1
|
Nguyen ML, Demri N, Lapin B, Di Federico F, Gropplero G, Cayrac F, Hennig K, Gomes ER, Wilhelm C, Roman W, Descroix S. Studying the impact of geometrical and cellular cues on myogenesis with a skeletal muscle-on-chip. LAB ON A CHIP 2024; 24:4147-4160. [PMID: 39072529 DOI: 10.1039/d4lc00417e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
In the skeletal muscle tissue, cells are organized following an anisotropic architecture, which is both required during myogenesis when muscle precursor cells fuse to generate myotubes and for its contractile function. To build an in vitro skeletal muscle tissue, it is therefore essential to develop methods to organize cells in an anisotropic fashion, which can be particularly challenging, especially in 3D. In this study, we present a versatile muscle-on-chip system with adjustable collagen hollow tubes that can be seeded with muscle precursor cells. The collagen acts both as a tube-shaped hollow mold and as an extracellular matrix scaffold that can house other cell types for co-culture. We found that the diameter of the channel affects the organization of the muscle cells and that proper myogenesis was obtained at a diameter of 75 μm. In these conditions, muscle precursor cells fused into long myotubes aligned along these collagen channels, resulting in a fascicle-like structure. These myotubes exhibited actin striations and upregulation of multiple myogenic genes, reflecting their maturation. Moreover, we showed that our chip allowed muscle tissue culture and maturation over a month, with the possibility of fibroblast co-culture embedding in the collagen matrix.
Collapse
Affiliation(s)
- M-L Nguyen
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - N Demri
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - B Lapin
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - F Di Federico
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - G Gropplero
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - F Cayrac
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - K Hennig
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Edgar R Gomes
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - C Wilhelm
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - W Roman
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Australian Regenerative Medicine Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - S Descroix
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| |
Collapse
|
2
|
Bagdasarian IA, Tonmoy TI, Park BH, Morgan JT. In vitro formation and extended culture of highly metabolically active and contractile tissues. PLoS One 2023; 18:e0293609. [PMID: 37910543 PMCID: PMC10619834 DOI: 10.1371/journal.pone.0293609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/16/2023] [Indexed: 11/03/2023] Open
Abstract
3D cell culture models have gained popularity in recent years as an alternative to animal and 2D cell culture models for pharmaceutical testing and disease modeling. Polydimethylsiloxane (PDMS) is a cost-effective and accessible molding material for 3D cultures; however, routine PDMS molding may not be appropriate for extended culture of contractile and metabolically active tissues. Failures can include loss of culture adhesion to the PDMS mold and limited culture surfaces for nutrient and waste diffusion. In this study, we evaluated PDMS molding materials and surface treatments for highly contractile and metabolically active 3D cell cultures. PDMS functionalized with polydopamine allowed for extended culture duration (14.8 ± 3.97 days) when compared to polyethylamine/glutaraldehyde functionalization (6.94 ± 2.74 days); Additionally, porous PDMS extended culture duration (16.7 ± 3.51 days) compared to smooth PDMS (6.33 ± 2.05 days) after treatment with TGF-β2 to increase culture contraction. Porous PDMS additionally allowed for large (13 mm tall × 8 mm diameter) constructs to be fed by diffusion through the mold, resulting in increased cell density (0.0210 ± 0.0049 mean nuclear fraction) compared to controls (0.0045 ± 0.0016 mean nuclear fraction). As a practical demonstration of the flexibility of porous PDMS, we engineered a vascular bioartificial muscle model (VBAM) and demonstrated extended culture of VBAMs anchored with porous PDMS posts. Using this model, we assessed the effect of feeding frequency on VBAM cellularity. Feeding 3×/week significantly increased nuclear fraction at multiple tissue depths relative to 2×/day. VBAM maturation was similarly improved in 3×/week feeding as measured by nuclear alignment (23.49° ± 3.644) and nuclear aspect ratio (2.274 ± 0.0643) relative to 2x/day (35.93° ± 2.942) and (1.371 ± 0.1127), respectively. The described techniques are designed to be simple and easy to implement with minimal training or expense, improving access to dense and/or metabolically active 3D cell culture models.
Collapse
Affiliation(s)
- Isabella A. Bagdasarian
- Department of Bioengineering, University of California, Riverside, CA, United States of America
| | - Thamidul Islam Tonmoy
- Department of Bioengineering, University of California, Riverside, CA, United States of America
| | - B. Hyle Park
- Department of Bioengineering, University of California, Riverside, CA, United States of America
| | - Joshua T. Morgan
- Department of Bioengineering, University of California, Riverside, CA, United States of America
| |
Collapse
|
3
|
Suresh Kumar H, Barnett EN, Fowlkes JL, Kalaitzoglou E, Annamalai RT. Biomechanical Stimulation of Muscle Constructs Influences Phenotype of Bone Constructs by Modulating Myokine Secretion. JBMR Plus 2023; 7:e10804. [PMID: 38025033 PMCID: PMC10652181 DOI: 10.1002/jbm4.10804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/22/2023] [Accepted: 07/24/2023] [Indexed: 12/01/2023] Open
Abstract
Diabetes is a chronic metabolic disorder that can lead to diabetic myopathy and bone diseases. The etiology of musculoskeletal complications in such metabolic disorders and the interplay between the muscular and osseous systems are not well understood. Exercise training promises to prevent diabetic myopathy and bone disease and offer protection. Although the muscle-bone interaction is largely biomechanical, the muscle secretome has significant implications for bone biology. Uncoupling effects of biophysical and biochemical stimuli on the adaptive response of bone during exercise training may offer therapeutic targets for diabetic bone disease. Here, we have developed an in vitro model to elucidate the effects of mechanical strain on myokine secretion and its impact on bone metabolism decoupled from physical stimuli. We developed bone constructs using cross-linked gelatin, which facilitated osteogenic differentiation of osteoprogenitor cells. Then muscle constructs were made from fibrin, which enabled myoblast differentiation and myotube formation. We investigated the myokine expression by muscle constructs under strain regimens replicating endurance (END) and high-intensity interval training (HIIT) in hyperglycemic conditions. In monocultures, both regimens induced higher expression of Il15 and Igf1, whereas END supported more myoblast differentiation and myotube maturation than HIIT. When co-cultured with bone constructs, HIIT regimen increased Glut4 expression in muscle constructs more than END, supporting higher glucose uptake. Likewise, the muscle constructs under the HIIT regimen promoted a healthier and more matured bone phenotype than END. Under static conditions, myostatin (Mstn) expression was significantly downregulated in muscle constructs co-cultured with bone constructs compared with monocultures. Together, our in vitro co-culture system allowed orthogonal manipulation of mechanical strain on muscle constructs while facilitating bone-muscle biochemical cross-talk. Such systems can provide an individualized microenvironment that allows decoupled biomechanical manipulation, help identify molecular targets, and develop engineered therapies for metabolic bone disease. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
| | - Edwina N. Barnett
- Department of Biomedical EngineeringUniversity of KentuckyLexingtonKYUSA
| | - John L. Fowlkes
- Barnstable Brown Diabetes CenterLexingtonKYUSA
- Department of PediatricsUniversity of KentuckyLexingtonKYUSA
| | - Evangelia Kalaitzoglou
- Barnstable Brown Diabetes CenterLexingtonKYUSA
- Department of PediatricsUniversity of KentuckyLexingtonKYUSA
| | | |
Collapse
|
4
|
Kozan NG, Joshi M, Sicherer ST, Grasman JM. Porous biomaterial scaffolds for skeletal muscle tissue engineering. Front Bioeng Biotechnol 2023; 11:1245897. [PMID: 37854885 PMCID: PMC10579822 DOI: 10.3389/fbioe.2023.1245897] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 09/18/2023] [Indexed: 10/20/2023] Open
Abstract
Volumetric muscle loss is a traumatic injury which overwhelms the innate repair mechanisms of skeletal muscle and results in significant loss of muscle functionality. Tissue engineering seeks to regenerate these injuries through implantation of biomaterial scaffolds to encourage endogenous tissue formation and to restore mechanical function. Many types of scaffolds are currently being researched for this purpose. Scaffolds are typically made from either natural, synthetic, or conductive polymers, or any combination therein. A major criterion for the use of scaffolds for skeletal muscle is their porosity, which is essential for myoblast infiltration and myofiber ingrowth. In this review, we summarize the various methods of fabricating porous biomaterial scaffolds for skeletal muscle regeneration, as well as the various types of materials used to make these scaffolds. We provide guidelines for the fabrication of scaffolds based on functional requirements of skeletal muscle tissue, and discuss the general state of the field for skeletal muscle tissue engineering.
Collapse
Affiliation(s)
| | | | | | - Jonathan M. Grasman
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, United States
| |
Collapse
|
5
|
Loi G, Scocozza F, Aliberti F, Rinvenuto L, Cidonio G, Marchesi N, Benedetti L, Ceccarelli G, Conti M. 3D Co-Printing and Substrate Geometry Influence the Differentiation of C2C12 Skeletal Myoblasts. Gels 2023; 9:595. [PMID: 37504474 PMCID: PMC10378771 DOI: 10.3390/gels9070595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/07/2023] [Accepted: 07/22/2023] [Indexed: 07/29/2023] Open
Abstract
Cells are influenced by several biomechanical aspects of their microenvironment, such as substrate geometry. According to the literature, substrate geometry influences the behavior of muscle cells; in particular, the curvature feature improves cell proliferation. However, the effect of substrate geometry on the myogenic differentiation process is not clear and needs to be further investigated. Here, we show that the 3D co-printing technique allows the realization of substrates. To test the influence of the co-printing technique on cellular behavior, we realized linear polycaprolactone substrates with channels in which a fibrinogen-based hydrogel loaded with C2C12 cells was deposited. Cell viability and differentiation were investigated up to 21 days in culture. The results suggest that this technology significantly improves the differentiation at 14 days. Therefore, we investigate the substrate geometry influence by comparing three different co-printed geometries-linear, circular, and hybrid structures (linear and circular features combined). Based on our results, all structures exhibit optimal cell viability (>94%), but the linear pattern allows to increase the in vitro cell differentiation, in particular after 14 days of culture. This study proposes an endorsed approach for creating artificial muscles for future skeletal muscle tissue engineering applications.
Collapse
Affiliation(s)
- Giada Loi
- Department of Civil Engineering and Architecture, University of Pavia, Via Adolfo Ferrata 3, 27100 Pavia, Italy
| | - Franca Scocozza
- Department of Civil Engineering and Architecture, University of Pavia, Via Adolfo Ferrata 3, 27100 Pavia, Italy
| | - Flaminia Aliberti
- Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Via Forlanini 2, 27100 Pavia, Italy
- Fondazione IRCCS Policlinico San Matteo, Center for Inherited Cardiovascular Diseases, Transplant Research Area, 27100 Pavia, Italy
| | - Lorenza Rinvenuto
- Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Via Forlanini 2, 27100 Pavia, Italy
| | - Gianluca Cidonio
- Center for Life Nano- & Neuro-Science (CLN2S), Fondazione Istituto Italiano di Tecnologia, 00161 Rome, Italy
| | - Nicola Marchesi
- Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Via Forlanini 2, 27100 Pavia, Italy
| | - Laura Benedetti
- Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Via Forlanini 2, 27100 Pavia, Italy
| | - Gabriele Ceccarelli
- Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Via Forlanini 2, 27100 Pavia, Italy
| | - Michele Conti
- Department of Civil Engineering and Architecture, University of Pavia, Via Adolfo Ferrata 3, 27100 Pavia, Italy
| |
Collapse
|
6
|
Lee J, Lee H, Jin EJ, Ryu D, Kim GH. 3D bioprinting using a new photo-crosslinking method for muscle tissue restoration. NPJ Regen Med 2023; 8:18. [PMID: 37002225 PMCID: PMC10066283 DOI: 10.1038/s41536-023-00292-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 03/17/2023] [Indexed: 04/03/2023] Open
Abstract
Three-dimensional (3D) bioprinting is a highly effective technique for fabricating cell-loaded constructs in tissue engineering. However, the versatility of fabricating precise and complex cell-loaded hydrogels is limited owing to the poor crosslinking ability of cell-containing hydrogels. Herein, we propose an optic-fiber-assisted bioprinting (OAB) process to efficiently crosslink methacrylated hydrogels. By selecting appropriate processing conditions for the photo-crosslinking technique, we fabricated biofunctional cell-laden structures including methacrylated gelatin (Gelma), collagen, and decellularized extracellular matrix. To apply the method to skeletal muscle regeneration, cell-laden Gelma constructs were processed with a functional nozzle having a topographical cue and an OAB process that could induce a uniaxial alignment of C2C12 and human adipose stem cells (hASCs). Significantly higher degrees of cell alignment and myogenic activities in the cell-laden Gelma structure were observed compared with those in the cell construct that was printed using a conventional crosslinking method. Moreover, an in vivo regenerative potential was observed in volumetric muscle defects in a mouse model. The hASC-laden construct significantly induced greater muscle regeneration than the cell construct without topographical cues. Based on the results, the newly designed bioprinting process can prove to be highly effective in fabricating biofunctional cell-laden constructs for various tissue engineering applications.
Collapse
Affiliation(s)
- JaeYoon Lee
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Hyeongjin Lee
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, Republic of Korea
| | - Eun-Ju Jin
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Dongryeol Ryu
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea.
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea.
| | - Geun Hyung Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea.
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
7
|
Ayran M, Karabulut H, Deniz KI, Akcanli GC, Ulag S, Croitoru AM, Tihăuan BM, Sahin A, Ficai D, Gunduz O, Ficai A. Electrically Triggered Quercetin Release from Polycaprolactone/Bismuth Ferrite Microfibrous Scaffold for Skeletal Muscle Tissue. Pharmaceutics 2023; 15:pharmaceutics15030920. [PMID: 36986781 PMCID: PMC10056538 DOI: 10.3390/pharmaceutics15030920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/27/2023] [Accepted: 03/03/2023] [Indexed: 03/16/2023] Open
Abstract
Skeletal muscle tissue engineering presents a promising avenue to address the limitations pertaining to the regenerative potential of stem cells in case of injury or damage. The objective of this research was to evaluate the effects of utilizing novel microfibrous scaffolds, containing the compound quercetin (Q), on skeletal muscle regeneration. Morphological test results showed us that the combination of bismuth ferrite (BFO), polycaprolactone (PCL), and Q were bonded and well-ordered with each other, and a uniform microfibrous structure was obtained. Antimicrobial susceptibility testing of PCL/BFO/Q was conducted, and microbial reduction was found to be over 90% in the highest concentration of Q-loaded microfibrous scaffolds with the most inhibitory effect on S. aureus strains. Further, biocompatibility was investigated by performing MTT testing, fluorescence testing, and SEM imaging on mesenchymal stem cells (MSCs) to determine whether they could act as suitable microfibrous scaffolds for skeletal muscle tissue engineering. Incremental changes in the concentration of Q led to increased strength and strain, allowing muscles to withstand stretching during the healing process. In addition, electrically conductive microfibrous scaffolds enhanced the drug release capability by revealing that Q can be released significantly more quickly by applying the appropriate electric field, compared with conventional drug-release techniques. These findings suggest a possible use for PCL/BFO/Q microfibrous scaffolds in skeletal muscle regeneration by demonstrating that the combined action of both guidance biomaterials was more successful than Q itself acting alone.
Collapse
Affiliation(s)
- Musa Ayran
- Center for Nanotechnology & Biomaterials Application and Research (NBUAM), Marmara University, Istanbul 34722, Turkey
- Institute of Pure and Applied Sciences, Department of Metallurgical and Materials Engineering, Faculty of Technology, Marmara University, Istanbul 34722, Turkey
| | - Hatice Karabulut
- Center for Nanotechnology & Biomaterials Application and Research (NBUAM), Marmara University, Istanbul 34722, Turkey
- Institute of Pure and Applied Sciences, Department of Metallurgical and Materials Engineering, Faculty of Technology, Marmara University, Istanbul 34722, Turkey
| | - Kudret Irem Deniz
- Center for Nanotechnology & Biomaterials Application and Research (NBUAM), Marmara University, Istanbul 34722, Turkey
- Institute of Pure and Applied Sciences, Department of Metallurgical and Materials Engineering, Faculty of Technology, Marmara University, Istanbul 34722, Turkey
| | - Gamze Ceren Akcanli
- Center for Nanotechnology & Biomaterials Application and Research (NBUAM), Marmara University, Istanbul 34722, Turkey
| | - Songul Ulag
- Center for Nanotechnology & Biomaterials Application and Research (NBUAM), Marmara University, Istanbul 34722, Turkey
- Institute of Pure and Applied Sciences, Department of Metallurgical and Materials Engineering, Faculty of Technology, Marmara University, Istanbul 34722, Turkey
| | - Alexa-Maria Croitoru
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 060042 Bucharest, Romania
- National Centre for Micro- and Nanomaterials, University Politehnica of Bucharest, 060042 Bucharest, Romania
- National Centre for Food Safety, University Politehnica of Bucharest, 060042 Bucharest, Romania
| | - Bianca-Maria Tihăuan
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 060042 Bucharest, Romania
- Research Institute of the University of Bucharest—ICUB, 050567 Bucharest, Romania
- Research & Development for Advanced Biotechnologies and Medical Devices, SC Sanimed International Impex SRL, 087040 Calugareni, Romania
| | - Ali Sahin
- Department of Biochemistry, Faculty of Medicine, Marmara University, Istanbul 34722, Turkey
| | - Denisa Ficai
- National Centre for Micro- and Nanomaterials, University Politehnica of Bucharest, 060042 Bucharest, Romania
- National Centre for Food Safety, University Politehnica of Bucharest, 060042 Bucharest, Romania
- Department of Inorganic Chemistry, Physical Chemistry and Electrochemistry, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 060042 Bucharest, Romania
| | - Oguzhan Gunduz
- Center for Nanotechnology & Biomaterials Application and Research (NBUAM), Marmara University, Istanbul 34722, Turkey
- Institute of Pure and Applied Sciences, Department of Metallurgical and Materials Engineering, Faculty of Technology, Marmara University, Istanbul 34722, Turkey
- Correspondence:
| | - Anton Ficai
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 060042 Bucharest, Romania
- National Centre for Micro- and Nanomaterials, University Politehnica of Bucharest, 060042 Bucharest, Romania
- National Centre for Food Safety, University Politehnica of Bucharest, 060042 Bucharest, Romania
- Academy of Romanian Scientists, Ilfov St. 3, 050044 Bucharest, Romania
| |
Collapse
|
8
|
Lee H, Kim SH, Lee JS, Lee YJ, Lee OJ, Ajiteru O, Sultan MT, Lee SW, Park CH. Functional Skeletal Muscle Regeneration Using Muscle Mimetic Tissue Fabricated by Microvalve-Assisted Coaxial 3D Bioprinting. Adv Healthc Mater 2023; 12:e2202664. [PMID: 36469728 DOI: 10.1002/adhm.202202664] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/23/2022] [Indexed: 12/12/2022]
Abstract
3D-printed artificial skeletal muscle, which mimics the structural and functional characteristics of native skeletal muscle, is a promising treatment method for muscle reconstruction. Although various fabrication techniques for skeletal muscle using 3D bio-printers are studied, it is still challenging to build a functional muscle structure. A strategy using microvalve-assisted coaxial 3D bioprinting in consideration of functional skeletal muscle fabrication is reported. The unit (artificial muscle fascicle: AMF) of muscle mimetic tissue is composed of a core filled with medium-based C2C12 myoblast aggregates as a role of muscle fibers and a photo cross-linkable hydrogel-based shell as a role of connective tissue in muscles that enhances printability and cell adhesion and proliferation. Especially, a microvalve system is applied for the core part with even cell distribution and strong cell-cell interaction. This system enhances myotube formation and consequently shows spontaneous contraction. A multi-printed AMF (artificial muscle tissue: AMT) as a piece of muscle is implanted into the anterior tibia (TA) muscle defect site of immunocompromised rats. As a result, the TA-implanted AMT responds to electrical stimulation and represents histologically regenerated muscle tissue. This microvalve-assisted coaxial 3D bioprinting shows a significant step forward to mimicking native skeletal muscle tissue.
Collapse
Affiliation(s)
- Hanna Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Soon Hee Kim
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Ji Seung Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Young Jin Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Ok Joo Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Olatunji Ajiteru
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Md Tipu Sultan
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Suk Woo Lee
- Department of Obstetrics and Gynecology, Hallym University Sacred Heart Hospital, Anyang, 14068, Republic of Korea
| | - Chan Hum Park
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea.,Department of Otorhinolaryngology-Head and Neck Surgery, Chuncheon Sacred Heart Hospital, School of Medicine, Hallym University, Chuncheon, 24252, Republic of Korea
| |
Collapse
|
9
|
Scala P, Manzo P, Lamparelli EP, Lovecchio J, Ciardulli MC, Giudice V, Selleri C, Giordano E, Rehak L, Maffulli N, Della Porta G. Peripheral blood mononuclear cells contribute to myogenesis in a 3D bioengineered system of bone marrow mesenchymal stem cells and myoblasts. Front Bioeng Biotechnol 2023; 10:1075715. [PMID: 36704300 PMCID: PMC9871311 DOI: 10.3389/fbioe.2022.1075715] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/28/2022] [Indexed: 01/12/2023] Open
Abstract
In this work, a 3D environment obtained using fibrin scaffold and two cell populations, such as bone marrow-derived mesenchymal stem cells (BM-MSCs), and primary skeletal muscle cells (SkMs), was assembled. Peripheral blood mononuclear cells (PBMCs) fraction obtained after blood filtration with HemaTrate® filter was then added to the 3D culture system to explore their influence on myogenesis. The best cell ratio into a 3D fibrin hydrogel was 1:1 (BM-MSCs plus SkMs:PBMCs) when cultured in a perfusion bioreactor; indeed, excellent viability and myogenic event induction were observed. Myogenic genes were significantly overexpressed when cultured with PBMCs, such as MyoD1 of 118-fold at day 14 and Desmin 6-fold at day 21. Desmin and Myosin Heavy Chain were also detected at protein level by immunostaining along the culture. Moreover, the presence of PBMCs in 3D culture induced a significant downregulation of pro-inflammatory cytokine gene expression, such as IL6. This smart biomimetic environment can be an excellent tool for investigation of cellular crosstalk and PBMC influence on myogenic processes.
Collapse
Affiliation(s)
- Pasqualina Scala
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy
| | - Paola Manzo
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy,Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, Salerno, Italy
| | | | - Joseph Lovecchio
- Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi” (DEI), University of Bologna, Bologna, Italy
| | | | - Valentina Giudice
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy,Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, Salerno, Italy
| | - Carmine Selleri
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy,Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, Salerno, Italy
| | - Emanuele Giordano
- Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi” (DEI), University of Bologna, Bologna, Italy
| | - Laura Rehak
- Athena Biomedical innovations, Florence, Italy
| | - Nicola Maffulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy,Centre for Sports and Exercise Medicine, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, England
| | - Giovanna Della Porta
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy,Interdepartment Centre BIONAM, University of Salerno, Fisciano, Italy,*Correspondence: Giovanna Della Porta,
| |
Collapse
|
10
|
Ahmad SS, Chun HJ, Ahmad K, Shaikh S, Lim JH, Ali S, Han SS, Hur SJ, Sohn JH, Lee EJ, Choi I. The roles of growth factors and hormones in the regulation of muscle satellite cells for cultured meat production. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2023; 65:16-31. [PMID: 37093925 PMCID: PMC10119461 DOI: 10.5187/jast.2022.e114] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/25/2022] [Accepted: 11/27/2022] [Indexed: 12/24/2022]
Abstract
Cultured meat is a potential sustainable food generated by the in vitro myogenesis of muscle satellite (stem) cells (MSCs). The self-renewal and differentiation properties of MSCs are of primary interest for cultured meat production. MSC proliferation and differentiation are influenced by a variety of growth factors such as insulin-like growth factors (IGF-1 and IGF-2), transforming growth factor beta (TGF-β), fibroblast growth factors (FGF-2 and FGF-21), platelet-derived growth factor (PDGF) and hepatocyte growth factor (HGF) and by hormones like insulin, testosterone, glucocorticoids, and thyroid hormones. In this review, we investigated the roles of growth factors and hormones during cultured meat production because these factors provide signals for MSC growth and structural stability. The aim of this article is to provide the important idea about different growth factors such as FGF (enhance the cell proliferation and differentiation), IGF-1 (increase the number of myoblasts), PDGF (myoblast proliferation), TGF-β1 (muscle repair) and hormones such as insulin (cell survival and growth), testosterone (muscle fiber size), dexamethasone (myoblast proliferation and differentiation), and thyroid hormones (amount and diameter of muscle fibers and determine the usual pattern of fiber distributions) as media components during myogenesis for cultured meat production.
Collapse
Affiliation(s)
- Syed Sayeed Ahmad
- Department of Medical Biotechnology,
Yeungnam University, Gyeongsan 38541, Korea
- Research Institute of Cell Culture,
Yeungnam University, Gyeongsan 38541, Korea
| | - Hee Jin Chun
- Department of Medical Biotechnology,
Yeungnam University, Gyeongsan 38541, Korea
| | - Khurshid Ahmad
- Department of Medical Biotechnology,
Yeungnam University, Gyeongsan 38541, Korea
- Research Institute of Cell Culture,
Yeungnam University, Gyeongsan 38541, Korea
| | - Sibhghatulla Shaikh
- Department of Medical Biotechnology,
Yeungnam University, Gyeongsan 38541, Korea
- Research Institute of Cell Culture,
Yeungnam University, Gyeongsan 38541, Korea
| | - Jeong Ho Lim
- Department of Medical Biotechnology,
Yeungnam University, Gyeongsan 38541, Korea
- Research Institute of Cell Culture,
Yeungnam University, Gyeongsan 38541, Korea
| | - Shahid Ali
- Department of Medical Biotechnology,
Yeungnam University, Gyeongsan 38541, Korea
- Research Institute of Cell Culture,
Yeungnam University, Gyeongsan 38541, Korea
| | - Sung Soo Han
- Research Institute of Cell Culture,
Yeungnam University, Gyeongsan 38541, Korea
- School of Chemical Engineering, Yeungnam
University, Gyeongsan 38541, Korea
| | - Sun Jin Hur
- Department of Animal Science and
Technology, Chung-Ang University, Anseong 17546, Korea
| | - Jung Hoon Sohn
- Synthetic Biology and Bioengineering
Research Center, Korea Research Institute of Bioscience and Biotechnology
(KRIBB), Daejeon 34141, Korea
| | - Eun Ju Lee
- Department of Medical Biotechnology,
Yeungnam University, Gyeongsan 38541, Korea
- Research Institute of Cell Culture,
Yeungnam University, Gyeongsan 38541, Korea
| | - Inho Choi
- Department of Medical Biotechnology,
Yeungnam University, Gyeongsan 38541, Korea
- Research Institute of Cell Culture,
Yeungnam University, Gyeongsan 38541, Korea
| |
Collapse
|
11
|
Charbe NB, Tambuwala M, Palakurthi SS, Warokar A, Hromić‐Jahjefendić A, Bakshi H, Zacconi F, Mishra V, Khadse S, Aljabali AA, El‐Tanani M, Serrano‐Aroca Ã, Palakurthi S. Biomedical applications of three-dimensional bioprinted craniofacial tissue engineering. Bioeng Transl Med 2023; 8:e10333. [PMID: 36684092 PMCID: PMC9842068 DOI: 10.1002/btm2.10333] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 02/06/2023] Open
Abstract
Anatomical complications of the craniofacial regions often present considerable challenges to the surgical repair or replacement of the damaged tissues. Surgical repair has its own set of limitations, including scarcity of the donor tissues, immune rejection, use of immune suppressors followed by the surgery, and restriction in restoring the natural aesthetic appeal. Rapid advancement in the field of biomaterials, cell biology, and engineering has helped scientists to create cellularized skeletal muscle-like structures. However, the existing method still has limitations in building large, highly vascular tissue with clinical application. With the advance in the three-dimensional (3D) bioprinting technique, scientists and clinicians now can produce the functional implants of skeletal muscles and bones that are more patient-specific with the perfect match to the architecture of their craniofacial defects. Craniofacial tissue regeneration using 3D bioprinting can manage and eliminate the restrictions of the surgical transplant from the donor site. The concept of creating the new functional tissue, exactly mimicking the anatomical and physiological function of the damaged tissue, looks highly attractive. This is crucial to reduce the donor site morbidity and retain the esthetics. 3D bioprinting can integrate all three essential components of tissue engineering, that is, rehabilitation, reconstruction, and regeneration of the lost craniofacial tissues. Such integration essentially helps to develop the patient-specific treatment plans and damage site-driven creation of the functional implants for the craniofacial defects. This article is the bird's eye view on the latest development and application of 3D bioprinting in the regeneration of the skeletal muscle tissues and their application in restoring the functional abilities of the damaged craniofacial tissue. We also discussed current challenges in craniofacial bone vascularization and gave our view on the future direction, including establishing the interactions between tissue-engineered skeletal muscle and the peripheral nervous system.
Collapse
Affiliation(s)
- Nitin Bharat Charbe
- Irma Lerma Rangel College of PharmacyTexas A&M Health Science CenterKingsvilleTexasUSA
| | - Murtaza Tambuwala
- School of Pharmacy and Pharmaceutical ScienceUlster UniversityColeraineUK
| | | | - Amol Warokar
- Department of PharmacyDadasaheb Balpande College of PharmacyNagpurIndia
| | - Altijana Hromić‐Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural SciencesInternational University of SarajevoSarajevoBosnia and Herzegovina
| | - Hamid Bakshi
- School of Pharmacy and Pharmaceutical ScienceUlster UniversityColeraineUK
| | - Flavia Zacconi
- Departamento de Quimica Orgánica, Facultad de Química y de FarmaciaPontificia Universidad Católica de ChileSantiagoChile
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological SciencesPontificia Universidad Católica de ChileSantiagoChile
| | - Vijay Mishra
- School of Pharmaceutical SciencesLovely Professional UniversityPhagwaraIndia
| | - Saurabh Khadse
- Department of Pharmaceutical ChemistryR.C. Patel Institute of Pharmaceutical Education and ResearchDhuleIndia
| | - Alaa A. Aljabali
- Faculty of Pharmacy, Department of Pharmaceutical SciencesYarmouk UniversityIrbidJordan
| | - Mohamed El‐Tanani
- Pharmacological and Diagnostic Research Centre, Faculty of PharmacyAl‐Ahliyya Amman UniversityAmmanJordan
| | - Ãngel Serrano‐Aroca
- Biomaterials and Bioengineering Lab Translational Research Centre San Alberto MagnoCatholic University of Valencia San Vicente MártirValenciaSpain
| | - Srinath Palakurthi
- Irma Lerma Rangel College of PharmacyTexas A&M Health Science CenterKingsvilleTexasUSA
| |
Collapse
|
12
|
Vesga-Castro C, Aldazabal J, Vallejo-Illarramendi A, Paredes J. Contractile force assessment methods for in vitro skeletal muscle tissues. eLife 2022; 11:e77204. [PMID: 35604384 PMCID: PMC9126583 DOI: 10.7554/elife.77204] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/27/2022] [Indexed: 02/06/2023] Open
Abstract
Over the last few years, there has been growing interest in measuring the contractile force (CF) of engineered muscle tissues to evaluate their functionality. However, there are still no standards available for selecting the most suitable experimental platform, measuring system, culture protocol, or stimulation patterns. Consequently, the high variability of published data hinders any comparison between different studies. We have identified that cantilever deflection, post deflection, and force transducers are the most commonly used configurations for CF assessment in 2D and 3D models. Additionally, we have discussed the most relevant emerging technologies that would greatly complement CF evaluation with intracellular and localized analysis. This review provides a comprehensive analysis of the most significant advances in CF evaluation and its critical parameters. In order to compare contractile performance across experimental platforms, we have used the specific force (sF, kN/m2), CF normalized to the calculated cross-sectional area (CSA). However, this parameter presents a high variability throughout the different studies, which indicates the need to identify additional parameters and complementary analysis suitable for proper comparison. We propose that future contractility studies in skeletal muscle constructs report detailed information about construct size, contractile area, maturity level, sarcomere length, and, ideally, the tetanus-to-twitch ratio. These studies will hopefully shed light on the relative impact of these variables on muscle force performance of engineered muscle constructs. Prospective advances in muscle tissue engineering, particularly in muscle disease models, will require a joint effort to develop standardized methodologies for assessing CF of engineered muscle tissues.
Collapse
Affiliation(s)
- Camila Vesga-Castro
- University of Navarra, Tecnun School of Engineering, Manuel de LardizábalSan SebastianSpain
- University of Navarra, Biomedical Engineering Center, Campus UniversitarioPamplonaSpain
- Group of Neurosciences, Department of Pediatrics, UPV/EHU, Hospital Donostia - IIS BiodonostiaSan SebastianSpain
| | - Javier Aldazabal
- University of Navarra, Tecnun School of Engineering, Manuel de LardizábalSan SebastianSpain
- University of Navarra, Biomedical Engineering Center, Campus UniversitarioPamplonaSpain
| | - Ainara Vallejo-Illarramendi
- Group of Neurosciences, Department of Pediatrics, UPV/EHU, Hospital Donostia - IIS BiodonostiaSan SebastianSpain
- CIBERNED, Instituto de Salud Carlos III, Ministry of Science, Innovation, and UniversitiesMadridSpain
| | - Jacobo Paredes
- University of Navarra, Tecnun School of Engineering, Manuel de LardizábalSan SebastianSpain
- University of Navarra, Biomedical Engineering Center, Campus UniversitarioPamplonaSpain
| |
Collapse
|
13
|
Tomasch J, Maleiner B, Heher P, Rufin M, Andriotis OG, Thurner PJ, Redl H, Fuchs C, Teuschl-Woller AH. Changes in Elastic Moduli of Fibrin Hydrogels Within the Myogenic Range Alter Behavior of Murine C2C12 and Human C25 Myoblasts Differently. Front Bioeng Biotechnol 2022; 10:836520. [PMID: 35669058 PMCID: PMC9164127 DOI: 10.3389/fbioe.2022.836520] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Fibrin hydrogels have proven highly suitable scaffold materials for skeletal muscle tissue engineering in the past. Certain parameters of those types of scaffolds, however, greatly affect cellular mechanobiology and therefore the myogenic outcome. The aim of this study was to identify the influence of apparent elastic properties of fibrin scaffolds in 2D and 3D on myoblasts and evaluate if those effects differ between murine and human cells. Therefore, myoblasts were cultured on fibrin-coated multiwell plates ("2D") or embedded in fibrin hydrogels ("3D") with different elastic moduli. Firstly, we established an almost linear correlation between hydrogels' fibrinogen concentrations and apparent elastic moduli in the range of 7.5 mg/ml to 30 mg/ml fibrinogen (corresponds to a range of 7.7-30.9 kPa). The effects of fibrin hydrogel elastic modulus on myoblast proliferation changed depending on culture type (2D vs 3D) with an inhibitory effect at higher fibrinogen concentrations in 3D gels and vice versa in 2D. The opposite effect was evident in differentiating myoblasts as shown by gene expression analysis of myogenesis marker genes and altered myotube morphology. Furthermore, culture in a 3D environment slowed down proliferation compared to 2D, with a significantly more pronounced effect on human myoblasts. Differentiation potential was also substantially impaired upon incorporation into 3D gels in human, but not in murine, myoblasts. With this study, we gained further insight in the influence of apparent elastic modulus and culture type on cellular behavior and myogenic outcome of skeletal muscle tissue engineering approaches. Furthermore, the results highlight the need to adapt parameters of 3D culture setups established for murine cells when applied to human cells.
Collapse
Affiliation(s)
- Janine Tomasch
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Babette Maleiner
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Philipp Heher
- Ludwig Randall Centre for Cell and Molecular Biophysics, King’s College London, Guy’s Campus, London, United Kingdom
| | - Manuel Rufin
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Institute of Lightweight Design and Structural Biomechanics, TU Wien, Vienna, Austria
| | - Orestis G. Andriotis
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Institute of Lightweight Design and Structural Biomechanics, TU Wien, Vienna, Austria
| | - Philipp J. Thurner
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Institute of Lightweight Design and Structural Biomechanics, TU Wien, Vienna, Austria
| | - Heinz Redl
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria
| | - Christiane Fuchs
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Wellman Center for Photomedicine, MGH, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Andreas H. Teuschl-Woller
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
14
|
Colapicchioni V, Millozzi F, Parolini O, Palacios D. Nanomedicine, a valuable tool for skeletal muscle disorders: Challenges, promises, and limitations. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1777. [PMID: 35092179 PMCID: PMC9285803 DOI: 10.1002/wnan.1777] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 12/24/2021] [Accepted: 01/06/2022] [Indexed: 12/15/2022]
Abstract
Muscular dystrophies are a group of rare genetic disorders characterized by progressive muscle weakness, which, in the most severe forms, leads to the patient's death due to cardiorespiratory problems. There is still no cure available for these diseases and significant effort is being placed into developing new strategies to either correct the genetic defect or to compensate muscle loss by stimulating skeletal muscle regeneration. However, the vast anatomical extension of the target tissue poses great challenges to these goals, highlighting the need for complementary strategies. Nanomedicine is an actively evolving field that merges nanotechnologies with biomedical and pharmaceutical sciences. It holds great potential in regenerative medicine, both in supporting tissue engineering and regeneration, and in optimizing drug and oligonucleotide delivery and gene therapy strategies. In this review, we will summarize the state‐of‐the‐art in the field of nanomedicine applied to skeletal muscle regeneration. We will discuss the recent work toward the development of nanopatterned scaffolds for tissue engineering, the efforts in the synthesis of organic and inorganic nanoparticles for gene therapy and drug delivery applications, as well as their use as immune modulators. Although nanomedicine holds great promise for muscle and other degenerative diseases, many challenges still need to be systematically addressed to assure a smooth transition from the bench to the bedside. This article is categorized under:Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement
Collapse
Affiliation(s)
- Valentina Colapicchioni
- Italian National Research Council, Institute for Atmospheric Pollution Research (CNR-IIA), Rome, Italy.,Mhetra LLC, Miami, Florida, USA
| | - Francesco Millozzi
- Histology and Embryology Unit, DAHFMO, Sapienza University, Rome, Italy.,IRCCS Santa Lucia Foundation, Rome, Italy
| | - Ornella Parolini
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy.,IRCCS Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Daniela Palacios
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy.,IRCCS Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
15
|
Cheesbrough A, Sciscione F, Riccio F, Harley P, R'Bibo L, Ziakas G, Darbyshire A, Lieberam I, Song W. Biobased Elastomer Nanofibers Guide Light-Controlled Human-iPSC-Derived Skeletal Myofibers. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2110441. [PMID: 35231133 PMCID: PMC9131876 DOI: 10.1002/adma.202110441] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/25/2022] [Indexed: 05/07/2023]
Abstract
Generating skeletal muscle tissue that mimics the cellular alignment, maturation, and function of native skeletal muscle is an ongoing challenge in disease modeling and regenerative therapies. Skeletal muscle cultures require extracellular guidance and mechanical support to stabilize contractile myofibers. Existing microfabrication-based solutions are limited by complex fabrication steps, low throughput, and challenges in measuring dynamic contractile function. Here, the synthesis and characterization of a new biobased nanohybrid elastomer, which is electrospun into aligned nanofiber sheets to mimic the skeletal muscle extracellular matrix, is presented. The polymer exhibits remarkable hyperelasticity well-matched to that of native skeletal muscle (≈11-50 kPa), with ultimate strain ≈1000%, and elastic modulus ≈25 kPa. Uniaxially aligned nanofibers guide myoblast alignment, enhance sarcomere formation, and promote a ≈32% increase in myotube fusion and ≈50% increase in myofiber maturation. The elastomer nanofibers stabilize optogenetically controlled human induced pluripotent stem cell derived skeletal myofibers. When activated by blue light, the myofiber-nanofiber hybrid constructs maintain a significantly higher (>200%) contraction velocity and specific force (>280%) compared to conventional culture methods. The engineered myofibers exhibit a power density of ≈35 W m-3 . This system is a promising new skeletal muscle tissue model for applications in muscular disease modeling, drug discovery, and muscle regeneration.
Collapse
Affiliation(s)
- Aimee Cheesbrough
- UCL Centre for Biomaterials in Surgical Reconstruction and RegenerationDepartment of Surgical BiotechnologyDivision of Surgery and Interventional ScienceUniversity College LondonLondonNW3 2PFUK
- Centre for Gene Therapy and Regenerative MedicineMRC Centre for Neurodevelopmental DisordersCentre for Developmental NeurobiologyKings College LondonLondonSE1 9RTUK
| | - Fabiola Sciscione
- UCL Centre for Biomaterials in Surgical Reconstruction and RegenerationDepartment of Surgical BiotechnologyDivision of Surgery and Interventional ScienceUniversity College LondonLondonNW3 2PFUK
| | - Federica Riccio
- Centre for Gene Therapy and Regenerative MedicineMRC Centre for Neurodevelopmental DisordersCentre for Developmental NeurobiologyKings College LondonLondonSE1 9RTUK
| | - Peter Harley
- Centre for Gene Therapy and Regenerative MedicineMRC Centre for Neurodevelopmental DisordersCentre for Developmental NeurobiologyKings College LondonLondonSE1 9RTUK
| | - Lea R'Bibo
- Centre for Gene Therapy and Regenerative MedicineMRC Centre for Neurodevelopmental DisordersCentre for Developmental NeurobiologyKings College LondonLondonSE1 9RTUK
| | - Georgios Ziakas
- UCL Centre for Biomaterials in Surgical Reconstruction and RegenerationDepartment of Surgical BiotechnologyDivision of Surgery and Interventional ScienceUniversity College LondonLondonNW3 2PFUK
| | - Arnold Darbyshire
- UCL Centre for Biomaterials in Surgical Reconstruction and RegenerationDepartment of Surgical BiotechnologyDivision of Surgery and Interventional ScienceUniversity College LondonLondonNW3 2PFUK
| | - Ivo Lieberam
- Centre for Gene Therapy and Regenerative MedicineMRC Centre for Neurodevelopmental DisordersCentre for Developmental NeurobiologyKings College LondonLondonSE1 9RTUK
| | - Wenhui Song
- UCL Centre for Biomaterials in Surgical Reconstruction and RegenerationDepartment of Surgical BiotechnologyDivision of Surgery and Interventional ScienceUniversity College LondonLondonNW3 2PFUK
| |
Collapse
|
16
|
Yamamoto K, Ohsumi S, Nagashima T, Akiyama H, Honda H, Shimizu K. Screening of anti-atrophic peptides by using photo-cleavable peptide array and 96-well scale contractile human skeletal muscle atrophy models. Biotechnol Bioeng 2022; 119:2196-2205. [PMID: 35478456 DOI: 10.1002/bit.28125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 04/08/2022] [Accepted: 04/26/2022] [Indexed: 11/08/2022]
Abstract
Skeletal muscle atrophy is characterized by decreases in protein content, myofiber diameter, and contractile force generation. As muscle atrophy worsens the quality of life, the development of anti-atrophic substances is desirable. In this study, we aimed to demonstrate a screening process for anti-atrophic peptides using photo-cleavable peptide array technology and human contractile atrophic muscle models. We developed a 96-well system, and established a screening process with less variability. Dexamethasone-induced human atrophic tissue was constructed on the system. Eight peptides were selected from the literature and used for the screening of peptides for preventing the decrease of the contractile forces of tissues. The peptide QIGFIW, which showed preventive activity, was selected as the seed sequence. As a result of amino acid substitution, we obtained QIGFIQ as a peptide with higher anti-atrophic activity. These results indicate that the combinatorial use of the photo-cleavable peptide array technology and 96-well screening system could comprise a powerful approach to obtaining anti-atrophic peptides, and suggest that the 96-well screening system and atrophic model represent a practical and powerful tool for the development of drugs/functional food ingredients. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Kazuki Yamamoto
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, 464-8603, Japan
| | - Saki Ohsumi
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, 464-8603, Japan
| | - Takunori Nagashima
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, 464-8603, Japan
| | - Hirokazu Akiyama
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, 464-8603, Japan
| | - Hiroyuki Honda
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, 464-8603, Japan
| | - Kazunori Shimizu
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, 464-8603, Japan
| |
Collapse
|
17
|
Sugimoto T, Imai S, Yoshikawa M, Fujisato T, Hashimoto T, Nakamura T. Mechanical unloading in 3D-engineered muscle leads to muscle atrophy by suppressing protein synthesis. J Appl Physiol (1985) 2022; 132:1091-1103. [PMID: 35297688 DOI: 10.1152/japplphysiol.00323.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Three dimensional (3D)-engineered muscle is an useful approach to a more comprehensive understanding of molecular mechanisms underlying unloading-induced muscle atrophy. We investigated the effects of mechanical unloading on molecular muscle protein synthesis (MPS)- and muscle protein breakdown (MPB)-related signaling pathways involved in muscle atrophy in 3D-engineered muscle, and to better understand in vitro model of muscle disuse. The 3D-engineered muscle consisting of C2C12 myoblasts and type-1 collagen gel was allowed to differentiate for 2 weeks and divided into three groups: 0 days of stretched-on control (CON), 2 and/or 7 days of stretched-on (ON), in which both ends of the muscle were fixed with artificial tendons, and the stretched-off group (OFF), in which one side of the artificial tendon was detached. Muscle weight (-38.1 to -48.4%), length (-67.0 to -73.5%), twitch contractile force (-70.5 to -75.0%) and myosin heavy chain expression (-32.5 to -50.5%) in the OFF group were significantly decreased on days 2 and 7 compared with the ON group (P < 0.05, respectively), despite that ON group was stable over time. Although determinative molecular signaling could not be identified, the MPS rate reflected by puromysin labeled protein was significantly decreased following mechanical unloading (P < 0.05, -38.5 to -51.1%). Meanwhile, MPB, particularly the ubiquitin-proteasome pathway, was not impacted. Hence, mechanical unloading of 3D-engineered muscle in vitro leads to muscle atrophy by suppressing MPS, cell differentiation, and cell growth rather than the promotion of MPB.
Collapse
Affiliation(s)
- Takeshi Sugimoto
- Faculty of Sport and Health Science, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Shoma Imai
- Division of Human Sciences, Faculty of Engineering, Osaka Institute of Technology, Ohmiya, Osaka, Japan
| | - Maki Yoshikawa
- Faculty of Sport and Health Science, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Toshia Fujisato
- Biomedical Engineering Graduate School of Engineering, Osaka Institute of Technology, Ohmiya, Osaka, Japan
| | - Takeshi Hashimoto
- Faculty of Sport and Health Science, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Tomohiro Nakamura
- Division of Human Sciences, Faculty of Engineering, Osaka Institute of Technology, Ohmiya, Osaka, Japan
| |
Collapse
|
18
|
Ziemkiewicz N, Hilliard GM, Dunn AJ, Madsen J, Haas G, Au J, Genovese PC, Chauvin HM, West C, Paoli A, Garg K. Laminin-111-Enriched Fibrin Hydrogels Enhance Functional Muscle Regeneration Following Trauma. Tissue Eng Part A 2022; 28:297-311. [PMID: 34409846 DOI: 10.1089/ten.tea.2021.0096] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Volumetric muscle loss (VML) is the surgical or traumatic loss of skeletal muscle, which can cause loss of limb function or permanent disability. VML injuries overwhelms the endogenous regenerative capacity of skeletal muscle and results in poor functional healing outcomes. Currently, there are no approved tissue engineering treatments for VML injuries. In this study, fibrin hydrogels enriched with laminin-111 (LM-111; 50-450 μg/mL) were used for the treatment of VML of the tibialis anterior in a rat model. Treatment with fibrin hydrogel containing 450 μg/mL of LM-111 (FBN450) improved muscle regeneration following VML injury. FBN450 hydrogel treatment increased the relative proportion of contractile to fibrotic tissue as indicated by the myosin: collagen ratio on day 28 post-VML injury. FBN450 hydrogels also enhanced myogenic protein expression and increased the quantity of small to medium size myofibers (500-2000 μm2) as well as innervated myofibers. Improved contractile tissue deposition due to FBN450 hydrogel treatment resulted in a significant improvement (∼60%) in torque production at day 28 postinjury. Taken together, these results suggest that the acellular FBN450 hydrogels provide a promising therapeutic strategy for VML that is worthy of further investigation.
Collapse
Affiliation(s)
- Natalia Ziemkiewicz
- Department of Biomedical Engineering, Parks College of Engineering, Aviation and Technology, Saint Louis University, St. Louis, Missouri, USA
| | - Genevieve M Hilliard
- Department of Biomedical Engineering, Parks College of Engineering, Aviation and Technology, Saint Louis University, St. Louis, Missouri, USA
| | - Andrew J Dunn
- Department of Biomedical Engineering, Parks College of Engineering, Aviation and Technology, Saint Louis University, St. Louis, Missouri, USA
| | - Josh Madsen
- Department of Biomedical Engineering, Parks College of Engineering, Aviation and Technology, Saint Louis University, St. Louis, Missouri, USA
| | - Gabriel Haas
- Department of Biomedical Engineering, Parks College of Engineering, Aviation and Technology, Saint Louis University, St. Louis, Missouri, USA
| | - Jeffrey Au
- Department of Biomedical Engineering, Parks College of Engineering, Aviation and Technology, Saint Louis University, St. Louis, Missouri, USA
| | - Peter C Genovese
- Department of Biomedical Engineering, Parks College of Engineering, Aviation and Technology, Saint Louis University, St. Louis, Missouri, USA
| | - Hannah M Chauvin
- Department of Biomedical Engineering, Parks College of Engineering, Aviation and Technology, Saint Louis University, St. Louis, Missouri, USA
| | - Charles West
- Department of Biomedical Engineering, Parks College of Engineering, Aviation and Technology, Saint Louis University, St. Louis, Missouri, USA
| | - Allison Paoli
- Department of Biomedical Engineering, Parks College of Engineering, Aviation and Technology, Saint Louis University, St. Louis, Missouri, USA
| | - Koyal Garg
- Department of Biomedical Engineering, Parks College of Engineering, Aviation and Technology, Saint Louis University, St. Louis, Missouri, USA
| |
Collapse
|
19
|
The contracture-in-a-well. An in vitro model distinguishes bulk and interfacial processes of irreversible (fibrotic) cell-mediated contraction. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2022; 133:112661. [DOI: 10.1016/j.msec.2022.112661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 11/21/2022]
|
20
|
Romagnoli C, Iantomasi T, Brandi ML. Available In Vitro Models for Human Satellite Cells from Skeletal Muscle. Int J Mol Sci 2021; 22:ijms222413221. [PMID: 34948017 PMCID: PMC8706222 DOI: 10.3390/ijms222413221] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/01/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Skeletal muscle accounts for almost 40% of the total adult human body mass. This tissue is essential for structural and mechanical functions such as posture, locomotion, and breathing, and it is endowed with an extraordinary ability to adapt to physiological changes associated with growth and physical exercise, as well as tissue damage. Moreover, skeletal muscle is the most age-sensitive tissue in mammals. Due to aging, but also to several diseases, muscle wasting occurs with a loss of muscle mass and functionality, resulting from disuse atrophy and defective muscle regeneration, associated with dysfunction of satellite cells, which are the cells responsible for maintaining and repairing adult muscle. The most established cell lines commonly used to study muscle homeostasis come from rodents, but there is a need to study skeletal muscle using human models, which, due to ethical implications, consist primarily of in vitro culture, which is the only alternative way to vertebrate model organisms. This review will survey in vitro 2D/3D models of human satellite cells to assess skeletal muscle biology for pre-clinical investigations and future directions.
Collapse
Affiliation(s)
- Cecilia Romagnoli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (C.R.); (T.I.)
| | - Teresa Iantomasi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (C.R.); (T.I.)
| | - Maria Luisa Brandi
- F.I.R.M.O. Italian Foundation for the Research on Bone Diseases, Via Reginaldo Giuliani 195/A, 50141 Florence, Italy
- Correspondence:
| |
Collapse
|
21
|
Yeo M, Kim G. Electrohydrodynamic-direct-printed cell-laden microfibrous structure using alginate-based bioink for effective myotube formation. Carbohydr Polym 2021; 272:118444. [PMID: 34420709 DOI: 10.1016/j.carbpol.2021.118444] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 11/26/2022]
Abstract
In this study, a fully aligned microfibrous structure fabricated using fibrin-assisted alginate bioink and electrohydrodynamic direct-printing was proposed for skeletal muscle tissue engineering. To safely construct the aligned alginate/fibrin microfibrous structure laden with myoblasts or endothelial cells, various printing conditions, such as an applied electric field, distance between the nozzle and target, and nozzle moving speed, were selected appropriately. Furthermore, to accelerate the formation of myotubes more efficiently, the alginate/fibrin bioink with vascular endothelial cells was co-printed into a spatially patterned structure within a myoblast-laden structure. The myoblast-laden structure co-cultured with endothelial cells presented fully aligned myotube formation and significantly greater myogenic differentiation compared to the myoblast-laden structure without the endothelial cells owing to the more abundant secretion of angiogenic cytokines. Also, when adipose stem cell- and endothelial cell-laden fibrous structure was implanted in a mouse volumetric muscle loss model, accelerated volumetric muscle repair was observed compared to the defect model. Based on the results, this study demonstrates an alginate-based bioink and new bio-fabricating method to obtain microfibrous cell-laden alginate/fibrin structures with mechanically stable and topographical cues. The proposed method can provide a myoblast/endothelial cell-laden fibrous alginate structure to efficiently induce engineering of skeletal muscle tissue, which could be used in muscle-on-a-chip or recovering structures of volumetric muscle defects.
Collapse
Affiliation(s)
- Miji Yeo
- Department of Biomechatronic Engineering, College of Biotechnology and Bioengineering, Sungkyunkwan University (SKKU), Suwon 16419, South Korea
| | - GeunHyung Kim
- Department of Biomechatronic Engineering, College of Biotechnology and Bioengineering, Sungkyunkwan University (SKKU), Suwon 16419, South Korea; Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, South Korea.
| |
Collapse
|
22
|
Hameed P, Manivasagam G. An overview of bio-actuation in collagen hydrogels: a mechanobiological phenomenon. Biophys Rev 2021; 13:387-403. [PMID: 34178172 PMCID: PMC8214648 DOI: 10.1007/s12551-021-00804-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 05/04/2021] [Indexed: 12/26/2022] Open
Abstract
Due to their congruity with the native extracellular matrix and their ability to assist in soft tissue repair, hydrogels have been touted as a matrix mimicking biomaterial. Hydrogels are one of the prevalent scaffolds used for 3D cell culture. They can exhibit actuation in response to various stimuli like a magnetic field, electric field, mechanical force, temperature, or pH. In 3D cell culture, the traction exerted by cells on hydrogel can induce non-periodic mechanobiological movements (shrinking or folding) called 'bio-actuation'. Interestingly, this hydrogel 'tropism' phenomenon in 3D cell cultures can be exploited to devise hydrogel-cell-based actuators for tissue engineering. This review briefs about the discrepancies in 2D vs. 3D cell culturing on hydrogels and discusses on different types of cell migration occurring inside the hydrogel matrix. It substantiates the role of mechanical stimuli (such as stiffness) exhibited by the collagen-based hydrogel used for 3D cell culture and its influence in governing the lineage commitment of stem cells. Lastly, the review also audits the cytoskeleton proteins present in cells responsible for influencing the actuation of collagen hydrogel and also elaborates on the cellular signaling pathways responsible for actuation of collagen hydrogels.
Collapse
Affiliation(s)
- Pearlin Hameed
- Centre for Biomaterials Cellular and Molecular Theranostics, Vellore Institute of Technology, Vellore, 632014 India
| | - Geetha Manivasagam
- Centre for Biomaterials Cellular and Molecular Theranostics, Vellore Institute of Technology, Vellore, 632014 India
| |
Collapse
|
23
|
Dessauge F, Schleder C, Perruchot MH, Rouger K. 3D in vitro models of skeletal muscle: myopshere, myobundle and bioprinted muscle construct. Vet Res 2021; 52:72. [PMID: 34011392 PMCID: PMC8136231 DOI: 10.1186/s13567-021-00942-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/15/2021] [Indexed: 12/25/2022] Open
Abstract
Typical two-dimensional (2D) culture models of skeletal muscle-derived cells cannot fully recapitulate the organization and function of living muscle tissues, restricting their usefulness in in-depth physiological studies. The development of functional 3D culture models offers a major opportunity to mimic the living tissues and to model muscle diseases. In this respect, this new type of in vitro model significantly increases our understanding of the involvement of the different cell types present in the formation of skeletal muscle and their interactions, as well as the modalities of response of a pathological muscle to new therapies. This second point could lead to the identification of effective treatments. Here, we report the significant progresses that have been made the last years to engineer muscle tissue-like structures, providing useful tools to investigate the behavior of resident cells. Specifically, we interest in the development of myopshere- and myobundle-based systems as well as the bioprinting constructs. The electrical/mechanical stimulation protocols and the co-culture systems developed to improve tissue maturation process and functionalities are presented. The formation of these biomimetic engineered muscle tissues represents a new platform to study skeletal muscle function and spatial organization in large number of physiological and pathological contexts.
Collapse
|
24
|
Ajiteru O, Choi KY, Lim TH, Kim DY, Hong H, Lee YJ, Lee JS, Lee H, Suh YJ, Sultan MT, Lee OJ, Kim SH, Park CH. A digital light processing 3D printed magnetic bioreactor system using silk magnetic bioink. Biofabrication 2021; 13. [PMID: 33887719 DOI: 10.1088/1758-5090/abfaee] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 04/22/2021] [Indexed: 12/13/2022]
Abstract
Among various bioreactors used in the field of tissue engineering and regenerative medicine, a magnetic bioreactor is more capable of providing steady force to the cells while avoiding direct manipulation of the materials. However, most of them are complex and difficult to fabricate, with drawbacks in terms of consistency and biocompatibility. In this study, a magnetic bioreactor system and a magnetic hydrogel were manufactured by single-stage three-dimensional (3D) printing with digital light processing (DLP) technique for differentiation of myoblast cells. The hydrogel was composed of a magnetic part containing iron oxide and glycidyl-methacrylated silk fibroin, and a cellular part printed by adding mouse myoblast cell (C2C12) to gelatin glycidyl methacrylate, that was placed in the magnetic bioreactor system to stimulate the cells in the hydrogel. The composite hydrogel was steadily printed by a one-stage layering technique using a DLP printer. The magnetic bioreactor offered mechanical stretching of the cells in the hydrogel in 3D ways, so that the cellular differentiation could be executed in three dimensions just like the human environment. Cell viability, as well as gene expression using quantitative reverse transcription-polymerase chain reaction, were assessed after magneto-mechanical stimulation of the myoblast cell-embedded hydrogel in the magnetic bioreactor system. Comparison with the control group revealed that the magnetic bioreactor system accelerated differentiation of mouse myoblast cells in the hydrogel and increased myotube diameter and lengthin vitro. The DLP-printed magnetic bioreactor and the hydrogel were simply manufactured and easy-to-use, providing an efficient environment for applying noninvasive mechanical force via FDA-approved silk fibroin and iron oxide biocomposite hydrogel, to stimulate cells without any evidence of cytotoxicity, demonstrating the potential for application in muscle tissue engineering.
Collapse
Affiliation(s)
- Olatunji Ajiteru
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Kyu Young Choi
- Department of Otorhinolaryngology-Head and Neck Surgery, Hallym University College of Medicine, Kangnam Sacred Heart Hospital, Seoul 07441, Republic of Korea
| | - Tae Hyeon Lim
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Do Yeon Kim
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Heesun Hong
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Young Jin Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Ji Seung Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Hanna Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Ye Ji Suh
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Md Tipu Sultan
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Ok Joo Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Soon Hee Kim
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Chan Hum Park
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do 24252, Republic of Korea.,Department of Otorhinolaryngology-Head and Neck Surgery, Chuncheon Sacred Heart Hospital, School of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| |
Collapse
|
25
|
Acosta FM, Jia UTA, Stojkova K, Howland KK, Guda T, Pacelli S, Brey EM, Rathbone CR. Diabetic Conditions Confer Metabolic and Structural Modifications to Tissue-Engineered Skeletal Muscle. Tissue Eng Part A 2021; 27:549-560. [PMID: 32878567 PMCID: PMC8126424 DOI: 10.1089/ten.tea.2020.0138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/06/2020] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle is a tissue that is directly involved in the progression and persistence of type 2 diabetes (T2D), a disease that is becoming increasingly common. Gaining better insight into the mechanisms that are affecting skeletal muscle dysfunction in the context of T2D has the potential to lead to novel treatments for a large number of patients. Through its ability to emulate skeletal muscle architecture while also incorporating aspects of disease, tissue-engineered skeletal muscle (TE-SkM) has the potential to provide a means for rapid high-throughput discovery of therapies to treat skeletal muscle dysfunction, to include that which occurs with T2D. Muscle precursor cells isolated from lean or obese male Zucker diabetic fatty rats were used to generate TE-SkM constructs. Some constructs were treated with adipogenic induction media to accentuate the presence of adipocytes that is a characteristic feature of T2D skeletal muscle. The maturity (compaction and creatine kinase activity), mechanical integrity (Young's modulus), organization (myotube orientation), and metabolic capacity (insulin-stimulated glucose uptake) were all reduced by diabetes. Treating constructs with adipogenic induction media increased the quantity of lipid within the diabetic TE-SkM constructs, and caused changes in construct compaction, cell orientation, and insulin-stimulated glucose uptake in both lean and diabetic samples. Collectively, the findings herein suggest that the recapitulation of structural and metabolic aspects of T2D can be accomplished by engineering skeletal muscle in vitro.
Collapse
Affiliation(s)
- Francisca M. Acosta
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, USA
- UTSA-UTHSCSA Joint Graduate Program in Biomedical Engineering, San Antonio, Texas, USA
| | - U-Ter Aonda Jia
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, USA
- UTSA-UTHSCSA Joint Graduate Program in Biomedical Engineering, San Antonio, Texas, USA
| | - Katerina Stojkova
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Kennedy K. Howland
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Teja Guda
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Settimio Pacelli
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Eric M. Brey
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Christopher R. Rathbone
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
26
|
Khodabukus A. Tissue-Engineered Skeletal Muscle Models to Study Muscle Function, Plasticity, and Disease. Front Physiol 2021; 12:619710. [PMID: 33716768 PMCID: PMC7952620 DOI: 10.3389/fphys.2021.619710] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/25/2021] [Indexed: 12/20/2022] Open
Abstract
Skeletal muscle possesses remarkable plasticity that permits functional adaptations to a wide range of signals such as motor input, exercise, and disease. Small animal models have been pivotal in elucidating the molecular mechanisms regulating skeletal muscle adaptation and plasticity. However, these small animal models fail to accurately model human muscle disease resulting in poor clinical success of therapies. Here, we review the potential of in vitro three-dimensional tissue-engineered skeletal muscle models to study muscle function, plasticity, and disease. First, we discuss the generation and function of in vitro skeletal muscle models. We then discuss the genetic, neural, and hormonal factors regulating skeletal muscle fiber-type in vivo and the ability of current in vitro models to study muscle fiber-type regulation. We also evaluate the potential of these systems to be utilized in a patient-specific manner to accurately model and gain novel insights into diseases such as Duchenne muscular dystrophy (DMD) and volumetric muscle loss. We conclude with a discussion on future developments required for tissue-engineered skeletal muscle models to become more mature, biomimetic, and widely utilized for studying muscle physiology, disease, and clinical use.
Collapse
Affiliation(s)
- Alastair Khodabukus
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
27
|
Abstract
Tissue engineering refers to the attempt to create functional human tissue from cells in a laboratory. This is a field that uses living cells, biocompatible materials, suitable biochemical and physical factors, and their combinations to create tissue-like structures. To date, no tissue engineered skeletal muscle implants have been developed for clinical use, but they may represent a valid alternative for the treatment of volumetric muscle loss in the near future. Herein, we reviewed the literature and showed different techniques to produce synthetic tissues with the same architectural, structural and functional properties as native tissues.
Collapse
|
28
|
Barrett P, Quick TJ, Mudera V, Player DJ. Generating intrafusal skeletal muscle fibres in vitro: Current state of the art and future challenges. J Tissue Eng 2020; 11:2041731420985205. [PMID: 34956586 PMCID: PMC8693220 DOI: 10.1177/2041731420985205] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/12/2020] [Indexed: 01/18/2023] Open
Abstract
Intrafusal fibres are a specialised cell population in skeletal muscle, found within the muscle spindle. These fibres have a mechano-sensory capacity, forming part of the monosynaptic stretch-reflex arc, a key component responsible for proprioceptive function. Impairment of proprioception and associated dysfunction of the muscle spindle is linked with many neuromuscular diseases. Research to-date has largely been undertaken in vivo or using ex vivo preparations. These studies have provided a foundation for our understanding of muscle spindle physiology, however, the cellular and molecular mechanisms which underpin physiological changes are yet to be fully elucidated. Therefrom, the use of in vitro models has been proposed, whereby intrafusal fibres can be generated de novo. Although there has been progress, it is predominantly a developing and evolving area of research. This narrative review presents the current state of art in this area and proposes the direction of future work, with the aim of providing novel pre-clinical and clinical applications.
Collapse
Affiliation(s)
- Philip Barrett
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, London, UK
| | - Tom J Quick
- Peripheral Nerve Injury Research Unit, Royal National Orthopaedic Hospital, Stanmore, UK
- UCL Centre for Nerve Engineering, University College London, London, UK
| | - Vivek Mudera
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, London, UK
| | - Darren J Player
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, London, UK
| |
Collapse
|
29
|
Nagashima T, Hadiwidjaja S, Ohsumi S, Murata A, Hisada T, Kato R, Okada Y, Honda H, Shimizu K. In Vitro Model of Human Skeletal Muscle Tissues with Contractility Fabricated by Immortalized Human Myogenic Cells. ACTA ACUST UNITED AC 2020; 4:e2000121. [DOI: 10.1002/adbi.202000121] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 10/04/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Takunori Nagashima
- Department of Biomolecular Engineering Graduate School of Engineering Nagoya University Nagoya 464‐8603 Japan
| | - Stacy Hadiwidjaja
- Department of Biomolecular Engineering Graduate School of Engineering Nagoya University Nagoya 464‐8603 Japan
| | - Saki Ohsumi
- Department of Biomolecular Engineering Graduate School of Engineering Nagoya University Nagoya 464‐8603 Japan
| | - Akari Murata
- Department of Biomolecular Engineering Graduate School of Engineering Nagoya University Nagoya 464‐8603 Japan
| | - Takumi Hisada
- Department of Basic Medicinal Sciences Graduate School of Pharmaceutical Sciences Nagoya University Nagoya 464‐8601 Japan
| | - Ryuji Kato
- Department of Basic Medicinal Sciences Graduate School of Pharmaceutical Sciences Nagoya University Nagoya 464‐8601 Japan
| | - Yohei Okada
- Department of Neurology Aichi Medical University School of Medicine Aichi 480‐1195 Japan
| | - Hiroyuki Honda
- Department of Biomolecular Engineering Graduate School of Engineering Nagoya University Nagoya 464‐8603 Japan
| | - Kazunori Shimizu
- Department of Biomolecular Engineering Graduate School of Engineering Nagoya University Nagoya 464‐8603 Japan
| |
Collapse
|
30
|
Fleming JW, Capel AJ, Rimington RP, Wheeler P, Leonard AN, Bishop NC, Davies OG, Lewis MP. Bioengineered human skeletal muscle capable of functional regeneration. BMC Biol 2020; 18:145. [PMID: 33081771 PMCID: PMC7576716 DOI: 10.1186/s12915-020-00884-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/30/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Skeletal muscle (SkM) regenerates following injury, replacing damaged tissue with high fidelity. However, in serious injuries, non-regenerative defects leave patients with loss of function, increased re-injury risk and often chronic pain. Progress in treating these non-regenerative defects has been slow, with advances only occurring where a comprehensive understanding of regeneration has been gained. Tissue engineering has allowed the development of bioengineered models of SkM which regenerate following injury to support research in regenerative physiology. To date, however, no studies have utilised human myogenic precursor cells (hMPCs) to closely mimic functional human regenerative physiology. RESULTS Here we address some of the difficulties associated with cell number and hMPC mitogenicity using magnetic association cell sorting (MACS), for the marker CD56, and media supplementation with fibroblast growth factor 2 (FGF-2) and B-27 supplement. Cell sorting allowed extended expansion of myogenic cells and supplementation was shown to improve myogenesis within engineered tissues and force generation at maturity. In addition, these engineered human SkM regenerated following barium chloride (BaCl2) injury. Following injury, reductions in function (87.5%) and myotube number (33.3%) were observed, followed by a proliferative phase with increased MyoD+ cells and a subsequent recovery of function and myotube number. An expansion of the Pax7+ cell population was observed across recovery suggesting an ability to generate Pax7+ cells within the tissue, similar to the self-renewal of satellite cells seen in vivo. CONCLUSIONS This work outlines an engineered human SkM capable of functional regeneration following injury, built upon an open source system adding to the pre-clinical testing toolbox to improve the understanding of basic regenerative physiology.
Collapse
Affiliation(s)
- J W Fleming
- School of Sports, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - A J Capel
- School of Sports, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - R P Rimington
- School of Sports, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - P Wheeler
- School of Sports, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - A N Leonard
- School of Sports, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - N C Bishop
- School of Sports, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - O G Davies
- School of Sports, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - M P Lewis
- School of Sports, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK.
| |
Collapse
|
31
|
Urciuolo A, Serena E, Ghua R, Zatti S, Giomo M, Mattei N, Vetralla M, Selmin G, Luni C, Vitulo N, Valle G, Vitiello L, Elvassore N. Engineering a 3D in vitro model of human skeletal muscle at the single fiber scale. PLoS One 2020; 15:e0232081. [PMID: 32374763 PMCID: PMC7202609 DOI: 10.1371/journal.pone.0232081] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 04/07/2020] [Indexed: 01/07/2023] Open
Abstract
The reproduction of reliable in vitro models of human skeletal muscle is made harder by the intrinsic 3D structural complexity of this tissue. Here we coupled engineered hydrogel with 3D structural cues and specific mechanical properties to derive human 3D muscle constructs (“myobundles”) at the scale of single fibers, by using primary myoblasts or myoblasts derived from embryonic stem cells. To this aim, cell culture was performed in confined, laminin-coated micrometric channels obtained inside a 3D hydrogel characterized by the optimal stiffness for skeletal muscle myogenesis. Primary myoblasts cultured in our 3D culture system were able to undergo myotube differentiation and maturation, as demonstrated by the proper expression and localization of key components of the sarcomere and sarcolemma. Such approach allowed the generation of human myobundles of ~10 mm in length and ~120 μm in diameter, showing spontaneous contraction 7 days after cell seeding. Transcriptome analyses showed higher similarity between 3D myobundles and skeletal signature, compared to that found between 2D myotubes and skeletal muscle, mainly resulting from expression in 3D myobundles of categories of genes involved in skeletal muscle maturation, including extracellular matrix organization. Moreover, imaging analyses confirmed that structured 3D culture system was conducive to differentiation/maturation also when using myoblasts derived from embryonic stem cells. In conclusion, our structured 3D model is a promising tool for modelling human skeletal muscle in healthy and diseases conditions.
Collapse
Affiliation(s)
- Anna Urciuolo
- Industrial Engineering Department, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy.,Women's and Children's Health Department, University of Padova, Padova, Italy
| | - Elena Serena
- Industrial Engineering Department, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Rusha Ghua
- Department of Biology, University of Padova, Padova, Italy
| | - Susi Zatti
- Industrial Engineering Department, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Monica Giomo
- Industrial Engineering Department, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Nicolò Mattei
- Industrial Engineering Department, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Massimo Vetralla
- Industrial Engineering Department, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy
| | - Giulia Selmin
- Industrial Engineering Department, University of Padova, Padova, Italy
| | - Camilla Luni
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Nicola Vitulo
- Department of Biotechnologies, University of Verona, Verona, Italy
| | - Giorgio Valle
- Department of Biology, University of Padova, Padova, Italy
| | - Libero Vitiello
- Department of Biology, University of Padova, Padova, Italy.,Interuniversity Institute of Myology (IIM), Assisi, Italy
| | - Nicola Elvassore
- Industrial Engineering Department, University of Padova, Padova, Italy.,Venetian Institute of Molecular Medicine, Padova, Italy.,Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China.,University College London ICH, London, England, United Kingdom
| |
Collapse
|
32
|
Shimizu K, Ohsumi S, Kishida T, Mazda O, Honda H. Fabrication of contractile skeletal muscle tissues using directly converted myoblasts from human fibroblasts. J Biosci Bioeng 2020; 129:632-637. [DOI: 10.1016/j.jbiosc.2019.11.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/30/2019] [Accepted: 11/26/2019] [Indexed: 12/28/2022]
|
33
|
Kim W, Lee H, Lee J, Atala A, Yoo JJ, Lee SJ, Kim GH. Efficient myotube formation in 3D bioprinted tissue construct by biochemical and topographical cues. Biomaterials 2020; 230:119632. [PMID: 31761486 PMCID: PMC7141931 DOI: 10.1016/j.biomaterials.2019.119632] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/31/2019] [Accepted: 11/13/2019] [Indexed: 01/08/2023]
Abstract
Biochemical and biophysical cues directly affect cell morphology, adhesion, proliferation, and phenotype, as well as differentiation; thus, they have been commonly utilized for designing and developing biomaterial systems for tissue engineering applications. To bioengineer skeletal muscle tissues, the efficient and stable formation of aligned fibrous multinucleated myotubes is essential. To achieve this goal, we employed a decellularized extracellular matrix (dECM) as a biochemical component and a modified three-dimensional (3D) cell-printing process to produce an in situ uniaxially aligned/micro-topographical structure. The dECM was derived from the decellularization of porcine skeletal muscles and chemically modified by methacrylate process to enhance mechanical stability. By using this ECM-based material and the 3D printing capability, we were able to produce a cell-laden dECM-based structure with unique topographical cues. The myoblasts (C2C12 cell line) laden in the printed structure were aligned and differentiated with a high degree of myotube formation, owing to the synergistic effect of the skeletal muscle-specific biochemical and topographical cues. In particular, the increase of the gene-expression levels of the dECM structure with topographical cues was approximately 1.5-1.8-fold compared with those of a gelatin methacrylate (GelMA)-based structure with the same topographical cues and a dECM-based structure without topographical cues. According to these in vitro cellular responses, the 3D printed dECM-based structures with topographical cues have the potential for bioengineering functional skeletal muscle tissues, and this strategy can be extended for many musculoskeletal tissues, such as tendons and ligaments and utilized for developing in vitro tissue-on-a-chip models in drug screening and development.
Collapse
Affiliation(s)
- WonJin Kim
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA; Department of Biomechatronic Engineering, College of Biotechnology and Bioengineering, Sungkyunkwan University (SKKU), Suwon, 16419, South Korea
| | - Hyeongjin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - JiUn Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA; Department of Biomechatronic Engineering, College of Biotechnology and Bioengineering, Sungkyunkwan University (SKKU), Suwon, 16419, South Korea
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA.
| | - Geun Hyung Kim
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA; Department of Biomechatronic Engineering, College of Biotechnology and Bioengineering, Sungkyunkwan University (SKKU), Suwon, 16419, South Korea.
| |
Collapse
|
34
|
Roberts IV, Bukhary D, Valdivieso CYL, Tirelli N. Fibrin Matrices as (Injectable) Biomaterials: Formation, Clinical Use, and Molecular Engineering. Macromol Biosci 2019; 20:e1900283. [PMID: 31769933 DOI: 10.1002/mabi.201900283] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/14/2019] [Indexed: 12/19/2022]
Abstract
This review focuses on fibrin, starting from biological mechanisms (its production from fibrinogen and its enzymatic degradation), through its use as a medical device and as a biomaterial, and finally discussing the techniques used to add biological functions and/or improve its mechanical performance through its molecular engineering. Fibrin is a material of biological (human, and even patient's own) origin, injectable, adhesive, and remodellable by cells; further, it is nature's most common choice for an in situ forming, provisional matrix. Its widespread use in the clinic and in research is therefore completely unsurprising. There are, however, areas where its biomedical performance can be improved, namely achieving a better control over mechanical properties (and possibly higher modulus), slowing down degradation or incorporating cell-instructive functions (e.g., controlled delivery of growth factors). The authors here specifically review the efforts made in the last 20 years to achieve these aims via biomimetic reactions or self-assembly, as much via formation of hybrid materials.
Collapse
Affiliation(s)
- Iwan Vaughan Roberts
- Division of Pharmacy and Optometry, School of Health Science, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Deena Bukhary
- Division of Pharmacy and Optometry, School of Health Science, University of Manchester, Oxford Road, Manchester, M13 9PT, UK.,Department of Pharmaceutical Science, Faculty of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | | | - Nicola Tirelli
- Division of Pharmacy and Optometry, School of Health Science, University of Manchester, Oxford Road, Manchester, M13 9PT, UK.,Laboratory of Polymers and Biomaterials, Fondazione Istituto Italiano di Tecnologia, via Morego 30, 16163, Genova, Italy
| |
Collapse
|
35
|
Wragg NM, Mosqueira D, Blokpeol-Ferreras L, Capel A, Player DJ, Martin NRW, Liu Y, Lewis MP. Development of a 3D Tissue-Engineered Skeletal Muscle and Bone Co-culture System. Biotechnol J 2019; 15:e1900106. [PMID: 31468704 DOI: 10.1002/biot.201900106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 07/05/2019] [Indexed: 12/26/2022]
Abstract
In vitro 3D tissue-engineered (TE) structures have been shown to better represent in vivo tissue morphology and biochemical pathways than monolayer culture, and are less ethically questionable than animal models. However, to create systems with even greater relevance, multiple integrated tissue systems should be recreated in vitro. In the present study, the effects and conditions most suitable for the co-culture of TE skeletal muscle and bone are investigated. High-glucose Dulbecco's modified Eagle medium (HG-DMEM) supplemented with 20% fetal bovine serum followed by HG-DMEM with 2% horse serum is found to enable proliferation of both C2C12 muscle precursor cells and TE85 human osteosarcoma cells, fusion of C2C12s into myotubes, as well as an upregulation of RUNX2/CBFa1 in TE85s. Myotube formation is also evident within indirect contact monolayer cultures. Finally, in 3D co-cultures, TE85 collagen/hydroxyapatite constructs have significantly greater expression of RUNX2/CBFa1 and osteocalcin/BGLAP in the presence of collagen-based C2C12 skeletal muscle constructs; however, fusion within these constructs appears reduced. This work demonstrates the first report of the simultaneous co-culture and differentiation of 3D TE skeletal muscle and bone, and represents a significant step toward a full in vitro 3D musculoskeletal junction model.
Collapse
Affiliation(s)
- Nicholas M Wragg
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.,Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, UK
| | - Diogo Mosqueira
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Lia Blokpeol-Ferreras
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Andrew Capel
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Darren J Player
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.,Institute of Orthopaedics and Musculoskeletal Sciences, RNOH University College London, Stanmore, UK
| | - Neil R W Martin
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Yang Liu
- Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, UK
| | - Mark P Lewis
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| |
Collapse
|
36
|
Maffioletti SM, Sarcar S, Henderson ABH, Mannhardt I, Pinton L, Moyle LA, Steele-Stallard H, Cappellari O, Wells KE, Ferrari G, Mitchell JS, Tyzack GE, Kotiadis VN, Khedr M, Ragazzi M, Wang W, Duchen MR, Patani R, Zammit PS, Wells DJ, Eschenhagen T, Tedesco FS. Three-Dimensional Human iPSC-Derived Artificial Skeletal Muscles Model Muscular Dystrophies and Enable Multilineage Tissue Engineering. Cell Rep 2019; 23:899-908. [PMID: 29669293 PMCID: PMC5917451 DOI: 10.1016/j.celrep.2018.03.091] [Citation(s) in RCA: 198] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 02/21/2018] [Accepted: 03/20/2018] [Indexed: 01/07/2023] Open
Abstract
Generating human skeletal muscle models is instrumental for investigating muscle pathology and therapy. Here, we report the generation of three-dimensional (3D) artificial skeletal muscle tissue from human pluripotent stem cells, including induced pluripotent stem cells (iPSCs) from patients with Duchenne, limb-girdle, and congenital muscular dystrophies. 3D skeletal myogenic differentiation of pluripotent cells was induced within hydrogels under tension to provide myofiber alignment. Artificial muscles recapitulated characteristics of human skeletal muscle tissue and could be implanted into immunodeficient mice. Pathological cellular hallmarks of incurable forms of severe muscular dystrophy could be modeled with high fidelity using this 3D platform. Finally, we show generation of fully human iPSC-derived, complex, multilineage muscle models containing key isogenic cellular constituents of skeletal muscle, including vascular endothelial cells, pericytes, and motor neurons. These results lay the foundation for a human skeletal muscle organoid-like platform for disease modeling, regenerative medicine, and therapy development. Human iPSC-derived 3D artificial muscles show features of normal skeletal muscle Multiple muscular dystrophy iPSC lines can be differentiated in 3D artificial muscles Artificial muscle constructs model severe, incurable forms of muscular dystrophy Isogenic vascular-like networks and motor neurons develop within artificial muscles
Collapse
Affiliation(s)
| | - Shilpita Sarcar
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK
| | - Alexander B H Henderson
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK
| | - Ingra Mannhardt
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf (UKE), 20246 Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany
| | - Luca Pinton
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK; Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Louise Anne Moyle
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK
| | - Heather Steele-Stallard
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK; Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Ornella Cappellari
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK
| | - Kim E Wells
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK
| | - Giulia Ferrari
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK
| | - Jamie S Mitchell
- Institute of Neurology, University College London, London WC1N 3BG, UK; The Francis Crick Institute, London NW1 1AT, UK
| | - Giulia E Tyzack
- Institute of Neurology, University College London, London WC1N 3BG, UK; The Francis Crick Institute, London NW1 1AT, UK
| | - Vassilios N Kotiadis
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK
| | - Moustafa Khedr
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK
| | - Martina Ragazzi
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK
| | - Weixin Wang
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK
| | - Michael R Duchen
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK
| | - Rickie Patani
- Institute of Neurology, University College London, London WC1N 3BG, UK; The Francis Crick Institute, London NW1 1AT, UK
| | - Peter S Zammit
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Dominic J Wells
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf (UKE), 20246 Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Germany
| | | |
Collapse
|
37
|
Fleming JW, Capel AJ, Rimington RP, Player DJ, Stolzing A, Lewis MP. Functional regeneration of tissue engineered skeletal muscle in vitro is dependent on the inclusion of basement membrane proteins. Cytoskeleton (Hoboken) 2019; 76:371-382. [PMID: 31376315 PMCID: PMC6790946 DOI: 10.1002/cm.21553] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 06/28/2019] [Accepted: 07/31/2019] [Indexed: 12/23/2022]
Abstract
Skeletal muscle has a high regenerative capacity, injuries trigger a regenerative program which restores tissue function to a level indistinguishable to the pre-injury state. However, in some cases where significant trauma occurs, such as injuries seen in military populations, the regenerative process is overwhelmed and cannot restore full function. Limited clinical interventions exist which can be used to promote regeneration and prevent the formation of non-regenerative defects following severe skeletal muscle trauma. Robust and reproducible techniques for modelling complex tissue responses are essential to promote the discovery of effective clinical interventions. Tissue engineering has been highlighted as an alternative method, allowing the generation of three-dimensional in vivo like tissues without laboratory animals. Reducing the requirement for animal models promotes rapid screening of potential clinical interventions, as these models are more easily manipulated, genetically and pharmacologically, and reduce the associated cost and complexity, whilst increasing access to models for laboratories without animal facilities. In this study, an in vitro chemical injury using barium chloride is validated using the C2C12 myoblast cell line, and is shown to selectively remove multinucleated myotubes, whilst retaining a regenerative mononuclear cell population. Monolayer cultures showed limited regenerative capacity, with basement membrane supplementation or extended regenerative time incapable of improving the regenerative response. Conversely tissue engineered skeletal muscles, supplemented with basement membrane proteins, showed full functional regeneration, and a broader in vivo like inflammatory response. This work outlines a freely available and open access methodology to produce a cell line-based tissue engineered model of skeletal muscle regeneration.
Collapse
Affiliation(s)
- Jacob W Fleming
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Andrew J Capel
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Rowan P Rimington
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Darren J Player
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Alexandra Stolzing
- Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, United Kingdom
| | - Mark P Lewis
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| |
Collapse
|
38
|
Wang J, Khodabukus A, Rao L, Vandusen K, Abutaleb N, Bursac N. Engineered skeletal muscles for disease modeling and drug discovery. Biomaterials 2019; 221:119416. [PMID: 31419653 DOI: 10.1016/j.biomaterials.2019.119416] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 08/01/2019] [Accepted: 08/05/2019] [Indexed: 01/04/2023]
Abstract
Skeletal muscle is the largest organ of human body with several important roles in everyday movement and metabolic homeostasis. The limited ability of small animal models of muscle disease to accurately predict drug efficacy and toxicity in humans has prompted the development in vitro models of human skeletal muscle that fatefully recapitulate cell and tissue level functions and drug responses. We first review methods for development of three-dimensional engineered muscle tissues and organ-on-a-chip microphysiological systems and discuss their potential utility in drug discovery research and development of new regenerative therapies. Furthermore, we describe strategies to increase the functional maturation of engineered muscle, and motivate the importance of incorporating multiple tissue types on the same chip to model organ cross-talk and generate more predictive drug development platforms. Finally, we review the ability of available in vitro systems to model diseases such as type II diabetes, Duchenne muscular dystrophy, Pompe disease, and dysferlinopathy.
Collapse
Affiliation(s)
- Jason Wang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Lingjun Rao
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Keith Vandusen
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nadia Abutaleb
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
39
|
Natarajan A, Sethumadhavan A, Krishnan UM. Toward Building the Neuromuscular Junction: In Vitro Models To Study Synaptogenesis and Neurodegeneration. ACS OMEGA 2019; 4:12969-12977. [PMID: 31460423 PMCID: PMC6682064 DOI: 10.1021/acsomega.9b00973] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/04/2019] [Indexed: 06/10/2023]
Abstract
The neuromuscular junction (NMJ) is a unique, specialized chemical synapse that plays a crucial role in transmitting and amplifying information from spinal motor neurons to skeletal muscles. NMJ complexity ensures closely intertwined interactions between numerous synaptic vesicles, signaling molecules, ion channels, motor neurons, glia, and muscle fibers, making it difficult to dissect the underlying mechanisms and factors affecting neurodegeneration and muscle loss. Muscle fiber or motor neuron cell death followed by rapid axonal degeneration due to injury or disease has a debilitating effect on movement and behavior, which adversely affects the quality of life. It thus becomes imperative to study the synapse and intercellular signaling processes that regulate plasticity at the NMJ and elucidate mechanisms and pathways at the cellular level. Studies using in vitro 2D cell cultures have allowed us to gain a fundamental understanding of how the NMJ functions. However, they do not provide information on the intricate signaling networks that exist between NMJs and the biological environment. The advent of 3D cell cultures and microfluidic lab-on-a-chip technologies has opened whole new avenues to explore the NMJ. In this perspective, we look at the challenges involved in building a functional NMJ and the progress made in generating models for studying the NMJ, highlighting the current and future applications of these models.
Collapse
Affiliation(s)
- Anupama Natarajan
- Centre
for Nanotechnology & Advanced Biomaterials (CeNTAB), School of Chemical
& Biotechnology, and School of Arts, Science & Humanities, SASTRA Deemed University, Thanjavur 613 401, India
| | - Anjali Sethumadhavan
- Centre
for Nanotechnology & Advanced Biomaterials (CeNTAB), School of Chemical
& Biotechnology, and School of Arts, Science & Humanities, SASTRA Deemed University, Thanjavur 613 401, India
| | - Uma Maheswari Krishnan
- Centre
for Nanotechnology & Advanced Biomaterials (CeNTAB), School of Chemical
& Biotechnology, and School of Arts, Science & Humanities, SASTRA Deemed University, Thanjavur 613 401, India
| |
Collapse
|
40
|
Pal A, Tripathi K, Pathak C, Vernon BL. Plasma-based fast-gelling biohybrid gels for biomedical applications. Sci Rep 2019; 9:10881. [PMID: 31350449 PMCID: PMC6659638 DOI: 10.1038/s41598-019-47366-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 07/16/2019] [Indexed: 12/14/2022] Open
Abstract
Blood based biomaterials are widely researched and used in different biomedical applications including cell therapy, drug delivery, sealants etc. due to their biocompatibility and biodegradability. Blood derived gels are successfully used in clinical studies due to the presence of fibrinogen and several platelet growth factors. In spite of their wide applications, it is challenging to use blood-based biomaterials due to their low mechanical stability, poor adhesive property and contamination risk. In this study, we used porcine plasma to form gel in presence of biodegradable synthetic crosslinkers. Mechanical strength of this plasma gel could be tailored by altering the amount of crosslinkers for any desired biomedical applications. These plasma gels, formed by the synthetic crosslinkers, were utilized as a drug delivery platform for wound healing due to their low cytotoxicity. A model drug release study with these plasma gels indicated slow and sustained release of the drugs.
Collapse
Affiliation(s)
- Amrita Pal
- Arizona State University, Tempe, AZ, 85287, USA
| | | | | | | |
Collapse
|
41
|
Natesan S, Stone R, Coronado RE, Wrice NL, Kowalczewski AC, Zamora DO, Christy RJ. PEGylated Platelet-Free Blood Plasma-Based Hydrogels for Full-Thickness Wound Regeneration. Adv Wound Care (New Rochelle) 2019; 8:323-340. [PMID: 31737420 DOI: 10.1089/wound.2018.0844] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/23/2018] [Indexed: 02/03/2023] Open
Abstract
Objective: To develop a cost-effective and clinically usable therapy to treat full-thickness skin injuries. We accomplished this by preparing a viscoelastic hydrogel using polyethylene glycol (PEG)-modified platelet-free plasma (PEGylated PFP) combined with human adipose-derived stem cells (ASCs). Approach: PEGylated PFP hydrogels were prepared by polymerizing the liquid mixture of PEG and PFP±ASCs and gelled either by adding calcium chloride (CaCl2) or thrombin. Rheological and in vitro studies were performed to assess viscoelasticity and the ability of hydrogels to direct ASCs toward a vasculogenic phenotype, respectively. Finally, a pilot study evaluated the efficacy of hydrogels±ASCs using an athymic rat full-thickness skin wound model. Results: Hydrogels prepared within the range of 11 to 27 mM for CaCl2 or 5 to 12.5 U/mL for thrombin exhibited a storage modulus of ∼62 to 87 Pa and ∼47 to 92 Pa, respectively. The PEGylated PFP hydrogels directed ASCs to form network-like structures resembling vasculature, with a fourfold increase in perivascular specific genes that were confirmed by immunofluorescent staining. Hydrogels combined with ASCs exhibited an increase in blood vessel density when applied to excisional rat wounds compared with those treated with hydrogels (110.3 vs. 95.6 BV/mm2; p < 0.05). Furthermore, ASCs were identified in the perivascular region associated with newly forming blood vessels. Innovation: This study demonstrates that PFP modified with PEG along with ASCs can be used to prepare cost-effective stable hydrogels, at the bed-side, to treat extensive skin wounds. Conclusion: These results indicate that PEGylated plasma-based hydrogels combined with ASCs may be a potential regenerative therapy for full-thickness skin wounds.
Collapse
Affiliation(s)
- Shanmugasundaram Natesan
- Combat Trauma and Burn Injury Research, U.S. Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - Randolph Stone
- Combat Trauma and Burn Injury Research, U.S. Army Institute of Surgical Research, Fort Sam Houston, Texas
| | | | - Nicole L. Wrice
- Ocular Trauma & Vision Restoration, U.S. Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - Andrew C. Kowalczewski
- Combat Trauma and Burn Injury Research, U.S. Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - David O. Zamora
- Ocular Trauma & Vision Restoration, U.S. Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - Robert J. Christy
- Combat Trauma and Burn Injury Research, U.S. Army Institute of Surgical Research, Fort Sam Houston, Texas
| |
Collapse
|
42
|
Wragg NM, Player DJ, Martin NRW, Liu Y, Lewis MP. Development of tissue‐engineered skeletal muscle manufacturing variables. Biotechnol Bioeng 2019; 116:2364-2376. [DOI: 10.1002/bit.27074] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 05/03/2019] [Accepted: 05/21/2019] [Indexed: 11/09/2022]
Affiliation(s)
- Nicholas M. Wragg
- School of Sport, Exercise, and Health SciencesLoughborough UniversityLoughborough UK
- Wolfson School of Mechanical, Electrical, and Manufacturing EngineeringLoughborough UniversityLoughborough UK
- Centre for Biological EngineeringLoughborough UniversityLoughborough UK
| | - Darren J. Player
- School of Sport, Exercise, and Health SciencesLoughborough UniversityLoughborough UK
- Centre for Sport, Exercise, and OsteoarthritisArthritis Research UK UK
- Division of Surgery and Interventional ScienceUniversity College LondonLondon UK
| | - Neil R. W. Martin
- School of Sport, Exercise, and Health SciencesLoughborough UniversityLoughborough UK
| | - Yang Liu
- Wolfson School of Mechanical, Electrical, and Manufacturing EngineeringLoughborough UniversityLoughborough UK
- Centre for Biological EngineeringLoughborough UniversityLoughborough UK
| | - Mark P. Lewis
- School of Sport, Exercise, and Health SciencesLoughborough UniversityLoughborough UK
- Centre for Sport, Exercise, and OsteoarthritisArthritis Research UK UK
- National Centre for Sport and Exercise MedicineLoughborough UK
| |
Collapse
|
43
|
Aguilar-Agon KW, Capel AJ, Martin NRW, Player DJ, Lewis MP. Mechanical loading stimulates hypertrophy in tissue-engineered skeletal muscle: Molecular and phenotypic responses. J Cell Physiol 2019; 234:23547-23558. [PMID: 31180593 PMCID: PMC6771594 DOI: 10.1002/jcp.28923] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/17/2019] [Accepted: 05/20/2019] [Indexed: 12/17/2022]
Abstract
Mechanical loading of skeletal muscle results in molecular and phenotypic adaptations typified by enhanced muscle size. Studies on humans are limited by the need for repeated sampling, and studies on animals have methodological and ethical limitations. In this investigation, three‐dimensional skeletal muscle was tissue‐engineered utilizing the murine cell line C2C12, which bears resemblance to native tissue and benefits from the advantages of conventional in vitro experiments. The work aimed to determine if mechanical loading induced an anabolic hypertrophic response, akin to that described in vivo after mechanical loading in the form of resistance exercise. Specifically, we temporally investigated candidate gene expression and Akt‐mechanistic target of rapamycin 1 signalling along with myotube growth and tissue function. Mechanical loading (construct length increase of 15%) significantly increased insulin‐like growth factor‐1 and MMP‐2 messenger RNA expression 21 hr after overload, and the levels of the atrophic gene MAFbx were significantly downregulated 45 hr after mechanical overload. In addition, p70S6 kinase and 4EBP‐1 phosphorylation were upregulated immediately after mechanical overload. Maximal contractile force was augmented 45 hr after load with a 265% increase in force, alongside significant hypertrophy of the myotubes within the engineered muscle. Overall, mechanical loading of tissue‐engineered skeletal muscle induced hypertrophy and improved force production.
Collapse
Affiliation(s)
- Kathryn W Aguilar-Agon
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Andrew J Capel
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Neil R W Martin
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Darren J Player
- Division of Surgery, University College London, London, United Kingdom
| | - Mark P Lewis
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| |
Collapse
|
44
|
Miri AK, Mirzaee I, Hassan S, Mesbah Oskui S, Nieto D, Khademhosseini A, Zhang YS. Effective bioprinting resolution in tissue model fabrication. LAB ON A CHIP 2019; 19:2019-2037. [PMID: 31080979 PMCID: PMC6554720 DOI: 10.1039/c8lc01037d] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Recent advancements in bioprinting techniques have enabled convenient fabrication of micro-tissues in organ-on-a-chip platforms. In a sense, the success of bioprinted micro-tissues depends on how close their architectures are to the anatomical features of their native counterparts. The bioprinting resolution largely relates to the technical specifications of the bioprinter platforms and the physicochemical properties of the bioinks. In this article, we compare inkjet, extrusion, and light-assisted bioprinting technologies for fabrication of micro-tissues towards construction of biomimetic organ-on-a-chip platforms. Our theoretical analyses reveal that for a given printhead diameter, surface contact angle dominates inkjet bioprinting resolution, while nozzle moving speed and the nonlinearity of viscosity for bioinks regulate extrusion bioprinting resolution. The resolution of light-assisted bioprinting is strongly affected by the photocrosslinking behavior and light characteristics. Our tutorial guideline for optimizing bioprinting resolution would potentially help model the complex microenvironment of biological tissues in organ-on-a-chip platforms.
Collapse
Affiliation(s)
- Amir K Miri
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA. and Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA and Department of Mechanical Engineering, Rowan University, Glassboro, NJ 08028, USA
| | - Iman Mirzaee
- Department of Mechanical Engineering, University of Massachusetts, Lowell, MA 01854, USA
| | - Shabir Hassan
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA. and Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Shirin Mesbah Oskui
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA. and Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA and Bioengineering Program, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Daniel Nieto
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA. and Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, CA 90095, USA. and Department of Radiology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, USA and Department of Bioengineering, Department of Chemical and Biomolecular Engineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, CA 90095, USA and California NanoSystems Institute (CNSI), University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA. and Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
45
|
Ostrovidov S, Salehi S, Costantini M, Suthiwanish K, Ebrahimi M, Sadeghian RB, Fujie T, Shi X, Cannata S, Gargioli C, Tamayol A, Dokmeci MR, Orive G, Swieszkowski W, Khademhosseini A. 3D Bioprinting in Skeletal Muscle Tissue Engineering. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1805530. [PMID: 31012262 PMCID: PMC6570559 DOI: 10.1002/smll.201805530] [Citation(s) in RCA: 157] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 01/31/2019] [Indexed: 05/13/2023]
Abstract
Skeletal muscle tissue engineering (SMTE) aims at repairing defective skeletal muscles. Until now, numerous developments are made in SMTE; however, it is still challenging to recapitulate the complexity of muscles with current methods of fabrication. Here, after a brief description of the anatomy of skeletal muscle and a short state-of-the-art on developments made in SMTE with "conventional methods," the use of 3D bioprinting as a new tool for SMTE is in focus. The current bioprinting methods are discussed, and an overview of the bioink formulations and properties used in 3D bioprinting is provided. Finally, different advances made in SMTE by 3D bioprinting are highlighted, and future needs and a short perspective are provided.
Collapse
Affiliation(s)
- Serge Ostrovidov
- Department of Radiological Sciences, Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, California 90095, United States
| | - Sahar Salehi
- Department of Biomaterials, Faculty of Engineering Science, University of Bayreuth, Bayreuth 95440, Germany
| | - Marco Costantini
- Institute of Physical Chemistry – Polish Academy of Sciences, 01-224 Warsaw, Poland
| | - Kasinan Suthiwanish
- Department of Radiological Sciences, Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, California 90095, United States
| | - Majid Ebrahimi
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto ON M5S3G9, Canada
| | - Ramin Banan Sadeghian
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto 615-8540, Japan
| | - Toshinori Fujie
- School of Life Science and Technology, Tokyo Institute of Technology, B-50, 4259 Nagatsuta -cho, Midori-ku, Yokohama 226-8501, Japan
- PRESTO, Japan Science and Technology Agency, 4-1-8, Honcho, Kawaguchi-shi, Saitama 332-0012, Japan
| | - Xuetao Shi
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China, University of Technology, Guangzhou 510006, PR China
| | - Stefano Cannata
- Department of Biology, Tor Vergata Rome University, Rome 00133, Italy
| | - Cesare Gargioli
- Department of Biology, Tor Vergata Rome University, Rome 00133, Italy
| | - Ali Tamayol
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, NE 68588, USA
| | - Mehmet Remzi Dokmeci
- Department of Radiological Sciences, Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, California 90095, United States
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN). Vitoria-Gasteiz, Spain
- University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain; BTI Biotechnology Institute, Vitoria, Spain
| | - Wojciech Swieszkowski
- Faculty of Materials Science and Engineering, Warsaw University of Technology, 02-106 Warsaw, Poland
| | - Ali Khademhosseini
- Department of Radiological Sciences, Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, California 90095, United States
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Hwayang-dong, Kwangjin-gu, Seoul 05029, Republic of Korea
- Department of Chemical and Biomolecular Engineering, California NanoSystems Institute (CNSI), Department of Bioengineering, and Jonsson Comprehensive Cancer Centre University of California, Los Angeles, California 90095, United States
| |
Collapse
|
46
|
Dunn A, Talovic M, Patel K, Patel A, Marcinczyk M, Garg K. Biomaterial and stem cell-based strategies for skeletal muscle regeneration. J Orthop Res 2019; 37:1246-1262. [PMID: 30604468 DOI: 10.1002/jor.24212] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 12/13/2018] [Indexed: 02/04/2023]
Abstract
Adult skeletal muscle can regenerate effectively after mild physical or chemical insult. Muscle trauma or disease can overwhelm this innate capacity for regeneration and result in heightened inflammation and fibrotic tissue deposition resulting in loss of structure and function. Recent studies have focused on biomaterial and stem cell-based therapies to promote skeletal muscle regeneration following injury and disease. Many stem cell populations besides satellite cells are implicated in muscle regeneration. These stem cells include but are not limited to mesenchymal stem cells, adipose-derived stem cells, hematopoietic stem cells, pericytes, fibroadipogenic progenitors, side population cells, and CD133+ stem cells. However, several challenges associated with their isolation, availability, delivery, survival, engraftment, and differentiation have been reported in recent studies. While acellular scaffolds offer a relatively safe and potentially off-the-shelf solution to cell-based therapies, they are often unable to stimulate host cell migration and activity to a level that would result in clinically meaningful regeneration of traumatized muscle. Combining stem cells and biomaterials may offer a viable therapeutic strategy that may overcome the limitations associated with these therapies when they are used in isolation. In this article, we review the stem cell populations that can stimulate muscle regeneration in vitro and in vivo. We also discuss the regenerative potential of combination therapies that utilize both stem cell and biomaterials for the treatment of skeletal muscle injury and disease. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1246-1262, 2019.
Collapse
Affiliation(s)
- Andrew Dunn
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Muhamed Talovic
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Krishna Patel
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Anjali Patel
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Madison Marcinczyk
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Koyal Garg
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| |
Collapse
|
47
|
Nakayama KH, Quarta M, Paine P, Alcazar C, Karakikes I, Garcia V, Abilez OJ, Calvo NS, Simmons CS, Rando TA, Huang NF. Treatment of volumetric muscle loss in mice using nanofibrillar scaffolds enhances vascular organization and integration. Commun Biol 2019; 2:170. [PMID: 31098403 PMCID: PMC6505043 DOI: 10.1038/s42003-019-0416-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 04/05/2019] [Indexed: 12/15/2022] Open
Abstract
Traumatic skeletal muscle injuries cause irreversible tissue damage and impaired revascularization. Engineered muscle is promising for enhancing tissue revascularization and regeneration in injured muscle. Here we fabricated engineered skeletal muscle composed of myotubes interspersed with vascular endothelial cells using spatially patterned scaffolds that induce aligned cellular organization, and then assessed their therapeutic benefit for treatment of murine volumetric muscle loss. Murine skeletal myoblasts co-cultured with endothelial cells in aligned nanofibrillar scaffolds form endothelialized and aligned muscle with longer myotubes, more synchronized contractility, and more abundant secretion of angiogenic cytokines, compared to endothelialized engineered muscle formed from randomly-oriented scaffolds. Treatment of traumatically injured muscle with endothelialized and aligned skeletal muscle promotes the formation of highly organized myofibers and microvasculature, along with greater vascular perfusion, compared to treatment of muscle derived from randomly-oriented scaffolds. This work demonstrates the potential of endothelialized and aligned engineered skeletal muscle to promote vascular regeneration following transplantation.
Collapse
Affiliation(s)
- Karina H. Nakayama
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 USA
- The Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305 USA
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305 USA
| | - Marco Quarta
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94304 USA
| | - Patrick Paine
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94304 USA
| | - Cynthia Alcazar
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 USA
| | - Ioannis Karakikes
- The Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305 USA
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305 USA
| | - Victor Garcia
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 USA
| | - Oscar J. Abilez
- The Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305 USA
| | - Nicholas S. Calvo
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainsville, FL 32611 USA
| | - Chelsey S. Simmons
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainsville, FL 32611 USA
| | - Thomas A. Rando
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94304 USA
| | - Ngan F. Huang
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304 USA
- The Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305 USA
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305 USA
| |
Collapse
|
48
|
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: 106] [Impact Index Per Article: 21.2] [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
|
49
|
Nakayama KH, Shayan M, Huang NF. Engineering Biomimetic Materials for Skeletal Muscle Repair and Regeneration. Adv Healthc Mater 2019; 8:e1801168. [PMID: 30725530 PMCID: PMC6589032 DOI: 10.1002/adhm.201801168] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/21/2018] [Indexed: 11/12/2022]
Abstract
Although skeletal muscle is highly regenerative following injury or disease, endogenous self-regeneration is severely impaired in conditions of volume traumatic muscle loss. Consequently, tissue engineering approaches are a promising means to regenerate skeletal muscle. Biological scaffolds serve as not only structural support for the promotion of cellular ingrowth but also impart potent modulatory signaling cues that may be beneficial for tissue regeneration. In this work, the progress of tissue engineering approaches for skeletal muscle engineering and regeneration is overviewed, with a focus on the techniques to create biomimetic engineered tissue using extracellular cues. These factors include mechanical and electrical stimulation, geometric patterning, and delivery of growth factors or other bioactive molecules. The progress of evaluating the therapeutic efficacy of these approaches in preclinical models of muscle injury is further discussed.
Collapse
Affiliation(s)
- Karina H Nakayama
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
- Veterans Affairs Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA, 94304, USA
- The Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
| | - Mahdis Shayan
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
- Veterans Affairs Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA, 94304, USA
- The Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
| | - Ngan F Huang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
- Veterans Affairs Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA, 94304, USA
- The Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
50
|
Three-Dimensional Hydrogel-Based Culture to Study the Effects of Toxicants on Ovarian Follicles. Methods Mol Biol 2019; 1758:55-72. [PMID: 29679322 DOI: 10.1007/978-1-4939-7741-3_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Various toxicants, such as drugs and their metabolites, can cause potential ovarian toxicity. As the functional units of the ovary, ovarian follicles are susceptible to this type of damage at all developmental stages. Studying the effects of toxicants on ovarian follicles is an important task. Three-dimensional (3D) hydrogels, such as fibrin alginate interpenetrating networks (FA-IPNs), can support ovarian follicle culture in vitro for extended periods of time and serve as a suitable tool for studying ovotoxicity. Growing follicles encapsulated in the FA-IPN can proteolytically degrade the fibrin component in the FA-IPN. The degradation of fibrin mirrors the follicle growth and serves as a surrogate reporter for follicle health. The speed of fibrin degradation can be further controlled by aprotinin, a small molecule that inhibits plasmin-driven proteolytic degradation, which further expands the application of the described system. In this chapter, we describe methods to (1) isolate and encapsulate mouse ovarian follicles in FA-IPN, (2) follow follicle growth and development in vitro, and (3) evaluate the effects of toxicants on folliculogenesis using fibrin degradation.
Collapse
|