragon), Zaragoza, Spain
- CIBERehd, Zaragoza, Spain
| |
Collapse
|
45
|
Yang Y, Wang K, Gu X, Leong KW. Biophysical Regulation of Cell Behavior-Cross Talk between Substrate Stiffness and Nanotopography. ENGINEERING (BEIJING, CHINA) 2017; 3:36-54. [PMID: 29071164 PMCID: PMC5653318 DOI: 10.1016/j.eng.2017.01.014] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The stiffness and nanotopographical characteristics of the extracellular matrix (ECM) influence numerous developmental, physiological, and pathological processes in vivo. These biophysical cues have therefore been applied to modulate almost all aspects of cell behavior, from cell adhesion and spreading to proliferation and differentiation. Delineation of the biophysical modulation of cell behavior is critical to the rational design of new biomaterials, implants, and medical devices. The effects of stiffness and topographical cues on cell behavior have previously been reviewed, respectively; however, the interwoven effects of stiffness and nanotopographical cues on cell behavior have not been well described, despite similarities in phenotypic manifestations. Herein, we first review the effects of substrate stiffness and nanotopography on cell behavior, and then focus on intracellular transmission of the biophysical signals from integrins to nucleus. Attempts are made to connect extracellular regulation of cell behavior with the biophysical cues. We then discuss the challenges in dissecting the biophysical regulation of cell behavior and in translating the mechanistic understanding of these cues to tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Yong Yang
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA
| | - Kai Wang
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Kam W. Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| |
Collapse
|
46
|
Duffey OJ, Smart N. Approaches to augment vascularisation and regeneration of the adult heart via the reactivated epicardium. Glob Cardiol Sci Pract 2016; 2016:e201628. [PMID: 28979901 PMCID: PMC5624183 DOI: 10.21542/gcsp.2016.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 12/15/2016] [Indexed: 11/05/2022] Open
Abstract
Survival rates following myocardial infarction have increased in recent years but current treatments for post-infarction recovery are inadequate and cannot induce regeneration of damaged hearts. Regenerative medicine could provide disease-reversing treatments by harnessing modern concepts in cell and developmental biology. A recently-established paradigm in regenerative medicine is that regeneration of a tissue can be achieved by reactivation of the coordinated developmental processes that originally formed the tissue. In the heart, the epicardium has emerged as an important regulator of cardiac development and reactivation of epicardial developmental processes may provide a means to enable cardiac regeneration. Indeed, in adult mouse hearts, treatment with thymosin β4 and other drug-like molecules reactivates the epicardium and improves outcomes after myocardial infarction by inducing regenerative paracrine signalling, neovascularisation and de novo cardiomyocyte production. However, there are considerable limitations to current methods of epicardial reactivation that prevent direct translation into clinical practice. Here, we describe the rationale for targeting the epicardium and the successes and limitations of this approach. We consider how several recent advances in epicardial biology could be used to overcome these limitations. These advances include insight into epicardial signalling and heterogeneity, epicardial modulation of inflammation and epicardial remodelling of extracellular matrix.
Collapse
Affiliation(s)
- Owen J. Duffey
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Nicola Smart
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
47
|
Huang TY, Chang CC, Cheng NC, Wang MH, Chiou LL, Lee KL, Lee HS. Re-epithelialization of large wound in paedomorphic and metamorphic axolotls. J Morphol 2016; 278:228-235. [PMID: 27859516 DOI: 10.1002/jmor.20631] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 10/26/2016] [Accepted: 11/01/2016] [Indexed: 11/11/2022]
Abstract
Axolotls (Ambystoma mexicanum) may heal their skin wounds scar-free in both paedomorphs and metamorphs. In previous studies on small punch skin wounds, rapid re-epithelialisation was noted in these two axolotl morphs. However, large wound size in mammals may affect wound healing. In this study, large circumferential full thickness excision wounds on the hind limbs were created on juvenile paedomorphic and metamorphic axolotls. The results showed re-epithelialisation was more quickly initiated in paedomorphs than in metamorphs after wounding. The migrating rate of epidermis on the wound bed was faster in paedomorphs than in metamorphs and thus completion of re-epithelialisation was faster in paedomorphs than in metamorphs. Within these re-epithelialisation periods, neither basement membrane nor dermis was reformed. Epidermal cell proliferation was detected by EdU-labelling technique. In the normal unwounded skin, epidermal proliferation rate was higher in paedomorphs than in metamorphs. After wounding, the epidermal proliferation rate was significantly lower in the migrating front on the wound bed than in the normal skin in paedomorphs. The EdU-labelling rate between normal skin and migration front was not different in metamorphs. Lacking of more proliferating epidermal cells on the wound bed indicated that the new epidermis here derived rather from migrating epidermal cells than from cell proliferation in situ. In conclusion, re-epithelialisation in the large wound might be fully completed in both morphs despite it was initiated earlier and with faster rate in paedomorphs than in metamorphs. The new epidermis on the wound bed derived mainly from cell migration than by cell proliferation in the re-epithelialisation period. J. Morphol. 278:228-235, 2017. © 2016 Wiley Periodicals,Inc.
Collapse
Affiliation(s)
- Ting-Yu Huang
- Liver Disease Prevention and Treatment Research Foundation, Taipei, Taiwan
| | - Chun-Che Chang
- Department of Entomology, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan.,Institute of Biotechnology, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan
| | - Nai-Chen Cheng
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Mu-Hui Wang
- Institute of Biotechnology, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan
| | - Ling-Ling Chiou
- Liver Disease Prevention and Treatment Research Foundation, Taipei, Taiwan
| | - Kuang-Lun Lee
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hsuan-Shu Lee
- Institute of Biotechnology, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.,Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
48
|
Hartman CD, Isenberg BC, Chua SG, Wong JY. Vascular smooth muscle cell durotaxis depends on extracellular matrix composition. Proc Natl Acad Sci U S A 2016; 113:11190-11195. [PMID: 27647912 PMCID: PMC5056055 DOI: 10.1073/pnas.1611324113] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Mechanical compliance has been demonstrated to be a key determinant of cell behavior, directing processes such as spreading, migration, and differentiation. Durotaxis, directional migration from softer to more stiff regions of a substrate, has been observed for a variety of cell types. Recent stiffness mapping experiments have shown that local changes in tissue stiffness in disease are often accompanied by an altered ECM composition in vivo. However, the importance of ECM composition in durotaxis has not yet been explored. To address this question, we have developed and characterized a polyacrylamide hydrogel culture platform featuring highly tunable gradients in mechanical stiffness. This feature, together with the ability to control ECM composition, allows us to isolate the effects of mechanical and biological signals on cell migratory behavior. Using this system, we have tracked vascular smooth muscle cell migration in vitro and quantitatively analyzed differences in cell migration as a function of ECM composition. Our results show that vascular smooth muscle cells undergo durotaxis on mechanical gradients coated with fibronectin but not on those coated with laminin. These findings indicate that the composition of the adhesion ligand is a critical determinant of a cell's migratory response to mechanical gradients.
Collapse
Affiliation(s)
| | - Brett C Isenberg
- Department of Biomedical Engineering, Boston University, Boston, MA 02215
| | - Samantha G Chua
- Department of Biomedical Engineering, Boston University, Boston, MA 02215
| | - Joyce Y Wong
- Department of Biomedical Engineering, Boston University, Boston, MA 02215
| |
Collapse
|
49
|
Quijano LM, Lynch KM, Allan CH, Badylak SF, Ahsan T. Looking Ahead to Engineering Epimorphic Regeneration of a Human Digit or Limb. TISSUE ENGINEERING. PART B, REVIEWS 2016; 22:251-62. [PMID: 26603349 PMCID: PMC4892205 DOI: 10.1089/ten.teb.2015.0401] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 11/24/2015] [Indexed: 01/08/2023]
Abstract
Approximately 2 million people have had limb amputations in the United States due to disease or injury, with more than 185,000 new amputations every year. The ability to promote epimorphic regeneration, or the regrowth of a biologically based digit or limb, would radically change the prognosis for amputees. This ambitious goal includes the regrowth of a large number of tissues that need to be properly assembled and patterned to create a fully functional structure. We have yet to even identify, let alone address, all the obstacles along the extended progression that limit epimorphic regeneration in humans. This review aims to present introductory fundamentals in epimorphic regeneration to facilitate design and conduct of research from a tissue engineering and regenerative medicine perspective. We describe the clinical scenario of human digit healing, featuring published reports of regenerative potential. We then broadly delineate the processes of epimorphic regeneration in nonmammalian systems and describe a few mammalian regeneration models. We give particular focus to the murine digit tip, which allows for comparative studies of regeneration-competent and regeneration-incompetent outcomes in the same animal. Finally, we describe a few forward-thinking opportunities for promoting epimorphic regeneration in humans.
Collapse
Affiliation(s)
- Lina M. Quijano
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana
| | - Kristen M. Lynch
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana
| | - Christopher H. Allan
- Department of Orthopedics and Sports Medicine, University of Washington, Seattle, Washington
| | - Stephen F. Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Tabassum Ahsan
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana
| |
Collapse
|
50
|
Xu X, Li Z, Cai L, Calve S, Neu CP. Mapping the Nonreciprocal Micromechanics of Individual Cells and the Surrounding Matrix Within Living Tissues. Sci Rep 2016; 6:24272. [PMID: 27067516 PMCID: PMC4828668 DOI: 10.1038/srep24272] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 03/11/2016] [Indexed: 12/14/2022] Open
Abstract
The biomechanical properties of the extracellular matrix (ECM) play an important role in cell migration, gene expression, and differentiation. Biomechanics measurements of ECM are usually performed on cryotomed tissue sections. However, studies on cell/matrix interplay are impossible to perform due to disruptions in cell viability and tissue architecture from freeze-thaw cycling. We developed a technique to map the stiffness of living cells and surrounding matrix by atomic force microscopy and use fluorescence microscopy to relate those properties to changes in matrix and cell structure in embryonic and adult tissues in situ. Stiffness mapping revealed significant differences between vibratomed (living) and cryotomed tissues. Isolated cells are softer than those in native matrix, suggesting that cell mechanics are profoundly influenced by their three-dimensional environment and processing state. Viable tissues treated by hyaluronidase and cytochalasin D displayed targeted disruption of matrix and cytoskeletal networks, respectively. While matrix stiffness affected cellular stiffness, changes in cell mechanics did not reciprocally influence matrix stiffness.
Collapse
Affiliation(s)
- Xin Xu
- Department of Mechanical Engineering 1111 Engineering Drive, 427 UCB University of Colorado Boulder Boulder, CO 80309-0427, USA
| | - Zhiyu Li
- Purdue University Weldon School of Biomedical Engineering 206 South Martin Jischke Drive West Lafayette, IN 47907, USA
| | - Luyao Cai
- Purdue University Weldon School of Biomedical Engineering 206 South Martin Jischke Drive West Lafayette, IN 47907, USA
| | - Sarah Calve
- Purdue University Weldon School of Biomedical Engineering 206 South Martin Jischke Drive West Lafayette, IN 47907, USA
| | - Corey P Neu
- Department of Mechanical Engineering 1111 Engineering Drive, 427 UCB University of Colorado Boulder Boulder, CO 80309-0427, USA
| |
Collapse
|