1
|
Vesga-Castro C, Mosqueira-Martín L, Ubiria-Urkola P, Marco-Moreno P, González-Imaz K, Rendon-Hinestroza J, Vallejo-Illarramendi A, Paredes J. Development of an in vitro platform for the analysis of contractile and calcium dynamics in single human myotubes. LAB ON A CHIP 2024; 24:4741-4754. [PMID: 39264341 DOI: 10.1039/d3lc00442b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
In vitro myotube cultures are widely used as models for studying muscle pathophysiology, but their limited maturation and heterogeneity pose significant challenges for functional analyses. While they remain the gold standard for studying muscle function in vitro, myotube cultures do not fully recapitulate the complexity and native features of muscle fibers, which may compromise their ability to predict in vivo outcomes. To promote maturation and decrease heterogeneity, we have incorporated engineered structures into myotube cultures, based on a PDMS thin layer with micrometer-sized grooves (μGrooves) placed over a glass substrate. Different sizes and shapes of μGrooves were tested for their ability to promote alignment and fusion of myoblasts and enhance their differentiation into myotubes. A 24 hour electrical field stimulation protocol (4 V, 6 ms, 0.1 Hz) was used to further promote myotube maturation, after which several myotube features were assessed, including myotube alignment, width, fusion index, contractile function, and calcium handling. Our results indicate superior calcium and contractile performance in μGrooved myotubes, particularly with the 100 μm-width 700 μm-long geometry (7 : 1). This platform generated homogeneous and isolated myotubes that reproduced native muscle features, such as excitation-contraction coupling and force-frequency responses. Overall, our 2D muscle platform enables robust high-content assays of calcium dynamics and contractile readouts with increased sensitivity and reproducibility compared to traditional myotube cultures, making it particularly suitable for screening therapeutic candidates for different muscle pathologies.
Collapse
Affiliation(s)
- Camila Vesga-Castro
- Tecnun School of Engineering, University of Navarra, Manuel de Lardizábal 13, 20018 San Sebastián, Spain.
- Biomedical Engineering Center, University of Navarra, Campus Universitario, 31080, Pamplona, Spain
| | - Laura Mosqueira-Martín
- Group of Neuroscience, Department of Pediatrics, Faculty of Medicine and Nursing, Donostia Hospital, UPV/EHU, 20014 San Sebastian, Spain.
- Group of Neuromuscular Diseases, IIS Biogipuzkoa, 20014 San Sebastian, Spain
- CIBERNED, Ministry of Science and Innovation, Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Paul Ubiria-Urkola
- Tecnun School of Engineering, University of Navarra, Manuel de Lardizábal 13, 20018 San Sebastián, Spain.
- Biomedical Engineering Center, University of Navarra, Campus Universitario, 31080, Pamplona, Spain
| | - Pablo Marco-Moreno
- Group of Neuroscience, Department of Pediatrics, Faculty of Medicine and Nursing, Donostia Hospital, UPV/EHU, 20014 San Sebastian, Spain.
- Group of Neuromuscular Diseases, IIS Biogipuzkoa, 20014 San Sebastian, Spain
- CIBERNED, Ministry of Science and Innovation, Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Klaudia González-Imaz
- Group of Neuroscience, Department of Pediatrics, Faculty of Medicine and Nursing, Donostia Hospital, UPV/EHU, 20014 San Sebastian, Spain.
- Group of Neuromuscular Diseases, IIS Biogipuzkoa, 20014 San Sebastian, Spain
- CIBERNED, Ministry of Science and Innovation, Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Jorge Rendon-Hinestroza
- Group of Neuroscience, Department of Pediatrics, Faculty of Medicine and Nursing, Donostia Hospital, UPV/EHU, 20014 San Sebastian, Spain.
- Group of Neuromuscular Diseases, IIS Biogipuzkoa, 20014 San Sebastian, Spain
| | - Ainara Vallejo-Illarramendi
- Group of Neuroscience, Department of Pediatrics, Faculty of Medicine and Nursing, Donostia Hospital, UPV/EHU, 20014 San Sebastian, Spain.
- Group of Neuromuscular Diseases, IIS Biogipuzkoa, 20014 San Sebastian, Spain
- CIBERNED, Ministry of Science and Innovation, Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Jacobo Paredes
- Tecnun School of Engineering, University of Navarra, Manuel de Lardizábal 13, 20018 San Sebastián, Spain.
- Biomedical Engineering Center, University of Navarra, Campus Universitario, 31080, Pamplona, Spain
| |
Collapse
|
2
|
Zhou Z, Liu J, Xiong T, Liu Y, Tuan RS, Li ZA. Engineering Innervated Musculoskeletal Tissues for Regenerative Orthopedics and Disease Modeling. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310614. [PMID: 38200684 DOI: 10.1002/smll.202310614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/28/2023] [Indexed: 01/12/2024]
Abstract
Musculoskeletal (MSK) disorders significantly burden patients and society, resulting in high healthcare costs and productivity loss. These disorders are the leading cause of physical disability, and their prevalence is expected to increase as sedentary lifestyles become common and the global population of the elderly increases. Proper innervation is critical to maintaining MSK function, and nerve damage or dysfunction underlies various MSK disorders, underscoring the potential of restoring nerve function in MSK disorder treatment. However, most MSK tissue engineering strategies have overlooked the significance of innervation. This review first expounds upon innervation in the MSK system and its importance in maintaining MSK homeostasis and functions. This will be followed by strategies for engineering MSK tissues that induce post-implantation in situ innervation or are pre-innervated. Subsequently, research progress in modeling MSK disorders using innervated MSK organoids and organs-on-chips (OoCs) is analyzed. Finally, the future development of engineering innervated MSK tissues to treat MSK disorders and recapitulate disease mechanisms is discussed. This review provides valuable insights into the underlying principles, engineering methods, and applications of innervated MSK tissues, paving the way for the development of targeted, efficacious therapies for various MSK conditions.
Collapse
Affiliation(s)
- Zhilong Zhou
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Jun Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Tiandi Xiong
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Yuwei Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, 518000, P. R. China
| | - Rocky S Tuan
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Zhong Alan Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518057, P. R. China
| |
Collapse
|
3
|
Takahashi H, Ishiyama K, Takeda N, Shimizu T. Nutrient Rescue of Early Maturing and Deteriorating Satellite Cell-Derived Engineered Muscle Tissue. Tissue Eng Part A 2023; 29:633-644. [PMID: 37694582 DOI: 10.1089/ten.tea.2023.0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023] Open
Abstract
Engineered human muscle tissue is a promising tool for tissue models to better understand muscle physiology and diseases, since they can replicate many biomimetic structures and functions of skeletal muscle in vitro. We have developed a method to produce contractile muscle sheet tissues from human myoblasts, based on our cell sheet fabrication technique. This study reports that our tissue engineering technique allowed us to discover unique characteristics of human muscle satellite cells as a cell source for our muscle sheet tissue. The tissues engineered from satellite cells functionally matured within several days, which is earlier than those created from myoblasts. On the other hand, satellite cell-derived muscle sheet tissues were unable to maintain the contractile ability, whereas the myoblast-derived tissues showed muscle contractions for several weeks. The sarcomere structures and membrane-like structures of laminin and dystrophin were lost along with early functional deterioration. Based on a hypothesis that an insufficiency of nutrients caused a shortened lifetime, we supplemented the culture medium for the satellite cell-derived muscle sheet tissues with 10% serum, although a lower serum medium is commonly used to produce muscle tissues. Further combined with the transforming growth factor (TGF-β1) receptor inhibitor, SB431542, the contractile ability of the muscle tissues was increased remarkably and the tissue microstructures were maintained for a longer term, while retaining the early functionalization and the enriched culture conditions prevented early deterioration. These results strengthened our understanding of the biology of myoblasts and satellite cells in muscle tissue formation and provided new insights into the applications of muscle tissue engineering.
Collapse
Affiliation(s)
- Hironobu Takahashi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University (TWIns), Tokyo, Japan
| | - Kaho Ishiyama
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University (TWIns), Tokyo, Japan
| | - Naoya Takeda
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University (TWIns), Tokyo, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University (TWIns), Tokyo, Japan
| |
Collapse
|
4
|
Tejedera-Villafranca A, Montolio M, Ramón-Azcón J, Fernández-Costa JM. Mimicking sarcolemmal damage in vitro: a contractile 3D model of skeletal muscle for drug testing in Duchenne muscular dystrophy. Biofabrication 2023; 15:045024. [PMID: 37725998 DOI: 10.1088/1758-5090/acfb3d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/19/2023] [Indexed: 09/21/2023]
Abstract
Duchenne muscular dystrophy (DMD) is the most prevalent neuromuscular disease diagnosed in childhood. It is a progressive and wasting disease, characterized by a degeneration of skeletal and cardiac muscles caused by the lack of dystrophin protein. The absence of this crucial structural protein leads to sarcolemmal fragility, resulting in muscle fiber damage during contraction. Despite ongoing efforts, there is no cure available for DMD patients. One of the primary challenges is the limited efficacy of current preclinical tools, which fail in modeling the biological complexity of the disease. Human-based three-dimensional (3D) cell culture methods appear as a novel approach to accelerate preclinical research by enhancing the reproduction of pathophysiological processes in skeletal muscle. In this work, we developed a patient-derived functional 3D skeletal muscle model of DMD that reproduces the sarcolemmal damage found in the native DMD muscle. These bioengineered skeletal muscle tissues exhibit contractile functionality, as they responded to electrical pulse stimulation. Sustained contractile regimes induced the loss of myotube integrity, mirroring the pathological myotube breakdown inherent in DMD due to sarcolemmal instability. Moreover, damaged DMD tissues showed disease functional phenotypes, such as tetanic fatigue. We also evaluated the therapeutic effect of utrophin upregulator drug candidates on the functionality of the skeletal muscle tissues, thus providing deeper insight into the real impact of these treatments. Overall, our findings underscore the potential of bioengineered 3D skeletal muscle technology to advance DMD research and facilitate the development of novel therapies for DMD and related neuromuscular disorders.
Collapse
Affiliation(s)
- Ainoa Tejedera-Villafranca
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), C/Baldiri Reixac 10-12, E08028 Barcelona, Spain
| | - Marisol Montolio
- Duchenne Parent Project España, E28032 Madrid, Spain
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, E08027 Barcelona, Spain
| | - Javier Ramón-Azcón
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), C/Baldiri Reixac 10-12, E08028 Barcelona, Spain
- Institució Catalana de Reserca i Estudis Avançats (ICREA), Passeig de Lluís Companys, 23, E08010 Barcelona, Spain
| | - Juan M Fernández-Costa
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), C/Baldiri Reixac 10-12, E08028 Barcelona, Spain
| |
Collapse
|
5
|
Takahashi H, Wakayama H, Nagase K, Shimizu T. Engineered Human Muscle Tissue from Multilayered Aligned Myofiber Sheets for Studies of Muscle Physiology and Predicting Drug Response. SMALL METHODS 2023; 7:e2200849. [PMID: 36562139 DOI: 10.1002/smtd.202200849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 10/22/2022] [Indexed: 06/17/2023]
Abstract
In preclinical drug testing, human muscle tissue models are critical to understanding the complex physiology, including drug effects in the human body. This study reports that a multilayering approach to cell sheet-based engineering produces an engineered human muscle tissue with sufficient contractile force suitable for measurement. A thermoresponsive micropatterned substrate regulates the biomimetic alignment of myofiber structures enabling the harvest of the aligned myofibers as a single cell sheet. The functional muscle tissue is produced by layering multiple myofiber sheets on a fibrin-based gel. This gel environment promotes myofiber maturation, provides the tissue an elastic platform for contraction, and allows the attachment of a measurement device. Since this multilayering approach is effective in enhancing the contractile ability of the muscle tissue, this muscle tissue generates a significantly high contractile force that can be measured quantitatively. The multilayered muscle tissue shows unidirectional contraction from electrical and chemical stimulation. In addition, their physiological responses to representative drugs can be determined quantitatively in real time by changes in contractile force and fatigue resistance. These physiological properties indicate that the engineered muscle tissue can become a promising tissue model for preclinical in vitro studies in muscle physiology and drug discovery.
Collapse
Affiliation(s)
- Hironobu Takahashi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, 162-8666, Japan
| | - Haruno Wakayama
- Faculty of Pharmacy, Keio University, Tokyo, 105-8512, Japan
| | - Kenichi Nagase
- Faculty of Pharmacy, Keio University, Tokyo, 105-8512, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, 162-8666, Japan
| |
Collapse
|
6
|
Rexius-Hall ML, Khalil NN, Escopete SS, Li X, Hu J, Yuan H, Parker SJ, McCain ML. A myocardial infarct border-zone-on-a-chip demonstrates distinct regulation of cardiac tissue function by an oxygen gradient. SCIENCE ADVANCES 2022; 8:eabn7097. [PMID: 36475790 PMCID: PMC9728975 DOI: 10.1126/sciadv.abn7097] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
After a myocardial infarction, the boundary between the injured, hypoxic tissue and the adjacent viable, normoxic tissue, known as the border zone, is characterized by an oxygen gradient. Yet, the impact of an oxygen gradient on cardiac tissue function is poorly understood, largely due to limitations of existing experimental models. Here, we engineered a microphysiological system to controllably expose engineered cardiac tissue to an oxygen gradient that mimics the border zone and measured the effects of the gradient on electromechanical function and the transcriptome. The gradient delayed calcium release, reuptake, and propagation; decreased diastolic and peak systolic stress; and increased expression of inflammatory cascades that are hallmarks of myocardial infarction. These changes were distinct from those observed in tissues exposed to uniform normoxia or hypoxia, demonstrating distinct regulation of cardiac tissue phenotypes by an oxygen gradient. Our border-zone-on-a-chip model advances functional and mechanistic insight into oxygen-dependent cardiac tissue pathophysiology.
Collapse
Affiliation(s)
- Megan L. Rexius-Hall
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Natalie N. Khalil
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Sean S. Escopete
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Xin Li
- Department of Mechanics and Aerospace Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Jiayi Hu
- Department of Mechanics and Aerospace Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Hongyan Yuan
- Department of Mechanics and Aerospace Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Sarah J. Parker
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Megan L. McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
- Corresponding author.
| |
Collapse
|
7
|
Tissue Engineering Applied to Skeletal Muscle: Strategies and Perspectives. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9120744. [PMID: 36550950 PMCID: PMC9774646 DOI: 10.3390/bioengineering9120744] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 12/04/2022]
Abstract
Muscle tissue is formed by elongated and contractile cells with specific morphofunctional characteristics. Thus, it is divided into three basic types: smooth muscle tissue, cardiac striated muscle tissue and skeletal striated muscle tissue. The striated skeletal muscle tissue presents high plasticity, regeneration and growth capacity due to the presence of satellite cells, quiescent myoblasts that are activated in case of injury to the tissue and originate new muscle fibers when they differentiate. In more severe deficiencies or injuries there is a loss of their regenerative capacity, thus compromising the body's functionality at different levels. Tissue engineering studies the development of biomaterials capable of stimulating the recovery of cellular activity in injured body tissues, as well as the activity of cells with muscle differentiation potential in injury repair. However, the need for three-dimensional re-assembly in a complex organization makes it difficult to mimic this tissue and fully regenerate it for the sake of precise and effective movements. Thus, this article aims to provide a narrative review of tissue engineering strategies applied to the regeneration of skeletal muscle, in a critical evaluation of research, whether aimed at injury or atrophies such as spinal muscular atrophy.
Collapse
|
8
|
Choi Y, Morlino G, Toboso-Navasa A, Hopf R, Pramotton FM, Bigot A, Taddei A, Cesarovic N, Falk V, Mazza E, Giampietro C. A novel bistable device to study mechanosensitive cell responses to instantaneous stretch. BIOMATERIALS ADVANCES 2022; 141:213134. [PMID: 36191540 DOI: 10.1016/j.bioadv.2022.213134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 08/17/2022] [Accepted: 09/25/2022] [Indexed: 06/16/2023]
Abstract
The behavior of cells and tissues in vivo is determined by the integration of multiple biochemical and mechanical signals. Of the mechanical signals, stretch has been studied for decades and shown to contribute to pathophysiological processes. Several different stretch devices have been developed for in vitro investigations of cell stretch. In this work, we describe a new 3D-printed uniaxial stretching device for studying cell response to rapid deformation. The device is a bistable compliant mechanism holding two equilibrium states-an unstretched and stretched configuration-without the need of an external actuator. Furthermore, it allows multiple simultaneous measurements of different levels of stretch on a single substrate and is compatible with standard immunofluorescence imaging of fixed cells as well as live-cell imaging. To demonstrate the effectiveness of the device to stretch cells, a test case using aligned myotubes is presented. Leveraging material area changes associated with deformation of the substrate, changes in nuclei density provided evidence of affine deformation between cells and substrate. Furthermore, intranuclear deformations were also assessed and shown to deform non-affinely. As a proof-of-principle of the use of the device for mechanobiological studies, we uniaxially stretched aligned healthy and dystrophic myotubes that displayed different passive mechanical responses, consistent with previous literature in the field. We also identified a new feature in the mechanoresponse of dystrophic myotubes, which is of potential interest for identifying the diseased cells based on a quick mechanical readout. While some applications of the device for elucidating passive mechanical responses are demonstrated, the simplicity of the device allows it to be potentially used for other modes of deformation with little modifications.
Collapse
Affiliation(s)
- Young Choi
- Department of Mechanical and Process Engineering, ETH Zurich, Zurich 8092, Switzerland
| | | | | | - Raoul Hopf
- Department of Mechanical and Process Engineering, ETH Zurich, Zurich 8092, Switzerland; Swiss Federal Laboratories for Materials Science and Technology (EMPA), Dübendorf 8600, Switzerland; Senecell AG, Zurich 8057, Switzerland
| | - Francesca Michela Pramotton
- Department of Mechanical and Process Engineering, ETH Zurich, Zurich 8092, Switzerland; Swiss Federal Laboratories for Materials Science and Technology (EMPA), Dübendorf 8600, Switzerland
| | - Anne Bigot
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | | | - Nikola Cesarovic
- Department of Health Sciences and Technology, ETH Zurich, Zurich 8092, Switzerland; Charité - Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Volkmar Falk
- Department of Health Sciences and Technology, ETH Zurich, Zurich 8092, Switzerland; Charité - Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Edoardo Mazza
- Department of Mechanical and Process Engineering, ETH Zurich, Zurich 8092, Switzerland; Swiss Federal Laboratories for Materials Science and Technology (EMPA), Dübendorf 8600, Switzerland.
| | - Costanza Giampietro
- Department of Mechanical and Process Engineering, ETH Zurich, Zurich 8092, Switzerland; Swiss Federal Laboratories for Materials Science and Technology (EMPA), Dübendorf 8600, Switzerland; Senecell AG, Zurich 8057, Switzerland.
| |
Collapse
|
9
|
Gosselin MRF, Mournetas V, Borczyk M, Verma S, Occhipinti A, Róg J, Bozycki L, Korostynski M, Robson SC, Angione C, Pinset C, Gorecki DC. Loss of full-length dystrophin expression results in major cell-autonomous abnormalities in proliferating myoblasts. eLife 2022; 11:e75521. [PMID: 36164827 PMCID: PMC9514850 DOI: 10.7554/elife.75521] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 09/02/2022] [Indexed: 12/05/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) affects myofibers and muscle stem cells, causing progressive muscle degeneration and repair defects. It was unknown whether dystrophic myoblasts-the effector cells of muscle growth and regeneration-are affected. Using transcriptomic, genome-scale metabolic modelling and functional analyses, we demonstrate, for the first time, convergent abnormalities in primary mouse and human dystrophic myoblasts. In Dmdmdx myoblasts lacking full-length dystrophin, the expression of 170 genes was significantly altered. Myod1 and key genes controlled by MyoD (Myog, Mymk, Mymx, epigenetic regulators, ECM interactors, calcium signalling and fibrosis genes) were significantly downregulated. Gene ontology analysis indicated enrichment in genes involved in muscle development and function. Functionally, we found increased myoblast proliferation, reduced chemotaxis and accelerated differentiation, which are all essential for myoregeneration. The defects were caused by the loss of expression of full-length dystrophin, as similar and not exacerbated alterations were observed in dystrophin-null Dmdmdx-βgeo myoblasts. Corresponding abnormalities were identified in human DMD primary myoblasts and a dystrophic mouse muscle cell line, confirming the cross-species and cell-autonomous nature of these defects. The genome-scale metabolic analysis in human DMD myoblasts showed alterations in the rate of glycolysis/gluconeogenesis, leukotriene metabolism, and mitochondrial beta-oxidation of various fatty acids. These results reveal the disease continuum: DMD defects in satellite cells, the myoblast dysfunction affecting muscle regeneration, which is insufficient to counteract muscle loss due to myofiber instability. Contrary to the established belief, our data demonstrate that DMD abnormalities occur in myoblasts, making these cells a novel therapeutic target for the treatment of this lethal disease.
Collapse
Affiliation(s)
- Maxime RF Gosselin
- School of Pharmacy and Biomedical Sciences, University of PortsmouthPortsmouthUnited Kingdom
| | | | - Malgorzata Borczyk
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology PASKrakowPoland
| | - Suraj Verma
- School of Computing, Engineering and Digital Technologies, Teesside UniversityMiddlesbroughUnited Kingdom
| | - Annalisa Occhipinti
- School of Computing, Engineering and Digital Technologies, Teesside UniversityMiddlesbroughUnited Kingdom
| | - Justyna Róg
- School of Pharmacy and Biomedical Sciences, University of PortsmouthPortsmouthUnited Kingdom
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental BiologyWarsawPoland
| | - Lukasz Bozycki
- School of Pharmacy and Biomedical Sciences, University of PortsmouthPortsmouthUnited Kingdom
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental BiologyWarsawPoland
| | - Michal Korostynski
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology PASKrakowPoland
| | - Samuel C Robson
- School of Pharmacy and Biomedical Sciences, University of PortsmouthPortsmouthUnited Kingdom
- Centre for Enzyme Innovation, University of PortsmouthPortsmouthUnited Kingdom
| | - Claudio Angione
- School of Computing, Engineering and Digital Technologies, Teesside UniversityMiddlesbroughUnited Kingdom
| | | | - Dariusz C Gorecki
- School of Pharmacy and Biomedical Sciences, University of PortsmouthPortsmouthUnited Kingdom
| |
Collapse
|
10
|
Takahashi H, Yoshida A, Gao B, Yamanaka K, Shimizu T. Harvest of quality-controlled bovine myogenic cells and biomimetic bovine muscle tissue engineering for sustainable meat production. Biomaterials 2022; 287:121649. [PMID: 35779482 DOI: 10.1016/j.biomaterials.2022.121649] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 05/19/2022] [Accepted: 06/22/2022] [Indexed: 11/20/2022]
Abstract
Alternative technology for meat production holds the potential to alleviate ethical, environmental, and public health concerns associated with conventional meat production. Cultured meat produced using cell culture technology promises to become a viable alternative to animal-raised meat for the future of the food industry. In this study, biomimetic bovine muscle tissue was artificially fabricated from myogenic cells extracted from bovine meat. Our primary culture method relies on three key factors; a sequential digesting process, enzymatic treatment with pronase, and coating with laminin fragment on culture dishes. This method allows the efficient collection of large numbers of primary cells from bovine cheek meat, purifies the myogenic cells from the cell mixture, and then continuously grows the myogenic cells in vitro. In addition, using our "quality control" methods, we were able to determine the "cell quality", including the proliferative and differentiation capability in each step of the primary culture. Furthermore, to mimic native bovine meat, the quality-controlled bovine myogenic cells were cultured on a micropatterned thermoresponsive substrate stimulating a native-like aligned structure of cells, which were then transferred onto a fibrin-based gel. This gel-based culture environment promoted structural and functional maturation of the myogenic cells, resulting in the production of bovine muscle tissues with sarcomere structures, native-like membrane structures, and contractile ability. We believe that these biomimetic features of "tissue-engineered meat" are important for the production of future cultured meat, which will need native-like nutrients, texture and taste. Therefore, our meat production approach will provide a new platform to produce more native biomimetic tissue-engineered meat in the near future.
Collapse
Affiliation(s)
- Hironobu Takahashi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666 Japan.
| | - Azumi Yoshida
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666 Japan
| | - Botao Gao
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666 Japan
| | - Kumiko Yamanaka
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666 Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666 Japan
| |
Collapse
|
11
|
Carraro E, Rossi L, Maghin E, Canton M, Piccoli M. 3D in vitro Models of Pathological Skeletal Muscle: Which Cells and Scaffolds to Elect? Front Bioeng Biotechnol 2022; 10:941623. [PMID: 35898644 PMCID: PMC9313593 DOI: 10.3389/fbioe.2022.941623] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/21/2022] [Indexed: 12/29/2022] Open
Abstract
Skeletal muscle is a fundamental tissue of the human body with great plasticity and adaptation to diseases and injuries. Recreating this tissue in vitro helps not only to deepen its functionality, but also to simulate pathophysiological processes. In this review we discuss the generation of human skeletal muscle three-dimensional (3D) models obtained through tissue engineering approaches. First, we present an overview of the most severe myopathies and the two key players involved: the variety of cells composing skeletal muscle tissue and the different components of its extracellular matrix. Then, we discuss the peculiar characteristics among diverse in vitro models with a specific focus on cell sources, scaffold composition and formulations, and fabrication techniques. To conclude, we highlight the efficacy of 3D models in mimicking patient-specific myopathies, deepening muscle disease mechanisms or investigating possible therapeutic effects.
Collapse
Affiliation(s)
- Eugenia Carraro
- Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Lucia Rossi
- Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Edoardo Maghin
- Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Marcella Canton
- Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Martina Piccoli
- Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
- *Correspondence: Martina Piccoli,
| |
Collapse
|
12
|
Vesga-Castro C, Aldazabal J, Vallejo-Illarramendi A, Paredes J. Contractile force assessment methods for in vitro skeletal muscle tissues. eLife 2022; 11:e77204. [PMID: 35604384 PMCID: PMC9126583 DOI: 10.7554/elife.77204] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/27/2022] [Indexed: 02/06/2023] Open
Abstract
Over the last few years, there has been growing interest in measuring the contractile force (CF) of engineered muscle tissues to evaluate their functionality. However, there are still no standards available for selecting the most suitable experimental platform, measuring system, culture protocol, or stimulation patterns. Consequently, the high variability of published data hinders any comparison between different studies. We have identified that cantilever deflection, post deflection, and force transducers are the most commonly used configurations for CF assessment in 2D and 3D models. Additionally, we have discussed the most relevant emerging technologies that would greatly complement CF evaluation with intracellular and localized analysis. This review provides a comprehensive analysis of the most significant advances in CF evaluation and its critical parameters. In order to compare contractile performance across experimental platforms, we have used the specific force (sF, kN/m2), CF normalized to the calculated cross-sectional area (CSA). However, this parameter presents a high variability throughout the different studies, which indicates the need to identify additional parameters and complementary analysis suitable for proper comparison. We propose that future contractility studies in skeletal muscle constructs report detailed information about construct size, contractile area, maturity level, sarcomere length, and, ideally, the tetanus-to-twitch ratio. These studies will hopefully shed light on the relative impact of these variables on muscle force performance of engineered muscle constructs. Prospective advances in muscle tissue engineering, particularly in muscle disease models, will require a joint effort to develop standardized methodologies for assessing CF of engineered muscle tissues.
Collapse
Affiliation(s)
- Camila Vesga-Castro
- University of Navarra, Tecnun School of Engineering, Manuel de LardizábalSan SebastianSpain
- University of Navarra, Biomedical Engineering Center, Campus UniversitarioPamplonaSpain
- Group of Neurosciences, Department of Pediatrics, UPV/EHU, Hospital Donostia - IIS BiodonostiaSan SebastianSpain
| | - Javier Aldazabal
- University of Navarra, Tecnun School of Engineering, Manuel de LardizábalSan SebastianSpain
- University of Navarra, Biomedical Engineering Center, Campus UniversitarioPamplonaSpain
| | - Ainara Vallejo-Illarramendi
- Group of Neurosciences, Department of Pediatrics, UPV/EHU, Hospital Donostia - IIS BiodonostiaSan SebastianSpain
- CIBERNED, Instituto de Salud Carlos III, Ministry of Science, Innovation, and UniversitiesMadridSpain
| | - Jacobo Paredes
- University of Navarra, Tecnun School of Engineering, Manuel de LardizábalSan SebastianSpain
- University of Navarra, Biomedical Engineering Center, Campus UniversitarioPamplonaSpain
| |
Collapse
|
13
|
Cakal SD, Radeke C, Alcala JF, Ellman DG, Butdayev S, Andersen DC, Calloe K, Lind JU. A simple and scalable 3D printing methodology for generating aligned and extended human and murine skeletal muscle tissues. Biomed Mater 2022; 17. [PMID: 35483352 DOI: 10.1088/1748-605x/ac6b71] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/28/2022] [Indexed: 11/11/2022]
Abstract
Preclinical biomedical and pharmaceutical research on disease causes, drug targets, and side effects increasingly relies on in vitro models of human tissue. 3D printing offers unique opportunities for generating models of superior physiological accuracy, as well as for automating their fabrication. Towards these goals, we here describe a simple and scalable methodology for generating physiologically relevant models of skeletal muscle. Our approach relies on dual-material micro-extrusion of two types of gelatin hydrogel into patterned soft substrates with locally alternating stiffness. We identify minimally complex patterns capable of guiding the large-scale self-assembly of aligned, extended, and contractile human and murine skeletal myotubes. Interestingly, we find high-resolution patterning is not required, as even patterns with feature sizes of several hundred micrometers is sufficient. Consequently, the procedure is rapid and compatible with any low-cost extrusion-based 3D printer. The generated myotubes easily span several millimeters, and various myotube patterns can be generated in a predictable and reproducible manner. The compliant nature and adjustable thickness of the hydrogel substrates, serves to enable extended culture of contractile myotubes. The method is further readily compatible with standard cell-culturing platforms as well as commercially available electrodes for electrically induced exercise and monitoring of the myotubes.
Collapse
Affiliation(s)
- Selgin D Cakal
- Department of Health Technology, Technical University of Denmark, Produktionstorvet, Building 423, Lyngby, 2800, DENMARK
| | - Carmen Radeke
- Department of Health Technology, Technical University of Denmark, Produktionstorvet, Building 423, Lyngby, 2800, DENMARK
| | - Juan F Alcala
- Department of Health Technology, Technical University of Denmark, Produktionstorvet, Building 423, Lyngby, 2800, DENMARK
| | - Ditte G Ellman
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, J. B. Winsløwsvej 25, Odense, Syddanmark, 5000, DENMARK
| | - Sarkhan Butdayev
- Department of Health Technology, Technical University of Denmark, Produktionstorvet, Building 423, Lyngby, 2800, DENMARK
| | - Ditte C Andersen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, J. B. Winsløwsvej 25, Odense, Syddanmark, 5000, DENMARK
| | - Kirstine Calloe
- Department of Veterinary and Animal Sciences, Section for Pathobiological Sciences, University of Copenhagen, Grønnegårdsvej 7, Frederiksberg C, 1870, DENMARK
| | - Johan Ulrik Lind
- Institut for Sundhedsteknologi, Danmarks Tekniske Universitet, Produktionstorvet, Building 423, Lyngby, 2800, DENMARK
| |
Collapse
|
14
|
Barthélémy F, Santoso JW, Rabichow L, Jin R, Little I, Nelson SF, McCain ML, Miceli MC. Modeling Patient-Specific Muscular Dystrophy Phenotypes and Therapeutic Responses in Reprogrammed Myotubes Engineered on Micromolded Gelatin Hydrogels. Front Cell Dev Biol 2022; 10:830415. [PMID: 35465312 PMCID: PMC9020228 DOI: 10.3389/fcell.2022.830415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/22/2022] [Indexed: 11/24/2022] Open
Abstract
In vitro models of patient-derived muscle allow for more efficient development of genetic medicines for the muscular dystrophies, which often present mutation-specific pathologies. One popular strategy to generate patient-specific myotubes involves reprogramming dermal fibroblasts to a muscle lineage through MyoD induction. However, creating physiologically relevant, reproducible tissues exhibiting multinucleated, aligned myotubes with organized striations is dependent on the introduction of physicochemical cues that mimic the native muscle microenvironment. Here, we engineered patient-specific control and dystrophic muscle tissues in vitro by culturing and differentiating MyoD–directly reprogrammed fibroblasts isolated from one healthy control subject, three patients with Duchenne muscular dystrophy (DMD), and two Limb Girdle 2A/R1 (LGMD2A/R1) patients on micromolded gelatin hydrogels. Engineered DMD and LGMD2A/R1 tissues demonstrated varying levels of defects in α-actinin expression and organization relative to control, depending on the mutation. In genetically relevant DMD tissues amenable to mRNA reframing by targeting exon 44 or 45 exclusion, exposure to exon skipping antisense oligonucleotides modestly increased myotube coverage and alignment and rescued dystrophin protein expression. These findings highlight the value of engineered culture substrates in guiding the organization of reprogrammed patient fibroblasts into aligned muscle tissues, thereby extending their value as tools for exploration and dissection of the cellular and molecular basis of genetic muscle defects, rescue, and repair.
Collapse
Affiliation(s)
- Florian Barthélémy
- Department of Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jeffrey W. Santoso
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
| | - Laura Rabichow
- Department of Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, Los Angeles, CA, United States
| | - Rongcheng Jin
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
| | - Isaiah Little
- Department of Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, Los Angeles, CA, United States
| | - Stanley F. Nelson
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Megan L. McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States
- *Correspondence: M. Carrie Miceli, ; Megan L. McCain,
| | - M. Carrie Miceli
- Department of Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, Los Angeles, CA, United States
- *Correspondence: M. Carrie Miceli, ; Megan L. McCain,
| |
Collapse
|
15
|
Zschüntzsch J, Meyer S, Shahriyari M, Kummer K, Schmidt M, Kummer S, Tiburcy M. The Evolution of Complex Muscle Cell In Vitro Models to Study Pathomechanisms and Drug Development of Neuromuscular Disease. Cells 2022; 11:1233. [PMID: 35406795 PMCID: PMC8997482 DOI: 10.3390/cells11071233] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/25/2022] [Accepted: 03/31/2022] [Indexed: 12/04/2022] Open
Abstract
Many neuromuscular disease entities possess a significant disease burden and therapeutic options remain limited. Innovative human preclinical models may help to uncover relevant disease mechanisms and enhance the translation of therapeutic findings to strengthen neuromuscular disease precision medicine. By concentrating on idiopathic inflammatory muscle disorders, we summarize the recent evolution of the novel in vitro models to study disease mechanisms and therapeutic strategies. A particular focus is laid on the integration and simulation of multicellular interactions of muscle tissue in disease phenotypes in vitro. Finally, the requirements of a neuromuscular disease drug development workflow are discussed with a particular emphasis on cell sources, co-culture systems (including organoids), functionality, and throughput.
Collapse
Affiliation(s)
- Jana Zschüntzsch
- Department of Neurology, University Medical Center Goettingen, 37075 Goettingen, Germany; (S.M.); (K.K.); (M.S.)
| | - Stefanie Meyer
- Department of Neurology, University Medical Center Goettingen, 37075 Goettingen, Germany; (S.M.); (K.K.); (M.S.)
| | - Mina Shahriyari
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, 37075 Goettingen, Germany;
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Goettingen, Germany
| | - Karsten Kummer
- Department of Neurology, University Medical Center Goettingen, 37075 Goettingen, Germany; (S.M.); (K.K.); (M.S.)
| | - Matthias Schmidt
- Department of Neurology, University Medical Center Goettingen, 37075 Goettingen, Germany; (S.M.); (K.K.); (M.S.)
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, 37075 Goettingen, Germany;
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Goettingen, Germany
| | - Susann Kummer
- Risk Group 4 Pathogens–Stability and Persistence, Biosafety Level-4 Laboratory, Center for Biological Threats and Special Pathogens, Robert Koch Institute, 13353 Berlin, Germany;
| | - Malte Tiburcy
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, 37075 Goettingen, Germany;
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Goettingen, Germany
| |
Collapse
|
16
|
Luciano M, Versaevel M, Vercruysse E, Procès A, Kalukula Y, Remson A, Deridoux A, Gabriele S. Appreciating the role of cell shape changes in the mechanobiology of epithelial tissues. BIOPHYSICS REVIEWS 2022; 3:011305. [PMID: 38505223 PMCID: PMC10903419 DOI: 10.1063/5.0074317] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/23/2022] [Indexed: 03/21/2024]
Abstract
The wide range of epithelial cell shapes reveals the complexity and diversity of the intracellular mechanisms that serve to construct their morphology and regulate their functions. Using mechanosensitive steps, epithelial cells can sense a variety of different mechanochemical stimuli and adapt their behavior by reshaping their morphology. These changes of cell shape rely on a structural reorganization in space and time that generates modifications of the tensional state and activates biochemical cascades. Recent studies have started to unveil how the cell shape maintenance is involved in mechanical homeostatic tasks to sustain epithelial tissue folding, identity, and self-renewal. Here, we review relevant works that integrated mechanobiology to elucidate some of the core principles of how cell shape may be conveyed into spatial information to guide collective processes such as epithelial morphogenesis. Among many other parameters, we show that the regulation of the cell shape can be understood as the result of the interplay between two counteracting mechanisms: actomyosin contractility and intercellular adhesions, and that both do not act independently but are functionally integrated to operate on molecular, cellular, and tissue scales. We highlight the role of cadherin-based adhesions in force-sensing and mechanotransduction, and we report recent developments that exploit physics of liquid crystals to connect cell shape changes to orientational order in cell aggregates. Finally, we emphasize that the further intermingling of different disciplines to develop new mechanobiology assays will lead the way toward a unified picture of the contribution of cell shape to the pathophysiological behavior of epithelial tissues.
Collapse
Affiliation(s)
- Marine Luciano
- University of Mons, Interfaces and Complex Fluids Laboratory, Mechanobiology and Biomaterials Group, Research Institute for Biosciences, CIRMAP, 20 Place du Parc, B-7000 Mons, Belgium
| | - Marie Versaevel
- University of Mons, Interfaces and Complex Fluids Laboratory, Mechanobiology and Biomaterials Group, Research Institute for Biosciences, CIRMAP, 20 Place du Parc, B-7000 Mons, Belgium
| | - Eléonore Vercruysse
- University of Mons, Interfaces and Complex Fluids Laboratory, Mechanobiology and Biomaterials Group, Research Institute for Biosciences, CIRMAP, 20 Place du Parc, B-7000 Mons, Belgium
| | - Anthony Procès
- University of Mons, Interfaces and Complex Fluids Laboratory, Mechanobiology and Biomaterials Group, Research Institute for Biosciences, CIRMAP, 20 Place du Parc, B-7000 Mons, Belgium
| | - Yohalie Kalukula
- University of Mons, Interfaces and Complex Fluids Laboratory, Mechanobiology and Biomaterials Group, Research Institute for Biosciences, CIRMAP, 20 Place du Parc, B-7000 Mons, Belgium
| | - Alexandre Remson
- University of Mons, Interfaces and Complex Fluids Laboratory, Mechanobiology and Biomaterials Group, Research Institute for Biosciences, CIRMAP, 20 Place du Parc, B-7000 Mons, Belgium
| | - Amandine Deridoux
- University of Mons, Interfaces and Complex Fluids Laboratory, Mechanobiology and Biomaterials Group, Research Institute for Biosciences, CIRMAP, 20 Place du Parc, B-7000 Mons, Belgium
| | - Sylvain Gabriele
- University of Mons, Interfaces and Complex Fluids Laboratory, Mechanobiology and Biomaterials Group, Research Institute for Biosciences, CIRMAP, 20 Place du Parc, B-7000 Mons, Belgium
| |
Collapse
|
17
|
Takahashi H, Oikawa F, Takeda N, Shimizu T. Contraction Control of Aligned Myofiber Sheet Tissue by Parallel Oriented iPS Cell-derived Neurons. Tissue Eng Part A 2022; 28:661-671. [PMID: 35057641 DOI: 10.1089/ten.tea.2021.0202] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Fabrication and application of engineered complex tissues composed of different types of cells is a crucial milestone in the next phase of tissue engineering. The delicate organization structure of each tissue component and their physiological connections enable all the functions in the human body. In this study, cell sheet-based engineering allowed us to fabricate a complex myofiber sheet tissue using motor neurons derived from human iPS cells. In contrast with previous studies of other groups, a myofiber sheet with a biomimetic aligned structure was produced from human myoblasts using a striped-patterned thermoresponsive dish, which enabled manipulation of the sheet tissue by simply lowering the culture temperature. The myofiber sheet was transferred onto a gel that promotes functional maturation of human myofibers, resulting in production of contractile human muscle tissue. Just by seeding motor neurons onto the sheet tissue, all the neurons physically contacted to the aligned myofibers, and autonomously elongated in parallel to the myofiber orientation. In addition, the neurite outgrowth was enlarged by co-culturing on the myofiber sheet. The presence of the neurons enhanced clustering of myofiber acetylcholine receptors (AChR), typically found at the neuromuscular junctions (NMJs). Consequently, contraction behaviors of the myofiber sheet were regulated by neuronal signal transduction through NMJs. Muscle contraction was induced when the motor neurons were stimulated by glutamic acid, and effectively blocked by administration of d-tubocurarine as an antagonistic inhibitor for the AChR. The fibrin-based gel was useful as a culture environment for tissue maturation and as a favorable substrate for unobstructed contractions. Our neuron-muscle sheet tissue will be scalable by simply enlarging the micropatterned substrate and manipulable three-dimensionally; fabrication of a thick tissue and a bundle-like structured tissue will be possible just by layering multiple sheets or rolling up the sheet. Simplified control over self-orientation of neurite elongation will be advantageous for fabrication of such a large and complex tissue. Therefore, our methodology, established in this study, will be instrumental in future applications of regenerative medicine for locomotion apparatus.
Collapse
Affiliation(s)
| | - Fumiko Oikawa
- Waseda University, 13148, Shinjuku-ku, Tokyo, Japan;
| | - Naoya Takeda
- Waseda University, 13148, Shinjuku-ku, Tokyo, Japan;
| | | |
Collapse
|
18
|
The contracture-in-a-well. An in vitro model distinguishes bulk and interfacial processes of irreversible (fibrotic) cell-mediated contraction. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2022; 133:112661. [DOI: 10.1016/j.msec.2022.112661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 11/21/2022]
|
19
|
Ajalik RE, Alenchery RG, Cognetti JS, Zhang VZ, McGrath JL, Miller BL, Awad HA. Human Organ-on-a-Chip Microphysiological Systems to Model Musculoskeletal Pathologies and Accelerate Therapeutic Discovery. Front Bioeng Biotechnol 2022; 10:846230. [PMID: 35360391 PMCID: PMC8964284 DOI: 10.3389/fbioe.2022.846230] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Human Microphysiological Systems (hMPS), otherwise known as organ- and tissue-on-a-chip models, are an emerging technology with the potential to replace in vivo animal studies with in vitro models that emulate human physiology at basic levels. hMPS platforms are designed to overcome limitations of two-dimensional (2D) cell culture systems by mimicking 3D tissue organization and microenvironmental cues that are physiologically and clinically relevant. Unlike animal studies, hMPS models can be configured for high content or high throughput screening in preclinical drug development. Applications in modeling acute and chronic injuries in the musculoskeletal system are slowly developing. However, the complexity and load bearing nature of musculoskeletal tissues and joints present unique challenges related to our limited understanding of disease mechanisms and the lack of consensus biomarkers to guide biological therapy development. With emphasis on examples of modeling musculoskeletal tissues, joints on chips, and organoids, this review highlights current trends of microphysiological systems technology. The review surveys state-of-the-art design and fabrication considerations inspired by lessons from bioreactors and biological variables emphasizing the role of induced pluripotent stem cells and genetic engineering in creating isogenic, patient-specific multicellular hMPS. The major challenges in modeling musculoskeletal tissues using hMPS chips are identified, including incorporating biological barriers, simulating joint compartments and heterogenous tissue interfaces, simulating immune interactions and inflammatory factors, simulating effects of in vivo loading, recording nociceptors responses as surrogates for pain outcomes, modeling the dynamic injury and healing responses by monitoring secreted proteins in real time, and creating arrayed formats for robotic high throughput screens. Overcoming these barriers will revolutionize musculoskeletal research by enabling physiologically relevant, predictive models of human tissues and joint diseases to accelerate and de-risk therapeutic discovery and translation to the clinic.
Collapse
Affiliation(s)
- Raquel E. Ajalik
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Rahul G. Alenchery
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - John S. Cognetti
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Victor Z. Zhang
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - James L. McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Benjamin L. Miller
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
- Department of Dermatology, University of Rochester, Rochester, NY, United States
| | - Hani A. Awad
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
- *Correspondence: Hani A. Awad,
| |
Collapse
|
20
|
Cho S, Jang J. Recent Trends in Biofabrication Technologies for Studying Skeletal Muscle Tissue-Related Diseases. Front Bioeng Biotechnol 2021; 9:782333. [PMID: 34778240 PMCID: PMC8578921 DOI: 10.3389/fbioe.2021.782333] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 10/18/2021] [Indexed: 01/15/2023] Open
Abstract
In native skeletal muscle, densely packed myofibers exist in close contact with surrounding motor neurons and blood vessels, which are embedded in the fibrous connective tissue. In comparison to conventional two-dimensional (2D) cultures, the three-dimensional (3D) engineered skeletal muscle models allow structural and mechanical resemblance with native skeletal muscle tissue by providing geometric confinement and physiological matrix stiffness to the cells. In addition, various external stimuli applied to these models enhance muscle maturation along with cell-cell and cell-extracellular matrix interaction. Therefore, 3D in vitro muscle models can adequately recapitulate the pathophysiologic events occurring in tissue-tissue interfaces inside the native skeletal muscle such as neuromuscular junction. Moreover, 3D muscle models can induce pathological phenotype of human muscle dystrophies such as Duchenne muscular dystrophy by incorporating patient-derived induced pluripotent stem cells and human primary cells. In this review, we discuss the current biofabrication technologies for modeling various skeletal muscle tissue-related diseases (i.e., muscle diseases) including muscular dystrophies and inflammatory muscle diseases. In particular, these approaches would enable the discovery of novel phenotypic markers and the mechanism study of human muscle diseases with genetic mutations.
Collapse
Affiliation(s)
- Seungyeun Cho
- Department of Convergence IT Engineering, Pohang University of Science and Technology, Pohang, South Korea
| | - Jinah Jang
- Department of Convergence IT Engineering, Pohang University of Science and Technology, Pohang, South Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, South Korea
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, South Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, South Korea
| |
Collapse
|
21
|
Fralish Z, Lotz EM, Chavez T, Khodabukus A, Bursac N. Neuromuscular Development and Disease: Learning From in vitro and in vivo Models. Front Cell Dev Biol 2021; 9:764732. [PMID: 34778273 PMCID: PMC8579029 DOI: 10.3389/fcell.2021.764732] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/06/2021] [Indexed: 01/02/2023] Open
Abstract
The neuromuscular junction (NMJ) is a specialized cholinergic synaptic interface between a motor neuron and a skeletal muscle fiber that translates presynaptic electrical impulses into motor function. NMJ formation and maintenance require tightly regulated signaling and cellular communication among motor neurons, myogenic cells, and Schwann cells. Neuromuscular diseases (NMDs) can result in loss of NMJ function and motor input leading to paralysis or even death. Although small animal models have been instrumental in advancing our understanding of the NMJ structure and function, the complexities of studying this multi-tissue system in vivo and poor clinical outcomes of candidate therapies developed in small animal models has driven the need for in vitro models of functional human NMJ to complement animal studies. In this review, we discuss prevailing models of NMDs and highlight the current progress and ongoing challenges in developing human iPSC-derived (hiPSC) 3D cell culture models of functional NMJs. We first review in vivo development of motor neurons, skeletal muscle, Schwann cells, and the NMJ alongside current methods for directing the differentiation of relevant cell types from hiPSCs. We further compare the efficacy of modeling NMDs in animals and human cell culture systems in the context of five NMDs: amyotrophic lateral sclerosis, myasthenia gravis, Duchenne muscular dystrophy, myotonic dystrophy, and Pompe disease. Finally, we discuss further work necessary for hiPSC-derived NMJ models to function as effective personalized NMD platforms.
Collapse
Affiliation(s)
| | | | | | | | - Nenad Bursac
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
22
|
Nguyen CT, Ebrahimi M, Gilbert PM, Stewart BA. Electrophysiological analysis of healthy and dystrophic 3-D bioengineered skeletal muscle tissues. Am J Physiol Cell Physiol 2021; 321:C749-C759. [PMID: 34406904 DOI: 10.1152/ajpcell.00049.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recently, methods for creating three-dimensional (3-D) human skeletal muscle tissues from myogenic cell lines have been reported. Bioengineered muscle tissues are contractile and respond to electrical and chemical stimulation. In this study, we provide an electrophysiological analysis of healthy and dystrophic 3-D bioengineered skeletal muscle tissues, focusing on Duchenne muscular dystrophy (DMD). We enlist the 3-D in vitro model of DMD muscle tissue to evaluate muscle cell electrical properties uncoupled from presynaptic neural inputs, an understudied aspect of DMD. Our data show that previously reported electrophysiological aspects of DMD, including effects on membrane potential and membrane resistance, are replicated in the 3-D muscle tissue model. Furthermore, we test a potential therapeutic compound, poloxamer 188, and demonstrate capacity for improving the membrane potential in DMD muscle. Therefore, this study serves as a baseline for a new in vitro method to examine potential therapies for muscular disorders.
Collapse
Affiliation(s)
- Christine T Nguyen
- Department of Biology, University of Toronto Mississauga, Toronto, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Majid Ebrahimi
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada.,Donnelly Centre for Cellular and Biomolecular Research, Toronto, Canada
| | - Penney M Gilbert
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada.,Institute of Biomedical Engineering, University of Toronto, Toronto, Canada.,Donnelly Centre for Cellular and Biomolecular Research, Toronto, Canada
| | - Bryan A Stewart
- Department of Biology, University of Toronto Mississauga, Toronto, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| |
Collapse
|
23
|
Ebrahimi M, Lad H, Fusto A, Tiper Y, Datye A, Nguyen CT, Jacques E, Moyle LA, Nguyen T, Musgrave B, Chávez-Madero C, Bigot A, Chen C, Turner S, Stewart BA, Pegoraro E, Vitiello L, Gilbert PM. De novo revertant fiber formation and therapy testing in a 3D culture model of Duchenne muscular dystrophy skeletal muscle. Acta Biomater 2021; 132:227-244. [PMID: 34048976 DOI: 10.1016/j.actbio.2021.05.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 05/16/2021] [Accepted: 05/18/2021] [Indexed: 12/18/2022]
Abstract
The biological basis of Duchenne muscular dystrophy (DMD) pathology is only partially characterized and there are still few disease-modifying therapies available, therein underlying the value of strategies to model and study DMD. Dystrophin, the causative gene of DMD, is responsible for linking the cytoskeleton of muscle fibers to the extracellular matrix beyond the sarcolemma. We posited that disease-associated phenotypes not yet captured by two-dimensional culture methods would arise by generating multinucleated muscle cells within a three-dimensional (3D) extracellular matrix environment. Herein we report methods to produce 3D human skeletal muscle microtissues (hMMTs) using clonal, immortalized myoblast lines established from healthy and DMD donors. We also established protocols to evaluate immortalized hMMT self-organization and myotube maturation, as well as calcium handling, force generation, membrane stability (i.e., creatine kinase activity and Evans blue dye permeability) and contractile apparatus organization following electrical-stimulation. In examining hMMTs generated with a cell line wherein the dystrophin gene possessed a duplication of exon 2, we observed rare dystrophin-positive myotubes, which were not seen in 2D cultures. Further, we show that treating DMD hMMTs with a β1-integrin activating antibody, improves contractile apparatus maturation and stability. Hence, immortalized myoblast-derived DMD hMMTs offer a pre-clinical system with which to investigate the potential of duplicated exon skipping strategies and those that protect muscle cells from contraction-induced injury. STATEMENT OF SIGNIFICANCE: Duchenne muscular dystrophy (DMD) is a progressive muscle-wasting disorder that is caused by mutation of the dystrophin gene. The biological basis of DMD pathology is only partially characterized and there is no cure for this fatal disease. Here we report a method to produce 3D human skeletal muscle microtissues (hMMTs) using immortalized human DMD and healthy myoblasts. Morphological and functional assessment revealed DMD-associated pathophysiology including impaired calcium handling and de novo formation of dystrophin-positive revertant muscle cells in immortalized DMD hMMTs harbouring an exon 2 duplication, a feature of many DMD patients that has not been recapitulated in culture prior to this report. We further demonstrate that this "DMD in a dish" system can be used as a pre-clinical assay to test a putative DMD therapeutic and study the mechanism of action.
Collapse
Affiliation(s)
- Majid Ebrahimi
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Heta Lad
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Aurora Fusto
- Department of Neuroscience, University of Padua, Padua, 35128, Italy
| | - Yekaterina Tiper
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Asiman Datye
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Christine T Nguyen
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S3G5, Canada; Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L1C6, Canada
| | - Erik Jacques
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Louise A Moyle
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Thy Nguyen
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Brennen Musgrave
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Carolina Chávez-Madero
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Anne Bigot
- Sorbonne Universite, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, Paris UMRS974, France
| | - Chun Chen
- Pliant Therapeutics, Inc, South San Francisco, California 94080, USA
| | - Scott Turner
- Pliant Therapeutics, Inc, South San Francisco, California 94080, USA
| | - Bryan A Stewart
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S3G5, Canada; Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L1C6, Canada
| | - Elena Pegoraro
- Department of Neuroscience, University of Padua, Padua, 35128, Italy
| | - Libero Vitiello
- Department of Biology, University of Padua, Padua 35131, Italy; Interuniversity Institute of Myology (IIM), Italy
| | - Penney M Gilbert
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S3G5, Canada.
| |
Collapse
|
24
|
Santoso JW, Li X, Gupta D, Suh GC, Hendricks E, Lin S, Perry S, Ichida JK, Dickman D, McCain ML. Engineering skeletal muscle tissues with advanced maturity improves synapse formation with human induced pluripotent stem cell-derived motor neurons. APL Bioeng 2021; 5:036101. [PMID: 34286174 PMCID: PMC8282350 DOI: 10.1063/5.0054984] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022] Open
Abstract
To develop effective cures for neuromuscular diseases, human-relevant in vitro models of neuromuscular tissues are critically needed to probe disease mechanisms on a cellular and molecular level. However, previous attempts to co-culture motor neurons and skeletal muscle have resulted in relatively immature neuromuscular junctions (NMJs). In this study, NMJs formed by human induced pluripotent stem cell (hiPSC)-derived motor neurons were improved by optimizing the maturity of the co-cultured muscle tissue. First, muscle tissues engineered from the C2C12 mouse myoblast cell line, cryopreserved primary human myoblasts, and freshly isolated primary chick myoblasts on micromolded gelatin hydrogels were compared. After three weeks, only chick muscle tissues remained stably adhered to hydrogels and exhibited progressive increases in myogenic index and stress generation, approaching values generated by native muscle tissue. After three weeks of co-culture with hiPSC-derived motor neurons, engineered chick muscle tissues formed NMJs with increasing co-localization of pre- and postsynaptic markers as well as increased frequency and magnitude of synaptic activity, surpassing structural and functional maturity of previous in vitro models. Engineered chick muscle tissues also demonstrated increased expression of genes related to sarcomere maturation and innervation over time, revealing new insights into the molecular pathways that likely contribute to enhanced NMJ formation. These approaches for engineering advanced neuromuscular tissues with relatively mature NMJs and interrogating their structure and function have many applications in neuromuscular disease modeling and drug development.
Collapse
Affiliation(s)
- Jeffrey W. Santoso
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | - Xiling Li
- Department of Biological Sciences, Dornsife College of Arts and Letters, University of Southern California, Los Angeles, California 90089, USA
| | - Divya Gupta
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | - Gio C. Suh
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | - Eric Hendricks
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, California 90033, USA
| | - Shaoyu Lin
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, California 90033, USA
| | - Sarah Perry
- Department of Biological Sciences, Dornsife College of Arts and Letters, University of Southern California, Los Angeles, California 90089, USA
| | - Justin K. Ichida
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, California 90033, USA
| | - Dion Dickman
- Department of Biological Sciences, Dornsife College of Arts and Letters, University of Southern California, Los Angeles, California 90089, USA
| | - Megan L. McCain
- Author to whom correspondence should be addressed:. Tel: +1 2138210791. URL:https://livingsystemsengineering.usc.edu
| |
Collapse
|
25
|
Jahagirdar D, Bangde P, Jain R, Dandekar P. Degenerative disease-on-a-chip: Developing microfluidic models for rapid availability of newer therapies. Biotechnol J 2021; 16:e2100154. [PMID: 34390543 DOI: 10.1002/biot.202100154] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 08/09/2021] [Accepted: 08/11/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Understanding the pathophysiology of degenerative diseases pertaining to nervous system, ocular region, bone/cartilage and muscle are still being comprehended, thus delaying the availability of targeted therapies. PURPOSE AND SCOPE Newer micro-physiological systems (organ-on-chip technology) involves development of more sophisticated devices, modelling a range of in vitro human tissues and an array of models for diseased conditions. These models expand opportunities for high throughput screening (HTS) of drugs and are likely to be rapid and cost-effective, thus reducing extensive usage of animal models. CONCLUSION Through this review article, we aim to present an overview of the degenerative disease models that are presently being developed using microfluidic platforms with the aim of mimicking in vivo tissue physiology and micro-architecture. The manuscript provides an overview of the degenerative disease models and their potential for testing and screening of possible biotherapeutic molecules and drugs. It highlights the perspective of the regulatory bodies with respect to the established-on chip models and thereby enhancing its translational potential. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Devashree Jahagirdar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Matunga, Mumbai, 400019, India
| | - Prachi Bangde
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Matunga, Mumbai, 400019, India
| | - Ratnesh Jain
- Department of Chemical Engineering, Institute of Chemical Technology, Nathalal Parekh Marg, Matunga, Mumbai, 400019, India
| | - Prajakta Dandekar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Matunga, Mumbai, 400019, India
| |
Collapse
|
26
|
Al Tanoury Z, Zimmerman JF, Rao J, Sieiro D, McNamara HM, Cherrier T, Rodríguez-delaRosa A, Hick-Colin A, Bousson F, Fugier-Schmucker C, Marchiano F, Habermann B, Chal J, Nesmith AP, Gapon S, Wagner E, Gupta VA, Bassel-Duby R, Olson EN, Cohen AE, Parker KK, Pourquié O. Prednisolone rescues Duchenne muscular dystrophy phenotypes in human pluripotent stem cell-derived skeletal muscle in vitro. Proc Natl Acad Sci U S A 2021; 118:e2022960118. [PMID: 34260377 PMCID: PMC8285911 DOI: 10.1073/pnas.2022960118] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a devastating genetic disease leading to degeneration of skeletal muscles and premature death. How dystrophin absence leads to muscle wasting remains unclear. Here, we describe an optimized protocol to differentiate human induced pluripotent stem cells (iPSC) to a late myogenic stage. This allows us to recapitulate classical DMD phenotypes (mislocalization of proteins of the dystrophin-associated glycoprotein complex, increased fusion, myofiber branching, force contraction defects, and calcium hyperactivation) in isogenic DMD-mutant iPSC lines in vitro. Treatment of the myogenic cultures with prednisolone (the standard of care for DMD) can dramatically rescue force contraction, fusion, and branching defects in DMD iPSC lines. This argues that prednisolone acts directly on myofibers, challenging the largely prevalent view that its beneficial effects are caused by antiinflammatory properties. Our work introduces a human in vitro model to study the onset of DMD pathology and test novel therapeutic approaches.
Collapse
Affiliation(s)
- Ziad Al Tanoury
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104, INSERM U964, Université de Strasbourg, 67411 Illkirch Graffenstaden, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - John F Zimmerman
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Boston, MA 02134
| | - Jyoti Rao
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - Daniel Sieiro
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - Harold M McNamara
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
- Department of Physics, Harvard University, Cambridge, MA 02138
| | - Thomas Cherrier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104, INSERM U964, Université de Strasbourg, 67411 Illkirch Graffenstaden, France
| | - Alejandra Rodríguez-delaRosa
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | | | - Fanny Bousson
- Anagenesis Biotechnologies, 67400 Illkirch Graffenstaden, France
| | | | - Fabio Marchiano
- Aix-Marseille University, CNRS, Institut de Biologie du Développement de Marseille UMR 7288, The Turing Center for Living Systems, 13009 Marseille, France
| | - Bianca Habermann
- Aix-Marseille University, CNRS, Institut de Biologie du Développement de Marseille UMR 7288, The Turing Center for Living Systems, 13009 Marseille, France
| | - Jérome Chal
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104, INSERM U964, Université de Strasbourg, 67411 Illkirch Graffenstaden, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - Alexander P Nesmith
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Boston, MA 02134
| | - Svetlana Gapon
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
| | - Erica Wagner
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
| | - Vandana A Gupta
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Eric N Olson
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Adam E Cohen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
- Department of Physics, Harvard University, Cambridge, MA 02138
| | - Kevin Kit Parker
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Boston, MA 02134
| | - Olivier Pourquié
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104, INSERM U964, Université de Strasbourg, 67411 Illkirch Graffenstaden, France;
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| |
Collapse
|
27
|
Canonico F, Chirivi M, Maiullari F, Milan M, Rizzi R, Arcudi A, Galli M, Pane M, Gowran A, Pompilio G, Mercuri E, Crea F, Bearzi C, D'Amario D. Focus on the road to modelling cardiomyopathy in muscular dystrophy. Cardiovasc Res 2021; 118:1872-1884. [PMID: 34254111 DOI: 10.1093/cvr/cvab232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 07/07/2021] [Indexed: 11/13/2022] Open
Abstract
Alterations in the DMD gene, which codes for the protein dystrophin, cause forms of dystrophinopathies such as Duchenne muscular dystrophy, an X-linked disease. Cardiomyopathy linked to DMD mutations is becoming the leading cause of death in patients with dystrophinopathy. Since phenotypic pathophysiological mechanisms are not fully understood, the improvement and development of new disease models, considering their relative advantages and disadvantages, is essential. The application of genetic engineering approaches on induced pluripotent stem cells, such as gene editing technology, enables the development of physiologically relevant human cell models for in vitro dystrophinopathy studies. The combination of induced pluripotent stem cells-derived cardiovascular cell types and 3 D bioprinting technologies hold great promise for the study of dystrophin-linked cardiomyopathy. This combined approach enables the assessment of responses to physical or chemical stimuli, and the influence of pharmaceutical approaches. The critical objective of in vitro microphysiological systems is to more accurately reproduce the microenvironment observed in vivo. Ground-breaking methodology involving the connection of multiple microphysiological systems comprised of different tissues would represent a move toward precision body-on-chip disease modelling could lead to a critical expansion in what is known about inter-organ responses to disease and novel therapies that have the potential to replace animal models. In this review, we will focus on the generation, development, and application of current cellular, animal and potential for bio-printed models, in the study of the pathophysiological mechanisms underlying dystrophin-linked cardiomyopathy in the direction of personalized medicine.
Collapse
Affiliation(s)
- Francesco Canonico
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Department of Cardiovascular Sciences, Rome, Italy
| | - Maila Chirivi
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Monterotondo, Rome, Italy.,Istituto Nazionale Genetica Molecolare (INGM) "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Fabio Maiullari
- Istituto Nazionale Genetica Molecolare (INGM) "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Marika Milan
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Monterotondo, Rome, Italy.,Istituto Nazionale Genetica Molecolare (INGM) "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Roberto Rizzi
- Istituto Nazionale Genetica Molecolare (INGM) "Romeo ed Enrica Invernizzi", Milan, Italy.,Institute of Biomedical Technologies, National Research Council of Italy (ITB-CNR), Segrate, Milan, Italy
| | - Alessandra Arcudi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Department of Cardiovascular Sciences, Rome, Italy
| | - Mattia Galli
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Department of Cardiovascular Sciences, Rome, Italy
| | - Marika Pane
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Department of Women, Children and Public Health Sciences, Rome, Italy
| | - Aoife Gowran
- Centro Cardiologico Monzino IRCCS, Unit of Vascular Biology and Regenerative Medicine, Milan, Italy
| | - Giulio Pompilio
- Centro Cardiologico Monzino IRCCS, Unit of Vascular Biology and Regenerative Medicine, Milan, Italy.,Department of Biomedical, Surgical and Dental Sciences, University of Milan, Italy
| | - Eugenio Mercuri
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Department of Women, Children and Public Health Sciences, Rome, Italy
| | - Filippo Crea
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Department of Cardiovascular Sciences, Rome, Italy
| | - Claudia Bearzi
- Istituto Nazionale Genetica Molecolare (INGM) "Romeo ed Enrica Invernizzi", Milan, Italy.,Institute of Genetic and Biomedical Research, National Research Council (IRGB-CNR), Milan, Italy
| | - Domenico D'Amario
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Department of Cardiovascular Sciences, Rome, Italy
| |
Collapse
|
28
|
Uchimura T, Asano T, Nakata T, Hotta A, Sakurai H. A muscle fatigue-like contractile decline was recapitulated using skeletal myotubes from Duchenne muscular dystrophy patient-derived iPSCs. CELL REPORTS MEDICINE 2021; 2:100298. [PMID: 34195678 PMCID: PMC8233665 DOI: 10.1016/j.xcrm.2021.100298] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 01/28/2021] [Accepted: 05/10/2021] [Indexed: 02/07/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a muscle degenerating disease caused by dystrophin deficiency, for which therapeutic options are limited. To facilitate drug development, it is desirable to develop in vitro disease models that enable the evaluation of DMD declines in contractile performance. Here, we show MYOD1-induced differentiation of hiPSCs into functional skeletal myotubes in vitro with collagen gel and electrical field stimulation (EFS). Long-term EFS training (0.5 Hz, 20 V, 2 ms, continuous for 2 weeks) mimicking muscle overuse recapitulates declines in contractile performance in dystrophic myotubes. A screening of clinically relevant drugs using this model detects three compounds that ameliorate this decline. Furthermore, we validate the feasibility of adapting the model to a 96-well culture system using optogenetic technology for large-scale screening. Our results support a disease model using patient-derived iPSCs that allows for the recapitulation of the contractile pathogenesis of DMD and a screening strategy for drug development.
Collapse
Affiliation(s)
- Tomoya Uchimura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.,Takeda-CiRA Joint Program, Fujisawa, Kanagawa 251-8555, Japan
| | - Toshifumi Asano
- Department of Cell Biology, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo 113-8510, Japan.,The Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Takao Nakata
- Department of Cell Biology, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo 113-8510, Japan.,The Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Akitsu Hotta
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.,Takeda-CiRA Joint Program, Fujisawa, Kanagawa 251-8555, Japan
| | - Hidetoshi Sakurai
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.,Takeda-CiRA Joint Program, Fujisawa, Kanagawa 251-8555, Japan
| |
Collapse
|
29
|
Yamamoto K, Yamaoka N, Imaizumi Y, Nagashima T, Furutani T, Ito T, Okada Y, Honda H, Shimizu K. Development of a human neuromuscular tissue-on-a-chip model on a 24-well-plate-format compartmentalized microfluidic device. LAB ON A CHIP 2021; 21:1897-1907. [PMID: 34008665 DOI: 10.1039/d1lc00048a] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Engineered three-dimensional models of neuromuscular tissues are promising for use in mimicking their disorder states in vitro. Although several models have been developed, it is still challenging to mimic the physically separated structures of motor neurons (MNs) and skeletal muscle (SkM) fibers in the motor units in vivo. In this study, we aimed to develop microdevices for precisely compartmentalized coculturing of MNs and engineered SkM tissues. The developed microdevices, which fit a well of 24 well plates, had a chamber for MNs and chamber for SkM tissues. The two chambers were connected by microtunnels for axons, permissive to axons but not to cell bodies. Human iPSC (hiPSC)-derived MN spheroids in one chamber elongated their axons into microtunnels, which reached the tissue-engineered human SkM in the SkM chamber, and formed functional neuromuscular junctions with the muscle fibers. The cocultured SkM tissues with MNs on the device contracted spontaneously in response to spontaneous firing of MNs. The addition of a neurotransmitter, glutamate, into the MN chamber induced contraction of the cocultured SkM tissues. Selective addition of tetrodotoxin or vecuronium bromide into either chamber induced SkM tissue relaxation, which could be explained by the inhibitory mechanisms. We also demonstrated the application of chemical or mechanical stimuli to the middle of the axons of cocultured tissues on the device. Thus, compartmentalized neuromuscular tissue models fabricated on the device could be used for phenotypic screening to evaluate the cellular type specific efficacy of drug candidates and would be a useful tool in fundamental research and drug development for neuromuscular disorders.
Collapse
Affiliation(s)
- Kazuki Yamamoto
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan.
| | - Nao Yamaoka
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan.
| | - Yu Imaizumi
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan.
| | - Takunori Nagashima
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan.
| | - Taiki Furutani
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan.
| | - Takuji Ito
- Department of Neurology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Yohei Okada
- Department of Neurology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Hiroyuki Honda
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan.
| | - Kazunori Shimizu
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan.
| |
Collapse
|
30
|
Brunetti J, Koenig S, Monnier A, Frieden M. Nanopattern surface improves cultured human myotube maturation. Skelet Muscle 2021; 11:12. [PMID: 33952323 PMCID: PMC8097894 DOI: 10.1186/s13395-021-00268-3] [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: 01/25/2021] [Accepted: 04/19/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In vitro maturation of human primary myoblasts using 2D culture remains a challenging process and leads to immature fibers with poor internal organization and function. This would however represent a valuable system to study muscle physiology or pathophysiology from patient myoblasts, at a single-cell level. METHODS Human primary myoblasts were cultured on 800-nm wide striated surface between two layers of Matrigel, and in a media supplemented with an inhibitor of TGFβ receptor. Gene expression, immunofluorescence, and Ca2+ measurements upon electrical stimulations were performed at various time points during maturation to assess the organization and function of the myotubes. RESULTS We show that after 10 days in culture, myotubes display numerous functional acetylcholine receptor clusters and express the adult isoforms of myosin heavy chain and dihydropyridine receptor. In addition, the myotubes are internally well organized with striations of α-actinin and STIM1, and occasionally ryanodine receptor 1. We also demonstrate that the myotubes present robust Ca2+ responses to repetitive electrical stimulations. CONCLUSION The present method describes a fast and efficient system to obtain well matured and functional myotubes in 2D culture allowing thorough analysis of single-cell Ca2+ signals.
Collapse
Affiliation(s)
- Jessica Brunetti
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Stéphane Koenig
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Arthur Monnier
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Maud Frieden
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
31
|
Compliant 3D frameworks instrumented with strain sensors for characterization of millimeter-scale engineered muscle tissues. Proc Natl Acad Sci U S A 2021; 118:2100077118. [PMID: 33941674 DOI: 10.1073/pnas.2100077118] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Tissue-on-chip systems represent promising platforms for monitoring and controlling tissue functions in vitro for various purposes in biomedical research. The two-dimensional (2D) layouts of these constructs constrain the types of interactions that can be studied and limit their relevance to three-dimensional (3D) tissues. The development of 3D electronic scaffolds and microphysiological devices with geometries and functions tailored to realistic 3D tissues has the potential to create important possibilities in advanced sensing and control. This study presents classes of compliant 3D frameworks that incorporate microscale strain sensors for high-sensitivity measurements of contractile forces of engineered optogenetic muscle tissue rings, supported by quantitative simulations. Compared with traditional approaches based on optical microscopy, these 3D mechanical frameworks and sensing systems can measure not only motions but also contractile forces with high accuracy and high temporal resolution. Results of active tension force measurements of engineered muscle rings under different stimulation conditions in long-term monitoring settings for over 5 wk and in response to various chemical and drug doses demonstrate the utility of such platforms in sensing and modulation of muscle and other tissues. Possibilities for applications range from drug screening and disease modeling to biohybrid robotic engineering.
Collapse
|
32
|
Liu Y, Ren J, Zhang R, Hu S, Pang SW, Lam RHW. Spreading and Migration of Nasopharyngeal Normal and Cancer Cells on Microgratings. ACS APPLIED BIO MATERIALS 2021; 4:3224-3231. [DOI: 10.1021/acsabm.0c01610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Yi Liu
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
| | - Jifeng Ren
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- School of Biomedical Engineering, Capital Medical University, Beijing 100069, China
| | - Ruolin Zhang
- Department of Electrical Engineering, City University of Hong Kong, Kowloon, Hong Kong
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Shuhuan Hu
- BGI-Shenzhen, Shenzhen 518083, Guangdong China
| | - Stella W. Pang
- Department of Electrical Engineering, City University of Hong Kong, Kowloon, Hong Kong
- Centre for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Kowloon, Hong Kong
| | - Raymond H. W. Lam
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- Centre for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, Kowloon, Hong Kong
| |
Collapse
|
33
|
Kim H, Jeong JH, Fendereski M, Lee HS, Kang DY, Hur SS, Amirian J, Kim Y, Pham NT, Suh N, Hwang NSY, Ryu S, Yoon JK, Hwang Y. Heparin-Mimicking Polymer-Based In Vitro Platform Recapitulates In Vivo Muscle Atrophy Phenotypes. Int J Mol Sci 2021; 22:ijms22052488. [PMID: 33801235 PMCID: PMC7957884 DOI: 10.3390/ijms22052488] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 12/13/2022] Open
Abstract
The cell–cell/cell–matrix interactions between myoblasts and their extracellular microenvironment have been shown to play a crucial role in the regulation of in vitro myogenic differentiation and in vivo skeletal muscle regeneration. In this study, by harnessing the heparin-mimicking polymer, poly(sodium-4-styrenesulfonate) (PSS), which has a negatively charged surface, we engineered an in vitro cell culture platform for the purpose of recapitulating in vivo muscle atrophy-like phenotypes. Our initial findings showed that heparin-mimicking moieties inhibited the fusion of mononucleated myoblasts into multinucleated myotubes, as indicated by the decreased gene and protein expression levels of myogenic factors, myotube fusion-related markers, and focal adhesion kinase (FAK). We further elucidated the underlying molecular mechanism via transcriptome analyses, observing that the insulin/PI3K/mTOR and Wnt signaling pathways were significantly downregulated by heparin-mimicking moieties through the inhibition of FAK/Cav3. Taken together, the easy-to-adapt heparin-mimicking polymer-based in vitro cell culture platform could be an attractive platform for potential applications in drug screening, providing clear readouts of changes in insulin/PI3K/mTOR and Wnt signaling pathways.
Collapse
Affiliation(s)
- Hyunbum Kim
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Korea;
| | - Ji Hoon Jeong
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Mona Fendereski
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Hyo-Shin Lee
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Da Yeon Kang
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan-si 31538, Korea; (D.Y.K.); (N.S.)
| | - Sung Sik Hur
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
| | - Jhaleh Amirian
- Institute of Tissue Regeneration, Soonchunhyang University, Asan-si 31538, Korea;
| | - Yunhye Kim
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Nghia Thi Pham
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Nayoung Suh
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan-si 31538, Korea; (D.Y.K.); (N.S.)
| | - Nathaniel Suk-Yeon Hwang
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Korea;
| | - Seongho Ryu
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Jeong Kyo Yoon
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
- Correspondence: (J.K.Y.); (Y.H.); Tel.: +82-41-413-5016 (J.K.Y.); +82-41-413-5017 (Y.H.)
| | - Yongsung Hwang
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
- Correspondence: (J.K.Y.); (Y.H.); Tel.: +82-41-413-5016 (J.K.Y.); +82-41-413-5017 (Y.H.)
| |
Collapse
|
34
|
Khodabukus A. Tissue-Engineered Skeletal Muscle Models to Study Muscle Function, Plasticity, and Disease. Front Physiol 2021; 12:619710. [PMID: 33716768 PMCID: PMC7952620 DOI: 10.3389/fphys.2021.619710] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/25/2021] [Indexed: 12/20/2022] Open
Abstract
Skeletal muscle possesses remarkable plasticity that permits functional adaptations to a wide range of signals such as motor input, exercise, and disease. Small animal models have been pivotal in elucidating the molecular mechanisms regulating skeletal muscle adaptation and plasticity. However, these small animal models fail to accurately model human muscle disease resulting in poor clinical success of therapies. Here, we review the potential of in vitro three-dimensional tissue-engineered skeletal muscle models to study muscle function, plasticity, and disease. First, we discuss the generation and function of in vitro skeletal muscle models. We then discuss the genetic, neural, and hormonal factors regulating skeletal muscle fiber-type in vivo and the ability of current in vitro models to study muscle fiber-type regulation. We also evaluate the potential of these systems to be utilized in a patient-specific manner to accurately model and gain novel insights into diseases such as Duchenne muscular dystrophy (DMD) and volumetric muscle loss. We conclude with a discussion on future developments required for tissue-engineered skeletal muscle models to become more mature, biomimetic, and widely utilized for studying muscle physiology, disease, and clinical use.
Collapse
Affiliation(s)
- Alastair Khodabukus
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
35
|
Mournetas V, Massouridès E, Dupont JB, Kornobis E, Polvèche H, Jarrige M, Dorval ARL, Gosselin MRF, Manousopoulou A, Garbis SD, Górecki DC, Pinset C. Myogenesis modelled by human pluripotent stem cells: a multi-omic study of Duchenne myopathy early onset. J Cachexia Sarcopenia Muscle 2021; 12:209-232. [PMID: 33586340 PMCID: PMC7890274 DOI: 10.1002/jcsm.12665] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/25/2020] [Accepted: 12/16/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) causes severe disability of children and death of young men, with an incidence of approximately 1/5000 male births. Symptoms appear in early childhood, with a diagnosis made mostly around 4 years old, a time where the amount of muscle damage is already significant, preventing early therapeutic interventions that could be more efficient at halting disease progression. In the meantime, the precise moment at which disease phenotypes arise-even asymptomatically-is still unknown. Thus, there is a critical need to better define DMD onset as well as its first manifestations, which could help identify early disease biomarkers and novel therapeutic targets. METHODS We have used both human tissue-derived myoblasts and human induced pluripotent stem cells (hiPSCs) from DMD patients to model skeletal myogenesis and compared their differentiation dynamics with that of healthy control cells by a comprehensive multi-omic analysis at seven time points. Results were strengthened with the analysis of isogenic CRISPR-edited human embryonic stem cells and through comparisons against published transcriptomic and proteomic datasets from human DMD muscles. The study was completed with DMD knockdown/rescue experiments in hiPSC-derived skeletal muscle progenitor cells and adenosine triphosphate measurement in hiPSC-derived myotubes. RESULTS Transcriptome and miRnome comparisons combined with protein analyses demonstrated that hiPSC differentiation (i) leads to embryonic/foetal myotubes that mimic described DMD phenotypes at the differentiation endpoint and (ii) homogeneously and robustly recapitulates key developmental steps-mesoderm, somite, and skeletal muscle. Starting at the somite stage, DMD dysregulations concerned almost 10% of the transcriptome. These include mitochondrial genes whose dysregulations escalate during differentiation. We also describe fibrosis as an intrinsic feature of DMD skeletal muscle cells that begins early during myogenesis. All the omics data are available online for exploration through a graphical interface at https://muscle-dmd.omics.ovh/. CONCLUSIONS Our data argue for an early developmental manifestation of DMD whose onset is triggered before the entry into the skeletal muscle compartment, data leading to a necessary reconsideration of dystrophin roles during muscle development. This hiPSC model of skeletal muscle differentiation offers the possibility to explore these functions as well as find earlier DMD biomarkers and therapeutic targets.
Collapse
Affiliation(s)
| | | | | | - Etienne Kornobis
- Biomics, C2RT, Institut Pasteur, Paris, France.,Hub de Bioinformatique et Biostatistique - Département BiologieComputationnelle, Paris, France
| | | | | | | | - Maxime R F Gosselin
- Molecular Medicine, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Antigoni Manousopoulou
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Spiros D Garbis
- Unit for Cancer Sciences, Centre for Proteomics Research, Institute for Life Sciences, University of Southampton, Southampton, UK.,Proteas Bioanalytics Inc., BioLabs at The Lundquist Institute, Torrance, CA, USA
| | - Dariusz C Górecki
- Molecular Medicine, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK.,Military Institute of Hygiene and Epidemiology, Warsaw, Poland
| | | |
Collapse
|
36
|
Rugowska A, Starosta A, Konieczny P. Epigenetic modifications in muscle regeneration and progression of Duchenne muscular dystrophy. Clin Epigenetics 2021; 13:13. [PMID: 33468200 PMCID: PMC7814631 DOI: 10.1186/s13148-021-01001-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/14/2020] [Indexed: 02/08/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a multisystemic disorder that affects 1:5000 boys. The severity of the phenotype varies dependent on the mutation site in the DMD gene and the resultant dystrophin expression profile. In skeletal muscle, dystrophin loss is associated with the disintegration of myofibers and their ineffective regeneration due to defective expansion and differentiation of the muscle stem cell pool. Some of these phenotypic alterations stem from the dystrophin absence-mediated serine-threonine protein kinase 2 (MARK2) misplacement/downregulation in activated muscle stem (satellite) cells and neuronal nitric oxide synthase loss in cells committed to myogenesis. Here, we trace changes in DNA methylation, histone modifications, and expression of regulatory noncoding RNAs during muscle regeneration, from the stage of satellite cells to myofibers. Furthermore, we describe the abrogation of these epigenetic regulatory processes due to changes in signal transduction in DMD and point to therapeutic treatments increasing the regenerative potential of diseased muscles based on this acquired knowledge.
Collapse
Affiliation(s)
- Anna Rugowska
- Institute of Human Biology and Evolution, Faculty of Biology, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 6, 61-614, Poznan, Poland
| | - Alicja Starosta
- Institute of Human Biology and Evolution, Faculty of Biology, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 6, 61-614, Poznan, Poland
| | - Patryk Konieczny
- Institute of Human Biology and Evolution, Faculty of Biology, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 6, 61-614, Poznan, Poland.
| |
Collapse
|
37
|
Gupta D, Santoso JW, McCain ML. Characterization of Gelatin Hydrogels Cross-Linked with Microbial Transglutaminase as Engineered Skeletal Muscle Substrates. Bioengineering (Basel) 2021; 8:bioengineering8010006. [PMID: 33418892 PMCID: PMC7825108 DOI: 10.3390/bioengineering8010006] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/18/2020] [Accepted: 12/30/2020] [Indexed: 12/12/2022] Open
Abstract
Engineered in vitro models of skeletal muscle are essential for efficiently screening drug safety and efficacy. However, conventional culture substrates poorly replicate physical features of native muscle and do not support long-term culture, which limits tissue maturity. Micromolded gelatin hydrogels cross-linked with microbial transglutaminase (gelatin-MTG hydrogels) have previously been shown to induce C21C2 myotube alignment and improve culture longevity. However, several properties of gelatin-MTG hydrogels have not been systematically characterized, such as changes in elastic modulus during incubation in culture-like conditions and their ability to support sarcomere maturation. In this study, various gelatin-MTG hydrogels were fabricated and incubated in ambient or culture-like conditions. Elastic modulus, mass, and transmittance were measured over a one- or two-week period. Compared to hydrogels in phosphate buffered saline (PBS) or ambient air, hydrogels in Dulbecco’s Modified Eagle Medium (DMEM) and 5% CO2 demonstrated the most stable elastic modulus. A subset of gelatin-MTG hydrogels was micromolded and seeded with C2C12 or primary chick myoblasts, which aligned and fused into multinucleated myotubes with relatively mature sarcomeres. These data are important for fabricating gelatin-MTG hydrogels with predictable and stable mechanical properties and highlight their advantages as culture substrates for engineering relatively mature and stable muscle tissues.
Collapse
Affiliation(s)
- Divya Gupta
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, 1042 Downey Way, DRB 140, Los Angeles, CA 90089, USA; (D.G.); (J.W.S.)
| | - Jeffrey W. Santoso
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, 1042 Downey Way, DRB 140, Los Angeles, CA 90089, USA; (D.G.); (J.W.S.)
| | - Megan L. McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, 1042 Downey Way, DRB 140, Los Angeles, CA 90089, USA; (D.G.); (J.W.S.)
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, 1975 Zonal Ave, Los Angeles, CA 90033, USA
- Correspondence:
| |
Collapse
|
38
|
Modelling Neuromuscular Diseases in the Age of Precision Medicine. J Pers Med 2020; 10:jpm10040178. [PMID: 33080928 PMCID: PMC7712305 DOI: 10.3390/jpm10040178] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 12/20/2022] Open
Abstract
Advances in knowledge resulting from the sequencing of the human genome, coupled with technological developments and a deeper understanding of disease mechanisms of pathogenesis are paving the way for a growing role of precision medicine in the treatment of a number of human conditions. The goal of precision medicine is to identify and deliver effective therapeutic approaches based on patients’ genetic, environmental, and lifestyle factors. With the exception of cancer, neurological diseases provide the most promising opportunity to achieve treatment personalisation, mainly because of accelerated progress in gene discovery, deep clinical phenotyping, and biomarker availability. Developing reproducible, predictable and reliable disease models will be key to the rapid delivery of the anticipated benefits of precision medicine. Here we summarize the current state of the art of preclinical models for neuromuscular diseases, with particular focus on their use and limitations to predict safety and efficacy treatment outcomes in clinical trials.
Collapse
|
39
|
Jensen JH, Cakal SD, Li J, Pless CJ, Radeke C, Jepsen ML, Jensen TE, Dufva M, Lind JU. Large-scale spontaneous self-organization and maturation of skeletal muscle tissues on ultra-compliant gelatin hydrogel substrates. Sci Rep 2020; 10:13305. [PMID: 32764726 PMCID: PMC7411013 DOI: 10.1038/s41598-020-69936-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 07/15/2020] [Indexed: 11/09/2022] Open
Abstract
Cellular self-organization is the fundamental driving force behind the complex architectures of native tissue. Yet, attempts at replicating native tissue architectures in vitro often involve complex micro-fabrication methods and materials. While impressive progress has been made within engineered models of striated muscle, the wide adaptation of these models is held back by the need for specific tools and knowhow. In this report, we show that C2C12 myoblasts spontaneously organize into highly aligned myotube tissues on the mm to cm scale, when cultured on sufficiently soft yet fully isotropic gelatin hydrogel substrates. Interestingly, we only observed this phenomenon for hydrogels with Young’s modulus of 6 kPa and below. For slightly more rigid compositions, only local micrometer-scale myotube organization was observed, similar to that seen in conventional polystyrene dishes. The hydrogel-supported myotubes could be cultured for multiple weeks and matured into highly contractile phenotypes with notable upregulation of myosin heavy chain, as compared to myotubes developed in conventional petri dishes. The procedure for casting the ultra-soft gelatin hydrogels is straight forward and compatible with standardized laboratory tools. It may thus serve as a simple, yet versatile, approach to generating skeletal muscle tissue of improved physiological relevance for applied and basic research.
Collapse
Affiliation(s)
- Joen H Jensen
- Department of Health Technology, Technical University of Denmark, Building 423, 2800, Kgs. Lyngby, Denmark
| | - Selgin D Cakal
- Department of Health Technology, Technical University of Denmark, Building 423, 2800, Kgs. Lyngby, Denmark
| | - Jingwen Li
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, 2100, København Ø, Denmark
| | - Christian J Pless
- Department of Health Technology, Technical University of Denmark, Building 423, 2800, Kgs. Lyngby, Denmark
| | - Carmen Radeke
- Department of Health Technology, Technical University of Denmark, Building 423, 2800, Kgs. Lyngby, Denmark
| | - Morten Leth Jepsen
- Department of Health Technology, Technical University of Denmark, Building 423, 2800, Kgs. Lyngby, Denmark.,The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Technical University of Denmark, Building 423, 2800, Kgs. Lyngby, Denmark
| | - Thomas E Jensen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, 2100, København Ø, Denmark
| | - Martin Dufva
- Department of Health Technology, Technical University of Denmark, Building 423, 2800, Kgs. Lyngby, Denmark. .,The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Technical University of Denmark, Building 423, 2800, Kgs. Lyngby, Denmark.
| | - Johan U Lind
- Department of Health Technology, Technical University of Denmark, Building 423, 2800, Kgs. Lyngby, Denmark.
| |
Collapse
|
40
|
Ariyasinghe NR, Santoso JW, Gupta D, Pincus MJ, August PR, McCain ML. Optical Clearing of Skeletal Muscle Bundles Engineered in 3-D Printed Templates. Ann Biomed Eng 2020; 49:523-535. [PMID: 32748107 DOI: 10.1007/s10439-020-02583-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 07/22/2020] [Indexed: 01/23/2023]
Abstract
Many techniques for engineering and interrogating three-dimensional (3-D) muscle bundles from animal- or patient-derived myoblasts have recently been developed to overcome the limitations of existing in vitro and in vivo model systems. However, many approaches for engineering 3-D muscle bundles rely on specialized and time-consuming techniques, such as photolithography for fabrication and cryosectioning for histology. Cryosectioning also limits visualization to a single plane instead of the entire 3-D structure. To address these challenges, we first implemented a consumer-grade 3-D-printer to rapidly prototype multiple templates for engineering muscle bundles. We then employed our templates to engineer 3D muscle bundles and identify template geometries that promoted bundle survival over three weeks. Subsequently, we implemented tissue clearing, immunostaining, and confocal imaging to acquire z-stacks of intact muscle bundles labelled for myogenic markers. With this approach, we could select the imaging plane on-demand and visualize the intact 3-D structure of bundles. However, tissue clearing did cause some tissue degradation that should be considered. Together, these advances in muscle tissue engineering and imaging will accelerate the use of these 3-D tissue platforms for disease modeling and therapeutic discovery.
Collapse
Affiliation(s)
- Nethika R Ariyasinghe
- Smidt Heart Institute, Cedars-Sinai Medical Center, 127 San Vicente Blvd, AHSP A9228, Los Angeles, CA, 90048, USA.,Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, 1042 Downey Way, DRB 140, Los Angeles, CA, 90089, USA.,Icagen, 2090 E. Innovation Park Dr, Oro Valley, AZ, 85755, USA
| | - Jeffrey W Santoso
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, 1042 Downey Way, DRB 140, Los Angeles, CA, 90089, USA
| | - Divya Gupta
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, 1042 Downey Way, DRB 140, Los Angeles, CA, 90089, USA
| | - Mark J Pincus
- Icagen, 2090 E. Innovation Park Dr, Oro Valley, AZ, 85755, USA.,Department of Science & CTE, Ironwood Ridge High School, 2475 W Naranja Dr, Oro Valley, AZ, 85742, USA
| | - Paul R August
- Icagen, 2090 E. Innovation Park Dr, Oro Valley, AZ, 85755, USA.,Agios Pharmaceuticals, 88 Sidney Street, Cambridge, MA, 02139, USA
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, 1042 Downey Way, DRB 140, Los Angeles, CA, 90089, USA. .,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, 1975 Zonal Ave, Los Angeles, CA, 90033, USA.
| |
Collapse
|
41
|
Rexius-Hall ML, Khalil NN, Andres AM, McCain ML. Mitochondrial division inhibitor 1 (mdivi-1) increases oxidative capacity and contractile stress generated by engineered skeletal muscle. FASEB J 2020; 34:11562-11576. [PMID: 32652761 DOI: 10.1096/fj.201901039rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/03/2020] [Accepted: 06/08/2020] [Indexed: 12/16/2022]
Abstract
In skeletal muscle fibers, mitochondria are densely packed adjacent to myofibrils because adenosine triphosphate (ATP) is needed to fuel sarcomere shortening. However, despite this close physical and biochemical relationship, the effects of mitochondrial dynamics on skeletal muscle contractility are poorly understood. In this study, we analyzed the effects of Mitochondrial Division Inhibitor 1 (mdivi-1), an inhibitor of mitochondrial fission, on the structure and function of both mitochondria and myofibrils in skeletal muscle tissues engineered on micromolded gelatin hydrogels. Treatment with mdivi-1 did not alter myotube morphology, but did increase the mitochondrial turbidity and oxidative capacity, consistent with reduced mitochondrial fission. Mdivi-1 also significantly increased basal, twitch, and tetanus stresses, as measured using the Muscular Thin Film (MTF) assay. Finally, mdivi-1 increased sarcomere length, potentially due to mdivi-1-induced changes in mitochondrial volume and compression of myofibrils. Together, these results suggest that mdivi-1 increases contractile stress generation, which may be caused by an increase in maximal respiration and/or sarcomere length due to increased volume of individual mitochondria. These data reinforce that mitochondria have both biochemical and biomechanical roles in skeletal muscle and that mitochondrial dynamics can be manipulated to alter muscle contractility.
Collapse
Affiliation(s)
- Megan L Rexius-Hall
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Natalie N Khalil
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Allen M Andres
- Smidt Heart Institute and Barbra Streisand Women's Heart Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
42
|
Abstract
Organs-on-chips are broadly defined as microfabricated surfaces or devices designed to engineer cells into microscale tissues with native-like features and then extract physiologically relevant readouts at scale. Because they are generally compatible with patient-derived cells, these technologies can address many of the human relevance limitations of animal models. As a result, organs-on-chips have emerged as a promising new paradigm for patient-specific disease modeling and drug development. Because neuromuscular diseases span a broad range of rare conditions with diverse etiology and complex pathophysiology, they have been especially challenging to model in animals and thus are well suited for organ-on-chip approaches. In this Review, we first briefly summarize the challenges in neuromuscular disease modeling with animal models. Next, we describe a variety of existing organ-on-chip approaches for neuromuscular tissues, including a survey of cell sources for both muscle and nerve, and two- and three-dimensional neuromuscular tissue-engineering techniques. Although researchers have made tremendous advances in modeling neuromuscular diseases on a chip, the remaining challenges in cell sourcing, cell maturity, tissue assembly and readout capabilities limit their integration into the drug development pipeline today. However, as the field advances, models of healthy and diseased neuromuscular tissues on a chip, coupled with animal models, have vast potential as complementary tools for modeling multiple aspects of neuromuscular diseases and identifying new therapeutic strategies. Summary: Modeling neuromuscular diseases is challenging due to their complex etiology and pathophysiology. Here, we review the cell sources and tissue-engineering procedures that are being integrated as emerging neuromuscular disease models.
Collapse
Affiliation(s)
- Jeffrey W Santoso
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA .,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
43
|
O’Connor BB, Pope BD, Peters MM, Ris-Stalpers C, Parker KK. The role of extracellular matrix in normal and pathological pregnancy: Future applications of microphysiological systems in reproductive medicine. Exp Biol Med (Maywood) 2020; 245:1163-1174. [PMID: 32640894 PMCID: PMC7400725 DOI: 10.1177/1535370220938741] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
IMPACT STATEMENT Extracellular matrix in the womb regulates the initiation, progression, and completion of a healthy pregnancy. The composition and physical properties of extracellular matrix in the uterus and at the maternal-fetal interface are remodeled at each gestational stage, while maladaptive matrix remodeling results in obstetric disease. As in vitro models of uterine and placental tissues, including micro-and milli-scale versions of these organs on chips, are developed to overcome the inherent limitations of studying human development in vivo, we can isolate the influence of cellular and extracellular components in healthy and pathological pregnancies. By understanding and recreating key aspects of the extracellular microenvironment at the maternal-fetal interface, we can engineer microphysiological systems to improve assisted reproduction, obstetric disease treatment, and prenatal drug safety.
Collapse
Affiliation(s)
- Blakely B O’Connor
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering; Harvard John A. Paulson School of Engineering and Applied Sciences; Harvard University, Cambridge, MA 02138, USA
| | - Benjamin D Pope
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering; Harvard John A. Paulson School of Engineering and Applied Sciences; Harvard University, Cambridge, MA 02138, USA
| | - Michael M Peters
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering; Harvard John A. Paulson School of Engineering and Applied Sciences; Harvard University, Cambridge, MA 02138, USA
| | - Carrie Ris-Stalpers
- Department of Gynecology and Obstetrics, Academic Reproduction and Development, Amsterdam UMC, University of Amsterdam, Amsterdam 1105, The Netherlands
| | - Kevin K Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering; Harvard John A. Paulson School of Engineering and Applied Sciences; Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
44
|
Actomyosin contractility scales with myoblast elongation and enhances differentiation through YAP nuclear export. Sci Rep 2019; 9:15565. [PMID: 31664178 PMCID: PMC6820726 DOI: 10.1038/s41598-019-52129-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 10/10/2019] [Indexed: 01/14/2023] Open
Abstract
Skeletal muscle fibers are formed by the fusion of mononucleated myoblasts into long linear myotubes, which differentiate and reorganize into multinucleated myofibers that assemble in bundles to form skeletal muscles. This fundamental process requires the elongation of myoblasts into a bipolar shape, although a complete understanding of the mechanisms governing skeletal muscle fusion is lacking. To address this question, we consider cell aspect ratio, actomyosin contractility and the Hippo pathway member YAP as potential regulators of the fusion of myoblasts into myotubes. Using fibronectin micropatterns of different geometries and traction force microscopy, we investigated how myoblast elongation affects actomyosin contractility. Our findings indicate that cell elongation enhances actomyosin contractility in myoblasts, which regulate their actin network to their spreading area. Interestingly, we found that the contractility of cell pairs increased after their fusion and raise on elongated morphologies. Furthermore, our findings indicate that myoblast elongation modulates nuclear orientation and triggers cytoplasmic localization of YAP, increasing evidence that YAP is a key regulator of mechanotransduction in myoblasts. Taken together, our findings support a mechanical model where actomyosin contractility scales with myoblast elongation and enhances the differentiation of myoblasts into myotubes through YAP nuclear export.
Collapse
|
45
|
Muscle tissue engineering in fibrous gelatin: implications for meat analogs. NPJ Sci Food 2019; 3:20. [PMID: 31646181 PMCID: PMC6803664 DOI: 10.1038/s41538-019-0054-8] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 08/16/2019] [Indexed: 12/17/2022] Open
Abstract
Bioprocessing applications that derive meat products from animal cell cultures require food-safe culture substrates that support volumetric expansion and maturation of adherent muscle cells. Here we demonstrate scalable production of microfibrous gelatin that supports cultured adherent muscle cells derived from cow and rabbit. As gelatin is a natural component of meat, resulting from collagen denaturation during processing and cooking, our extruded gelatin microfibers recapitulated structural and biochemical features of natural muscle tissues. Using immersion rotary jet spinning, a dry-jet wet-spinning process, we produced gelatin fibers at high rates (~ 100 g/h, dry weight) and, depending on process conditions, we tuned fiber diameters between ~ 1.3 ± 0.1 μm (mean ± SEM) and 8.7 ± 1.4 μm (mean ± SEM), which are comparable to natural collagen fibers. To inhibit fiber degradation during cell culture, we crosslinked them either chemically or by co-spinning gelatin with a microbial crosslinking enzyme. To produce meat analogs, we cultured bovine aortic smooth muscle cells and rabbit skeletal muscle myoblasts in gelatin fiber scaffolds, then used immunohistochemical staining to verify that both cell types attached to gelatin fibers and proliferated in scaffold volumes. Short-length gelatin fibers promoted cell aggregation, whereas long fibers promoted aligned muscle tissue formation. Histology, scanning electron microscopy, and mechanical testing demonstrated that cultured muscle lacked the mature contractile architecture observed in natural muscle but recapitulated some of the structural and mechanical features measured in meat products.
Collapse
|
46
|
Wang J, Khodabukus A, Rao L, Vandusen K, Abutaleb N, Bursac N. Engineered skeletal muscles for disease modeling and drug discovery. Biomaterials 2019; 221:119416. [PMID: 31419653 DOI: 10.1016/j.biomaterials.2019.119416] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 08/01/2019] [Accepted: 08/05/2019] [Indexed: 01/04/2023]
Abstract
Skeletal muscle is the largest organ of human body with several important roles in everyday movement and metabolic homeostasis. The limited ability of small animal models of muscle disease to accurately predict drug efficacy and toxicity in humans has prompted the development in vitro models of human skeletal muscle that fatefully recapitulate cell and tissue level functions and drug responses. We first review methods for development of three-dimensional engineered muscle tissues and organ-on-a-chip microphysiological systems and discuss their potential utility in drug discovery research and development of new regenerative therapies. Furthermore, we describe strategies to increase the functional maturation of engineered muscle, and motivate the importance of incorporating multiple tissue types on the same chip to model organ cross-talk and generate more predictive drug development platforms. Finally, we review the ability of available in vitro systems to model diseases such as type II diabetes, Duchenne muscular dystrophy, Pompe disease, and dysferlinopathy.
Collapse
Affiliation(s)
- Jason Wang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Lingjun Rao
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Keith Vandusen
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nadia Abutaleb
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
47
|
Wang Y, Hao L, Li H, Cleary JD, Tomac MP, Thapa A, Guo X, Zeng D, Wang H, McRae M, Jastrzemski O, Smith-Fassler AM, Xu Y, Xia G. Abnormal nuclear aggregation and myotube degeneration in myotonic dystrophy type 1. Neurol Sci 2019; 40:1255-1265. [PMID: 30891637 DOI: 10.1007/s10072-019-03783-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 02/20/2019] [Indexed: 12/21/2022]
Abstract
Myotonic dystrophy type 1 (DM1) is caused by CTG nucleotide repeat expansions in the 3'-untranslated region (3'-UTR) of the dystrophia myotonica protein kinase (DMPK) gene. The expanded CTG repeats encode toxic CUG RNAs that cause disease, largely through RNA gain-of-function. DM1 is a fatal disease characterized by progressive muscle wasting, which has no cure. Regenerative medicine has emerged as a promising therapeutic modality for DM1, especially with the advancement of induced pluripotent stem (iPS) cell technology and therapeutic genome editing. However, there is an unmet need to identify in vitro outcome measures to demonstrate the therapeutic effects prior to in vivo clinical trials. In this study, we examined the muscle regeneration (myotube formation) in normal and DM1 myoblasts in vitro to establish outcome measures for therapeutic monitoring. We found normal proliferation of DM1 myoblasts, but abnormal nuclear aggregation during the early stage myotube formation, as well as myotube degeneration during the late stage of myotube formation. We concluded that early abnormal nuclear aggregation and late myotube degeneration offer easy and sensitive outcome measures to monitor therapeutic effects in vitro.
Collapse
Affiliation(s)
- Yanlin Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan, 450000, China
| | - Lei Hao
- Department of Neurology, The Fifth People's Hospital of Chongqing, Chongqing, 400062, China
| | - Hui Li
- Department of Neurology, University of Florida, Gainesville, FL, USA
| | - John D Cleary
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, USA
| | - Michael P Tomac
- Department of Neurology and Neuroscience, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Arjun Thapa
- Department of Neurology and Neuroscience, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Xiuming Guo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Desmond Zeng
- Department of Neurology, University of Florida, Gainesville, FL, USA
| | - Hongcai Wang
- Department of Neurology and Neuroscience, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - MacKezie McRae
- Department of Neurology and Neuroscience, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Olivia Jastrzemski
- Department of Neurology and Neuroscience, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Ali Marichen Smith-Fassler
- Department of Neurology and Neuroscience, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan, 450000, China.
| | - Guangbin Xia
- Department of Neurology and Neuroscience, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA.
| |
Collapse
|
48
|
Truskey GA. Development and application of human skeletal muscle microphysiological systems. LAB ON A CHIP 2018; 18:3061-3073. [PMID: 30183050 PMCID: PMC6177290 DOI: 10.1039/c8lc00553b] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
A number of major disease states involve skeletal muscle, including type 2 diabetes, muscular dystrophy, sarcopenia and cachexia arising from cancer or heart disease. Animals do not accurately represent many of these disease states. Human skeletal muscle microphysiological systems derived from primary or induced pluripotent stem cells (hPSCs) can provide an in vitro model of genetic and chronic diseases and assess individual variations. Three-dimensional culture systems more accurately represent skeletal muscle function than do two-dimensional cultures. While muscle biopsies enable culture of primary muscle cells, hPSCs provide the opportunity to sample a wider population of donors. Recent advances to promote maturation of PSC-derived skeletal muscle provide an alternative to primary cells. While contractile function is often measured in three-dimensional cultures and several systems exist to characterize contraction of small numbers of muscle fibers, there is a need for functional measures of metabolism suited for microphysiological systems. Future research should address generation of well-differentiated hPSC-derived muscle cells, enabling muscle repair in vitro, and improved disease models.
Collapse
Affiliation(s)
- George A Truskey
- Department of Biomedical Engineering, Duke University, 1427 CIEMAS, 101 Science Drive, Durham, NC 27708-0281, USA.
| |
Collapse
|
49
|
Takahashi H, Shimizu T, Okano T. Engineered Human Contractile Myofiber Sheets as a Platform for Studies of Skeletal Muscle Physiology. Sci Rep 2018; 8:13932. [PMID: 30224737 PMCID: PMC6141563 DOI: 10.1038/s41598-018-32163-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 09/03/2018] [Indexed: 12/25/2022] Open
Abstract
Skeletal muscle physiology and the mechanisms of muscle diseases can be effectively studied by an in-vitro tissue model produced by muscle tissue engineering. Engineered human cell-based tissues are required more than ever because of the advantages they bring as tissue models in research studies. This study reports on a production method of a human skeletal myofiber sheet that demonstrates biomimetic properties including the aligned structure of myofibers, basement membrane-like structure of the extracellular matrix, and unidirectional contractile ability. The contractile ability and drug responsibility shown in this study indicate that this engineered muscle tissue has potential as a human cell-based tissue model for clinically relevant in-vitro studies in muscle physiology and drug discovery. Moreover, this engineered tissue can be used to better understand the relationships between mechanical stress and myogenesis, including muscle growth and regeneration. In this study, periodic exercise induced by continuous electrical pulse stimulation enhanced the contractile ability of the engineered myofibers and the secretion of interleukin-6 (IL-6) and vascular endothelial growth factor (VEGF) from the exercising myofibers. Since the physiology of skeletal muscle is directly related to mechanical stress, these features point to application as a tissue model and platform for future biological studies of skeletal muscle including muscle metabolism, muscle atrophy and muscle regeneration.
Collapse
Affiliation(s)
- Hironobu Takahashi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan.
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
50
|
Zhang X, Hong S, Yen R, Kondash M, Fernandez CE, Truskey GA. A system to monitor statin-induced myopathy in individual engineered skeletal muscle myobundles. LAB ON A CHIP 2018; 18:2787-2796. [PMID: 30112530 PMCID: PMC6145090 DOI: 10.1039/c8lc00654g] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Microphysiological tissue engineering models of human skeletal muscle (myobundles) provide a platform to investigate the mechanism of muscle diseases and to study the response to drugs and toxins in vitro. To examine the dynamic response to drugs, which often take several days to induce responses, we developed a system to monitor the contractile force of the same human skeletal muscle myobundles over time before and after treatment with drugs. Myobundles were formed in series with Ecoflex films (platinum-catalyzed silicones) with embedded microbeads. The displacement of the microbeads in Ecoflex exhibited a linear relation between muscle force production and Ecoflex film stretch. Forces measured with the microbeads embedded in Ecoflex agreed well with simultaneous measurements with a force transducer. Application of the Hill model for the myobundles showed that the Ecoflex affected the magnitude of the response, but not the kinetics. After continuous exposure to 100 nM cerivastatin, both active and passive forces were reduced relative to controls after 2-4 days. The decline in force was associated with a decline in the muscle myofiber organization. The inhibitory effect of cerivastatin was reduced when 0.1-1 mM mevalonate was added with cerivastatin. Although addition of co-enzyme Q10 with cerivastatin inhibited degradation of sarcomeric α-actinin (SAA) in myoblasts, the contractile force still declined, suggesting that statin-induced myopathy was related to mevalonate pathway but the addition of co-enzyme Q10 was insufficient to overcome the effect of statins on the mevalonate pathway. Thus, cerivastatin rapidly induces myopathy which can be reversds with mevalonate but not co-enzyme Q10.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA.
| | | | | | | | | | | |
Collapse
|