1
|
Galindo AN, Chi AK, Liashenko I, O’Neill KL, Sharma R, Khachatourian JD, Hajarizadeh A, Dalton PD, Hettiaratchi MH. Hyaluronic Acid-Coated Melt Electrowritten Scaffolds Promote Myoblast Attachment, Alignment, and Differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.06.641880. [PMID: 40161586 PMCID: PMC11952302 DOI: 10.1101/2025.03.06.641880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Purpose In muscle tissues, anisotropic cell alignment is essential for optimal muscle fiber development and function. Biomaterials for muscle tissue engineering must guide cellular alignment while supporting cell proliferation and myogenic differentiation. Methods Here, we describe the fabrication of a tissue-engineered construct consisting of a scaffold of aligned poly(ε-caprolactone) (PCL) microfibers coated in a dynamic covalent hydrazone crosslinked hyaluronic acid (HA) hydrogel to support myoblast attachment, alignment, and differentiation. Norbornene modification of HA further enabled functionalization with fibronectin-derived arginine-glycine-aspartic acid (RGD) peptide. Scaffolds were fabricated using melt electrowriting (MEW), a three-dimensional (3D)-printing technique that uses stabilization of fluid columns to produce precisely aligned polymeric microfibers. We evaluated scaffolds with fiber diameters of 10 μm, 20 μm, and 30 μm of non-coated, HA-coated, and HA-RGD-coated MEW scaffolds through immunocytochemistry and creatine kinase activity assays. Results HA-coated and HA-RGD-coated scaffolds showed increased cellular attachment of C2C12 mouse skeletal myoblasts on all fiber diameters compared to non-coated scaffolds, with HA-RGD-coated scaffolds demonstrating the highest cell attachment. All scaffolds supported cellular alignment along the fibers. Cells differentiated on scaffolds showed anisotropic alignment with increased myotube formation on HA-RGD-coated scaffolds as seen by myosin heavy chain (MHC) staining. Highest creatine kinase (CK) activity on day 5 signified the successful differentiation of C2C12 cells into mature myotubes. Conclusion This unique combination of tunable biophysical and biochemical cues enables the creation of a biomimetic tissue engineered scaffold, providing a platform for new therapeutic approaches for muscle regeneration.
Collapse
Affiliation(s)
- Alycia N. Galindo
- Department of Bioengineering, Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon
| | - Alyssa K. Chi
- Department of Bioengineering, Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon
- Department of Chemistry and Biochemistry, University of Oregon
| | - Ievgenii Liashenko
- Department of Bioengineering, Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon
| | - Kelly L. O’Neill
- Department of Bioengineering, Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon
| | - Ruchi Sharma
- Department of Bioengineering, Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon
| | - Jenna D. Khachatourian
- Department of Bioengineering, Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon
- Department of Human Physiology, University of Oregon
| | - Armaan Hajarizadeh
- Department of Bioengineering, Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon
- Department of Computer and Data Sciences, University of Oregon
| | - Paul D. Dalton
- Department of Bioengineering, Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon
| | - Marian H. Hettiaratchi
- Department of Bioengineering, Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon
- Department of Chemistry and Biochemistry, University of Oregon
| |
Collapse
|
2
|
Lee HS, Samolyk BL, Pins GD. Extrusion-Based Printing of Myoblast-Loaded Fibrin Microthreads to Induce Myogenesis. J Funct Biomater 2025; 16:21. [PMID: 39852577 PMCID: PMC11765554 DOI: 10.3390/jfb16010021] [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: 09/30/2024] [Revised: 12/19/2024] [Accepted: 01/04/2025] [Indexed: 01/26/2025] Open
Abstract
Large skeletal muscle injuries such as volumetric muscle loss (VML) disrupt native tissue structures, including biophysical and biochemical signaling cues that promote the regeneration of functional skeletal muscle. Various biofabrication strategies have been developed to create engineered skeletal muscle constructs that mimic native matrix and cellular microenvironments to enhance muscle regeneration; however, there remains a need to create scalable engineered tissues that provide mechanical stability as well as structural and spatiotemporal signaling cues to promote cell-mediated regeneration of contractile skeletal muscle. We describe a novel strategy for bioprinting multifunctional myoblast-loaded fibrin microthreads (myothreads) that recapitulate the cellular microniches to drive myogenesis and aligned myotube formation. We characterized myoblast alignment, myotube formation, and tensile properties of myothreads as a function of cell-loading density and culture time. We showed that increasing myoblast loading densities enhances myotube formation. Additionally, alignment analyses indicate that the bioprinting process confers myoblast alignment in the constructs. Finally, tensile characterizations suggest that myothreads possess the structural stability to serve as a potential platform for developing scalable muscle scaffolds. We anticipate that our myothread biofabrication approach will enable us to strategically investigate biophysical and biochemical signaling cues and cellular mechanisms that enhance functional skeletal muscle regeneration for the treatment of VML.
Collapse
Affiliation(s)
| | | | - George D. Pins
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA 01609, USA; (H.S.L.); (B.L.S.)
| |
Collapse
|
3
|
Jo B, Motoi K, Morimoto Y, Takeuchi S. Dynamic and Static Workout of In Vitro Skeletal Muscle Tissue through a Weight Training Device. Adv Healthc Mater 2024; 13:e2401844. [PMID: 39212188 PMCID: PMC11670278 DOI: 10.1002/adhm.202401844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/28/2024] [Indexed: 09/04/2024]
Abstract
Enhancing muscle strength through workouts is a key factor in improving physical activity and maintaining metabolic profiles. The controversial results concerning the impacts of weight training often arise from clinical experiments that require controlled experimental conditions. In this study, a weight training system for a muscle development model is presented, which is capable of performing weight training motions with adjustable weight loads. Through the implementation of cultured skeletal muscle tissue with floating structures and a flexible ribbon, both isotonic (dynamic change in muscle length) and isometric (static in muscle length) exercises can be performed without the deflection of the tissue. Quantitative analysis of contraction force, changes in metabolic processes, and muscle morphology under different weight training conditions demonstrates the effectiveness of the proposed system. Our proposed system holds potential for establishing effective muscle development and for further applications in rehabilitation training methods.
Collapse
Affiliation(s)
- Byeongwook Jo
- Mechano‐InformaticsGraduate School of Information Science and TechnologyThe University of Tokyo7‐3‐1 Hongo, Bunkyo‐kuTokyo113‐8656Japan
| | - Kentaro Motoi
- Mechano‐InformaticsGraduate School of Information Science and TechnologyThe University of Tokyo7‐3‐1 Hongo, Bunkyo‐kuTokyo113‐8656Japan
| | - Yuya Morimoto
- Electronic and Physical SystemsSchool of Fundamental Science and EngineeringWaseda University3‐4‐1 Okubo, Shinjuku‐kuTokyo169‐8555Japan
| | - Shoji Takeuchi
- Mechano‐InformaticsGraduate School of Information Science and TechnologyThe University of Tokyo7‐3‐1 Hongo, Bunkyo‐kuTokyo113‐8656Japan
- International Research for Center for Neurointelligence (WPI‐IRCN)The University of Tokyo Institutes for Advanced StudyThe University of Tokyo7‐3‐1 Hongo Bunkyo‐kuTokyo113‐0033Japan
- Institute of Industrial ScienceThe University of Tokyo4‐6‐1 Komaba, Meguro‐kuTokyo153‐8505Japan
| |
Collapse
|
4
|
Kowalczuk K, Dasgupta A, Páez Larios F, Ulrich HF, Wegner V, Brendel JC, Eggeling C, Mosig AS, Schacher FH. Self-Degrading Multifunctional PEG-Based Hydrogels-Tailormade Substrates for Cell Culture. Macromol Biosci 2024; 24:e2300383. [PMID: 38102978 DOI: 10.1002/mabi.202300383] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/11/2023] [Indexed: 12/17/2023]
Abstract
The use of PEG-based hydrogels as cell culture matrix to mimic the natural extracellular matrix (ECM) has been realized using a range of well-defined, tunable, and dynamic scaffolds, although they require cell adhesion ligands such as RGDS-peptide (Arg-Gly-Asp-Ser) to promote cell adhesion. Herein the synthesis of ionic and degradable hydrogels is demonstrated for cell culture by crosslinking [PEG-SH]4 with the zwitterionic crosslinker N,N-bis(acryloxyethyl)-N-methyl-N-(3-sulfopropyl) ammonium betaine (BMSAB) and the cationic crosslinker N,N-bis(acryloxyethyl)-N,N-dimethyl-1-ammonium iodide (BDMAI). Depending on the amount of ionic crosslinker used in gel formation, the hydrogels show tunable gelation time and stiffness. At the same time, the ionic groups act as catalysts for hydrolytic degradation, thereby allowing to define a stability window. The latter could be tailored in a straightforward manner by introducing the non-degradable crosslinker tri(ethylene glycol) divinyl ether. In addition, both ionic crosslinkers favor cell attachment in comparison to the pristine PEG hydrogels. The degradation is examined by swelling behavior, rheology, and fluorescence correlation spectroscopy indicating degradation kinetics depending on diffusion of incorporated fluorescent molecules.
Collapse
Affiliation(s)
- Kathrin Kowalczuk
- Institute of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich-Schiller-University Jena, Lessingstraße 8, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich-Schiller-University Jena, Philosophenweg 7, 07743, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Grüne Aue, D-07754, Jena, Germany
| | - Anindita Dasgupta
- Leibniz Institute of Photonic Technology, Albert-Einstein-Straße 9, 07745, Jena, Germany
- Institute of Applied Optics and Biophysics, Friedrich-Schiller-University Jena, Helmholtzweg 4, 07743, Jena, Germany
| | - Francisco Páez Larios
- Leibniz Institute of Photonic Technology, Albert-Einstein-Straße 9, 07745, Jena, Germany
- Institute of Applied Optics and Biophysics, Friedrich-Schiller-University Jena, Helmholtzweg 4, 07743, Jena, Germany
| | - Hans F Ulrich
- Institute of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich-Schiller-University Jena, Lessingstraße 8, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich-Schiller-University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Valentin Wegner
- Institute of Biochemistry II, Jena University Hospital, Am Nonnenplan 2-4, 07743, Jena, Germany
| | - Johannes C Brendel
- Institute of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich-Schiller-University Jena, Lessingstraße 8, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich-Schiller-University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Christian Eggeling
- Jena Center for Soft Matter (JCSM), Friedrich-Schiller-University Jena, Philosophenweg 7, 07743, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Grüne Aue, D-07754, Jena, Germany
- Leibniz Institute of Photonic Technology, Albert-Einstein-Straße 9, 07745, Jena, Germany
- Institute of Applied Optics and Biophysics, Friedrich-Schiller-University Jena, Helmholtzweg 4, 07743, Jena, Germany
| | - Alexander S Mosig
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Grüne Aue, D-07754, Jena, Germany
- Institute of Biochemistry II, Jena University Hospital, Am Nonnenplan 2-4, 07743, Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Felix H Schacher
- Institute of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich-Schiller-University Jena, Lessingstraße 8, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich-Schiller-University Jena, Philosophenweg 7, 07743, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Grüne Aue, D-07754, Jena, Germany
| |
Collapse
|
5
|
de Barros NR, Darabi MA, Ma X, Diltemiz SE, Ermis M, Hassani Najafabasi A, Nadine S, Banton EA, Mandal K, Abbasgholizadeh R, Falcone N, Mano JF, Nasiri R, Herculano RD, Zhu Y, Ostrovidov S, Lee J, Kim HJ, Hosseini V, Dokmeci MR, Ahadian S, Khademhosseini A. Enhanced Maturation of 3D Bioprinted Skeletal Muscle Tissue Constructs Encapsulating Soluble Factor-Releasing Microparticles. Macromol Biosci 2023; 23:e2300276. [PMID: 37534566 PMCID: PMC10837326 DOI: 10.1002/mabi.202300276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Indexed: 08/04/2023]
Abstract
Several microfabrication technologies have been used to engineer native-like skeletal muscle tissues. However, the successful development of muscle remains a significant challenge in the tissue engineering field. Muscle tissue engineering aims to combine muscle precursor cells aligned within a highly organized 3D structure and biological factors crucial to support cell differentiation and maturation into functional myotubes and myofibers. In this study, the use of 3D bioprinting is proposed for the fabrication of muscle tissues using gelatin methacryloyl (GelMA) incorporating sustained insulin-like growth factor-1 (IGF-1)-releasing microparticles and myoblast cells. This study hypothesizes that functional and mature myotubes will be obtained more efficiently using a bioink that can release IGF-1 sustainably for in vitro muscle engineering. Synthesized microfluidic-assisted polymeric microparticles demonstrate successful adsorption of IGF-1 and sustained release of IGF-1 at physiological pH for at least 21 days. Incorporating the IGF-1-releasing microparticles in the GelMA bioink assisted in promoting the alignment of myoblasts and differentiation into myotubes. Furthermore, the myotubes show spontaneous contraction in the muscle constructs bioprinted with IGF-1-releasing bioink. The proposed bioprinting strategy aims to improve the development of new therapies applied to the regeneration and maturation of muscle tissues.
Collapse
Affiliation(s)
| | - Mohammad Ali Darabi
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Xin Ma
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Sibel Emir Diltemiz
- Department of Chemistry, Eskisehir Technical University, Eskisehir, 26470, Turkey
| | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | | | - Sara Nadine
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
- Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Ethan A. Banton
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | | | - Natashya Falcone
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - João F. Mano
- Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Rohollah Nasiri
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | | | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Serge Ostrovidov
- Department of Radiological Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Junmin Lee
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Han-Jun Kim
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Vahid Hosseini
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Mehmet R. Dokmeci
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Samad Ahadian
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA, 90095, USA
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Radiological Sciences, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Chemical and Biomolecular Engineering, University of California-Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
6
|
Sanchez MM, Bagdasarian IA, Darch W, Morgan JT. Organotypic cultures as aging associated disease models. Aging (Albany NY) 2022; 14:9338-9383. [PMID: 36435511 PMCID: PMC9740367 DOI: 10.18632/aging.204361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/21/2022] [Indexed: 11/24/2022]
Abstract
Aging remains a primary risk factor for a host of diseases, including leading causes of death. Aging and associated diseases are inherently multifactorial, with numerous contributing factors and phenotypes at the molecular, cellular, tissue, and organismal scales. Despite the complexity of aging phenomena, models currently used in aging research possess limitations. Frequently used in vivo models often have important physiological differences, age at different rates, or are genetically engineered to match late disease phenotypes rather than early causes. Conversely, routinely used in vitro models lack the complex tissue-scale and systemic cues that are disrupted in aging. To fill in gaps between in vivo and traditional in vitro models, researchers have increasingly been turning to organotypic models, which provide increased physiological relevance with the accessibility and control of in vitro context. While powerful tools, the development of these models is a field of its own, and many aging researchers may be unaware of recent progress in organotypic models, or hesitant to include these models in their own work. In this review, we describe recent progress in tissue engineering applied to organotypic models, highlighting examples explicitly linked to aging and associated disease, as well as examples of models that are relevant to aging. We specifically highlight progress made in skin, gut, and skeletal muscle, and describe how recently demonstrated models have been used for aging studies or similar phenotypes. Throughout, this review emphasizes the accessibility of these models and aims to provide a resource for researchers seeking to leverage these powerful tools.
Collapse
Affiliation(s)
- Martina M. Sanchez
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| | | | - William Darch
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| | - Joshua T. Morgan
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| |
Collapse
|
7
|
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: 0.7] [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
|
8
|
Khalili MH, Afsar A, Zhang R, Wilson S, Dossi E, Goel S, Impey SA, Aria AI. Thermal response of multi-layer UV crosslinked PEGDA hydrogels. Polym Degrad Stab 2022. [DOI: 10.1016/j.polymdegradstab.2021.109805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
9
|
Araf Y, Galib M, Naser IB, Promon SK. Prospects of 3D Bioprinting as a Possible Treatment for Cancer Cachexia. JOURNAL OF CLINICAL AND EXPERIMENTAL INVESTIGATIONS 2021. [DOI: 10.29333/jcei/11289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
10
|
Abstract
Abstract
In the past few decades, robotics research has witnessed an increasingly high interest in miniaturized, intelligent, and integrated robots. The imperative component of a robot is the actuator that determines its performance. Although traditional rigid drives such as motors and gas engines have shown great prevalence in most macroscale circumstances, the reduction of these drives to the millimeter or even lower scale results in a significant increase in manufacturing difficulty accompanied by a remarkable performance decline. Biohybrid robots driven by living cells can be a potential solution to overcome these drawbacks by benefiting from the intrinsic microscale self-assembly of living tissues and high energy efficiency, which, among other unprecedented properties, also feature flexibility, self-repair, and even multiple degrees of freedom. This paper systematically reviews the development of biohybrid robots. First, the development of biological flexible drivers is introduced while emphasizing on their advantages over traditional drivers. Second, up-to-date works regarding biohybrid robots are reviewed in detail from three aspects: biological driving sources, actuator materials, and structures with associated control methodologies. Finally, the potential future applications and major challenges of biohybrid robots are explored.
Graphic abstract
Collapse
|
11
|
Magarotto F, Sgrò A, Dorigo Hochuli AH, Andreetta M, Grassi M, Saggioro M, Nogara L, Tolomeo AM, Francescato R, Collino F, Germano G, Caicci F, Maghin E, Piccoli M, Jurga M, Blaauw B, Gamba P, Muraca M, Pozzobon M. Muscle functional recovery is driven by extracellular vesicles combined with muscle extracellular matrix in a volumetric muscle loss murine model. Biomaterials 2021; 269:120653. [PMID: 33461058 DOI: 10.1016/j.biomaterials.2021.120653] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 12/26/2020] [Accepted: 01/02/2021] [Indexed: 12/23/2022]
Abstract
Biological scaffolds derived from decellularized tissues are being investigated as a promising approach to repair volumetric muscle losses (VML). Indeed, extracellular matrix (ECM) from decellularized tissues is highly biocompatible and mimics the original tissue. However, the development of fibrosis and the muscle stiffness still represents a major problem. Intercellular signals mediating tissue repair are conveyed via extracellular vesicles (EVs), biologically active nanoparticles secreted by the cells. This work aimed at using muscle ECM and human EVs derived from Wharton Jelly mesenchymal stromal cells (MSC EVs) to boost tissue regeneration in a VML murine model. Mice transplanted with muscle ECM and treated with PBS or MSC EVs were analyzed after 7 and 30 days. Flow cytometry, tissue analysis, qRT-PCR and physiology test were performed. We demonstrated that angiogenesis and myogenesis were enhanced while fibrosis was reduced after EV treatment. Moreover, the inflammation was directed toward tissue repair. M2-like, pro-regenerative macrophages were significantly increased in the MSC EVs treated group compared to control. Strikingly, the histological improvements were associated with enhanced functional recovery. These results suggest that human MSC EVs can be a naturally-derived boost able to ameliorate the efficacy of tissue-specific ECM in muscle regeneration up to the restored tissue function.
Collapse
Affiliation(s)
- Fabio Magarotto
- Stem Cells and Regenerative Medicine Lab, Institute of Pediatric Research Città Della Speranza, Padova, Italy; Department of Women and Children Health, University of Padova, Italy
| | - Alberto Sgrò
- Department of Women and Children Health, University of Padova, Italy
| | | | - Marina Andreetta
- Department of Women and Children Health, University of Padova, Italy
| | - Michele Grassi
- Department of Women and Children Health, University of Padova, Italy
| | - Mattia Saggioro
- Stem Cells and Regenerative Medicine Lab, Institute of Pediatric Research Città Della Speranza, Padova, Italy; Department of Women and Children Health, University of Padova, Italy
| | - Leonardo Nogara
- Biomedical Sciences Department, University of Padova, Italy; Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Anna Maria Tolomeo
- Department of Women and Children Health, University of Padova, Italy; L.i.f.e.L.a.b. Program, Consorzio per La Ricerca Sanitaria (CORIS), Veneto Region, Padova, Italy
| | - Riccardo Francescato
- Stem Cells and Regenerative Medicine Lab, Institute of Pediatric Research Città Della Speranza, Padova, Italy
| | - Federica Collino
- Laboratory of Translational Research in Paediatric Nephro-urology, Fondazione Ca' Granada IRCCS Ospedale Maggiore Policlinico, Milano, Italy
| | - Giuseppe Germano
- Institute of Pediatric Research Città Della Speranza, Padova, Italy
| | | | - Edoardo Maghin
- Department of Women and Children Health, University of Padova, Italy; Tissue Engineering Lab, Institute of Pediatric Research Città Della Speranza, Padova, Italy
| | - Martina Piccoli
- Tissue Engineering Lab, Institute of Pediatric Research Città Della Speranza, Padova, Italy
| | | | - Bert Blaauw
- Biomedical Sciences Department, University of Padova, Italy; Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Piergiorgio Gamba
- Department of Women and Children Health, University of Padova, Italy
| | - Maurizio Muraca
- Department of Women and Children Health, University of Padova, Italy; Institute of Pediatric Research Città Della Speranza, Padova, Italy; L.i.f.e.L.a.b. Program, Consorzio per La Ricerca Sanitaria (CORIS), Veneto Region, Padova, Italy
| | - Michela Pozzobon
- Stem Cells and Regenerative Medicine Lab, Institute of Pediatric Research Città Della Speranza, Padova, Italy; Department of Women and Children Health, University of Padova, Italy.
| |
Collapse
|
12
|
Intensive Care Unit-Acquired Weakness: Not just Another Muscle Atrophying Condition. Int J Mol Sci 2020; 21:ijms21217840. [PMID: 33105809 PMCID: PMC7660068 DOI: 10.3390/ijms21217840] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/18/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023] Open
Abstract
Intensive care unit-acquired weakness (ICUAW) occurs in critically ill patients stemming from the critical illness itself, and results in sustained disability long after the ICU stay. Weakness can be attributed to muscle wasting, impaired contractility, neuropathy, and major pathways associated with muscle protein degradation such as the ubiquitin proteasome system and dysregulated autophagy. Furthermore, it is characterized by the preferential loss of myosin, a distinct feature of the condition. While many risk factors for ICUAW have been identified, effective interventions to offset these changes remain elusive. In addition, our understanding of the mechanisms underlying the long-term, sustained weakness observed in a subset of patients after discharge is minimal. Herein, we discuss the various proposed pathways involved in the pathophysiology of ICUAW, with a focus on the mechanisms underpinning skeletal muscle wasting and impaired contractility, and the animal models used to study them. Furthermore, we will explore the contributions of inflammation, steroid use, and paralysis to the development of ICUAW and how it pertains to those with the corona virus disease of 2019 (COVID-19). We then elaborate on interventions tested as a means to offset these decrements in muscle function that occur as a result of critical illness, and we propose new strategies to explore the molecular mechanisms of ICUAW, including serum-related biomarkers and 3D human skeletal muscle culture models.
Collapse
|
13
|
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: 15] [Impact Index Per Article: 3.0] [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
|
14
|
Lee JM, Yeong WY. Engineering macroscale cell alignment through coordinated toolpath design using support-assisted 3D bioprinting. J R Soc Interface 2020; 17:20200294. [PMID: 32674709 DOI: 10.1098/rsif.2020.0294] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aligned cells provide direction-dependent mechanical properties that influence biological and mechanical function in native tissues. Alignment techniques such as casting and uniaxial stretching cannot fully replicate the complex fibre orientation of native tissue such as the heart. In this study, bioprinting is used to direct the orientation of cell alignment. A 0°-90° grid structure was printed to assess the robustness of the support-assisted bioprinting technique. The variation in the angles of the grid pattern is designed to mimic the differences in fibril orientation of native tissues, where angles of cell alignment vary across the different layers. Through bioprinting of a cell-hydrogel mixture, C2C12 cells displayed directed alignment along the longitudinal axis of printed struts. Cell alignment is induced through firstly establishing structurally stable constructs (i.e. distinct 0°-90° structures) and secondly, allowing cells to dynamically remodel the bioprinted construct. Herein reports a method of inducing a macroscale level of controlled cell alignment with angle variation. This was not achievable both in terms of methods (i.e. conventional alignment techniques such as stretching and electrical stimulation) and magnitude (i.e. hydrogel features with less than 100 µm features).
Collapse
Affiliation(s)
- Jia Min Lee
- Singapore Centre for 3D Printing (SC3DP), Mechanical and Aerospace Engineering, Nanyang Technological University (NTU), 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Wai Yee Yeong
- Singapore Centre for 3D Printing (SC3DP), Mechanical and Aerospace Engineering, Nanyang Technological University (NTU), 50 Nanyang Avenue, Singapore 639798, Singapore
| |
Collapse
|
15
|
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: 70] [Impact Index Per Article: 11.7] [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
|
16
|
Grant L, Raman R, Cvetkovic C, Ferrall-Fairbanks MC, Pagan-Diaz GJ, Hadley P, Ko E, Platt MO, Bashir R. Long-Term Cryopreservation and Revival of Tissue-Engineered Skeletal Muscle. Tissue Eng Part A 2019; 25:1023-1036. [PMID: 30412045 PMCID: PMC6916121 DOI: 10.1089/ten.tea.2018.0202] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 11/06/2018] [Indexed: 01/13/2023] Open
Abstract
IMPACT STATEMENT The ability to freeze, revive, and prolong the lifetime of tissue-engineered skeletal muscle without incurring any loss of function represents a significant advancement in the field of tissue engineering. Cryopreservation enables the efficient fabrication, storage, and shipment of these tissues. This in turn facilitates multidisciplinary collaboration between research groups, enabling advances in skeletal muscle regenerative medicine, organ-on-a-chip models of disease, drug testing, and soft robotics. Furthermore, the observation that freezing undifferentiated skeletal muscle enhances functional performance may motivate future studies developing stronger and more clinically relevant engineered muscle.
Collapse
Affiliation(s)
- Lauren Grant
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Ritu Raman
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Caroline Cvetkovic
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Meghan C. Ferrall-Fairbanks
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | - Gelson J. Pagan-Diaz
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Pierce Hadley
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Eunkyung Ko
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Manu O. Platt
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | - Rashid Bashir
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois
| |
Collapse
|
17
|
Von den Hoff JW, Carvajal Monroy PL, Ongkosuwito EM, van Kuppevelt TH, Daamen WF. Muscle fibrosis in the soft palate: Delivery of cells, growth factors and anti-fibrotics. Adv Drug Deliv Rev 2019; 146:60-76. [PMID: 30107211 DOI: 10.1016/j.addr.2018.08.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/29/2018] [Accepted: 08/06/2018] [Indexed: 02/07/2023]
Abstract
The healing of skeletal muscle injuries after major trauma or surgical reconstruction is often complicated by the development of fibrosis leading to impaired function. Research in the field of muscle regeneration is mainly focused on the restoration of muscle mass while far less attention is paid to the prevention of fibrosis. In this review, we take as an example the reconstruction of the muscles in the soft palate of cleft palate patients. After surgical closure of the soft palate, muscle function during speech is often impaired by a shortage of muscle tissue as well as the development of fibrosis. We will give a short overview of the most common approaches to generate muscle mass and then focus on strategies to prevent fibrosis. These include anti-fibrotic strategies that have been developed for muscle and other organs by the delivery of small molecules, decorin and miRNAs. Anti-fibrotic compounds should be delivered in aligned constructs in order to obtain the organized architecture of muscle tissue. The available techniques for the preparation of aligned muscle constructs will be discussed. The combination of approaches to generate muscle mass with anti-fibrotic components in an aligned muscle construct may greatly improve the functional outcome of regenerative therapies for muscle injuries.
Collapse
Affiliation(s)
- Johannes W Von den Hoff
- Department of Orthodontics and Craniofacial Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500HB Nijmegen, The Netherlands.
| | - Paola L Carvajal Monroy
- Department of Oral and Maxillofacial Surgery, Special Dental Care and Orthodontics, Erasmus Medical Center, P.O. Box 2060, 3000CB Rotterdam, The Netherlands.
| | - Edwin M Ongkosuwito
- Department of Orthodontics and Craniofacial Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500HB Nijmegen, The Netherlands.
| | - Toin H van Kuppevelt
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500HB Nijmegen, The Netherlands.
| | - Willeke F Daamen
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500HB Nijmegen, The Netherlands.
| |
Collapse
|
18
|
Vajanthri KY, Sidu RK, Poddar S, Singh AK, Mahto SK. Combined substrate micropatterning and FFT analysis reveals myotube size control and alignment by contact guidance. Cytoskeleton (Hoboken) 2019; 76:269-285. [DOI: 10.1002/cm.21527] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 04/23/2019] [Accepted: 05/02/2019] [Indexed: 12/30/2022]
Affiliation(s)
- Kiran Yellappa Vajanthri
- Tissue Engineering and Biomicrofluidics Laboratory, School of Biomedical EngineeringIndian Institute of Technology (Banaras Hindu University) Varanasi Uttar Pradesh India
| | - Rakesh Kumar Sidu
- Tissue Engineering and Biomicrofluidics Laboratory, School of Biomedical EngineeringIndian Institute of Technology (Banaras Hindu University) Varanasi Uttar Pradesh India
| | - Suruchi Poddar
- Tissue Engineering and Biomicrofluidics Laboratory, School of Biomedical EngineeringIndian Institute of Technology (Banaras Hindu University) Varanasi Uttar Pradesh India
| | - Ashish Kumar Singh
- School of Biochemical EngineeringIndian Institute of Technology (Banaras Hindu University) Varanasi Uttar Pradesh India
| | - Sanjeev Kumar Mahto
- Tissue Engineering and Biomicrofluidics Laboratory, School of Biomedical EngineeringIndian Institute of Technology (Banaras Hindu University) Varanasi Uttar Pradesh India
- Center for Advanced Biomaterials and Tissue EngineeringIndian Institute of Technology (Banaras Hindu University) Varanasi Uttar Pradesh India
| |
Collapse
|
19
|
Khodabukus A, Madden L, Prabhu NK, Koves TR, Jackman CP, Muoio DM, Bursac N. Electrical stimulation increases hypertrophy and metabolic flux in tissue-engineered human skeletal muscle. Biomaterials 2019; 198:259-269. [PMID: 30180985 PMCID: PMC6395553 DOI: 10.1016/j.biomaterials.2018.08.058] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/05/2018] [Accepted: 08/27/2018] [Indexed: 02/08/2023]
Abstract
In vitro models of contractile human skeletal muscle hold promise for use in disease modeling and drug development, but exhibit immature properties compared to native adult muscle. To address this limitation, 3D tissue-engineered human muscles (myobundles) were electrically stimulated using intermittent stimulation regimes at 1 Hz and 10 Hz. Dystrophin in myotubes exhibited mature membrane localization suggesting a relatively advanced starting developmental maturation. One-week stimulation significantly increased myobundle size, sarcomeric protein abundance, calcium transient amplitude (∼2-fold), and tetanic force (∼3-fold) resulting in the highest specific force generation (19.3mN/mm2) reported for engineered human muscles to date. Compared to 1 Hz electrical stimulation, the 10 Hz stimulation protocol resulted in greater myotube hypertrophy and upregulated mTORC1 and ERK1/2 activity. Electrically stimulated myobundles also showed a decrease in fatigue resistance compared to control myobundles without changes in glycolytic or mitochondrial protein levels. Greater glucose consumption and decreased abundance of acetylcarnitine in stimulated myobundles indicated increased glycolytic and fatty acid metabolic flux. Moreover, electrical stimulation of myobundles resulted in a metabolic shift towards longer-chain fatty acid oxidation as evident from increased abundances of medium- and long-chain acylcarnitines. Taken together, our study provides an advanced in vitro model of human skeletal muscle with improved structure, function, maturation, and metabolic flux.
Collapse
Affiliation(s)
| | - Lauran Madden
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Neel K Prabhu
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Timothy R Koves
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | | | - Deborah M Muoio
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
20
|
Huang T, Xiao J, Wang S, Liao Z, Huang T, Gu R, Li J, Wu G, Liao H. The thickness of poly-phenoxyethyl methacrylate brush interferes with cellular behavior and function of myofibers. J Biomed Mater Res A 2019; 107:1264-1272. [PMID: 30724032 DOI: 10.1002/jbm.a.36636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 12/19/2018] [Accepted: 12/30/2018] [Indexed: 11/07/2022]
Abstract
Introducing or grafting molecules onto biomaterial surfaces to regulate muscle cell destination via biophysical cues is one of the important steps for biomaterial design in muscle tissue engineering. Therefore, it is important to understand the interaction between myoblasts and myofibers with substrates modified by biomimetic layer with different thicknesses. In this study, we used a surface-induced atom transfer radical polymerization method to synthetize and graft poly-phenoxyethyl methacrylate (PHEMA) brushes having different lengths on the glass substrates. C2C12 myoblasts were seeded on the PHEMA brushes and differentiated using horse serum, for analyzing the sensibility of muscle cells to feel environment changing, and further investigating whether the depths of grafting layer on the biomaterial surface are important factors in regulating muscle cell behaviors. Our results demonstrated that on the thicker PHEMA brushes surface (200 and 450 nm), C2C12 myoblasts showed a better survival and proliferation and were favorable for cell fusion and myotube formation. Furthermore, myofibers survived on the thicker brushes were more functional and upregulated cytoskeleton proteins (tubulin, vimentin, and vinculin) and FAK levels, and enhanced the expression levels for mechanical stress molecules (HGF, NOS-1, and c-Met). These results suggest that grafting thickness of PHEMA layer on the substrate led to the myoblasts/myofiber behavior change, which would be valuable for the design and preparation of the modified layer on muscle tissue engineering scaffolds. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 1264-1272, 2019.
Collapse
Affiliation(s)
- Tao Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China
| | - Jiangwei Xiao
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shuhao Wang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
| | - Zhaohong Liao
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Tao Huang
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ruicai Gu
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Junhua Li
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Gang Wu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
| | - Hua Liao
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
21
|
Liu L, Zhang C, Wang W, Xi N, Wang Y. Regulation of C2C12 Differentiation and Control of the Beating Dynamics of Contractile Cells for a Muscle-Driven Biosyncretic Crawler by Electrical Stimulation. Soft Robot 2018; 5:748-760. [DOI: 10.1089/soro.2018.0017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Lianqing Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China
| | - Chuang Zhang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Wenxue Wang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China
| | - Ning Xi
- Department of Industrial and Manufacturing Systems Engineering, Emerging Technologies Institute, University of Hong Kong Pokfulam, Hong Kong, Hong Kong
| | - Yuechao Wang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China
| |
Collapse
|
22
|
Khodabukus A, Prabhu N, Wang J, Bursac N. In Vitro Tissue-Engineered Skeletal Muscle Models for Studying Muscle Physiology and Disease. Adv Healthc Mater 2018; 7:e1701498. [PMID: 29696831 PMCID: PMC6105407 DOI: 10.1002/adhm.201701498] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 02/18/2018] [Indexed: 12/18/2022]
Abstract
Healthy skeletal muscle possesses the extraordinary ability to regenerate in response to small-scale injuries; however, this self-repair capacity becomes overwhelmed with aging, genetic myopathies, and large muscle loss. The failure of small animal models to accurately replicate human muscle disease, injury and to predict clinically-relevant drug responses has driven the development of high fidelity in vitro skeletal muscle models. Herein, the progress made and challenges ahead in engineering biomimetic human skeletal muscle tissues that can recapitulate muscle development, genetic diseases, regeneration, and drug response is discussed. Bioengineering approaches used to improve engineered muscle structure and function as well as the functionality of satellite cells to allow modeling muscle regeneration in vitro are also highlighted. Next, a historical overview on the generation of skeletal muscle cells and tissues from human pluripotent stem cells, and a discussion on the potential of these approaches to model and treat genetic diseases such as Duchenne muscular dystrophy, is provided. Finally, the need to integrate multiorgan microphysiological systems to generate improved drug discovery technologies with the potential to complement or supersede current preclinical animal models of muscle disease is described.
Collapse
Affiliation(s)
- Alastair Khodabukus
- Department of Biomedical Engineering Duke University 101 Science Drive, FCIEMAS 1427, Durham, NC 27708-90281, USA
| | - Neel Prabhu
- Department of Biomedical Engineering Duke University 101 Science Drive, FCIEMAS 1427, Durham, NC 27708-90281, USA
| | - Jason Wang
- Department of Biomedical Engineering Duke University 101 Science Drive, FCIEMAS 1427, Durham, NC 27708-90281, USA
| | - Nenad Bursac
- Department of Biomedical Engineering Duke University 101 Science Drive, FCIEMAS 1427, Durham, NC 27708-90281, USA
| |
Collapse
|
23
|
Biomaterials in Tendon and Skeletal Muscle Tissue Engineering: Current Trends and Challenges. MATERIALS 2018; 11:ma11071116. [PMID: 29966303 PMCID: PMC6073924 DOI: 10.3390/ma11071116] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 06/20/2018] [Accepted: 06/25/2018] [Indexed: 12/17/2022]
Abstract
Tissue engineering is a promising approach to repair tendon and muscle when natural healing fails. Biohybrid constructs obtained after cells’ seeding and culture in dedicated scaffolds have indeed been considered as relevant tools for mimicking native tissue, leading to a better integration in vivo. They can also be employed to perform advanced in vitro studies to model the cell differentiation or regeneration processes. In this review, we report and analyze the different solutions proposed in literature, for the reconstruction of tendon, muscle, and the myotendinous junction. They classically rely on the three pillars of tissue engineering, i.e., cells, biomaterials and environment (both chemical and physical stimuli). We have chosen to present biomimetic or bioinspired strategies based on understanding of the native tissue structure/functions/properties of the tissue of interest. For each tissue, we sorted the relevant publications according to an increasing degree of complexity in the materials’ shape or manufacture. We present their biological and mechanical performances, observed in vitro and in vivo when available. Although there is no consensus for a gold standard technique to reconstruct these musculo-skeletal tissues, the reader can find different ways to progress in the field and to understand the recent history in the choice of materials, from collagen to polymer-based matrices.
Collapse
|
24
|
Cardiopatch platform enables maturation and scale-up of human pluripotent stem cell-derived engineered heart tissues. Nat Commun 2017; 8:1825. [PMID: 29184059 PMCID: PMC5705709 DOI: 10.1038/s41467-017-01946-x] [Citation(s) in RCA: 292] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 10/27/2017] [Indexed: 12/25/2022] Open
Abstract
Despite increased use of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) for drug development and disease modeling studies, methods to generate large, functional heart tissues for human therapy are lacking. Here we present a “Cardiopatch” platform for 3D culture and maturation of hiPSC-CMs that after 5 weeks of differentiation show robust electromechanical coupling, consistent H-zones, I-bands, and evidence for T-tubules and M-bands. Cardiopatch maturation markers and functional output increase during culture, approaching values of adult myocardium. Cardiopatches can be scaled up to clinically relevant dimensions, while preserving spatially uniform properties with high conduction velocities and contractile stresses. Within window chambers in nude mice, cardiopatches undergo vascularization by host vessels and continue to fire Ca2+ transients. When implanted onto rat hearts, cardiopatches robustly engraft, maintain pre-implantation electrical function, and do not increase the incidence of arrhythmias. These studies provide enabling technology for future use of hiPSC-CM tissues in human heart repair. Cardiomyocytes derived from human induced pluripotent stem cells could be used to generate cardiac tissues for regenerative purposes. Here the authors describe a method to obtain large bioengineered heart tissues showing advanced maturation, functional features and engraftment capacity.
Collapse
|
25
|
Jagla K, Kalman B, Boudou T, Hénon S, Batonnet-Pichon S. Beyond mice: Emerging and transdisciplinary models for the study of early-onset myopathies. Semin Cell Dev Biol 2017; 64:171-180. [DOI: 10.1016/j.semcdb.2016.09.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 09/06/2016] [Accepted: 09/22/2016] [Indexed: 01/23/2023]
|
26
|
Choi YJ, Kim TG, Jeong J, Yi HG, Park JW, Hwang W, Cho DW. 3D Cell Printing of Functional Skeletal Muscle Constructs Using Skeletal Muscle-Derived Bioink. Adv Healthc Mater 2016; 5:2636-2645. [PMID: 27529631 DOI: 10.1002/adhm.201600483] [Citation(s) in RCA: 171] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 06/15/2016] [Indexed: 12/21/2022]
Abstract
Engineered skeletal muscle tissues that mimic the structure and function of native muscle have been considered as an alternative strategy for the treatment of various muscular diseases and injuries. Here, it is demonstrated that 3D cell-printing of decellularized skeletal muscle extracellular matrix (mdECM)-based bioink facilitates the fabrication of functional skeletal muscle constructs. The cellular alignment and the shape of the tissue constructs are controlled by 3D cell-printing technology. mdECM bioink provides the 3D cell-printed muscle constructs with a myogenic environment that supports high viability and contractility as well as myotube formation, differentiation, and maturation. More interestingly, the preservation of agrin is confirmed in the mdECM, and significant increases in the formation of acetylcholine receptor clusters are exhibited in the 3D cell-printed muscle constructs. In conclusion, mdECM bioink and 3D cell-printing technology facilitate the mimicking of both the structural and functional properties of native muscle and hold great promise for producing clinically relevant engineered muscle for the treatment of muscular injuries.
Collapse
Affiliation(s)
- Yeong-Jin Choi
- Division of Integrative Biosciences and Biotechnology; Pohang University of Science and Technology (POSTECH); 77 Cheongam-ro Nam-guPohang Kyungbuk 790-784 Korea
| | - Taek Gyoung Kim
- Department of Mechanical Engineering; Pohang University of Science and Technology (POSTECH); 77 Cheongam-ro Nam-gu, Pohang Kyungbuk 790-784 Korea
| | - Jonghyeon Jeong
- Department of Mechanical Engineering; Pohang University of Science and Technology (POSTECH); 77 Cheongam-ro Nam-gu, Pohang Kyungbuk 790-784 Korea
| | - Hee-Gyeong Yi
- Department of Mechanical Engineering; Pohang University of Science and Technology (POSTECH); 77 Cheongam-ro Nam-gu, Pohang Kyungbuk 790-784 Korea
| | - Ji Won Park
- Department of Mechanical Engineering; Pohang University of Science and Technology (POSTECH); 77 Cheongam-ro Nam-gu, Pohang Kyungbuk 790-784 Korea
| | - Woonbong Hwang
- Department of Mechanical Engineering; Pohang University of Science and Technology (POSTECH); 77 Cheongam-ro Nam-gu, Pohang Kyungbuk 790-784 Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering; Pohang University of Science and Technology (POSTECH); 77 Cheongam-ro Nam-gu, Pohang Kyungbuk 790-784 Korea
| |
Collapse
|
27
|
Bursac N, Juhas M, Rando TA. Synergizing Engineering and Biology to Treat and Model Skeletal Muscle Injury and Disease. Annu Rev Biomed Eng 2016; 17:217-42. [PMID: 26643021 DOI: 10.1146/annurev-bioeng-071114-040640] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Although skeletal muscle is one of the most regenerative organs in our body, various genetic defects, alterations in extrinsic signaling, or substantial tissue damage can impair muscle function and the capacity for self-repair. The diversity and complexity of muscle disorders have attracted much interest from both cell biologists and, more recently, bioengineers, leading to concentrated efforts to better understand muscle pathology and develop more efficient therapies. This review describes the biological underpinnings of muscle development, repair, and disease, and discusses recent bioengineering efforts to design and control myomimetic environments, both to study muscle biology and function and to aid in the development of new drug, cell, and gene therapies for muscle disorders. The synergy between engineering-aided biological discovery and biology-inspired engineering solutions will be the path forward for translating laboratory results into clinical practice.
Collapse
Affiliation(s)
- Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708;
| | - Mark Juhas
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708;
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305.,Rehabilitation Research & Development Service, VA Palo Alto Health Care System, Palo Alto, California 94304
| |
Collapse
|
28
|
Morgan KY, Sklaviadis D, Tochka ZL, Fischer KM, Hearon K, Morgan TD, Langer R, Freed LE. Multi-Material Tissue Engineering Scaffold with Hierarchical Pore Architecture. ADVANCED FUNCTIONAL MATERIALS 2016; 26:5873-5883. [PMID: 27942257 PMCID: PMC5142531 DOI: 10.1002/adfm.201601146] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Multi-material polymer scaffolds with multiscale pore architectures were characterized and tested with vascular and heart cells as part of a platform for replacing damaged heart muscle. Vascular and muscle scaffolds were constructed from a new material, poly(limonene thioether) (PLT32i), which met the design criteria of slow biodegradability, elastomeric mechanical properties, and facile processing. The vascular-parenchymal interface was a poly(glycerol sebacate) (PGS) porous membrane that met different criteria of rapid biodegradability, high oxygen permeance, and high porosity. A hierarchical architecture of primary (macroscale) and secondary (microscale) pores was created by casting the PLT32i prepolymer onto sintered spheres of poly(methyl methacrylate) (PMMA) within precisely patterned molds followed by photocuring, de-molding, and leaching out the PMMA. Pre-fabricated polymer templates were cellularized, assembled, and perfused in order to engineer spatially organized, contractile heart tissue. Structural and functional analyses showed that the primary pores guided heart cell alignment and enabled robust perfusion while the secondary pores increased heart cell retention and reduced polymer volume fraction.
Collapse
Affiliation(s)
- Kathy Ye Morgan
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Research, and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Demetra Sklaviadis
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Research, and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Zachary L. Tochka
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Research, and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kristin M. Fischer
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Research, and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Keith Hearon
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Research, and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Thomas D. Morgan
- Harvard University School of Engineering & Applied Science, Cambridge, MA 02138, USA
| | - Robert Langer
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Research, and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Lisa E. Freed
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Research, and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Materials Engineering Division, Draper, Cambridge, MA 02139, USA
| |
Collapse
|
29
|
Wu S, Duan B, Liu P, Zhang C, Qin X, Butcher JT. Fabrication of Aligned Nanofiber Polymer Yarn Networks for Anisotropic Soft Tissue Scaffolds. ACS APPLIED MATERIALS & INTERFACES 2016; 8:16950-60. [PMID: 27304080 DOI: 10.1021/acsami.6b05199] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Nanofibrous scaffolds with defined architectures and anisotropic mechanical properties are attractive for many tissue engineering and regenerative medicine applications. Here, a novel electrospinning system is developed and implemented to fabricate continuous processable uniaxially aligned nanofiber yarns (UANY). UANY were processed into fibrous tissue scaffolds with defined anisotropic material properties using various textile-forming technologies, i.e., braiding, weaving, and knitting techniques. UANY braiding dramatically increased overall stiffness and strength compared to the same number of UANY unbraided. Human adipose derived stem cells (HADSC) cultured on UANY or woven and knitted 3D scaffolds aligned along local fiber direction and were >90% viable throughout 21 days. Importantly, UANY supported biochemical induction of HADSC differentiation toward smooth muscle and osteogenic lineages. Moreover, we integrated an anisotropic woven fiber mesh within a bioactive hydrogel to mimic the complex microstructure and mechanical behavior of valve tissues. Human aortic valve interstitial cells (HAVIC) and human aortic root smooth muscle cells (HASMC) were separately encapsulated within hydrogel/woven fabric composite scaffolds for generating scaffolds with anisotropic biomechanics and valve ECM like microenvironment for heart valve tissue engineering. UANY have great potential as building blocks for generating fiber-shaped tissues or tissue microstructures with complex architectures.
Collapse
Affiliation(s)
- Shaohua Wu
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles, Donghua University , No. 2999 North Renmin Road, Songjiang, Shanghai 201620, China
- Department of Biomedical Engineering, Cornell University , Ithaca, New York 14850, United States
| | - Bin Duan
- Department of Biomedical Engineering, Cornell University , Ithaca, New York 14850, United States
| | - Penghong Liu
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles, Donghua University , No. 2999 North Renmin Road, Songjiang, Shanghai 201620, China
| | - Caidan Zhang
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles, Donghua University , No. 2999 North Renmin Road, Songjiang, Shanghai 201620, China
| | - Xiaohong Qin
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles, Donghua University , No. 2999 North Renmin Road, Songjiang, Shanghai 201620, China
- Key Laboratory of Shanghai Micro & Nano Technology , Shanghai 201620, China
| | - Jonathan T Butcher
- Department of Biomedical Engineering, Cornell University , Ithaca, New York 14850, United States
| |
Collapse
|
30
|
Abstract
There is growing appreciation of the role that the extracellular environment plays in regulating cell behavior. Mechanical, structural, and compositional cues, either alone or in concert, can drastically alter cell function. Biomaterials, and particularly hydrogels, have been developed and implemented to present defined subsets of these cues for investigating countless cellular processes as a means of understanding morphogenesis, aging, and disease. Although most scientists concede that standard cell culture materials (tissue culture plastic and glass) do a poor job of recapitulating native cellular milieus, there is currently a knowledge barrier for many researchers in regard to the application of hydrogels for cell culture. Here, we introduce hydrogels to those who may be unfamiliar with procedures to culture and study cells with these systems, with a particular focus on commercially available hydrogels.
Collapse
Affiliation(s)
- Steven R Caliari
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jason A Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
31
|
Gribova V, Liu CY, Nishiguchi A, Matsusaki M, Boudou T, Picart C, Akashi M. Construction and myogenic differentiation of 3D myoblast tissues fabricated by fibronectin-gelatin nanofilm coating. Biochem Biophys Res Commun 2016; 474:515-521. [PMID: 27125461 DOI: 10.1016/j.bbrc.2016.04.130] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 04/24/2016] [Indexed: 11/16/2022]
Abstract
In this study, we used a recently developed approach of coating the cells with fibronectin-gelatin nanofilms to build 3D skeletal muscle tissue models. We constructed the microtissues from C2C12 myoblasts and subsequently differentiated them to form muscle-like tissue. The thickness of the constructs could be successfully controlled by altering the number of seeded cells. We were able to build up to ∼76 μm thick 3D constructs that formed multinucleated myotubes. We also found that Rho-kinase inhibitor Y27632 improved myotube formation in thick constructs. Our approach makes it possible to rapidly form 3D muscle tissues and is promising for the in vitro construction of physiologically relevant human skeletal muscle tissue models.
Collapse
Affiliation(s)
- Varvara Gribova
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Chun-Yen Liu
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Akihiro Nishiguchi
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Thomas Boudou
- Grenoble Institute of Technology, CNRS UMR 5628, Laboratory of Materials and Physical Engineering, 3 Parvis L. Néel, 38016 Grenoble, France
| | - Catherine Picart
- Grenoble Institute of Technology, CNRS UMR 5628, Laboratory of Materials and Physical Engineering, 3 Parvis L. Néel, 38016 Grenoble, France.
| | - Mitsuru Akashi
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
32
|
Juhas M, Ye J, Bursac N. Design, evaluation, and application of engineered skeletal muscle. Methods 2016; 99:81-90. [PMID: 26455485 PMCID: PMC4821818 DOI: 10.1016/j.ymeth.2015.10.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 09/03/2015] [Accepted: 10/04/2015] [Indexed: 12/17/2022] Open
Abstract
For over two decades, research groups have been developing methods to engineer three-dimensional skeletal muscle tissues. These tissues hold great promise for use in disease modeling and pre-clinical drug development, and have potential to serve as therapeutic grafts for functional muscle repair. Recent advances in the field have resulted in the engineering of regenerative muscle constructs capable of survival, vascularization, and functional maturation in vivo as well as the first-time creation of functional human engineered muscles for screening of therapeutics in vitro. In this review, we will discuss the methodologies that have progressed work in the muscle tissue engineering field to its current state. The emphasis will be placed on the existing procedures to generate myogenic cell sources and form highly functional muscle tissues in vitro, techniques to monitor and evaluate muscle maturation and performance in vitro and in vivo, and surgical strategies to both create diseased environments and ensure implant survival and rapid integration into the host. Finally, we will suggest the most promising methodologies that will enable continued progress in the field.
Collapse
Affiliation(s)
- Mark Juhas
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - Jean Ye
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, United States.
| |
Collapse
|
33
|
Fischer KM, Morgan KY, Hearon K, Sklaviadis D, Tochka ZL, Fenton OS, Anderson DG, Langer R, Freed LE. Poly(Limonene Thioether) Scaffold for Tissue Engineering. Adv Healthc Mater 2016; 5:813-21. [PMID: 26890480 PMCID: PMC4828277 DOI: 10.1002/adhm.201500892] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Indexed: 01/14/2023]
Abstract
A photocurable thiol-ene network polymer, poly(limonene thioether) (PLT32o), is synthesized, characterized, fabricated into tissue engineering scaffolds, and demonstrated in vitro and in vivo. Micromolded PLT32o grids exhibit compliant, elastomeric mechanical behavior similar to grids made of poly(glycerol sebacate) (PGS), an established biomaterial. Multilayered PL32o scaffolds with regular, geometrically defined pore architectures support heart cell seeding and culture in a manner similar to multilayered PGS scaffolds. Subcutaneous implantation of multilayered PLT32o scaffolds with cultured heart cells provides long-term 3D structural support and retains the exogenous cells, whereas PGS scaffolds lose both their structural integrity and the exogenous cells over 31 d in vivo. PLT32o membrane implants retain their dry mass, whereas PGS implants lose 70 percent of their dry mass by day 31. Macrophages are initially recruited to PLT32o and PGS membrane implants but are no longer present by day 31. Facile synthesis and processing in combination with the capability to support heart cells in vitro and in vivo suggest that PLT32o can offer advantages for tissue engineering applications where prolonged in vivo maintenance of 3D structural integrity and elastomeric mechanical behavior are required.
Collapse
Affiliation(s)
- Kristin M Fischer
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Researchand Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Kathy Ye Morgan
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Researchand Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Keith Hearon
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Researchand Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Demetra Sklaviadis
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Researchand Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Zachary L Tochka
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Researchand Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Owen S Fenton
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Researchand Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Daniel G Anderson
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Researchand Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Robert Langer
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Researchand Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Lisa E Freed
- Harvard-MIT Division of Health Sciences and Technology, David H. Koch Institute for Integrative Cancer Researchand Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Materials Engineering Division, Draper, Cambridge, MA, 02139, USA
| |
Collapse
|
34
|
Mazelet L, Parker MO, Li M, Arner A, Ashworth R. Role of Active Contraction and Tropomodulins in Regulating Actin Filament Length and Sarcomere Structure in Developing Zebrafish Skeletal Muscle. Front Physiol 2016; 7:91. [PMID: 27065876 PMCID: PMC4814503 DOI: 10.3389/fphys.2016.00091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/26/2016] [Indexed: 01/13/2023] Open
Abstract
Whilst it is recognized that contraction plays an important part in maintaining the structure and function of mature skeletal muscle, its role during development remains undefined. In this study the role of movement in skeletal muscle maturation was investigated in intact zebrafish embryos using a combination of genetic and pharmacological approaches. An immotile mutant line (cacnb1 (ts25) ) which lacks functional voltage-gated calcium channels (dihydropyridine receptors) in the muscle and pharmacological immobilization of embryos with a reversible anesthetic (Tricaine), allowed the study of paralysis (in mutants and anesthetized fish) and recovery of movement (reversal of anesthetic treatment). The effect of paralysis in early embryos (aged between 17 and 24 hours post-fertilization, hpf) on skeletal muscle structure at both myofibrillar and myofilament level was determined using both immunostaining with confocal microscopy and small angle X-ray diffraction. The consequences of paralysis and subsequent recovery on the localization of the actin capping proteins Tropomodulin 1 & 4 (Tmod) in fish aged from 17 hpf until 42 hpf was also assessed. The functional consequences of early paralysis were investigated by examining the mechanical properties of the larval muscle. The length-force relationship, active and passive tension, was measured in immotile, recovered and control skeletal muscle at 5 and 7 day post-fertilization (dpf). Recovery of muscle function was also assessed by examining swimming patterns in recovered and control fish. Inhibition of the initial embryonic movements (up to 24 hpf) resulted in an increase in myofibril length and a decrease in width followed by almost complete recovery in both moving and paralyzed fish by 42 hpf. In conclusion, myofibril organization is regulated by a dual mechanism involving movement-dependent and movement-independent processes. The initial contractile event itself drives the localization of Tmod1 to its sarcomeric position, capping the actin pointed ends and ultimately regulating actin length. This study demonstrates that both contraction and contractile-independent mechanisms are important for the regulation of myofibril organization, which in turn is necessary for establishing proper skeletal muscle structure and function during development in vivo in zebrafish.
Collapse
Affiliation(s)
- Lise Mazelet
- School of Biological and Chemical Sciences, Queen Mary, University of London London, UK
| | - Matthew O Parker
- School of Health Sciences and Social Work, University of Portsmouth Portsmouth, UK
| | - Mei Li
- Department of Physiology and Pharmacology, Karolinska Institutet Stockholm, Sweden
| | - Anders Arner
- Department of Physiology and Pharmacology, Karolinska Institutet Stockholm, Sweden
| | - Rachel Ashworth
- The Blizard Institute/Institute of Health Sciences Education, Barts and The London School of Medicine and Dentistry London, UK
| |
Collapse
|
35
|
Kang HW, Lee SJ, Ko IK, Kengla C, Yoo JJ, Atala A. A 3D bioprinting system to produce human-scale tissue constructs with structural integrity. Nat Biotechnol 2016; 34:312-9. [PMID: 26878319 DOI: 10.1038/nbt.3413] [Citation(s) in RCA: 1506] [Impact Index Per Article: 167.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/19/2015] [Indexed: 02/07/2023]
Abstract
A challenge for tissue engineering is producing three-dimensional (3D), vascularized cellular constructs of clinically relevant size, shape and structural integrity. We present an integrated tissue-organ printer (ITOP) that can fabricate stable, human-scale tissue constructs of any shape. Mechanical stability is achieved by printing cell-laden hydrogels together with biodegradable polymers in integrated patterns and anchored on sacrificial hydrogels. The correct shape of the tissue construct is achieved by representing clinical imaging data as a computer model of the anatomical defect and translating the model into a program that controls the motions of the printer nozzles, which dispense cells to discrete locations. The incorporation of microchannels into the tissue constructs facilitates diffusion of nutrients to printed cells, thereby overcoming the diffusion limit of 100-200 μm for cell survival in engineered tissues. We demonstrate capabilities of the ITOP by fabricating mandible and calvarial bone, cartilage and skeletal muscle. Future development of the ITOP is being directed to the production of tissues for human applications and to the building of more complex tissues and solid organs.
Collapse
Affiliation(s)
- Hyun-Wook Kang
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, USA
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, USA
| | - In Kap Ko
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, USA
| | - Carlos Kengla
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, USA
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, USA
| |
Collapse
|
36
|
Affiliation(s)
- Adam W. Feinberg
- Department of Biomedical Engineering and Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213;
| |
Collapse
|
37
|
Takahashi H, Okano T. Cell Sheet-Based Tissue Engineering for Organizing Anisotropic Tissue Constructs Produced Using Microfabricated Thermoresponsive Substrates. Adv Healthc Mater 2015; 4:2388-407. [PMID: 26033874 DOI: 10.1002/adhm.201500194] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 04/22/2015] [Indexed: 11/12/2022]
Abstract
In some native tissues, appropriate microstructures, including orientation of the cell/extracellular matrix, provide specific mechanical and biological functions. For example, skeletal muscle is made of oriented myofibers that is responsible for the mechanical function. Native artery and myocardial tissues are organized three-dimensionally by stacking sheet-like tissues of aligned cells. Therefore, to construct any kind of complex tissue, the microstructures of cells such as myotubes, smooth muscle cells, and cardiomyocytes also need to be organized three-dimensionally just as in the native tissues of the body. Cell sheet-based tissue engineering allows the production of scaffold-free engineered tissues through a layer-by-layer construction technique. Recently, using microfabricated thermoresponsive substrates, aligned cells are being harvested as single continuous cell sheets. The cell sheets act as anisotropic tissue units to build three-dimensional tissue constructs with the appropriate anisotropy. This cell sheet-based technology is straightforward and has the potential to engineer a wide variety of complex tissues. In addition, due to the scaffold-free cell-dense environment, the physical and biological cell-cell interactions of these cell sheet constructs exhibit unique cell behaviors. These advantages will provide important clues to enable the production of well-organized tissues that closely mimic the structure and function of native tissues, required for the future of tissue engineering.
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
| | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University; 8-1 Kawada-cho, Shinjuku-ku; Tokyo 162-8666 Japan
| |
Collapse
|
38
|
Xu Y, Li Z, Li X, Fan Z, Liu Z, Xie X, Guan J. Regulating myogenic differentiation of mesenchymal stem cells using thermosensitive hydrogels. Acta Biomater 2015; 26:23-33. [PMID: 26277379 DOI: 10.1016/j.actbio.2015.08.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 07/30/2015] [Accepted: 08/11/2015] [Indexed: 01/02/2023]
Abstract
Stem cell therapy has potential to regenerate skeletal muscle tissue in ischemic limb. However, the delivered stem cells experience low rate of myogenic differentiation. Employing injectable hydrogels as stem cell carriers may enhance the myogenic differentiation as their modulus may be tailored to induce the differentiation. Yet current approaches used to manipulate hydrogel modulus often simultaneously vary other properties that also affect stem cell differentiation, such as chemical structure, composition and water content. Thus it is challenging to demonstrate the decoupled effect of hydrogel modulus on stem cell differentiation. In this report, we decoupled the hydrogel modulus from chemical structure, composition, and water content using injectable and thermosensitive hydrogels. The hydrogels were synthesized from N-isopropylacrylamide (NIPAAm), acrylic acid (AAc), and degradable macromer 2-hydroxyethyl methacrylate-oligomer [oligolatide, oligohydroxybutyrate, or oligo(trimethylene carbonate)]. We found that using the same monomer composition and oligomer chemical structure but different oligomer length can independently vary hydrogel modulus. Rat bone marrow mesenchymal stem cells (MSCs) were encapsulated in the hydrogels with elastic expansion moduli of 11, 20, and 40 kPa, respectively. After 14 days of culture, significant myogenic differentiation was achieved for the hydrogel with elastic expansion modulus of 20 kPa, as judged from both the gene and protein expression. In addition, MSCs exhibited an elastic expansion modulus-dependent proliferation rate. The most significant proliferation was observed in the hydrogel with elastic expansion modulus of 40 kPa. These results demonstrate that the developed injectable and thermosensitive hydrogels with suitable modulus has the potential to deliver stem cells into ischemic limb for enhanced myogenic differentiation and muscle regeneration. STATEMENT OF SIGNIFICANCE Stem cell therapy for skeletal muscle regeneration in ischemic limb experiences low rate of myogenic differentiation. Employing injectable hydrogels as stem cell carriers may enhance the myogenic differentiation as hydrogel modulus may be modulated to induce the differentiation. Yet current approaches used to modulate hydrogel modulus may simultaneously vary other properties that also affect stem cell myogenic differentiation, such as chemistry, composition and water content. In this report, we decoupled the hydrogel modulus from chemistry, composition, and water content using injectable and thermosensitive hydrogels. We found that mesenchymal stem cells best differentiated into myogenic lineage in the hydrogel with elastic modulus of 20 kPa.
Collapse
Affiliation(s)
- Yanyi Xu
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States
| | - Zhenqing Li
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States
| | - Xiaofei Li
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States
| | - Zhaobo Fan
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States
| | - Zhenguo Liu
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, United States
| | - Xiaoyun Xie
- Department of Gerontology, Tongji Hospital, Tongji University, Shanghai, China
| | - Jianjun Guan
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, United States; Tongji Hospital, Tongji University, Shanghai, China.
| |
Collapse
|
39
|
Wolf MT, Dearth CL, Sonnenberg SB, Loboa EG, Badylak SF. Naturally derived and synthetic scaffolds for skeletal muscle reconstruction. Adv Drug Deliv Rev 2015; 84:208-21. [PMID: 25174309 DOI: 10.1016/j.addr.2014.08.011] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Revised: 07/22/2014] [Accepted: 08/20/2014] [Indexed: 12/15/2022]
Abstract
Skeletal muscle tissue has an inherent capacity for regeneration following injury. However, severe trauma, such as volumetric muscle loss, overwhelms these natural muscle repair mechanisms prompting the search for a tissue engineering/regenerative medicine approach to promote functional skeletal muscle restoration. A desirable approach involves a bioscaffold that simultaneously acts as an inductive microenvironment and as a cell/drug delivery vehicle to encourage muscle ingrowth. Both biologically active, naturally derived materials (such as extracellular matrix) and carefully engineered synthetic polymers have been developed to provide such a muscle regenerative environment. Next generation naturally derived/synthetic "hybrid materials" would combine the advantageous properties of these materials to create an optimal platform for cell/drug delivery and possess inherent bioactive properties. Advances in scaffolds using muscle tissue engineering are reviewed herein.
Collapse
Affiliation(s)
- Matthew T Wolf
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA 15219, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Christopher L Dearth
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA 15219, USA; Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Sonya B Sonnenberg
- Joint Department of Biomedical Engineering at University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
| | - Elizabeth G Loboa
- Joint Department of Biomedical Engineering at University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA; Department of Materials Science & Engineering, North Carolina State University, Raleigh, NC 27695, USA
| | - Stephen F Badylak
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA 15219, USA; Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
40
|
Farouz Y, Chen Y, Terzic A, Menasché P. Concise Review: Growing Hearts in the Right Place: On the Design of Biomimetic Materials for Cardiac Stem Cell Differentiation. Stem Cells 2015; 33:1021-35. [DOI: 10.1002/stem.1929] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 11/10/2014] [Accepted: 12/01/2014] [Indexed: 12/25/2022]
Affiliation(s)
- Yohan Farouz
- Department of Chemistry, Paris Sciences et Lettres, Ecole Normale Supérieure de Paris; CNRS UMR; Paris France
- Sorbonne Paris Cité; Paris Descartes University; Paris France
- INSERM U970; Paris France
| | - Yong Chen
- Department of Chemistry, Paris Sciences et Lettres, Ecole Normale Supérieure de Paris; CNRS UMR; Paris France
| | | | - Philippe Menasché
- Sorbonne Paris Cité; Paris Descartes University; Paris France
- INSERM U970; Paris France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou; Department of Cardiovascular Surgery; Paris France
| |
Collapse
|
41
|
Carlsen RW, Sitti M. Bio-hybrid cell-based actuators for microsystems. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2014; 10:3831-51. [PMID: 24895215 DOI: 10.1002/smll.201400384] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 04/10/2014] [Indexed: 05/25/2023]
Abstract
As we move towards the miniaturization of devices to perform tasks at the nano and microscale, it has become increasingly important to develop new methods for actuation, sensing, and control. Over the past decade, bio-hybrid methods have been investigated as a promising new approach to overcome the challenges of scaling down robotic and other functional devices. These methods integrate biological cells with artificial components and therefore, can take advantage of the intrinsic actuation and sensing functionalities of biological cells. Here, the recent advancements in bio-hybrid actuation are reviewed, and the challenges associated with the design, fabrication, and control of bio-hybrid microsystems are discussed. As a case study, focus is put on the development of bacteria-driven microswimmers, which has been investigated as a targeted drug delivery carrier. Finally, a future outlook for the development of these systems is provided. The continued integration of biological and artificial components is envisioned to enable the performance of tasks at a smaller and smaller scale in the future, leading to the parallel and distributed operation of functional systems at the microscale.
Collapse
Affiliation(s)
- Rika Wright Carlsen
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | | |
Collapse
|
42
|
McAleer CW, Smith AST, Najjar S, Pirozzi K, Long CJ, Hickman JJ. Mechanistic investigation of adult myotube response to exercise and drug treatment in vitro using a multiplexed functional assay system. J Appl Physiol (1985) 2014; 117:1398-405. [PMID: 25301895 DOI: 10.1152/japplphysiol.00612.2014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The ability to accurately measure skeletal muscle functional performance at the single-cell level would be advantageous for exercise physiology studies and disease modeling applications. To that end, this study characterizes the functional response of individual skeletal muscle myotubes derived from adult rodent tissue to creatine treatment and chronic exercise. The observed improvements to functional performance in response to these treatments appear to correlate with alterations in hypertrophic and mitochondrial biogenesis pathways, supporting previously published in vivo and in vitro data, which highlights the role of these pathways in augmenting skeletal muscle output. The developed system represents a multiplexed functional in vitro assay capable of long-term assessment of contractile cellular outputs in real-time that is compatible with concomitant molecular biology analysis. Adoption of this system in drug toxicity and efficacy studies would improve understanding of compound activity on physical cellular outputs and provide more streamlined and predictive data for future preclinical analyses.
Collapse
Affiliation(s)
- C W McAleer
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| | - A S T Smith
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| | - S Najjar
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| | - K Pirozzi
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| | - C J Long
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| | - J J Hickman
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| |
Collapse
|
43
|
Cittadella Vigodarzere G, Mantero S. Skeletal muscle tissue engineering: strategies for volumetric constructs. Front Physiol 2014; 5:362. [PMID: 25295011 PMCID: PMC4170101 DOI: 10.3389/fphys.2014.00362] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 09/03/2014] [Indexed: 12/21/2022] Open
Abstract
Skeletal muscle tissue is characterized by high metabolic requirements, defined structure and high regenerative potential. As such, it constitutes an appealing platform for tissue engineering to address volumetric defects, as proven by recent works in this field. Several issues common to all engineered constructs constrain the variety of tissues that can be realized in vitro, principal among them the lack of a vascular system and the absence of reliable cell sources; as it is, the only successful tissue engineering constructs are not characterized by active function, present limited cellular survival at implantation and possess low metabolic requirements. Recently, functionally competent constructs have been engineered, with vascular structures supporting their metabolic requirements. In addition to the use of biochemical cues, physical means, mechanical stimulation and the application of electric tension have proven effective in stimulating the differentiation of cells and the maturation of the constructs; while the use of co-cultures provided fine control of cellular developments through paracrine activity. This review will provide a brief analysis of some of the most promising improvements in the field, with particular attention to the techniques that could prove easily transferable to other branches of tissue engineering.
Collapse
Affiliation(s)
| | - Sara Mantero
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano Milano, Italy
| |
Collapse
|
44
|
Uzel SGM, Pavesi A, Kamm RD. Microfabrication and microfluidics for muscle tissue models. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2014; 115:279-93. [PMID: 25175338 DOI: 10.1016/j.pbiomolbio.2014.08.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Accepted: 08/19/2014] [Indexed: 12/14/2022]
Abstract
The relatively recent development of microfluidic systems with wide-ranging capabilities for generating realistic 2D or 3D systems with single or multiple cell types has given rise to an extensive collection of platform technologies useful in muscle tissue engineering. These new systems are aimed at (i) gaining fundamental understanding of muscle function, (ii) creating functional muscle constructs in vitro, and (iii) utilizing these constructs a variety of applications. Use of microfluidics to control the various stimuli that promote differentiation of multipotent cells into cardiac or skeletal muscle is first discussed. Next, systems that incorporate muscle cells to produce either 2D sheets or 3D tissues of contractile muscle are described with an emphasis on the more recent 3D platforms. These systems are useful for fundamental studies of muscle biology and can also be incorporated into drug screening assays. Applications are discussed for muscle actuators in the context of microrobotics and in miniaturized biological pumps. Finally, an important area of recent study involves coculture with cell types that either activate muscle or facilitate its function. Limitations of current designs and the potential for improving functionality for a wider range of applications is also discussed, with a look toward using current understanding and capabilities to design systems of greater realism, complexity and functionality.
Collapse
Affiliation(s)
- Sebastien G M Uzel
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Andrea Pavesi
- Singapore MIT Alliance for Research and Technology, BioSystems and Micromechanics, 1 CREATE way, #04-13/14 Enterprise Wing, Singapore 138602, Singapore
| | - Roger D Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Singapore MIT Alliance for Research and Technology, BioSystems and Micromechanics, 1 CREATE way, #04-13/14 Enterprise Wing, Singapore 138602, Singapore; Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA.
| |
Collapse
|
45
|
Juhas M, Bursac N. Roles of adherent myogenic cells and dynamic culture in engineered muscle function and maintenance of satellite cells. Biomaterials 2014; 35:9438-46. [PMID: 25154662 DOI: 10.1016/j.biomaterials.2014.07.035] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 07/21/2014] [Indexed: 01/02/2023]
Abstract
Highly functional engineered skeletal muscle constructs could serve as physiological models of muscle function and regeneration and have utility in therapeutic replacement of damaged or diseased muscle tissue. In this study, we examined the roles of different myogenic cell fractions and culturing conditions in the generation of highly functional engineered muscle. Fibrin-based muscle bundles were fabricated using either freshly-isolated myogenic cells or their adherent fraction pre-cultured for 36 h. Muscle bundles made of these cells were cultured in both static and dynamic conditions and systematically characterized with respect to early myogenic events and contractile function. Following 2 weeks of culture, we observed both individual and synergistic benefits of using the adherent cell fraction and dynamic culture on muscle formation and function. In particular, optimal culture conditions resulted in significant increase in the total cross-sectional muscle area (- 3-fold), myofiber size (- 1.6-fold), myonuclei density (- 1.2-fold), and force generation (- 9-fold) compared to traditional use of freshly-isolated cells and static culture. Curiously, we observed that only a simultaneous use of the adherent cell fraction and dynamic culture resulted in accelerated formation of differentiated myofibers which were critical for providing a niche-like environment for maintenance of a satellite cell pool early during culture. Our study identifies key parameters for engineering large-size, highly functional skeletal muscle tissues with improved ability for retention of functional satellite cells.
Collapse
Affiliation(s)
- Mark Juhas
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
46
|
Three-dimensionally printed biological machines powered by skeletal muscle. Proc Natl Acad Sci U S A 2014; 111:10125-30. [PMID: 24982152 DOI: 10.1073/pnas.1401577111] [Citation(s) in RCA: 224] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Combining biological components, such as cells and tissues, with soft robotics can enable the fabrication of biological machines with the ability to sense, process signals, and produce force. An intuitive demonstration of a biological machine is one that can produce motion in response to controllable external signaling. Whereas cardiac cell-driven biological actuators have been demonstrated, the requirements of these machines to respond to stimuli and exhibit controlled movement merit the use of skeletal muscle, the primary generator of actuation in animals, as a contractile power source. Here, we report the development of 3D printed hydrogel "bio-bots" with an asymmetric physical design and powered by the actuation of an engineered mammalian skeletal muscle strip to result in net locomotion of the bio-bot. Geometric design and material properties of the hydrogel bio-bots were optimized using stereolithographic 3D printing, and the effect of collagen I and fibrin extracellular matrix proteins and insulin-like growth factor 1 on the force production of engineered skeletal muscle was characterized. Electrical stimulation triggered contraction of cells in the muscle strip and net locomotion of the bio-bot with a maximum velocity of ∼ 156 μm s(-1), which is over 1.5 body lengths per min. Modeling and simulation were used to understand both the effect of different design parameters on the bio-bot and the mechanism of motion. This demonstration advances the goal of realizing forward-engineered integrated cellular machines and systems, which can have a myriad array of applications in drug screening, programmable tissue engineering, drug delivery, and biomimetic machine design.
Collapse
|
47
|
Cheng CS, Davis BNJ, Madden L, Bursac N, Truskey GA. Physiology and metabolism of tissue-engineered skeletal muscle. Exp Biol Med (Maywood) 2014; 239:1203-14. [PMID: 24912506 DOI: 10.1177/1535370214538589] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Skeletal muscle is a major target for tissue engineering, given its relative size in the body, fraction of cardiac output that passes through muscle beds, as well as its key role in energy metabolism and diabetes, and the need for therapies for muscle diseases such as muscular dystrophy and sarcopenia. To date, most studies with tissue-engineered skeletal muscle have utilized murine and rat cell sources. On the other hand, successful engineering of functional human muscle would enable different applications including improved methods for preclinical testing of drugs and therapies. Some of the requirements for engineering functional skeletal muscle include expression of adult forms of muscle proteins, comparable contractile forces to those produced by native muscle, and physiological force-length and force-frequency relations. This review discusses the various strategies and challenges associated with these requirements, specific applications with cultured human myoblasts, and future directions.
Collapse
Affiliation(s)
- Cindy S Cheng
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Brittany N J Davis
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Lauran Madden
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| |
Collapse
|
48
|
Bian W, Jackman CP, Bursac N. Controlling the structural and functional anisotropy of engineered cardiac tissues. Biofabrication 2014; 6:024109-24109. [PMID: 24717534 DOI: 10.1088/1758-5082/6/2/024109] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The ability to control the degree of structural and functional anisotropy in 3D engineered cardiac tissues would have high utility for both in vitro studies of cardiac muscle physiology and pathology as well as potential tissue engineering therapies for myocardial infarction. Here, we applied a high aspect ratio soft lithography technique to generate network-like tissue patches seeded with neonatal rat cardiomyocytes. Fabricating longer elliptical pores within the patch networks increased the overall cardiomyocyte and extracellular matrix alignment within the patch. Improved uniformity of cell and matrix alignment yielded an increase in anisotropy of action potential propagation and faster longitudinal conduction velocity (LCV). Cardiac tissue patches with a higher degree of cardiomyocyte alignment and electrical anisotropy also demonstrated greater isometric twitch forces. After two weeks of culture, specific measures of electrical and contractile function (LCV = 26.8 ± 0.8 cm s(-1), specific twitch force = 8.9 ± 1.1 mN mm(-2) for the longest pores studied) were comparable to those of neonatal rat myocardium. We have thus described methodology for engineering of highly functional 3D engineered cardiac tissues with controllable degree of anisotropy.
Collapse
Affiliation(s)
- W Bian
- Department of Anesthesia and Medicine and Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - C P Jackman
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - N Bursac
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| |
Collapse
|
49
|
Bian W, Badie N, Himel HD, Bursac N. Robust T-tubulation and maturation of cardiomyocytes using tissue-engineered epicardial mimetics. Biomaterials 2014; 35:3819-28. [PMID: 24508078 DOI: 10.1016/j.biomaterials.2014.01.045] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 01/20/2014] [Indexed: 01/27/2023]
Abstract
Complex three-dimensional (3-D) heart structure is an important determinant of cardiac electrical and mechanical function. In this study, we set to develop a versatile tissue-engineered system that can promote important aspects of cardiac functional maturation and reproduce variations in myofiber directions present in native ventricular epicardium. We cultured neonatal rat cardiomyocytes within a 3-D hydrogel environment using microfabricated elastomeric molds with hexagonal posts. By varying individual post orientations along the directions derived from diffusion tensor magnetic resonance imaging (DTMRI) maps of human ventricle, we created large (2.5 × 2.5 cm(2)) 3-D cardiac tissue patches with cardiomyocyte alignment that replicated human epicardial fiber orientations. After 3 weeks of culture, the advanced structural and functional maturation of the engineered 3-D cardiac tissues compared to age-matched 2-D monolayers was evident from: 1) the presence of dense, aligned and electromechanically-coupled cardiomyocytes, quiescent fibroblasts, and interspersed capillary-like structures, 2) action potential propagation with near-adult conduction velocity and directional dependence on local cardiomyocyte orientation, and 3) robust formation of T-tubules aligned with Z-disks, co-localization of L-type Ca(2+) channels and ryanodine receptors, and accelerated Ca(2+) transient kinetics. This biomimetic tissue-engineered platform can enable systematic in vitro studies of cardiac structure-function relationships and promote the development of advanced tissue engineering strategies for cardiac repair and regeneration.
Collapse
Affiliation(s)
- Weining Bian
- Department of Biomedical Engineering, Duke University, 3000 Science Dr, Hudson Hall 136, Durham, NC 27708, USA
| | - Nima Badie
- Department of Biomedical Engineering, Duke University, 3000 Science Dr, Hudson Hall 136, Durham, NC 27708, USA
| | - Herman D Himel
- Department of Biomedical Engineering, Duke University, 3000 Science Dr, Hudson Hall 136, Durham, NC 27708, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, 3000 Science Dr, Hudson Hall 136, Durham, NC 27708, USA.
| |
Collapse
|
50
|
Sarvazyan N. Thinking Outside the Heart: Use of Engineered Cardiac Tissue for the Treatment of Chronic Deep Venous Insufficiency. J Cardiovasc Pharmacol Ther 2014; 19:394-401. [PMID: 24500906 DOI: 10.1177/1074248413520343] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This article considers the use of autologous stem cell-derived cardiomyocytes as a novel means to aid venous return. The approach consists of creating external cuffs of engineered heart tissue around vein segments with incompetent or poorly competent valves. The engineered heart tissue cuff prevents distention of the impaired vein segments and aids unidirectional flow by its rhythmic contractions. There appear to be no fundamental limitations to this approach as feasibility of all of the individual components has already been shown. Here, we underline the clinical need for novel ways to treat chronic deep venous insufficiency, review previous research that enabled this approach, consider potential designs of engineered heart tissue cuffs, and outline its advantages and future challenges.
Collapse
Affiliation(s)
- Narine Sarvazyan
- Pharmacology and Physiology Department, The George Washington University School of Medicine and Health Sciences, Washington DC, USA
| |
Collapse
|