1
|
Alonso-Puyo J, Izagirre-Fernandez O, Crende O, Valdivia A, García-Gallastegui P, Sanz B. Experimental models as a tool for research on sarcopenia: A narrative review. Ageing Res Rev 2024; 101:102534. [PMID: 39369798 DOI: 10.1016/j.arr.2024.102534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
Sarcopenia is a musculoskeletal disorder related to muscle mass and function; as the worldwide population ages, its growing prevalence means a decline in quality of life and an increased burden for public health systems. As sarcopenia is a reversible condition, its early diagnosis is of utmost importance. Consensus definitions and diagnosis protocols for sarcopenia have been evolving for a long time, and the identification of molecular pathways subjacent to sarcopenia is a growing research area. The use of liquid biopsies to identify circulating molecules does not provide information about specific regulatory pathways or biomarkers in relevant tissue, and the use of skeletal muscle biopsies from older people has many limitations. Complementary tools are therefore necessary to advance the knowledge of relevant molecular aspects. The development of experimental models, such as animal, cellular, or bioengineered tissue, together with knock-in or knock-out strategies, could therefore be of great interest. This narrative review will explore experimental models of healthy muscle and aged muscle cells as a tool for research on sarcopenia. We will summarize the literature and present relevant experimental models in terms of their advantages and disadvantages. All of the presented approaches could potentially contribute to the accurate and early diagnosis, follow-up, and possible treatment of sarcopenia.
Collapse
Affiliation(s)
- Janire Alonso-Puyo
- Physiology Department, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., Leioa 48940, Spain
| | - Oihane Izagirre-Fernandez
- Physiology Department, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., Leioa 48940, Spain
| | - Olatz Crende
- Cell Biology and Histology Department, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., Leioa 48940, Spain
| | - Asier Valdivia
- Cell Biology and Histology Department, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., Leioa 48940, Spain
| | - Patricia García-Gallastegui
- Physiology Department, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., Leioa 48940, Spain.
| | - Begoña Sanz
- Physiology Department, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., Leioa 48940, Spain; Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia 48903, Spain.
| |
Collapse
|
2
|
Davenport ML, Fong A, Albury KN, Henley-Beasley CS, Barton ER, Maden M, Swanson MS. Spiny mice are primed but fail to regenerate volumetric skeletal muscle loss injuries. Skelet Muscle 2024; 14:26. [PMID: 39468576 PMCID: PMC11520498 DOI: 10.1186/s13395-024-00358-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/20/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND In recent years, the African spiny mouse Acomys cahirinus has been shown to regenerate a remarkable array of severe internal and external injuries in the absence of a fibrotic response, including the ability to regenerate full-thickness skin excisions, ear punches, severe kidney injuries, and complete transection of the spinal cord. While skeletal muscle is highly regenerative in adult mammals, Acomys displays superior muscle regeneration properties compared with standard laboratory mice following several injuries, including serial cardiotoxin injections of skeletal muscle and volumetric muscle loss (VML) of the panniculus carnosus muscle following full-thickness excision injuries. VML is an extreme muscle injury defined as the irrecoverable ablation of muscle mass, most commonly resulting from combat injuries or surgical debridement. Barriers to the treatment of VML injury include early and prolonged inflammatory responses that promote fibrotic repair and the loss of structural and mechanical cues that promote muscle regeneration. While the regeneration of the panniculus carnosus in Acomys is impressive, its direct relevance to the study of VML in patients is less clear as this muscle has largely been lost in humans, and, while striated, is not a true skeletal muscle. We therefore sought to test the ability of Acomys to regenerate a skeletal muscle more commonly used in VML injury models. METHODS We performed two different VML injuries of the Acomys tibialis anterior muscle and compared the regenerative response to a standard laboratory mouse strain, Mus C57BL6/J. RESULTS Neither Acomys nor Mus recovered lost muscle mass or myofiber number within three months following VML injury, and Acomys also failed to recover force production better than Mus. In contrast, Acomys continued to express eMHC within the injured area even three months following injury, whereas Mus ceased expressing eMHC less than one-month post-injury, suggesting that Acomys muscle was primed, but failed, to regenerate. CONCLUSIONS While the panniculus carnosus muscle in Acomys regenerates following VML injury in the context of full-thickness skin excision, this regenerative ability does not translate to regenerative repair of a skeletal muscle.
Collapse
Affiliation(s)
- Mackenzie L Davenport
- Department of Molecular Genetics and Microbiology, University of Florida, College of Medicine, Gainesville, FL, 32610, USA.
- Center for NeuroGenetics, University of Florida, Gainesville, FL, 32610, USA.
- UF Genetics Institute, University of Florida, Gainesville, FL, 32610, USA.
- Myology Institute, University of Florida, Gainesville, FL, USA.
| | - Amaya Fong
- Department of Molecular Genetics and Microbiology, University of Florida, College of Medicine, Gainesville, FL, 32610, USA
- Center for NeuroGenetics, University of Florida, Gainesville, FL, 32610, USA
- UF Genetics Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Kaela N Albury
- Department of Molecular Genetics and Microbiology, University of Florida, College of Medicine, Gainesville, FL, 32610, USA
- Center for NeuroGenetics, University of Florida, Gainesville, FL, 32610, USA
- UF Genetics Institute, University of Florida, Gainesville, FL, 32610, USA
| | - C Spencer Henley-Beasley
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA
- Myology Institute, University of Florida, Gainesville, FL, USA
| | - Elisabeth R Barton
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA
- Myology Institute, University of Florida, Gainesville, FL, USA
| | - Malcolm Maden
- UF Genetics Institute, University of Florida, Gainesville, FL, 32610, USA
- Department of Biology, University of Florida, Gainesville, FL, 32611, USA
| | - Maurice S Swanson
- Department of Molecular Genetics and Microbiology, University of Florida, College of Medicine, Gainesville, FL, 32610, USA.
- Center for NeuroGenetics, University of Florida, Gainesville, FL, 32610, USA.
- UF Genetics Institute, University of Florida, Gainesville, FL, 32610, USA.
- Myology Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
3
|
Basurto IM, Boudreau RD, Bandara GC, Muhammad SA, Christ GJ, Caliari SR. Freeze-dried porous collagen scaffolds for the repair of volumetric muscle loss injuries. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.30.610194. [PMID: 39282357 PMCID: PMC11398406 DOI: 10.1101/2024.08.30.610194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Volumetric muscle loss (VML) injuries are characterized by the traumatic loss of skeletal muscle resulting in permanent damage to both tissue architecture and electrical excitability. To address this challenge, we previously developed a 3D aligned collagen-glycosaminoglycan (CG) scaffold platform that supported in vitro myotube alignment and maturation. In this work, we assessed the ability of CG scaffolds to facilitate functional muscle recovery in a rat tibialis anterior (TA) model of VML. Functional muscle recovery was assessed following implantation of either non-conductive CG or electrically conductive CG-polypyrrole (PPy) scaffolds at 4, 8, and 12 weeks post-injury by in vivo electrical stimulation of the peroneal nerve. After 12 weeks, scaffold-treated muscles produced maximum isometric torque that was significantly greater than non-treated tissues. Histological analysis further supported these reparative outcomes with evidence of regenerating muscle fibers at the material-tissue interface in scaffold-treated tissues that was not observed in non-repaired muscles. Scaffold-treated muscles possessed higher numbers of M1 and M2 macrophages at the injury while conductive CG-PPy scaffold-treated muscles showed significantly higher levels of neovascularization as indicated by the presence of pericytes and endothelial cells, suggesting a persistent wound repair response not observed in non-treated tissues. Finally, only tissues treated with non-conductive CG scaffolds displayed neurofilament staining similar to native muscle, further corroborating isometric contraction data. Together, these findings show that CG scaffolds can facilitate improved skeletal muscle function and endogenous cellular repair, highlighting their potential use as therapeutics for VML injuries.
Collapse
Affiliation(s)
- Ivan M. Basurto
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22903
| | - Ryann D. Boudreau
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22903
| | - Geshani C. Bandara
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22903
| | - Samir A. Muhammad
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22903
| | - George J. Christ
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22903
- Department of Orthopedic Surgery, University of Virginia, Charlottesville, Virginia 22903
| | - Steven R. Caliari
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22903
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22903
| |
Collapse
|
4
|
Zhou Z, Liu J, Xiong T, Liu Y, Tuan RS, Li ZA. Engineering Innervated Musculoskeletal Tissues for Regenerative Orthopedics and Disease Modeling. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310614. [PMID: 38200684 DOI: 10.1002/smll.202310614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/28/2023] [Indexed: 01/12/2024]
Abstract
Musculoskeletal (MSK) disorders significantly burden patients and society, resulting in high healthcare costs and productivity loss. These disorders are the leading cause of physical disability, and their prevalence is expected to increase as sedentary lifestyles become common and the global population of the elderly increases. Proper innervation is critical to maintaining MSK function, and nerve damage or dysfunction underlies various MSK disorders, underscoring the potential of restoring nerve function in MSK disorder treatment. However, most MSK tissue engineering strategies have overlooked the significance of innervation. This review first expounds upon innervation in the MSK system and its importance in maintaining MSK homeostasis and functions. This will be followed by strategies for engineering MSK tissues that induce post-implantation in situ innervation or are pre-innervated. Subsequently, research progress in modeling MSK disorders using innervated MSK organoids and organs-on-chips (OoCs) is analyzed. Finally, the future development of engineering innervated MSK tissues to treat MSK disorders and recapitulate disease mechanisms is discussed. This review provides valuable insights into the underlying principles, engineering methods, and applications of innervated MSK tissues, paving the way for the development of targeted, efficacious therapies for various MSK conditions.
Collapse
Affiliation(s)
- Zhilong Zhou
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Jun Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Tiandi Xiong
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Yuwei Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, 518000, P. R. China
| | - Rocky S Tuan
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Zhong Alan Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518057, P. R. China
| |
Collapse
|
5
|
Li LK, Huang WC, Hsueh YY, Yamauchi K, Olivares N, Davila R, Fang J, Ding X, Zhao W, Soto J, Hasani M, Novitch B, Li S. Intramuscular delivery of neural crest stem cell spheroids enhances neuromuscular regeneration after denervation injury. Stem Cell Res Ther 2022; 13:205. [PMID: 35578348 PMCID: PMC9109326 DOI: 10.1186/s13287-022-02877-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 03/28/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Muscle denervation from trauma and motor neuron disease causes disabling morbidities. A limiting step in functional recovery is the regeneration of neuromuscular junctions (NMJs) for reinnervation. Stem cells have the potential to promote these regenerative processes, but current approaches have limited success, and the optimal types of stem cells remain to be determined. Neural crest stem cells (NCSCs), as the developmental precursors of the peripheral nervous system, are uniquely advantageous, but the role of NCSCs in neuromuscular regeneration is not clear. Furthermore, a cell delivery approach that can maintain NCSC survival upon transplantation is critical. METHODS We established a streamlined protocol to derive, isolate, and characterize functional p75+ NCSCs from human iPSCs without genome integration of reprogramming factors. To enhance survival rate upon delivery in vivo, NCSCs were centrifuged in microwell plates to form spheroids of desirable size by controlling suspension cell density. Human bone marrow mesenchymal stem cells (MSCs) were also studied for comparison. NCSC or MSC spheroids were injected into the gastrocnemius muscle with denervation injury, and the effects on NMJ formation and functional recovery were investigated. The spheroids were also co-cultured with engineered neuromuscular tissue to assess effects on NMJ formation in vitro. RESULTS NCSCs cultured in spheroids displayed enhanced secretion of soluble factors involved in neuromuscular regeneration. Intramuscular transplantation of spheroids enabled long-term survival and retention of NCSCs, in contrast to the transplantation of single-cell suspensions. Furthermore, NCSC spheroids significantly improved functional recovery after four weeks as shown by gait analysis, electrophysiology, and the rate of NMJ innervation. MSC spheroids, on the other hand, had insignificant effect. In vitro co-culture of NCSC or MSC spheroids with engineered myotubes and motor neurons further evidenced improved innervated NMJ formation with NCSC spheroids. CONCLUSIONS We demonstrate that stem cell type is critical for neuromuscular regeneration and that NCSCs have a distinct advantage and therapeutic potential to promote reinnervation following peripheral nerve injury. Biophysical effects of spheroidal culture, in particular, enable long-term NCSC survival following in vivo delivery. Furthermore, synthetic neuromuscular tissue, or "tissues-on-a-chip," may offer a platform to evaluate stem cells for neuromuscular regeneration.
Collapse
Affiliation(s)
- LeeAnn K Li
- Departments of Bioengineering and Department of Medicine, University of California, Los Angeles, USA
- David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Wen-Chin Huang
- Departments of Bioengineering and Department of Medicine, University of California, Los Angeles, USA
| | - Yuan-Yu Hsueh
- Departments of Bioengineering and Department of Medicine, University of California, Los Angeles, USA
- Division of Plastic and Reconstructive Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ken Yamauchi
- Department of Neurobiology, University of California, Los Angeles, USA
| | - Natalie Olivares
- Departments of Bioengineering and Department of Medicine, University of California, Los Angeles, USA
| | - Raul Davila
- Departments of Bioengineering and Department of Medicine, University of California, Los Angeles, USA
| | - Jun Fang
- Departments of Bioengineering and Department of Medicine, University of California, Los Angeles, USA
| | - Xili Ding
- Departments of Bioengineering and Department of Medicine, University of California, Los Angeles, USA
| | - Weikang Zhao
- Departments of Bioengineering and Department of Medicine, University of California, Los Angeles, USA
| | - Jennifer Soto
- Departments of Bioengineering and Department of Medicine, University of California, Los Angeles, USA
| | - Mahdi Hasani
- Departments of Bioengineering and Department of Medicine, University of California, Los Angeles, USA
| | - Bennett Novitch
- Department of Neurobiology, University of California, Los Angeles, USA
| | - Song Li
- Departments of Bioengineering and Department of Medicine, University of California, Los Angeles, USA.
| |
Collapse
|
6
|
Lopez TCC, Malavazzi TCDS, Rodrigues MFSD, Bach EE, Silva DT, Hi EMB, França CM, Bussadori SK, Mesquita-Ferrari RA, Fernandes KPS. Histological and biochemical effects of preventive and therapeutic vascular photobiomodulation on rat muscle injury. JOURNAL OF BIOPHOTONICS 2022; 15:e202100271. [PMID: 34978386 DOI: 10.1002/jbio.202100271] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/26/2021] [Accepted: 12/24/2021] [Indexed: 06/14/2023]
Abstract
The intravascular or transcutaneous application of photobiomodulation (PBM) over blood vessels (vascular photobiomodulation, VPBM) has been used for the treatment of inflammatory and chronic conditions with promising systemic results. This study evaluated the VPBM effects on a model of muscle regeneration after acute injury and compared the outcomes of preventive and therapeutic VPBM. Transcutaneous VPBM was administered over the rat's main tail vein. Serum levels of creatine kinase (CK), aspartate aminotransferase (AST), and lactate were evaluated and muscles were processed for macroscopic and microscopic analysis. Preventive and therapeutic VPBM led to decreased inflammatory infiltrate, edema, and myonecrosis but with an increase in immature muscle fibers. CK, AST, and lactate levels were lower in the groups treated with VPBM (lowest concentrations in preventive VPBM application). Preventive and therapeutic VPBM were capable of exerting a positive effect on acute muscle injury repair, with more accentuated results when preventive VPBM was administered.
Collapse
Affiliation(s)
- Talita Christine Camillo Lopez
- Postgraduate Program in Biophotonics Applied to the Health Sciences, Universidade Nove de Julho (UNINOVE), São Paulo, São Paulo, Brazil
| | | | | | | | - Daniela Teixeira Silva
- Postgraduate Program in Biophotonics Applied to the Health Sciences, Universidade Nove de Julho (UNINOVE), São Paulo, São Paulo, Brazil
| | - Edgar Matias Bach Hi
- Experimental Biochemistry Academic Nucleum (NABEX), UNILUS (Centro Universitário Lusiada), Santos, São Paulo, Brazil
| | - Cristiane Miranda França
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon, USA
| | - Sandra Kalil Bussadori
- Postgraduate Program in Biophotonics Applied to the Health Sciences, Universidade Nove de Julho (UNINOVE), São Paulo, São Paulo, Brazil
- Postgraduate Program in Rehabilitation Sciences, UNINOVE, São Paulo, São Paulo, Brazil
| | - Raquel Agnelli Mesquita-Ferrari
- Postgraduate Program in Biophotonics Applied to the Health Sciences, Universidade Nove de Julho (UNINOVE), São Paulo, São Paulo, Brazil
- Postgraduate Program in Rehabilitation Sciences, UNINOVE, São Paulo, São Paulo, Brazil
| | - Kristianne Porta Santos Fernandes
- Postgraduate Program in Biophotonics Applied to the Health Sciences, Universidade Nove de Julho (UNINOVE), São Paulo, São Paulo, Brazil
| |
Collapse
|
7
|
Iberite F, Gruppioni E, Ricotti L. Skeletal muscle differentiation of human iPSCs meets bioengineering strategies: perspectives and challenges. NPJ Regen Med 2022; 7:23. [PMID: 35393412 PMCID: PMC8991236 DOI: 10.1038/s41536-022-00216-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 03/01/2022] [Indexed: 12/31/2022] Open
Abstract
Although skeletal muscle repairs itself following small injuries, genetic diseases or severe damages may hamper its ability to do so. Induced pluripotent stem cells (iPSCs) can generate myogenic progenitors, but their use in combination with bioengineering strategies to modulate their phenotype has not been sufficiently investigated. This review highlights the potential of this combination aimed at pushing the boundaries of skeletal muscle tissue engineering. First, the overall organization and the key steps in the myogenic process occurring in vivo are described. Second, transgenic and non-transgenic approaches for the myogenic induction of human iPSCs are compared. Third, technologies to provide cells with biophysical stimuli, biomaterial cues, and biofabrication strategies are discussed in terms of recreating a biomimetic environment and thus helping to engineer a myogenic phenotype. The embryonic development process and the pro-myogenic role of the muscle-resident cell populations in co-cultures are also described, highlighting the possible clinical applications of iPSCs in the skeletal muscle tissue engineering field.
Collapse
Affiliation(s)
- Federica Iberite
- The BioRobotics Institute, Scuola Superiore Sant'Anna, 56127, Pisa (PI), Italy. .,Department of Excellence in Robotics & AI, Scuola Superiore Sant'Anna, 56127, Pisa (PI), Italy.
| | - Emanuele Gruppioni
- Centro Protesi INAIL, Istituto Nazionale per l'Assicurazione contro gli Infortuni sul Lavoro, 40054, Vigorso di Budrio (BO), Italy
| | - Leonardo Ricotti
- The BioRobotics Institute, Scuola Superiore Sant'Anna, 56127, Pisa (PI), Italy.,Department of Excellence in Robotics & AI, Scuola Superiore Sant'Anna, 56127, Pisa (PI), Italy
| |
Collapse
|
8
|
Jeong GJ, Castels H, Kang I, Aliya B, Jang YC. Nanomaterial for Skeletal Muscle Regeneration. Tissue Eng Regen Med 2022; 19:253-261. [PMID: 35334091 PMCID: PMC8971233 DOI: 10.1007/s13770-022-00446-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/15/2022] [Accepted: 02/20/2022] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle has an innate regenerative capacity to restore their structure and function following acute damages and injuries. However, in congenital muscular dystrophies, large volumetric muscle loss, cachexia, or aging, the declined regenerative capacity of skeletal muscle results in muscle wasting and functional impairment. Recent studies indicate that muscle mass and function are closely correlated with morbidity and mortality due to the large volume and location of skeletal muscle. However, the options for treating neuromuscular disorders are limited. Biomedical engineering strategies such as nanotechnologies have been implemented to address this issue.In this review, we focus on recent studies leveraging nano-sized materials for regeneration of skeletal muscle. We look at skeletal muscle pathologies and describe various proof-of-concept and pre-clinical studies that have used nanomaterials, with a focus on how nano-sized materials can be used for skeletal muscle regeneration depending on material dimensionality.Depending on the dimensionality of nano-sized materials, their application have been changed because of their different physical and biochemical properties.Nanomaterials have been spotlighted as a great candidate for addressing the unmet needs of regenerative medicine. Nanomaterials could be applied to several types of tissues and diseases along with the unique characteristics of nanomaterials. However, when confined to muscle tissue, the targets of nanomaterial applications are limited and can be extended in future research.
Collapse
Affiliation(s)
- Gun-Jae Jeong
- Department of Orthopedics, Emory Musculoskeletal Institute, Emory School of Medicine, Atlanta, GA, 30329, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory School of Medicine, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Hannah Castels
- Department of Orthopedics, Emory Musculoskeletal Institute, Emory School of Medicine, Atlanta, GA, 30329, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Innie Kang
- Department of Orthopedics, Emory Musculoskeletal Institute, Emory School of Medicine, Atlanta, GA, 30329, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Berna Aliya
- Department of Orthopedics, Emory Musculoskeletal Institute, Emory School of Medicine, Atlanta, GA, 30329, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Young C Jang
- Department of Orthopedics, Emory Musculoskeletal Institute, Emory School of Medicine, Atlanta, GA, 30329, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory School of Medicine, Atlanta, GA, 30332, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| |
Collapse
|
9
|
Cho S, Jang J. Recent Trends in Biofabrication Technologies for Studying Skeletal Muscle Tissue-Related Diseases. Front Bioeng Biotechnol 2021; 9:782333. [PMID: 34778240 PMCID: PMC8578921 DOI: 10.3389/fbioe.2021.782333] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 10/18/2021] [Indexed: 01/15/2023] Open
Abstract
In native skeletal muscle, densely packed myofibers exist in close contact with surrounding motor neurons and blood vessels, which are embedded in the fibrous connective tissue. In comparison to conventional two-dimensional (2D) cultures, the three-dimensional (3D) engineered skeletal muscle models allow structural and mechanical resemblance with native skeletal muscle tissue by providing geometric confinement and physiological matrix stiffness to the cells. In addition, various external stimuli applied to these models enhance muscle maturation along with cell-cell and cell-extracellular matrix interaction. Therefore, 3D in vitro muscle models can adequately recapitulate the pathophysiologic events occurring in tissue-tissue interfaces inside the native skeletal muscle such as neuromuscular junction. Moreover, 3D muscle models can induce pathological phenotype of human muscle dystrophies such as Duchenne muscular dystrophy by incorporating patient-derived induced pluripotent stem cells and human primary cells. In this review, we discuss the current biofabrication technologies for modeling various skeletal muscle tissue-related diseases (i.e., muscle diseases) including muscular dystrophies and inflammatory muscle diseases. In particular, these approaches would enable the discovery of novel phenotypic markers and the mechanism study of human muscle diseases with genetic mutations.
Collapse
Affiliation(s)
- Seungyeun Cho
- Department of Convergence IT Engineering, Pohang University of Science and Technology, Pohang, South Korea
| | - Jinah Jang
- Department of Convergence IT Engineering, Pohang University of Science and Technology, Pohang, South Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, South Korea
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, South Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, South Korea
| |
Collapse
|
10
|
Abdel-Raouf KMA, Rezgui R, Stefanini C, Teo JCM, Christoforou N. Transdifferentiation of Human Fibroblasts into Skeletal Muscle Cells: Optimization and Assembly into Engineered Tissue Constructs through Biological Ligands. BIOLOGY 2021; 10:biology10060539. [PMID: 34208436 PMCID: PMC8235639 DOI: 10.3390/biology10060539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 11/16/2022]
Abstract
Simple Summary Engineered human skeletal muscle tissue is a platform tool that can help scientists and physicians better understand human physiology, pharmacology, and disease modeling. Over the past few years this area of research has been actively being pursued by many labs worldwide. Significant challenges remain, including accessing an adequate cell source, and achieving proper physiological-like architecture of the engineered tissue. To address cell resourcing we aimed at further optimizing a process called transdifferentiation which involves the direct conversion of fibroblasts into skeletal muscle cells. The opportunity here is that fibroblasts are readily available and can be expanded sufficiently to meet the needs of a tissue engineering approach. Additionally, we aimed to demonstrate the applicability of transdifferentiation in assembling tissue engineered skeletal muscle. We implemented a screening process of protein ligands in an effort to refine transdifferentiation, and identified that most proteins resulted in a deficit in transdifferentiation efficiency, although one resulted in robust expansion of cultured cells. We were also successful in assembling engineered constructs consisting of transdifferentiated cells. Future directives involve demonstrating that the engineered tissues are capable of contractile and functional activity, and pursuit of optimizing factors such as electrical and chemical exposure, towards achieving physiological parameters observed in human muscle. Abstract The development of robust skeletal muscle models has been challenging due to the partial recapitulation of human physiology and architecture. Reliable and innovative 3D skeletal muscle models recently described offer an alternative that more accurately captures the in vivo environment but require an abundant cell source. Direct reprogramming or transdifferentiation has been considered as an alternative. Recent reports have provided evidence for significant improvements in the efficiency of derivation of human skeletal myotubes from human fibroblasts. Herein we aimed at improving the transdifferentiation process of human fibroblasts (tHFs), in addition to the differentiation of murine skeletal myoblasts (C2C12), and the differentiation of primary human skeletal myoblasts (HSkM). Differentiating or transdifferentiating cells were exposed to single or combinations of biological ligands, including Follistatin, GDF8, FGF2, GDF11, GDF15, hGH, TMSB4X, BMP4, BMP7, IL6, and TNF-α. These were selected for their critical roles in myogenesis and regeneration. C2C12 and tHFs displayed significant differentiation deficits when exposed to FGF2, BMP4, BMP7, and TNF-α, while proliferation was significantly enhanced by FGF2. When exposed to combinations of ligands, we observed consistent deficit differentiation when TNF-α was included. Finally, our direct reprogramming technique allowed for the assembly of elongated, cross-striated, and aligned tHFs within tissue-engineered 3D skeletal muscle constructs. In conclusion, we describe an efficient system to transdifferentiate human fibroblasts into myogenic cells and a platform for the generation of tissue-engineered constructs. Future directions will involve the evaluation of the functional characteristics of these engineered tissues.
Collapse
Affiliation(s)
- Khaled M. A. Abdel-Raouf
- Department of Biomedical Engineering, Khalifa University, Abu Dhabi 127788, United Arab Emirates;
- Department of Biology, American University in Cairo, New Cairo 11835, Egypt
- Correspondence: (K.M.A.A.-R.); (N.C.)
| | - Rachid Rezgui
- Core Technology Platforms, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates;
| | - Cesare Stefanini
- Department of Biomedical Engineering, Khalifa University, Abu Dhabi 127788, United Arab Emirates;
- Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Jeremy C. M. Teo
- Department of Mechanical and Biomedical Engineering, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates;
| | - Nicolas Christoforou
- Pfizer Inc., Rare Disease Research Unit, 610 Main Street, Cambridge, MA 02139, USA
- Correspondence: (K.M.A.A.-R.); (N.C.)
| |
Collapse
|
11
|
Basurto IM, Mora MT, Gardner GM, Christ GJ, Caliari SR. Aligned and electrically conductive 3D collagen scaffolds for skeletal muscle tissue engineering. Biomater Sci 2021; 9:4040-4053. [PMID: 33899845 DOI: 10.1039/d1bm00147g] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Skeletal muscle is characterized by its three-dimensional (3D) anisotropic architecture composed of highly aligned and electrically-excitable muscle fibers that enable normal movement. Biomaterial-based tissue engineering approaches to repair skeletal muscle are limited due to difficulties combining 3D structural alignment (to guide cell/matrix organization) and electrical conductivity (to enable electrically-excitable myotube assembly and maturation). In this work we successfully produced aligned and electrically conductive 3D collagen scaffolds using a freeze-drying approach. Conductive polypyrrole (PPy) nanoparticles were synthesized and directly mixed into a suspension of type I collagen and chondroitin sulfate followed by directional lyophilization. Scanning electron microscopy (SEM), energy-dispersive spectroscopy (EDS), and confocal microscopy showed that directional solidification resulted in scaffolds with longitudinally aligned pores with homogeneously-distributed PPy content. Chronopotentiometry verified that PPy incorporation resulted in a five-fold increase in conductivity compared to non-PPy-containing collagen scaffolds without detrimentally affecting myoblast metabolic activity. Furthermore, the aligned scaffold microstructure provided contact guidance cues that directed myoblast growth and organization. Incorporation of PPy also promoted enhanced myotube formation and maturation as measured by myosin heavy chain (MHC) expression and number of nuclei per myotube. Together these data suggest that aligned and electrically conductive 3D collagen scaffolds could be useful for skeletal muscle tissue engineering.
Collapse
Affiliation(s)
| | | | | | - George J Christ
- Department of Biomedical Engineering, USA. and Department of Orthopedic Surgery, University of Virginia, USA
| | - Steven R Caliari
- Department of Biomedical Engineering, USA. and Department of Chemical Engineering, USA
| |
Collapse
|
12
|
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
|
13
|
Intermittent pressure imitating rolling manipulation ameliorates injury in skeletal muscle cells through oxidative stress and lipid metabolism signalling pathways. Gene 2021; 778:145460. [PMID: 33515727 DOI: 10.1016/j.gene.2021.145460] [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: 05/17/2020] [Revised: 11/25/2020] [Accepted: 01/20/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND Traditional Chinese medicine manipulation (TCMM) is often used to treat human skeletal muscle injury, but its mechanism remains unclear due to difficulty standardizing and quantifying manipulation parameters. METHODS Here, dexamethasone sodium phosphate (DSP) was utilized to induce human skeletal muscle cell (HSkMC) impairments. Cells in a three-dimensional environment were divided into the control normal group (CNG), control injured group (CIG) and rolling manipulation group (RMG). The RMG was exposed to intermittent pressure imitating rolling manipulation (IPIRM) of TCMM via the FX‑5000™ compression system. Skeletal muscle damage was assessed via the cell proliferation rate, superoxide dismutase (SOD) activity, malondialdehyde (MDA) content and creatine kinase (CK) activity. Isobaric tagging for relative and absolute protein quantification (iTRAQ) and bioinformatic analysis were used to evaluate differentially expressed proteins (DEPs). RESULTS Higher-pressure IPIRM ameliorated the skeletal muscle cell injury induced by 1.2 mM DSP. Thirteen common DEPs after IPIRM were selected. Key biological processes, molecular functions, cellular components, and pathways were identified as mechanisms underlying the protective effect of TCMM against skeletal muscle damage. Some processes (response to oxidative stress, response to wounding, response to stress and lipid metabolism signalling pathways) were related to skeletal muscle cell injury. Western blotting for 4 DEPs confirmed the reliability of iTRAQ. CONCLUSIONS Higher-pressure IPIRM downregulated the CD36, Hsp27 and FABP4 proteins in oxidative stress and lipid metabolism pathways, alleviating excessive oxidative stress and lipid metabolism disorder in injured HSkMCs. The techniques used in this study might provide novel insights into the mechanism of TCMM.
Collapse
|
14
|
Ding R, Horie M, Nagasaka S, Ohsumi S, Shimizu K, Honda H, Nagamori E, Fujita H, Kawamoto T. Effect of cell-extracellular matrix interaction on myogenic characteristics and artificial skeletal muscle tissue. J Biosci Bioeng 2020; 130:98-105. [PMID: 32278672 DOI: 10.1016/j.jbiosc.2020.02.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 01/23/2023]
Abstract
Although various types of artificial skeletal muscle tissue have been reported, the contractile forces generated by tissue-engineered artificial skeletal muscles remain to be improved for biological model and clinical applications. In this study, we investigated the effects of extracellular matrix (ECM) and supplementation of a small molecule, which has been reported to enhance α7β1 integrin expression (SU9516), on cell migration speed, cell fusion rate, myoblast (mouse C2C12 cells) differentiation and contractile force generation of tissue-engineered artificial skeletal muscles. When cells were cultured on varying ECM coated-surfaces, we observed significant enhancement in the migration speed, while the myotube formation (differentiation ratio) decreased in all except for cells cultured on Matrigel coated-surfaces. In contrast, SU9516 supplementation resulted in an increase in both the myotube width and differentiation ratio. Following combined culture with a Matrigel-coated surface and SU9516 supplementation, myotube width was further increased. Additionally, contractile forces produced by the tissue-engineered artificial skeletal muscles was augmented following combined culture. These findings indicate that regulation of the cell-ECM interaction is a promising approach to improve the function of tissue-engineered artificial skeletal muscles.
Collapse
Affiliation(s)
- Ran Ding
- Graduate School of Human and Environmental, Kyoto University, Yoshida-Konoe-Cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Masanobu Horie
- Division of Biochemical Engineering, Radioisotope Research Center, Kyoto University, Yoshida-Konoe-Cho, Sakyo-ku, Kyoto 606-8507, Japan.
| | - Sumire Nagasaka
- 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
| | - Kazunori Shimizu
- 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; Innovative Research Center for Preventive Medical Engineering, Nagoya University, Nagoya 464-8601, Japan
| | - Eiji Nagamori
- Department of Biomedical Engineering, Osaka Institute of Technology, 5-16-1 Omiya, Asahi-ku, Osaka 535-8585, Japan
| | - Hideaki Fujita
- WPI, Immunology Frontier Research Center, Institute of Scientific and Industrial Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Takuo Kawamoto
- Graduate School of Human and Environmental, Kyoto University, Yoshida-Konoe-Cho, Sakyo-ku, Kyoto 606-8507, Japan; Division of Biological Chemistry, Radioisotope Research Center, Kyoto University, Yoshida-Konoe-Cho, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
15
|
Gilbert-Honick J, Grayson W. Vascularized and Innervated Skeletal Muscle Tissue Engineering. Adv Healthc Mater 2020; 9:e1900626. [PMID: 31622051 PMCID: PMC6986325 DOI: 10.1002/adhm.201900626] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 09/27/2019] [Indexed: 12/12/2022]
Abstract
Volumetric muscle loss (VML) is a devastating loss of muscle tissue that overwhelms the native regenerative properties of skeletal muscle and results in lifelong functional deficits. There are currently no treatments for VML that fully recover the lost muscle tissue and function. Tissue engineering presents a promising solution for VML treatment and significant research has been performed using tissue engineered muscle constructs in preclinical models of VML with a broad range of defect locations and sizes, tissue engineered construct characteristics, and outcome measures. Due to the complex vascular and neural anatomy within skeletal muscle, regeneration of functional vasculature and nerves is vital for muscle recovery following VML injuries. This review aims to summarize the current state of the field of skeletal muscle tissue engineering using 3D constructs for VML treatment with a focus on studies that have promoted vascular and neural regeneration within the muscle tissue post-VML.
Collapse
Affiliation(s)
- Jordana Gilbert-Honick
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Warren Grayson
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Material Sciences & Engineering, Johns Hopkins University, School of Engineering, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology (INBT), Johns Hopkins University School of Engineering, Baltimore, MD 21218, USA
| |
Collapse
|
16
|
Tiburcy M, Markov A, Kraemer LK, Christalla P, Rave‐Fraenk M, Fischer HJ, Reichardt HM, Zimmermann W. Regeneration competent satellite cell niches in rat engineered skeletal muscle. FASEB Bioadv 2019; 1:731-746. [PMID: 32123818 PMCID: PMC6996344 DOI: 10.1096/fba.2019-00013] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 02/21/2019] [Accepted: 10/30/2019] [Indexed: 12/22/2022] Open
Abstract
Satellite cells reside in defined niches and are activated upon skeletal muscle injury to facilitate regeneration. Mechanistic studies of skeletal muscle regeneration are hampered by the inability to faithfully simulate satellite cell biology in vitro. We sought to overcome this limitation by developing tissue engineered skeletal muscle (ESM) with (1) satellite cell niches and (2) the capacity to regenerate after injury. ESMs contained quiescent Pax7-positive satellite cells in morphologically defined niches. Satellite cells could be activated to repair (i) cardiotoxin and (ii) mechanical crush injuries. Activation of the Wnt-pathway was essential for muscle regeneration. Finally, muscle progenitors from the engineered niche developed de novo ESM in vitro and regenerated skeletal muscle after cardiotoxin-induced injury in vivo. We conclude that ESM with functional progenitor niches reminiscent of the in vivo satellite cell niches can be engineered in vitro. ESM may ultimately be exploited in disease modeling, drug screening, or muscle regeneration.
Collapse
Affiliation(s)
- Malte Tiburcy
- Institute of Pharmacology and ToxicologyGeorg‐August University GöttingenGöttingenGermany
- DZHK (German Center for Cardiovascular Research), partner site GöttingenGöttingenGermany
| | - Alex Markov
- Institute of Pharmacology and ToxicologyGeorg‐August University GöttingenGöttingenGermany
- DZHK (German Center for Cardiovascular Research), partner site GöttingenGöttingenGermany
| | - Lena K. Kraemer
- Institute of Pharmacology and ToxicologyGeorg‐August University GöttingenGöttingenGermany
- DZHK (German Center for Cardiovascular Research), partner site GöttingenGöttingenGermany
| | - Peter Christalla
- Institute of Pharmacology and ToxicologyGeorg‐August University GöttingenGöttingenGermany
| | - Margret Rave‐Fraenk
- Department of Radiation Therapy and RadiooncologyGeorg‐August University GöttingenGöttingenGermany
| | - Henrike J. Fischer
- Institute for Cellular and Molecular ImmunologyGeorg‐August University GöttingenGöttingenGermany
- Present address:
Institute for ImmunologyMedical FacultyRWTH Aachen UniversityAachenGermany
| | - Holger M. Reichardt
- Institute for Cellular and Molecular ImmunologyGeorg‐August University GöttingenGöttingenGermany
| | - Wolfram‐Hubertus Zimmermann
- Institute of Pharmacology and ToxicologyGeorg‐August University GöttingenGöttingenGermany
- DZHK (German Center for Cardiovascular Research), partner site GöttingenGöttingenGermany
| |
Collapse
|
17
|
Greising SM, Corona BT, McGann C, Frankum JK, Warren GL. Therapeutic Approaches for Volumetric Muscle Loss Injury: A Systematic Review and Meta-Analysis. TISSUE ENGINEERING PART B-REVIEWS 2019; 25:510-525. [PMID: 31578930 DOI: 10.1089/ten.teb.2019.0207] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Our goal was to understand the impact of regenerative therapies on the functional capacity of skeletal muscle following volumetric muscle loss (VML) injury. An extensive database search (e.g., PubMed, Cochrane Library, and ClinicalTrials.gov) was conducted up through January 2019 to evaluate the following: "In humans or animals with VML injury, is treatment better than no treatment at recovering functional capacity?" Study eligibility criteria required studies to have both an untreated and at least one treated VML injury group. From 2312 study reports, 44 studies met the inclusion criteria. Quantitative functional capacity data (absolute and/or normalized strength) or proportional measures (histological analysis quantifying viable muscle tissue, mitochondrial function, and/or exhaustive treadmill running) were extracted for use. While both human and animal studies were included in the searches, only animal studies met the eligibility criteria. Using a random-effects model, Hedges' g was used as the effect size (ES) and calculated such that a positive ES indicated treatment efficacy. The overall ES was 0.75 (95% confidence interval: 0.53-0.96; p < 0.0000001), indicating that the treatments, on average, resulted in a significant improvement in functional capacity. From network meta-analyses, it was determined that an acellular biomaterial combined with stem and/or progenitor cells had the greatest treatment effectiveness. The findings indicate that various treatments in animal models of VML improve the functional capacity of muscle compared to leaving the injury untreated; however, the ∼16% beneficial effect is small. Our results suggest that current regenerative therapy paradigms require further maturation to achieve clinically meaningful improvements in the functional capacity of the muscle. Impact Statement Our most salient findings are that (1) various treatment approaches used in animal models of volumetric muscle loss (VML) injury improve functional capacity compared to leaving the injury untreated and (2) an acellular biomaterial in combination with cellular components was the most effective treatment to improve functional capacity following VML injury to date. The nature of our findings has substantial implications for regenerative medicine, biomedical engineering, and rehabilitative techniques currently being evaluated and developed for VML injury repair, and are pivotal to the progression of the regenerative medicine effort aimed at restoring maximal function to traumatized and disabled limbs.
Collapse
Affiliation(s)
- Sarah M Greising
- School of Kinesiology, University of Minnesota, Minneapolis, Minnesota
| | - Benjamin T Corona
- School of Medicine, Wake Forest University, Winston-Salem, North Carolina
| | - Christopher McGann
- Department of Physical Therapy, Georgia State University, Atlanta, Georgia
| | - Jeremy K Frankum
- Department of Physical Therapy, Georgia State University, Atlanta, Georgia
| | - Gordon L Warren
- Department of Physical Therapy, Georgia State University, Atlanta, Georgia
| |
Collapse
|
18
|
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
|
19
|
Somers SM, Zhang NY, Morrissette-McAlmon JB, Tran K, Mao HQ, Grayson WL. Myoblast maturity on aligned microfiber bundles at the onset of strain application impacts myogenic outcomes. Acta Biomater 2019; 94:232-242. [PMID: 31212110 DOI: 10.1016/j.actbio.2019.06.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 05/24/2019] [Accepted: 06/14/2019] [Indexed: 10/26/2022]
Abstract
Engineered skeletal muscle grafts may be employed in various applications including the treatment of volumetric muscle loss (VML) and pharmacological drug screening. To recapitulate the well-defined structure of native muscle, tensile strains have been applied to the grafts. In this study, we cultured C2C12 murine myoblasts on electrospun fibrin microfiber bundles for 7 days in custom-built bioreactor units and investigated the impact of strain regimen and delayed onset of tensile straining on myogenic outcomes. The substrate topography induced uniaxial alignment of cells in all (strained and unstrained) groups. The engineered grafts in strained groups were subjected to 10% strain amplitude for 6 h per day. We found that both static and cyclic uniaxial strains resulted in similar morphological and gene expression outcomes. However, relative to 0% strain groups, there were stark increases in myotube diameter, myosin heavy chain (MHC) coverage, and expression of key myogenic genes (Pax 7, Troponin, MHC I, MHC IIb, MHC IIx) only if strain was applied at Days 5-7 rather than Days 3-7. This finding suggests that a critical indicator of myogenic improvement under strain in our system is the phenotype of the cells at the onset of strain and suggests that this is a key parameter that should be considered in studies where myoblasts are subjected to biophysical stimulation to promote tissue formation. STATEMENT OF SIGNIFICANCE: This is the first report on the impact of the timing of the initial application of mechanical strain for improving the myogenic outcomes of 3D engineered skeletal muscle grafts. In this work, immature skeletal myoblasts were grown on topographically aligned, electrospun fibrin microfiber bundles and we applied 10% uniaxial static or cyclic strain. We concluded that the maturity of myoblasts prior to strain application, rather than strain waveform, was the primary predictor of improved myogenic outcomes, including myogenic gene expression and myotube morphology. Elucidating the optimal conditions for strain application is a vital step in recapitulating physiological myogenic properties in tissue engineered skeletal muscle constructs, with applications for treating volumetric muscle loss, disease modeling, and drug testing.
Collapse
|
20
|
Von den Hoff JW, Carvajal Monroy PL, Ongkosuwito EM, van Kuppevelt TH, Daamen WF. Muscle fibrosis in the soft palate: Delivery of cells, growth factors and anti-fibrotics. Adv Drug Deliv Rev 2019; 146:60-76. [PMID: 30107211 DOI: 10.1016/j.addr.2018.08.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/29/2018] [Accepted: 08/06/2018] [Indexed: 02/07/2023]
Abstract
The healing of skeletal muscle injuries after major trauma or surgical reconstruction is often complicated by the development of fibrosis leading to impaired function. Research in the field of muscle regeneration is mainly focused on the restoration of muscle mass while far less attention is paid to the prevention of fibrosis. In this review, we take as an example the reconstruction of the muscles in the soft palate of cleft palate patients. After surgical closure of the soft palate, muscle function during speech is often impaired by a shortage of muscle tissue as well as the development of fibrosis. We will give a short overview of the most common approaches to generate muscle mass and then focus on strategies to prevent fibrosis. These include anti-fibrotic strategies that have been developed for muscle and other organs by the delivery of small molecules, decorin and miRNAs. Anti-fibrotic compounds should be delivered in aligned constructs in order to obtain the organized architecture of muscle tissue. The available techniques for the preparation of aligned muscle constructs will be discussed. The combination of approaches to generate muscle mass with anti-fibrotic components in an aligned muscle construct may greatly improve the functional outcome of regenerative therapies for muscle injuries.
Collapse
Affiliation(s)
- Johannes W Von den Hoff
- Department of Orthodontics and Craniofacial Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500HB Nijmegen, The Netherlands.
| | - Paola L Carvajal Monroy
- Department of Oral and Maxillofacial Surgery, Special Dental Care and Orthodontics, Erasmus Medical Center, P.O. Box 2060, 3000CB Rotterdam, The Netherlands.
| | - Edwin M Ongkosuwito
- Department of Orthodontics and Craniofacial Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500HB Nijmegen, The Netherlands.
| | - Toin H van Kuppevelt
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500HB Nijmegen, The Netherlands.
| | - Willeke F Daamen
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500HB Nijmegen, The Netherlands.
| |
Collapse
|
21
|
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
|
22
|
Estrellas KM, Chung L, Cheu LA, Sadtler K, Majumdar S, Mula J, Wolf MT, Elisseeff JH, Wagner KR. Biological scaffold-mediated delivery of myostatin inhibitor promotes a regenerative immune response in an animal model of Duchenne muscular dystrophy. J Biol Chem 2018; 293:15594-15605. [PMID: 30139748 DOI: 10.1074/jbc.ra118.004417] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/16/2018] [Indexed: 01/16/2023] Open
Abstract
Recent studies have reported that the immune system significantly mediates skeletal muscle repair and regeneration. Additionally, biological scaffolds have been shown to play a role in polarizing the immune microenvironment toward pro-myogenic outcomes. Moreover, myostatin inhibitors are known to promote muscle regeneration and ameliorate fibrosis in animal models of Duchenne muscular dystrophy (DMD), a human disease characterized by chronic muscle degeneration. Biological scaffolds and myostatin inhibition can potentially influence immune-mediated regeneration in the dystrophic environment, but have not been evaluated together. Toward this end, here we created an injectable biological scaffold composed of hyaluronic acid and processed skeletal muscle extracellular matrix. This material formed a cytocompatible hydrogel at physiological temperatures in vitro When injected subfascially above the tibialis anterior muscles of both WT and dystrophic mdx-5Cv mice, a murine model of DMD, the hydrogel spreads across the entire muscle before completely degrading at 3 weeks in vivo We found that the hydrogel is associated with CD206+ pro-regenerative macrophage polarization and elevated anti-inflammatory cytokine expression in both WT and dystrophic mice. Co-injection of both hydrogel and myostatin inhibitor significantly increased FoxP3+ regulatory T cell modulation and Foxp3 gene expression in the scaffold immune microenvironment. Finally, delivery of myostatin inhibitor with the hydrogel increased its bioactivity in vivo, and transplantation of immortalized human myoblasts with the hydrogel promoted their survival in vivo This study identifies a key role for biological scaffolds and myostatin inhibitors in modulating a pro-regenerative immune microenvironment in dystrophic muscle.
Collapse
Affiliation(s)
- Kenneth M Estrellas
- From the Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland 21205.,the Translational Tissue Engineering Center and
| | - Liam Chung
- the Translational Tissue Engineering Center and.,Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
| | - Lindsay A Cheu
- the Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| | - Kaitlyn Sadtler
- the David H. Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142.,the Department of Anesthesiology, Boston Children's Hospital, Boston, Massachusetts 02115
| | | | - Jyothi Mula
- the NCI at Frederick, National Institutes of Health, Frederick, Maryland 21702, and
| | - Matthew T Wolf
- the Translational Tissue Engineering Center and.,Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
| | - Jennifer H Elisseeff
- the Translational Tissue Engineering Center and .,Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
| | - Kathryn R Wagner
- From the Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland 21205, .,the Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
| |
Collapse
|
23
|
Maleiner B, Tomasch J, Heher P, Spadiut O, Rünzler D, Fuchs C. The Importance of Biophysical and Biochemical Stimuli in Dynamic Skeletal Muscle Models. Front Physiol 2018; 9:1130. [PMID: 30246791 PMCID: PMC6113794 DOI: 10.3389/fphys.2018.01130] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 07/30/2018] [Indexed: 12/31/2022] Open
Abstract
Classical approaches to engineer skeletal muscle tissue based on current regenerative and surgical procedures still do not meet the desired outcome for patient applications. Besides the evident need to create functional skeletal muscle tissue for the repair of volumetric muscle defects, there is also growing demand for platforms to study muscle-related diseases, such as muscular dystrophies or sarcopenia. Currently, numerous studies exist that have employed a variety of biomaterials, cell types and strategies for maturation of skeletal muscle tissue in 2D and 3D environments. However, researchers are just at the beginning of understanding the impact of different culture settings and their biochemical (growth factors and chemical changes) and biophysical cues (mechanical properties) on myogenesis. With this review we intend to emphasize the need for new in vitro skeletal muscle (disease) models to better recapitulate important structural and functional aspects of muscle development. We highlight the importance of choosing appropriate system components, e.g., cell and biomaterial type, structural and mechanical matrix properties or culture format, and how understanding their interplay will enable researchers to create optimized platforms to investigate myogenesis in healthy and diseased tissue. Thus, we aim to deliver guidelines for experimental designs to allow estimation of the potential influence of the selected skeletal muscle tissue engineering setup on the myogenic outcome prior to their implementation. Moreover, we offer a workflow to facilitate identifying and selecting different analytical tools to demonstrate the successful creation of functional skeletal muscle tissue. Ultimately, a refinement of existing strategies will lead to further progression in understanding important aspects of muscle diseases, muscle aging and muscle regeneration to improve quality of life of patients and enable the establishment of new treatment options.
Collapse
Affiliation(s)
- Babette Maleiner
- Department of Biochemical Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria.,The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Janine Tomasch
- Department of Biochemical Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria.,The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Philipp Heher
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Ludwig Boltzmann Institute for Experimental and Clinical Traumatology/AUVA Research Center, Vienna, Austria.,Trauma Care Consult GmbH, Vienna, Austria
| | - Oliver Spadiut
- Institute of Chemical Engineering, Vienna University of Technology, Vienna, Austria
| | - Dominik Rünzler
- Department of Biochemical Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria.,The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Christiane Fuchs
- Department of Biochemical Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria.,The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
24
|
Rahman SU, Nagrath M, Ponnusamy S, Arany PR. Nanoscale and Macroscale Scaffolds with Controlled-Release Polymeric Systems for Dental Craniomaxillofacial Tissue Engineering. MATERIALS (BASEL, SWITZERLAND) 2018; 11:E1478. [PMID: 30127246 PMCID: PMC6120038 DOI: 10.3390/ma11081478] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 08/03/2018] [Accepted: 08/10/2018] [Indexed: 12/11/2022]
Abstract
Tremendous progress in stem cell biology has resulted in a major current focus on effective modalities to promote directed cellular behavior for clinical therapy. The fundamental principles of tissue engineering are aimed at providing soluble and insoluble biological cues to promote these directed biological responses. Better understanding of extracellular matrix functions is ensuring optimal adhesive substrates to promote cell mobility and a suitable physical niche to direct stem cell responses. Further, appreciation of the roles of matrix constituents as morphogen cues, termed matrikines or matricryptins, are also now being directly exploited in biomaterial design. These insoluble topological cues can be presented at both micro- and nanoscales with specific fabrication techniques. Progress in development and molecular biology has described key roles for a range of biological molecules, such as proteins, lipids, and nucleic acids, to serve as morphogens promoting directed behavior in stem cells. Controlled-release systems involving encapsulation of bioactive agents within polymeric carriers are enabling utilization of soluble cues. Using our efforts at dental craniofacial tissue engineering, this narrative review focuses on outlining specific biomaterial fabrication techniques, such as electrospinning, gas foaming, and 3D printing used in combination with polymeric nano- or microspheres. These avenues are providing unprecedented therapeutic opportunities for precision bioengineering for regenerative applications.
Collapse
Affiliation(s)
- Saeed Ur Rahman
- Departments of Oral Biology and Biomedical Engineering, School of Dentistry, University at Buffalo, Buffalo, NY 14214, USA.
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad, Lahore Campus, Lahore 54000, Pakistan.
| | - Malvika Nagrath
- Departments of Oral Biology and Biomedical Engineering, School of Dentistry, University at Buffalo, Buffalo, NY 14214, USA.
- Department of Biomedical Engineering, Ryerson University, Toronto, ON M5B 2K3, Canada.
| | - Sasikumar Ponnusamy
- Departments of Oral Biology and Biomedical Engineering, School of Dentistry, University at Buffalo, Buffalo, NY 14214, USA.
| | - Praveen R Arany
- Departments of Oral Biology and Biomedical Engineering, School of Dentistry, University at Buffalo, Buffalo, NY 14214, USA.
| |
Collapse
|
25
|
Khodabukus A, Prabhu N, Wang J, Bursac N. In Vitro Tissue-Engineered Skeletal Muscle Models for Studying Muscle Physiology and Disease. Adv Healthc Mater 2018; 7:e1701498. [PMID: 29696831 PMCID: PMC6105407 DOI: 10.1002/adhm.201701498] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 02/18/2018] [Indexed: 12/18/2022]
Abstract
Healthy skeletal muscle possesses the extraordinary ability to regenerate in response to small-scale injuries; however, this self-repair capacity becomes overwhelmed with aging, genetic myopathies, and large muscle loss. The failure of small animal models to accurately replicate human muscle disease, injury and to predict clinically-relevant drug responses has driven the development of high fidelity in vitro skeletal muscle models. Herein, the progress made and challenges ahead in engineering biomimetic human skeletal muscle tissues that can recapitulate muscle development, genetic diseases, regeneration, and drug response is discussed. Bioengineering approaches used to improve engineered muscle structure and function as well as the functionality of satellite cells to allow modeling muscle regeneration in vitro are also highlighted. Next, a historical overview on the generation of skeletal muscle cells and tissues from human pluripotent stem cells, and a discussion on the potential of these approaches to model and treat genetic diseases such as Duchenne muscular dystrophy, is provided. Finally, the need to integrate multiorgan microphysiological systems to generate improved drug discovery technologies with the potential to complement or supersede current preclinical animal models of muscle disease is described.
Collapse
Affiliation(s)
- Alastair Khodabukus
- Department of Biomedical Engineering Duke University 101 Science Drive, FCIEMAS 1427, Durham, NC 27708-90281, USA
| | - Neel Prabhu
- Department of Biomedical Engineering Duke University 101 Science Drive, FCIEMAS 1427, Durham, NC 27708-90281, USA
| | - Jason Wang
- Department of Biomedical Engineering Duke University 101 Science Drive, FCIEMAS 1427, Durham, NC 27708-90281, USA
| | - Nenad Bursac
- Department of Biomedical Engineering Duke University 101 Science Drive, FCIEMAS 1427, Durham, NC 27708-90281, USA
| |
Collapse
|
26
|
Dinulovic I, Furrer R, Handschin C. Plasticity of the Muscle Stem Cell Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1041:141-169. [PMID: 29204832 DOI: 10.1007/978-3-319-69194-7_8] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Satellite cells (SCs) are adult muscle stem cells capable of repairing damaged and creating new muscle tissue throughout life. Their functionality is tightly controlled by a microenvironment composed of a wide variety of factors, such as numerous secreted molecules and different cell types, including blood vessels, oxygen, hormones, motor neurons, immune cells, cytokines, fibroblasts, growth factors, myofibers, myofiber metabolism, the extracellular matrix and tissue stiffness. This complex niche controls SC biology-quiescence, activation, proliferation, differentiation or renewal and return to quiescence. In this review, we attempt to give a brief overview of the most important players in the niche and their mutual interaction with SCs. We address the importance of the niche to SC behavior under physiological and pathological conditions, and finally survey the significance of an artificial niche both for basic and translational research purposes.
Collapse
|
27
|
van der Wal E, Herrero-Hernandez P, Wan R, Broeders M, In 't Groen SLM, van Gestel TJM, van IJcken WFJ, Cheung TH, van der Ploeg AT, Schaaf GJ, Pijnappel WWMP. Large-Scale Expansion of Human iPSC-Derived Skeletal Muscle Cells for Disease Modeling and Cell-Based Therapeutic Strategies. Stem Cell Reports 2018; 10:1975-1990. [PMID: 29731431 PMCID: PMC5993675 DOI: 10.1016/j.stemcr.2018.04.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 03/31/2018] [Accepted: 04/03/2018] [Indexed: 01/06/2023] Open
Abstract
Although skeletal muscle cells can be generated from human induced pluripotent stem cells (iPSCs), transgene-free protocols include only limited options for their purification and expansion. In this study, we found that fluorescence-activated cell sorting-purified myogenic progenitors generated from healthy controls and Pompe disease iPSCs can be robustly expanded as much as 5 × 1011-fold. At all steps during expansion, cells could be cryopreserved or differentiated into myotubes with a high fusion index. In vitro, cells were amenable to maturation into striated and contractile myofibers. Insertion of acid α-glucosidase cDNA into the AAVS1 locus in iPSCs using CRISPR/Cas9 prevented glycogen accumulation in myotubes generated from a patient with classic infantile Pompe disease. In vivo, the expression of human-specific nuclear and sarcolemmar antigens indicated that myogenic progenitors engraft into murine muscle to form human myofibers. This protocol is useful for modeling of skeletal muscle disorders and for using patient-derived, gene-corrected cells to develop cell-based strategies. Transgene-free protocol for generation and expansion of myogenic progenitors Differentiation into contractile skeletal muscle cells in vitro Correction of glycogen accumulation in Pompe disease using CRISPR/cas9 Contribution to muscle regeneration in vivo
Collapse
Affiliation(s)
- Erik van der Wal
- Department of Clinical Genetics, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands; Department of Pediatrics, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands; Center for Lysosomal and Metabolic Diseases, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands
| | - Pablo Herrero-Hernandez
- Department of Clinical Genetics, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands; Department of Pediatrics, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands; Center for Lysosomal and Metabolic Diseases, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands
| | - Raymond Wan
- Division of Life Science, Center for Stem Cell Research, Center of Systems Biology and Human Health, State Key Laboratory in Molecular Neuroscience, Hong Kong University of Science & Technology, Clear Water Bay, Kowloon, Hong Kong 999077, China
| | - Mike Broeders
- Department of Clinical Genetics, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands; Department of Pediatrics, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands; Center for Lysosomal and Metabolic Diseases, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands
| | - Stijn L M In 't Groen
- Department of Clinical Genetics, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands; Department of Pediatrics, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands; Center for Lysosomal and Metabolic Diseases, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands
| | - Tom J M van Gestel
- Department of Clinical Genetics, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands; Department of Pediatrics, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands; Center for Lysosomal and Metabolic Diseases, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands
| | - Wilfred F J van IJcken
- Erasmus Center for Biomics, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - Tom H Cheung
- Division of Life Science, Center for Stem Cell Research, Center of Systems Biology and Human Health, State Key Laboratory in Molecular Neuroscience, Hong Kong University of Science & Technology, Clear Water Bay, Kowloon, Hong Kong 999077, China
| | - Ans T van der Ploeg
- Department of Pediatrics, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands; Center for Lysosomal and Metabolic Diseases, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands
| | - Gerben J Schaaf
- Department of Clinical Genetics, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands; Department of Pediatrics, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands; Center for Lysosomal and Metabolic Diseases, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands
| | - W W M Pim Pijnappel
- Department of Clinical Genetics, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands; Department of Pediatrics, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands; Center for Lysosomal and Metabolic Diseases, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands.
| |
Collapse
|
28
|
Somers SM, Spector AA, DiGirolamo DJ, Grayson WL. Biophysical Stimulation for Engineering Functional Skeletal Muscle. TISSUE ENGINEERING PART B-REVIEWS 2018; 23:362-372. [PMID: 28401807 DOI: 10.1089/ten.teb.2016.0444] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tissue engineering is a promising therapeutic strategy to regenerate skeletal muscle. However, ex vivo cultivation methods typically result in a low differentiation efficiency of stem cells as well as grafts that resemble the native tissues morphologically, but lack contractile function. The application of biomimetic tensile strain provides a potent stimulus for enhancing myogenic differentiation and engineering functional skeletal muscle grafts. We reviewed integrin-dependent mechanisms that potentially link mechanotransduction pathways to the upregulation of myogenic genes. Yet, gaps in our understanding make it challenging to use these pathways to theoretically determine optimal ex vivo strain regimens. A multitude of strain protocols have been applied to in vitro cultures for the cultivation of myogenic progenitors (adipose- and bone marrow-derived stem cells and satellite cells) and transformed murine myoblasts, C2C12s. Strain regimens are characterized by orientation, amplitude, and time-dependent factors (effective frequency, duration, and the rest period between successive strain cycles). Analysis of published data has identified possible minimum/maximum values for these parameters and suggests that uniaxial strains may be more potent than biaxial strains, possibly because they more closely mimic physiologic strain profiles. The application of these biophysical stimuli for engineering 3D skeletal muscle grafts is nontrivial and typically requires custom-designed bioreactors used in combination with biomaterial scaffolds. Consideration of the physical properties of these scaffolds is critical for effective transmission of the applied strains to encapsulated cells. Taken together, these studies demonstrate that biomimetic tensile strain generally results in improved myogenic outcomes in myogenic progenitors and differentiated myoblasts. However, for 3D systems, the optimization of the strain regimen may require the entire system including cells, biomaterials, and bioreactor, to be considered in tandem.
Collapse
Affiliation(s)
- Sarah M Somers
- 1 Department of Biomedical Engineering, Johns Hopkins University School of Medicine , Baltimore, Maryland.,2 Translational Tissue Engineering Center, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Alexander A Spector
- 1 Department of Biomedical Engineering, Johns Hopkins University School of Medicine , Baltimore, Maryland.,2 Translational Tissue Engineering Center, Johns Hopkins University School of Medicine , Baltimore, Maryland.,3 Institute for Nanobiotechnology (INBT), Johns Hopkins University Whiting School of Engineering , Baltimore, Maryland
| | - Douglas J DiGirolamo
- 4 Department of Orthopedics, Johns Hopkins University School of Medicine , Baltimore Maryland
| | - Warren L Grayson
- 1 Department of Biomedical Engineering, Johns Hopkins University School of Medicine , Baltimore, Maryland.,2 Translational Tissue Engineering Center, Johns Hopkins University School of Medicine , Baltimore, Maryland.,3 Institute for Nanobiotechnology (INBT), Johns Hopkins University Whiting School of Engineering , Baltimore, Maryland.,5 Department of Material Sciences and Engineering, Johns Hopkins University , Whiting School of Engineering, Baltimore, Maryland
| |
Collapse
|
29
|
Quarta M. Volumetric muscle loss: Including nerves into the equation. Muscle Nerve 2018; 57:705-706. [PMID: 29365348 DOI: 10.1002/mus.26080] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 01/19/2018] [Accepted: 01/22/2018] [Indexed: 01/17/2023]
Affiliation(s)
- Marco Quarta
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.,Paul F. Glenn Laboratories for the Biology of Aging, Stanford University School of Medicine, Stanford, California, USA.,Center for Tissue Regeneration, Restoration and Repair, Veterans Affairs Hospital, 3801 Miranda Avenue, Palo Alto, California, 94036, USA
| |
Collapse
|
30
|
Young J, Margaron Y, Fernandes M, Duchemin-Pelletier E, Michaud J, Flaender M, Lorintiu O, Degot S, Poydenot P. MyoScreen, a High-Throughput Phenotypic Screening Platform Enabling Muscle Drug Discovery. SLAS DISCOVERY 2018; 23:790-806. [PMID: 29498891 DOI: 10.1177/2472555218761102] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Despite the need for more effective drug treatments to address muscle atrophy and disease, physiologically accurate in vitro screening models and higher information content preclinical assays that aid in the discovery and development of novel therapies are lacking. To this end, MyoScreen was developed: a robust and versatile high-throughput high-content screening (HT/HCS) platform that integrates a physiologically and pharmacologically relevant micropatterned human primary skeletal muscle model with a panel of pertinent phenotypic and functional assays. MyoScreen myotubes form aligned, striated myofibers, and they show nerve-independent accumulation of acetylcholine receptors (AChRs), excitation-contraction coupling (ECC) properties characteristic of adult skeletal muscle and contraction in response to chemical stimulation. Reproducibility and sensitivity of the fully automated MyoScreen platform are highlighted in assays that quantitatively measure myogenesis, hypertrophy and atrophy, AChR clusterization, and intracellular calcium release dynamics, as well as integrating contractility data. A primary screen of 2560 compounds to identify stimulators of myofiber regeneration and repair, followed by further biological characterization of two hits, validates MyoScreen for the discovery and testing of novel therapeutics. MyoScreen is an improvement of current in vitro muscle models, enabling a more predictive screening strategy for preclinical selection of the most efficacious new chemical entities earlier in the discovery pipeline process.
Collapse
|
31
|
Polstein LR, Juhas M, Hanna G, Bursac N, Gersbach CA. An Engineered Optogenetic Switch for Spatiotemporal Control of Gene Expression, Cell Differentiation, and Tissue Morphogenesis. ACS Synth Biol 2017; 6:2003-2013. [PMID: 28793186 DOI: 10.1021/acssynbio.7b00147] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The precise spatial and temporal control of gene expression, cell differentiation, and tissue morphogenesis has widespread application in regenerative medicine and the study of tissue development. In this work, we applied optogenetics to control cell differentiation and new tissue formation. Specifically, we engineered an optogenetic "on" switch that provides permanent transgene expression following a transient dose of blue light illumination. To demonstrate its utility in controlling cell differentiation and reprogramming, we incorporated an engineered form of the master myogenic factor MyoD into this system in multipotent cells. Illumination of cells with blue light activated myogenic differentiation, including upregulation of myogenic markers and fusion into multinucleated myotubes. Cell differentiation was spatially patterned by illumination of cell cultures through a photomask. To demonstrate the application of the system to controlling in vivo tissue development, the light inducible switch was used to control the expression of VEGF and angiopoietin-1, which induced angiogenic sprouting in a mouse dorsal window chamber model. Live intravital microscopy showed illumination-dependent increases in blood-perfused microvasculature. This optogenetic switch is broadly useful for applications in which sustained and patterned gene expression is desired following transient induction, including tissue engineering, gene therapy, synthetic biology, and fundamental studies of morphogenesis.
Collapse
Affiliation(s)
- Lauren R. Polstein
- Department of Biomedical
Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Mark Juhas
- Department of Biomedical
Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Gabi Hanna
- Department of Radiation Oncology and Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Nenad Bursac
- Department of Biomedical
Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Charles A. Gersbach
- Department of Biomedical
Engineering, Duke University, Durham, North Carolina 27708, United States
- Center for
Genomic and Computational Biology, Duke University, Durham, North Carolina 27708, United States
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, North Carolina 27710, United States
| |
Collapse
|
32
|
Abstract
Skeletal muscle is the largest tissue in the body and loss of its function or its regenerative properties results in debilitating musculoskeletal disorders. Understanding the mechanisms that drive skeletal muscle formation will not only help to unravel the molecular basis of skeletal muscle diseases, but also provide a roadmap for recapitulating skeletal myogenesis in vitro from pluripotent stem cells (PSCs). PSCs have become an important tool for probing developmental questions, while differentiated cell types allow the development of novel therapeutic strategies. In this Review, we provide a comprehensive overview of skeletal myogenesis from the earliest premyogenic progenitor stage to terminally differentiated myofibers, and discuss how this knowledge has been applied to differentiate PSCs into muscle fibers and their progenitors in vitro.
Collapse
Affiliation(s)
- Jérome Chal
- Department of Pathology, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Harvard Stem Cell Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Olivier Pourquié
- Department of Pathology, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, Boston, MA 02115, USA .,Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Harvard Stem Cell Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, 67400 Illkirch-Graffenstaden, France
| |
Collapse
|
33
|
Redondo PA, Pavlou M, Loizidou M, Cheema U. Elements of the niche for adult stem cell expansion. J Tissue Eng 2017; 8:2041731417725464. [PMID: 28890779 PMCID: PMC5574483 DOI: 10.1177/2041731417725464] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 07/18/2017] [Indexed: 12/21/2022] Open
Abstract
Adult stem cells are crucial for tissue homeostasis. These cells reside within exclusive locations in tissues, termed niches, which protect adult stem cell fidelity and regulate their many functions through biophysical-, biochemical- and cellular-mediated mechanisms. There is a growing understanding of how these mechanisms and their components contribute towards maintaining stem cell quiescence, self-renewal, expansion and differentiation patterns. In vitro expansion of adult stem cells is a powerful tool for understanding stem cell biology, and for tissue engineering and regenerative medicine applications. However, it is technically challenging, since adult stem cell removal from their native microenvironment has negative repercussions on their sustainability. In this review, we overview specific elements of the biomimetic niche and how recreating such elements can help in vitro propagation of adult stem cells.
Collapse
Affiliation(s)
- Patricia A Redondo
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Marina Pavlou
- Institute of Orthopaedics & Musculoskeletal Science, University College London, London, UK
| | - Marilena Loizidou
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Umber Cheema
- Institute of Orthopaedics & Musculoskeletal Science, University College London, London, UK
| |
Collapse
|
34
|
Ikeda K, Ito A, Imada R, Sato M, Kawabe Y, Kamihira M. In vitro drug testing based on contractile activity of C2C12 cells in an epigenetic drug model. Sci Rep 2017; 7:44570. [PMID: 28300163 PMCID: PMC5353687 DOI: 10.1038/srep44570] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 02/09/2017] [Indexed: 11/23/2022] Open
Abstract
Skeletal muscle tissue engineering holds great promise for pharmacological studies. Herein, we demonstrated an in vitro drug testing system using tissue-engineered skeletal muscle constructs. In response to epigenetic drugs, myotube differentiation of C2C12 myoblast cells was promoted in two-dimensional cell cultures, but the levels of contractile force generation of tissue-engineered skeletal muscle constructs prepared by three-dimensional cell cultures were not correlated with the levels of myotube differentiation in two-dimensional cell cultures. In contrast, sarcomere formation and contractile activity in two-dimensional cell cultures were highly correlated with contractile force generation of tissue-engineered skeletal muscle constructs. Among the epigenetic drugs tested, trichostatin A significantly improved contractile force generation of tissue-engineered skeletal muscle constructs. Follistatin expression was also enhanced by trichostatin A treatment, suggesting the importance of follistatin in sarcomere formation of muscular tissues. These observations indicate that contractility data are indispensable for in vitro drug screening.
Collapse
Affiliation(s)
- Kazushi Ikeda
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Akira Ito
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Ryusuke Imada
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Masanori Sato
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Yoshinori Kawabe
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Masamichi Kamihira
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.,Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| |
Collapse
|
35
|
Chal J, Al Tanoury Z, Hestin M, Gobert B, Aivio S, Hick A, Cherrier T, Nesmith AP, Parker KK, Pourquié O. Generation of human muscle fibers and satellite-like cells from human pluripotent stem cells in vitro. Nat Protoc 2016; 11:1833-50. [PMID: 27583644 DOI: 10.1038/nprot.2016.110] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Progress toward finding a cure for muscle diseases has been slow because of the absence of relevant cellular models and the lack of a reliable source of muscle progenitors for biomedical investigation. Here we report an optimized serum-free differentiation protocol to efficiently produce striated, millimeter-long muscle fibers together with satellite-like cells from human pluripotent stem cells (hPSCs) in vitro. By mimicking key signaling events leading to muscle formation in the embryo, in particular the dual modulation of Wnt and bone morphogenetic protein (BMP) pathway signaling, this directed differentiation protocol avoids the requirement for genetic modifications or cell sorting. Robust myogenesis can be achieved in vitro within 1 month by personnel experienced in hPSC culture. The differentiating culture can be subcultured to produce large amounts of myogenic progenitors amenable to numerous downstream applications. Beyond the study of myogenesis, this differentiation method offers an attractive platform for the development of relevant in vitro models of muscle dystrophies and drug screening strategies, as well as providing a source of cells for tissue engineering and cell therapy approaches.
Collapse
Affiliation(s)
- Jérome Chal
- Institut de Génétique et de Biologie Moléculaireet Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch-Graffenstaden, France
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Boston, Massachusetts, USA
| | - Ziad Al Tanoury
- Institut de Génétique et de Biologie Moléculaireet Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch-Graffenstaden, France
| | - Marie Hestin
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Boston, Massachusetts, USA
| | - Bénédicte Gobert
- Institut de Génétique et de Biologie Moléculaireet Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch-Graffenstaden, France
| | - Suvi Aivio
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Boston, Massachusetts, USA
| | - Aurore Hick
- Anagenesis Biotechnologies, Parc d'innovation, Illkirch-Graffenstaden, France
| | - Thomas Cherrier
- Institut de Génétique et de Biologie Moléculaireet Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch-Graffenstaden, France
| | - Alexander P Nesmith
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
| | - Kevin K Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
| | - Olivier Pourquié
- Institut de Génétique et de Biologie Moléculaireet Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch-Graffenstaden, France
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Boston, Massachusetts, USA
| |
Collapse
|
36
|
Matrix Metalloproteinase Responsive Delivery of Myostatin Inhibitors. Pharm Res 2016; 34:58-72. [DOI: 10.1007/s11095-016-2038-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 09/06/2016] [Indexed: 12/21/2022]
|
37
|
Bersini S, Arrigoni C, Lopa S, Bongio M, Martin I, Moretti M. Engineered miniaturized models of musculoskeletal diseases. Drug Discov Today 2016; 21:1429-1436. [PMID: 27132520 DOI: 10.1016/j.drudis.2016.04.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 03/31/2016] [Accepted: 04/18/2016] [Indexed: 01/07/2023]
Abstract
The musculoskeletal system is an incredible machine that protects, supports and moves the human body. However, several diseases can limit its functionality, compromising patient quality of life. Designing novel pathological models would help to clarify the mechanisms driving such diseases, identify new biomarkers and screen potential drug candidates. Miniaturized models in particular can mimic the structure and function of basic tissue units within highly controlled microenvironments, overcoming the limitations of traditional macroscale models and complementing animal studies, which despite being closer to the in vivo situation, are affected by species-specific differences. Here, we discuss the miniaturized models engineered over the past few years to analyze osteochondral and skeletal muscle pathologies, demonstrating how the rationale design of novel systems could provide key insights into the pathological mechanisms behind diseases of the musculoskeletal system.
Collapse
Affiliation(s)
- Simone Bersini
- Cell and Tissue Engineering Lab, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
| | - Chiara Arrigoni
- Cell and Tissue Engineering Lab, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
| | - Silvia Lopa
- Cell and Tissue Engineering Lab, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
| | - Matilde Bongio
- Cell and Tissue Engineering Lab, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
| | - Ivan Martin
- Department of Surgery and Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Matteo Moretti
- Cell and Tissue Engineering Lab, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy; Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale (EOC), Lugano, Switzerland; Swiss Institute for Regenerative Medicine, Lugano, Switzerland; Fondazione Cardiocentro Ticino, Lugano, Switzerland.
| |
Collapse
|